1
|
Pun S, Prakash A, Demaree D, Krummel DP, Sciumè G, Sengupta S, Barrile R. Rapid Biofabrication of an Advanced Microphysiological System Mimicking Phenotypical Heterogeneity and Drug Resistance in Glioblastoma. Adv Healthc Mater 2024:e2401876. [PMID: 39101329 DOI: 10.1002/adhm.202401876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/10/2024] [Indexed: 08/06/2024]
Abstract
Microphysiological systems (MPSs) reconstitute tissue interfaces and organ functions, presenting a promising alternative to animal models in drug development. However, traditional materials like polydimethylsiloxane (PDMS) often interfere by absorbing hydrophobic molecules, affecting drug testing accuracy. Additive manufacturing, including 3D bioprinting, offers viable solutions. GlioFlow3D, a novel microfluidic platform combining extrusion bioprinting and stereolithography (SLA) is introduced. GlioFlow3D integrates primary human cells and glioblastoma (GBM) lines in hydrogel-based microchannels mimicking vasculature, within an SLA resin framework using cost-effective materials. The study introduces a robust protocol to mitigate SLA resin cytotoxicity. Compared to PDMS, GlioFlow3D demonstrated lower small molecule absorption, which is relevant for accurate testing of small molecules like Temozolomide (TMZ). Computational modeling is used to optimize a pumpless setup simulating interstitial fluid flow dynamics in tissues. Co-culturing GBM with brain endothelial cells in GlioFlow3D showed enhanced CD133 expression and TMZ resistance near vascular interfaces, highlighting spatial drug resistance mechanisms. This PDMS-free platform promises advanced drug testing, improving preclinical research and personalized therapy by elucidating complex GBM drug resistance mechanisms influenced by the tissue microenvironment.
Collapse
Affiliation(s)
- Sirjana Pun
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Anusha Prakash
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
- Abbvie, Worcester, Massachusetts, 01605, USA
| | - Dalee Demaree
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
- Thermo Fisher Scientific, Waltham, Massachusetts, 02451, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology, University of Cincinnati, Cincinnati, OH, 45219, USA
- Department of Neurosurgery, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Giuseppe Sciumè
- Institute of Mechanics and Engineering-12 M, University of Bordeaux, Bordeaux, 33607, France
| | - Soma Sengupta
- Department of Neurology, University of Cincinnati, Cincinnati, OH, 45219, USA
- Department of Neurosurgery, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, 27599-7025, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599-7295, USA
| | - Riccardo Barrile
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, 45221, USA
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| |
Collapse
|
2
|
Ahmed T. Biomaterial-based in vitro 3D modeling of glioblastoma multiforme. CANCER PATHOGENESIS AND THERAPY 2023; 1:177-194. [PMID: 38327839 PMCID: PMC10846340 DOI: 10.1016/j.cpt.2023.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/24/2022] [Accepted: 01/04/2023] [Indexed: 02/09/2024]
Abstract
Adult-onset brain cancers, such as glioblastomas, are particularly lethal. People with glioblastoma multiforme (GBM) do not anticipate living for more than 15 months if there is no cure. The results of conventional treatments over the past 20 years have been underwhelming. Tumor aggressiveness, location, and lack of systemic therapies that can penetrate the blood-brain barrier are all contributing factors. For GBM treatments that appear promising in preclinical studies, there is a considerable rate of failure in phase I and II clinical trials. Unfortunately, access becomes impossible due to the intricate architecture of tumors. In vitro, bioengineered cancer models are currently being used by researchers to study disease development, test novel therapies, and advance specialized medications. Many different techniques for creating in vitro systems have arisen over the past few decades due to developments in cellular and tissue engineering. Later-stage research may yield better results if in vitro models that resemble brain tissue and the blood-brain barrier are used. With the use of 3D preclinical models made available by biomaterials, researchers have discovered that it is possible to overcome these limitations. Innovative in vitro models for the treatment of GBM are possible using biomaterials and novel drug carriers. This review discusses the benefits and drawbacks of 3D in vitro glioblastoma modeling systems.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| |
Collapse
|
3
|
van Echten-Deckert G. The role of sphingosine 1-phosphate metabolism in brain health and disease. Pharmacol Ther 2023; 244:108381. [PMID: 36907249 DOI: 10.1016/j.pharmthera.2023.108381] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Lipids are essential structural and functional components of the central nervous system (CNS). Sphingolipids are ubiquitous membrane components which were discovered in the brain in the late 19th century. In mammals, the brain contains the highest concentration of sphingolipids in the body. Sphingosine 1-phosphate (S1P) derived from membrane sphingolipids evokes multiple cellular responses which, depending on its concentration and localization, make S1P a double-edged sword in the brain. In the present review we highlight the role of S1P in brain development and focus on the often contrasting findings regarding its contributions to the initiation, progression and potential recovery of different brain pathologies, including neurodegeneration, multiple sclerosis (MS), brain cancers, and psychiatric illnesses. A detailed understanding of the critical implications of S1P in brain health and disease may open the door for new therapeutic options. Thus, targeting S1P-metabolizing enzymes and/or signaling pathways might help overcome, or at least ameliorate, several brain illnesses.
Collapse
|
4
|
Hong X, Zhang J, Zou J, Ouyang J, Xiao B, Wang P, Peng X. Role of COL6A2 in malignant progression and temozolomide resistance of glioma. Cell Signal 2023; 102:110560. [PMID: 36521657 DOI: 10.1016/j.cellsig.2022.110560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Gliomas are one of the most common primary malignant tumors of the central nervous system, and have an unfavorable prognosis. Even combining precise surgery, chemotherapy and radiotherapy, the survival rate is still unsatisfactory. Chemotherapy resistance is one of main reasons for its adverse prognosis. As shown by several studies, glioma stem cells (GSCs) were correlated with radiotherapy/chemotherapy resistance and high relapse rate. This study aimed to find a new biomarker related to GSCs and chemotherapy resistance. METHODS TCGA, CGGA, GSE16011, GSE23806 and GDSC datasets were used to screen the genes related to GSCs, Temozolomide (TMZ) resistance, and survival. In the TCGA, GTEx, GSE16011 and CGGA datasets, mRNA level, prognostic value, and correlation with immune infiltration in the selected genes were analyzed through methods including Kaplan-Meier analysis, Cox analysis, the ESTIMATE algorithm, and the CIBERSORT algorithm. The expression of COL6A2 mRNA and protein in different groups was detected by RT-qPCR and western blot. A MTT assay and flow cytometry were used to measure the effect of COL6A2 on proliferation and apoptosis of glioma cells. RESULTS COL6A2 was positively correlated with glioma stemness and TMZ resistance. Its expression was up-regulated in GBM, and high expression was correlated with adverse prognosis. As shown by Cox analysis, COL6A2 was an independent prognostic factor for glioma. COL6A2 mRNA was increased with the glioma grade. It was over-expressed in MGMT non-methylation and IDH wild-type specimens. The results of in vitro experiments showed that COL6A2 promots proliferation of glioma cells and inhibits their apoptosis. Meanwhile, the expression of COL6A2 in TMZ-resistant glioma cells was significantly higher than that in ordinary glioma cells. As shown by GO and KEGG pathway analysis, COL6A2 was correlated with glioma proliferation, migration, invasion, and immunity. In particular, it was significantly positively correlated with PD-1, PD-L2, PD-L1, B7-H3, CTLA-4, IDO1 and TIM-3 expression, further verifying that it may play an important role in immune response. In addition, COL6A2 might influence immune cell infiltration in the glioma microenvironment. CONCLUSION COL6A2 high-expression is an indicator for adverse glioma prognosis, and is correlated with TMZ-resistant and immune response. Meanwhile, it may be a prospective biomarker for treatment.
Collapse
Affiliation(s)
- Xia Hong
- Medical School of Jingchu University of Technology, Jingmen 448000, China
| | - Jingjing Zhang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Jianmin Zou
- The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, China
| | - Jiecai Ouyang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Boan Xiao
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Peng Wang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China.
| | - Xiaobin Peng
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China.
| |
Collapse
|
5
|
Effects of a monoclonal antibody against (pro)renin receptor on gliomagenesis. Sci Rep 2023; 13:808. [PMID: 36646875 PMCID: PMC9842725 DOI: 10.1038/s41598-023-28133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Glioblastoma is characterized by a strong self-renewal potential and poor differentiated state. We have reported previously that the (pro)renin receptor [(P)RR] is a potential target for glioma therapy by silencing the (P)RR gene. Here, we have examined the effects of a monoclonal antibody against (P)RR on gliomagenesis. Human glioma cell lines (U251MG and U87MG) and a glioma stem cell line (MGG23) were used for the in vitro study. The expressions of the Wnt/β-catenin signaling pathway (Wnt signaling pathway) components and stemness markers were measured by Western blotting. The effects of the (P)RR antibody on cell proliferation, sphere formation, apoptosis and migration were also examined. Subcutaneous xenografts were also examined in nude mice. Treatment with the (P)RR antibody reduced expression of Wnt signaling pathway components and stemness markers. Furthermore, the (P)RR antibody reduced cell proliferation and decreased sphere formation significantly. The treatment also suppressed migration and induced apoptosis. In a subcutaneous xenograft model, systemic administration of the (P)RR antibody reduced tumor volume significantly. These data show that treatment with the (P)RR antibody is a potential therapeutic strategy for treating glioblastoma.
Collapse
|
6
|
Li D, Zhang Q, Li L, Chen K, Yang J, Dixit D, Gimple RC, Ci S, Lu C, Hu L, Gao J, Shan D, Li Y, Zhang J, Shi Z, Gu D, Yuan W, Wu Q, Yang K, Zhao L, Qiu Z, Lv D, Gao W, Yang H, Lin F, Wang Q, Man J, Li C, Tao W, Agnihotri S, Qian X, Shi Y, You Y, Zhang N, Rich JN, Wang X. β2-Microglobulin Maintains Glioblastoma Stem Cells and Induces M2-like Polarization of Tumor-Associated Macrophages. Cancer Res 2022; 82:3321-3334. [PMID: 35841593 DOI: 10.1158/0008-5472.can-22-0507] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 06/08/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is a complex ecosystem that includes a heterogeneous tumor population and the tumor-immune microenvironment (TIME), prominently containing tumor-associated macrophages (TAM) and microglia. Here, we demonstrated that β2-microglobulin (B2M), a subunit of the class I major histocompatibility complex (MHC-I), promotes the maintenance of stem-like neoplastic populations and reprograms the TIME to an anti-inflammatory, tumor-promoting state. B2M activated PI3K/AKT/mTOR signaling by interacting with PIP5K1A in GBM stem cells (GSC) and promoting MYC-induced secretion of transforming growth factor-β1 (TGFβ1). Inhibition of B2M attenuated GSC survival, self-renewal, and tumor growth. B2M-induced TGFβ1 secretion activated paracrine SMAD and PI3K/AKT signaling in TAMs and promoted an M2-like macrophage phenotype. These findings reveal tumor-promoting functions of B2M and suggest that targeting B2M or its downstream axis may provide an effective approach for treating GBM. SIGNIFICANCE β2-microglobulin signaling in glioblastoma cells activates a PI3K/AKT/MYC/TGFβ1 axis that maintains stem cells and induces M2-like macrophage polarization, highlighting potential therapeutic strategies for targeting tumor cells and the immunosuppressive microenvironment in glioblastoma.
Collapse
Affiliation(s)
- Daqi Li
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Zhang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lu Li
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kexin Chen
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junlei Yang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Deobrat Dixit
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Ryan C Gimple
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Shusheng Ci
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenfei Lu
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lang Hu
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiancheng Gao
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danyang Shan
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yangqing Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danling Gu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Wei Yuan
- Department of Pathology, The Fourth Affiliated Hospital of Nantong University, The First people's Hospital of Yancheng, Yancheng, China
| | - Qiulian Wu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Linjie Zhao
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Zhixin Qiu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Institute for Translational Brain Research, Fudan University, Shanghai, China
| | - Deguan Lv
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Wei Gao
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fan Lin
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianghu Wang
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Chaojun Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing, China
| | - Weiwei Tao
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Shi
- Institute of Pathology, Ministry of Education Key Laboratory of Tumor Immunopathology, Southwest Hospital, Chongqing, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, China
| | - Jeremy N Rich
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiuxing Wang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, California
- Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Role of Transmembrane Water Exchange in Glioma Invasion/Migration: In Vivo Preclinical Study by Relaxometry at Very Low Magnetic Field. Cancers (Basel) 2022; 14:cancers14174180. [PMID: 36077717 PMCID: PMC9454706 DOI: 10.3390/cancers14174180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
This work shows that the longitudinal relaxation differences observed at very low magnetic fields between invasion/migration and proliferation processes on glioma mouse models in vivo are related to differences in the transmembrane water exchange basically linked to the aquaporin expression changes. Three glioma mouse models were used: Glio6 and Glio96 as invasion/migration models and U87 as cell proliferation model. In vivo proton longitudinal relaxation-rate constants (R1) at very low fields were measured by fast field cycling NMR (FFC-NMR). The tumor contribution to the observed proton relaxation rate, R1tum (U87: 12.26 ± 0.64 s−1; Glio6: 3.76 ± 0.88 s−1; Glio96: 6.90 ± 0.64 s−1 at 0.01 MHz), and the intracellular water lifetime, τin (U87: 826 ± 19 ms; Glio6: 516 ± 8 ms; Glio96: 596 ± 15 ms), were found to be good diagnostic hallmarks to distinguish invasion/migration from proliferation (p < 0.01 and 0.001). Overexpression of AQP4 and AQP1 were assessed in invasion/migration models, highlighting the pathophysiological role of these two aquaporins in water exchange that, in turn, determine the lower values in the observed R1 relaxation rate constant in glioma invasion/migration. Overall, our findings demonstrate that τin and R1 (measured at very low fields) are relevant biomarkers, discriminating invasion/migration from proliferation in vivo. These results highlight the use of FFC-NMR and FFC-imaging to assess the efficiency of drugs that could modulate aquaporin functions.
