1
|
Sun Q, Xu J, Yuan F, Liu Y, Chen Q, Guo L, Dong H, Liu B. RND1 inhibits epithelial-mesenchymal transition and temozolomide resistance of glioblastoma via AKT/GSK3-β pathway. Cancer Biol Ther 2024; 25:2321770. [PMID: 38444223 PMCID: PMC10936657 DOI: 10.1080/15384047.2024.2321770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 02/18/2024] [Indexed: 03/07/2024] Open
Abstract
GBM is one of the most malignant tumor in central nervous system. The resistance to temozolomide (TMZ) is inevitable in GBM and the characterization of TMZ resistance seriously hinders clinical treatment. It is worthwhile exploring the underlying mechanism of aggressive invasion and TMZ resistance in GBM treatment. Bioinformatic analysis was used to analyze the association between RND1 and a series of EMT-related genes. Colony formation assay and cell viability assay were used to assess the growth of U87 and U251 cells. The cell invasion status was evaluated based on transwell and wound-healing assays. Western blot was used to detect the protein expression in GBM cells. Treatment targeted RND1 combined with TMZ therapy was conducted in nude mice to evaluate the potential application of RND1 as a clinical target for GBM. The overexpression of RND1 suppressed the progression and migration of U87 and U251 cells. RND1 knockdown facilitated the growth and invasion of GBM cells. RND1 regulated the EMT of GBM cells via inhibiting the phosphorylation of AKT and GSK3-β. The promoted effects of RND1 on TMZ sensitivity was identified both in vitro and in vivo. This research demonstrated that the overexpression of RND1 suppressed the migration and EMT status by downregulating AKT/GSK3-β pathway in GBM. RND1 enhanced the TMZ sensitivity of GBM cells both in vitro and in vivo. Our findings may contribute to the targeted therapy for GBM and the understanding of mechanisms of TMZ resistance in GBM.
Collapse
Affiliation(s)
- Qian Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junjie Xu
- Office of director, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Fan’en Yuan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lirui Guo
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huimin Dong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
2
|
Fujii T, Nakano Y, Hagita D, Onishi N, Endo A, Nakagawa M, Yoshiura T, Otsuka Y, Takeuchi S, Suzuki M, Shimizu Y, Toyooka T, Matsushita Y, Hibiya Y, Tomura S, Kondo A, Wada K, Ichimura K, Tomiyama A. KLC1-ROS1 Fusion Exerts Oncogenic Properties of Glioma Cells via Specific Activation of JAK-STAT Pathway. Cancers (Basel) 2023; 16:9. [PMID: 38201436 PMCID: PMC10778328 DOI: 10.3390/cancers16010009] [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: 11/07/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Here, we investigated the detailed molecular oncogenic mechanisms of a novel receptor tyrosine kinase (RTK) fusion, KLC1-ROS1, with an adapter molecule, KLC1, and an RTK, ROS1, discovered in pediatric glioma, and we explored a novel therapeutic target for glioma that possesses oncogenic RTK fusion. When wild-type ROS1 and KLC1-ROS1 fusions were stably expressed in the human glioma cell lines A172 and U343MG, immunoblotting revealed that KLC1-ROS1 fusion specifically activated the JAK2-STAT3 pathway, a major RTK downstream signaling pathway, when compared with wild-type ROS1. Immunoprecipitation of the fractionated cell lysates revealed a more abundant association of the KLC1-ROS1 fusion with JAK2 than that observed for wild-type ROS1 in the cytosolic fraction. A mutagenesis study of the KLC1-ROS1 fusion protein demonstrated the fundamental roles of both the KLC1 and ROS1 domains in the constitutive activation of KLC1-ROS1 fusion. Additionally, in vitro assays demonstrated that KLC1-ROS1 fusion upregulated cell proliferation, invasion, and chemoresistance when compared to wild-type ROS1. Combination treatment with the chemotherapeutic agent temozolomide and an inhibitor of ROS1, JAK2, or a downstream target of STAT3, demonstrated antitumor effects against KLC1-ROS1 fusion-expressing glioma cells. Our results demonstrate that KLC1-ROS1 fusion exerts oncogenic activity through serum-independent constitutive activation, resulting in specific activation of the JAK-STAT pathway. Our data suggested that molecules other than RTKs may serve as novel therapeutic targets for RTK fusion in gliomas.
Collapse
Affiliation(s)
- Takashi Fujii
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Yoshiko Nakano
- Department of Pediatrics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan;
| | - Daichi Hagita
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Nobuyuki Onishi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan;
| | - Arumu Endo
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Masaya Nakagawa
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Toru Yoshiura
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Yohei Otsuka
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Satoru Takeuchi
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Mario Suzuki
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Yuzaburo Shimizu
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Terushige Toyooka
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Yuko Matsushita
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Yuko Hibiya
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Satoshi Tomura
- Division of Traumatology, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan;
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Kojiro Wada
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
| | - Koichi Ichimura
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| | - Arata Tomiyama
- Department of Brain Disease Translational Research, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (T.F.); (D.H.); (Y.M.); (Y.H.); (K.I.)
- Department of Neurosurgery, National Defense Medical College, 3-2 Namiki, Tokorozawa 359-8513, Saitama, Japan; (A.E.); (M.N.); (T.Y.); (Y.O.); (S.T.); (T.T.); (K.W.)
