1
|
Teng W, Ling Y, Long N, Cen W, Zhang H, Jiang L, Liu J, Zhou X, Chu L. Repurposing flubendazole for glioblastoma ferroptosis by affecting xCT and TFRC proteins. J Cell Mol Med 2024; 28:e70188. [PMID: 39543084 PMCID: PMC11563996 DOI: 10.1111/jcmm.70188] [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: 01/14/2024] [Revised: 10/05/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
New uses of old drugs hold great promise for clinical translation. Flubendazole, an FDA-approved antiparasitic drug, has been shown to target p53 and promote apoptosis in glioblastoma (GBM) cells. However, its damaging mechanism in GBM remains elusive. Herein, we explored the ferroptosis-inducing ability of flubendazole on GBM cells. After treating glioma cell lines U251 and LN229 with the flubendazole (DMSO <1‰), cell viability was inhibited in a concentration-dependent manner (IC50 for LN229 = 0.5331 μM, IC50 for U251 = 0.6809 μM), attributed to the induction of ferroptosis, as evidenced by increased MDA levels, accumulation of ROS and lipid peroxides, change in mitochondrial membrane potential and structure. Protein analysis related to ferroptosis showed upregulation of TFRC, DMT1 and p53, alongside downregulation of xCT, FHC and GPX4 (p < 0.05). All-atom docking studies demonstrated that flubendazole bound closely with xCT, and TFRC, validating its role in inducing glioma ferroptosis via modulation of these proteins. Notably, flubendazole could damage the glioblastoma stem cells (GSC) that are typically resistant to other therapies, thereby possessing advantages in stopping glioma recurrence. This study delved into the mechanisms of flubendazole-induced ferroptosis in glioma, broadening its application and providing new ideas for new uses of other old drugs.
Collapse
Affiliation(s)
- Wei Teng
- Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
- Department of Clinical MedicineGuizhou Medical UniversityGuiyangGuizhouChina
| | - Yuanguo Ling
- Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
- Department of Clinical MedicineGuizhou Medical UniversityGuiyangGuizhouChina
| | - Niya Long
- Department of Clinical MedicineGuizhou Medical UniversityGuiyangGuizhouChina
| | - Wu Cen
- Department of Clinical MedicineGuizhou Medical UniversityGuiyangGuizhouChina
| | - Hongzhi Zhang
- Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Lishi Jiang
- Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Jian Liu
- Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
- Department of Neurosurgery, Guizhou Provincial People's HospitalGuiyangGuizhouChina
| | - Xingwang Zhou
- Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
- Department of Clinical MedicineGuizhou Medical UniversityGuiyangGuizhouChina
| | - Liangzhao Chu
- Department of NeurosurgeryThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
- Department of Clinical MedicineGuizhou Medical UniversityGuiyangGuizhouChina
| |
Collapse
|
2
|
Wang Z, Chen F, Cao Y, Zhang F, Sun L, Yang C, Xie X, Wu Z, Sun M, Ma F, Shao D, Leong KW, Pei R. An Engineered Nanoplatform with Tropism Toward Irradiated Glioblastoma Augments Its Radioimmunotherapy Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314197. [PMID: 38713519 DOI: 10.1002/adma.202314197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/27/2024] [Indexed: 05/09/2024]
Abstract
Combining radiotherapy with immune checkpoint blockade therapy offers a promising approach to treat glioblastoma multiforme (GBM), yet challenges such as limited effectiveness and immune-related adverse events (irAEs) persist. These issues are largely due to the failure in targeting immunomodulators directly to the tumor microenvironment. To address this, a biomimetic nanoplatform that combines a genetically modified mesenchymal stem cell (MSC) membrane with a bioactive nanoparticle core for chemokine-directed radioimmunotherapy of GBM is developed. The CC chemokine receptor 2 (CCR2)-overexpressing MSC membrane acts as a tactical tentacle to achieve radiation-induced tropism toward the abundant chemokine (CC motif) ligand 2 (CCL2) in irradiated gliomas. The nanoparticle core, comprising diselenide-bridged mesoporous silica nanoparticles (MSNs) and PD-L1 antibodies (αPD-L1), enables X-ray-responsive drug release and radiosensitization. In two murine models with orthotopic GBM tumors, this nanoplatform reinvigorated immunogenic cell death, and augmented the efficacy and specificity of GBM radioimmunotherapy, with reduced occurrence of irAEs. This study suggests a promising radiation-induced tropism strategy for targeted drug delivery, and presents a potent nanoplatform that enhances the efficacy and safety of radio-immunotherapy.
Collapse
Affiliation(s)
- Zheng Wang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Yi Cao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Fan Zhang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Lina Sun
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Chao Yang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Ziping Wu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Madi Sun
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Fanshu Ma
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Dan Shao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| |
Collapse
|
3
|
Zhang G, Hu J, Li A, Zhang H, Guo Z, Li X, You Z, Wang Y, Jing Z. Ginsenoside Rg5 inhibits glioblastoma by activating ferroptosis via NR3C1/HSPB1/NCOA4. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155631. [PMID: 38640858 DOI: 10.1016/j.phymed.2024.155631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/02/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND The utilization of Chinese medicine as an adjunctive therapy for cancer has recently gained significant attention. Ferroptosis, a newly regulated cell death process depending on the ferrous ions, has been proved to be participated in glioma stem cells inactivation. PURPOSE We aim to study whether ginsenoside Rg5 exerted inhibitory effects on crucial aspects of glioma stem cells, including cell viability, tumor initiation, invasion, self-renewal ability, neurosphere formation, and stemness. METHODS Through comprehensive sequencing analysis, we identified a compelling association between ginsenoside Rg5 and the ferroptosis pathway, which was further validated through subsequent experiments demonstrating its ability to activate this pathway. RESULTS To elucidate the precise molecular targets affected by ginsenoside Rg5 in gliomas, we conducted an intersection analysis between differentially expressed genes obtained from sequencing and a database-predicted list of transcription factors and potential targets of ginsenoside Rg5. This rigorous approach led us to unequivocally confirm NR3C1 (Nuclear Receptor Subfamily 3 Group C Member 1) as a direct target of ginsenoside Rg5, a finding consistently supported by subsequent experimental investigations. Moreover, we uncovered NR3C1's capacity to transcriptionally regulate ferroptosis -related genes HSPB1 and NCOA4. Strikingly, ginsenoside Rg5 induced notable alterations in the expression levels of both HSPB1 (Heat Shock Protein Family B Member 1) and NCOA4 (Nuclear Receptor Coactivator 4). Finally, our intracranial xenograft assays served to reaffirm the inhibitory effect of ginsenoside Rg5 on the malignant progression of glioblastoma. CONCLUSION These collective findings strongly suggest that ginsenoside Rg5 hampers glioblastoma progression by activating ferroptosis through NR3C1, which subsequently modulates HSPB1 and NCOA4. Importantly, this novel therapeutic direction holds promise for advancing the treatment of glioblastoma.
Collapse
Affiliation(s)
- Guoqing Zhang
- Department of Neurosurgery, the First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Jinpeng Hu
- Department of Neurosurgery, the First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Ao Li
- Emergency department, Liaoning Provincial People Hospital, Shenyang, 110016, China
| | - Haiying Zhang
- International Education College, Liaoning University of Traditional Chinese Medicine, No. 79 Chongshan East Road, Shenyang, Liaoning, 110042, China
| | - Zhengting Guo
- Department of Neurosurgery, the First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Xinqiao Li
- Department of Neurosurgery, the First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Zinan You
- Department of Neurosurgery, the First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China
| | - Yongfeng Wang
- Department of Radiology, the First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, PR China.
| | - Zhitao Jing
- Department of Neurosurgery, the First Hospital of China Medical University, No. 155 North Nanjing Street, Shenyang, 110001, China.
| |
Collapse
|
4
|
Mao M, Wu Y, He Q. Recent advances in targeted drug delivery for the treatment of glioblastoma. NANOSCALE 2024; 16:8689-8707. [PMID: 38606460 DOI: 10.1039/d4nr01056f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Glioblastoma multiforme (GBM) is one of the highly malignant brain tumors characterized by significant morbidity and mortality. Despite the recent advancements in the treatment of GBM, major challenges persist in achieving controlled drug delivery to tumors. The management of GBM poses considerable difficulties primarily due to unresolved issues in the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB) and GBM microenvironment. These factors limit the uptake of anti-cancer drugs by the tumor, thus limiting the therapeutic options. Current breakthroughs in nanotechnology provide new prospects concerning unconventional drug delivery approaches for GBM treatment. Specifically, swimming nanorobots show great potential in active targeted delivery, owing to their autonomous propulsion and improved navigation capacities across biological barriers, which further facilitate the development of GBM-targeted strategies. This review presents an overview of technological progress in different drug administration methods for GBM. Additionally, the limitations in clinical translation and future research prospects in this field are also discussed. This review aims to provide a comprehensive guideline for researchers and offer perspectives on further development of new drug delivery therapies to combat GBM.
Collapse
Affiliation(s)
- Meng Mao
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
5
|
Wang Y, Wang B, Cao W, Xu X. PTX3 activates POSTN and promotes the progression of glioblastoma via the MAPK/ERK signalling axis. Biochem Biophys Res Commun 2024; 703:149665. [PMID: 38359612 DOI: 10.1016/j.bbrc.2024.149665] [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: 12/24/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Intrinsic brain tumours such as glioblastoma (GBM) are believed to develop from neuroglial stem or progenitor cells. GBM accounts for approximately half of gliomas. GBM has a poor prognosis and a low 5-year survival rate. Pentraxin 3 (PTX3) is overexpressed in GBM, but the potential mechanism is unclear. METHODS Glioblastoma data from the TCGA and CGGA databases were used to analyse PTX3 expression. Subsequently, in vivo and in vitro experiments were conducted to verify the effect of PTX3 silencing in glioma cells on EMT like process and GSC maintenance. The JASPAR database was used to predict the downstream genes of PTX3. POSTN is a novel target gene of PTX3 in gliomas, and this finding was validated using a luciferase reporter gene assay. Western blotting and KEGG enrichment analysis were used to predict the downstream pathway of POSTN, and it was found that the MAPK/ERK pathway might be related to the function of POSTN. RESULTS GBM tissues have higher levels of PTX3 expression than normal brain tissues (NBTs). In functional tests, PTX3 promoted the EMT like process of GBM cells while maintaining the stem cell characteristics of GBM stem cells and enhancing their self-renewal. Moreover, we performed a dual luciferase reporter experiment to confirm that PTX3 binds to the POSTN promoter region. In addition, the expression of key proteins in the MAPK/ERK signalling pathway was increased after PTX3 overexpression. CONCLUSION POSTN is a direct target of PTX3 that promotes GBM growth via the MAPK/ERK signalling pathway.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Binbin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
| | - Wenping Cao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| | - Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China.
| |
Collapse
|
6
|
Wang Y, Wang B, Cao W, Xu X. TGF-β-activated circRYK drives glioblastoma progression by increasing VLDLR mRNA expression and stability in a ceRNA- and RBP-dependent manner. J Exp Clin Cancer Res 2024; 43:73. [PMID: 38454465 PMCID: PMC10921701 DOI: 10.1186/s13046-024-03000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/01/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The TGF-β signalling pathway is intricately associated with the progression of glioblastoma (GBM). The objective of this study was to examine the role of circRNAs in the TGF-β signalling pathway. METHODS In our research, we used transcriptome analysis to search for circRNAs that were activated by TGF-β. After confirming the expression pattern of the selected circRYK, we carried out in vitro and in vivo cell function assays. The underlying mechanisms were analysed via RNA pull-down, luciferase reporter, and RNA immunoprecipitation assays. RESULTS CircRYK expression was markedly elevated in GBM, and this phenotype was strongly associated with a poor prognosis. Functionally, circRYK promotes epithelial-mesenchymal transition and GSC maintenance in GBM. Mechanistically, circRYK sponges miR-330-5p and promotes the expression of the oncogene VLDLR. In addition, circRYK could enhance the stability of VLDLR mRNA via the RNA-binding protein HuR. CONCLUSION Our findings show that TGF-β promotes epithelial-mesenchymal transition and GSC maintenance in GBM through the circRYK-VLDLR axis, which may provide a new therapeutic target for the treatment of GBM.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China
| | - Binbin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China
| | - Wenping Cao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China.
| | - Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China.
| |
Collapse
|
7
|
Li D, Cai S, Wang P, Cheng H, Cheng B, Zhang Y, Liu G. Innovative Design Strategies Advance Biomedical Applications of Phthalocyanines. Adv Healthc Mater 2023; 12:e2300263. [PMID: 37039069 DOI: 10.1002/adhm.202300263] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/30/2023] [Indexed: 04/12/2023]
Abstract
Owing to their long absorption wavelengths, high molar absorptivity, and tunable photosensitivity, phthalocyanines have been widely used in photodynamic therapy (PDT). However, phthalocyanines still face the drawbacks of poor targeting, "always-on" photosensitizing properties, and unsatisfactory therapeutic efficiency, which limit their wide applications in biomedical fields. Thus, new design strategies such as modification of targeting molecules, formation of nanoparticles, and activating photosensitizers are developed to improve the above defects. Notably, recent studies have shown that novel phthalocyanines are not only used in fluorescence imaging and PDT, but also in photoacoustic imaging, photothermal imaging, sonodynamic therapy, and photothermal therapy. This review focuses on recent design strategies, applications in biomedicine, and clinical development of phthalocyanines, providing ideas and references for the design and application of phthalocyanine, so as to promote their future transformation into clinical applications.
Collapse
Affiliation(s)
- Dong Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021, China
| | - Shundong Cai
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Peiyu Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Bingwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- Shen Zhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
8
|
Bao Y, Lu W. Targeting cerebral diseases with enhanced delivery of therapeutic proteins across the blood-brain barrier. Expert Opin Drug Deliv 2023; 20:1681-1698. [PMID: 36945117 DOI: 10.1080/17425247.2023.2193390] [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: 01/14/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Cerebral diseases have been threatening public physical and psychological health in the recent years. With the existence of the blood-brain barrier (BBB), it is particularly hard for therapeutic proteins like peptides, enzymes, antibodies, etc. to enter the central nervous system (CNS) and function in diagnosis and treatment in cerebral diseases. Fortunately, the past decade has witnessed some emerging strategies of delivering macromolecular therapeutic proteins across the BBB. AREAS COVERED Based on the structure, functions, and substances transport mechanisms, various enhanced delivery strategies of therapeutic proteins were reviewed, categorized by molecule-mediated delivery strategies, carrier-mediated delivery strategies, and other delivery strategies. EXPERT OPINION As for molecule-mediated delivery strategies, development of genetic engineering technology, optimization of protein expression and purification techniques, and mature of quality control systems all help to realize large-scale production of recombinant antibodies, making it possible to apply to the clinical practice. In terms of carrier-mediated delivery strategies and others, although nano-carriers/adeno-associated virus (AAV) are also promising candidates for delivering therapeutic proteins or genes across the BBB, some issues still remain to be further investigated, including safety concerns related to applied materials, large-scale production costs, quality control standards, combination therapies with auxiliary delivery strategies like focused ultrasound, etc.
Collapse
Affiliation(s)
- Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
- Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd. Lingang of Shanghai, China
| |
Collapse
|
9
|
Chartouni A, Mouawad A, Boutros M, Attieh F, Medawar N, Kourie HR. Mesenchymal stem cells: a trojan horse to treat glioblastoma. Invest New Drugs 2023; 41:240-250. [PMID: 37017885 DOI: 10.1007/s10637-023-01352-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/23/2023] [Indexed: 04/06/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and lethal primary tumor of the central nervous system. What makes it so dreadful is the very low survival rate, despite the existence of a standard treatment plan. An innovative and more effective way to treat glioblastoma based on Mesenchymal Stem Cells (MSCs) has been explored recently. MSCs are a group of endogenous multipotent stem cells that could mainly be harvested from adipose tissue, bone marrow, and umbilical cord. Having the ability to migrate toward the tumor using multiple types of binding receptors, they could be used either as a direct treatment (whether they are enhanced or not) or as a delivery vehicle carrying various anti-tumoral agents. Some of these agents are: chemotherapy drugs, prodrug activating therapy, oncolytic viruses, nanoparticles, human artificial chromosome… Promising results have started to surface; however, more evidence is needed to perfect their use as a glioblastoma multiforme treatment option. Alternative treatment, using unloaded or loaded MSCs, leading to a better outcome.
Collapse
Affiliation(s)
- Antoine Chartouni
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon.
| | - Antoine Mouawad
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
| | - Marc Boutros
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
| | - Fouad Attieh
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
| | - Nicolas Medawar
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
| | - Hampig Raphaël Kourie
- Faculty of Medicine, Saint Joseph University of Beirut, Riad El Solh, 11 - 5076, Beirut, Lebanon
- Department of hematology-oncology, Faculty of medicine, Saint-joseph university of beirut, Beirut, Lebanon
| |
Collapse
|
10
|
Wang Y, Wang B, Zhou F, Lv K, Xu X, Cao W. CircNDC80 promotes glioblastoma multiforme tumorigenesis via the miR-139-5p/ECE1 pathway. J Transl Med 2023; 21:22. [PMID: 36635757 PMCID: PMC9837923 DOI: 10.1186/s12967-022-03852-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/24/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) have been shown to be essential for the emergence and growth of different cancers. However, further research is required to validate the function of circRNA in glioblastoma (GBM). METHODS CircNDC80 expression in both normal brain tissues (NBTs) and glioma tissues was determined using real-time PCR. The impact of circNDC80 on GBM cell proliferation, migration, and invasion was then confirmed by CCK-8, colony formation, EdU incorporation, Transwell, and wound healing assays. To determine how circNDC80 affects the capacity of glioma stem cells (GSCs) to maintain their stemness and self-renewal, a CellTiter-Glo assay, clonogenic assay and extreme limiting dilution assay were utilized. To ascertain the impact of circNDC80 in vivo, intracranial xenograft models were established. RESULTS When compared to NBT, glioblastoma tissue had a higher level of circNDC80 expression. In functional assays, circNDC80 promoted glioblastoma cell proliferation, migration, and invasion, while sustaining the stemness and fostering the self-renewal of glioma stem cells. In addition, a dual luciferase reporter assay and circRIP were used to verify that circNDC80 simultaneously affects the expression of ECE1 mRNA by sponging miR-139-5p, and a rescue experiment was used to verify the above results further. CONCLUSIONS According to our research, circNDC80 is an oncogenic factor that promotes glioblastoma through the miR-139-5p/ECE1 pathway. This implies that circNDC80 may be employed as a novel therapeutic target and a possible predictive biomarker.
Collapse
Affiliation(s)
- Yuhang Wang
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000 Jiangsu China
| | - Binbin Wang
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000 Jiangsu China
| | - Fengqi Zhou
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000 Jiangsu China
| | - Kun Lv
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000 Jiangsu China
| | - Xiupeng Xu
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000 Jiangsu China
| | - Wenping Cao
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000 Jiangsu China
| |
Collapse
|
11
|
Pham TC, Nguyen VN, Choi Y, Lee S, Yoon J. Recent Strategies to Develop Innovative Photosensitizers for Enhanced Photodynamic Therapy. Chem Rev 2021; 121:13454-13619. [PMID: 34582186 DOI: 10.1021/acs.chemrev.1c00381] [Citation(s) in RCA: 803] [Impact Index Per Article: 200.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review presents a robust strategy to design photosensitizers (PSs) for various species. Photodynamic therapy (PDT) is a photochemical-based treatment approach that involves the use of light combined with a light-activated chemical, referred to as a PS. Attractively, PDT is one of the alternatives to conventional cancer treatment due to its noninvasive nature, high cure rates, and low side effects. PSs play an important factor in photoinduced reactive oxygen species (ROS) generation. Although the concept of photosensitizer-based photodynamic therapy has been widely adopted for clinical trials and bioimaging, until now, to our surprise, there has been no relevant review article on rational designs of organic PSs for PDT. Furthermore, most of published review articles in PDT focused on nanomaterials and nanotechnology based on traditional PSs. Therefore, this review aimed at reporting recent strategies to develop innovative organic photosensitizers for enhanced photodynamic therapy, with each example described in detail instead of providing only a general overview, as is typically done in previous reviews of PDT, to provide intuitive, vivid, and specific insights to the readers.
Collapse
Affiliation(s)
- Thanh Chung Pham
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Van-Nghia Nguyen
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Yeonghwan Choi
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Songyi Lee
- Department of Chemistry, Pukyong National University, Busan 48513, Korea.,Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
12
|
Sokolov AV, Dostdar SA, Attwood MM, Krasilnikova AA, Ilina AA, Nabieva AS, Lisitsyna AA, Chubarev VN, Tarasov VV, Schiöth HB. Brain Cancer Drug Discovery: Clinical Trials, Drug Classes, Targets, and Combinatorial Therapies. Pharmacol Rev 2021; 73:1-32. [PMID: 34663683 DOI: 10.1124/pharmrev.121.000317] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Brain cancer is a formidable challenge for drug development, and drugs derived from many cutting-edge technologies are being tested in clinical trials. We manually characterized 981 clinical trials on brain tumors that were registered in ClinicalTrials.gov from 2010 to 2020. We identified 582 unique therapeutic entities targeting 581 unique drug targets and 557 unique treatment combinations involving drugs. We performed the classification of both the drugs and drug targets based on pharmacological and structural classifications. Our analysis demonstrates a large diversity of agents and targets. Currently, we identified 32 different pharmacological directions for therapies that are based on 42 structural classes of agents. Our analysis shows that kinase inhibitors, chemotherapeutic agents, and cancer vaccines are the three most common classes of agents identified in trials. Agents in clinical trials demonstrated uneven distribution in combination approaches; chemotherapy agents, proteasome inhibitors, and immune modulators frequently appeared in combinations, whereas kinase inhibitors, modified immune effector cells did not as was shown by combination networks and descriptive statistics. This analysis provides an extensive overview of the drug discovery field in brain cancer, shifts that have been happening in recent years, and challenges that are likely to come. SIGNIFICANCE STATEMENT: This review provides comprehensive quantitative analysis and discussion of the brain cancer drug discovery field, including classification of drug, targets, and therapies.
Collapse
Affiliation(s)
- Aleksandr V Sokolov
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Samira A Dostdar
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Misty M Attwood
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aleksandra A Krasilnikova
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anastasia A Ilina
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Amina Sh Nabieva
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anna A Lisitsyna
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir N Chubarev
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vadim V Tarasov
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden (A.V.S., S.A.D., M.M.A., H.B.S.); and Department of Pharmacology, Institute of Pharmacy (A.V.S., S.A.D., A.A.K., A.A.I., A.S.N., A.A.L., V.N.C., V.V.T.) and Institute of Translational Medicine and Biotechnology (V.V.T., H.B.S.), I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
13
|
Rat Adipose-Derived Stromal Cells (ADSCs) Increases the Glioblastoma Growth and Decreases the Animal Survival. Stem Cell Rev Rep 2021; 18:1495-1509. [PMID: 34403074 DOI: 10.1007/s12015-021-10227-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2021] [Indexed: 12/22/2022]
Abstract
Many studies have shown that mesenchymal stromal cells (MSCs) and their secreted factors may modulate the biology of tumor cells. However, how these interactions happen in vivo remains unclear. In the present study, we investigated the effects of rat adipose-derived stromal cells (ADSCs) and their conditioned medium (ADSC-CM) in glioma tumor growth and malignancy in vivo. Our results showed that when we co-injected C6 cells plus ADSCs into the rat brains, the tumors generated were larger and the animals exhibited shorter survival, when compared with tumors of the animals that received only C6 cells or C6 cells pre-treated with ADSC-CM. We further showed that the animals that received C6 plus ADSC did not present enhanced expression of CD73 (a gene highly expressed in ADSCs), indicating that the tumor volume observed in these animals was not a mere consequence of the higher density of cells administered in this group. Finally, we showed that the animals that received C6 + ADSC presented tumors with larger necrosis areas and greater infiltration of immune cells. These results indicate that the immunoregulatory properties of ADSCs and its contribution to tumor stroma can support tumor growth leading to larger zones of necrosis, recruitment of immune cells, thus facilitating tumor progression. Our data provide new insights into the way by which ADSCs and tumor cells interact and highlight the importance of understanding the fate and roles of MSCs in tumor sites in vivo, as well as their intricate crosstalk with cancer cells.
