1
|
Jing Y, Kobayashi M, Shoulkamy MI, Zhou M, Thi Vu H, Arakawa H, Sabit H, Iwabuchi S, Quang Vu C, Kasahara A, Ueno M, Tadokoro Y, Kurayoshi K, Chen X, Yan Y, Arai S, Hashimoto S, Soga T, Todo T, Nakada M, Hirao A. Lysine-arginine imbalance overcomes therapeutic tolerance governed by the transcription factor E3-lysosome axis in glioblastoma. Nat Commun 2025; 16:2876. [PMID: 40169552 PMCID: PMC11962137 DOI: 10.1038/s41467-025-56946-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Recent advances in cancer therapy have underscored the importance of targeting specific metabolic pathways. In this study, we propose a precision nutrition approach aimed at lysosomal function in glioblastoma multiforme (GBM). Using patient-derived GBM cells, we identify lysosomal activity as a unique metabolic biomarker of tumorigenesis, controlling the efficacy of temozolomide (TMZ), a standard GBM therapy. Employing combined analyses of clinical patient samples and xenograft models, we further elucidate the pivotal role of Transcription Factor Binding To IGHM Enhancer 3 (TFE3), a master regulator of lysosomal biogenesis, in modulating malignant properties, particularly TMZ tolerance, by regulating peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1α)-mediated mitochondrial activity. Notably, we find that lysine protects GBM cells from lysosomal stress by counteracting arginine's effects on nitric oxide production. The lysine restriction mimetic, homoarginine administration, significantly enhances the efficacy of anticancer therapies through lysosomal dysfunction. This study underscores the critical role of lysosomal function modulated by amino acid metabolism in GBM pathogenesis and treatment.
Collapse
Affiliation(s)
- Yongwei Jing
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Masahiko Kobayashi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Mahmoud I Shoulkamy
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
- Department of Zoology, Faculty of Science, Minia University, Minia, Egypt
| | - Meiqi Zhou
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Ha Thi Vu
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
- Department of Medical Biology and Genetics, Hanoi Medical University, Ha Noi, Vietnam
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hemragul Sabit
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Cong Quang Vu
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Atsuko Kasahara
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Masaya Ueno
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuko Tadokoro
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kenta Kurayoshi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Xi Chen
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuhang Yan
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Satoshi Arai
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo, Japan
| | - Tomoki Todo
- Division of Innovative Cancer Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan.
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa, Ishikawa, Japan.
| |
Collapse
|
2
|
Amaro F, Carvalho M, Bastos MDL, Guedes de Pinho P, Pinto J. Metabolomics Reveals Tyrosine Kinase Inhibitor Resistance-Associated Metabolic Events in Human Metastatic Renal Cancer Cells. Int J Mol Sci 2024; 25:6328. [PMID: 38928035 PMCID: PMC11204329 DOI: 10.3390/ijms25126328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The development of resistance to tyrosine kinase inhibitors (TKIs) is a major cause of treatment failure in metastatic renal cell carcinoma (mRCC). A deeper understanding of the metabolic mechanisms associated with TKI resistance is critical for refining therapeutic strategies. In this study, we established resistance to sunitinib and pazopanib by exposing a parental Caki-1 cell line to increasing concentrations of sunitinib and pazopanib. The intracellular and extracellular metabolome of sunitinib- and pazopanib-resistant mRCC cells were investigated using a nuclear magnetic resonance (NMR)-based metabolomics approach. Data analysis included multivariate and univariate methods, as well as pathway and network analyses. Distinct metabolic signatures in sunitinib- and pazopanib-resistant RCC cells were found for the first time in this study. A common metabolic reprogramming pattern was observed in amino acid, glycerophospholipid, and nicotinate and nicotinamide metabolism. Sunitinib-resistant cells exhibited marked alterations in metabolites involved in antioxidant defence mechanisms, while pazopanib-resistant cells showed alterations in metabolites associated with energy pathways. Sunitinib-resistant RCC cells demonstrated an increased ability to proliferate, whereas pazopanib-resistant cells appeared to restructure their energy metabolism and undergo alterations in pathways associated with cell death. These findings provide potential targets for novel therapeutic strategies to overcome TKI resistance in mRCC through metabolic regulation.
Collapse
Affiliation(s)
- Filipa Amaro
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Márcia Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- RISE-UFP, Health Research Network, Faculty of Health Sciences, University Fernando Pessoa, 4200-150 Porto, Portugal
| | - Maria de Lourdes Bastos
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana Pinto
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; (M.C.); (M.d.L.B.); (P.G.d.P.)
- UCIBIO-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
3
|
Cela I, Capone E, Trevisi G, Sala G. Extracellular vesicles in glioblastoma: Biomarkers and therapeutic tools. Semin Cancer Biol 2024; 101:25-43. [PMID: 38754752 DOI: 10.1016/j.semcancer.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/19/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024]
Abstract
Glioblastoma (GBM) is the most aggressive tumor among the gliomas and intracranial tumors and to date prognosis for GBM patients remains poor, with a median survival typically measured in months to a few years depending on various factors. Although standardized therapies are routinely employed, it is clear that these strategies are unable to cope with heterogeneity and invasiveness of GBM. Furthermore, diagnosis and monitoring of responses to therapies are directly dependent on tissue biopsies or magnetic resonance imaging (MRI) techniques. From this point of view, liquid biopsies are arising as key sources of a variety of biomarkers with the advantage of being easily accessible and monitorable. In this context, extracellular vesicles (EVs), physiologically shed into body fluids by virtually all cells, are gaining increasing interest both as natural carriers of biomarkers and as specific signatures even for GBM. What makes these vesicles particularly attractive is they are also emerging as therapeutical vehicles to treat GBM given their native ability to cross the blood-brain barrier (BBB). Here, we reviewed recent advances on the use of EVs as biomarker for liquid biopsy and nanocarriers for targeted delivery of anticancer drugs in glioblastoma.
Collapse
Affiliation(s)
- Ilaria Cela
- Department of Innovative Technologies in Medicine & Dentistry, University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Emily Capone
- Department of Innovative Technologies in Medicine & Dentistry, University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Gianluca Trevisi
- Department of Neurosciences, Imaging and Clinical Sciences, "G. D'Annunzio" University, Chieti, Italy; Neurosurgical Unit, Santo Spirito Hospital, Pescara 65121, Italy
| | - Gianluca Sala
- Department of Innovative Technologies in Medicine & Dentistry, University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), University "G. D'Annunzio" of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
4
|
Li D, Yan J, Li K, Yang Q, Bian L, Lin B, Liu X, Xi Z. Identification of potential glioma drug resistance target proteins based on ultra-performance liquid chromatography-mass spectrometry differential proteomics. PeerJ 2023; 11:e16426. [PMID: 38054015 PMCID: PMC10695112 DOI: 10.7717/peerj.16426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 10/18/2023] [Indexed: 12/07/2023] Open
Abstract
In this study, to screen for candidate markers of temozolomide (TMZ) resistance in glioblastoma, we artificially established TMZ drug-resistant glioblastoma (GBM) cell lines, U251-TMZ and U87-TMZ. In the U251-TMZ and U87-TMZ cell lines, we screened and analyzed differentially expressed proteins using ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) differential proteomics. Compared with the U251 and U87 control cell lines, 95 differential proteins were screened in the U251-TMZ and U87-TMZ cell lines, of which 28 proteins were upregulated and 67 proteins were down-regulated. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of the co-upregulated proteins showed that most of the differentially expressed proteins were located in the cytoplasm and were significantly upregulated in the biological processes related to vesicular transport in the intimal system and inflammatory response mediated by myeloid leukocytes. Seven candidates were identified as potential GBM markers of TMZ resistance. Combined with existing research findings, our study supports that UAP1L1 and BCKDK are promising potential markers of TMZ resistance in GBM. This is important for further understanding the molecular mechanisms that drive the development and enhancement of TMZ resistance.