Collapse
|
8
|
Adjei‐Sowah EA, O'Connor SA, Veldhuizen J, Lo Cascio C, Plaisier C, Mehta S, Nikkhah M. Investigating the Interactions of Glioma Stem Cells in the Perivascular Niche at Single-Cell Resolution using a Microfluidic Tumor Microenvironment Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201436. [PMID: 35619544 PMCID: PMC9313491 DOI: 10.1002/advs.202201436] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/25/2022] [Indexed: 05/03/2023]
Abstract
The perivascular niche (PVN) is a glioblastoma tumor microenvironment (TME) that serves as a safe haven for glioma stem cells (GSCs), and acts as a reservoir that inevitably leads to tumor recurrence. Understanding cellular interactions in the PVN that drive GSC treatment resistance and stemness is crucial to develop lasting therapies for glioblastoma. The limitations of in vivo models and in vitro assays have led to critical knowledge gaps regarding the influence of various cell types in the PVN on GSCs behavior. This study developed an organotypic triculture microfluidic model as a means to recapitulate the PVN and study its impact on GSCs. This triculture platform, comprised of endothelial cells (ECs), astrocytes, and GSCs, is used to investigate GSC invasion, proliferation and stemness. Both ECs and astrocytes significantly increased invasiveness of GSCs. This study futher identified 15 ligand-receptor pairs using single-cell RNAseq with putative chemotactic mechanisms of GSCs, where the receptor is up-regulated in GSCs and the diffusible ligand is expressed in either astrocytes or ECs. Notably, the ligand-receptor pair SAA1-FPR1 is demonstrated to be involved in chemotactic invasion of GSCs toward PVN. The novel triculture platform presented herein can be used for therapeutic development and discovery of molecular mechanisms driving GSC biology.
Collapse
Affiliation(s)
| | - Samantha A. O'Connor
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ85287‐9709USA
| | - Jaimeson Veldhuizen
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ85287‐9709USA
| | - Costanza Lo Cascio
- Ivy Brain Tumor Center, Barrow Neurological InstituteSt. Joseph's Hospital and Medical Center350 W Thomas RdPhoenixAZ85013USA
| | - Christopher Plaisier
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ85287‐9709USA
| | - Shwetal Mehta
- Ivy Brain Tumor Center, Barrow Neurological InstituteSt. Joseph's Hospital and Medical Center350 W Thomas RdPhoenixAZ85013USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZ85287‐9709USA
- Virginia G. Piper Biodesign Center for Personalized DiagnosticsArizona State UniversityTempeAZ85287‐9709USA
| |
Collapse
|
9
|
Stanke KM, Kidambi S. Direct-Contact Co-culture of Astrocytes and Glioblastoma Cells Patterned using Polyelectrolyte Multilayer Templates. J Vis Exp 2022:10.3791/63420. [PMID: 35815979 PMCID: PMC10363413 DOI: 10.3791/63420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Abstract
Glioblastoma Multiforme (GBM) is the most abundant and fatal malignant brain cancer. There are more than 13,000 cases projected in the United States in 2020 and 2021. GBM tumors most often arise from astrocytes and are characterized by their invasive nature, often recruiting healthy tissues into tumor tissue. Understanding communication between astrocytes and glioblastoma cells is vital for the molecular understanding of tumor progression. This protocol demonstrates a novel patterned co-culture method to investigate contact-mediated effects of astrocytes on GBM employing layer-by-layer assembly and micro-capillary-force driven patterning. Advantages include a protein-free cell culture environment and precise control of cellular interaction dictated by the pattern dimensions. This technique provides a versatile, economical, reproducible protocol for mimicking cellular interaction between glioma and astrocytes in glioma tumors. This model can further be used to tease apart changes in GBM molecular biology due to physical contact with astrocytes or with non-contact mediated soluble cofactor communication.
Collapse
Affiliation(s)
- Kimberly M Stanke
- Complex Biosystems Graduate Program, University of Nebraska, Lincoln; Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln
| | - Srivatsan Kidambi
- Complex Biosystems Graduate Program, University of Nebraska, Lincoln; Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center; Nebraska Center for Integrated Biomolecular Communication, University of Nebraska, Lincoln; Nebraska Center for the Prevention of Obesity Diseases, University of Nebraska, Lincoln; Nebraska Center for Materials and Nanoscience, University of Nebraska, Lincoln; Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center;
| |
Collapse
|
10
|
Xing Z, Liu Z, Fu X, Zhou S, Liu L, Dang Q, Guo C, Ge X, Lu T, Zheng Y, Dai L, Han X, Wang X. Clinical Significance and Immune Landscape of a Pyroptosis-Derived LncRNA Signature for Glioblastoma. Front Cell Dev Biol 2022; 10:805291. [PMID: 35223836 PMCID: PMC8866949 DOI: 10.3389/fcell.2022.805291] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/12/2022] [Indexed: 01/15/2023] Open
Abstract
Introduction: Pyroptosis was recently implicated in the initiation and progression of tumors, including glioblastoma (GBM). This study aimed to explore the clinical significance of pyroptosis-related lncRNAs (PRLs) in GBM. Methods: Three independent cohorts were retrieved from the TCGA and CGGA databases. The consensus clustering and weighted gene coexpression network analysis (WGCNA) were applied to identify PRLs. The LASSO algorithm was employed to develop and validate a pyroptosis-related lncRNA signature (PRLS) in three independent cohorts. The molecular characteristics, clinical significances, tumor microenvironment, immune checkpoints profiles, and benefits of chemotherapy and immunotherapy regarding to PRLS were also explored. Results: In the WGCNA framework, a key module that highly correlated with pyroptosis was extracted for identifying PRLs. Univariate Cox analysis further revealed the associations between PRLs and overall survival. Based on the expression profiles of PRLs, the PRLS was initially developed in TCGA cohort (n = 143) and then validated in two CGGA cohorts (n = 374). Multivariate Cox analysis demonstrated that our PRLS model was an independent risk factor. More importantly, this signature displayed a stable and accurate performance in predicting prognosis at 1, 3, and 5 years, with all AUCs above 0.7. The decision curve analysis also indicated that our signature had promising clinical application. In addition, patients with high PRLS score suggested a more abundant immune infiltration, higher expression of immune checkpoint genes, and better response to immunotherapy but worse to chemotherapy. Conclusion: A novel pyroptosis-related lncRNA signature with a robust performance was constructed and validated in multiple cohorts. This signature provided new perspectives for clinical management and precise treatments of GBM.
Collapse
Affiliation(s)
- Zhe Xing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xudong Fu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Shaolong Zhou
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunguang Guo
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyong Ge
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Taoyuan Lu
- Department of Cerebrovascular Disease, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Youyang Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lirui Dai
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Correspondence: Xinjun Wang, ; Xinwei Han,
| | - Xinjun Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Glioma Metabolism and Microenvironment Research, Zhengzhou, China
- Correspondence: Xinjun Wang, ; Xinwei Han,
| |
Collapse
|
11
|
Coniglio SJ, Segall JE. Microglial-stimulation of glioma invasion involves the EGFR ligand amphiregulin. PLoS One 2021; 16:e0260252. [PMID: 34843542 PMCID: PMC8629255 DOI: 10.1371/journal.pone.0260252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022] Open
Abstract
High grade glioma is one of the deadliest human cancers with a median survival rate of only one year following diagnosis. The highly motile and invasive nature of high grade glioma makes it difficult to completely remove surgically. Therefore, increasing our knowledge of the mechanisms glioma cells use to invade normal brain is of critical importance in designing novel therapies. It was previously shown by our laboratory that tumor-associated microglia (TAMs) stimulate glioma cell invasion and this process is dependent on CSF-1R signaling. In this study, we seek to identify pro-invasive factors that are upregulated in microglia in a CSF-1R-dependent manner. We assayed cDNA and protein from microglia treated with conditioned media from the murine glioma cell line GL261, and discovered that several EGFR ligands including amphiregulin (AREG) are strongly upregulated. This upregulation is blocked by addition of a pharmacological CSF-1R inhibitor. Using RNA interference, we show that AREG-depleted microglia are less effective at promoting invasion of GL261 cells into Matrigel-coated invasion chambers. In addition, an AREG blocking antibody strongly attenuates the ability of THP-1 macrophages to activate human glioma cell line U87 invasion. Furthermore, we have identified a signaling pathway which involves CSF-1 signaling through ERK to upregulate AREG expression in microglia. Interfering with ERK using pharmacological inhibitors prevents AREG upregulation in microglia and microglia-stimulated GL261 invasion. These data highlight AREG as a key factor in produced by tumor associated microglia in promoting glioma invasion.
Collapse
Affiliation(s)
- Salvatore J. Coniglio
- New Jersey Center for Science Technology and Mathematics, Kean University, Union, NJ, United States of America
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Jeffrey E. Segall
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Gruss Lipper Biophotonics Center, Bronx, NY, United States of America
| |
Collapse
|
12
|
Nayak C, Singh SK. In silico identification of natural product inhibitors against Octamer-binding transcription factor 4 (Oct4) to impede the mechanism of glioma stem cells. PLoS One 2021; 16:e0255803. [PMID: 34613998 PMCID: PMC8494328 DOI: 10.1371/journal.pone.0255803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Octamer-binding transcription factor 4 (Oct4) is a core regulator in the retention of stemness, invasive, and self-renewal properties in glioma initiating cells (GSCs) and its overexpression inhibits the differentiation of glioma cells promoting tumor cell proliferation. The Pit-Oct-Unc (POU) domain comprising POU-specific domain (POUS) and POU-type homeodomain (POUHD) subdomains is the most critical part of the Oct4 for the generation of induced pluripotent stem cells from somatic cells that lead to tumor initiation, invasion, posttreatment relapse, and therapeutic resistance. Therefore, the present investigation hunts for natural product inhibitors (NPIs) against the POUHD domain of Oct4 by employing receptor-based virtual screening (RBVS) followed by binding free energy calculation and molecular dynamics simulation (MDS). RBVS provided 13 compounds with acceptable ranges of pharmacokinetic properties and good docking scores having key interactions with the POUHD domain. More Specifically, conformational and interaction stability analysis of 13 compounds through MDS unveiled two compounds ZINC02145000 and ZINC32124203 which stabilized the backbone of protein even in the presence of linker and POUS domain. Additionally, ZINC02145000 and ZINC32124203 exhibited stable and strong interactions with key residues W277, R242, and R234 of the POUHD domain even in dynamic conditions. Interestingly, ZINC02145000 and ZINC32124203 established communication not only with the POUHD domain but also with the POUS domain indicating their incredible potency toward thwarting the function of Oct4. ZINC02145000 and ZINC32124203 also reduced the flexibility and escalated the correlations between the amino acid residues of Oct4 evidenced by PCA and DCCM analysis. Finally, our examination proposed two NPIs that can impede the Oct4 function and may help to improve overall survival, diminish tumor relapse, and achieve a cure not only in deadly disease GBM but also in other cancers with minimal side effects.
Collapse
Affiliation(s)
- Chirasmita Nayak
- Computer-Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi Tamil Nadu, India
| | - Sanjeev Kumar Singh
- Computer-Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics, Alagappa University, Karaikudi Tamil Nadu, India
| |
Collapse
|
13
|
Huang K, Yue X, Zheng Y, Zhang Z, Cheng M, Li L, Chen Z, Yang Z, Bian E, Zhao B. Development and Validation of an Mesenchymal-Related Long Non-Coding RNA Prognostic Model in Glioma. Front Oncol 2021; 11:726745. [PMID: 34540695 PMCID: PMC8446619 DOI: 10.3389/fonc.2021.726745] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/16/2021] [Indexed: 12/29/2022] Open
Abstract
Glioma is well known as the most aggressive and prevalent primary malignant tumor in the central nervous system. Molecular subtypes and prognosis biomarkers remain a promising research area of gliomas. Notably, the aberrant expression of mesenchymal (MES) subtype related long non-coding RNAs (lncRNAs) is significantly associated with the prognosis of glioma patients. In this study, MES-related genes were obtained from The Cancer Genome Atlas (TCGA) and the Ivy Glioblastoma Atlas Project (Ivy GAP) data sets of glioma, and MES-related lncRNAs were acquired by performing co-expression analysis of these genes. Next, Cox regression analysis was used to establish a prognostic model, that integrated ten MES-related lncRNAs. Glioma patients in TCGA were divided into high-risk and low-risk groups based on the median risk score; compared with the low-risk groups, patients in the high-risk group had shorter survival times. Additionally, we measured the specificity and sensitivity of our model with the ROC curve. Univariate and multivariate Cox analyses showed that the prognostic model was an independent prognostic factor for glioma. To verify the predictive power of these candidate lncRNAs, the corresponding RNA-seq data were downloaded from the Chinese Glioma Genome Atlas (CGGA), and similar results were obtained. Next, we performed the immune cell infiltration profile of patients between two risk groups, and gene set enrichment analysis (GSEA) was performed to detect functional annotation. Finally, the protective factors DGCR10 and HAR1B, and risk factor SNHG18 were selected for functional verification. Knockdown of DGCR10 and HAR1B promoted, whereas knockdown of SNHG18 inhibited the migration and invasion of gliomas. Collectively, we successfully constructed a prognostic model based on a ten MES-related lncRNAs signature, which provides a novel target for predicting the prognosis for glioma patients.
