1
|
del Rocío Aguilera-Marquez J, Manzanares-Guzmán A, García-Uriostegui L, Canales-Aguirre AA, Camacho-Villegas TA, Lugo-Fabres PH. Alginate-Gelatin Hydrogel Scaffold Model for Hypoxia Induction in Glioblastoma Embedded Spheroids. Gels 2025; 11:263. [PMID: 40277699 PMCID: PMC12026674 DOI: 10.3390/gels11040263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and malignant brain tumor, characterized by hypoxia in its microenvironment, which drives its growth and resistance to treatments. Hypoxia-inducible factor 1 (HIF-1) plays a central role in GBM progression by regulating cellular adaptation to low oxygen availability, promoting processes such as angiogenesis and cell invasion. However, studying and modeling GBM under hypoxic conditions is complex, especially due to the limitations of animal models. In this study, we developed a glioma spheroid model using an alginate-gelatin hydrogel scaffold, which enabled the simulation of hypoxic conditions within the tumor. The scaffold-based model demonstrated high reproducibility, facilitating the analysis of HIF-1α expression, a key protein in the hypoxic response of GBM. Furthermore, cell viability, the microstructural features of the encapsulated spheroids, and the water absorption rate of the hydrogel were assessed. Our findings validate the three-dimensional (3D) glioblastoma spheroids model as a valuable platform for studying hypoxia in GBM and evaluating new therapies. This approach could offer a more accessible and specific alternative for studying the tumor microenvironment and therapeutic resistance in GBM.
Collapse
Affiliation(s)
- Janette del Rocío Aguilera-Marquez
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico; (J.d.R.A.-M.); (A.M.-G.); (A.A.C.-A.); (T.A.C.-V.)
| | - Alejandro Manzanares-Guzmán
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico; (J.d.R.A.-M.); (A.M.-G.); (A.A.C.-A.); (T.A.C.-V.)
| | - Lorena García-Uriostegui
- SECIHTI-Secretaría de Ciencia, Humanidades, Tecnología e Innovación-Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara 44430, Mexico;
| | - Alejandro A. Canales-Aguirre
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico; (J.d.R.A.-M.); (A.M.-G.); (A.A.C.-A.); (T.A.C.-V.)
| | - Tanya A. Camacho-Villegas
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico; (J.d.R.A.-M.); (A.M.-G.); (A.A.C.-A.); (T.A.C.-V.)
| | - Pavel H. Lugo-Fabres
- SECIHTI-Secretaría de Ciencia, Humanidades, Tecnología e Innovación-Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara 44270, Mexico
| |
Collapse
|
2
|
Orasanu CI, Aschie M, Deacu M, Bosoteanu M, Vamesu S, Enciu M, Cozaru GC, Mitroi AF, Ghitoi SA, Cretu AM, Ursica OA, Voda RI. Comparative Clinical-Imaging and Histogenetic Analysis Between Astrocytoma IDH-Mutant Grade 4 and Glioblastoma IDH-Wildtype-Is There Really a Worse One? Diagnostics (Basel) 2025; 15:438. [PMID: 40002588 PMCID: PMC11854731 DOI: 10.3390/diagnostics15040438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Brain tumors pose a significant health threat, leading to high morbidity and mortality rates. Astrocytoma IDH-mutant grade 4 (A4IDHmt) and glioblastoma IDH-wildtype (G4IDHwt) exhibit similar clinical and imaging characteristics. This study aims to highlight the differences in their clinical evolution and histogenetic aspects with the possible therapeutic impact, as well as the adverse prognostic factors in patient survival. Methods: We performed a 10-year retrospective study of grade 4 gliomas, evaluating immunomarkers and FISH tests. We also quantified tumor necrosis and microvascular density. Results: A total of 81 cases were identified; 54.32% were A4IDHmt. We observed that A4IDHmt patients were younger (34.10% under 50) and had a higher survival rate (4.55%). This group also exhibited a more pronounced microvascular density (p = 0.010) and proliferative index (p = 0.026). G4IDHwt was associated with larger tumor volumes (94.84 cm3 vs. 86.14 cm3), lower resectability rates (82.88% vs. 87.67%), and a more significant immature cell population (83.78% vs. 68.18%). In the case of both, the negative risk on survival in the univariate analysis is given by advanced age (A4IDHmt: HR = 1.035, G4IDHwt: HR = 1.045) and p53 immunopositivity (A4IDHmt: HR = 6.962, G4IDHwt: HR = 4.680). Conclusions: The negative risk factors for A4IDHmt include the rapid onset of clinical symptoms (HR = 2.038), diabetes mellitus (HR = 2.311), arterial hypertension (HR = 2.325), residual tumor (HR = 2.662), increased residual tumor volume (HR = 1.060), increased microvascular density (HR = 1.096), and high tumor necrosis (HR = 1.097). For G4IDHwt, the negative risk factors consist of increased residual volume (HR = 1.023), lost PTEN immunoreaction (HR = 33.133), and unmethylated DNA status (HR = 6.765, respectively HR = 20.573). Even if it has more risk factors, A4IDHmt is the lesser evil.
Collapse
Affiliation(s)
- Cristian Ionut Orasanu
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University, 900591 Constanta, Romania
| | - Mariana Aschie
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University, 900591 Constanta, Romania
- Department of Anatomy, Academy of Medical Sciences of Romania, 030171 Bucharest, Romania
- Department VIII—Medical Sciences, The Romanian Academy of Scientists, 030167 Bucharest, Romania
| | - Mariana Deacu
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
| | - Madalina Bosoteanu
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
| | - Sorin Vamesu
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
| | - Manuela Enciu
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
| | - Georgeta Camelia Cozaru
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University, 900591 Constanta, Romania
- Department of Genetics, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
| | - Anca Florentina Mitroi
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University, 900591 Constanta, Romania
- Department of Genetics, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
| | - Sinziana Andra Ghitoi
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
| | - Ana Maria Cretu
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University, 900591 Constanta, Romania
| | - Oana Andreea Ursica
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
| | - Raluca Ioana Voda
- Department of Pathology, Clinical Service of Pathology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University, 900591 Constanta, Romania
| |
Collapse
|
3
|
Chida D, Okita Y, Utsugi R, Kuroda H, Hirayama R, Kijima N, Arisawa A, Kagawa N, Kanemura Y, Yoshimura S, Tomiyama N, Kishima H. Dynamic susceptibility contrast‑enhanced perfusion magnetic resonance imaging parameters for predicting MGMT promoter methylation and prognostic value in newly diagnosed patients with glioblastoma. Oncol Lett 2024; 28:610. [PMID: 39493435 PMCID: PMC11528182 DOI: 10.3892/ol.2024.14741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/09/2024] [Indexed: 11/05/2024] Open
Abstract
O6-methylguanine DNA methyltransferase (MGMT) promoter methylation is an important clinical biomarker of newly diagnosed glioblastoma. Previous radiological studies using dynamic susceptibility contrast (DSC) magnetic resonance imaging (MRI) perfusion have aimed to predict MGMT methylation status non-invasively in gliomas with radiological characteristics. The possibility of predicting MGMT methylation status using DSC-MRI perfusion with a radiological approach remains controversial. The present study aimed to evaluate the usefulness of MRI perfusion parameters as non-invasive markers to predict MGMT methylation status and prognosis in newly diagnosed glioblastoma patients. Thus, 50 patients with histologically confirmed primary glioblastoma, IDH-wildtype who underwent tumor resection at Osaka University Hospital (Suita, Japan) between January 2017 and January 2023 were included in this study. The mean cerebral blood volume (CBV) ratio (rCBV) and cerebral blood flow (CBF) ratio (rCBF) for tumors with MGMT methylation (mean rCBV:2.09 and mean rCBF:3.08) were significantly higher compared with those for tumors without MGMT methylation (mean rCBV:1.33 and mean rCBF:1.85; P<0.05). While patients with MGMT methylation had longer progression-free survival (PFS) compared with those without MGMT methylation (P<0.05), there was no significant difference in OS with or without MGMT methylation (P=0.06). By contrast, there was no association between MRI perfusion parameters and OS or PFS in patients with glioblastoma. Furthermore, the association between CBV, CBF, MGMT promotor methylation status, OS, and PFS were explored. There was no significant prognostic difference between low vascularity tumors (rCBV <1.3 or rCBF <1.8) with or without MGMT methylation. On the other hand, high vascularity tumors (rCBF ≥1.8) with MGMT promotor methylation were associated to longer OS and PFS compared with those without. However, there was no association between MGMT methylation status and OS or PFS in patients with high rCBV (rCBV ≥1.3). The present study indicated that CBV and CBF could be used to predict the MGMT methylation status in glioblastomas. However, the prognostic value of tumor vascularity and MGMT methylation status may be limited.