Collapse
|
14
|
Glioblastoma Therapy: Rationale for a Mesenchymal Stem Cell-based Vehicle to Carry Recombinant Viruses. Stem Cell Rev Rep 2021; 18:523-543. [PMID: 34319509 DOI: 10.1007/s12015-021-10207-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2021] [Indexed: 12/12/2022]
Abstract
Evasion of growth suppression is among the prominent hallmarks of cancer. Phosphatase and tensin homolog (PTEN) and p53 tumor-suppressive pathways are compromised in most human cancers, including glioblastoma (GB). Hence, these signaling pathways are an ideal point of focus for novel cancer therapeutics. Recombinant viruses can selectivity kill cancer cells and carry therapeutic genes to tumors. Specifically, oncolytic viruses (OV) have been successfully employed for gene delivery in GB animal models and showed potential to neutralize immunosuppression at the tumor site. However, the associated systemic immunogenicity, inefficient transduction of GB cells, and inadequate distribution to metastatic tumors have been the major bottlenecks in clinical studies. Mesenchymal stem cells (MSCs), with tumor-tropic properties and immune privilege, can improve OVs targeting. Remarkably, combining the two approaches can address their individual issues. Herein, we summarize findings to advocate the reactivation of tumor suppressors p53 and PTEN in GB treatment and use MSCs as a "Trojan horse" to carry oncolytic viral cargo to disseminated tumor beds. The integration of MSCs and OVs can emerge as the new paradigm in cancer treatment.
Collapse
|
15
|
Hypoxia Engineered Bone Marrow Mesenchymal Stem Cells Targeting System with Tumor Microenvironment Regulation for Enhanced Chemotherapy of Breast Cancer. Biomedicines 2021; 9:biomedicines9050575. [PMID: 34069607 PMCID: PMC8160638 DOI: 10.3390/biomedicines9050575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/02/2021] [Accepted: 05/16/2021] [Indexed: 12/12/2022] Open
Abstract
Improving the tumor targeting of docetaxel (DTX) would not only be favored for the chemotherapeutic efficacy, but also reduce its side effects. However, the regulation of the tumor microenvironment could further inhibit the growth of tumors. In this study, we introduced a system consisting of hypoxia-engineered bone marrow mesenchymal stem cells (H-bMSCs) and DTX micelles (DTX-M) for breast cancer treatment. First, the stem cell chemotherapy complex system (DTX@H-bMSCs) with tumor-targeting ability was constructed according to the uptake of DTX-M by hypoxia-induced bMSCs (H-bMSCs). DTX micellization improved the uptake efficiency of DTX by H-bMSCs, which equipped DTX@H-bMSCs with satisfactory drug loading and stability. Furthermore, the migration of DTX@H-bMSCs revealed that it could effectively target the tumor site and facilitate the drug transport between cells. Moreover, in vitro and in vivo pharmacodynamics of DTX@H-bMSCs exhibited a superior antitumor effect, which could promote the apoptosis of 4T1 cells and upregulate the expression of inflammatory factors at the tumor site. In brief, DTX@H-bMSCs enhanced the chemotherapeutic effect in breast cancer treatment.
Collapse
|
16
|
Iturrioz-Rodríguez N, Bertorelli R, Ciofani G. Lipid-Based Nanocarriers for The Treatment of Glioblastoma. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000054. [PMID: 33623931 PMCID: PMC7116796 DOI: 10.1002/anbr.202000054] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant neoplasia having origin in the brain. The current treatments involve surgery, radiotherapy, and chemotherapy, being complete surgical resection the best option for the patient survival chances. However, in those cases where a complete removal is not possible, radiation and chemotherapy are applied. Herein, the main challenges of chemotherapy, and how they can be overcome with the help of nanomedicine, are approached. Natural pathways to cross the blood-brain barrier (BBB) are detailed, and different in vivo studies where these pathways are mimicked functionalizing the nanomaterial surface are shown. Later, lipid-based nanocarriers, such as liposomes, solid lipid nanoparticles, and nanostructured lipid carriers, are presented. To finish, recent studies that have used lipid-based nanosystems carrying not only therapeutic agents, yet also magnetic nanoparticles, are described. Although the advantages of using these types of nanosystems are explained, including their biocompatibility, the possibility of modifying their surface to enhance the cell targeting, and their intrinsic ability of BBB crossing, it is important to mention that research in this field is still at its early stage, and extensive preclinical and clinical investigations are mandatory in the close future.
Collapse
Affiliation(s)
- Nerea Iturrioz-Rodríguez
- Smart Bio-Interfaces Istituto Italiano di Tecnologia Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| | - Rosalia Bertorelli
- Translational Pharmacology Istituto Italiano di Tecnologia Via Morego 30, Genova 16163, Italy
| | - Gianni Ciofani
- Smart Bio-Interfaces Istituto Italiano di Tecnologia Viale Rinaldo Piaggio 34, Pontedera 56025, Italy
| |
Collapse
|
17
|
Hosseindoost S, Hashemizadeh S, Gharaylou Z, Dehpour AR, Javadi SAH, Arjmand B, Hadjighassem M. β2-Adrenergic Receptor Stimulation Upregulates Cx43 Expression on Glioblastoma Multiforme and Olfactory Ensheathing Cells. J Mol Neurosci 2020; 70:1451-1460. [PMID: 32506304 DOI: 10.1007/s12031-020-01542-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022]
Abstract
Glioblastoma multiforme (GBM) is described as an invasive astrocytic tumor in adults. Despite current standard treatment approaches, the outcome of GBM remains unfavorable. The downregulation of connexin 43 (Cx43) expression is one of the molecular transformations in GBM cells. The Cx43 levels and subsequently gap junctional intercellular communication (GJIC) have an important role in the efficient transfer of cytotoxic drugs to whole tumor cells. As shown in our previous study, the stimulation of the β2-adrenergic receptor (β2-AR) leads to the modulation of Cx43 expression level in the GBM cell line. Here we further examine the effect of clenbuterol hydrochloride as a selective β2-AR agonist on the Cx43 expression in human GBM-derived astrocyte cells and human olfactory ensheathing cells (OECs) as a potent vector for future gene therapy. In this experiment, first we established a primary culture of astrocytes from GBM samples and verified the purity using immunocytofluorescent staining. Western blot analysis was performed to evaluate the Cx43 protein level. Our western blot findings reveal that clenbuterol hydrochloride upregulates the Cx43 protein level in both primary human astrocyte cells and human OECs. Conversely, ICI 118551 as a β2-AR antagonist inhibits these effects. Moreover, clenbuterol hydrochloride increases the Cx43 expression in primary human astrocyte cells and OECs co-culture systems, and ICI 118551 reverses these effects. To confirm the western blot results, immunocytofluorescent staining was performed to evaluate the β2-AR agonist effect on Cx43 expression. Our immunocytofluorescent results supported western blot analysis in primary human astrocyte cells and the OECs co-culture system. The results of this study suggest that the activation of β2-AR with regard to Cx43 protein levels enhancement in GBM cells and OECs might be a promising approach for GBM treatment in the future.
Collapse
Affiliation(s)
- Saereh Hosseindoost
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Hashemizadeh
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Gharaylou
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Hossein Javadi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Neurosurgery department, Imam Khomeini hospital complex, TUMS, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Srinivasan VM, Gumin J, Camstra KM, Chen SR, Johnson JN, Shimizu Y, Parker Kerrigan BC, Shpall EJ, Lang FF, Kan P. Microcatheter delivery of neurotherapeutics: compatibility with mesenchymal stem cells. J Neurosurg 2020; 133:1182-1190. [PMID: 31491769 DOI: 10.3171/2019.6.jns19327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/11/2019] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Bone marrow-derived human mesenchymal stem cells (BM-hMSCs) have been used in clinical trials for the treatment of several neurological disorders. MSCs have been explored as a delivery modality for targeted viral therapeutic agents in the treatment of intracranial pathologies. Delta-24-RGD, a tumor-selective oncolytic adenovirus designed to target malignant glioma cells, has been shown to be effective in animal models and in a recent clinical trial. However, the most efficient strategy for delivering oncolytic therapies remains unclear. BM-hMSCs have been shown to home toward glioma xenografts after intracarotid delivery. The feasibility of selective intraarterial infusion of BM-hMSCs loaded with Delta-24-RGD (BM-hMSC-Delta-24) to deliver the virus to the tumor is being investigated. To evaluate the feasibility of endovascular intraarterial delivery, the authors tested in vitro the compatibility of BM-hMSC-Delta-24 with a variety of commercially available, clinically common microcatheters. METHODS BM-hMSCs were cultured, transfected with Delta-24-RGD, and resuspended in 1% human serum albumin. The solution was then injected via 4 common neuroendovascular microcatheters of different inner diameters (Marathon, Echelon-14, Marksman, and SL-10). Cell count and viability after injection through the microcatheters were assessed, including tests of injection velocity and catheter configuration. Transwell assays were performed with the injected cells to test the efficacy of BM-hMSC-Delta-24 activity against U87 glioma cells. BM-hMSC-Delta-24 compatibility was also tested with common neuroendovascular medications: Omnipaque, verapamil, and heparin. RESULTS The preinfusion BM-hMSC-Delta-24 cell count was 1.2 × 105 cells/ml, with 98.7% viability. There was no significant difference in postinfusion cell count or viability for any of the catheters. Increasing the injection velocity from 1.0 ml/min to 73.2 ml/min, or modifying the catheter shape from straight to tortuous, did not significantly reduce cell count or viability. Cell count and viability remained stable for up to 5 hours when the cell solution was stored on ice. Mixing BM-hMSC-Delta-24 with clinical concentrations of Omnipaque, verapamil, and heparin prior to infusion did not alter cell count or viability. Transwell experiments demonstrated that the antiglioma activity of BM-hMSC-Delta-24 was maintained after infusion. CONCLUSIONS BM-hMSC-Delta-24 is compatible with a wide variety of microcatheters and medications commonly used in neuroendovascular therapy. Stem cell viability and viral agent activity do not appear to be affected by catheter configuration or injection velocity. Commercially available microcatheters can be used to deliver stem cell neurotherapeutics via intraarterial routes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Elizabeth J Shpall
- 4Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | |
Collapse
|
19
|
Identification of Glioma Cancer Stem Cell Characteristics Based on Weighted Gene Prognosis Module Co-Expression Network Analysis of Transcriptome Data Stemness Indices. J Mol Neurosci 2020; 70:1512-1520. [PMID: 32451841 DOI: 10.1007/s12031-020-01590-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Glioma is the most common primary brain tumor in humans and the most deadly. Stem cells, which are characterized by therapeutic resistance and self-renewal, play a critical role in glioma, and therefore the identification of stem cell-related genes in glioma is important. In this study, we collected and evaluated the epigenetically regulated-mRNA expression-based stemness index (EREG-mRNAsi) of The Cancer Genome Atlas (TCGA, http://www.ncbi.nlm.nih.gov/ ) for glioma patient samples, corrected through tumor purity. After EREG-mRNAsi correction, glioma pathological grade and survival were analyzed. The differentially expressed gene (DEG) co-expression network was constructed by weighted gene co-expression network analysis (WGCNA) in TCGA glioma samples to find modules of interest and key genes. Gene ontology (GO) and pathway-enrichment analysis were performed to identify the function of significant genetic modules. Protein-protein interaction (PPI) and co-expression network analysis of key genes was performed for further analysis. In this experiment, we found that corrected EREG-mRNAsi was significantly up-regulated in glioma samples and increased with glioma grade, with G4 having the highest stemness index. Patients with higher corrected EREG-mRNAsi scores had worse overall survival. Fifty-one DEGs in the brown gene module were found to be positively related to EREG-mRNAsi via WGCNA. GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that chromosome segregation and cell cycle molecular function were the major functions in key DEGs. Among these key DEGs, BUB1 showed high connectivity and co-expression, and also high connectivity in PPI. Fifty-one key genes were verified to play a critical role in glioma stem cells. These genes may serve as primary therapeutic targets to inhibit the activity of glioma stem cells.