Collapse
Affiliation(s)
- Kebing Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Xiaoyu Yue
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Yinfei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Zhengwei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Meng Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Lianxin Li
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Zhigang Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Erbao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, China
| |
Collapse
|
14
|
Mazurek M, Rola R. The implications of nitric oxide metabolism in the treatment of glial tumors. Neurochem Int 2021; 150:105172. [PMID: 34461111 DOI: 10.1016/j.neuint.2021.105172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/03/2021] [Accepted: 08/21/2021] [Indexed: 12/20/2022]
Abstract
Glial tumors are the most common intracranial malignancies. Unfortunately, despite such a high prevalence, patients' prognosis is usually poor. It is related to the high invasiveness, tendency to relapse and the resistance of tumors to traditional methods of treatment. An important link in the aspect of these issues may be nitric oxide (NO) metabolism. It is a very complex mechanism with multidirectional effects on the neoplastic process. Depending on the concentration axis, it can both exert pro-tumor action as well as contribute to the inhibition of tumorigenesis. The latest observations show that the control of its metabolism can be very helpful in the development of new methods of treating gliomas, as well as in increasing the effectiveness of the agents currently used. The influence of nitric oxide and nitric oxide synthase (NOS) activity on glioma stem cells seem to be of particular importance. The use of specific inhibitors may allow the reduction of tumor growth and its tendency to relapse. Another important feature of GSCs is their conditioning of glioma resistance to traditional forms of treatment. Recent studies have shown that modulation of NO metabolism can suppress this effect, preventing the induction of radio and chemoresistance. Moreover, nitric oxide is involved in the regulation of a number of immune mechanisms. Adequate modulation of its metabolism may contribute to the induction of an anti-tumor response in the patients' immune system.
Collapse
Affiliation(s)
- Marek Mazurek
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University in Lublin, Poland.
| | - Radosław Rola
- Chair and Department of Neurosurgery and Paediatric Neurosurgery, Medical University in Lublin, Poland
| |
Collapse
|
15
|
Zheng ZQ, Chen JT, Zheng MC, Yang LJ, Wang JM, Liu QL, Chen LF, Ye ZC, Lin JM, Lin ZX. Nestin+/CD31+ cells in the hypoxic perivascular niche regulate glioblastoma chemoresistance by upregulating JAG1 and DLL4. Neuro Oncol 2021; 23:905-919. [PMID: 33249476 DOI: 10.1093/neuonc/noaa265] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Failure of glioblastoma (GBM) therapy is often ascribed to different types of glioblastoma stem-like cell (GSLC) niche; in particular, a hypoxic perivascular niche (HPVN) is involved in GBM progression. However, the cells responsible for HPVNs remain unclear. METHODS Immunostaining was performed to determine the cells involved in HPVNs. A hypoxic chamber and 3-dimensional (3D) microfluidic chips were designed to simulate a HPVN based on the pathological features of GBM. The phenotype of GSLCs was evaluated by fluorescence scanning in real time and proliferation and apoptotic assays. The expression of JAG1, DLL4, and Hes1 was determined by immunostaining, ELISA, Western blotting, and quantitative PCR. Their clinical prognostic significance in GBM HPVNs and total tumor tissues were verified by clinical data and The Cancer Genome Atlas databases. RESULTS Nestin+/CD31+ cells and pericytes constitute the major part of microvessels in the HPVN, and the high ratio of nestin+/CD31+ cells rather than pericytes are responsible for the poor prognosis of GBM. A more real HPVN was simulated by a hypoxic coculture system in vitro, which consisted of 3D microfluidic chips and a hypoxic chamber. Nestin+/CD31+ cells in the HPVN were derived from GSLC transdifferentiation and promoted GSLC chemoresistance by providing more JAG1 and DLL4 to induce downstream Hes1 overexpression. Poor GBM prognosis correlated with Hes1 expression of tumor cells in the GBM HPVN, and not with total Hes1 expression in GBM tissues. CONCLUSIONS These results highlight the critical role of nestin+/CD31+ cells in HPVNs that acts in GBM chemoresistance and reveal the distinctive prognostic value of these molecular markers in HPVNs.
Collapse
Affiliation(s)
- Zong-Qing Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, P.R. China
- Department of Neurosurgery, Sanbo Brain Hospital of Capital Medical University, Beijing, P.R. China
| | - Jin-Tao Chen
- Department of Neurosurgery, Fujian Sanbo Funeng Brain Hospital, Fuzhou, Fujian, P.R. China
| | - Ming-Cheng Zheng
- Department of Neurosurgery, the Fifth Hospital of Hospital of Xiamen, Xiamen, Fujian, P.R. China
| | - Li-Juan Yang
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Jun-Ming Wang
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing, P.R. China
| | - Quan-Li Liu
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing, P.R. China
| | - Lu-Fei Chen
- Fujian Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Zu-Cheng Ye
- Fujian Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, P.R. China
| | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing, P.R. China
| | - Zhi-Xiong Lin
- Department of Neurosurgery, Sanbo Brain Hospital of Capital Medical University, Beijing, P.R. China
| |
Collapse
|
16
|
Gerigk M, Bulstrode H, Shi HH, Tönisen F, Cerutti C, Morrison G, Rowitch D, Huang YYS. On-chip perivascular niche supporting stemness of patient-derived glioma cells in a serum-free, flowable culture. LAB ON A CHIP 2021; 21:2343-2358. [PMID: 33969368 PMCID: PMC8204159 DOI: 10.1039/d1lc00271f] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/03/2021] [Indexed: 05/05/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and the most aggressive type of primary brain malignancy. Glioblastoma stem-like cells (GSCs) can migrate in vascular niches within or away from the tumour mass, increasing tumour resistance to treatments and contributing to relapses. To study individual GSC migration and their interactions with the perivasculature of the tumour microenvironment, there is a need to develop a human organotypic in vitro model. Herein, we demonstrated a perivascular niche-on-a-chip, in a serum-free condition with gravity-driven flow, that supported the stemness of patient-derived GSCs and foetal neural stem cells grown in a three-dimensional environment (3D). Endothelial cells from three organ origins, (i) human brain microvascular endothelial cells (hCMEC/D3), (ii) human umbilical vein endothelial cells (HUVECs) and, (iii) human lung microvascular endothelial cells (HMVEC-L) formed rounded microvessels within the extracellular-matrix integrated microfluidic chip. By optimising cell extraction protocols, systematic studies were performed to evaluate the effects of serum-free media, 3D cell cultures, and the application of gravity-driven flow on the characteristics of endothelial cells and their co-culture with GSCs. Our results showed the maintenance of adherent and tight junction markers of hCMEC/D3 in the serum-free culture and that gravity-driven flow was essential to support adequate viability of both the microvessel and the GSCs in co-culture (>80% viability at day 3). Endpoint biological assays showed upregulation of neovascularization-related genes (e.g., angiopoietins, vascular endothelial growth factor receptors) in endothelial cells co-cultured with GSCs in contrast to the neural stem cell reference that showed insignificant changes. The on-chip platform further permitted live-cell imaging of GSC - microvessel interaction, enabling quantitative analysis of GSC polarization and migration. Overall, our comparative genotypic (i.e. qPCR) and phenotypic (i.e. vessel permeability and GSC migration) studies showed that organotypic (brain cancer cells-brain endothelial microvessel) interactions differed from those within non-tissue specific vascular niches of human origin. The development and optimization of this on-chip perivascular niche, in a serum-free flowable culture, could provide the next level of complexity of an in vitro system to study the influence of glioma stem cells on brain endothelium.
Collapse
Affiliation(s)
- Magda Gerigk
- Department of Engineering, University of Cambridge, UK. and The Nanoscience Centre, University of Cambridge, UK
| | - Harry Bulstrode
- Department of Clinical Neuroscience, University of Cambridge, UK
| | - HaoTian Harvey Shi
- Department of Mechanical & Industrial Engineering, University of Toronto, Canada and Department of Engineering, University of Cambridge, UK.
| | - Felix Tönisen
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboudumc, Netherlands and Department of Engineering, University of Cambridge, UK.
| | - Camilla Cerutti
- Randall Centre of Cell & Molecular Biophysics, King's College London, UK
| | | | - David Rowitch
- Department of Paediatrics, University of Cambridge, UK
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, UK. and The Nanoscience Centre, University of Cambridge, UK
| |
Collapse
|
17
|
Paolillo M, Comincini S, Schinelli S. In Vitro Glioblastoma Models: A Journey into the Third Dimension. Cancers (Basel) 2021; 13:cancers13102449. [PMID: 34070023 PMCID: PMC8157833 DOI: 10.3390/cancers13102449] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In this review, the thorny issue of glioblastoma models is addressed, with a focus on 3D in vitro models. In the first part of the manuscript, glioblastoma features and classification are recapitulated, in order to highlight the major critical aspects that should be taken into account when choosing a glioblastoma 3D model. In the second part of the review, the 3D models described in the literature are critically discussed, considering the advantages, disadvantages, and feasibility for each experimental model, in the light of the potential issues that researchers want to address. Abstract Glioblastoma multiforme (GBM) is the most lethal primary brain tumor in adults, with an average survival time of about one year from initial diagnosis. In the attempt to overcome the complexity and drawbacks associated with in vivo GBM models, together with the need of developing systems dedicated to screen new potential drugs, considerable efforts have been devoted to the implementation of reliable and affordable in vitro GBM models. Recent findings on GBM molecular features, revealing a high heterogeneity between GBM cells and also between other non-tumor cells belonging to the tumoral niche, have stressed the limitations of the classical 2D cell culture systems. Recently, several novel and innovative 3D cell cultures models for GBM have been proposed and implemented. In this review, we first describe the different populations and their functional role of GBM and niche non-tumor cells that could be used in 3D models. An overview of the current available 3D in vitro systems for modeling GBM, together with their major weaknesses and strengths, is presented. Lastly, we discuss the impact of groundbreaking technologies, such as bioprinting and multi-omics single cell analysis, on the future implementation of 3D in vitro GBM models.
Collapse
Affiliation(s)
- Mayra Paolillo
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy;
- Correspondence:
| | - Sergio Comincini
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Sergio Schinelli
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy;
| |
Collapse
|
18
|
Piper K, DePledge L, Karsy M, Cobbs C. Glioma Stem Cells as Immunotherapeutic Targets: Advancements and Challenges. Front Oncol 2021; 11:615704. [PMID: 33718170 PMCID: PMC7945033 DOI: 10.3389/fonc.2021.615704] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most common and lethal primary brain malignancy. Despite major investments in research into glioblastoma biology and drug development, treatment remains limited and survival has not substantially improved beyond 1-2 years. Cancer stem cells (CSC) or glioma stem cells (GSC) refer to a population of tumor originating cells capable of self-renewal and differentiation. While controversial and challenging to study, evidence suggests that GCSs may result in glioblastoma tumor recurrence and resistance to treatment. Multiple treatment strategies have been suggested at targeting GCSs, including immunotherapy, posttranscriptional regulation, modulation of the tumor microenvironment, and epigenetic modulation. In this review, we discuss recent advances in glioblastoma treatment specifically focused on targeting of GCSs as well as their potential integration into current clinical pathways and trials.
Collapse
Affiliation(s)
- Keenan Piper
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States.,Sidney Kimmel Medical College, Philadelphia, PA, United States
| | - Lisa DePledge
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States.,University of Washington School of Medicine, Spokane, WA, United States
| | - Michael Karsy
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Charles Cobbs
- Ben & Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA, United States
| |
Collapse
|
19
|
Coelho BP, Fernandes CFDL, Boccacino JM, Souza MCDS, Melo-Escobar MI, Alves RN, Prado MB, Iglesia RP, Cangiano G, Mazzaro GLR, Lopes MH. Multifaceted WNT Signaling at the Crossroads Between Epithelial-Mesenchymal Transition and Autophagy in Glioblastoma. Front Oncol 2020; 10:597743. [PMID: 33312955 PMCID: PMC7706883 DOI: 10.3389/fonc.2020.597743] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
Tumor cells can employ epithelial-mesenchymal transition (EMT) or autophagy in reaction to microenvironmental stress. Importantly, EMT and autophagy negatively regulate each other, are able to interconvert, and both have been shown to contribute to drug-resistance in glioblastoma (GBM). EMT has been considered one of the mechanisms that confer invasive properties to GBM cells. Autophagy, on the other hand, may show dual roles as either a GBM-promoter or GBM-suppressor, depending on microenvironmental cues. The Wingless (WNT) signaling pathway regulates a plethora of developmental and biological processes such as cellular proliferation, adhesion and motility. As such, GBM demonstrates deregulation of WNT signaling in favor of tumor initiation, proliferation and invasion. In EMT, WNT signaling promotes induction and stabilization of different EMT activators. WNT activity also represses autophagy, while nutrient deprivation induces β-catenin degradation via autophagic machinery. Due to the importance of the WNT pathway to GBM, and the role of WNT signaling in EMT and autophagy, in this review we highlight the effects of the WNT signaling in the regulation of both processes in GBM, and discuss how the crosstalk between EMT and autophagy may ultimately affect tumor biology.