Collapse
Affiliation(s)
- Daiki Chida
- Department of Neurosurgery, Hyogo Medical University, Nishinomiya, Hyogo 663-8501, Japan
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yoshiko Okita
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Reina Utsugi
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hideki Kuroda
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Ryuichi Hirayama
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Noriyuki Kijima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Atsuko Arisawa
- Department of Diagnostic Radiology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Naoki Kagawa
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Yonehiro Kanemura
- Department of Neurosurgery, NHO Osaka National Hospital, Osaka 540-0006, Japan
- Division of Regenerative Medicine, Institute for Clinical Research, NHO Osaka National Hospital, Osaka 540-0006, Japan
| | - Shinichi Yoshimura
- Department of Neurosurgery, Hyogo Medical University, Nishinomiya, Hyogo 663-8501, Japan
| | - Noriyuki Tomiyama
- Department of Diagnostic Radiology, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Haruhiko Kishima
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
4
|
Peddinti V, Rout B, Agnihotri TG, Gomte SS, Jain A. Functionalized liposomes: an enticing nanocarrier for management of glioma. J Liposome Res 2024; 34:349-367. [PMID: 37855432 DOI: 10.1080/08982104.2023.2270060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/07/2023] [Indexed: 10/20/2023]
Abstract
Glioma is one of the most severe central nervous systems (CNS)-specific tumors, with rapidly growing malignant glial cells accounting for roughly half of all brain tumors and having a poor survival rate ranging from 12 to 15 months. Despite being the most often used technique for glioma therapy, conventional chemotherapy suffers from low permeability of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) to anticancer drugs. When it comes to nanocarriers, liposomes are thought of as one of the most promising nanocarrier systems for glioma treatment. However, owing to BBB tight junctions, non-targeted liposomes, which passively accumulate in most cancer cells primarily via the increased permeability and retention effect (EPR), would not be suitable for glioma treatment. The surface modification of liposomes with various active targeting ligands has shown encouraging outcomes in the recent times by allowing various chemotherapy drugs to pass across the BBB and BBTB and enter glioma cells. This review article introduces by briefly outlining the landscape of glioma, its classification, and some of the pathogenic causes. Further, it discusses major barriers for delivering drugs to glioma such as the BBB, BBTB, and tumor microenvironment. It further discusses modified liposomes such as long-acting circulating liposomes, actively targeted liposomes, stimuli responsive liposomes. Finally, it highlighted the limitations of liposomes in the treatment of glioma and the various actively targeted liposomes undergoing clinical trials for the treatment of glioma.
Collapse
Affiliation(s)
- Vasu Peddinti
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Biswajit Rout
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India
| |
Collapse
|
5
|
Dreute J, Pfisterer M, Schmitz ML. A reductionist perspective on HIF-1α's role in cell proliferation under non-hypoxic conditions. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119683. [PMID: 38301905 DOI: 10.1016/j.bbamcr.2024.119683] [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: 10/18/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/03/2024]
Abstract
The role of hypoxia-inducible factor (HIF)-1α in the control of proliferation under non-hypoxic conditions has been investigated in numerous studies, but does not yield a coherent picture. Therefore, we conducted this meta-analysis of existing literature to systematically evaluate the role of HIF-1α, based on a number of inclusion and exclusion criteria. Studies analyzing non-transformed, primary cells showed a largely heterogeneous distribution of pro-proliferative, anti-proliferative or absent functions for HIF-1α, which are co-determined by several parameters, including the type and age of the cell and its localization in tissues and organs. In contrast, the analyses of tumor cells showed a predominantly pro-proliferative role of HIF-1α by cell-intrinsic and cell-extrinsic molecular mechanism not yet understood.
Collapse
Affiliation(s)
- Jan Dreute
- Institute of Biochemistry, Justus-Liebig-University Giessen, Germany
| | | | | |
Collapse
|
6
|
Zeng Y, Zhao L, Li K, Ma J, Chen D, Liu C, Zhan W, Zhan Y. Aptamer-functionalized nanoplatforms overcoming temozolomide resistance in synergistic chemo/photothermal therapy through alleviating tumor hypoxia. NANO RESEARCH 2023; 16:9859-9872. [DOI: 10.1007/s12274-023-5742-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 01/03/2025]
|
7
|
Eatmann AI, Hamouda E, Hamouda H, Farouk HK, Jobran AWM, Omar AA, Madeeh AK, Al-Dardery NM, Elnoamany S, Abd-Elnasser EG, Koraiem AM, Ahmed AA, Abouzid M, Karaźniewicz-Łada M. Potential Use of Thalidomide in Glioblastoma Treatment: An Updated Brief Overview. Metabolites 2023; 13:metabo13040543. [PMID: 37110201 PMCID: PMC10146416 DOI: 10.3390/metabo13040543] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/05/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults. Thalidomide is a vascular endothelial growth factor inhibitor that demonstrates antiangiogenic activity, and may provide additive or synergistic anti-tumor effects when co-administered with other antiangiogenic medications. This study is a comprehensive review that highlights the potential benefits of using thalidomide, in combination with other medications, to treat glioblastoma and its associated inflammatory conditions. Additionally, the review examines the mechanism of action of thalidomide in different types of tumors, which may be beneficial in treating glioblastoma. To our knowledge, a similar study has not been conducted. We found that thalidomide, when used in combination with other medications, has been shown to produce better outcomes in several conditions or symptoms, such as myelodysplastic syndromes, multiple myeloma, Crohn's disease, colorectal cancer, renal failure carcinoma, breast cancer, glioblastoma, and hepatocellular carcinoma. However, challenges may persist for newly diagnosed or previously treated patients, with moderate side effects being reported, particularly with the various mechanisms of action observed for thalidomide. Therefore, thalidomide, used alone, may not receive significant attention for use in treating glioblastoma in the future. Conducting further research by replicating current studies that show improved outcomes when thalidomide is combined with other medications, using larger sample sizes, different demographic groups and ethnicities, and implementing enhanced therapeutic protocol management, may benefit these patients. A meta-analysis of the combinations of thalidomide with other medications in treating glioblastoma is also needed to investigate its potential benefits further.