Collapse
|
20
|
Partridge B, Rossmeisl JH. Companion animal models of neurological disease. J Neurosci Methods 2020; 331:108484. [PMID: 31733285 PMCID: PMC6942211 DOI: 10.1016/j.jneumeth.2019.108484] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023]
Abstract
Clinical translation of novel therapeutics that improve the survival and quality of life of patients with neurological disease remains a challenge, with many investigational drug and device candidates failing in advanced stage clinical trials. Naturally occurring inherited and acquired neurological diseases, such as epilepsy, inborn errors of metabolism, brain tumors, spinal cord injury, and stroke occur frequently in companion animals, and many of these share epidemiologic, pathophysiologic and clinical features with their human counterparts. As companion animals have a relatively abbreviated lifespan and genetic background, are immunocompetent, share their environment with human caregivers, and can be clinically managed using techniques and tools similar to those used in humans, they have tremendous potential for increasing the predictive value of preclinical drug and device studies. Here, we review comparative features of spontaneous neurological diseases in companion animals with an emphasis on neuroimaging methods and features, illustrate their historical use in translational studies, and discuss inherent limitations associated with each disease model. Integration of companion animals with naturally occurring disease into preclinical studies can complement and expand the knowledge gained from studies in other animal models, accelerate or improve the manner in which research is translated to the human clinic, and ultimately generate discoveries that will benefit the health of humans and animals.
Collapse
Affiliation(s)
- Brittanie Partridge
- Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA; Brain Tumor Center of Excellence, Wake Forest University Comprehensive Cancer Center, Medical Center Blvd, NRC 405, Winston Salem, NC, 27157, USA
| | - John H Rossmeisl
- Veterinary and Comparative Neuro-Oncology Laboratory, Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA; Brain Tumor Center of Excellence, Wake Forest University Comprehensive Cancer Center, Medical Center Blvd, NRC 405, Winston Salem, NC, 27157, USA.
| |
Collapse
|
21
|
Dong H, Xu X, Wang L, Mo R. Advances in living cell-based anticancer therapeutics. Biomater Sci 2020; 8:2344-2365. [DOI: 10.1039/d0bm00036a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review summarizes recent advances in the applications of living cells as drug carriers or active drugs for anticancer drug delivery and cancer therapy.
Collapse
Affiliation(s)
- He Dong
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Xiao Xu
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Leikun Wang
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| | - Ran Mo
- State Key Laboratory of Natural Medicines
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
| |
Collapse
|
22
|
Qi SG, Quan LQ, Cui XY, Li HM, Zhao XD, Li RT. A natural compound obtained from Valeriana jatamansi selectively inhibits glioma stem cells. Oncol Lett 2019; 19:1384-1392. [PMID: 32002029 PMCID: PMC6960388 DOI: 10.3892/ol.2019.11239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 10/22/2019] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma is one of the most malignant tumors with very poor prognosis. Glioma stem cells (GSCs) occupy a small proportion in glioma, but they are closely associated with radiotherapy and chemotherapy resistance, promoting tumor angiogenesis, hypoxia response, invasion and recurrence. Therefore, GSCs have become a new target for tumor treatment and are used in drug screening. Rupesin E is a natural compound obtained from Valeriana jatamansi, and its antitumor activity has not been reported. In the present study, the antitumor activity of rupesin E was investigated, and the results demonstrated that it inhibited the proliferation of GSCs (GSC-3#, GSC-12#, GSC-18#) with the IC50 values of 7.13±1.41, 13.51±1.46 and 4.44±0.22 µg/ml, respectively. In addition, immunofluorescence cell staining and flow cytometry techniques demonstrated that rupesin E inhibited GSC proliferation and induced apoptosis. Furthermore, rupesin E inhibited the ability of GSC colony formation, indicating its antitumor activity against GSCs in vitro.
Collapse
Affiliation(s)
- Shi-Gang Qi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China
| | - Li-Qiu Quan
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Xiao-Yue Cui
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China
| | - Hong-Mei Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Xu-Dong Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
23
|
Nasal Drug Delivery of Anticancer Drugs for the Treatment of Glioblastoma: Preclinical and Clinical Trials. Molecules 2019; 24:molecules24234312. [PMID: 31779126 PMCID: PMC6930669 DOI: 10.3390/molecules24234312] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/22/2019] [Accepted: 11/24/2019] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal form of brain tumor, being characterized by the rapid growth and invasion of the surrounding tissue. The current standard treatment for glioblastoma is surgery, followed by radiotherapy and concurrent chemotherapy, typically with temozolomide. Although extensive research has been carried out over the past years to develop a more effective therapeutic strategy for the treatment of GBM, efforts have not provided major improvements in terms of the overall survival of patients. Consequently, new therapeutic approaches are urgently needed. Overcoming the blood–brain barrier (BBB) is a major challenge in the development of therapies for central nervous system (CNS) disorders. In this context, the intranasal route of drug administration has been proposed as a non-invasive alternative route for directly targeting the CNS. This route of drug administration bypasses the BBB and reduces the systemic side effects. Recently, several formulations have been developed for further enhancing nose-to-brain transport, mainly with the use of nano-sized and nanostructured drug delivery systems. The focus of this review is to provide an overview of the strategies that have been developed for delivering anticancer compounds for the treatment of GBM while using nasal administration. In particular, the specific properties of nanomedicines proposed for nose-to-brain delivery will be critically evaluated. The preclinical and clinical data considered supporting the idea that nasal delivery of anticancer drugs may represent a breakthrough advancement in the fight against GBM.
Collapse
|
24
|
Lu Y, Sun W, Zhang L, Li J. Silencing Of MAGI1 Promotes The Proliferation And Inhibits Apoptosis Of Glioma Cells Via The Wnt/β-Catenin And PTEN/AKT Signaling Pathways. Onco Targets Ther 2019; 12:9639-9650. [PMID: 32009799 PMCID: PMC6859429 DOI: 10.2147/ott.s215400] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 10/15/2019] [Indexed: 12/27/2022] Open
Abstract
Background Membrane-associated guanylate kinase (MAGUK) with inverted orientation protein 1 (MAGI1) is a novel member of the MAGUK family with a vital role in tumor progression related to invasion and metastasis. However, the function of MAGI1 in glioma is currently unknown. We therefore analyzed the expression of MAGI1 protein in human glioma samples, glioma cell lines and glioma stem cells (GSCs), and explored its effects on glioma cell proliferation and apoptosis. Methods MAGI1 expression in glioma tissues was examined by Western blotting and real-time polymerase chain reaction and its relationships with clinical pathological features were analyzed. The effects of MAGI1 knockdown on the proliferation of glioma cell lines and GSCs were detected by CCK8 and colony-formation assays, and apoptosis was assessed by flow cytometry. We also investigated the effects of MAGI1 silencing on protein expression levels of epithelial-mesenchymal transition biomarkers, as well as β-catenin, cyclin D1, PTEN and phospho-Akt by Western blotting. Results MAGI1 was significantly downregulated in glioma tissues and its expression was related to cancer progression. Silencing of MAGI1 in both glioma cell lines and GSCs enhanced proliferation and inhibited apoptosis. MAGI1 knockdown also significantly increased the expression levels of N-cadherin, vimentin, β-catenin, cyclin D1 and phospho-Akt and reduced the expression of E-cadherin and PTEN. Conclusions Our results indicated that MAGI1 might play a vital role in glioma progression and may represent a potential therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Yongzhi Lu
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, Shandong Province 266000, People's Republic of China
| | - Wei Sun
- Department of Neurology, Qingdao Third People's Hospital, Qingdao, Shandong Province 266000, People's Republic of China
| | - Liang Zhang
- Department of Critical Care Medicine, Qingdao Eighth People's Hospital, Qingdao, Shandong Province 266000, People's Republic of China
| | - Junyao Li
- Department of Emergency, Qingdao Municipal Hospital, Qingdao, Shandong Province 266000, People's Republic of China
| |
Collapse
|
25
|
Miyata S, Tominaga K, Sakashita E, Urabe M, Onuki Y, Gomi A, Yamaguchi T, Mieno M, Mizukami H, Kume A, Ozawa K, Watanabe E, Kawai K, Endo H. Comprehensive Metabolomic Analysis of IDH1 R132H Clinical Glioma Samples Reveals Suppression of β-oxidation Due to Carnitine Deficiency. Sci Rep 2019; 9:9787. [PMID: 31278288 PMCID: PMC6611790 DOI: 10.1038/s41598-019-46217-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 06/25/2019] [Indexed: 11/13/2022] Open
Abstract
Gliomas with Isocitrate dehydrogenase 1 (IDH1) mutation have alterations in several enzyme activities, resulting in various metabolic changes. The aim of this study was to determine a mechanism for the better prognosis of gliomas with IDH mutation by performing metabolomic analysis. To understand the metabolic state of human gliomas, we analyzed clinical samples obtained from surgical resection of glioma patients (grades II–IV) with or without the IDH1 mutation, and compared the results with U87 glioblastoma cells overexpressing IDH1 or IDH1R132H. In clinical samples of gliomas with IDH1 mutation, levels of D-2-hydroxyglutarate (D-2HG) were increased significantly compared with gliomas without IDH mutation. Gliomas with IDH mutation also showed decreased intermediates in the tricarboxylic acid cycle and pathways involved in the production of energy, amino acids, and nucleic acids. The marked difference in the metabolic profile in IDH mutant clinical glioma samples compared with that of mutant IDH expressing cells includes a decrease in β-oxidation due to acyl-carnitine and carnitine deficiencies. These metabolic changes may explain the lower cell division rate observed in IDH mutant gliomas and may provide a better prognosis in IDH mutant gliomas.