Collapse
Affiliation(s)
- Bárbara Paranhos Coelho
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Camila Felix de Lima Fernandes
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Jacqueline Marcia Boccacino
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Maria Clara da Silva Souza
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Maria Isabel Melo-Escobar
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Rodrigo Nunes Alves
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Mariana Brandão Prado
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Rebeca Piatniczka Iglesia
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Giovanni Cangiano
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Giulia La Rocca Mazzaro
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Marilene Hohmuth Lopes
- Laboratory of Neurobiology and Stem Cells, Institute of Biomedical Sciences, Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Birzu C, Tran S, Bielle F, Touat M, Mokhtari K, Younan N, Psimaras D, Hoang‐Xuan K, Sanson M, Delattre J, Idbaih A. Leptomeningeal Spread in Glioblastoma: Diagnostic and Therapeutic Challenges. Oncologist 2020; 25:e1763-e1776. [PMID: 33394574 PMCID: PMC7648332 DOI: 10.1634/theoncologist.2020-0258] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common and aggressive primary malignant brain tumor. Leptomeningeal spread (LMS) is a severe complication of GBM, raising diagnostic and therapeutic challenges in clinical routine. METHODS We performed a review of the literature focused on LMS in GBM. MEDLINE and EMBASE databases were queried from 1989 to 2019 for articles describing diagnosis and therapeutic options in GBM LMS, as well as risk factors and pathogenic mechanisms. RESULTS We retrieved 155 articles, including retrospective series, case reports, and early phase clinical trials, as well as preclinical studies. These articles confirmed that LMS in GBM remains (a) a diagnostic challenge with cytological proof of LMS obtained in only 35% of cases and (b) a therapeutic challenge with a median overall survival below 2 months with best supportive care alone. For patients faced with suggestive clinical symptoms, whole neuroaxis magnetic resonance imaging and cerebrospinal fluid analysis are both recommended. Liquid biopsies are under investigation and may help prompt a reliable diagnosis. Based on the literature, a multimodal and personalized therapeutic approach of LMS, including surgery, radiotherapy, systemic cytotoxic chemotherapy, and intrathecal chemotherapies, may provide benefits to selected patients. Interestingly, molecular targeted therapies appear promising in case of actionable molecular target and should be considered. CONCLUSION As the prognosis of glioblastoma is improving over time, LMS becomes a more common complication. Our review highlights the need for translational studies and clinical trials dedicated to this challenging condition in order to improve diagnostic and therapeutic strategies. IMPLICATIONS FOR PRACTICE This review summarizes the diagnostic tools and applied treatments for leptomeningeal spread, a complication of glioblastoma, as well as their outcomes. The importance of exhaustive molecular testing for molecular targeted therapies is discussed. New diagnostic and therapeutic strategies are outlined, and the need for translational studies and clinical trials dedicated to this challenging condition is highlighted.
Collapse
Affiliation(s)
- Cristina Birzu
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neurologie 2‐MazarinParisFrance
| | - Suzanne Tran
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neuropathologie‐EscourolleParisFrance
| | - Franck Bielle
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neuropathologie‐EscourolleParisFrance
| | - Mehdi Touat
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neurologie 2‐MazarinParisFrance
| | - Karima Mokhtari
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neuropathologie‐EscourolleParisFrance
| | - Nadia Younan
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neurologie 2‐MazarinParisFrance
| | - Dimitri Psimaras
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neurologie 2‐MazarinParisFrance
| | - Khe Hoang‐Xuan
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neurologie 2‐MazarinParisFrance
| | - Marc Sanson
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neurologie 2‐MazarinParisFrance
| | - Jean‐Yves Delattre
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neurologie 2‐MazarinParisFrance
| | - Ahmed Idbaih
- Sorbonne Université, INSERM, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) S 1127, Institut du Cerveau et de la Moelle épinière (ICM), Assistance Publique–Hôpitaux de Paris (AP‐HP), Hôpitaux Universitaires La Pitié Salpêtrière—Charles Foix Service de Neurologie 2‐MazarinParisFrance
| |
Collapse
|
21
|
Miyai M, Kanayama T, Hyodo F, Kinoshita T, Ishihara T, Okada H, Suzuki H, Takashima S, Wu Z, Hatano Y, Egashira Y, Enomoto Y, Nakayama N, Soeda A, Yano H, Hirata A, Niwa M, Sugie S, Mori T, Maekawa Y, Iwama T, Matsuo M, Hara A, Tomita H. Glucose transporter Glut1 controls diffuse invasion phenotype with perineuronal satellitosis in diffuse glioma microenvironment. Neurooncol Adv 2020; 3:vdaa150. [PMID: 33506198 PMCID: PMC7817894 DOI: 10.1093/noajnl/vdaa150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Gliomas typically escape surgical resection and recur due to their “diffuse invasion” phenotype, enabling them to infiltrate diffusely into the normal brain parenchyma. Over the past 80 years, studies have revealed 2 key features of the “diffuse invasion” phenotype, designated the Scherer’s secondary structure, and include perineuronal satellitosis (PS) and perivascular satellitosis (PVS). However, the mechanisms are still unknown. Methods We established a mouse glioma cell line (IG27) by manipulating the histone H3K27M mutation, frequently harboring in diffuse intrinsic pontine gliomas, that reproduced the diffuse invasion phenotype, PS and PVS, following intracranial transplantation in the mouse brain. Further, to broadly apply the results in this mouse model to human gliomas, we analyzed data from 66 glioma patients. Results Increased H3K27 acetylation in IG27 cells activated glucose transporter 1 (Glut1) expression and induced aerobic glycolysis and TCA cycle activation, leading to lactate, acetyl-CoA, and oncometabolite production irrespective of oxygen and glucose levels. Gain- and loss-of-function in vivo experiments demonstrated that Glut1 controls the PS of glioma cells, that is, attachment to and contact with neurons. GLUT1 is also associated with early progression in glioma patients. Conclusions Targeting the transporter Glut1 suppresses the unique phenotype, “diffuse invasion” in the diffuse glioma mouse model. This work leads to promising therapeutic and potential useful imaging targets for anti-invasion in human gliomas widely.
Collapse
Affiliation(s)
- Masafumi Miyai
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Fuminori Hyodo
- Department of Radiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takamasa Kinoshita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan.,Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takuma Ishihara
- Gifu University Hospital, Innovative and Clinical Research Promotion Center, Gifu University, Gifu, Japan
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroki Suzuki
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shigeo Takashima
- Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan
| | - Zhiliang Wu
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yuichiro Hatano
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yusuke Egashira
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yukiko Enomoto
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Noriyuki Nakayama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akio Soeda
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hirohito Yano
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akihiro Hirata
- Division of Animal Experiment, Life Science Research Center, Gifu University, Gifu, Japan
| | - Masayuki Niwa
- Medical Science Division, United Graduate School of Drug Discovery and Medical Information Sciences, Gifu, Japan
| | - Shigeyuki Sugie
- Department of Pathology, Asahi University Hospital, Gifu, Japan
| | - Takashi Mori
- Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University (G-CHAIN), Gifu, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan.,Domain of Integrated Life Systems, Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masayuki Matsuo
- Department of Radiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
22
|
2D and 3D in vitro assays to quantify the invasive behavior of glioblastoma stem cells in response to SDF-1α. Biotechniques 2020; 69:339-346. [PMID: 32867513 DOI: 10.2144/btn-2020-0046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Invasion is a hallmark of cancer and therefore in vitro invasion assays are important tools in cancer research. We aimed to describe in vitro 2D transwell assays and 3D spheroid assays to quantitatively determine the invasive behavior of glioblastoma stem cells in response to the chemoattractant SDF-1α. Matrigel was used as a matrix in both assays. We demonstrated quantitatively that SDF-1α increased invasive behavior of glioblastoma stem cells in both assays. We conclude that the 2D transwell invasion assay is easy to perform, fast and less complex whereas the more time-consuming 3D spheroid invasion assay is physiologically closer to the in vivo situation.
Collapse
|
23
|
Immune-Related lncRNA Risk Signatures Predict Survival of IDH Wild-Type and MGMT Promoter Unmethylated Glioblastoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1971284. [PMID: 32851059 PMCID: PMC7441444 DOI: 10.1155/2020/1971284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/02/2020] [Accepted: 06/24/2020] [Indexed: 11/24/2022]
Abstract
Introduction Glioblastoma is the most malignant grade of glioma, and it is also the most common primary tumor in the brain. Immunotherapy is a kind of precise tumor treatment. However, there are limited studies about immune-related lncRNA. This study is aimed at analyzing immune-related lncRNAs in glioblastoma and screening out prognostic factors, providing new potential targets for glioblastoma immunology research. Material and Methods. Gene expression data and clinical data of IDH wild-type with MGMT promoter unmethylated glioblastoma were acquired from the TCGA and CGGA databases. Immune-related lncRNAs were identified with the help of data from the InnateDB database. Immune prognostic factors were recognized by Cox regression analysis. GSEA analysis pursued their potential functions. Results We found 318 immune-related lncRNAs. Among them, there were 137 immune-related lncRNAs that were upregulated and 181 that were downregulated. 15 prognostic lncRNAs were identified by Cox regression, and a total of 6 molecules were included in the following risk scoring model. GSEA showed that these lncRNAs participated in functions such as protein digestion and absorption and the PPAR signaling pathway. Conclusion There are limited studies about immune regulation mechanisms of lncRNA in IDH wild-type with MGMT promoter unmethylated glioblastoma. The identified immune-related lncRNAs in glioblastoma might contribute new targets and research directions for immunological molecular studies of glioblastoma.
Collapse
|
24
|
Bhargav AG, Mondal SK, Garcia CA, Green JJ, Quiñones‐Hinojosa A. Nanomedicine Revisited: Next Generation Therapies for Brain Cancer. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Adip G. Bhargav
- Mayo Clinic College of Medicine and Science Mayo Clinic 200 First Street SW Rochester MN 55905 USA
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| | - Sujan K. Mondal
- Department of Pathology University of Pittsburgh School of Medicine 200 Lothrop Street Pittsburgh PA 15213 USA
| | - Cesar A. Garcia
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| | - Jordan J. Green
- Departments of Biomedical Engineering, Neurosurgery, Oncology, Ophthalmology, Materials Science and Engineering, and Chemical and Biomolecular Engineering, Translational Tissue Engineering Center, Bloomberg‐Kimmel Institute for Cancer Immunotherapy, Institute for Nanobiotechnology Johns Hopkins University School of Medicine 400 N. Broadway, Smith 5017 Baltimore MD 21231 USA
| | - Alfredo Quiñones‐Hinojosa
- Department of Neurologic Surgery Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
- Departments of Otolaryngology‐Head and Neck Surgery/Audiology Neuroscience, Cancer Biology, and Anatomy Mayo Clinic 4500 San Pablo Rd. Jacksonville FL 32224 USA
| |
Collapse
|
25
|
Giotta Lucifero A, Luzzi S, Brambilla I, Schena L, Mosconi M, Foiadelli T, Savasta S. Potential roads for reaching the summit: an overview on target therapies for high-grade gliomas. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:61-78. [PMID: 32608376 PMCID: PMC7975828 DOI: 10.23750/abm.v91i7-s.9956] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022]
Abstract
Background: The tailored targeting of specific oncogenes represents a new frontier in the treatment of high-grade glioma in the pursuit of innovative and personalized approaches. The present study consists in a wide-ranging overview of the target therapies and related translational challenges in neuro-oncology. Methods: A review of the literature on PubMed/MEDLINE on recent advances concerning the target therapies for treatment of central nervous system malignancies was carried out. In the Medical Subject Headings, the terms “Target Therapy”, “Target drug” and “Tailored Therapy” were combined with the terms “High-grade gliomas”, “Malignant brain tumor” and “Glioblastoma”. Articles published in the last five years were further sorted, based on the best match and relevance. The ClinicalTrials.gov website was used as a source of the main trials, where the search terms were “Central Nervous System Tumor”, “Malignant Brain Tumor”, “Brain Cancer”, “Brain Neoplasms” and “High-grade gliomas”. Results: A total of 137 relevant articles and 79 trials were selected. Target therapies entailed inhibitors of tyrosine kinases, PI3K/AKT/mTOR pathway, farnesyl transferase enzymes, p53 and pRB proteins, isocitrate dehydrogenases, histone deacetylases, integrins and proteasome complexes. The clinical trials mostly involved combined approaches. They were phase I, II, I/II and III in 33%, 42%, 16%, and 9% of the cases, respectively. Conclusion: Tyrosine kinase and angiogenesis inhibitors, in combination with standard of care, have shown most evidence of the effectiveness in glioblastoma. Resistance remains an issue. A deeper understanding of the molecular pathways involved in gliomagenesis is the key aspect on which the translational research is focusing, in order to optimize the target therapies of newly diagnosed and recurrent brain gliomas. (www.actabiomedica.it)
Collapse
Affiliation(s)
- Alice Giotta Lucifero
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Ilaria Brambilla
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Lucia Schena
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Mario Mosconi
- Orthopaedic and Traumatology Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - Thomas Foiadelli
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Salvatore Savasta
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| |
Collapse
|
26
|
Giotta Lucifero A, Luzzi S, Brambilla I, Trabatti C, Mosconi M, Savasta S, Foiadelli T. Innovative therapies for malignant brain tumors: the road to a tailored cure. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:5-17. [PMID: 32608372 PMCID: PMC7975829 DOI: 10.23750/abm.v91i7-s.9951] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022]
Abstract
Background: Immune tolerance, immune escape, neoangiogenesis, phenotypic changes, and glioma stem cells are all responsible for the resistance of malignant brain tumors to current therapies and persistent recurrence. The present study provides a panoramic view of innovative therapies for malignant brain tumors, especially glioblastoma, aimed at achieving a tailored approach. Methods: PubMed/Medline and ClinicalTrials.gov were the main sources of an extensive literature review in which “Regenerative Medicine,” “Cell-Based Therapy,” “Chemotherapy,” “Vaccine,” “Cell Engineering,” “Immunotherapy, Active,” “Immunotherapy, Adoptive,” “Stem Cells,” “Gene Therapy,” “Target Therapy,” “Brain Cancer,” “Glioblastoma,” and “Malignant Brain Tumor” were the search terms. Only articles in English published in the last 5 years were included. A further selection was made according to the quality of the studies and level of evidence. Results: Cell-based and targeted therapies represent the newest frontiers of brain cancer treatment. Active and adoptive immunotherapies, stem cell therapies, and gene therapies represent a tremendous evolution in recent years due to many preclinical and clinical studies. Clinical trials have validated the effectiveness of antibody-based immunotherapies, including an in-depth study of bevacizumab, in combination with standard of care. Preclinical data highlights the role of vaccines, stem cells, and gene therapies to prevent recurrence. Conclusion: Monoclonal antibodies strengthen the first-line therapy for high grade gliomas. Vaccines, engineered cells, stem cells, and gene and targeted therapies are good candidates for second-line treatment of both newly diagnosed and recurrent gliomas. Further data are necessary to validate this tailored approach at the bedside. (www.actabiomedica.it)
Collapse
Affiliation(s)
| | - Sabino Luzzi
- Neurosurgery Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Neurosurgery Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Ilaria Brambilla
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Chiara Trabatti
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Mario Mosconi
- Orthopaedic and Traumatology Unit, Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
| | - Salvatore Savasta
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| | - Thomas Foiadelli
- Pediatric Clinic, Department of Pediatrics, Fondazione IRCCS Policlinico San Matteo, Uni-versity of Pavia, Pavia, Italy.