Collapse
Affiliation(s)
- Da Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jun Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Qingcheng Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Liping Bian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiaohua Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| |
Collapse
|
5
|
Ahmed M, Semreen AM, El-Huneidi W, Bustanji Y, Abu-Gharbieh E, Alqudah MAY, Alhusban A, Shara M, Abuhelwa AY, Soares NC, Semreen MH, Alzoubi KH. Preclinical and Clinical Applications of Metabolomics and Proteomics in Glioblastoma Research. Int J Mol Sci 2022; 24:ijms24010348. [PMID: 36613792 PMCID: PMC9820403 DOI: 10.3390/ijms24010348] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Glioblastoma (GB) is a primary malignancy of the central nervous system that is classified by the WHO as a grade IV astrocytoma. Despite decades of research, several aspects about the biology of GB are still unclear. Its pathogenesis and resistance mechanisms are poorly understood, and methods to optimize patient diagnosis and prognosis remain a bottle neck owing to the heterogeneity of the malignancy. The field of omics has recently gained traction, as it can aid in understanding the dynamic spatiotemporal regulatory network of enzymes and metabolites that allows cancer cells to adjust to their surroundings to promote tumor development. In combination with other omics techniques, proteomic and metabolomic investigations, which are a potent means for examining a variety of metabolic enzymes as well as intermediate metabolites, might offer crucial information in this area. Therefore, this review intends to stress the major contribution these tools have made in GB clinical and preclinical research and highlights the crucial impacts made by the integrative "omics" approach in reducing some of the therapeutic challenges associated with GB research and treatment. Thus, our study can purvey the use of these powerful tools in research by serving as a hub that particularly summarizes studies employing metabolomics and proteomics in the realm of GB diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Munazza Ahmed
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahlam M. Semreen
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Yasser Bustanji
- Department of Basic and Clinical Pharmacology, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Eman Abu-Gharbieh
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad A. Y. Alqudah
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ahmed Alhusban
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohd Shara
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahmad Y. Abuhelwa
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence: (M.H.S.); (K.H.A.)
| | - Karem H. Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence: (M.H.S.); (K.H.A.)
| |
Collapse
|
6
|
Abdul Rashid K, Ibrahim K, Wong JHD, Mohd Ramli N. Lipid Alterations in Glioma: A Systematic Review. Metabolites 2022; 12:metabo12121280. [PMID: 36557318 PMCID: PMC9783089 DOI: 10.3390/metabo12121280] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Gliomas are highly lethal tumours characterised by heterogeneous molecular features, producing various metabolic phenotypes leading to therapeutic resistance. Lipid metabolism reprogramming is predominant and has contributed to the metabolic plasticity in glioma. This systematic review aims to discover lipids alteration and their biological roles in glioma and the identification of potential lipids biomarker. This systematic review was conducted using the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Extensive research articles search for the last 10 years, from 2011 to 2021, were conducted using four electronic databases, including PubMed, Web of Science, CINAHL and ScienceDirect. A total of 158 research articles were included in this study. All studies reported significant lipid alteration between glioma and control groups, impacting glioma cell growth, proliferation, drug resistance, patients' survival and metastasis. Different lipids demonstrated different biological roles, either beneficial or detrimental effects on glioma. Notably, prostaglandin (PGE2), triacylglycerol (TG), phosphatidylcholine (PC), and sphingosine-1-phosphate play significant roles in glioma development. Conversely, the most prominent anti-carcinogenic lipids include docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and vitamin D3 have been reported to have detrimental effects on glioma cells. Furthermore, high lipid signals were detected at 0.9 and 1.3 ppm in high-grade glioma relative to low-grade glioma. This evidence shows that lipid metabolisms were significantly dysregulated in glioma. Concurrent with this knowledge, the discovery of specific lipid classes altered in glioma will accelerate the development of potential lipid biomarkers and enhance future glioma therapeutics.
Collapse
Affiliation(s)
- Khairunnisa Abdul Rashid
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Kamariah Ibrahim
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Jeannie Hsiu Ding Wong
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Norlisah Mohd Ramli
- Department of Biomedical Imaging, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence: ; Tel.: +60-379673238
| |
Collapse
|
7
|
BET protein inhibition sensitizes glioblastoma cells to temozolomide treatment by attenuating MGMT expression. Cell Death Dis 2022; 13:1037. [PMID: 36513631 PMCID: PMC9747918 DOI: 10.1038/s41419-022-05497-y] [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: 07/06/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Bromodomain and extra-terminal tail (BET) proteins have been identified as potential epigenetic targets in cancer, including glioblastoma. These epigenetic modifiers link the histone code to gene transcription that can be disrupted with small molecule BET inhibitors (BETi). With the aim of developing rational combination treatments for glioblastoma, we analyzed BETi-induced differential gene expression in glioblastoma derived-spheres, and identified 6 distinct response patterns. To uncover emerging actionable vulnerabilities that can be targeted with a second drug, we extracted the 169 significantly disturbed DNA Damage Response genes and inspected their response pattern. The most prominent candidate with consistent downregulation, was the O-6-methylguanine-DNA methyltransferase (MGMT) gene, a known resistance factor for alkylating agent therapy in glioblastoma. BETi not only reduced MGMT expression in GBM cells, but also inhibited its induction, typically observed upon temozolomide treatment. To determine the potential clinical relevance, we evaluated the specificity of the effect on MGMT expression and MGMT mediated treatment resistance to temozolomide. BETi-mediated attenuation of MGMT expression was associated with reduction of BRD4- and Pol II-binding at the MGMT promoter. On the functional level, we demonstrated that ectopic expression of MGMT under an unrelated promoter was not affected by BETi, while under the same conditions, pharmacologic inhibition of MGMT restored the sensitivity to temozolomide, reflected in an increased level of γ-H2AX, a proxy for DNA double-strand breaks. Importantly, expression of MSH6 and MSH2, which are required for sensitivity to unrepaired O6-methylguanine-lesions, was only briefly affected by BETi. Taken together, the addition of BET-inhibitors to the current standard of care, comprising temozolomide treatment, may sensitize the 50% of patients whose glioblastoma exert an unmethylated MGMT promoter.