- Department of Neurosurgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (M.S.); (Y.S.); (A.K.)
| |
Collapse
|
3
|
Despotović A, Janjetović K, Zogović N, Tovilović-Kovačević G. Pharmacological Akt and JNK Kinase Inhibitors 10-DEBC and SP600125 Potentiate Anti-Glioblastoma Effect of Menadione and Ascorbic Acid Combination in Human U251 Glioblastoma Cells. Biomedicines 2023; 11:2652. [PMID: 37893026 PMCID: PMC10604608 DOI: 10.3390/biomedicines11102652] [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: 08/17/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most lethal primary brain tumor in adults, characterized by a highly invasive nature and therapy resistance. Combination of menadione and ascorbic acid (AA+MD) exerts strong ROS-mediated anti-GBM activity in vitro. The objective of this study was to improve AA+MD anti-GBM potential by modulating the activity of Akt and c-Jun N-terminal kinase (JNK), molecules with an important role in GBM development. The effects of Akt and JNK modulation on AA+MD toxicity in U251 human glioblastoma cells were assessed by cell viability assays, flow cytometry, RNA interference and plasmid overexpression, and immunoblot analysis. The AA+MD induced severe oxidative stress, an early increase in Akt phosphorylation followed by its strong inhibition, persistent JNK activation, and U251 cell death. Small molecule Akt kinase inhibitor 10-DEBC enhanced, while pharmacological and genetic Akt activation decreased, AA+MD-induced toxicity. The U251 cell death potentiation by 10-DEBC correlated with an increase in the combination-induced autophagic flux and was abolished by genetic autophagy silencing. Additionally, pharmacological JNK inhibitor SP600125 augmented combination toxicity toward U251 cells, an effect linked with increased ROS accumulation. These results indicate that small Akt and JNK kinase inhibitors significantly enhance AA+MD anti-GBM effects by autophagy potentiation and amplifying deleterious ROS levels.
Collapse
Affiliation(s)
- Ana Despotović
- Department of Neurophysiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000 Belgrade, Serbia; (A.D.); (K.J.)
| | - Kristina Janjetović
- Department of Neurophysiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000 Belgrade, Serbia; (A.D.); (K.J.)
| | - Nevena Zogović
- Department of Neurophysiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000 Belgrade, Serbia; (A.D.); (K.J.)
| | - Gordana Tovilović-Kovačević
- Department of Biochemistry, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000 Belgrade, Serbia
| |
Collapse
|
4
|
Whitehead CA, Fang H, Su H, Morokoff AP, Kaye AH, Hanssen E, Nowell CJ, Drummond KJ, Greening DW, Vella LJ, Mantamadiotis T, Stylli SS. Small extracellular vesicles promote invadopodia activity in glioblastoma cells in a therapy-dependent manner. Cell Oncol (Dordr) 2023; 46:909-931. [PMID: 37014551 PMCID: PMC10356899 DOI: 10.1007/s13402-023-00786-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 04/05/2023] Open
Abstract
PURPOSE The therapeutic efficacy of radiotherapy/temozolomide treatment for glioblastoma (GBM) is limited by the augmented invasiveness mediated by invadopodia activity of surviving GBM cells. As yet, however the underlying mechanisms remain poorly understood. Due to their ability to transport oncogenic material between cells, small extracellular vesicles (sEVs) have emerged as key mediators of tumour progression. We hypothesize that the sustained growth and invasion of cancer cells depends on bidirectional sEV-mediated cell-cell communication. METHODS Invadopodia assays and zymography gels were used to examine the invadopodia activity capacity of GBM cells. Differential ultracentrifugation was utilized to isolate sEVs from conditioned medium and proteomic analyses were conducted on both GBM cell lines and their sEVs to determine the cargo present within the sEVs. In addition, the impact of radiotherapy and temozolomide treatment of GBM cells was studied. RESULTS We found that GBM cells form active invadopodia and secrete sEVs containing the matrix metalloproteinase MMP-2. Subsequent proteomic studies revealed the presence of an invadopodia-related protein sEV cargo and that sEVs from highly invadopodia active GBM cells (LN229) increase invadopodia activity in sEV recipient GBM cells. We also found that GBM cells displayed increases in invadopodia activity and sEV secretion post radiation/temozolomide treatment. Together, these data reveal a relationship between invadopodia and sEV composition/secretion/uptake in promoting the invasiveness of GBM cells. CONCLUSIONS Our data indicate that sEVs secreted by GBM cells can facilitate tumour invasion by promoting invadopodia activity in recipient cells, which may be enhanced by treatment with radio-chemotherapy. The transfer of pro-invasive cargos may yield important insights into the functional capacity of sEVs in invadopodia.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Haoyun Fang
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Huaqi Su
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew P Morokoff
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia
| | - Andrew H Kaye
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Eric Hanssen
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Advanced Microscopy Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, 3052, Australia
| | - Katharine J Drummond
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Laura J Vella
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Theo Mantamadiotis
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia.