Collapse
Affiliation(s)
- Ahmed Ismail Eatmann
- Department of Cell Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, 31-007 Kraków, Poland
| | - Esraa Hamouda
- Faculty of Medicine, Menoufia University, Menoufia P.O. Box 5744, Egypt
| | - Heba Hamouda
- Faculty of Medicine, Menoufia University, Menoufia P.O. Box 5744, Egypt
| | | | - Afnan W M Jobran
- Faculty of Medicine, Al Quds University, Jerusalem P.O. Box 51000, Palestine
| | - Abdallah A Omar
- Department of Pharmaceutical Services and Sciences, Children's Cancer Hospital Egypt (CCHE-57357), Cairo 11617, Egypt
| | | | | | - Salma Elnoamany
- Faculty of Medicine, Menoufia University, Menoufia P.O. Box 5744, Egypt
| | | | | | - Alhassan Ali Ahmed
- Department of Bioinformatics and Computational Biology, Poznan University of Medical Sciences, 60-812 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Mohamed Abouzid
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| | - Marta Karaźniewicz-Łada
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
| |
Collapse
|
8
|
Photodynamic therapy enhances the cytotoxicity of temozolomide against glioblastoma via reprogramming anaerobic glycolysis. Photodiagnosis Photodyn Ther 2023; 42:103342. [PMID: 36781008 DOI: 10.1016/j.pdpdt.2023.103342] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/21/2022] [Accepted: 02/10/2023] [Indexed: 02/13/2023]
Abstract
The successful application of photodynamic therapy in the treatment of glioma (CNS WHO grade 4) depends in large part to the effect of killing cells in the infiltrating area after tumor had been removed, when combined with radiotherapy, chemotherapy, and targeted drug therapy. The purpose of this study was to investigate the potential mechanism of TMZ's involvement in the glioma's glycolytic metabolic pathway during photodynamic therapy. The low dose of photodynamic therapy treatment on the cell viability of gliomas was investigated by CCK8. Alterations in reactive oxygen species were detected by flow cytometer. The differentially expressed proteins related to glucose transporter 1 (GLUT-1), matrix metalloproteinase-2 (MMP-2)/actively MMP-2 and apoptosis-associated caspase-3/cleaved caspase-3 were evaluated by Western Blot experiment. Additionally, transmission electron microscopy observed apoptosis, necrosis and the changes of the ultrastructure in U251 cells. In addition, antitumor effects in vivo were tested using orthotopic BALB/c mice with the glioma U87 model. The findings showed that low dose PDT affected mitochondrial function by inducing radical oxygen, hindered cellular glucose transport and metabolism, and induced apoptosis. The results also showed that cell viability considerably decreased and increased cell apoptosis under the PDT therapy. The HIF-1/GLUT-1 axis enhanced the cytotoxicity of temozolomide in gliomas as a result of PDT treatment, which was influenced by ROS. As a result, this study presents PDT as a potential therapeutic approach for treating malignant glioma, and enhanced antitumor effect of TMZ by inhibiting glycolytic pathway.
Collapse
|
9
|
Beylerli O, Sufianova G, Shumadalova A, Zhang D, Gareev I. MicroRNAs-mediated regulation of glucose transporter (GLUT) expression in glioblastoma. Noncoding RNA Res 2022; 7:205-211. [PMID: 36157351 PMCID: PMC9467858 DOI: 10.1016/j.ncrna.2022.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/23/2022] [Accepted: 09/01/2022] [Indexed: 12/01/2022] Open
Abstract
Current knowledge about the role of microRNAs (miRNAs) in tumor glucose metabolism is growing, and a number of studies regularly confirm the impact miRNAs can have on glucose metabolism reprogramming in tumors. However, there remains a lack of understanding of the broader perspective on the role of miRNAs in energy reprogramming in glioblastoma. An important role in the metabolism of glucose is played by carrier proteins that ensure its transmembrane movement. Carrier proteins in mammalian cells are glucose transporters (GLUTs). In total, 12 types of GLUTs are distinguished, differing in localization, affinity for glucose and ability to regulate. The fact of increased consumption of glucose in tumors compared to non-proliferating normal tissues is known. Tumor cells need glucose to ensure their survival and growth, so the type of transport proteins like GLUT are critical for them. Previous studies have shown that GLUT-1 and GLUT-3 may play an important role in the development of some types of malignant tumors, including glioblastoma. In addition, there is evidence of how GLUT-1 and GLUT-3 expression is regulated by miRNAs in glioblastoma. Thus, the aim of this study is to highlight the role of specific miRNAs in modulating GLUT levels in order to take into account the use of miRNAs expression modulators as a useful strategy to increase the sensitivity of glioblastoma to current therapies.
Collapse
Affiliation(s)
- Ozal Beylerli
- Рeoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| | - Galina Sufianova
- Department of Pharmacology, Tyumen State Medical University, 54 Odesskaya Street, 625023, Tyumen, Russia
| | - Alina Shumadalova
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Daming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Ilgiz Gareev
- Рeoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| |
Collapse
|
10
|
Glioma diagnosis and therapy: Current challenges and nanomaterial-based solutions. J Control Release 2022; 352:338-370. [PMID: 36206948 DOI: 10.1016/j.jconrel.2022.09.065] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022]
Abstract
Glioma is often referred to as one of the most dreadful central nervous system (CNS)-specific tumors with rapidly-proliferating cancerous glial cells, accounting for nearly half of the brain tumors at an annual incidence rate of 30-80 per a million population. Although glioma treatment remains a significant challenge for researchers and clinicians, the rapid development of nanomedicine provides tremendous opportunities for long-term glioma therapy. However, several obstacles impede the development of novel therapeutics, such as the very tight blood-brain barrier (BBB), undesirable hypoxia, and complex tumor microenvironment (TME). Several efforts have been dedicated to exploring various nanoformulations for improving BBB permeation and precise tumor ablation to address these challenges. Initially, this article briefly introduces glioma classification and various pathogenic factors. Further, currently available therapeutic approaches are illustrated in detail, including traditional chemotherapy, radiotherapy, and surgical practices. Then, different innovative treatment strategies, such as tumor-treating fields, gene therapy, immunotherapy, and phototherapy, are emphasized. In conclusion, we summarize the article with interesting perspectives, providing suggestions for future glioma diagnosis and therapy improvement.
Collapse
|
11
|
Ren P, Wang JY, Zeng ZR, Li NX, Chen HL, Peng XG, Bhawal UK, Guo WZ. A novel hypoxia-driven gene signature that can predict the prognosis and drug resistance of gliomas. Front Genet 2022; 13:976356. [PMID: 36118887 PMCID: PMC9478203 DOI: 10.3389/fgene.2022.976356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Hypoxia spontaneously forms in the interior of glioma tissues and regulates the expression of various genes. However, the status of hypoxia-driven genes in glioma tissues is not completely known. In the current study, RNA-seq data of 695 glioma tissues in The Cancer Genome Atlas (TCGA) were set as a discovery cohort and were used to identify hypoxia-driven genes and construct a novel gene signature. The prognostic values of that signature were verified in data from the TCGA and the Chinese Glioma Genome Atlas (CGGA). The expression and diagnostic values of hypoxia-driven genes were analyzed using immunohistochemistry and receiver operator characteristic curves. Finally, the effects of hypoxia-driven genes on temozolomide (TMZ) resistance were analyzed by western blot, CCK-8 and colony formation assay. A total of 169 hypoxia-driven genes were identified, which were associated with a poor outcome in glioma patients. Among them, 22 genes had a degree score ≥10 and 6 genes (WT1, HOXA2, HOXC6, MMP9, SHOX2 and MYOD1) were selected to construct a signature to classify glioma patients into low- or high-risk groups. That signature had a remarkable prognostic value for glioma patients in TCGA and CGGA. The expression of HOXC6, MMP9, SHOX2 and MYOD1 was associated with hypoxia degree in glioma tissues and in recurrent cases, had a remarkable diagnostic value and a significant relationship with disease free survival in glioma patients. Moreover, SHOX2 was highly expressed in glioma tissues with O-6-methylguanine-DNA methyltransferase (MGMT)-unmethylation and temozolomide (TMZ) resistant glioma cell lines, and associated with MGMT expression. Knockdown the expression of SHOX2 significantly reduced the TMZ-resistance induced by hypoxia in glioma cells. Ultimately, we identified six novel hypoxia-driven genes for reliable prognostic prediction in gliomas and found that SHOX2 might be a potential target to overcome the TMZ resistance induced by hypoxia.