Collapse
|
8
|
Semreen AM, Alsoud LO, El-Huneidi W, Ahmed M, Bustanji Y, Abu-Gharbieh E, El-Awady R, Ramadan WS, Alqudah MA, Shara M, Abuhelwa AY, Soares NC, Semreen MH, Alzoubi KH. Metabolomics Analysis Revealed Significant Metabolic Changes in Brain Cancer Cells Treated with Paclitaxel and/or Etoposide. Int J Mol Sci 2022; 23:13940. [PMID: 36430415 PMCID: PMC9693830 DOI: 10.3390/ijms232213940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer of the central nervous system (CNS) is ranked as the 19th most prevalent form of the disease in 2020. This study aims to identify candidate biomarkers and metabolic pathways affected by paclitaxel and etoposide, which serve as potential treatments for glioblastoma, and are linked to the pathogenesis of glioblastoma. We utilized an untargeted metabolomics approach using the highly sensitive ultra-high-performance liquid chromatography-electrospray ionization quadrupole time-of-flight mass spectrometry (UHPLC-ESI-QTOF-MS) for identification. In this study, 92 and 94 metabolites in U87 and U373 cell lines were profiled, respectively. The produced metabolites were then analyzed utilizing t-tests, volcano plots, and enrichment analysis modules. Our analysis revealed distinct metabolites to be significantly dysregulated (nutriacholic acid, L-phenylalanine, L-arginine, guanosine, ADP, hypoxanthine, and guanine), and to a lesser extent, mevalonic acid in paclitaxel and/or etoposide treated cells. Furthermore, both urea and citric acid cycles, and metabolism of polyamines and amino acids (aspartate, arginine, and proline) were significantly enriched. These findings can be used to create a map that can be utilized to assess the antitumor effect of paclitaxel and/or etoposide within the studied cancer cells.
Collapse
Affiliation(s)
- Ahlam M. Semreen
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Leen Oyoun Alsoud
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Waseem El-Huneidi
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Munazza Ahmed
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Yasser Bustanji
- Department of Basic and Clinical Pharmacology, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Eman Abu-Gharbieh
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Raafat El-Awady
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Wafaa S. Ramadan
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad A.Y. Alqudah
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohd Shara
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ahmad Y. Abuhelwa
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nelson C. Soares
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad H. Semreen
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Karem H. Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
9
|
Metabolomic and Lipidomic Profiling of Gliomas-A New Direction in Personalized Therapies. Cancers (Basel) 2022; 14:cancers14205041. [PMID: 36291824 PMCID: PMC9599495 DOI: 10.3390/cancers14205041] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Gliomas comprise an extremely diverse category of brain tumors that are difficult to diagnose and treat. As a result, scientists continue to search for new treatment solutions, with personalized medicine having emerged as a particularly promising therapeutic approach. Metabolomics and its sub-discipline, lipidomics, are two scientific fields well-suited to support this search. Metabolomics focuses on the physicochemical changes in the metabolome, which include all of the small endogenous and exogenous compounds in a biological system. As such, metabolic analysis can help identify important biochemical pathways which could be the targets for new therapeutic approaches. This review examines the new directions of personalized therapies for gliomas and how metabolomic and lipidomic analysis assists in developing these strategies and monitoring their effectiveness. The discussion of new strategies is preceded by a brief overview of the current “gold standard” treatment for gliomas and the obstacles that new treatment approaches must overcome. Abstract In addition to being the most common primary brain tumor, gliomas are also among the most difficult to diagnose and treat. At present, the “gold standard” in glioma treatment entails the surgical resection of the largest possible portion of the tumor, followed by temozolomide therapy and radiation. However, this approach does not always yield the desired results. Additionally, the ability to cross the blood-brain barrier remains a major challenge for new potential drugs. Thus, researchers continue to search for targeted therapies that can be individualized based on the specific characteristics of each case. Metabolic and lipidomic research may represent two of the best ways to achieve this goal, as they enable detailed insights into the changes in the profile of small molecules in a biological system/specimen. This article reviews the new approaches to glioma therapy based on the analysis of alterations to biochemical pathways, and it provides an overview of the clinical results that may support personalized therapies in the future.
Collapse
|
10
|
Carneiro TJ, Vojtek M, Gonçalves-Monteiro S, Neves JR, de Carvalho ALMB, Marques MPM, Diniz C, Gil AM. Metabolic Impact of Anticancer Drugs Pd 2Spermine and Cisplatin on the Brain of Healthy Mice. Pharmaceutics 2022; 14:259. [PMID: 35213994 PMCID: PMC8880159 DOI: 10.3390/pharmaceutics14020259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 12/21/2022] Open
Abstract
The new palladium agent Pd2Spermine (Spm) has been reported to exhibit promising cytotoxic properties, while potentially circumventing the known disadvantages associated to cisplatin therapeutics, namely acquired resistance and high toxicity. This work presents a nuclear magnetic resonance (NMR) metabolomics study of brain extracts obtained from healthy mice, to assess the metabolic impacts of the new Pd2Spm complex in comparison to that of cisplatin. The proton NMR spectra of both polar and nonpolar brain extracts were analyzed by multivariate and univariate statistics, unveiling several metabolite variations during the time course of exposition to each drug (1-48 h). The distinct time-course dependence of such changes revealed useful information on the drug-induced dynamics of metabolic disturbances and recovery periods, namely regarding amino acids, nucleotides, fatty acids, and membrane precursors and phospholipids. Putative biochemical explanations were proposed, based on existing pharmacokinetics data and previously reported metabolic responses elicited by the same metal complexes in the liver of the same animals. Generally, results suggest a more effective response of brain metabolism towards the possible detrimental effects of Pd2Spm, with more rapid recovery back to metabolites' control levels and, thus, indicating that the palladium drug may exert a more beneficial role than cDDP in relation to brain toxicity.
Collapse
Affiliation(s)
- Tatiana J. Carneiro
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal; (T.J.C.); (J.R.N.)
| | - Martin Vojtek
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal; (M.V.); (S.G.-M.); (C.D.)
| | - Salomé Gonçalves-Monteiro
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal; (M.V.); (S.G.-M.); (C.D.)
| | - João R. Neves
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal; (T.J.C.); (J.R.N.)
| | - Ana L. M. Batista de Carvalho
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal; (A.L.M.B.d.C.); (M.P.M.M.)
| | - Maria Paula M. Marques
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal; (A.L.M.B.d.C.); (M.P.M.M.)
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Carmen Diniz
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal; (M.V.); (S.G.-M.); (C.D.)
| | - Ana M. Gil
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal; (T.J.C.); (J.R.N.)
| |
Collapse
|
11
|
Shukla A, Srivastava S, Darokar J, Kulshreshtha R. HIF1α and p53 Regulated MED30, a Mediator Complex Subunit, is Involved in Regulation of Glioblastoma Pathogenesis and Temozolomide Resistance. Cell Mol Neurobiol 2021; 41:1521-1535. [PMID: 32705436 PMCID: PMC11448581 DOI: 10.1007/s10571-020-00920-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/10/2020] [Indexed: 10/23/2022]
Abstract
Glioblastoma (GBM) is the most common, malignant, and aggressive form of glial cell cancer with unfavorable clinical outcomes. It is believed that a better understanding of the mechanisms of gene deregulation may lead to novel therapeutic approaches for this yet incurable cancer. Mediator complex is a crucial component of enhancer-based gene expression and works as a transcriptional co-activator. Many of the mediator complex subunits are found to be deregulated/mutated in various malignancies; however, their status and role in GBM remains little studied. We report that MED30, a core subunit of the head module, is overexpressed in GBM tissues and cell lines. MED30 was found to be induced by conditions present in the tumor microenvironment such as hypoxia, serum, and glucose deprivation. MED30 harbors hypoxia response elements (HREs) and p53 binding site in its promoter and is induced in a HIF1α and p53 dependent manner. Further, MED30 levels also significantly positively correlated with p53 and HIF1α levels in GBM tissues. Using both MED30 overexpression and knockdown approach, we show that MED30 promotes cell proliferation while reduces the migration capabilities in GBM cell lines. Notably, MED30 was also found to confer sensitivity to chemodrug, temozolomide, in GBM cells and modulate the level of p53 in vitro. Overall, this is the first report showing MED30 overexpression in GBM and its involvement in GBM pathogenesis suggesting its diagnostic and therapeutic potential urging the need for further systematic exploration of MED30 interactome and target networks.