| |
Collapse
|
5
|
Li J, Ma J, Huang S, Li J, Zhou L, Sun J, Chen L. Circ TTLL13 Promotes TMZ Resistance in Glioma via Modulating OLR1-Mediated Activation of the Wnt/β-Catenin Pathway. Mol Cell Biol 2023; 43:354-369. [PMID: 37427890 PMCID: PMC10348032 DOI: 10.1080/10985549.2023.2210032] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/19/2023] [Indexed: 07/11/2023] Open
Abstract
Glioma, originating from neuroglial progenitor cells, is a type of intrinsic brain tumor with poor prognosis. temozolomide (TMZ) is the first-line chemotherapeutic agent for glioma. Exploring the mechanisms of circTTLL13 underlying TMZ resistance in glioma is of great significance to improve glioma treatment. Bioinformatics was adopted to identify target genes. The circular structure of circTTLL13 and its high expression in glioma cells were disclosed by quantitative real time-PCR (qRT-PCR) and PCR-agarose gel electrophoresis. Functional experiments proved that oxidized LDL receptor 1 (OLR1) promotes TMZ resistance of glioma cells. CircTTLL13 enhances TMZ resistance of glioma cells via modulating OLR1. Luciferase reporter, RNA-binding protein immunoprecipitation (RIP), RNA pulldown, mRNA stability, N6-methyladenosine (m6A) dot blot and RNA total m6A quantification assays were implemented, indicating that circTTLL13 stabilizes OLR1 mRNA via recruiting YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) and promotes m6A methylation of OLR1 pre-mRNA through recruiting methyltransferase-like 3 (METTL3). TOP/FOP-flash reporter assay and western blot verified that circTTLL13 activates Wnt/β-catenin signaling pathway by regulating OLR1. CircTTLL13 promotes TMZ resistance in glioma through regulating OLR1-mediated Wnt/β-catenin pathway activation. This study offers an insight into the efficacy improvement of TMZ for glioma treatment.
Collapse
Affiliation(s)
- Jun Li
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Junfeng Ma
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Shan Huang
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Jun Li
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Liang Zhou
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Jiahua Sun
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Lin Chen
- Department of Neurosurgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Shi Y, Wu M, Liu Y, Hu L, Wu H, Xie L, Liu Z, Wu A, Chen L, Xu C. ITGA5 Predicts Dual-Drug Resistance to Temozolomide and Bevacizumab in Glioma. Front Oncol 2021; 11:769592. [PMID: 34976814 PMCID: PMC8719456 DOI: 10.3389/fonc.2021.769592] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
AIMS Anti-angiotherapy (Bevacizumab) is currently regarded as a promising option for glioma patients who are resistant to temozolomide (TMZ) treatment. But ongoing clinical research failed to meet therapeutic expectations. This study aimed to explore the pivotal genetic feature responsible for TMZ and Bevacizumab resistance in glioma patients. METHODS We downloaded the transcriptomic and methylation data of glioma patients from The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), and Gene Expression Omnibus (GEO) databases and grouped these patients into resistant and non-resistant groups based on their clinical profiles. Differentially expressed genes and pathways were identified and exhibited with software in R platform. A TMZ-resistant cell line was constructed for validating the expression change of the candidate gene, ITGA5. An ITGA5-overexpressing cell line was also constructed to investigate its biological function using the CCK8 assay, Western blot, periodic acid-Schiff (PAS) staining, and transcriptional sequencing. RESULTS Change of the cell morphology and polarity was closely associated with TMZ mono-resistance and TMZ/Bevacizumab dual resistance in glioma patients. The expression level of ITGA5 was effective in determining drug resistance and the outcome of glioma patients, which is regulated by methylation on two distinct sites. ITGA5 was augmented in TMZ-resistant glioma cells, while overexpressing ITGA5 altered the cell-promoted TMZ resistance through enhancing vascular mimicry (VM) formation correspondingly. CONCLUSIONS Both the epigenetic and transcriptional levels of ITGA5 are effective in predicting TMZ and Bevacizumab resistance, indicating that ITGA5 may serve as a predictor of the treatment outcomes of glioma patients.