Collapse
Affiliation(s)
- Peng Ren
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jing-Ya Wang
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhi-Rui Zeng
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, China
| | - Nan-Xi Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hong-Lei Chen
- Hengyang Medical College, University of South China, Hengyang, China
| | - Xin-Ge Peng
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guizhou, China
| | - Ujjal K. Bhawal
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Chiba, Japan
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- *Correspondence: Ujjal K. Bhawal, ; Wen-Zhi Guo,
| | - Wen-Zhi Guo
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Ujjal K. Bhawal, ; Wen-Zhi Guo,
| |
Collapse
|
12
|
miR-138-5p Inhibits Vascular Mimicry by Targeting the HIF-1α/VEGFA Pathway in Hepatocellular Carcinoma. J Immunol Res 2022; 2022:7318950. [PMID: 35669101 PMCID: PMC9167126 DOI: 10.1155/2022/7318950] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 12/24/2022] Open
Abstract
Tumour vascular mimicry (VM) is the process by which new blood vessels are formed by tumour cells rather than endothelial cells. An increasing number of studies have revealed that the VM process is associated with cancer progression and metastasis. MiR-138-5p has been reported to act as a tumour suppressor in many cancers. However, the role and underlying mechanism of miR-138-5p in hepatocellular carcinoma (HCC) VM remain unclear. In this study, VM density was detected by CD31/periodic acid-Schiff double staining in HCC clinical specimens. We found that miR-138-5p expression correlated strongly and negatively with microvessel density. Additionally, the miR-138-5p mimic or inhibitor decreased or increased, respectively, tube formation capacity in HepG2 and Hep3B cells. Consistent with this finding, miR-138-5p repressed vessel density in vivo. Moreover, miR-138-5p targeted hypoxia-inducible factor 1α (HIF-1α) and regulated the expression of HIF-1α and vascular endothelial growth factor A (VEGFA), which are established classical master regulators for angiogenesis. Consistent with these findings, the HIF-1α inhibitor CAY10585 effectively blocked HCC cell VM and VEGFA expression. In conclusion, miR-138-5p inhibits HepG2 and Hep3B cell VM by blocking the HIF-1α/VEGFA pathway. Therefore, miR-138-5p may serve as a useful therapeutic target for miRNA-based HCC therapy.
Collapse
|
13
|
Domènech M, Hernández A, Plaja A, Martínez-Balibrea E, Balañà C. Hypoxia: The Cornerstone of Glioblastoma. Int J Mol Sci 2021; 22:12608. [PMID: 34830491 PMCID: PMC8620858 DOI: 10.3390/ijms222212608] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/19/2021] [Accepted: 11/19/2021] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma is the most aggressive form of brain tumor in adults and is characterized by the presence of hypervascularization and necrosis, both caused by a hypoxic microenvironment. In this review, we highlight that hypoxia-induced factor 1 (HIF-1), the main factor activated by hypoxia, is an important driver of tumor progression in GB patients. HIF-1α is a transcription factor regulated by the presence or absence of O2. The expression of HIF-1 has been related to high-grade gliomas and aggressive tumor behavior. HIF-1 promotes tumor progression via the activation of angiogenesis, immunosuppression, and metabolic reprogramming, promoting cell invasion and survival. Moreover, in GB, HIF-1 is not solely modulated by oxygen but also by oncogenic signaling pathways, such as MAPK/ERK, p53, and PI3K/PTEN. Therefore, the inhibition of the hypoxia pathway could represent an important treatment alternative in a disease with very few therapy options. Here, we review the roles of HIF-1 in GB progression and the inhibitors that have been studied thus far, with the aim of shedding light on this devastating disease.
Collapse
Affiliation(s)
- Marta Domènech
- B·ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, 08916 Badalona, Spain; (M.D.); (A.H.); (A.P.)
| | - Ainhoa Hernández
- B·ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, 08916 Badalona, Spain; (M.D.); (A.H.); (A.P.)
| | - Andrea Plaja
- B·ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, 08916 Badalona, Spain; (M.D.); (A.H.); (A.P.)
| | - Eva Martínez-Balibrea
- Germans Trias i Pujol Research Institute (IGTP), ProCURE Program, Catalan Institute of Oncology, 08916 Badalona, Spain;
| | - Carmen Balañà
- B·ARGO (Badalona Applied Research Group of Oncology) Medical Oncology Department, Catalan Institute of Oncology Badalona, 08916 Badalona, Spain; (M.D.); (A.H.); (A.P.)
| |
Collapse
|
14
|
Kayabolen A, Yilmaz E, Bagci-Onder T. IDH Mutations in Glioma: Double-Edged Sword in Clinical Applications? Biomedicines 2021; 9:799. [PMID: 34356864 PMCID: PMC8301439 DOI: 10.3390/biomedicines9070799] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 01/03/2023] Open
Abstract
Discovery of point mutations in the genes encoding isocitrate dehydrogenases (IDH) in gliomas about a decade ago has challenged our view of the role of metabolism in tumor progression and provided a new stratification strategy for malignant gliomas. IDH enzymes catalyze the conversion of isocitrate to alpha-ketoglutarate (α-KG), an intermediate in the citric acid cycle. Specific mutations in the genes encoding IDHs cause neomorphic enzymatic activity that produces D-2-hydroxyglutarate (2-HG) and result in the inhibition of α-KG-dependent enzymes such as histone and DNA demethylases. Thus, chromatin structure and gene expression profiles in IDH-mutant gliomas appear to be different from those in IDH-wildtype gliomas. IDH mutations are highly common in lower grade gliomas (LGG) and secondary glioblastomas, and they are among the earliest genetic events driving tumorigenesis. Therefore, inhibition of mutant IDH enzymes in LGGs is widely accepted as an attractive therapeutic strategy. On the other hand, the metabolic consequences derived from IDH mutations lead to selective vulnerabilities within tumor cells, making them more sensitive to several therapeutic interventions. Therefore, instead of shutting down mutant IDH enzymes, exploiting the selective vulnerabilities caused by them might be another attractive and promising strategy. Here, we review therapeutic options and summarize current preclinical and clinical studies on IDH-mutant gliomas.
Collapse
Affiliation(s)
- Alisan Kayabolen
- Brain Cancer Research and Therapy Lab, Koç University School of Medicine, 34450 Istanbul, Turkey; (A.K.); (E.Y.)
- Koç University Research Center for Translational Medicine (KUTTAM), 34450 Istanbul, Turkey
| | - Ebru Yilmaz
- Brain Cancer Research and Therapy Lab, Koç University School of Medicine, 34450 Istanbul, Turkey; (A.K.); (E.Y.)
- Koç University Research Center for Translational Medicine (KUTTAM), 34450 Istanbul, Turkey
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Lab, Koç University School of Medicine, 34450 Istanbul, Turkey; (A.K.); (E.Y.)
- Koç University Research Center for Translational Medicine (KUTTAM), 34450 Istanbul, Turkey
| |
Collapse
|
15
|
Mikkelsen VE, Dai HY, Stensjøen AL, Berntsen EM, Salvesen Ø, Solheim O, Torp SH. MGMT Promoter Methylation Status Is Not Related to Histological or Radiological Features in IDH Wild-type Glioblastomas. J Neuropathol Exp Neurol 2021; 79:855-862. [PMID: 32688383 DOI: 10.1093/jnen/nlaa060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/25/2020] [Accepted: 06/03/2020] [Indexed: 11/15/2022] Open
Abstract
O6-methylguanine DNA methyltransferase (MGMT) promoter methylation is an important favorable predictive marker in patients with glioblastoma (GBM). We hypothesized that MGMT status could be a surrogate marker of pretreatment tumor biology observed as histopathological and radiological features. Apart from some radiological studies aiming to noninvasively predict the MGMT status, few studies have investigated relationships between MGMT status and phenotypical tumor biology. We have therefore aimed to investigate such relationships in 85 isocitrate dehydrogenase (IDH) wild-type GBMs. MGMT status was determined by methylation-specific PCR and was assessed for associations with 22 histopathological features, immunohistochemical proliferative index and microvessel density measurements, conventional magnetic resonance imaging characteristics, preoperative speed of tumor growth, and overall survival. None of the investigated histological or radiological features were significantly associated with MGMT status. Methylated MGMT status was a significant independent predictor of improved overall survival. In conclusion, our results suggest that MGMT status is not related to the pretreatment phenotypical biology in IDH wild-type GBMs. Furthermore, our findings suggest the survival benefit of MGMT methylated GBMs is not due to an inherently less aggressive tumor biology, and that conventional magnetic resonance imaging features cannot be used to noninvasively predict the MGMT status.