Collapse
Affiliation(s)
- Anubha Shukla
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Srishti Srivastava
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Jayant Darokar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India.
| |
Collapse
|
12
|
Yin D, Jin G, He H, Zhou W, Fan Z, Gong C, Zhao J, Xiong H. Celecoxib reverses the glioblastoma chemo-resistance to temozolomide through mitochondrial metabolism. Aging (Albany NY) 2021; 13:21268-21282. [PMID: 34497154 PMCID: PMC8457578 DOI: 10.18632/aging.203443] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/01/2021] [Indexed: 01/04/2023]
Abstract
Temozolomide (TMZ) is used for the treatment of high-grade gliomas. Acquired chemoresistance is a serious limitation to the therapy with more than 90% of recurrent gliomas showing little response to a second line of chemotherapy. Therefore, it is necessary to explore an alternative strategy to enhance the sensitivity of glioblastoma (GBM) to TMZ in neuro-oncology. Celecoxib is well known and widely used in anti-inflammatory and analgesic. Cyclooxygenase-2 (COX-2) expression has been linked to the prognosis, angiogenesis, and radiation sensitivity of many malignancies such as primitive neuroectodermal tumor and advanced melanoma. The objective of this study was to explore the chemotherapy-sensitizing effect of celecoxib on TMZ in GBM cells and its potential mechanisms. From the study, we found that the combination therapy (TMZ 250uM+celecoxib 30uM) showed excellent inhibitory effect to the GBM, the LN229 and LN18, which were the TMZ resistant GBM cell lines. Our data suggest that the combination therapy may inhibits cell proliferation, increases apoptosis, and increases the autophagy on LN229 and LN18. The potential molecular mechanisms were related to mitochondrial metabolism and respiratory chain inhibition.
Collapse
Affiliation(s)
- Delong Yin
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Guoqing Jin
- Department of Intensive Care Unit, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Hong He
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Wei Zhou
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Zhenbo Fan
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
13
|
Gautam M, Gabrani R. Combinatorial Effect of Temozolomide and Naringenin in Human Glioblastoma Multiforme Cell Lines. Nutr Cancer 2021; 74:1071-1078. [PMID: 34431435 DOI: 10.1080/01635581.2021.1952438] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Glioblastoma multiforme (GBM) is a grade IV, lethal, and the most common type of brain tumor. GBM can acquire resistance to temozolomide (TMZ) recommended for its treatment. Naringenin (NAG), a flavonoid generally found in grapefruit, has antioxidant, anti-proliferative, and anti-inflammatory properties. It has been reported that phytochemicals can reduce resistance and improve the efficacy of a chemo-resistant drug. The combinatorial effect of TMZ and NAG on cell proliferation was evaluated using 3-4,5-dimethylthiazol-2-yl-2,5-diphenyltetrazolium bromide (MTT) assay, and the apoptosis in the U87MG and LN229 GBM cells were evaluated by change in fluorescence intensity. The effect of NAG and TMZ on anchorage-independent single-cell colony formation and cell migration was investigated. NAG and TMZ demonstrated enhanced cytotoxic effects on U87MG and LN229 cell lines. The combination index value being less than one indicated the synergistic action of the two drugs in restricting the growth of the cells. The NAG and TMZ together resulted in higher fluorescence intensity as compared to the alone drug. Further, the study showed a marked reduction in the migration of the cells and the formation of a single cell colony.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.1952438.
Collapse
Affiliation(s)
- Megha Gautam
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, India
| | - Reema Gabrani
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, India
| |
Collapse
|
14
|
Kirstein A, Schilling D, Combs SE, Schmid TE. Lomeguatrib Increases the Radiosensitivity of MGMT Unmethylated Human Glioblastoma Multiforme Cell Lines. Int J Mol Sci 2021; 22:ijms22136781. [PMID: 34202589 PMCID: PMC8268804 DOI: 10.3390/ijms22136781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Treatment resistance of glioblastoma multiforme to chemo- and radiotherapy remains a challenge yet to overcome. In particular, the O6-methylguanine-DNA-methyltransferase (MGMT) promoter unmethylated patients have only little benefit from chemotherapy treatment using temozolomide since MGMT counteracts its therapeutic efficacy. Therefore, new treatment options in radiotherapy need to be developed to inhibit MGMT and increase radiotherapy response. Methods: Lomeguatrib, a highly specific MGMT inhibitor, was used to inactivate MGMT protein in vitro. Radiosensitivity of established human glioblastoma multiforme cell lines in combination with lomeguatrib was investigated using the clonogenic survival assay. Inhibition of MGMT was analyzed using Western Blot. Cell cycle distribution and apoptosis were investigated to determine the effects of lomeguatrib alone as well as in combination with ionizing radiation. Results: Lomeguatrib significantly decreased MGMT protein and reduced radiation-induced G2/M arrest. A radiosensitizing effect of lomeguatrib was observed when administered at 1 µM and increased radioresistance at 20 µM. Conclusion: Low concentrations of lomeguatrib elicit radiosensitization, while high concentrations mediate a radioprotective effect.
Collapse
Affiliation(s)
- Anna Kirstein
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.K.); (D.S.); (S.E.C.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Daniela Schilling
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.K.); (D.S.); (S.E.C.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Stephanie E. Combs
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.K.); (D.S.); (S.E.C.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany
| | - Thomas E. Schmid
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.K.); (D.S.); (S.E.C.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-3187-43040
| |
Collapse
|
15
|
Jaroch K, Modrakowska P, Bojko B. Glioblastoma Metabolomics-In Vitro Studies. Metabolites 2021; 11:315. [PMID: 34068300 PMCID: PMC8153257 DOI: 10.3390/metabo11050315] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
In 2016, the WHO introduced new guidelines for the diagnosis of brain gliomas based on new genomic markers. The addition of these new markers to the pre-existing diagnostic methods provided a new level of precision for the diagnosis of glioma and the prediction of treatment effectiveness. Yet, despite this new classification tool, glioblastoma (GBM), a grade IV glioma, continues to have one of the highest mortality rates among central nervous system tumors. Metabolomics is a particularly promising tool for the analysis of GBM tumors and potential methods of treating them, as it is the only "omics" approach that is capable of providing a metabolic signature of a tumor's phenotype. With careful experimental design, cell cultures can be a useful matrix in GBM metabolomics, as they ensure stable conditions and, under proper conditions, are capable of capturing different tumor phenotypes. This paper reviews in vitro metabolomic profiling studies of high-grade gliomas, with a particular focus on sample-preparation techniques, crucial metabolites identified, cell culture conditions, in vitro-in vivo extrapolation, and pharmacometabolomics. Ultimately, this review aims to elucidate potential future directions for in vitro GBM metabolomics.