Collapse
Affiliation(s)
- Ying Shi
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mengwan Wu
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, China
| | - Yuyang Liu
- Chinese People’s Liberation Army (PLA) Institute of Neurosurgery, Chinese PLA General Hospital and PLA Medical College, Beijing, China
| | - Lanlin Hu
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, China
| | - Hong Wu
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, China
| | - Lei Xie
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhiwei Liu
- The Center for Advanced Semiconductor & Integrated Micro-System, University of Electronic Science and Technology of China, Chengdu, China
| | - Anhua Wu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ling Chen
- Chinese People’s Liberation Army (PLA) Institute of Neurosurgery, Chinese PLA General Hospital and PLA Medical College, Beijing, China
| | - Chuan Xu
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, China
| |
Collapse
|
7
|
Yamamuro S, Takahashi M, Satomi K, Sasaki N, Kobayashi T, Uchida E, Kawauchi D, Nakano T, Fujii T, Narita Y, Kondo A, Wada K, Yoshino A, Ichimura K, Tomiyama A. Lomustine and nimustine exert efficient antitumor effects against glioblastoma models with acquired temozolomide resistance. Cancer Sci 2021; 112:4736-4747. [PMID: 34536314 PMCID: PMC8586660 DOI: 10.1111/cas.15141] [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: 05/16/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 12/30/2022] Open
Abstract
Glioblastomas (GBM) often acquire resistance against temozolomide (TMZ) after continuous treatment and recur as TMZ‐resistant GBM (TMZ‐R‐GBM). Lomustine (CCNU) and nimustine (ACNU), which were previously used as standard therapeutic agents against GBM before TMZ, have occasionally been used for the salvage therapy of TMZ‐R‐GBM; however, their efficacy has not yet been thoroughly examined. Therefore, we investigated the antitumor effects of CCNU and ACNU against TMZ‐R‐GBM. As a model of TMZ‐R‐GBM, TMZ resistant clones of human GBM cell lines (U87, U251MG, and U343MG) were established (TMZ‐R‐cells) by the culture of each GBM cells under continuous TMZ treatment, and the antitumor effects of TMZ, CCNU, or ACNU against these cells were analyzed in vitro and in vivo. As a result, although growth arrest and apoptosis were triggered in all TMZ‐R‐cells after the administration of each drug, the antitumor effects of TMZ against TMZ‐R‐cells were significantly reduced compared to those of parental cells, whereas CCNU and ACNU demonstrated efficient antitumor effects on TMZ‐R‐cells as well as parental cells. It was also demonstrated that TMZ resistance of TMZ‐R‐cells was regulated at the initiation of DNA damage response. Furthermore, survival in mice was significantly prolonged by systemic treatment with CCNU or ACNU but not TMZ after implantation of TMZ‐R‐cells. These findings suggest that CCNU or ACNU may serve as a therapeutic agent in salvage treatment against TMZ‐R‐GBM.
Collapse
Affiliation(s)
- Shun Yamamuro
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Neurological Surgery, Nihon University School of Medicine, Itabashi-ku, Japan
| | - Masamichi Takahashi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-ku, Japan
| | - Kaishi Satomi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Diagnostic Pathology, National Cancer Center Hospital, Chuo-ku, Japan
| | - Nobuyoshi Sasaki
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Neurosurgery, Faculty of Medicine, Kyorin University, Mitaka, Japan
| | - Tatsuya Kobayashi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Neurosurgery, Tokyo Women's Medical University, Shinjuku-ku, Japan
| | - Eita Uchida
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka-City, Japan
| | - Daisuke Kawauchi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba-shi, Japan
| | - Tomoyuki Nakano
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Neurosurgery, Tokyo Medical and Dental University, Bunkyo-ku, Japan.,Department of Brain Disease Translational Research, Faculty of Medicine, Juntendo University, Bunkyo-ku, Japan
| | - Takashi Fujii
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Brain Disease Translational Research, Faculty of Medicine, Juntendo University, Bunkyo-ku, Japan.,Department of Neurosurgery, National Defense Medical College, Tokorozawa, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-ku, Japan
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University School of Medicine, Bunkyo-ku, Japan
| | - Kojiro Wada
- Department of Neurosurgery, National Defense Medical College, Tokorozawa, Japan
| | - Atsuo Yoshino
- Department of Neurological Surgery, Nihon University School of Medicine, Itabashi-ku, Japan
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Brain Disease Translational Research, Faculty of Medicine, Juntendo University, Bunkyo-ku, Japan
| | - Arata Tomiyama
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Chuo-ku, Japan.,Department of Brain Disease Translational Research, Faculty of Medicine, Juntendo University, Bunkyo-ku, Japan.,Department of Neurosurgery, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
8
|
Kawauchi D, Takahashi M, Satomi K, Yamamuro S, Kobayashi T, Uchida E, Honda-Kitahara M, Narita Y, Iwadate Y, Ichimura K, Tomiyama A. The ALK inhibitors, alectinib and ceritinib, induce ALK-independent and STAT3-dependent glioblastoma cell death. Cancer Sci 2021; 112:2442-2453. [PMID: 33728771 PMCID: PMC8177803 DOI: 10.1111/cas.14885] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/11/2021] [Accepted: 03/14/2021] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma (GBM) is the most common, but extremely malignant, brain tumor; thus, the development of novel therapeutic strategies for GBMs is imperative. Many tyrosine kinase inhibitors (TKIs) have been approved for various cancers, yet none has demonstrated clinical benefit against GBM. Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) that is confirmed only during the embryonic development period in humans. In addition, various ALK gene alterations are known to act as powerful oncogenes and therapeutic targets in various tumors. The antitumor activity of various TKIs was tested against three human GBM cell lines (U87MG, LN229, and GSC23), which expressed substantially low ALK levels; second‐generation ALK inhibitors, alectinib and ceritinib, effectively induced GBM cell death. In addition, treatment with either alectinib or ceritinib modulated the activation of various molecules downstream of RTK signaling and induced caspase‐dependent/‐independent cell death mainly by inhibiting signal transducer and activator of transcription 3 activation in human GBM cells. In addition, alectinib and ceritinib also showed antitumor activity against a U87MG cell line with acquired temozolomide resistance. Finally, oral administration of alectinib and ceritinib prolonged the survival of mice harboring intracerebral GBM xenografts compared with controls. These results suggested that treatment with the second‐generation ALK inhibitors, alectinib and ceritinib, might serve as a potent therapeutic strategy against GBM.