Collapse
Affiliation(s)
- Vilde Elisabeth Mikkelsen
- From the Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology
| | - Hong Yan Dai
- Department of Pathology, St Olav's University Hospital
| | - Anne Line Stensjøen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology
| | - Erik Magnus Berntsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology.,Department of Radiology and Nuclear Medicine, St. Olav's University Hospital
| | | | - Ole Solheim
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology.,Department of Neurosurgery, St. Olav's University Hospital, Trondheim, Norway
| | - Sverre Helge Torp
- From the Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology.,Department of Pathology, St Olav's University Hospital
| |
Collapse
|
16
|
The HIF1α/HIF2α-miR210-3p network regulates glioblastoma cell proliferation, dedifferentiation and chemoresistance through EGF under hypoxic conditions. Cell Death Dis 2020; 11:992. [PMID: 33208727 PMCID: PMC7674439 DOI: 10.1038/s41419-020-03150-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
Hypoxia-inducible factor 1α (HIF1α) promotes the malignant progression of glioblastoma under hypoxic conditions, leading to a poor prognosis for patients with glioblastoma; however, none of the therapies targeting HIF1α in glioblastoma have successfully eradicated the tumour. Therefore, we focused on the reason and found that treatments targeting HIF1α and HIF2α simultaneously increased tumour volume, but the combination of HIF1α/HIF2α-targeted therapies with temozolomide (TMZ) reduced tumourigenesis and significantly improved chemosensitization. Moreover, miR-210-3p induced HIF1α expression but inhibited HIF2α expression, suggesting that miR-210-3p regulates HIF1α/HIF2α expression. Epidermal growth factor (EGF) has been shown to upregulate HIF1α expression under hypoxic conditions. However, in the present study, in addition to the signalling pathways mentioned above, the upstream proteins HIF1α and HIF2α have been shown to induce EGF expression by binding to the sequences AGGCGTGG and GGGCGTGG. Briefly, in a hypoxic microenvironment the HIF1α/HIF2α-miR210-3p network promotes the malignant progression of glioblastoma through a positive feedback loop with EGF. Additionally, differentiated glioblastoma cells underwent dedifferentiation to produce glioma stem cells under hypoxic conditions, and simultaneous knockout of HIF1α and HIF2α inhibited cell cycle arrest but promoted proliferation with decreased stemness, promoting glioblastoma cell chemosensitization. In summary, both HIF1α and HIF2α regulate glioblastoma cell proliferation, dedifferentiation and chemoresistance through a specific pathway, which is important for glioblastoma treatments.
Collapse
|
17
|
Jnaidi R, Almeida AJ, Gonçalves LM. Solid Lipid Nanoparticles and Nanostructured Lipid Carriers as Smart Drug Delivery Systems in the Treatment of Glioblastoma Multiforme. Pharmaceutics 2020; 12:E860. [PMID: 32927610 PMCID: PMC7558650 DOI: 10.3390/pharmaceutics12090860] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant type of brain tumor. In fact, tumor recurrence usually appears a few months after surgical resection and chemotherapy, mainly due to many factors that make GBM treatment a real challenge, such as tumor location, heterogeneity, presence of the blood-brain barrier (BBB), and others. Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) represent the most promising carriers for therapeutics delivery into the central nervous system (CNS) owing to their inherent ability to cross the BBB. In this review, we present the main challenges in GBM treatment, a description of SLNs and NLCs and their valuable role as drug carriers in GBM treatment, and finally, a detailed description of all modification strategies that aim to change composition of SLNs and NLCs to enhance treatment outcomes. This includes modification of SLNs and NLCs to improve crossing the BBB, reduced GBM cell resistance, target GBM cells selectively minimizing side effects, and modification strategies to enhance SLNs and NLCs nose-to-brain delivery. Finally, future perspectives on their use are also be discussed, to provide insight about all strategies with SLNs and NLCs formulation that could result in drug delivery systems for GBM treatment with highly effective theraputic and minimum undesirable effects.
Collapse
Affiliation(s)
| | | | - Lídia M. Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (R.J.); (A.J.A.)
| |
Collapse
|
18
|
Huang GH, Pei YC, Yang L, Mou KJ, Tang JH, Xiang Y, Liu J, Lv SQ. Integrative transcriptome analysis identified a BMP signaling pathway-regulated lncRNA AC068643.1 in IDH mutant and wild-type glioblastomas. Oncol Lett 2020; 20:75-84. [PMID: 32565936 PMCID: PMC7285920 DOI: 10.3892/ol.2020.11542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/12/2019] [Indexed: 11/05/2022] Open
Abstract
Glioblastomas (GBMs) are classified into isocitrate dehydrogenase (IDH) mutant (IDH MT) and wild-type (IDH WT) subtypes, and each is associated with distinct tumor behavior and prognosis. The present study aimed to investigate differentially expressed long non-coding (lnc)RNAs and mRNAs between IDH MT and IDH WT GBMs, as well as to explore the interaction and potential functions of these RNAs. A total of 132 GBM samples with RNA profiling data (10 IDH MT and 122 IDH WT cases) were obtained from The Cancer Genome Atlas, and 62/78 and 142/219 up/downregulated lncRNAs and mRNAs between IDH MT and IDH WT GBMs were identified, respectively. Multivariate Cox analysis of the dysregulated lncRNAs/mRNAs identified three-lncRNA and fifteen-mRNA signatures with independent prognostic value, indicating that these RNAs may serve roles in determining distinct tumor behaviors and prognosis of patients with IDH MT/WT GBMs. Functional analysis of the three lncRNAs revealed that they were primarily associated with cell stemness or differentiation. Pearson's correlation analysis revealed that the protective lncRNA AC068643.1 was significantly positively correlated with two key bone morphogenetic protein (BMP) signaling-associated mRNAs, Bone morphogenetic protein 2 (BMP2) and Myostatin (MSTN), from the 15 mRNAs. Further in vitro studies demonstrated that BMP2 and MSTN directly stimulated AC068643.1 expression. In conclusion, the present study identified a BMP signaling pathway-regulated lncRNA AC068643.1, which may contribute to the different tumor behaviors observed between IDH MT and IDH WT GBMs.
Collapse
Affiliation(s)
- Guo-Hao Huang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yu-Chun Pei
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Lin Yang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Ke-Jie Mou
- Department of Neurosurgery, Bishan Hospital, Chongqing Medical University, Chongqing 402760, P.R. China
| | - Jun-Hai Tang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yan Xiang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Jun Liu
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
19
|
Wang S, Chen C, Li J, Xu X, Chen W, Li F. The CXCL12/CXCR4 axis confers temozolomide resistance to human glioblastoma cells via up-regulation of FOXM1. J Neurol Sci 2020; 414:116837. [PMID: 32334273 DOI: 10.1016/j.jns.2020.116837] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 11/18/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignancy in the adult central nervous, and is characterized by high aggressiveness and a high mortality rate. The high mortality rate is largely due to the development of drug resistance. Temozolomide (TMZ) resistance is considered to be one of the major reasons responsible for GBM therapy failure. CXCL12/CXCR4 has been demonstrated to be involved in cell proliferation, migration, invasion, angiogenesis, and radioresistance in GBM. However, its role in TMZ resistance in GBM is unknown. In this study, we aimed to evaluate the role of CXCL12/CXCR4 in mediating the TMZ resistance to GBM cells and explore the underlying mechanisms. We found that the CXCL12/CXCR4 axis enhanced TMZ resistance in GBM cells. Further study showed that CXCL12/CXCR4 conferred TMZ resistance and promoted the migration and invasion of GBM cells by up-regulating FOXM1. This resistance was partially reversed by suppressing CXCL12/CXCR4 and FOXM1 silencing. Our study revealed the vital role of CXCL12/CXCR4 in mediating the resistance of GBM cells to TMZ, and suggested that targeting CXCL12/CXCR4 axis may attenuate the resistance to TMZ in GBM.