Collapse
Affiliation(s)
| | | | - Barbara Bojko
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, dr A. Jurasza 2 Street, 85-089 Bydgoszcz, Poland; (K.J.); (P.M.)
| |
Collapse
|
16
|
Rahnama S, Bakhshinejad B, Farzam F, Bitaraf A, Ghazimoradi MH, Babashah S. Identification of dysregulated competing endogenous RNA networks in glioblastoma: A way toward improved therapeutic opportunities. Life Sci 2021; 277:119488. [PMID: 33862117 DOI: 10.1016/j.lfs.2021.119488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/16/2021] [Accepted: 04/04/2021] [Indexed: 12/17/2022]
Abstract
Glioblastoma is recognized as one of the leading causes of death worldwide. Although there have been considerable advancements in understanding the causative molecular mechanisms of this malignancy, effective therapeutic strategies are still in limited use. It has been revealed that non-coding RNAs (ncRNAs) play critical roles in glioblastoma development, while interactions between the regulatory molecules such as long ncRNAs (lncRNAs), microRNAs (miRNAs), transcribed pseudogenes, and circular RNAs (circRNAs) remain to be fully deciphered. Over the recent years, researchers have discovered a new category of RNA molecules called competing endogenous RNA (ceRNA). This kind of RNA can contribute to molecular interactions in the form of ceRNA networks (ceRNETs). Multiple lines of evidence have demonstrated that dysregulation of various ceRNA networks is involved in glioblastoma development. Therefore, gaining insights into these dysregulations might offer potential for the early diagnosis of glioblastoma patients and identification of efficient therapeutic targets. In this review, we provide an overview of recent discoveries on ceRNA networks and the involvement of dysregulated networks in posing limitations to temozolomide therapy. We also describe signaling pathways relevant to the progression of glioblastoma.
Collapse
Affiliation(s)
- Saghar Rahnama
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Babak Bakhshinejad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
17
|
Subramani E, Radoul M, Najac C, Batsios G, Molloy AR, Hong D, Gillespie AM, Santos RD, Viswanath P, Costello JF, Pieper RO, Ronen SM. Glutamate Is a Noninvasive Metabolic Biomarker of IDH1-Mutant Glioma Response to Temozolomide Treatment. Cancer Res 2020; 80:5098-5108. [PMID: 32958546 PMCID: PMC7669718 DOI: 10.1158/0008-5472.can-20-1314] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 08/11/2020] [Accepted: 09/16/2020] [Indexed: 02/04/2023]
Abstract
Although lower grade gliomas are driven by mutations in the isocitrate dehydrogenase 1 (IDH1) gene and are less aggressive than primary glioblastoma, they nonetheless generally recur. IDH1-mutant patients are increasingly being treated with temozolomide, but early detection of response remains a challenge and there is a need for complementary imaging methods to assess response to therapy prior to tumor shrinkage. The goal of this study was to determine the value of magnetic resonance spectroscopy (MRS)-based metabolic changes for detection of response to temozolomide in both genetically engineered and patient-derived mutant IDH1 models. Using 1H MRS in combination with chemometrics identified several metabolic alterations in temozolomide-treated cells, including a significant increase in steady-state glutamate levels. This was confirmed in vivo, where the observed 1H MRS increase in glutamate/glutamine occurred prior to tumor shrinkage. Cells labeled with [1-13C]glucose and [3-13C]glutamine, the principal sources of cellular glutamate, showed that flux to glutamate both from glucose via the tricarboxylic acid cycle and from glutamine were increased following temozolomide treatment. In line with these results, hyperpolarized [5-13C]glutamate produced from [2-13C]pyruvate and hyperpolarized [1-13C]glutamate produced from [1-13C]α-ketoglutarate were significantly higher in temozolomide-treated cells compared with controls. Collectively, our findings identify 1H MRS-detectable elevation of glutamate and hyperpolarized 13C MRS-detectable glutamate production from either pyruvate or α-ketoglutarate as potential translatable metabolic biomarkers of response to temozolomide treatment in mutant IDH1 glioma. SIGNIFICANCE: These findings show that glutamate can be used as a noninvasive, imageable metabolic marker for early assessment of tumor response to temozolomide, with the potential to improve treatment strategies for mutant IDH1 patients.
Collapse
Affiliation(s)
- Elavarasan Subramani
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Marina Radoul
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Chloe Najac
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Georgios Batsios
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Abigail R Molloy
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Donghyun Hong
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Romelyn Delos Santos
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Joseph F Costello
- Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Russell O Pieper
- Department of Neurological Surgery, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
- Brain Tumor Research Center, University of California San Francisco, San Francisco, California
| | - Sabrina M Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California.
- Brain Tumor Research Center, University of California San Francisco, San Francisco, California
| |
Collapse
|
18
|
Metabolic Reprogramming of Chemoresistant Cancer Cells and the Potential Significance of Metabolic Regulation in the Reversal of Cancer Chemoresistance. Metabolites 2020; 10:metabo10070289. [PMID: 32708822 PMCID: PMC7408410 DOI: 10.3390/metabo10070289] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/15/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is one of the hallmarks of tumors. Alterations of cellular metabolism not only contribute to tumor development, but also mediate the resistance of tumor cells to antitumor drugs. The metabolic response of tumor cells to various chemotherapy drugs can be analyzed by metabolomics. Although cancer cells have experienced metabolic reprogramming, the metabolism of drug resistant cancer cells has been further modified. Metabolic adaptations of drug resistant cells to chemotherapeutics involve redox, lipid metabolism, bioenergetics, glycolysis, polyamine synthesis and so on. The proposed metabolic mechanisms of drug resistance include the increase of glucose and glutamine demand, active pathways of glutaminolysis and glycolysis, promotion of NADPH from the pentose phosphate pathway, adaptive mitochondrial reprogramming, activation of fatty acid oxidation, and up-regulation of ornithine decarboxylase for polyamine production. Several genes are associated with metabolic reprogramming and drug resistance. Intervening regulatory points described above or targeting key genes in several important metabolic pathways may restore cell sensitivity to chemotherapy. This paper reviews the metabolic changes of tumor cells during the development of chemoresistance and discusses the potential of reversing chemoresistance by metabolic regulation.
Collapse
|
19
|
Shi Y, Wang X, Wang N, Li FF, You YL, Wang SQ. The effect of polysaccharides from Cibotium barometz on enhancing temozolomide-induced glutathione exhausted in human glioblastoma U87 cells, as revealed by 1H NMR metabolomics analysis. Int J Biol Macromol 2020; 156:471-484. [PMID: 32243933 DOI: 10.1016/j.ijbiomac.2020.03.243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) is the most malignant central nervous system tumor, with poor prognosis. Temozolomide (TMZ) has been used as a first-line drug for the treatment of GBM for over a decade, but its treatment benefits are limited by acquired resistance. Polysaccharides from Cibotium barometz (CBPs) are polysaccharides purified from the root of Cibotium barometz (L.) J. Sm., possessing sensitizing activity. The purpose of this study was to investigate the anti-cancer effect of CBP from different processing methods on U87 cells using a 1H NMR-based metabolic approach, complemented with qRT-PCR and flow cytometry, to identify potential markers and discover the targets to explore the underlying mechanism. Cibotium barometz is usually processed under sand heating in clinical applications. Polysaccharides from both the processed (PCBP) and raw (RCBP) C. barometz were prepared, and the effect on enhancing the sensitivity to TMZ was investigated in vitro. CBP can significantly increase the toxicity of TMZ to the U87 cell line, promote apoptosis, enhance cell cycle changes, and arrest cells in S phase, and RCBP demonstrated better activity. Multivariate statistical analyses, such as principal component analysis (PCA) and orthogonal projection to latent structure with discriminant analysis (OPLS-DA), were used to identify metabolic biomarkers, and 12 metabolites in the cell extract samples were clearly identified as altered after RCBP exposure. NMR-based cell metabolomics provided a holistic method for the identification of CBP's apoptosis-enhancing mechanisms and the exploration of its potential applications in preclinical and clinical studies.