Collapse
Affiliation(s)
- Satsuki Miyata
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan.
| | - Kaoru Tominaga
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan.
| | - Eiji Sakashita
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan
| | - Masashi Urabe
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Yoshiyuki Onuki
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Akira Gomi
- Department of Pediatric Neurosurgery, Jichi Children's Medical Center, Jichi Medical University, Tochigi, Japan
| | - Takashi Yamaguchi
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Makiko Mieno
- Department of Medical Informatics, Center for Information, Jichi Medical University, Tochigi, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Akihiro Kume
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Keiya Ozawa
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Eiju Watanabe
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Kensuke Kawai
- Department of Neurosurgery, Jichi Medical University, Tochigi, Japan
| | - Hitoshi Endo
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
26
|
Abdelrahman AE, Ibrahim HM, Elsebai EA, Ismail EI, Elmesallamy W. The clinicopathological significance of CD133 and Sox2 in astrocytic glioma. Cancer Biomark 2019; 23:391-403. [PMID: 30248046 DOI: 10.3233/cbm-181460] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The treatment strategies of astrocytoma have not changed considerably due to the restricted appreciation of its biology. OBJECTIVES This study aimed to evaluate the expression of the stem cell-related proteins (CD133 and Sox2) and their prognostic value in astrocytic glioma. METHODS The immunohistochemical expression of CD133 and Sox2 in 40 patients with an astrocytic glioma of different grades was studied. The recorded data on the overall survival (OS), progression-free survival (PFS) and the response to the therapeutic protocol were collected and lastly analyzed. RESULTS CD133 expression was observed in 87.5% of the cases, while positive Sox2 expression was found in all the studied cases. There was a significant association of CD133 expression with the histological grade and the tumor size (p< 0.001). A significant association of Sox2 with the histological grade and the tumor size was noted (p= 0.004, p= 0.006 respectively). Up-regulation of both CD133 and Sox2 had a significant association with poor clinical response to the therapy (p< 0.001 for each). Shorter OS and PFS were related to CD133 and Sox2 overexpression. CONCLUSIONS Astrocytoma with CD133 and Sox2 overexpression had an unfavorable prognosis and poor clinical response to the current therapeutic protocol.
Collapse
Affiliation(s)
- Aziza E Abdelrahman
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hanaa M Ibrahim
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman A Elsebai
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman I Ismail
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Wael Elmesallamy
- Neurosurgery Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
27
|
Wildes TJ, Flores CT, Mitchell DA. Concise Review: Modulating Cancer Immunity with Hematopoietic Stem and Progenitor Cells. Stem Cells 2019; 37:166-175. [PMID: 30353618 PMCID: PMC6368859 DOI: 10.1002/stem.2933] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are the progenitor cells that can regenerate the entire blood compartment, including the immune system. Recent studies have unearthed considerable immune-modulating potential of these cells. They can migrate through chemotactic gradients, differentiate into functional immune cells, and crosstalk with immune cells during infections, autoimmune diseases, and cancers. Although the primary role of HSPCs during solid malignancies is considered immunosuppressive, recent studies have discovered immune-activating HSPCs and progeny. In this review, we will discuss the recent evidence that HSPCs act as immunomodulators during solid cancers and highlight the future directions of discovery. Stem Cells 2019;37:166-175.
Collapse
Affiliation(s)
- Tyler J. Wildes
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of NeurosurgeryMcKnight Brain Institute, University of FloridaGainesvilleFloridaUSA
| | - Catherine T. Flores
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of NeurosurgeryMcKnight Brain Institute, University of FloridaGainesvilleFloridaUSA
| | - Duane A. Mitchell
- University of Florida Brain Tumor Immunotherapy Program, Preston A. Wells, Jr. Center for Brain Tumor Therapy, Lillian S. Wells Department of NeurosurgeryMcKnight Brain Institute, University of FloridaGainesvilleFloridaUSA
| |
Collapse
|
28
|
Kurogi R, Nakamizo A, Suzuki SO, Mizoguchi M, Yoshimoto K, Amano T, Amemiya T, Takagishi S, Iihara K. Inhibition of glioblastoma cell invasion by hsa-miR-145-5p and hsa-miR-31-5p co-overexpression in human mesenchymal stem cells. J Neurosurg 2019; 130:44-55. [PMID: 29521593 DOI: 10.3171/2017.8.jns1788] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 08/28/2017] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Human bone marrow–derived mesenchymal stem cells (hMSCs) show tropism for brain tumors and may be a useful vehicle for drug or gene delivery to malignant gliomas. Recently, some microRNAs (miRNAs) have been shown to suppress the invasiveness of malignant gliomas. METHODS To test their potential to become vehicles for the delivery of miRNA to malignant gliomas, hMSCs were engineered so that hMSC secretion of miRNAs that inhibit glioma cell invasion was enabled without altering the hMSC tropism for glioma cells. RESULTS In coculture, hMSCs cotransfected with hsa-miR-145-5p and -31-5p miRNAs showed markedly reduced invasion by U87 glioma cells in a contact-dependent manner both in vitro and ex vivo, with invasion of hMSCs cotransfected with these 2 miRNAs by the U87 cells reduced to 60.7% compared with control cells. According to a Matrigel invasion assay, the tropism of the hMSCs for U87 cells was not affected. In glioma cell lines U251 and LN229, hMSCs exhibited tropism in vivo, and invasion of hMSCs cotransfected with hsa-miR-145-5p and -31-5p was also significantly less than that of control cells. When U87 cells were coimplanted into the striatum of organotypic rat brain slices with hMSCs cotransfected with hsa-miR-145 and -31-5p, the relative invasive area decreased by 37.1%; interestingly, these U87 cells showed a change to a rounded morphology that was apparent at the invasion front. Whole-genome microarray analysis of the expression levels of 58,341 genes revealed that the co-overexpression of hsa-miR-145-5p and -31-5p downregulated FSCN1 expression in U87 cells. CONCLUSIONS This study demonstrates that miRNA overexpression in hMSCs can alter the function of glioma cells via contact-dependent transfer. Co-overexpression of multiple miRNAs may be a useful and novel therapeutic strategy. The study results suggest that hMSCs can be applied as a delivery vehicle for miRNAs.
Collapse
Affiliation(s)
- Ryota Kurogi
- Departments of1Neurosurgery and
- 2Department of Neurosurgery, National Hospital Organization, Clinical Research Institute, Kyushu Medical Center, Fukuoka, Japan
| | - Akira Nakamizo
- Departments of1Neurosurgery and
- 2Department of Neurosurgery, National Hospital Organization, Clinical Research Institute, Kyushu Medical Center, Fukuoka, Japan
| | - Satoshi O Suzuki
- 3Neuropathology, Graduate School of Medical Sciences, Kyushu University; and
| | | | | | | | | | | | | |
Collapse
|
29
|
Mesenchymal stem cell-driven activatable photosensitizers for precision photodynamic oncotherapy. Biomaterials 2018; 187:18-26. [DOI: 10.1016/j.biomaterials.2018.09.041] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/17/2018] [Accepted: 09/27/2018] [Indexed: 12/27/2022]
|
30
|
Shi S, Zhang M, Guo R, Miao Y, Li B. Bone Marrow-Derived Mesenchymal Stem Cell-Mediated Dual-Gene Therapy for Glioblastoma. Hum Gene Ther 2018; 30:106-117. [PMID: 29993289 DOI: 10.1089/hum.2018.092] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bone-marrow mesenchymal stem cells (BMSCs) have been used for systemic delivery of therapeutic genes to solid tumors. However, the optimal treatment time post-BMSC implantation and the assessment of the long-term fate of therapeutic BMSCs post-tumor treatment are critical if such promising therapies are to be translated into clinical practice. An efficient BMSC-based therapeutic strategy has been developed that simultaneously allows killing of tumor cells, inhibiting of tumor angiogenesis, and assessment and eradication of implanted BMSCs after treatment of glioblastoma. BMSCs were engineered to co-express the angiogenesis inhibitor kringle 5 (K5) of human plasminogen, under the control of the cytomegalovirus promoter (CMV) and the human sodium-iodide symporter (NIS), involved in uptake of radioisotopes, under the control of early growth response factor 1 (Egr1), a radiation-activated promoter. A significant decrease in tumor growth and tumor angiogenesis and a subsequent increase in survival were observed when mice bearing glioblastoma were treated with 188Re post-therapeutic intravenous BMSC implantation. Furthermore, the systemic administration of 188Re post-tumor treatment selectively eliminated therapeutic BMSCs expressing NIS, which was monitored in real time by 125I micro single photon emission computed tomography/computed tomography imaging. Meanwhile, the Egr1 promoter induced a 188Re radiation positive feedback effect absorbed by NIS. After intravenous BMSC implantation, BMSCs levels in the tumor and lung both peaked on day 10 and decreased to the lowest levels on days 24 and 17, respectively. These findings suggest that day 17 post-BMSC implantation could be an optimal time for 188Re treatment. These results provide a new adjuvant therapy mediated by BMSCs for glioblastoma treatment.
Collapse
Affiliation(s)
- Shuo Shi
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ying Miao
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
31
|
Guerra-Rebollo M, Nogueira de Moraes C, Alcoholado C, Soler-Botija C, Sanchez-Cid L, Vila OF, Meca-Cortés O, Ramos-Romero S, Rubio N, Becerra J, Blanco J, Garrido C. Glioblastoma Bystander Cell Therapy: Improvements in Treatment and Insights into the Therapy Mechanisms. MOLECULAR THERAPY-ONCOLYTICS 2018; 11:39-51. [PMID: 30364660 PMCID: PMC6197388 DOI: 10.1016/j.omto.2018.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/12/2018] [Indexed: 01/14/2023]
Abstract
A preclinical model of glioblastoma (GB) bystander cell therapy using human adipose mesenchymal stromal cells (hAMSCs) is used to address the issues of cell availability, quality, and feasibility of tumor cure. We show that a fast proliferating variety of hAMSCs expressing thymidine kinase (TK) has therapeutic capacity equivalent to that of TK-expressing hAMSCs and can be used in a multiple-inoculation procedure to reduce GB tumors to a chronically inhibited state. We also show that up to 25% of unmodified hAMSCs can be tolerated in the therapeutic procedure without reducing efficacy. Moreover, mimicking a clinical situation, tumor debulking previous to cell therapy inhibits GB tumor growth. To understand these striking results at a cellular level, we used a bioluminescence imaging strategy and showed that tumor-implanted therapeutic cells do not proliferate, are unaffected by GCV, and spontaneously decrease to a stable level. Moreover, using the CLARITY procedure for tridimensional visualization of fluorescent cells in transparent brains, we find therapeutic cells forming vascular-like structures that often associate with tumor cells. In vitro experiments show that therapeutic cells exposed to GCV produce cytotoxic extracellular vesicles and suggest that a similar mechanism may be responsible for the in vivo therapeutic effectiveness of TK-expressing hAMSCs.