| |
Collapse
|
27
|
Dual Role of WISP1 in maintaining glioma stem cells and tumor-supportive macrophages in glioblastoma. Nat Commun 2020; 11:3015. [PMID: 32541784 PMCID: PMC7295765 DOI: 10.1038/s41467-020-16827-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
The interplay between glioma stem cells (GSCs) and the tumor microenvironment plays crucial roles in promoting malignant growth of glioblastoma (GBM), the most lethal brain tumor. However, the molecular mechanisms underlying this crosstalk are incompletely understood. Here, we show that GSCs secrete the Wnt‐induced signaling protein 1 (WISP1) to facilitate a pro-tumor microenvironment by promoting the survival of both GSCs and tumor-associated macrophages (TAMs). WISP1 is preferentially expressed and secreted by GSCs. Silencing WISP1 markedly disrupts GSC maintenance, reduces tumor-supportive TAMs (M2), and potently inhibits GBM growth. WISP1 signals through Integrin α6β1-Akt to maintain GSCs by an autocrine mechanism and M2 TAMs through a paracrine manner. Importantly, inhibition of Wnt/β-catenin-WISP1 signaling by carnosic acid (CA) suppresses GBM tumor growth. Collectively, these data demonstrate that WISP1 plays critical roles in maintaining GSCs and tumor-supportive TAMs in GBM, indicating that targeting Wnt/β-catenin-WISP1 signaling may effectively improve GBM treatment and the patient survival. The tumour microenvironment plays an important role in promoting glioblastoma. Here, the authors show that glioma stem cells secrete WISP1, which promotes both the survival of the stem cells and tumour-associated macrophages.
Collapse
|
28
|
Torrisi F, Minafra L, Cammarata FP, Savoca G, Calvaruso M, Vicario N, Maccari L, Pérès EA, Özçelik H, Bernaudin M, Botta L, Russo G, Parenti R, Valable S. SRC Tyrosine Kinase Inhibitor and X-rays Combined Effect on Glioblastoma Cell Lines. Int J Mol Sci 2020; 21:ijms21113917. [PMID: 32486205 PMCID: PMC7312922 DOI: 10.3390/ijms21113917] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma (GBM) is one of the most lethal types of tumor due to its high recurrence level in spite of aggressive treatment regimens involving surgery, radiotherapy and chemotherapy. Hypoxia is a feature of GBM, involved in radioresistance, and is known to be at the origin of treatment failure. The aim of this work was to assess the therapeutic potential of a new targeted c-SRC inhibitor molecule, named Si306, in combination with X-rays on the human glioblastoma cell lines, comparing normoxia and hypoxia conditions. For this purpose, the dose modifying factor and oxygen enhancement ratio were calculated to evaluate the Si306 radiosensitizing effect. DNA damage and the repair capability were also studied from the kinetic of γ-H2AX immunodetection. Furthermore, motility processes being supposed to be triggered by hypoxia and irradiation, the role of c-SRC inhibition was also analyzed to evaluate the migration blockage by wound healing assay. Our results showed that inhibition of the c-SRC protein enhances the radiotherapy efficacy both in normoxic and hypoxic conditions. These data open new opportunities for GBM treatment combining radiotherapy with molecularly targeted drugs to overcome radioresistance.
Collapse
Affiliation(s)
- Filippo Torrisi
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95123 Catania, Italy; (F.T.); (N.V.)
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy; (L.M.); (G.S.); (M.C.); (G.R.)
| | - Luigi Minafra
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy; (L.M.); (G.S.); (M.C.); (G.R.)
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy
| | - Francesco P. Cammarata
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy; (L.M.); (G.S.); (M.C.); (G.R.)
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy
- Correspondence: (F.P.C.); (R.P.)
| | - Gaetano Savoca
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy; (L.M.); (G.S.); (M.C.); (G.R.)
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy
| | - Marco Calvaruso
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy; (L.M.); (G.S.); (M.C.); (G.R.)
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy
| | - Nunzio Vicario
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95123 Catania, Italy; (F.T.); (N.V.)
| | - Laura Maccari
- Lead Discovery Siena s.r.l. (LDS), via Vittorio Alfieri, 31, Castelnuovo Berardenga, 53019 Siena, Italy; (L.M.); (L.B.)
| | - Elodie A. Pérès
- UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Normandie University, 14074 Caen, France; (E.A.P.); (H.Ö.); (M.B.); (S.V.)
| | - Hayriye Özçelik
- UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Normandie University, 14074 Caen, France; (E.A.P.); (H.Ö.); (M.B.); (S.V.)
| | - Myriam Bernaudin
- UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Normandie University, 14074 Caen, France; (E.A.P.); (H.Ö.); (M.B.); (S.V.)
| | - Lorenzo Botta
- Lead Discovery Siena s.r.l. (LDS), via Vittorio Alfieri, 31, Castelnuovo Berardenga, 53019 Siena, Italy; (L.M.); (L.B.)
| | - Giorgio Russo
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy; (L.M.); (G.S.); (M.C.); (G.R.)
- Institute of Molecular Bioimaging and Physiology, National Research Council, IBFM-CNR, 90015 Cefalù, Italy
| | - Rosalba Parenti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95123 Catania, Italy; (F.T.); (N.V.)
- Correspondence: (F.P.C.); (R.P.)
| | - Samuel Valable
- UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Normandie University, 14074 Caen, France; (E.A.P.); (H.Ö.); (M.B.); (S.V.)
| |
Collapse
|
29
|
Shevchenko V, Arnotskaya N, Pak O, Sharma A, Sharma HS, Khotimchenko Y, Bryukhovetskiy A, Bryukhovetskiy I. Molecular determinants of the interaction between glioblastoma CD133 + cancer stem cells and the extracellular matrix. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 151:155-169. [PMID: 32448605 DOI: 10.1016/bs.irn.2020.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common primary tumor of the human brain. It is characterized by invasive growth and strong resistance to treatment, and the median survival time of patients is 15 months. The invasive growth of this tumor type is associated with tumor cells with an aggressive phenotype, while its treatment resistance is attributed to cancer stem cells (CSCs). It remains unclear if CSCs have a more invasive nature than differentiated glioblastoma cells (DGCs), and what contribution CSCs make to the aggressive phenotype of GBM. Interaction with the extracellular matrix (ECM) is a key factor in the development of invasion. The aim of the present study was to compare the expression levels of signaling pathway proteins involved in interaction of receptors with the ECM in CSCs and DGCs. The U-87MG GBM cell line was used in the present study CSCs were extracted from gliomaspheres through magnetic-activated cell sorting based on the expression of cluster of differentiation 133 (CD133); CD133-negative DCGs were used as a control. HPLC and mass spectrometry were also used, and biological and molecular functions, signaling pathways and protein-protein interactions were analyzed using publicly available databases. Increased expression levels of the following 10 proteins involved in interaction with the ECM were identified in CSCs, compared with expression levels in DGCs: COL6A1, COL6A3, FN1, ITGA2, ITGA5, ITGAV, ITGB1, ITGB3, LAMB1 and LAMC1. The proteome of CSCs was observed to have >2-fold higher expression of these key proteins, when compared with the DGC proteome. Increased expression levels of four proteins (FERMT2, LOXL2, HDAC2 and FBN1) involved in activating signaling in response to receptor interaction with the ECM was also observed, indicating that CSCs may have highly invasive nature. LOXL2 expression level was >9-fold higher in CSCs compared to DGCs, suggesting that this protein may have potential as an marker for CSCs and as a target for this cell type in GBM.
Collapse
Affiliation(s)
- Valeriy Shevchenko
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Oncoproteomics, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow, Russia
| | - Natalia Arnotskaya
- Laboratory of Oncoproteomics, Institute of Carcinogenesis, N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Moscow, Russia
| | - Oleg Pak
- Medical Center, Far Eastern Federal University, Vladivostok, Russia
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, University Hospital, Uppsala University, S-75185 Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, University Hospital, Uppsala University, S-75185 Uppsala, Sweden
| | - Yuri Khotimchenko
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Andrey Bryukhovetskiy
- NeuroVita Clinic of Interventional and Restorative Neurology and Therapy, Moscow, Russia
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Medical Center, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia.
| |
Collapse
|
30
|
Targeting RGS4 Ablates Glioblastoma Proliferation. Int J Mol Sci 2020; 21:ijms21093300. [PMID: 32392739 PMCID: PMC7247588 DOI: 10.3390/ijms21093300] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma (GBM) is the most common type of adult primary brain tumor with a median survival rate of less than 15 months, regardless of the current standard of care. Cellular heterogeneity, self-renewal ability and tumorigenic glioma cancer stem cell (GSC) populations contribute to the difficulty in treating GBM. G-protein-coupled receptors (GPCRs) are the largest group of membrane proteins and mediate many cellular responses. Regulators of G-protein signaling 4 (RGS4) are negative regulators of G-protein signaling, and elevated levels of RGS4 are reportedly linked with several human diseases, including cancer. This study investigates the effect of silencing RGS4, resulting in inhibition of GSC growth, invasion and migration. Data obtained from The Cancer Genome Atlas (TCGA) demonstrated poor patient survival with high expression of RGS4. Immunohistochemistry and immunoblot analysis conducted on GBM patient biopsy specimens demonstrated increased RGS4 expression correlative with the TCGA data. RNA sequencing confirmed a significant decrease in the expression of markers involved in GSC invasion and migration, particularly matrix metalloproteinase-2 (MMP2) in knockout of RGS4 using CRISPR plasmid (ko-RGS4)-treated samples compared to parental controls. Gelatin zymography confirmed the reduced activity of MMP2 in ko-RGS4-treated samples. Silencing RGS4 further reduced the invasive and migratory abilities and induction of apoptosis of GSCs as evidenced by Matrigel plug assay, wound healing assay and human apoptosis array. Collectively, our results showed that the silencing of RGS4 plays an important role in regulating multiple cellular functions, and is an important therapeutic target in GBM.
Collapse
|
31
|
Zhang M, Wang X, Chen X, Guo F, Hong J. Prognostic Value of a Stemness Index-Associated Signature in Primary Lower-Grade Glioma. Front Genet 2020; 11:441. [PMID: 32431729 PMCID: PMC7216823 DOI: 10.3389/fgene.2020.00441] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022] Open
Abstract
Objective As a prevalent and infiltrative cancer type of the central nervous system, the prognosis of lower-grade glioma (LGG) in adults is highly heterogeneous. Recent evidence has demonstrated the prognostic value of the mRNA expression-based stemness index (mRNAsi) in LGG. Our aim was to develop a stemness index-based signature (SI-signature) for risk stratification and survival prediction. Methods Differentially expressed genes (DEGs) between LGG in the Cancer Genome Atlas (TCGA) and normal brain tissue samples from the Genotype-Tissue Expression (GTEx) project were screened out, and the weighted gene correlation network analysis (WGCNA) was employed to identify the mRNAsi-related gene sets. Meanwhile, the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed for the functional annotation of the key genes. ESTIMATE was used to calculate tumor purity for acquiring the correct mRNAsi. Differences in overall survival (OS) between the high and low mRNAsi (corrected mRNAsi) groups were compared using the Kaplan Meier analysis. By combining the Lasso regression with univariate and multivariate Cox regression, the SI-signature was constructed and validated using the Chinese Glioma Genome Atlas (CGGA). Results There was a significant difference in OS between the high and low mRNAsi groups, which was also observed in the two corrected mRNAsi groups. Based on threshold limits, 86 DEGs were most significantly associated with mRNAsi via WGCNA. Seven genes (ADAP2, ALOX5AP, APOBEC3C, FCGRT, GNG5, LRRC25, and SP100) were selected to establish a risk signature for primary LGG. The ROC curves showed a fair performance in survival prediction in both the TCGA and the CGGA validation cohorts. Univariate and multivariate Cox regression revealed that the risk group was an independent prognostic factor in primary LGG. The nomogram was developed based on clinical parameters integrated with the risk signature, and its accuracy for predicting 3- and 5-years survival was assessed by the concordance index, the area under the curve of the time-dependent receiver operating characteristics curve, and calibration curves. Conclusion The SI-signature with seven genes could serve as an independent predictor, and suggests the importance of stemness features in risk stratification and survival prediction in primary LGG.