Collapse
Affiliation(s)
- Yue Shi
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xiao Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Ning Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Fei-Fei Li
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Yu-Lin You
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Shu-Qi Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
20
|
Jacob M, Lopata AL, Dasouki M, Abdel Rahman AM. Metabolomics toward personalized medicine. MASS SPECTROMETRY REVIEWS 2019; 38:221-238. [PMID: 29073341 DOI: 10.1002/mas.21548] [Citation(s) in RCA: 252] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/14/2017] [Indexed: 05/21/2023]
Abstract
Metabolomics, which is the metabolites profiling in biological matrices, is a key tool for biomarker discovery and personalized medicine and has great potential to elucidate the ultimate product of the genomic processes. Over the last decade, metabolomics studies have identified several relevant biomarkers involved in complex clinical phenotypes using diverse biological systems. Most diseases result in signature metabolic profiles that reflect the sums of external and internal cellular activities. Metabolomics has a major role in clinical practice as it represents >95% of the workload in clinical laboratories worldwide. Many of these metabolites require different analytical platforms, such as Nuclear Magnetic Resonance (NMR), Mass Spectrometry (MS), and Ultra Performance Liquid Chromatography (UPLC), while many clinically relevant metabolites are still not routinely amenable to detection using currently available assays. Combining metabolomics with genomics, transcriptomics, and proteomics studies will result in a significantly improved understanding of the disease mechanisms and the pathophysiology of the target clinical phenotype. This comprehensive approach will represent a major step forward toward providing precision medical care, in which individual is accounted for variability in genes, environment, and personal lifestyle. In this review, we compare and evaluate the metabolomics strategies and studies that focus on the discovery of biomarkers that have "personalized" diagnostic, prognostic, and therapeutic value, validated for monitoring disease progression and responses to various management regimens.
Collapse
Affiliation(s)
- Minnie Jacob
- Department of Genetics, King Faisal Specialist Hospital and Research Center (KFSH-RC), Riyadh, Saudi Arabia
- Department of Molecular and Cell Biology, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - Andreas L Lopata
- Department of Molecular and Cell Biology, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia
| | - Majed Dasouki
- Department of Genetics, King Faisal Specialist Hospital and Research Center (KFSH-RC), Riyadh, Saudi Arabia
| | - Anas M Abdel Rahman
- Department of Genetics, King Faisal Specialist Hospital and Research Center (KFSH-RC), Riyadh, Saudi Arabia
- College of Medicine, Al Faisal University, Riyadh, Saudi Arabia
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
21
|
Katsila T, Matsoukas MT, Patrinos GP, Kardamakis D. Pharmacometabolomics Informs Quantitative Radiomics for Glioblastoma Diagnostic Innovation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:429-439. [PMID: 28816643 DOI: 10.1089/omi.2017.0087] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Applications of omics systems biology technologies have enormous promise for radiology and diagnostics in surgical fields. In this context, the emerging fields of radiomics (a systems scale approach to radiology using a host of technologies, including omics) and pharmacometabolomics (use of metabolomics for patient and disease stratification and guiding precision medicine) offer much synergy for diagnostic innovation in surgery, particularly in neurosurgery. This synthesis of omics fields and applications is timely because diagnostic accuracy in central nervous system tumors still challenges decision-making. Considering the vast heterogeneity in brain tumors, disease phenotypes, and interindividual variability in surgical and chemotherapy outcomes, we believe that diagnostic accuracy can be markedly improved by quantitative radiomics coupled to pharmacometabolomics and related health information technologies while optimizing economic costs of traditional diagnostics. In this expert review, we present an innovation analysis on a systems-level multi-omics approach toward diagnostic accuracy in central nervous system tumors. For this, we suggest that glioblastomas serve as a useful application paradigm. We performed a literature search on PubMed for articles published in English between 2006 and 2016. We used the search terms "radiomics," "glioblastoma," "biomarkers," "pharmacogenomics," "pharmacometabolomics," "pharmacometabonomics/pharmacometabolomics," "collaborative informatics," and "precision medicine." A list of the top 4 insights we derived from this literature analysis is presented in this study. For example, we found that (i) tumor grading needs to be better refined, (ii) diagnostic precision should be improved, (iii) standardization in radiomics is lacking, and (iv) quantitative radiomics needs to prove clinical implementation. We conclude with an interdisciplinary call to the metabolomics, pharmacy/pharmacology, radiology, and surgery communities that pharmacometabolomics coupled to information technologies (chemoinformatics tools, databases, collaborative systems) can inform quantitative radiomics, thus translating Big Data and information growth to knowledge growth, rational drug development and diagnostics innovation for glioblastomas, and possibly in other brain tumors.
Collapse
Affiliation(s)
- Theodora Katsila
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece
| | | | - George P Patrinos
- 1 Department of Pharmacy, School of Health Sciences, University of Patras , Patras, Greece .,2 Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University , Al Ain, United Arab Emirates
| | - Dimitrios Kardamakis
- 3 Department of Radiation Oncology, University of Patras Medical School , Patras, Greece
| |
Collapse
|
22
|
Shen J, Song R, Hodges TR, Heimberger AB, Zhao H. Identification of metabolites in plasma for predicting survival in glioblastoma. Mol Carcinog 2018; 57:1078-1084. [PMID: 29603794 DOI: 10.1002/mc.22815] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/21/2018] [Accepted: 03/27/2018] [Indexed: 12/20/2022]
Abstract
Circulating metabolomics profiling holds prognostic potential. However, such efforts have not been extensively carried out in glioblastoma. In this study, two-step (training and testing) metabolomics profiling was conducted from the plasma samples of 159 glioblastoma patients. Metabolomics profiling was tested for correlation with 2-year overall and disease-free survivals. Arginine, methionine, and kynurenate levels were significantly associated with 2-year overall survival in both the training and testing sets. In the combined sets, elevated levels of arginine and methionine were associated with a 34% and 37% increased probability whereas kynurenate was associated with a 55% decreased probability of 2-year overall survival. These three metabolites were also significantly associated with 2-year disease-free survival. Risk scores were generated using the linear combination of levels of these significant metabolites. Glioblastoma patients with a high-risk score exhibited a 2.41-fold decreased probability of 2-year overall survival (hazard ratio (HR) = 2.41; 95% Confidence Interval (CI) = 1.20-4.93) and a 3.17-fold decreased probability of 2-year disease free survival (HR = 3.17, 95%CI = 1.42-7.54) relative to those with a low-risk score. In conclusion, we identified a unique plasma metabolite profile that is predictive of glioblastoma prognosis.
Collapse
Affiliation(s)
- Jie Shen
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Renduo Song
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tiffany R Hodges
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy B Heimberger
- Department of Neuro-Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hua Zhao
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
23
|
Libby CJ, Tran AN, Scott SE, Griguer C, Hjelmeland AB. The pro-tumorigenic effects of metabolic alterations in glioblastoma including brain tumor initiating cells. Biochim Biophys Acta Rev Cancer 2018; 1869:175-188. [PMID: 29378228 PMCID: PMC6596418 DOI: 10.1016/j.bbcan.2018.01.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 01/20/2018] [Accepted: 01/20/2018] [Indexed: 02/06/2023]
Abstract
De-regulated cellular energetics is an emerging hallmark of cancer with alterations to glycolysis, oxidative phosphorylation, the pentose phosphate pathway, lipid oxidation and synthesis and amino acid metabolism. Understanding and targeting of metabolic reprogramming in cancers may yield new treatment options, but metabolic heterogeneity and plasticity complicate this strategy. One highly heterogeneous cancer for which current treatments ultimately fail is the deadly brain tumor glioblastoma. Therapeutic resistance, within glioblastoma and other solid tumors, is thought to be linked to subsets of tumor initiating cells, also known as cancer stem cells. Recent profiling of glioblastoma and brain tumor initiating cells reveals changes in metabolism, as compiled here, that may be more broadly applicable. We will summarize the profound role for metabolism in tumor progression and therapeutic resistance and discuss current approaches to target glioma metabolism to improve standard of care.