Collapse
Affiliation(s)
- Marta Guerra-Rebollo
- Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), 08034 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
| | - Carolina Nogueira de Moraes
- Department of Animal Reproduction and Veterinary Radiology, College of Veterinary Medicine and Animal Science, São Paulo State University, UNESP, 18618-681 Botucatu, Brazil
| | - Cristina Alcoholado
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedical Research Institute of Málaga (IBIMA), 29071 Málaga, Spain
| | - Carolina Soler-Botija
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol, 08916 Badalona, Spain
- CIBER Cardiovascular, Carlos III Health Institute, 28029 Madrid, Spain
| | - Lourdes Sanchez-Cid
- Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), 08034 Barcelona, Spain
| | - Olaia F. Vila
- Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), 08034 Barcelona, Spain
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Oscar Meca-Cortés
- Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), 08034 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
| | - Sara Ramos-Romero
- Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), 08034 Barcelona, Spain
- Department of Cell Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - Nuria Rubio
- Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), 08034 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
| | - José Becerra
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, Biomedical Research Institute of Málaga (IBIMA), 29071 Málaga, Spain
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Andalusian Center for Nanomedicine and Biotechnology-BIONAND, 29590 Málaga, Spain
| | - Jeronimo Blanco
- Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), 08034 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
| | - Cristina Garrido
- Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), 08034 Barcelona, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain
- Corresponding author: Cristina Garrido, Cell Therapy Group, Catalonian Institute for Advanced Chemistry (IQAC-CSIC), Jordi Girona Street, 18-26, 08034 Barcelona, Spain.
| |
Collapse
|
32
|
Wang X, Gao J, Ouyang X, Wang J, Sun X, Lv Y. Mesenchymal stem cells loaded with paclitaxel-poly(lactic- co-glycolic acid) nanoparticles for glioma-targeting therapy. Int J Nanomedicine 2018; 13:5231-5248. [PMID: 30237710 PMCID: PMC6136913 DOI: 10.2147/ijn.s167142] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) possess inherent tropism towards tumor cells, and so have attracted increased attention as targeted-therapy vehicles for glioma treatment. Purpose The objective of this study was to demonstrate the injection of MSCs loaded with paclitaxel (Ptx)-encapsulated poly(d,l-lactide-co-glycolide) (PLGA) nanoparticles (NPs) for orthotopic glioma therapy in rats. Methods Ptx-PLGA NP-loaded MSC was obtained by incubating MSCs with Ptx-PLGA NPs. The drug transfer and cytotoxicity of Ptx-PLGA NP-loaded MSC against tumor cells were investigated in the transwell system. Biodistribution and antitumor activity was evaluated in the orthotopic glioma rats after contralateral injection. Results The optimal dose of MSC-loaded Ptx-PLGA NPs (1 pg/cell Ptx) had little effect on MSC-migration capacity, cell cycle, or multilineage-differentiation potential. Compared with Ptx-primed MSCs, Ptx-PLGA NP-primed MSCs had enhanced sustained Ptx release in the form of free Ptx and Ptx NPs. Ptx transfer from MSCs to glioma cells could induce tumor cell death in vitro. As for distribution in vivo, NP-loaded fluorescent MSCs were tracked throughout the tumor mass for 2 days after therapeutic injection. Survival was significantly longer after contralateral implantation of Ptx-PLGA NP-loaded MSCs than those injected with Ptx-primed MSCs or Ptx-PLGA NPs alone. Conclusion Based on timing and sufficient Ptx transfer from the MSCs to the tumor cells, Ptx-PLGA NP-loaded MSC is effective for glioma treatment. Incorporation of chemotherapeutic drug-loaded NPs into MSCs is a promising strategy for tumor-targeted therapy.
Collapse
Affiliation(s)
- Xiaoling Wang
- Department of Pharmacy, Zhejiang University City College, ;.,Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xumei Ouyang
- Department of Pharmacy, Zhejiang University City College, ;.,Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Junbo Wang
- Department of Pharmacy, Zhejiang University City College,
| | - Xiaoyi Sun
- Department of Pharmacy, Zhejiang University City College,
| | - Yuanyuan Lv
- Department of Pharmacy, Zhejiang University City College,
| |
Collapse
|
33
|
Liu XY, Zhou CB, Fang C. Nanomaterial-involved neural stem cell research: Disease treatment, cell labeling, and growth regulation. Biomed Pharmacother 2018; 107:583-597. [PMID: 30114642 DOI: 10.1016/j.biopha.2018.08.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) have been widely investigated for their potential in the treatment of various diseases and transplantation therapy. However, NSC growth regulation, labeling, and its application to disease diagnosis and treatment are outstanding challenges. Recently, nanomaterials have shown promise for various applications including genetic modification, imaging, and controlled drug release. Here we summarize the recent progress in the use of nanomaterials in combination with NSCs for disease treatment and diagnosis, cell labeling, and NSC growth regulation. The toxicity of nanomaterials to NSCs is also discussed.
Collapse
Affiliation(s)
- Xiang-Yu Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China
| | - Cheng-Bin Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Shanghai Key Laboratory of Psychotic Disorders, and Brain Science and Technology Research Center, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240 China
| | - Chao Fang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine (SJTU-SM), 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
34
|
Liang C, Yang Y, Guan J, Lv T, Qu S, Fu Q, Zhao H. LncRNA UCA1 sponges miR-204-5p to promote migration, invasion and epithelial-mesenchymal transition of glioma cells via upregulation of ZEB1. Pathol Res Pract 2018; 214:1474-1481. [PMID: 30107990 DOI: 10.1016/j.prp.2018.07.036] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/23/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022]
Abstract
Long non-coding RNA urothelial carcinoma associated 1 (lncRNA UCA1) promotes cancer progression and enhances chemoresistance through miR-204-5p in a few cancers. However, no studies have investigated whether UCA1 regulates glioma metastasis through miR-204-5p and its target. In the present study, cell migration, invasion and epithelial-mesenchymal transition (EMT) were evaluated in glioma cells overexpressing UCA1. The relationships among UCA1, miR-204-5p and ZEB1 were examined by real-time PCR, western blotting and dual-luciferase reporter assays. The effect of UCA1 knockdown on xenograft tumor growth was investigated. The levels of miR-204-5p, fibronectin, COL5 A1 and ZEB1 in tumor tissues were also determined. The results showed that UCA1 overexpression promoted cell migration, invasion and EMT. UCA1 interacted with miR-204-5p and decreased its level. ZEB1 was identified as a direct target of miR-204-5p and miR-204-5p negatively regulated ZEB1 expression. Moreover, UCA1 sponged miR-204-5p and partially rescued the inhibitory effect of miR-204-5p on ZEB1. In our in vivo studies, UCA1 knockdown reduced tumor volume and tumor weight. In addition, the levels of fibronectin, COL5 A1 and ZEB1 were decreased, while miR-204-5p level was increased. The present study provides the first evidence that UCA1 promotes glioma metastasis through the miR-204-5p/ZEB1 axis, contributing to the understanding of the pathogenesis of glioma.
Collapse
Affiliation(s)
- Chao Liang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China; Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Yang Yang
- Department of Neurosurgery, Jinzhou Central Hospital, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Junhong Guan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Tao Lv
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Shengtao Qu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Qiang Fu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China
| | - Hongyu Zhao
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, People's Republic of China.
| |
Collapse
|
35
|
Sheets KT, Bagó JR, Paulk IL, Hingtgen SD. Image-Guided Resection of Glioblastoma and Intracranial Implantation of Therapeutic Stem Cell-seeded Scaffolds. J Vis Exp 2018. [PMID: 30059037 DOI: 10.3791/57452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma (GBM), the most common and aggressive primary brain cancer, carries a life expectancy of 12-15 months. The short life expectancy is due in part to the inability of the current treatment, consisting of surgical resection followed by radiation and chemotherapy, to eliminate invasive tumor foci. Treatment of these foci may be improved with tumoricidal human mesenchymal stem cells (MSCs). MSCs exhibit potent tumor tropism and can be engineered to express therapeutic proteins that kill tumor cells. Advancements in preclinical models indicate that surgical resection induces premature MSC loss and reduces therapeutic efficacy. Efficacy of MSC treatment can be improved by seeding MSCs on a biodegradable poly(lactic acid) (PLA) scaffold. MSC delivery into the surgical resection cavity on a PLA scaffold restores cell retention, persistence, and tumor killing. To study the effects of MSC-seeded PLA implantation on GBM, an accurate preclinical model is needed. Here we provide a preclinical surgical protocol for image-guided tumor resection of GBM in immune-deficient mice followed by MSC-seeded scaffold implantation. MSCs are engineered with lentiviral constructs to constitutively express and secrete therapeutic TNFα-related apoptosis-inducing ligand (TRAIL) as well as green fluorescent protein (GFP) to allow fluorescent tracking. Similarly, the U87 tumor cells are engineered to express mCherry and firefly luciferase, providing dual fluorescent/luminescent tracking. While currently used for investigating stem cell mediated delivery of therapeutics, this protocol could be modified to investigate the impact of surgical resection on other GBM interventions.
Collapse
Affiliation(s)
- Kevin T Sheets
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Juli R Bagó
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Ivory L Paulk
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill;
| |
Collapse
|
36
|
Lv D, Yu SC, Ping YF, Wu H, Zhao X, Zhang H, Cui Y, Chen B, Zhang X, Dai J, Bian XW, Yao XH. A three-dimensional collagen scaffold cell culture system for screening anti-glioma therapeutics. Oncotarget 2018; 7:56904-56914. [PMID: 27486877 PMCID: PMC5302961 DOI: 10.18632/oncotarget.10885] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 06/30/2016] [Indexed: 12/17/2022] Open
Abstract
Three-dimensional (3D) culture, which can simulate in vivo microenvironments, has been increasingly used to study tumor cell biology. Since most preclinical anti-glioma drug tests still rely on conventional 2D cell culture, we established a collagen scaffold for 3D glioma cell culture. Glioma cells cultured on these 3D scaffolds showed greater degree of dedifferentiation and quiescence than cells in 2D culture. 3D-cultured cells also exhibited enhanced resistance to chemotherapeutic alkylating agents, with a much higher proportion of glioma stem cells and upregulation of O6-methylguanine DNA methyltransferase (MGMT). Importantly, tumor cells in 3D culture showed chemotherapy resistance patterns similar to those observed in glioma patients. Our results suggest that 3D collagen scaffolds are promising in vitro research platforms for screening new anti-glioma therapeutics.
Collapse
Affiliation(s)
- Donglai Lv
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Haibo Wu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xilong Zhao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Huarong Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Youhong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Bing Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, School of Military Preventive Medicine, Third Military Medical University, Chongqing, China.,Institute of Genetics and Development, Chinese Academy of Sciences, Beijing, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Jianwu Dai
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, School of Military Preventive Medicine, Third Military Medical University, Chongqing, China.,Institute of Genetics and Development, Chinese Academy of Sciences, Beijing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiao-Hong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| |
Collapse
|
37
|
Pang L, Zhang C, Qin J, Han L, Li R, Hong C, He H, Wang J. A novel strategy to achieve effective drug delivery: exploit cells as carrier combined with nanoparticles. Drug Deliv 2017; 24:83-91. [PMID: 28155538 PMCID: PMC8241159 DOI: 10.1080/10717544.2016.1230903] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/23/2016] [Accepted: 08/28/2016] [Indexed: 12/21/2022] Open
Abstract
Cell-mediated drug delivery systems employ specific cells as drug vehicles to deliver drugs to targeted sites. Therapeutics or imaging agents are loaded into these cells and then released in diseased sites. These specific cells mainly include red blood cells, leukocytes, stem cells and so on. The cell acts as a Trojan horse to transfer the drug from circulating blood to the diseased tissue. In such a system, these cells keep their original properties, which allow them to mimic the migration behavior of specific cells to carry drug to the targeted site after in vivo administration. This strategy elegantly combines the advantages of both carriers, i.e. the adjustability of nanoparticles (NPs) and the natural functions of active cells, which therefore provides a new perspective to challenge current obstacles in drug delivery. This review will describe a fundamental understanding of these cell-based drug delivery systems, and discuss the great potential of combinational application of cell carrier and NPs.
Collapse
Affiliation(s)
- Liang Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Chun Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Jing Qin
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Limei Han
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Ruixiang Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Chao Hong
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Jianxin Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China and
| |
Collapse
|
38
|
Zhang JB, Wang XQ, Lu GL, Huang HS, Xu SY. Adipose-derived mesenchymal stem cells therapy for acute kidney injury induced by ischemia-reperfusion in a rat model. Clin Exp Pharmacol Physiol 2017; 44:1232-1240. [PMID: 28688148 DOI: 10.1111/1440-1681.12811] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 06/10/2017] [Accepted: 06/14/2017] [Indexed: 12/28/2022]
Abstract
Acute kidney injury (AKI) represents a group of complicated syndromes with a high mortality rate. The administration of adipose-derived mesenchymal stem cells (ADMSCs) has been tested as a possible treatment method for AKI. The long-term evaluation of AKI induced by ischemia/reperfusion (IR) and the probable renal protection of ADMSCs are limited. In this study we have established a rat AKI model induced by IR and investigated the possible protective effects of ADMSCs. Adult Sprague-Dawley (SD) rats were divided into three groups (n = 6/each group). The MOCK group was as the normal control. Rats in the IR-AKI and IR-AKI+ADMSCs groups were subjected to IR injury by clamping both renal pedicles for 40 minutes. Rats in the MOCK and IR-AKI groups were injected with PBS via the tail vein as negative treatment controls. Rats in the IR-AKI+ADMSCs group received ADMSCs therapy (2 × 106 cells were injected into the rats via the tail vein). We found that ADMSC transplantation restored the pathologic morphology induced by IR-AKI to normal compared with the MOCK group, suggesting the reparative function of ADMSCs in kidney tissues. Compared with IR-induced AKI alone, ADMSC treatment significantly decreased the number of apoptotic cells, the level of total urinary protein and serum creatinine, the expression of pro-inflammatory cytokines (IL-6, TNF-α, IL-1β, IFN-γ, TNF-α, IFN-γ, and TGF-β), and the inflammation-associated proteins (HGF and SDF1), but increased the expression of the anti-inflammatory cytokine, IL-10, and the anti-apoptotic regulator, Bcl-2. Our data have indicated that ADMSC transplantation may protect against IR-induced AKI by anti-apoptotic and anti-inflammatory effects.