Collapse
Affiliation(s)
- Mingwei Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Institute of Immunotherapy, Fujian Medical University, Fuzhou, China.,Key Laboratory of Radiation Biology (Fujian Medical University), Fujian Province University, Fuzhou, China.,Fujian Key Laboratory of Individualized Active Immunotherapy, Fuzhou, China.,Fujian Medical University Union Hospital, Fuzhou, China
| | - Xuezhen Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaoping Chen
- Department of Statistics, College of Mathematics and Informatics & FJKLMAA, Fujian Normal University, Fuzhou, China
| | - Feibao Guo
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jinsheng Hong
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Key Laboratory of Radiation Biology (Fujian Medical University), Fujian Province University, Fuzhou, China.,Fujian Key Laboratory of Individualized Active Immunotherapy, Fuzhou, China
| |
Collapse
|
32
|
Vollmann-Zwerenz A, Leidgens V, Feliciello G, Klein CA, Hau P. Tumor Cell Invasion in Glioblastoma. Int J Mol Sci 2020; 21:E1932. [PMID: 32178267 PMCID: PMC7139341 DOI: 10.3390/ijms21061932] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/02/2020] [Accepted: 03/09/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is a particularly devastating tumor with a median survival of about 16 months. Recent research has revealed novel insights into the outstanding heterogeneity of this type of brain cancer. However, all GBM subtypes share the hallmark feature of aggressive invasion into the surrounding tissue. Invasive glioblastoma cells escape surgery and focal therapies and thus represent a major obstacle for curative therapy. This review aims to provide a comprehensive understanding of glioma invasion mechanisms with respect to tumor-cell-intrinsic properties as well as cues provided by the microenvironment. We discuss genetic programs that may influence the dissemination and plasticity of GBM cells as well as their different invasion patterns. We also review how tumor cells shape their microenvironment and how, vice versa, components of the extracellular matrix and factors from non-neoplastic cells influence tumor cell motility. We further discuss different research platforms for modeling invasion. Finally, we highlight the importance of accounting for the complex interplay between tumor cell invasion and treatment resistance in glioblastoma when considering new therapeutic approaches.
Collapse
Affiliation(s)
- Arabel Vollmann-Zwerenz
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, 93053 Regensburg, Germany; (A.V.-Z.); (V.L.)
| | - Verena Leidgens
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, 93053 Regensburg, Germany; (A.V.-Z.); (V.L.)
| | - Giancarlo Feliciello
- Fraunhofer-Institute for Toxicology and Experimental Medicine, Division of Personalized Tumor Therapy, 93053 Regensburg, Germany; (G.F.); (C.A.K.)
| | - Christoph A. Klein
- Fraunhofer-Institute for Toxicology and Experimental Medicine, Division of Personalized Tumor Therapy, 93053 Regensburg, Germany; (G.F.); (C.A.K.)
- Experimental Medicine and Therapy Research, University of Regensburg, 93053 Regensburg, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, 93053 Regensburg, Germany; (A.V.-Z.); (V.L.)
| |
Collapse
|
33
|
Behling F, Barrantes-Freer A, Behnes CL, Stockhammer F, Rohde V, Adel-Horowski A, Rodríguez-Villagra OA, Barboza MA, Brück W, Lehmann U, Stadelmann C, Hartmann C. Expression of Olig2, Nestin, NogoA and AQP4 have no impact on overall survival in IDH-wildtype glioblastoma. PLoS One 2020; 15:e0229274. [PMID: 32160197 PMCID: PMC7065747 DOI: 10.1371/journal.pone.0229274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 02/03/2020] [Indexed: 11/29/2022] Open
Abstract
Despite many years of research efforts and clinical trials the prognosis of patients diagnosed with glioblastoma remains very poor. The oligodendrocyte transcription factor 2 (Olig2) was identified as a marker for glioma stem cells, which are believed to be responsible for glioma recurrence and therapy resistance. In this retrospective analysis we assessed the prognostic value of oligodendroglial and glioma stem cell markers in 113 IDH-wildtype glioblastomas. Immunohistochemical staining for Olig2, NogoA, AQP4 and Nestin was performed in combination with sequencing of IDH1 and IDH2 as well as promotor methylation analysis of the MGMT gene. Even though differences in overall survival according to Olig2 expression were observed, univariate and multivariate survival analysis did not reveal a firm significant prognostic impact of Olig2, NogoA, AQP4 or Nestin expression. Additionally, no differences in the expression of these markers depending on clinical status, age or gender were found. The established independent prognostic factors age<65, Karnofsky Performance Status> = 70 and methylated MGMT gene promoter were significant in the multivariate analysis. In conclusion expression of oligodendroglial and glioma stem cell markers do not have an independent prognostic effect in IDH-wildtype glioblastoma.
Collapse
Affiliation(s)
- Felix Behling
- Institute of Neuropathology, University Medical Center Goettingen, Goettingen, Germany
- Department of Neurosurgery, University Hospital Tuebingen, Tuebingen, Germany
- Center for CNS Tumors, Comprehensive Cancer Center Tuebingen-Stuttgart, University Hospital Tuebingen, Tuebingen, Germany
| | - Alonso Barrantes-Freer
- Institute of Neuropathology, University Medical Center Goettingen, Goettingen, Germany
- Department of Neuropathology, Leipzig University Medicine, Leipzig, Germany
| | - Carl Ludwig Behnes
- Institute of Pathology, University Medical Center Goettingen, Goettingen, Germany
| | - Florian Stockhammer
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Veit Rohde
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Antonia Adel-Horowski
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Odir Antonio Rodríguez-Villagra
- Neuroscience Research Center, University of Costa Rica, San José, Costa Rica
- Institute for Psychological Research, University of Costa Rica, San José, Costa Rica
| | - Miguel Angel Barboza
- Neurosciences Department, Hospital Dr. Rafael A. Calderón Guardia, CCSS, University of Costa Rica, San José, Costa Rica
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center Goettingen, Goettingen, Germany
| | - Ulrich Lehmann
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Goettingen, Goettingen, Germany
| | - Christian Hartmann
- Department of Neuropathology, Institute of Pathology, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
34
|
Dundar B, Markwell SM, Sharma NV, Olson CL, Mukherjee S, Brat DJ. Methods for in vitro modeling of glioma invasion: Choosing tools to meet the need. Glia 2020; 68:2173-2191. [PMID: 32134155 DOI: 10.1002/glia.23813] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/24/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
Widespread tumor cell invasion is a fundamental property of diffuse gliomas and is ultimately responsible for their poor prognosis. A greater understanding of basic mechanisms underlying glioma invasion is needed to provide insights into therapies that could potentially counteract them. While none of the currently available in vitro models can fully recapitulate the complex interactions of glioma cells within the brain tumor microenvironment, if chosen and developed appropriately, these models can provide controlled experimental settings to study molecular and cellular phenomena that are challenging or impossible to model in vivo. Therefore, selecting the most appropriate in vitro model, together with its inherent advantages and limitations, for specific hypotheses and experimental questions achieves primary significance. In this review, we describe and discuss commonly used methods for modeling and studying glioma invasion in vitro, including platforms, matrices, cell culture, and visualization techniques, so that choices for experimental approach are informed and optimal.
Collapse
Affiliation(s)
- Bilge Dundar
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Steven M Markwell
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nitya V Sharma
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Cheryl L Olson
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Subhas Mukherjee
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniel J Brat
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
35
|
Riboni L, Abdel Hadi L, Navone SE, Guarnaccia L, Campanella R, Marfia G. Sphingosine-1-Phosphate in the Tumor Microenvironment: A Signaling Hub Regulating Cancer Hallmarks. Cells 2020; 9:E337. [PMID: 32024090 PMCID: PMC7072483 DOI: 10.3390/cells9020337] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
As a key hub of malignant properties, the cancer microenvironment plays a crucial role intimately connected to tumor properties. Accumulating evidence supports that the lysophospholipid sphingosine-1-phosphate acts as a key signal in the cancer extracellular milieu. In this review, we have a particular focus on glioblastoma, representative of a highly aggressive and deleterious neoplasm in humans. First, we highlight recent advances and emerging concepts for how tumor cells and different recruited normal cells contribute to the sphingosine-1-phosphate enrichment in the cancer microenvironment. Then, we describe and discuss how sphingosine-1-phosphate signaling contributes to favor cancer hallmarks including enhancement of proliferation, stemness, invasion, death resistance, angiogenesis, immune evasion and, possibly, aberrant metabolism. We also discuss the potential of how sphingosine-1-phosphate control mechanisms are coordinated across distinct cancer microenvironments. Further progress in understanding the role of S1P signaling in cancer will depend crucially on increasing knowledge of its participation in the tumor microenvironment.
Collapse
Affiliation(s)
- Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Loubna Abdel Hadi
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, via Fratelli Cervi, 93, 20090 Segrate, Milan, Italy
| | - Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
- Department of Clinical Sciences and Community Health, University of Milan, 20100 Milan, Italy
| | - Rolando Campanella
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122 Milan, Italy (L.G.)
| |
Collapse
|
36
|
Zeng F, Wang K, Liu X, Zhao Z. Comprehensive profiling identifies a novel signature with robust predictive value and reveals the potential drug resistance mechanism in glioma. Cell Commun Signal 2020; 18:2. [PMID: 31907037 PMCID: PMC6943920 DOI: 10.1186/s12964-019-0492-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background Gliomas are the most common and malignant brain tumors. The standard therapy is surgery combined with radiotherapy, chemotherapy, and/or other comprehensive methods. However, the emergence of chemoresistance is the main obstacle in treatment and its mechanism is still unclear. Methods We firstly developed a multi-gene signature by integrated analysis of cancer stem cell and drug resistance related genes. The Chinese Glioma Genome Atlas (CGGA, 325 samples) and The Cancer Genome Atlas (TCGA, 699 samples) datasets were then employed to verify the efficacy of the risk signature and investigate its significance in glioma prognosis. GraphPad Prism, SPSS and R language were used for statistical analysis and graphical work. Results This signature could distinguish the prognosis of patients, and patients with high risk score exhibited short survival time. The Cox regression and Nomogram model indicated the independent prognostic performance and high prognostic accuracy of the signature for survival. Combined with a well-known chemotherapy impact factor-MGMT promoter methylation status, this risk signature could further subdivide patients with distinct survival. Functional analysis of associated genes revealed signature-related biological process of cell proliferation, immune response and cell stemness. These mechanisms were confirmed in patient samples. Conclusions The signature was an independent and powerful prognostic biomarker in glioma, which would improve risk stratification and provide a more accurate assessment of personalized treatment. Additional file 8 Video abstract
Collapse
Affiliation(s)
- Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, No.119 South 4th Ring Road West, Fengtai District, Beijing, 100070, China
| | - Kuanyu Wang
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Xiu Liu
- Department of Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Zheng Zhao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, No.119 South 4th Ring Road West, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
37
|
Petővári G, Dankó T, Krencz I, Hujber Z, Rajnai H, Vetlényi E, Raffay R, Pápay J, Jeney A, Sebestyén A. Inhibition of Metabolic Shift can Decrease Therapy Resistance in Human High-Grade Glioma Cells. Pathol Oncol Res 2020; 26:23-33. [PMID: 31187466 PMCID: PMC7109188 DOI: 10.1007/s12253-019-00677-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022]
Abstract
The high-grade brain malignancy, glioblastoma multiforme (GBM), is one of the most aggressive tumours in central nervous system. The developing resistance against recent therapies and the recurrence rate of GBMs are extremely high. In spite several new ongoing trials, GBM therapies could not significantly increase the survival rate of the patients as significantly. The presence of inter- and intra-tumoral heterogeneity of GBMs arise the problem to find both the pre-existing potential resistant clones and the cellular processes which promote the adaptation mechanisms such as multidrug resistance, stem cell-ness or metabolic alterations, etc. In our work, the in situ metabolic heterogeneity of high-grade human glioblastoma cases were analysed by immunohistochemistry using tissue-microarray. The potential importance of the detected metabolic heterogeneity was tested in three glioma cell lines (grade III-IV) using protein expression analyses (Western blot and WES Simple) and therapeutic drug (temozolomide), metabolic inhibitor treatments (including glutaminase inhibitor) to compare the effects of rapamycin (RAPA) and glutaminase inhibitor combinations in vitro (Alamar Blue and SRB tests). The importance of individual differences and metabolic alterations were observed in mono-therapeutic failures, especially the enhanced Rictor expressions after different mono-treatments in correlation to lower sensitivity (temozolomide, doxycycline, etomoxir, BPTES). RAPA combinations with other metabolic inhibitors were the best strategies except for RAPA+glutaminase inhibitor. These observations underline the importance of multi-targeting metabolic pathways. Finally, our data suggest that the detected metabolic heterogeneity (the high mTORC2 complex activity, enhanced expression of Rictor, p-Akt, p-S6, CPT1A, and LDHA enzymes in glioma cases) and the microenvironmental or treatment induced metabolic shift can be potential targets in combination therapy. Therefore, it should be considered to map tissue heterogeneity and alterations with several cellular metabolism markers in biopsy materials after applying recently available or new treatments.