Collapse
Affiliation(s)
- Daisuke Kawauchi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masamichi Takahashi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kaishi Satomi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Shun Yamamuro
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Tatsuya Kobayashi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Eita Uchida
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka-City, Japan
| | - Mai Honda-Kitahara
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuo Iwadate
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Arata Tomiyama
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery, National Defense Medical College, Tokorozawa, Japan
| |
Collapse
|
9
|
Dekker LJM, Kannegieter NM, Haerkens F, Toth E, Kros JM, Steenhoff Hov DA, Fillebeen J, Verschuren L, Leenstra S, Ressa A, Luider TM. Multiomics profiling of paired primary and recurrent glioblastoma patient tissues. Neurooncol Adv 2020; 2:vdaa083. [PMID: 32793885 PMCID: PMC7415260 DOI: 10.1093/noajnl/vdaa083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Despite maximal therapy with surgery, chemotherapy, and radiotherapy, glioblastoma (GBM) patients have a median survival of only 15 months. Almost all patients inevitably experience symptomatic tumor recurrence. A hallmark of this tumor type is the large heterogeneity between patients and within tumors itself which relates to the failure of standardized tumor treatment. In this study, tissue samples of paired primary and recurrent GBM tumors were investigated to identify individual factors related to tumor progression. Methods Paired primary and recurrent GBM tumor tissues from 8 patients were investigated with a multiomics approach using transcriptomics, proteomics, and phosphoproteomics. Results In the studied patient cohort, large variations between and within patients are observed for all omics analyses. A few pathways affected at the different omics levels partly overlapped if patients are analyzed at the individual level, such as synaptogenesis (containing the SNARE complex) and cholesterol metabolism. Phosphoproteomics revealed increased STMN1(S38) phosphorylation as part of ERBB4 signaling. A pathway tool has been developed to visualize and compare different omics datasets per patient and showed potential therapeutic drugs, such as abobotulinumtoxinA (synaptogenesis) and afatinib (ERBB4 signaling). Afatinib is currently in clinical trials for GBM. Conclusions A large variation on all omics levels exists between and within GBM patients. Therefore, it will be rather unlikely to find a drug treatment that would fit all patients. Instead, a multiomics approach offers the potential to identify affected pathways on the individual patient level and select treatment options.
Collapse
Affiliation(s)
- Lennard J M Dekker
- Department of Neurology, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | | | - Emma Toth
- Department of Neurology, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Johan M Kros
- Department of Pathology, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | | | - Lars Verschuren
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Sieger Leenstra
- Department of Neurosurgery, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | - Theo M Luider
- Department of Neurology, Erasmus University Medical Centre Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
10
|
Identification of Key Pathways and Genes in the Orai2 Mediated Classical and Mesenchymal Subtype of Glioblastoma by Bioinformatic Analyses. DISEASE MARKERS 2019; 2019:7049294. [PMID: 31772693 PMCID: PMC6855003 DOI: 10.1155/2019/7049294] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/27/2019] [Accepted: 08/14/2019] [Indexed: 01/11/2023]
Abstract
Background Ca2+ release-activated Ca2+ channels (CRAC) are the main Ca2+ entry pathway regulating intracellular Ca2+ concentration in a variety of cancer types. Orai2 is the main pore-forming subunit of CRAC channels in central neurons. To explore the role of Orai2 in glioblastoma (GBM), we investigated the key pathways and genes in Orai2-mediated GBM by bioinformatic analyses. Methods Via The Cancer Genome Atlas (TCGA), French, Sun, and Gene Expression Omnibus (GEO) (GDS3885) datasets, we collected 1231 cases with RNA-seq data and analyzed the functional annotation of Orai2 by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Univariate and multivariate survival analyses were applied to 823 patients with survival data. Results We discovered that Orai2 was markedly upregulated in GBM compared to normal brain samples and lower-grade gliomas (LGG). Survival analysis found that higher expression of Orai2 was independently associated with a worse prognosis of patients with the classical and mesenchymal subtypes of GBM. Simultaneously, Orai2 expression was higher in tumors of the classical and mesenchymal subtypes than other subtypes and was significantly correlated with classical- and mesenchymal-related genes. GO and KEGG pathway analysis revealed that genes significantly correlated with Orai2 were involved in the JNK pathway. Through screening transcriptomic data, we found a strong association between Orai2 and apoptosis, stemness, and an epithelial-mesenchymal transition- (EMT-) like phenotype. Conclusion As a prognostic factor, Orai2 is obviously activated in the classical and mesenchymal subtypes of GBM and promotes glioma cell self-renewal, apoptosis, and EMT-like by the JNK pathway. These findings indicate that Orai2 could be a candidate prognostic and therapeutic target, especially for the classical and mesenchymal subtypes of GBM.