Collapse
Affiliation(s)
- Jian-Bo Zhang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Qiao Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guo-Lin Lu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Huan-Sen Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shi-Yuan Xu
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Clavreul A, Pourbaghi-Masouleh M, Roger E, Lautram N, Montero-Menei CN, Menei P. Human mesenchymal stromal cells as cellular drug-delivery vectors for glioblastoma therapy: a good deal? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:135. [PMID: 28962658 PMCID: PMC5622550 DOI: 10.1186/s13046-017-0605-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
Abstract
Background Glioblastoma (GB) is the most malignant brain tumor in adults. It is characterized by angiogenesis and a high proliferative and invasive capacity. Standard therapy (surgery, radiotherapy and chemotherapy with temozolomide) is of limited efficacy. Innovative anticancer drugs targeting both tumor cells and angiogenesis are urgently required, together with effective systems for their delivery to the brain. We assessed the ability of human mesenchymal stromal cells (MSCs) to uptake the multikinase inhibitor, sorafenib (SFN), and to carry this drug to a brain tumor following intranasal administration. Method MSCs were primed with SFN and drug content and release were quantified by analytical chemistry techniques. The ability of SFN-primed MSCs to inhibit the survival of the human U87MG GB cell line and endothelial cells was assessed in in vitro assays. These cells were then administered intranasally to nude mice bearing intracerebral U87MG xenografts. Their effect on tumor growth and angiogenesis was evaluated by magnetic resonance imaging and immunofluorescence analyses, and was compared with the intranasal administration of unprimed MSCs or SFN alone. Results MSCs took up about 9 pg SFN per cell, with no effect on viability, and were able to release 60% of the primed drug. The cytostatic activity of the released SFN was entirely conserved, resulting in a significant inhibition of U87MG and endothelial cell survival in vitro. Two intranasal administrations of SFN-primed MSCs in U87MG-bearing mice resulted in lower levels of tumor angiogenesis than the injection of unprimed MSCs or SFN alone, but had no effect on tumor volume. We also observed an increase in the proportion of small intratumoral vessels in animals treated with unprimed MSCs; this effect being abolished if the MSCs were primed with SFN. Conclusion We show the potential of MSCs to carry SFN to brain tumors following an intranasal administration. However, the therapeutic effect is modest probably due to the pro-tumorigenic properties of MSCs, which may limit the action of the released SFN. This calls into question the suitability of MSCs for use in GB therapy and renders it necessary to find methods guaranteeing the safety of this cellular vector after drug delivery.
Collapse
Affiliation(s)
- Anne Clavreul
- Département de Neurochirurgie, CHU, Angers, France. .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | - Milad Pourbaghi-Masouleh
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Emilie Roger
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | - Nolwenn Lautram
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | | | - Philippe Menei
- Département de Neurochirurgie, CHU, Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| |
Collapse
|
40
|
Razpotnik R, Novak N, Čurin Šerbec V, Rajcevic U. Targeting Malignant Brain Tumors with Antibodies. Front Immunol 2017; 8:1181. [PMID: 28993773 PMCID: PMC5622144 DOI: 10.3389/fimmu.2017.01181] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/06/2017] [Indexed: 12/31/2022] Open
Abstract
Antibodies have been shown to be a potent therapeutic tool. However, their use for targeting brain diseases, including neurodegenerative diseases and brain cancers, has been limited, particularly because the blood–brain barrier (BBB) makes brain tissue hard to access by conventional antibody-targeting strategies. In this review, we summarize new antibody therapeutic approaches to target brain tumors, especially malignant gliomas, as well as their potential drawbacks. Many different brain delivery platforms for antibodies have been studied such as liposomes, nanoparticle-based systems, cell-penetrating peptides (CPPs), and cell-based approaches. We have already shown the successful delivery of single-chain fragment variable (scFv) with CPP as a linker between two variable domains in the brain. Antibodies normally face poor penetration through the BBB, with some variants sufficiently passing the barrier on their own. A “Trojan horse” method allows passage of biomolecules, such as antibodies, through the BBB by receptor-mediated transcytosis (RMT). Such examples of therapeutic antibodies are the bispecific antibodies where one binding specificity recognizes and binds a BBB receptor, enabling RMT and where a second binding specificity recognizes an antigen as a therapeutic target. On the other hand, cell-based systems such as stem cells (SCs) are a promising delivery system because of their tumor tropism and ability to cross the BBB. Genetically engineered SCs can be used in gene therapy, where they express anti-tumor drugs, including antibodies. Different types and sources of SCs have been studied for the delivery of therapeutics to the brain; both mesenchymal stem cells (MSCs) and neural stem cells (NSCs) show great potential. Following the success in treatment of leukemias and lymphomas, the adoptive T-cell therapies, especially the chimeric antigen receptor-T cells (CAR-Ts), are making their way into glioma treatment as another type of cell-based therapy using the antibody to bind to the specific target(s). Finally, the current clinical trials are reviewed, showing the most recent progress of attractive approaches to deliver therapeutic antibodies across the BBB aiming at the specific antigen.
Collapse
Affiliation(s)
- Rok Razpotnik
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Neža Novak
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Vladka Čurin Šerbec
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Uros Rajcevic
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
41
|
|
42
|
Li B, McCrudden CM, Yuen HF, Xi X, Lyu P, Chan KW, Zhang SD, Kwok HF. CD133 in brain tumor: the prognostic factor. Oncotarget 2017; 8:11144-11159. [PMID: 28055976 PMCID: PMC5355253 DOI: 10.18632/oncotarget.14406] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 12/26/2016] [Indexed: 12/31/2022] Open
Abstract
CD133 has been shown to be an important stem cell factor that promotes glioma progression. However, the mechanism for CD133-mediated glioma progression has yet to be fully elucidated. In this study, we found that CD133 mRNA expression was a prognostic marker in three independent glioma patient cohorts, corroborating a putative role for CD133 in glioma progression. Importantly, we found that CD133 expression in glioma was highly correlated with the expression of HOX gene stem cell factors (HOXA5, HOXA7, HOXA10, HOXC4 and HOXC6). The expression of these HOX genes individually was significantly associated with survival. Interestingly, the prognostic significance of CD133 was dependent on the expression level of HOX genes, and vice versa. CD133 (p = 0.021) and HOXA7 (p = 0.001) were independent prognostic markers when the three glioma patient cohorts were combined (n = 231). Our results suggest that HOX genes may play a more important role in progression of glioma when CD133 expression is low. Furthermore, we showed that low-level expression of LIM2 in CD133-high glioma was associated with poorer survival, suggesting that LIM2 could be a therapeutic target for glioma expressing a high level of CD133. Connectivity mapping identified vinblastine and vincristine as agents that could reverse the CD133/HOX genes/LIM2-signature, and we confirmed this by in vitro analysis in glioma cell lines, demonstrating that CD133 and HOX genes were co-expressed and could be downregulated by vincristine. In conclusion, our data show that CD133 and HOX genes are important prognostic markers in glioma and shed light on possible treatment strategies for glioma expressing a high level of CD133.
Collapse
Affiliation(s)
- Bin Li
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Cian M McCrudden
- School of Pharmacy, Queen's University Belfast, Belfast, United Kingdom
| | - Hiu Fung Yuen
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Xinping Xi
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Peng Lyu
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Kwok Wah Chan
- Department of Pathology, University of Hong Kong, Hong Kong
| | - Shu Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, Londonderry, United Kingdom
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| |
Collapse
|
43
|
Wang Y, Ying X, Xu H, Yan H, Li X, Tang H. The functional curcumin liposomes induce apoptosis in C6 glioblastoma cells and C6 glioblastoma stem cells in vitro and in animals. Int J Nanomedicine 2017; 12:1369-1384. [PMID: 28260885 PMCID: PMC5325138 DOI: 10.2147/ijn.s124276] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma is a kind of malignant gliomas that is almost impossible to cure due to the poor drug transportation across the blood–brain barrier and the existence of glioma stem cells. We prepared a new kind of targeted liposomes in order to improve the drug delivery system onto the glioma cells and induce the apoptosis of glioma stem cells afterward. In this experiment, curcumin was chosen to kill gliomas, while quinacrine was used to induce apoptosis of the glioma stem cells. Also, p-aminophenyl-α-D-mannopyranoside could facilitate the transport of liposomes across the blood–brain barrier and finally target the brain glioma cells. The cell experiments in vitro indicated that the targeted liposomes could significantly improve the anti-tumor effects of the drugs, while enhancing the uptake effects, apoptosis effects, and endocytic effects of C6 glioma cells and C6 glioma stem cells. Given the animal experiments in vivo, we discovered that the targeted liposomes could obviously increase the survival period of brain glioma-bearing mice and inhibit the growth of gliomas. In summary, curcumin and quinacrine liposomes modified with p-aminophenyl-α-D-mannopyranoside is a potential preparation to treat brain glioma cells and brain glioma stem cells.
Collapse
Affiliation(s)
- Yahua Wang
- Key Laboratory of Xinjiang Phytomedicine Resources and Modernization of TCM, School of Pharmaceutical Sciences, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Xue Ying
- Key Laboratory of Xinjiang Phytomedicine Resources and Modernization of TCM, School of Pharmaceutical Sciences, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Haolun Xu
- Key Laboratory of Xinjiang Phytomedicine Resources and Modernization of TCM, School of Pharmaceutical Sciences, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Helu Yan
- Key Laboratory of Xinjiang Phytomedicine Resources and Modernization of TCM, School of Pharmaceutical Sciences, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Xia Li
- Key Laboratory of Xinjiang Phytomedicine Resources and Modernization of TCM, School of Pharmaceutical Sciences, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Hui Tang
- Key Laboratory of Xinjiang Phytomedicine Resources and Modernization of TCM, School of Pharmaceutical Sciences, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| |
Collapse
|
44
|
Identification of two distinct mesenchymal stromal cell populations in human malignant glioma. J Neurooncol 2016; 131:245-254. [PMID: 27757723 PMCID: PMC5306185 DOI: 10.1007/s11060-016-2302-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 10/09/2016] [Indexed: 12/25/2022]
Abstract
Gene profiling has revealed that malignant gliomas can be divided into four distinct molecular subtypes, where tumors with a mesenchymal gene expression are correlated with short survival. The present investigation was undertaken to clarify whether human malignant gliomas contain endogenous mesenchymal stromal cells (MSC), fulfilling consensus criteria defined by The International Society for Cellular Therapy, recruited from the host. We found that MSC-like cells can be isolated from primary human malignant gliomas. Two distinct MSC-like cell populations, differing in their expression of the CD90 surface marker, were discovered after cell sorting. RNA sequencing revealed further genetic differences between these two cell populations and MSC-like cells lacking CD90 produced higher amounts of VEGF and PGE2 compared to cells with the true MSC phenotype, implying that the CD90− MSC-like cells most probably are more active in tumor vascularization and immunosuppression than their CD90+ counterpart. The results highlight the CD90− subpopulation as an important tumor component, however, its functional effects in glioma remains to be resolved. Using the protocols presented here, it will be possible to isolate, characterize and analyze brain tumor-derived MSC-like cells in more detail and to further test their functions in vitro and in in vivo xenograft models of glioma.