Collapse
Affiliation(s)
- Catherine J. Libby
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA 35294
| | - Anh Nhat Tran
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA 35294
| | - Sarah E. Scott
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA 35294
| | - Corinne Griguer
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA 35294
| | - Anita B. Hjelmeland
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA 35294,, corresponding author, Anita Hjelmeland, Ph.D., Assistant Professor, University of Alabama at Birmingham, Department of Cell, Developmental, and Integrative Biology, 1900 University Blvd, THT 979, Birmingham Al 35294,
| |
Collapse
|
24
|
Gao P, Ji M, Fang X, Liu Y, Yu Z, Cao Y, Sun A, Zhao L, Zhang Y. Capillary electrophoresis - Mass spectrometry metabolomics analysis revealed enrichment of hypotaurine in rat glioma tissues. Anal Biochem 2017; 537:1-7. [PMID: 28847592 DOI: 10.1016/j.ab.2017.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 08/02/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022]
Abstract
Glioma is one of the most lethal brain malignancies with unknown etiologies. Many metabolomics analysis aiming at diverse kinds of samples had been performed. Due to the varied adopted analytical platforms, the reported disease-related metabolites were not consistent across different studies. Comparable metabolomics results are more likely to be acquired by analyzing the same sample types with identical analytical platform. For tumor researches, tissue samples metabolomics analysis own the unique advantage that it can gain more direct insight into disease-specific pathological molecules. In this light, a previous reported capillary electrophoresis - mass spectrometry human tissues metabolomics analysis method was employed to profile the metabolome of rat C6 cell implantation gliomas and the corresponding precancerous tissues. It was found that 9 metabolites increased in the glioma tissues. Of them, hypotaurine was the only metabolite that enriched in the malignant tissues as what had been reported in the relevant human tissues metabolomics analysis. Furthermore, hypotaurine was also proved to inhibit α-ketoglutarate-dependent dioxygenases (2-KDDs) through immunocytochemistry staining and in vitro enzymatic activity assays by using C6 cell cultures. This study reinforced the previous conclusion that hypotaurine acted as a competitive inhibitor of 2-KDDs and proved the value of metabolomics in oncology studies.
Collapse
Affiliation(s)
- Peng Gao
- Clinical Laboratory, Dalian Sixth People's Hospital, Dalian, 116031, China
| | - Min Ji
- Department of Interventional Therapy, Dalian Sixth People's Hospital, Dalian, 116031, China
| | - Xueyan Fang
- Department of Nursing, Dalian Sixth People's Hospital, Dalian, 116031, China
| | - Yingyang Liu
- Department of Nursing, Dalian Sixth People's Hospital, Dalian, 116031, China
| | - Zhigang Yu
- Clinical Technology Center, Dalian Medical University, Dalian, 116044, China
| | - Yunfeng Cao
- RSKT Biopharma Inc., Dalian, Dalian, 116023, China
| | - Aijun Sun
- Translational Medicine Center, Dalian Sixth People's Hospital, Dalian, 116031, China
| | - Liang Zhao
- Translational Medicine Center, Dalian Sixth People's Hospital, Dalian, 116031, China
| | - Yong Zhang
- Department of Surgery, Dalian Sixth People's Hospital, Dalian, 116031, China.
| |
Collapse
|
25
|
Zeng A, Ye T, Cao D, Huang X, Yang Y, Chen X, Xie Y, Yao S, Zhao C. Identify a Blood-Brain Barrier Penetrating Drug-TNB using Zebrafish Orthotopic Glioblastoma Xenograft Model. Sci Rep 2017; 7:14372. [PMID: 29085081 PMCID: PMC5662771 DOI: 10.1038/s41598-017-14766-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 10/12/2017] [Indexed: 02/05/2023] Open
Abstract
The blood-brain barrier (BBB) is necessary for maintaining brain homeostasis, but it also represents a major challenge for drug delivery to the brain tumors. A suitable in vivo Glioblastoma Multiforme (GBM) model is needed for efficient testing of BBB crossable pharmaceuticals. In this study, we firstly confirmed the BBB functionality in 3dpf zebrafish embryos by Lucifer Yellow, Evans Blue and DAPI microinjection. We then transplanted human GBM tumor cells into the zebrafish brain, in which implanted GBM cells (U87 and U251) were highly mitotic and invasive, mimicking their malignancy features in rodents' brain. Interestingly, we found that, although extensive endothelial proliferation and vessel dilation were observed in GBM xenografts, the BBB was still not disturbed. Next, using the zebrafish orthotopic GBM xenograft model as an in vivo visual readout, we successfully identified a promising small compound named TNB, which could efficiently cross the zebrafish BBB and inhibit the progression of orthotopic GBM xenografts. These results indicate that TNB is a promising BBB crossable GBM drug worth to be further characterized in human BBB setting, also suggest the zebrafish orthotopic GBM model as an efficient visual readout for the BBB penetrating anti-GBM drugs.
Collapse
Affiliation(s)
- Anqi Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China
| | - Tinghong Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China
| | - Dan Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China
| | - Xi Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China
| | - Yu Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China
| | - Xiuli Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China
| | - Shaohua Yao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China.
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, P.R. China.
| |
Collapse
|
26
|
Liu Q, Xue Y, Chen Q, Chen H, Zhang X, Wang L, Han C, Que S, Lou M, Lan J. PomGnT1 enhances temozolomide resistance by activating epithelial-mesenchymal transition signaling in glioblastoma. Oncol Rep 2017; 38:2911-2918. [PMID: 29048655 DOI: 10.3892/or.2017.5964] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/28/2017] [Indexed: 11/06/2022] Open
Abstract
Temozolomide (TMZ) is commonly used in glioblastoma (GBM) chemotherapy. However, a great challenge for TMZ treatment is the rapid development of resistance and subsequent tumor recurrence and poor outcome. In the present study we established TMZ-resistant GBM cells (U87-TR and U251-TR) and found that the expression of PomGnT1 was significantly upregulated in TMZ-resistant GBM cells compared with the TMZ-sensitive counterparts. Furthermore, overexpression of PomGnT1 in U87-MG and U251-MG cells led to increased IC50 values for TMZ and reduced apoptosis of cells. Knockdown of PomGnT1 in both U87-TR and U251-TR cells led to decreased IC50 values for TMZ and enhanced apoptosis. Biochemical analysis revealed that PomGnT1 regulates the expression of factors in epithelial-mesenchymal transition signaling including TCF8, vimentin, β-catenin and Slug in GBM cells. These findings demonstrate that PomGnT1 might be a new focus of GBM research for treatment of recurrent TMZ-resistant GBM.