Collapse
|
11
|
Signal transduction pathways and resistance to targeted therapies in glioma. Semin Cancer Biol 2019; 58:118-129. [PMID: 30685341 DOI: 10.1016/j.semcancer.2019.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023]
Abstract
Although surgical techniques and adjuvant therapies have undergone progressive development for decades, the therapeutic outcomes for treating glioblastoma (GBM) remain poor. The main reasons for the poor prognosis of gliomas are that limited tumor tissue that can be resected (to preserve brain functions) and that residual tumors are often resistant to irradiation and chemotherapy. Therefore, overcoming the resistance of residual tumors against adjuvant therapy is urgently needed for glioma treatment. Recent large cohort studies of genetic alterations in GBM demonstrated that both genetic information and intracellular molecular signaling are networked in gliomas and that such information may help clarify which molecules or signals serve essential roles in resistance against radiation or chemotherapy, highlighting them as potential novel therapeutic targets against refractory gliomas. In this review, we summarize the current understanding of molecular networks that govern glioma biology, mainly based on cohort studies or recent evidence, with a focus on how intracellular signaling molecules in gliomas associate with each other and regulate refractoriness against current therapy.
Collapse
|
12
|
Latré De Laté P, Haidar M, Ansari H, Tajeri S, Szarka E, Alexa A, Woods K, Reményi A, Pain A, Langsley G. Theileria highjacks JNK2 into a complex with the macroschizont GPI (GlycosylPhosphatidylInositol)-anchored surface protein p104. Cell Microbiol 2018; 21:e12973. [PMID: 30412643 DOI: 10.1111/cmi.12973] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/23/2018] [Accepted: 11/06/2018] [Indexed: 12/15/2022]
Abstract
Constitutive c-Jun N-terminal kinase (JNK) activity characterizes bovine T and B cells infected with Theileria parva, and B cells and macrophages infected with Theileria annulata. Here, we show that T. annulata infection of macrophages manipulates JNK activation by recruiting JNK2 and not JNK1 to the parasite surface, whereas JNK1 is found predominantly in the host cell nucleus. At the parasite's surface, JNK2 forms a complex with p104, a GPI-(GlycosylPhosphatidylInositol)-anchor T. annulata plasma membrane protein. Sequestration of JNK2 depended on Protein Kinase-A (PKA)-mediated phosphorylation of a JNK-binding motif common to T. parva and a cell penetrating peptide harbouring the conserved p104 JNK-binding motif competitively ablated binding, whereupon liberated JNK2 became ubiquitinated and degraded. Cytosolic sequestration of JNK2 suppressed small mitochondrial ARF-mediated autophagy, whereas it sustained nuclear JNK1 levels, c-Jun phosphorylation, and matrigel traversal. Therefore, T. annulata sequestration of JNK2 contributes to both survival and dissemination of Theileria-transformed macrophages.
Collapse
Affiliation(s)
- Perle Latré De Laté
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014, France.,Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France
| | - Malak Haidar
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014, France.,Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France.,Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Jeddah, 23955-6900, Kingdom of Saudi Arabia
| | - Hifzur Ansari
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Jeddah, 23955-6900, Kingdom of Saudi Arabia
| | - Shahin Tajeri
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014, France.,Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France
| | - Eszter Szarka
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anita Alexa
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Kerry Woods
- Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Attila Reményi
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Arnab Pain
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Jeddah, 23955-6900, Kingdom of Saudi Arabia.,Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Gordon Langsley
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014, France.,Inserm U1016, CNRS UMR8104, Cochin Institute, Paris, France
| |
Collapse
|
13
|
Yamamoto Y, Sasaki N, Kumagai K, Takeuchi S, Toyooka T, Otani N, Wada K, Narita Y, Ichimura K, Namba H, Mori K, Tomiyama A. Involvement of Intracellular Cholesterol in Temozolomide-Induced Glioblastoma Cell Death. Neurol Med Chir (Tokyo) 2018; 58:296-302. [PMID: 29899179 PMCID: PMC6048351 DOI: 10.2176/nmc.ra.2018-0040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma (GBM) still carries a poor prognosis due to the refractoriness against antitumor drugs. Temozolomide (TMZ), one of the few standard therapy drugs against GBM worldwide, has only limited effect due to acquired TMZ resistance of GBM. Therefore, development of novel therapeutic methods to overcome the TMZ resistance of GBM is urgent. The brain is the most cholesterol-rich organ in the human body, so modulation of cholesterol in tumor cells originating from the brain including GBM may be a tumor-specific therapeutic strategy including enhancement of TMZ effects. The unique lipid metabolism of glioma has recently been reported, but the involvement of intracellular cholesterol in TMZ therapy is yet to be fully elucidated. This review summarizes the effect of modulation of intracellular cholesterol level on cancer therapy including GBM treatment and the implications for TMZ therapy. Our recent findings about the involvement of intracellular cholesterol in TMZ-induced GBM cell death are described.