Collapse
|
45
|
Mangraviti A, Gullotti D, Tyler B, Brem H. Nanobiotechnology-based delivery strategies: New frontiers in brain tumor targeted therapies. J Control Release 2016; 240:443-453. [DOI: 10.1016/j.jconrel.2016.03.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 02/05/2016] [Accepted: 03/18/2016] [Indexed: 02/06/2023]
|
46
|
Lépinoux-Chambaud C, Barreau K, Eyer J. The Neurofilament-Derived Peptide NFL-TBS.40-63 Targets Neural Stem Cells and Affects Their Properties. Stem Cells Transl Med 2016; 5:901-13. [PMID: 27177578 DOI: 10.5966/sctm.2015-0221] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 02/23/2016] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED Targeting neural stem cells (NSCs) in the adult brain represents a promising approach for developing new regenerative strategies, because these cells can proliferate, self-renew, and differentiate into new neurons, astrocytes, and oligodendrocytes. Previous work showed that the NFL-TBS.40-63 peptide, corresponding to the sequence of a tubulin-binding site on neurofilaments, can target glioblastoma cells, where it disrupts their microtubules and inhibits their proliferation. We show that this peptide targets NSCs in vitro and in vivo when injected into the cerebrospinal fluid. Although neurosphere formation was not altered by the peptide, the NSC self-renewal capacity and proliferation were reduced and were associated with increased adhesion and differentiation. These results indicate that the NFL-TBS.40-63 peptide represents a new molecular tool to target NSCs to develop new strategies for regenerative medicine and the treatment of brain tumors. SIGNIFICANCE In the present study, the NFL-TBS.40-63 peptide targeted neural stem cells in vitro when isolated from the subventricular zone and in vivo when injected into the cerebrospinal fluid present in the lateral ventricle. The in vitro formation of neurospheres was not altered by the peptide; however, at a high concentration of the peptide, the neural stem cell (NSC) self-renewal capacity and proliferation were reduced and associated with increased adhesion and differentiation. These results indicate that the NFL-TBS.40-63 peptide represents a new molecular tool to target NSCs to develop new strategies for regenerative medicine and the treatment of brain tumors.
Collapse
Affiliation(s)
- Claire Lépinoux-Chambaud
- Laboratoire Neurobiologie et Transgenese, Université Nantes, Angers, Le Mans, Unité Propre de Recherche de l'Enseignement Supérieur EA-3143, Institut de Biologie en Santé, L'Université d'Angers, Centre Hospitalier Universitaire, Angers, France
| | - Kristell Barreau
- Laboratoire Neurobiologie et Transgenese, Université Nantes, Angers, Le Mans, Unité Propre de Recherche de l'Enseignement Supérieur EA-3143, Institut de Biologie en Santé, L'Université d'Angers, Centre Hospitalier Universitaire, Angers, France
| | - Joël Eyer
- Laboratoire Neurobiologie et Transgenese, Université Nantes, Angers, Le Mans, Unité Propre de Recherche de l'Enseignement Supérieur EA-3143, Institut de Biologie en Santé, L'Université d'Angers, Centre Hospitalier Universitaire, Angers, France
| |
Collapse
|
47
|
Korpusik A, Kolev M. Single injection of CD8+ T lymphocytes derived from hematopoietic stem cells - Mathematical and numerical insights. Biosystems 2016; 144:46-54. [PMID: 27095371 DOI: 10.1016/j.biosystems.2016.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 04/01/2016] [Accepted: 04/14/2016] [Indexed: 12/22/2022]
Abstract
Recently, hematopoietic stem cell (HSC) based therapy is being discussed as a possible treatment for HIV infection. The main advantage of this approach is that it limits the immune impairing effect of infection by introducing an independent influx of antigen-specific cytotoxic T lymphocytes (CTL). In this paper, we present a mathematical approach to predict the dynamics of HSC based therapy. We use a modification of a basic mathematical model for virus induced impairment of help to study how virus - immune system dynamics can be influenced by a single injection of CD8+ T lymphocytes derived from hematopoietic stem cells. Our mathematical and numerical results indicate that a single, large enough dose of genetically derived CTL may lead to restoration of the cellular immune response and result in long-term control of infection.
Collapse
Affiliation(s)
- Adam Korpusik
- Faculty of Technical Sciences, University of Warmia and Mazury, ul. Oczapowskiego 11, 10-719 Olsztyn, Poland.
| | - Mikhail Kolev
- Faculty of Mathematics and Computer Science, University of Warmia and Mazury, ul. Słoneczna 54, 10-710 Olsztyn, Poland.
| |
Collapse
|
48
|
Hulou MM, Cho CF, Chiocca EA, Bjerkvig R. Experimental therapies: gene therapies and oncolytic viruses. HANDBOOK OF CLINICAL NEUROLOGY 2016; 134:183-197. [PMID: 26948355 DOI: 10.1016/b978-0-12-802997-8.00011-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Glioblastoma is the most common and aggressive primary brain tumor in adults. Over the past three decades, the overall survival time has only improved by a few months, therefore novel alternative treatment modalities are needed to improve clinical management strategies. Such strategies should ultimately extend patient survival. At present, the extensive insight into the molecular biology of gliomas, as well as into genetic engineering techniques, has led to better decision processes when it comes to modifying the genome to accommodate suicide genes, cytokine genes, and tumor suppressor genes that may kill cancer cells, and boost the host defensive immune system against neoantigenic cytoplasmic and nuclear targets. Both nonreplicative viral vectors and replicating oncolytic viruses have been developed for brain cancer treatment. Stem cells, microRNAs, nanoparticles, and viruses have also been designed. These have been armed with transgenes or peptides, and have been used both in laboratory-based experiments as well as in clinical trials, with the aim of improving selective killing of malignant glioma cells while sparing normal brain tissue. This chapter reviews the current status of gene therapies for malignant gliomas and highlights the most promising viral and cell-based strategies under development.
Collapse
Affiliation(s)
- M Maher Hulou
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Choi-Fong Cho
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - E Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rolf Bjerkvig
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg; Department of Biomedicine, University of Bergen, Norway
| |
Collapse
|
49
|
Ropper AE, Zeng X, Haragopal H, Anderson JE, Aljuboori Z, Han I, Abd-El-Barr M, Lee HJ, Sidman RL, Snyder EY, Viapiano MS, Kim SU, Chi JH, Teng YD. Targeted Treatment of Experimental Spinal Cord Glioma With Dual Gene-Engineered Human Neural Stem Cells. Neurosurgery 2015; 79:481-91. [DOI: 10.1227/neu.0000000000001174] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Abstract
BACKGROUND
There are currently no satisfactory treatments or experimental models showing autonomic dysfunction for intramedullary spinal cord gliomas (ISCG).
OBJECTIVE
To develop a rat model of ISCG and investigate whether genetically engineered human neural stem cells (F3.hNSCs) could be developed into effective therapies for ISCG.
METHODS
Immunodeficient/Rowett Nude rats received C6 implantation of G55 human glioblastoma cells (10K/each). F3.hNSCs engineered to express either cytosine deaminase gene only (i.e., F3.CD) or dual genes of CD and thymidine kinase (i.e., F3.CD-TK) converted benign 5-fluorocytosine and ganciclovir into oncolytic 5-fluorouracil and ganciclovir-triphosphate, respectively. ISCG rats received injection of F3.CD-TK, F3.CD, or F3.CD-TK debris near the tumor epicenter 7 days after G55 seeding, followed with 5-FC (500 mg/kg/5 mL) and ganciclovir administrations (25 mg/kg/1 mL/day × 5/each repeat, intraperitoneal injection). Per humane standards for animals, loss of weight-bearing stepping in the hindlimb was used to determine post-tumor survival. Also evaluated were autonomic functions and tumor growth rate in vivo.
RESULTS
ISCG rats with F3.CD-TK treatment survived significantly longer (37.5 ± 4.78 days) than those receiving F3.CD (21.5 ± 1.75 days) or F3.CD-TK debris (19.3 ± 0.85 days; n = 4/group; P <.05, median rank test), with significantly improved autonomic function and reduced tumor growth rate. F3.DC-TK cells migrated diffusively into ISCG clusters to mediate oncolytic effect.
CONCLUSION
Dual gene-engineered human neural stem cell regimen markedly prolonged survival in a rat model that emulates somatomotor and autonomic dysfunctions of human cervical ISCG. F3.CD-TK may provide a novel approach to treating clinical ISCG.
Collapse
Affiliation(s)
- Alexander E. Ropper
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Xiang Zeng
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Hariprakash Haragopal
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Jamie E. Anderson
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Zaid Aljuboori
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Inbo Han
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Muhammad Abd-El-Barr
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hong Jun Lee
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | - Richard L. Sidman
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | - Evan Y. Snyder
- Stem Cell Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Mariano S. Viapiano
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Seung U. Kim
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John H. Chi
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Yang D. Teng
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Division of SCI Research, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Department of PM&R, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
50
|
Wu N, Liu J, Zhao X, Yan Z, Jiang B, Wang L, Cao S, Shi D, Lin X. Cardamonin induces apoptosis by suppressing STAT3 signaling pathway in glioblastoma stem cells. Tumour Biol 2015; 36:9667-9676. [PMID: 26150336 DOI: 10.1007/s13277-015-3673-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 06/15/2015] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma stem cells (GSCs) are the initiating cells in glioblastoma multiforme (GBM) and contribute to the resistance of GBM to chemotherapy and radiation. In the present study, we investigated the effects of cardamonin (3,4,2,4-tetrahydroxychalcone) on the self-renewal and apoptosis of GSCs, and if its action is associated with signal transducer and activator of transcription 3 (STAT3) pathway. CD133(+) GSCs, a kind of GSCs line, was established from human glioblastoma tissues. Cardamonin inhibited the proliferation and induced apoptosis in CD133+ GSCs. The proapoptotic effects of temozolomide (TMZ) were further enhanced by cardamonin in CD133+ GSCs and U87 cells in vitro. For in vivo study, injection of 5 × 10(5) cells of CD133+ GSCs subcutaneously (s.c.) into nude mice, 100 % of large tumors were developed within 8 weeks in all mice; in contrast, only one out of five mice developed a small tumor when 5 × 10(5) cells of CD133(-) GMBs cells were injected. Cardamonin also inhibited STAT3 activation by luciferase assay and suppressed the expression of the downstream genes of STAT3, such as Bcl-XL, Bcl-2, Mcl-1, survivin, and VEGF. Furthermore, cardamonin locked nuclear translocation and dimerization of STAT3 in CD133(+) GSCs. Docking analysis confirmed that cardamonin molecule was successfully docked into the active sites of STAT3 with a highly favorable binding energy of -10.78 kcal/mol. The study provides evidence that cardamonin is a novel inhibitor of STAT3 and has the potential to be developed as a new anticancer agent targeting GSCs. This study also reveals that targeting STAT3 signal pathway is an important strategy for the treatment of human GBM.
Collapse
Affiliation(s)
- Ning Wu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Jia Liu
- College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xiangzhong Zhao
- Qingdao Medical University Affiliated Hospital, Qingdao, 266070, China
| | - Zhiyong Yan
- Qingdao Medical University Affiliated Hospital, Qingdao, 266070, China
| | - Bo Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Lijun Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Shousong Cao
- Chifeng Saliont Pharmaceutical Co., Ltd, Chifeng, Inner Mongolia Autonomous Region, China
| | - Dayong Shi
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China.
| | - Xiukun Lin
- Department of Pharmacology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|