Collapse
Affiliation(s)
- Shengwen Wang
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Cheng Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Junliang Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Xinke Xu
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Wei Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Fangcheng Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China.
| |
Collapse
|
20
|
Li X, Zeng Z, Wang J, Wu Y, Chen W, Zheng L, Xi T, Wang A, Lu Y. MicroRNA-9 and breast cancer. Biomed Pharmacother 2020; 122:109687. [PMID: 31918267 DOI: 10.1016/j.biopha.2019.109687] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/14/2019] [Accepted: 11/16/2019] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is the most common cancer in women worldwide and seriously impairs patients' physical and mental health. Its incidence has been predicted to rise further. Mounting evidence indicates that microRNAs (miRNAs) play key roles in tumorigenesis and development. It is worth noting that miR-9 exerts critical functions in the initiation and progression of breast cancer, and the present research displays opposite roles of miR-9 in breast cancer. This article mainly reviews the roles of miR-9 in breast cancer progression.
Collapse
Affiliation(s)
- Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhu Zeng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiaer Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuanyuan Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Xi
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 210009, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
21
|
Sharma HS, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Tian ZR, Sahib S, Bryukhovetskiy I, Bryukhovetskiy A, Buzoianu AD, Patnaik R, Wiklund L, Sharma A. Pathophysiology of blood-brain barrier in brain tumor. Novel therapeutic advances using nanomedicine. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 151:1-66. [PMID: 32448602 DOI: 10.1016/bs.irn.2020.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Hu Q, Liu F, Yan T, Wu M, Ye M, Shi G, Lv S, Zhu X. MicroRNA‑576‑3p inhibits the migration and proangiogenic abilities of hypoxia‑treated glioma cells through hypoxia‑inducible factor‑1α. Int J Mol Med 2019; 43:2387-2397. [PMID: 31017266 PMCID: PMC6488173 DOI: 10.3892/ijmm.2019.4157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 03/26/2019] [Indexed: 12/24/2022] Open
Abstract
The most common and aggressive type of brain cancer in adults is glioblastoma multiforme (GBM), and hypoxia is a common feature of glioblastoma. As the histological features of glioma include capillary endothelial cell proliferation, they are highly prone to invading the surrounding normal brain tissue, which is often one of the reasons for the failure of treatment. However, the mechanisms involved in this process are not fully understood. MicroRNAs (miRs) are a class of non‑coding RNA that are able to inhibit the malignant progression of tumor cells through the regulation of downstream genes. In the present study, the low expression of miR‑576‑3p was detected in glioma samples and hypoxia‑treated glioma cells using a reverse transcription‑quantitative polymerase chain reaction. The present study focused on the effects of miR‑576‑3p on hypoxia‑induced glioma. The results of the functional experiments revealed that the overexpression of miR‑576‑3p significantly inhibited the migration and pro‑angiogenic abilities of the glioma cells under hypoxic conditions (P<0.05) compared with in the lentivirus‑miR‑negative control group. Furthermore, luciferase reporter gene assays were used to validate the hypothesis that miR‑576‑3p interacts with the 3'‑untranslated region of hypoxia‑inducible factor‑1α (HIF‑1α) and induces a reduction in the protein levels of matrix metalloproteinase‑2 and vascular endothelial growth factor. Rescue experiments demonstrated that the restoration of HIF‑1α expression attenuated the effect of miR‑576‑3p on the migration and proangiogenic abilities of glioma cells. In conclusion, the present study confirms that miR‑576‑3p is a novel GBM inhibitor and its inhibition of the migration and proangiogenic capacity of hypoxia‑induced glioma cells is mediated by HIF‑1α.
Collapse
Affiliation(s)
- Qing Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Feng Liu
- Department of Neurosurgery, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi 330006
| | - Tengfeng Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Miaojing Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Minhua Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Guangyao Shi
- Queen Mary School, Medical College, Nanchang University School of Medicine, Nanchang, Jiangxi 330031, P.R. China
| | - Shigang Lv
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University
| |
Collapse
|
23
|
Towner RA, Smith N, Saunders D, Brown CA, Cai X, Ziegler J, Mallory S, Dozmorov MG, Coutinho De Souza P, Wiley G, Kim K, Kang S, Kong DS, Kim YT, Fung KM, Wren JD, Battiste J. OKN-007 Increases temozolomide (TMZ) Sensitivity and Suppresses TMZ-Resistant Glioblastoma (GBM) Tumor Growth. Transl Oncol 2019; 12:320-335. [PMID: 30468988 PMCID: PMC6251232 DOI: 10.1016/j.tranon.2018.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 02/06/2023] Open
Abstract
Treatment of glioblastoma (GBM) remains a challenge using conventional chemotherapy, such as temozolomide (TMZ), and is often ineffective as a result of drug resistance. We have assessed a novel nitrone-based agent, OKN-007, and found it to be effective in decreasing tumor volumes and increasing survival in orthotopic GBM xenografts by decreasing cell proliferation and angiogenesis and increasing apoptosis. In this study, we assessed combining OKN-007 with TMZ in vivo in a human G55 GBM orthotopic xenograft model and in vitro in TMZ-resistant and TMZ-sensitive human GBM cell lines. For the in vivo studies, magnetic resonance imaging was used to assess tumor growth and vascular alterations. Percent animal survival was also determined. For the in vitro studies, cell growth, IC50 values, RNA-seq, RT-PCR, and ELISA were used to assess growth inhibition, possible mechanism-of actions (MOAs) associated with combined OKN-007 + TMZ versus TMZ alone, and gene and protein expression levels, respectively. Microarray analysis of OKN-007-treated rat F98 glioma tumors was also carried out to determine possible MOAs of OKN-007 in glioma-bearing animals either treated or not treated with OKN-007. OKN-007 seems to elicit its effect on GBM tumors via inhibition of tumorigenic TGF-β1, which affects the extracellular matrix. When combined with TMZ, OKN-007 significantly increases percent survival, decreases tumor volumes, and normalizes tumor blood vasculature in vivo compared to untreated tumors and seems to affect TMZ-resistant GBM cells possibly via IDO-1, SUMO2, and PFN1 in vitro. Combined OKN-007 + TMZ may be a potentially potent treatment strategy for GBM patients.