Collapse
Affiliation(s)
- Yutaro Yamamoto
- Department of Neurosurgery, National Defense Medical College.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| | - Nobuyoshi Sasaki
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute.,Department of Neurosurgery, Kyorin University School of Medicine
| | - Kosuke Kumagai
- Department of Neurosurgery, National Defense Medical College
| | - Satoru Takeuchi
- Department of Neurosurgery, National Defense Medical College
| | | | - Naoki Otani
- Department of Neurosurgery, National Defense Medical College
| | - Kojiro Wada
- Department of Neurosurgery, National Defense Medical College
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| | - Hiroki Namba
- Department of Neurosurgery, Hamamatsu University School of Medicine
| | - Kentaro Mori
- Department of Neurosurgery, National Defense Medical College
| | - Arata Tomiyama
- Department of Neurosurgery, National Defense Medical College.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute
| |
Collapse
|
14
|
Meirson T, Gil-Henn H. Targeting invadopodia for blocking breast cancer metastasis. Drug Resist Updat 2018; 39:1-17. [PMID: 30075834 DOI: 10.1016/j.drup.2018.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/04/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
Dissemination of cancer cells from the primary tumor and their spread to distant sites of the body is the leading cause of mortality in metastatic cancer patients. Metastatic cancer cells invade surrounding tissues and blood vessels by forming F-actin-rich protrusions known as invadopodia, which degrade the extracellular matrix and enable invasion of tumor cells through it. Invadopodia have now been observed in vivo, and recent evidence demonstrates direct molecular links between assembly of invadopodia and cancer metastasis in both mouse models and in human patients. While significant progress has been achieved in the last decade in understanding the molecular mechanisms and signaling pathways regulating invadopodia formation and function, the application of this knowledge to development of prognostic and therapeutic approaches for cancer metastasis has not been discussed before. Here, we provide a detailed overview of current prognostic markers and tests for cancer metastasis and discuss their advantages, disadvantages, and their predicted efficiency. Using bioinformatic patient database analysis, we demonstrate, for the first time, a significant correlation between invadopodia-associated genes to breast cancer metastasis, suggesting that invadopodia could be used as both a prognostic marker and as a therapeutic target for blocking cancer metastasis. We include here a novel network interaction map of invadopodia-associated proteins with currently available inhibitors, demonstrating a central role for the recently identified EGFR-Pyk2-Src-Arg-cortactin invadopodial pathway, to which re-purposing of existent inhibitors could be used to block breast cancer metastasis. We then present an updated overview of current cancer-related clinical trials, demonstrating the negligible number of trials focusing on cancer metastasis. We also discuss the difficulties and complexity of performing cancer metastasis clinical trials, and the possible development of anti-metastasis drug resistance when using a prolonged preventive treatment with invadopodia inhibitors. This review presents a new perspective on invadopodia-mediated tumor invasiveness and may lead to the development of novel prognostic and therapeutic approaches for cancer metastasis.
Collapse
Affiliation(s)
- Tomer Meirson
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel.
| |
Collapse
|
15
|
Abstract
Paxillin is a group III LIM domain protein that is best characterized as a cytoplasmic scaffold/adaptor protein that functions primarily as a mediator of focal adhesion. However, emerging studies indicate that paxillin's functions are far broader. Not only does paxillin appear to regulate cytoplasmic kinase signaling, but it also cycles between the cytoplasm and nucleus, and may serve as an important regulator of mRNA trafficking and subsequent translation. Herein, we provide some insights suggesting that paxillin, like its relative Hic-5, has nuclear binding partners and mediates critical processes within the nucleus, at least in part functioning as coregulator of nuclear receptors and nuclear kinases to mediate genomic signaling.
Collapse
Affiliation(s)
- Xiaoting Ma
- Department of Medicine, Division of Endocrinology and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States; Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States.
| | - Stephen R Hammes
- Department of Medicine, Division of Endocrinology and Metabolism, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States; Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States.
| |
Collapse
|
16
|
Ferrer VP, Moura Neto V, Mentlein R. Glioma infiltration and extracellular matrix: key players and modulators. Glia 2018; 66:1542-1565. [DOI: 10.1002/glia.23309] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Rolf Mentlein
- Department of Anatomy; University of Kiel; Kiel Germany
| |
Collapse
|
17
|
De Luca A, Carpanese D, Rapanotti MC, Viguria TMS, Forgione MA, Rotili D, Fulci C, Iorio E, Quintieri L, Chimenti S, Bianchi L, Rosato A, Caccuri AM. The nitrobenzoxadiazole derivative MC3181 blocks melanoma invasion and metastasis. Oncotarget 2017; 8:15520-15538. [PMID: 28107182 PMCID: PMC5362503 DOI: 10.18632/oncotarget.14690] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/27/2016] [Indexed: 02/01/2023] Open
Abstract
The novel nitrobenzoxadiazole (NBD) derivative MC3181 is endowed with remarkable therapeutic activity in mice bearing both sensitive and vemurafenib-resistant human melanoma xenografts. Here, we report that subtoxic concentrations of this compound significantly reduced invasiveness of BRAF-V600D mutated WM115 and WM266.4 melanoma cell lines derived from the primary lesion and related skin metastasis of the same patient, respectively. The strong antimetastatic activity of MC3181 was observed in both 2D monolayer cultures and 3D multicellular tumor spheroids, and confirmed in vivo by the significant decrease in the number of B16-F10 melanoma lung metastases in drug-treated mice. Our data also show that MC3181 affects the lactate production in the high glycolytic WM266.4 cell line. To unveil the MC3181 mechanism of action, we analyzed the ability of MC3181 to affect the degree of activation of different MAPK pathways, as well as the expression/activity levels of several proteins involved in angiogenesis, invasion, and survival (i.e. AP2, MCAM/MUC18, N-cadherin, VEGF and MMP-2). Our data disclosed both a decrease of the phospho-active form of JNK and an increased expression of the transcription factor AP2, events that occur in the very early phase of drug treatment and may be responsible of the antimetastatic effects of MC3181.