Collapse
Affiliation(s)
- Gábor Petővári
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Titanilla Dankó
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Ildikó Krencz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Zoltán Hujber
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Hajnalka Rajnai
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Enikő Vetlényi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Regina Raffay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Judit Pápay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - András Jeney
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary
| | - Anna Sebestyén
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, Budapest, H-1085, Hungary.
| |
Collapse
|
38
|
Xiong B, Chen S, Zhu P, Huang M, Gao W, Zhu R, Qian J, Peng Y, Zhang Y, Dai H, Ling Y. Design, Synthesis, and Biological Evaluation of Novel Thiazolyl Substituted Bis-pyrazole Oxime Derivatives with Potent Antitumor Activities by Selectively Inducing Apoptosis and ROS in Cancer Cells. Med Chem 2019; 15:743-754. [PMID: 30147012 DOI: 10.2174/1573406414666180827112724] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/06/2018] [Accepted: 07/26/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND A large number of pyrazole derivatives have different biological activities such as anticancer, antimicrobial, anti-inflammatory, analgesic and antiepileptic activity. Among them, pyrazole oximes have attracted much attention due to their potential pharmacological activities, particularly anticancer activities. OBJECTIVE Our goal is to synthesize novel thiazolyl substituted bis-pyrazole oxime derivatives with potent antitumor activities by selectively inducing apoptosis and Reactive Oxygen Species (ROS) accumulation in cancer cells. METHODS Eighteen bis-pyrazole oximes were synthesized by conjugating thiazolyl substituted pyrazoles with pyrazoxime. The target compounds were characterized by 1HNMR, 13C NMR, and HRMS, and screened for their antiproliferative activity against four cancer cells in MTT assay. The most potent compound was examined for its inhibitory effect and ROS accumulation in both cancer cells HCT116 and normal intestinal epithelial cells CCD841. Finally, the most potent compound was further evaluated for its apoptotic induction by flow cytometry analysis and immunoblot analysis of apoptosis-related proteins and DNA damage proteins. RESULTS Most compounds displayed potent antiproliferative activity against four cancer cell lines in vitro, displaying potencies superior to 5-FU. In particular, the most potent compound 13l selectively inhibited proliferation of colorectal cancer HCT116 cells but not normal colon CCD841 cells. Furthermore, compound 13l also selectively promoted intracellular ROS accumulation in HCT116 which was involved in 13l inhibition of cancer cell proliferation and induction of cell apoptosis. Finally, compound 13l also dose-dependently induced cancer cell apoptosis by regulating apoptotic and DNA damage related proteins expressions. CONCLUSION Our synthetic bis-pyrazole oxime derivatives possess potent antitumor activities by selectively inducing apoptosis and ROS accumulation in cancer cells, which may hold great promise as therapeutic agents for the treatment of human cancers.
Collapse
Affiliation(s)
- Biao Xiong
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China
| | - Shi Chen
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China
| | - Peng Zhu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China
| | - Meiling Huang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China.,College of Chemistry and Chemical Engineering, Nantong University, Nantong 226001, China
| | - Weijie Gao
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China
| | - Rui Zhu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China
| | - Jianqiang Qian
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China
| | - Yanfu Peng
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China
| | - Yanan Zhang
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China
| | - Hong Dai
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China.,College of Chemistry and Chemical Engineering, Nantong University, Nantong 226001, China
| | - Yong Ling
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China.,College of Chemistry and Chemical Engineering, Nantong University, Nantong 226001, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
39
|
van Dijken BRJ, Jan van Laar P, Li C, Yan JL, Boonzaier NR, Price SJ, van der Hoorn A. Ventricle contact is associated with lower survival and increased peritumoral perfusion in glioblastoma. J Neurosurg 2019; 131:717-723. [PMID: 30485234 DOI: 10.3171/2018.5.jns18340] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/02/2018] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The purpose of this study was to prospectively investigate outcome and differences in peritumoral MRI characteristics of glioblastomas (GBMs) that were in contact with the ventricles (ventricle-contacting tumors) and those that were not (noncontacting tumors). GBMs are heterogeneous tumors with variable survival. Lower survival is suggested for patients with ventricle-contacting tumors than for those with noncontacting tumors. This might be supported by aggressive peritumoral MRI features. However, differences in MRI characteristics of the peritumoral environment between ventricle-contacting and noncontacting GBMs have not yet been investigated. METHODS Patients with newly diagnosed GBM underwent preoperative MRI with contrast-enhanced T1-weighted, FLAIR, diffusion-weighted, and perfusion-weighted sequences. Tumors were categorized into ventricle-contacting or noncontacting based on contrast enhancement. Survival analysis was performed using log-rank for univariate analysis and Cox regression for multivariate analysis. Normalized perfusion (relative cerebral blood volume [rCBV]) and diffusion (apparent diffusion coefficient [ADC]) values were calculated in 2 regions: the peritumoral nonenhancing FLAIR region overlapping the subventricular zone and the remaining peritumoral nonenhancing FLAIR region. RESULTS Overall survival was significantly lower for patients with contacting tumors than for those with noncontacting tumors (434 vs 747 days, p < 0.001). Progression-free survival showed a comparable trend (260 vs 375 days, p = 0.094). Multivariate analysis confirmed a survival difference for both overall survival (HR 3.930, 95% CI 1.740-8.875, p = 0.001) and progression-free survival (HR 2.506, 95% CI 1.254-5.007, p = 0.009). Peritumoral perfusion was higher in contacting than in noncontacting tumors for both FLAIR regions (p = 0.04). There was no difference in peritumoral ADC values between the 2 groups. CONCLUSIONS Patients with ventricle-contacting tumors had poorer outcomes than patients with noncontacting tumors. This disadvantage of ventricle contact might be explained by higher peritumoral perfusion leading to more aggressive behavior.
Collapse
Affiliation(s)
- Bart Roelf Jan van Dijken
- 1Department of Radiology (EB44), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter Jan van Laar
- 1Department of Radiology (EB44), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Chao Li
- 2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom.,3Department of Neurosurgery, Shanghai General Hospital, Shanghai, China
| | - Jiun-Lin Yan
- 2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom.,4Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, Taiwan; and.,5Department of Neurosurgery, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Natalie Rosella Boonzaier
- 2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom
| | | | -
- 2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom
| | - Anouk van der Hoorn
- 1Department of Radiology (EB44), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,2Cambridge Brain Tumour Imaging Laboratory, Department of Clinical Neurosciences, Division of Neurosurgery, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
40
|
Eugenio-Pérez D, Briones-Herrera A, Martínez-Klimova E, Pedraza-Chaverri J. Divide et Impera: Drp1-mediated Mitochondrial Fission in Glioma Malignancy. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:423-433. [PMID: 31543706 PMCID: PMC6747948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Mitochondria are pivotal organelles involved in vital cellular functions, including energy generation, reactive oxygen species and calcium signaling, as well as intermediate biosynthesis. They are dynamic organelles that adapt their shape, size, and distribution to changes in intracellular conditions, being able to divide, fuse, or move along the cell, processes known as mitochondrial dynamics. Mitochondrial dynamics are involved in cell division and migration, as well as maintenance of pluripotency in stem (non-differentiated) cells. Thus, its central role in carcinogenesis is not surprising. Particularly, mitochondrial dynamics have been found to be pivotal to the development of gliomas, a lethal group of tumors developed from glial cells, which are nervous system cells that provide support to neurons. Unfortunately, prognosis of glioma patients is poor, most of them do not survive more than five years after diagnosis. In this context, it is fundamental to understand the cellular mechanisms involved in this pathology, in order to develop an appropriate clinical approach. As previously mentioned, mitochondrial dynamics is central to glioma development, particularly, mitochondrial division (fission) and one of its central effectors, dynamin-related protein 1 (Drp1), have been observed to be enhanced in gliomas and involved in the maintenance of stem cells (which initiate and maintain the tumor), as well as in migration and invasiveness, being central to gliomagenesis. In this review, we discuss the findings on mitochondrial fission role in these processes, further, we analyze the potential use of Drp1 as a novel prognostic biomarker in glioma patients.
Collapse
Affiliation(s)
| | | | | | - José Pedraza-Chaverri
- To whom all correspondence should be addressed: José Pedraza-Chaverri, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, 04510 Mexico;
| |
Collapse
|
41
|
Feng F, Zhang M, Yang C, Heng X, Wu X. The dual roles of autophagy in gliomagenesis and clinical therapy strategies based on autophagic regulation mechanisms. Biomed Pharmacother 2019; 120:109441. [PMID: 31541887 DOI: 10.1016/j.biopha.2019.109441] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/02/2019] [Accepted: 09/06/2019] [Indexed: 01/14/2023] Open
Abstract
Autophagy, a self-digestion intracellular catabolic process, plays a crucial role in cellular homeostasis under conditions of starvation, oxidative stress and genotoxic stress. The capability of maintaining homeostasis contributes to preventing malignant behavior in normal cells. Many studies have provided compelling evidence that autophagy is involved in brain tumor recurrence and chemotherapy and radiotherapy resistance. Gliomas, as the primary central nervous system (CNS) tumors, are characterized by rapid, aggressive growth and recurrence and have a poor prognosis and bleak outlook even with modern multimodality strategies involving maximal surgical resection, radiotherapy and alkylating agent-based chemotherapy. Autophagy-associated signaling pathways, such as the extracellular signal-regulated kinase1/2 (ERK1/2) pathway, class I phosphatidylinositol 3-phosphate kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway and nuclear factor kappa-B (NF-κB) pathway, act as tumor suppressors or protect tumor cells against chemotherapy/radiotherapy-induced cytotoxicity in gliomagenesis. Through these pathways, both lethal autophagy and protective autophagy play crucial roles in tumor initiation, chemoresistance and glioma stem cell differentiation. Moreover, lethal autophagy and protective autophagy have been identified as novel therapeutic targets in glioma according to the mechanisms described above. Here, we discuss the multiple impacts of the autophagic response on distinct phases of gliomagenesis and the advanced progress of therapies based on this concept.
Collapse
Affiliation(s)
- Fan Feng
- Institute of Clinical Medicine College, Qingdao University, # 38, Dengzhou Road, Qingdao 266071, Shandong, China
| | - Moxuan Zhang
- Weifang Medical University, 261042, # 7166, Baotong Western Road, Weifang, Shandong, China
| | - Chuanchao Yang
- Weifang Medical University, 261042, # 7166, Baotong Western Road, Weifang, Shandong, China
| | - Xueyuan Heng
- Department of Neurosurgery, Linyi People's Hospital, # 27, Jiefang Eastern Road, Linyi 276000, Shandong, China.
| | - Xiujie Wu
- Department of Neurosurgery, Linyi People's Hospital, # 27, Jiefang Eastern Road, Linyi 276000, Shandong, China.
| |
Collapse
|
42
|
Riva M, Wouters R, Weerasekera A, Belderbos S, Nittner D, Thal DR, Baert T, Giovannoni R, Gsell W, Himmelreich U, Van Ranst M, Coosemans A. CT-2A neurospheres-derived high-grade glioma in mice: a new model to address tumor stem cells and immunosuppression. Biol Open 2019; 8:bio.044552. [PMID: 31511246 PMCID: PMC6777368 DOI: 10.1242/bio.044552] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recently, several promising treatments for high-grade gliomas (HGGs) failed to provide significant benefit when translated from the preclinical setting to patients. Improving the animal models is fundamental to overcoming this translational gap. To address this need, we developed and comprehensively characterized a new in vivo model based on the orthotopic implantation of CT-2A cells cultured in neurospheres (NS/CT-2A). Murine CT-2A methylcholanthrene-induced HGG cells (C57BL/6 background) were cultured in monolayers (ML) or NS and orthotopically inoculated in syngeneic animals. ML/CT-2A and NS/CT-2A tumors' characterization included the analysis of tumor growth, immune microenvironment, glioma stem cells (GSCs), vascularization and metabolites. The immuno-modulating properties of NS/CT-2A and ML/CT-2A cells on splenocytes were tested in vitro. Mice harboring NS/CT-2A tumors had a shorter survival than those harboring ML/CT-2A tumors (P=0.0033). Compared to standard ML/CT-2A tumors, NS/CT-2A tumors showed more abundant GSCs (P=0.0002 and 0.0770 for Nestin and CD133, respectively) and regulatory T cells (Tregs, P=0.0074), and a strong tendency towards an increased vascularization (P=0.0503). There were no significant differences in metabolites' composition between NS/ and ML/CT-2A tumors. In vitro, NS were able to drive splenocytes towards a more immunosuppressive status by reducing CD8+ T cells (P=0.0354) and by promoting Tregs (P=0.0082), macrophages (MF, P=0.0019) and their M2 subset (P=0.0536). Compared to standard ML/CT-2A tumors, NS/CT-2A tumors show a more aggressive phenotype with increased immunosuppression and GSCs proliferation. Because of these specific features, the NS/CT-2A model represents a clinically relevant platform in the search for new HGG treatments aimed at reducing immunosuppression and eliminating GSCs. Summary: The NS/CT-2A tumor model represents a valuable research platform for the study of innovative treatments aimed at eliminating GSCs and reversing the tumor-induced immunosuppression in HGGs.