Collapse
Affiliation(s)
- Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Chase A Brown
- Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xue Cai
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jadith Ziegler
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Graham Wiley
- Clinical Genomics Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kyeongsoon Kim
- Department of Pharmaceutical Engineering, Inje University, Gimhae-si, Gyeongsangnam-do, Republic of Korea; Oblato, Inc., Princeton, NJ, USA
| | | | - Doo-Sik Kong
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Young-Tae Kim
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Kar-Ming Fung
- Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jonathan D Wren
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - James Battiste
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of NeurologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
24
|
Navone SE, Guarnaccia L, Cordiglieri C, Crisà FM, Caroli M, Locatelli M, Schisano L, Rampini P, Miozzo M, La Verde N, Riboni L, Campanella R, Marfia G. Aspirin Affects Tumor Angiogenesis and Sensitizes Human Glioblastoma Endothelial Cells to Temozolomide, Bevacizumab, and Sunitinib, Impairing Vascular Endothelial Growth Factor-Related Signaling. World Neurosurg 2018; 120:e380-e391. [DOI: 10.1016/j.wneu.2018.08.080] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/10/2018] [Accepted: 08/11/2018] [Indexed: 12/16/2022]
|
25
|
Guan X, Hasan MN, Begum G, Kohanbash G, Carney KE, Pigott VM, Persson AI, Castro MG, Jia W, Sun D. Blockade of Na/H exchanger stimulates glioma tumor immunogenicity and enhances combinatorial TMZ and anti-PD-1 therapy. Cell Death Dis 2018; 9:1010. [PMID: 30262908 PMCID: PMC6160445 DOI: 10.1038/s41419-018-1062-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 12/28/2022]
Abstract
The weak immunogenicity of gliomas presents a barrier for effective immunotherapy. Na/H exchanger isoform 1 (NHE1) maintains alkaline intracellular pH (pHi) of glioma cells and acidic microenvironment. In addition, NHE1 is expressed in tumor-associated microglia and tumor-associated macrophages (TAMs) and involved in protumoral communications between glioma and TAMs. Therefore, we hypothesize that NHE1 plays a role in developing tumor resistance and immunosuppressive tumor microenvironment. In this study, we investigated the efficacy of pharmacological inhibition of NHE1 on combinatorial therapies. Here we show that temozolomide (TMZ) treatment stimulates NHE1 protein expression in two intracranial syngeneic mouse glioma models (SB28, GL26). Pharmacological inhibition of NHE1 potentiated the cytotoxic effects of TMZ, leading to reduced tumor growth and increased median survival of mice. Blockade of NHE1 stimulated proinflammatory activation of TAM and increased cytotoxic T cell infiltration into tumors. Combining TMZ, anti-PD-1 antibody treatment with NHE1 blockade significantly prolonged the median survival in the mouse glioma model. These results demonstrate that pharmacological inhibition of NHE1 protein presents a new strategy for potentiating TMZ-induced cytotoxicity and increasing tumor immunogenicity for immunotherapy to improve glioma therapy.
Collapse
Affiliation(s)
- Xiudong Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Chinese National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Md Nabiul Hasan
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karen E Carney
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victoria M Pigott
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anders I Persson
- Department of Neurology, University of California, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Maria G Castro
- Department of Neurological Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Chinese National Clinical Research Center for Neurological Diseases, Beijing, China.
- Beijing Neurosurgical Institute, Beijing, China.
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
26
|
Methylation-mediated miR-155-FAM133A axis contributes to the attenuated invasion and migration of IDH mutant gliomas. Cancer Lett 2018; 432:93-102. [DOI: 10.1016/j.canlet.2018.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/30/2018] [Accepted: 06/01/2018] [Indexed: 12/26/2022]
|
27
|
Lo Dico A, Martelli C, Diceglie C, Lucignani G, Ottobrini L. Hypoxia-Inducible Factor-1α Activity as a Switch for Glioblastoma Responsiveness to Temozolomide. Front Oncol 2018; 8:249. [PMID: 30013951 PMCID: PMC6036118 DOI: 10.3389/fonc.2018.00249] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/19/2018] [Indexed: 11/13/2022] Open
Abstract
Rationale The activity of the transcription factor, hypoxia-inducible factor (HIF)-1α, is a common driver of a number of the pathways involved in the aggressiveness of glioblastomas (GBMs), and it has been suggested that the reduction in this activity observed, soon after the administration of temozolomide (TMZ), can be a biomarker of an early response in GBM models. As HIF-1α is a tightly regulated protein, studying the processes involved in its downregulation could shed new light on the mechanisms underlying GBM sensitivity or resistance to TMZ. Methods The effect of HIF-1α silencing on cell responsiveness to TMZ was assessed in four genetically different human GBM cell lines by evaluating cell viability and apoptosis-related gene balance. LAMP-2A silencing was used to evaluate the contribution of chaperone-mediated autophagy (CMA) to the modulation of HIF-1α activity in TMZ-sensitive and TMZ-resistant cells. Results The results showed that HIF-1α but not HIF-2α activity is associated with GBM responsiveness to TMZ: its downregulation improves the response of TMZ-resistant cells, while blocking CMA-mediated HIF-1α degradation induces resistance to TMZ in TMZ-sensitive cells. These findings are in line with the modulation of crucial apoptosis-related genes. Conclusion Our results demonstrate the central role played by HIF-1α activity in determining the sensitivity or resistance of GBMs to TMZ, and we suggest that CMA is the cellular mechanism responsible for modulating this activity after TMZ treatment.
Collapse
Affiliation(s)
- Alessia Lo Dico
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Cristina Martelli
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Cecilia Diceglie
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giovanni Lucignani
- Department of Health Sciences, University of Milan, Milan, Italy.,Department of Diagnostic Services, Unit of Nuclear Medicine, San Paolo Hospital, Milan, Italy
| | - Luisa Ottobrini
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
28
|
You CG, Sheng HS, Xie CR, Zhang N, Zheng XS. FM19G11 inhibits O 6 -methylguanine DNA-methyltransferase expression under both hypoxic and normoxic conditions. Cancer Med 2018; 7:3292-3300. [PMID: 29761922 PMCID: PMC6051152 DOI: 10.1002/cam4.1551] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/14/2018] [Accepted: 04/19/2018] [Indexed: 12/20/2022] Open
Abstract
FM19G11 is a small molecular agent that inhibits hypoxia-inducible factor-1-alpha (HIF-1α) and other signaling pathways. In this study, we characterized the modulating effects of FM19G11 on O6 -methylguanine DNA-methyltransferase (MGMT), the main regulator of temozolomide (TMZ) resistance in glioblastomas. This study included 2 MGMT-positive cell lines (GBM-XD and T98G). MGMT promoter methylation status, mRNA abundance, and protein levels were determined before and after FM19G11 treatment, and the roles of various signaling pathways were characterized. Under hypoxic conditions, MGMT mRNA and protein levels were significantly downregulated by FM19G11 via the HIF-1α pathway in both GBM-XD and T98G cells. In normoxic culture, T98G cells were strongly positive for MGMT, and MGMT expression was substantially downregulated by FM19G11 via the NF-κB pathway. In addition, TMZ resistance was reversed by treatment with FM19G11. Meanwhile, FM19G11 has no cytotoxicity at its effective dose. FM19G11 could potentially be used to counteract TMZ resistance in MGMT-positive glioblastomas.
Collapse
Affiliation(s)
- Chao-Guo You
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China.,Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Han-Song Sheng
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chao-Ran Xie
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China.,Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Nu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xue-Sheng Zheng
- Department of Neurosurgery, Xinhua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Xu Q, Ahmed AK, Zhu Y, Wang K, Lv S, Li Y, Jiang Y. Oncogenic MicroRNA-20a is downregulated by the HIF-1α/c-MYC pathway in IDH1 R132H-mutant glioma. Biochem Biophys Res Commun 2018; 499:882-888. [PMID: 29625108 DOI: 10.1016/j.bbrc.2018.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 12/13/2022]
Abstract
Mutations in the isocitrate dehydrogenase 1 (IDH1) gene have been identified as one of the earliest events in gliomagenesis, occurring in over 70% of low grade gliomas and are present in the vast majority of secondary glioblastoma (GBM) that develop from these low-grade lesions. The aim of this study was to investigate whether the IDH1 R132H mutation influences the expression of oncogenic miR-20a and shed light on the underlying molecular mechanisms. The findings of the current study demonstrate presence of the IDH1 R132H mutation in primary human glioblastoma cell lines with upregulated HIF-1α expression, downregulating c-MYC activity and resulting in a consequential decrease in miR-20a, which is responsible for cell proliferation and resistance to standard temozolomide treatment. Elucidating the mechanism of oncogenic miR-20a activity introduces its role among well-established signaling pathways (i.e. HIF/c-MYC) and may be a meaningful prognostic biomarker or target for novel therapies among patients with IDH1-mutant glioma.