Collapse
Affiliation(s)
- Qi Liu
- Department of Neurosurgery, Jingdezhen Second Hospital, Jingdezhen, Jiangxi, P.R. China
| | - Yajun Xue
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Qingshan Chen
- Department of Neurosurgery, The Second People's Hospital of Liaocheng City, Liaocheng, Shandong, P.R. China
| | - Huairui Chen
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Xiaofei Zhang
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Leiping Wang
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Cong Han
- Department of Neurosurgery, General Hospital, Shanghai, P.R. China
| | - Shuanglin Que
- Department of Neurosurgery, Longyan First Hospital, Fujian Medical University, Longyan, Fujian, P.R. China
| | - Meiqing Lou
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jin Lan
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| |
Collapse
|
27
|
Substituted Caffeic and Ferulic Acid Phenethyl Esters: Synthesis, Leukotrienes Biosynthesis Inhibition, and Cytotoxic Activity. Molecules 2017; 22:molecules22071124. [PMID: 28684707 PMCID: PMC6152019 DOI: 10.3390/molecules22071124] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/04/2017] [Accepted: 07/04/2017] [Indexed: 01/09/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor that correlates with short patient survival and for which therapeutic options are limited. Polyphenolic compounds, including caffeic acid phenethyl ester (CAPE, 1a), have been investigated for their anticancer properties in several types of cancer. To further explore these properties in brain cancer cells, a series of caffeic and ferulic acid esters bearing additional oxygens moieties (OH or OCH3) were designed and synthesized. (CAPE, 1a), but not ferulic acid phenethyl ester (FAPE, 1b), displayed substantial cytotoxicity against two glioma cell lines. Some but not all selected compounds derived from both (CAPE, 1a) and (FAPE, 1b) also displayed cytotoxicity. All CAPE-derived compounds were able to significantly inhibit 5-lipoxygenase (5-LO), however FAPE-derived compounds were largely ineffective 5-LO inhibitors. Molecular docking revealed new hydrogen bonds and π-π interactions between the enzyme and some of the investigated compounds. Overall, this work highlights the relevance of exploring polyphenolic compounds in cancer models and provides additional leads in the development of novel therapeutic strategies in gliomas.
Collapse
|
28
|
Zhang R, Hu P, Zang Q, Yue X, Zhou Z, Xu X, Xu J, Li S, Chen Y, Qiang B, Peng X, Han W, Zhang R, Abliz Z. LC-MS-based metabolomics reveals metabolic signatures related to glioma stem-like cell self-renewal and differentiation. RSC Adv 2017. [DOI: 10.1039/c7ra03781c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A metabolomic study of three glioma cell lines with different stemness were conducted. The specific metabolite signatures associated with SLC self-renewal and differentiation were characterized.
Collapse
|
29
|
Armitage EG, Southam AD. Monitoring cancer prognosis, diagnosis and treatment efficacy using metabolomics and lipidomics. Metabolomics 2016; 12:146. [PMID: 27616976 PMCID: PMC4987388 DOI: 10.1007/s11306-016-1093-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Cellular metabolism is altered during cancer initiation and progression, which allows cancer cells to increase anabolic synthesis, avoid apoptosis and adapt to low nutrient and oxygen availability. The metabolic nature of cancer enables patient cancer status to be monitored by metabolomics and lipidomics. Additionally, monitoring metabolic status of patients or biological models can be used to greater understand the action of anticancer therapeutics. OBJECTIVES Discuss how metabolomics and lipidomics can be used to (i) identify metabolic biomarkers of cancer and (ii) understand the mechanism-of-action of anticancer therapies. Discuss considerations that can maximize the clinical value of metabolic cancer biomarkers including case-control, prognostic and longitudinal study designs. METHODS A literature search of the current relevant primary research was performed. RESULTS Metabolomics and lipidomics can identify metabolic signatures that associate with cancer diagnosis, prognosis and disease progression. Discriminatory metabolites were most commonly linked to lipid or energy metabolism. Case-control studies outnumbered prognostic and longitudinal approaches. Prognostic studies were able to correlate metabolic features with future cancer risk, whereas longitudinal studies were most effective for studying cancer progression. Metabolomics and lipidomics can help to understand the mechanism-of-action of anticancer therapeutics and mechanisms of drug resistance. CONCLUSION Metabolomics and lipidomics can be used to identify biomarkers associated with cancer and to better understand anticancer therapies.
Collapse
Affiliation(s)
- Emily G. Armitage
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad CEU San Pablo, Campus Monteprincipe, Boadilla del Monte, 28668 Madrid, Spain
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA UK
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH UK
| | - Andrew D. Southam
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| |
Collapse
|
30
|
Yogev O, Barker K, Sikka A, Almeida GS, Hallsworth A, Smith LM, Jamin Y, Ruddle R, Koers A, Webber HT, Raynaud FI, Popov S, Jones C, Petrie K, Robinson SP, Keun HC, Chesler L. p53 Loss in MYC-Driven Neuroblastoma Leads to Metabolic Adaptations Supporting Radioresistance. Cancer Res 2016; 76:3025-35. [PMID: 27197232 DOI: 10.1158/0008-5472.can-15-1939] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 02/09/2016] [Indexed: 11/16/2022]
Abstract
Neuroblastoma is the most common childhood extracranial solid tumor. In high-risk cases, many of which are characterized by amplification of MYCN, outcome remains poor. Mutations in the p53 (TP53) tumor suppressor are rare at diagnosis, but evidence suggests that p53 function is often impaired in relapsed, treatment-resistant disease. To address the role of p53 loss of function in the development and pathogenesis of high-risk neuroblastoma, we generated a MYCN-driven genetically engineered mouse model in which the tamoxifen-inducible p53ER(TAM) fusion protein was expressed from a knock-in allele (Th-MYCN/Trp53(KI)). We observed no significant differences in tumor-free survival between Th-MYCN mice heterozygous for Trp53(KI) (n = 188) and Th-MYCN mice with wild-type p53 (n = 101). Conversely, the survival of Th-MYCN/Trp53(KI/KI) mice lacking functional p53 (n = 60) was greatly reduced. We found that Th-MYCN/Trp53(KI/KI) tumors were resistant to ionizing radiation (IR), as expected. However, restoration of functional p53ER(TAM) reinstated sensitivity to IR in only 50% of Th-MYCN/Trp53(KI/KI) tumors, indicating the acquisition of additional resistance mechanisms. Gene expression and metabolic analyses indicated that the principal acquired mechanism of resistance to IR in the absence of functional p53 was metabolic adaptation in response to chronic oxidative stress. Tumors exhibited increased antioxidant metabolites and upregulation of glutathione S-transferase pathway genes, including Gstp1 and Gstz1, which are associated with poor outcome in human neuroblastoma. Accordingly, glutathione depletion by buthionine sulfoximine together with restoration of p53 activity resensitized tumors to IR. Our findings highlight the complex pathways operating in relapsed neuroblastomas and the need for combination therapies that target the diverse resistance mechanisms at play. Cancer Res; 76(10); 3025-35. ©2016 AACR.
Collapse
Affiliation(s)
- Orli Yogev
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Karen Barker
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Arti Sikka
- Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Gilberto S Almeida
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom. Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Albert Hallsworth
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Laura M Smith
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Yann Jamin
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Ruth Ruddle
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Alexander Koers
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Hannah T Webber
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Florence I Raynaud
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Sergey Popov
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom. Department of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Chris Jones
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom. Department of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Kevin Petrie
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Hector C Keun
- Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
31
|
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
|