Collapse
Affiliation(s)
- Matteo Riva
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven 3000, Belgium .,Department of Neurosurgery, Erasme Hospital, Bruxelles 1070, Belgium
| | - Roxanne Wouters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven 3000, Belgium
| | - Akila Weerasekera
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven 3000, Belgium
| | - Sarah Belderbos
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven 3000, Belgium
| | - David Nittner
- Center for the Biology of Disease, KU Leuven Center for Human Genetics - InfraMouse, VIB, University of Leuven, Leuven 3000, Belgium
| | - Dietmar R Thal
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium.,Department of Pathology, UZ-Leuven, Leuven 3000, Belgium
| | - Thaïs Baert
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven 3000, Belgium.,Department of Gynecology and Gynecologic Oncology, Kliniken Essen Mitte (KEM), Essen 2910, Germany
| | - Roberto Giovannoni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
| | - Willy Gsell
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven 3000, Belgium
| | - Uwe Himmelreich
- Biomedical MRI, Department of Imaging and Pathology and Molecular Small Animal Imaging Center (MoSAIC), KU Leuven, Leuven 3000, Belgium
| | - Marc Van Ranst
- Laboratory for Clinical and Epidemiological Virology, Rega Institute for Medical Research, KU Leuven, Leuven 3000, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, KU Leuven, Leuven 3000, Belgium.,Department of Gynaecology and Obstetrics, Leuven Cancer Institute, UZ Leuven, Leuven 3000, Belgium
| |
Collapse
|
43
|
Derouiche A, Geiger KD. Perspectives for Ezrin and Radixin in Astrocytes: Kinases, Functions and Pathology. Int J Mol Sci 2019; 20:ijms20153776. [PMID: 31382374 PMCID: PMC6695708 DOI: 10.3390/ijms20153776] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are increasingly perceived as active partners in physiological brain function and behaviour. The structural correlations of the glia–synaptic interaction are the peripheral astrocyte processes (PAPs), where ezrin and radixin, the two astrocytic members of the ezrin-radixin-moesin (ERM) family of proteins are preferentially localised. While the molecular mechanisms of ERM (in)activation appear universal, at least in mammalian cells, and have been studied in great detail, the actual ezrin and radixin kinases, phosphatases and binding partners appear cell type specific and may be multiplexed within a cell. In astrocytes, ezrin is involved in process motility, which can be stimulated by the neurotransmitter glutamate, through activation of the glial metabotropic glutamate receptors (mGluRs) 3 or 5. However, it has remained open how this mGluR stimulus is transduced to ezrin activation. Knowing upstream signals of ezrin activation, ezrin kinase(s), and membrane-bound binding partners of ezrin in astrocytes might open new approaches to the glial role in brain function. Ezrin has also been implicated in invasive behaviour of astrocytomas, and glial activation. Here, we review data pertaining to potential molecular interaction partners of ezrin in astrocytes, with a focus on PKC and GRK2, and in gliomas and other diseases, to stimulate further research on their potential roles in glia-synaptic physiology and pathology.
Collapse
Affiliation(s)
- Amin Derouiche
- Institute of Anatomy II, Goethe-University Frankfurt, D-60590 Frankfurt am Main, Germany.
| | - Kathrin D Geiger
- Neuropathology, Institute for Pathology, Carl Gustav Carus University Hospital, TU Dresden, D-01307 Dresden, Germany
| |
Collapse
|
44
|
Irani S. Emerging insights into the biology of metastasis: A review article. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:833-847. [PMID: 31579438 PMCID: PMC6760483 DOI: 10.22038/ijbms.2019.32786.7839] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 02/16/2019] [Indexed: 12/12/2022]
Abstract
Metastasis means the dissemination of the cancer cells from one organ to another which is not directly connected to the primary site. Metastasis has a crucial role in the prognosis of cancer patients. A few theories, different types of cell and several molecular pathways have been proposed to explain the mechanism of metastasis. In this work, the related articles in the limited period of time, 2000-mid -2018 were reviewed, through search in PubMed, Google Scholar and Scopus database. The articles published in the last two decades related to the biology of cancer metastasis were selected and the most important factors were discussed. Metastasis is critical factor to predict survival in patients with advanced cancer and prognosis determines the treatment plan. Many different cell types and various signaling pathways control the metastatic process. Metastasis is a multistep process. Many signaling pathways and molecules are involved in metastasis. Increasing knowledge about the mechanism of metastasis can help in finding the promising targets of cancer therapy.
Collapse
Affiliation(s)
- Soussan Irani
- Dental Research Centre, Oral Pathology Department, Dental Faculty, Hamadan University of Medical Sciences, Hamadan,Iran, Lecturer at Griffith University, Gold Coast, Australia
| |
Collapse
|
45
|
Treatment Strategies Based on Histological Targets against Invasive and Resistant Glioblastoma. JOURNAL OF ONCOLOGY 2019; 2019:2964783. [PMID: 31320900 PMCID: PMC6610731 DOI: 10.1155/2019/2964783] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/02/2019] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is the most common and the most malignant primary brain tumor and is characterized by rapid proliferation, invasion into surrounding normal brain tissues, and consequent aberrant vascularization. In these characteristics of GBM, invasive properties are responsible for its recurrence after various therapies. The histomorphological patterns of glioma cell invasion have often been referred to as the “secondary structures of Scherer.” The “secondary structures of Scherer” can be classified mainly into four histological types as (i) perineuronal satellitosis, (ii) perivascular satellitosis, (iii) subpial spread, and (iv) invasion along the white matter tracts. In order to develop therapeutic interventions to mitigate glioma cell migration, it is important to understand the biological mechanism underlying the formation of these secondary structures. The main focus of this review is to examine new molecular pathways based on the histopathological evidence of GBM invasion as major prognostic factors for the high recurrence rate for GBMs. The histopathology-based pharmacological and biological targets for treatment strategies may improve the management of invasive and resistant GBMs.
Collapse
|
46
|
Barbieri F, Verduci I, Carlini V, Zona G, Pagano A, Mazzanti M, Florio T. Repurposed Biguanide Drugs in Glioblastoma Exert Antiproliferative Effects via the Inhibition of Intracellular Chloride Channel 1 Activity. Front Oncol 2019; 9:135. [PMID: 30918838 PMCID: PMC6424887 DOI: 10.3389/fonc.2019.00135] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
The lack of in-depth knowledge about the molecular determinants of glioblastoma (GBM) occurrence and progression, combined with few effective and BBB crossing-targeted compounds represents a major challenge for the discovery of novel and efficacious drugs for GBM. Among relevant molecular factors controlling the aggressive behavior of GBM, chloride intracellular channel 1 (CLIC1) represents an emerging prognostic and predictive biomarker, as well as a promising therapeutic target. CLIC1 is a metamorphic protein, co-existing as both soluble cytoplasmic and membrane-associated conformers, with the latter acting as chloride selective ion channel. CLIC1 is involved in several physiological cell functions and its abnormal expression triggers tumor development, favoring tumor cell proliferation, invasion, and metastasis. CLIC1 overexpression is associated with aggressive features of various human solid tumors, including GBM, in which its expression level is correlated with poor prognosis. Moreover, increasing evidence shows that modification of microglia ion channel activity, and CLIC1 in particular, contributes to the development of different neuropathological states and brain tumors. Intriguingly, CLIC1 is constitutively active within cancer stem cells (CSCs), while it seems less relevant for the survival of non-CSC GBM subpopulations and for normal cells. CSCs represent GBM development and progression driving force, being endowed with stem cell-like properties (self-renewal and differentiation), ability to survive therapies, to expand and differentiate, causing tumor recurrence. Downregulation of CLIC1 results in drastic inhibition of GBM CSC proliferation in vitro and in vivo, making the control of the activity this of channel a possible innovative pharmacological target. Recently, drugs belonging to the biguanide class (including metformin) were reported to selectively inhibit CLIC1 activity in CSCs, impairing their viability and invasiveness, but sparing normal stem cells, thus representing potential novel antitumor drugs with a safe toxicological profile. On these premises, we review the most recent insights into the biological role of CLIC1 as a potential selective pharmacological target in GBM. Moreover, we examine old and new drugs able to functionally target CLIC1 activity, discussing the challenges and potential development of CLIC1-targeted therapies.
Collapse
Affiliation(s)
- Federica Barbieri
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica, Università di Genoa, Genoa, Italy
| | - Ivan Verduci
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Valentina Carlini
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Gianluigi Zona
- Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Università di Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Aldo Pagano
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Dipartimento di Medicina Sperimentale, Università di Genoa, Genoa, Italy
| | - Michele Mazzanti
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan, Italy
| | - Tullio Florio
- Sezione di Farmacologia, Dipartimento di Medicina Interna & Centro di Eccellenza per la Ricerca Biomedica, Università di Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
47
|
Su BC, Pan CY, Chen JY. Antimicrobial Peptide TP4 Induces ROS-Mediated Necrosis by Triggering Mitochondrial Dysfunction in Wild-Type and Mutant p53 Glioblastoma Cells. Cancers (Basel) 2019; 11:cancers11020171. [PMID: 30717309 PMCID: PMC6406555 DOI: 10.3390/cancers11020171] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
Antimicrobial peptide tilapia piscidin 4 (TP4) from Oreochromis niloticus exhibits potent bactericidal and anti-tumorigenic effects. In a variety of cancers, the mutation status of p53 is a decisive factor for therapeutic sensitivity. Therefore, we investigated the impact of p53 status on TP4-induced cytotoxicity in glioblastoma cell lines and the molecular mechanisms that govern cytotoxic effects. Both U87MG (wild-type/WT p53) and U251 (mutant p53) glioblastoma cell lines were sensitive to TP4-induced cytotoxicity. The necrosis inhibitors Necrostatin-1 and GSK’872 attenuated TP4-induced cytotoxicity, and TP4 treatment induced the release of cyclophilin A, a biomarker of necrosis. Moreover, TP4 induced mitochondrial hyperpolarization and dysfunction, which preceded the elevation of intracellular reactive oxygen species, DNA damage, and necrotic cell death in both U87MG and U251 glioblastoma cells. p38 was also activated by TP4, but did not contribute to cytotoxicity. SB202190, a specific p38 inhibitor, enhanced TP4-induced oxidative stress, mitochondrial dysfunction, and cytotoxicity, suggesting a protective role of p38. Furthermore, TP4-induced cytotoxicity, oxidative stress, phosphorylation of p38, and DNA damage were all attenuated by the mitochondrial-targeted reactive oxygen species (ROS) scavenger MitoTEMPO, or the reactive oxygen species scavenger N-acetyl-L-cysteine. Based on these data, we conclude that TP4 induces necrosis in both WT and mutant p53 glioblastoma cells through a mitochondrial ROS-dependent pathway.
Collapse
Affiliation(s)
- Bor-Chyuan Su
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Road, Jiaushi, Ilan 262, Taiwan.
| | - Chieh-Yu Pan
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan.
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Road, Jiaushi, Ilan 262, Taiwan.
| |
Collapse
|
48
|
Geraldo LHM, Garcia C, da Fonseca ACC, Dubois LGF, de Sampaio e Spohr TCL, Matias D, de Camargo Magalhães ES, do Amaral RF, da Rosa BG, Grimaldi I, Leser FS, Janeiro JM, Macharia L, Wanjiru C, Pereira CM, Moura-Neto V, Freitas C, Lima FRS. Glioblastoma Therapy in the Age of Molecular Medicine. Trends Cancer 2019; 5:46-65. [DOI: 10.1016/j.trecan.2018.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022]
|
49
|
Eckert F, Schilbach K, Klumpp L, Bardoscia L, Sezgin EC, Schwab M, Zips D, Huber SM. Potential Role of CXCR4 Targeting in the Context of Radiotherapy and Immunotherapy of Cancer. Front Immunol 2018; 9:3018. [PMID: 30622535 PMCID: PMC6308162 DOI: 10.3389/fimmu.2018.03018] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/06/2018] [Indexed: 12/28/2022] Open
Abstract
Cancer immunotherapy has been established as standard of care in different tumor entities. After the first reports on synergistic effects with radiotherapy and the induction of abscopal effects-tumor shrinkage outside the irradiated volume attributed to immunological effects of radiotherapy-several treatment combinations have been evaluated. Different immunotherapy strategies (e.g., immune checkpoint inhibition, vaccination, cytokine based therapies) have been combined with local tumor irradiation in preclinical models. Clinical trials are ongoing in different cancer entities with a broad range of immunotherapeutics and radiation schedules. SDF-1 (CXCL12)/CXCR4 signaling has been described to play a major role in tumor biology, especially in hypoxia adaptation, metastasis and migration. Local tumor irradiation is a known inducer of SDF-1 expression and release. CXCR4 also plays a major role in immunological processes. CXCR4 antagonists have been approved for the use of hematopoietic stem cell mobilization from the bone marrow. In addition, several groups reported an influence of the SDF-1/CXCR4 axis on intratumoral immune cell subsets and anti-tumor immune response. The aim of this review is to merge the knowledge on the role of SDF-1/CXCR4 in tumor biology, radiotherapy and immunotherapy of cancer and in combinatorial approaches.
Collapse
Affiliation(s)
- Franziska Eckert
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Karin Schilbach
- Department of General Pediatrics/Pediatric Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Lukas Klumpp
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany.,Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Lilia Bardoscia
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany.,Department of Radiation Oncology, University of Brescia, Brescia, Italy
| | - Efe Cumhur Sezgin
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany.,Departments of Clinical Pharmacology, Pharmacy and Biochemistry, University Hospital and University Tuebingen, Tuebingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
50
|
Pišlar A, Jewett A, Kos J. Cysteine cathepsins: Their biological and molecular significance in cancer stem cells. Semin Cancer Biol 2018; 53:168-177. [DOI: 10.1016/j.semcancer.2018.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022]
|