Collapse
Affiliation(s)
- Qingfu Xu
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - A Karim Ahmed
- Department of Neurosurgery, The Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Yan Zhu
- Department of Obstetrics and Gynecology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Kimberly Wang
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA
| | - Shengqing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yunqing Li
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
30
|
Hombach-Klonisch S, Mehrpour M, Shojaei S, Harlos C, Pitz M, Hamai A, Siemianowicz K, Likus W, Wiechec E, Toyota BD, Hoshyar R, Seyfoori A, Sepehri Z, Ande SR, Khadem F, Akbari M, Gorman AM, Samali A, Klonisch T, Ghavami S. Glioblastoma and chemoresistance to alkylating agents: Involvement of apoptosis, autophagy, and unfolded protein response. Pharmacol Ther 2018; 184:13-41. [DOI: 10.1016/j.pharmthera.2017.10.017] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
31
|
Li B, Zhou C, Yi L, Xu L, Xu M. Effect and molecular mechanism of mTOR inhibitor rapamycin on temozolomide-induced autophagic death of U251 glioma cells. Oncol Lett 2017; 15:2477-2484. [PMID: 29434961 DOI: 10.3892/ol.2017.7537] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 11/08/2016] [Indexed: 01/10/2023] Open
Abstract
Glioma is a malignant tumor of the glial tissue that is difficult to excise through surgery, with poor patient prognosis. The use of chemotherapeutic drugs alone to treat glioma following surgery results in a high probability of sequelae, such as tumor recurrence. The present study investigated the effects of a novel treatment combination on glioma cells and determined the molecular mechanisms underlying its action. The effect of temozolomide (TMZ) combined with rapamycin (RAPA) on the TMZ-induced autophagic death of U251 glioma cells was examined. The U251 cell line was treated with TMZ combined with RAPA, and the cell survival rate and half maximal inhibitory concentration (IC50) of TMZ/RAPA was detected using the Cell Counting Kit-8 (CCK-8) assay. Flow cytometry was used to detect changes in cell cycle distribution. The formation of acidic vesicular organelles (AVOs) in the cytoplasm was identified using fluorescence microscopy and quantitatively analyzed. Western blotting was performed to detect the expression levels of autophagy-associated proteins Beclin-1 and microtubule associated protein 1 light chain 3 alpha (MAP1LC3A)-I and II. RAPA (1.25 nM) combined with 5 µM TMZ markedly inhibited U251 cell growth. RAPA reinforced TMZ-induced autophagic death, reducing the IC50 value of treatment when combined (TMZ alone, 22.5±3.23 µM vs. TMZ and RAPA, 10.35±2.81 µM). Compared with the control group, the proportion of cells in G2/M were markedly increased following treatment with TMZ combined with RAPA. Acridine orange staining demonstrated that TMZ combined with RAPA could markedly enhance the generation of intracellular AVOs compared with TMZ or RAPA alone. In addition, Beclin-1 and LC3-II protein expression was markedly increased compared with the control and single treatment groups (P<0.05). The results of the present study indicate that RAPA reinforces TMZ-induced autophagic death of U251 glioma cells, providing a novel therapeutic combination for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Bing Li
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Chun Zhou
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Liang Yi
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Lunshan Xu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Minhui Xu
- Department of Neurosurgery, Daping Hospital, Third Military Medical University, Yuzhong, Chongqing 400042, P.R. China
| |
Collapse
|
32
|
Du L, Tang JH, Huang GH, Xiang Y, Lv SQ. The progression of epithelial-mesenchymal transformation in gliomas. Chin Neurosurg J 2017. [DOI: 10.1186/s41016-017-0086-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
33
|
Zhu D, Tu M, Zeng B, Cai L, Zheng W, Su Z, Yu Z. Up-regulation of miR-497 confers resistance to temozolomide in human glioma cells by targeting mTOR/Bcl-2. Cancer Med 2017; 6:452-462. [PMID: 28064447 PMCID: PMC5313645 DOI: 10.1002/cam4.987] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/27/2016] [Accepted: 11/07/2016] [Indexed: 12/21/2022] Open
Abstract
The occurrence of an inherent or acquired resistance to temozolomide (TMZ) is a major burden for patients suffering from glioma. Recently, studies have demonstrated that microRNAs play an important role in the regulation of tumor properties in cancers. However, whether miR‐497 contributes to glioma resistance to chemotherapy is not fully understood. In this study, we showed that the expression of miR‐497 was markedly up‐regulated in TMZ‐resistant glioma cells; high miR‐497 expression level was associated with TMZ‐resistant phenotype of glioma cells. The down‐regulation of miR‐497 in glioma cells enhanced the apoptosis‐induction and growth inhibition effects of TMZ both in vitro and in vivo, whereas promotion of miR‐497 increased the chemosensitization of glioma cells to TMZ. The increased level of miR‐497 in TMZ‐resistant glioma cells was concurrent with the up‐regulation of insulin‐like growth factor 1 receptor (IGF1R)/insulin receptor substrate 1 (IRS1) pathway‐related proteins, that is, IGF1R, IRS1, mammalian target of rapamycin (mTOR), and Bcl‐2. In addition, the knockdown of mTOR and Bcl‐2 reduced the tolerance of glioma cells to TMZ. Our results demonstrated that overexpression of miR‐497 is significantly correlated with TMZ resistance in glioma cells by regulating the IGF1R/IRS1 pathway. Therefore, miR‐497 may be used as a new target for treatment of chemotherapy‐resistant glioma.
Collapse
Affiliation(s)
- Danhua Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Canglang District, Suzhou, Jiangsu, 215000, China.,Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ming Tu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Bo Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Lin Cai
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Weiming Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zhipeng Su
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zhengquan Yu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Canglang District, Suzhou, Jiangsu, 215000, China
| |
Collapse
|
34
|
Wang G, Wang JJ, Fu XL, Guang R, To SST. Advances in the targeting of HIF-1α and future therapeutic strategies for glioblastoma multiforme (Review). Oncol Rep 2016; 37:657-670. [PMID: 27959421 DOI: 10.3892/or.2016.5309] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 09/09/2016] [Indexed: 11/06/2022] Open
Abstract
Cell metabolism can be reprogrammed by tissue hypoxia leading to cell transformation and glioblastoma multiforme (GBM) progression. In response to hypoxia, GBM cells are able to express a transcription factor called hypoxia inducible factor-1 (HIF-1). HIF-1 belongs to a family of heterodimeric proteins that includes HIF-1α and HIF-1β subunits. HIF-1α has been reported to play a pivotal role in GBM development and progression. In the present review, we discuss the role of HIF-1α in glucose uptake, cancer proliferation, cell mobility and chemoresistance in GBM. Evidence from previous studies indicates that HIF-1α regulates angiogenesis, metabolic and transcriptional signaling pathways. Examples of such are the EGFR, PI3K/Akt and MAPK/ERK pathways. It affects cell migration and invasion by regulating glucose metabolism and growth in GBM cells. The present review focuses on the strategies through which to target HIF-1α and the related downstream genes highlighting their regulatory roles in angiogenesis, apoptosis, migration and glucose metabolism for the development of future GBM therapeutics. Combined treatment with inhibitors of HIF-1α and glycolysis may enhance antitumor effects in clinical settings.
Collapse
Affiliation(s)
- Gang Wang
- Department of Hospital Pharmacy, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai 200235, P.R. China
| | - Jun-Jie Wang
- Department of Hospital Pharmacy, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai 200235, P.R. China
| | - Xing-Li Fu
- Department of Hospital Pharmacy, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai 200235, P.R. China
| | - Rui Guang
- Department of Hospital Pharmacy, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai 200235, P.R. China
| | - Shing-Shun Tony To
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Kowloon Hong Kong, SAR, P.R. China
| |
Collapse
|