Collapse
Affiliation(s)
- Anastasia De Luca
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| | - Debora Carpanese
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| | | | | | | | - Dante Rotili
- Department of Drug Chemistry and Technologies, "Sapienza" University, 00185 Rome, Italy
| | - Chiara Fulci
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| | - Egidio Iorio
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Luigi Quintieri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Sergio Chimenti
- Department of Dermatology, University of Tor Vergata, 00133 Rome, Italy
| | - Luca Bianchi
- Department of Dermatology, University of Tor Vergata, 00133 Rome, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy.,Istituto Oncologico Veneto IOV-IRCCS, 35128 Padova, Italy
| | - Anna Maria Caccuri
- Department of Experimental Medicine and Surgery, University of Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
18
|
Sun LH, Yang FQ, Zhang CB, Wu YP, Liang JS, Jin S, Wang Z, Wang HJ, Bao ZS, Yang ZX, Jiang T. Overexpression of Paxillin Correlates with Tumor Progression and Predicts Poor Survival in Glioblastoma. CNS Neurosci Ther 2016; 23:69-75. [PMID: 27637748 DOI: 10.1111/cns.12606] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 07/25/2016] [Accepted: 08/11/2016] [Indexed: 12/22/2022] Open
Abstract
AIMS To explore the prognostic and clinicopathological features of glioma with Paxillin (PXN) expression based on a large number of samples. METHODS RNA sequencing data of 325 glioma samples from Chinese Glioma Genome Atlas (CGGA) database were obtained as discovery set. Three additional datasets were further obtained as validation sets. The protein expression pattern of PXN in glioma was measured by IHC. Kaplan-Meier survival and multivariate Cox analysis were used to estimate the survival distributions. Moreover, the functional annotation of PXN was also analyzed. RESULTS In the discovery set, PXN overexpression was significantly associated with high-grade glioma as well as the higher mortality in survival analysis (log-rank test, P < 0.01). The results of the other validation datasets showed similar findings. PXN also served as an independent prognostic biomarker in glioblastoma patients. Functional assays showed that PXN contributed to glioma cell proliferation and invasion. CONCLUSION PXN plays as an oncogene in glioma progression and suggests a new potential biotarget for therapy.
Collapse
Affiliation(s)
- Li-Hua Sun
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Fu-Qiang Yang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chuan-Bao Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yi-Ping Wu
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Jing-Shan Liang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shuai Jin
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Hong-Jun Wang
- Department of Neurosurgery, 2nd affiliated hospital of Harbin Medical University, Harbin, China
| | - Zhao-Shi Bao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zheng-Xiang Yang
- Department of Neurosurgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
19
|
Abstract
Temozolomide (TMZ) is an oral alkylating agent used to treat glioblastoma multiforme (GBM) and astrocytomas. However, at least 50% of TMZ treated patients do not respond to TMZ. This is due primarily to the over-expression of O6-methylguanine methyltransferase (MGMT) and/or lack of a DNA repair pathway in GBM cells. Multiple GBM cell lines are known to contain TMZ resistant cells and several acquired TMZ resistant GBM cell lines have been developed for use in experiments designed to define the mechanism of TMZ resistance and the testing of potential therapeutics. However, the characteristics of intrinsic and adaptive TMZ resistant GBM cells have not been systemically compared. This article reviews the characteristics and mechanisms of TMZ resistance in natural and adapted TMZ resistant GBM cell lines. It also summarizes potential treatment options for TMZ resistant GBMs.
Collapse
Key Words
- AGT (also known as MGMT), O6-methylguanine-DNA alkyltransferase
- AP-1, activator protein 1
- APE1, apurinic/apyrimidine endonuclease/redox factor-1
- APNG, Alkylpurine-DNA-N-glycosylase
- Adaptive
- BBB, blood-brain-barrier
- BCRP1, breast cancer resistance protein 1
- BER, base excision repair
- BG, benzylguanine
- C8orf4, Chromosome 8 open reading frame 4
- EGFR, epidermal growth factor receptor
- ERK1/2, Extracellular Signal Regulated Kinases 1 and 2
- FDA, Food and Drug Administration
- GBM, glioblastoma multiforme or glioblastoma
- Glioblastoma
- HDAC, histone deacetylase
- IFN-β, Interferon-β
- Intrinsic
- JNK, Jun N-terminal kinase
- KDM, Histone lysine demethylase
- LC50, 50% cell death concentration
- LIF, Leukemia inhibitory factor
- MGMT, O6-methylguanine methyltransferase
- MMR, DNA mismatch repair
- MSH6, mutS homolog 6
- MTIC, 5-(3-methyltriazen-1-yl) imidazole-4-carboxamide
- NAMPT, nicotinamide phosphoribosyl transferase
- NF-κB, nuclear factor-Kappa B
- NHA, normal human astrocytes
- PARP, poly ADP ribose polymerase
- Resistance
- SAHA, N-hydroxy-N′-phenyl-octanediamide
- STAT3, Signal Transducer and Activator of Transcription 3
- TMZ, Temozolomide
- TNFAIP3, Tumor necrosis factor-α-induced protein 3
- Temodar
- Temozolomide
- VPA, Valproic acid
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Sang Y Lee
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|