1
|
Xu X, Zhao H, Liu F, Xie Q, Yang J, Li S, Yang Z. Clinical Value of 18 F-(2S,4R)-4-Fluoroglutamine PET/CT in Glioma. Clin Nucl Med 2025; 50:125-132. [PMID: 39668484 DOI: 10.1097/rlu.0000000000005581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
PROPOSE The aim of this study was to evaluate the clinical application value of 18 F-FGln PET/CT in glioma. PATIENTS AND METHODS Patients with suspected gliomas by MRI were included in this study. Static and/or dynamic brain 18 F-FGln PET/CT was performed. The PET parameters SUV max , SUV mean , MTV, and TLG were evaluated. RESULTS Twenty-three patients were included in the analysis. Nineteen of 23 patients were positive for 18 F-FGln PET. The SUV max of high- and low-grade gliomas were 4.75 ± 2.21 and 1.00 ± 0.66 ( P < 0.001), respectively. FGln-PET SUV max , SUV mean , and TLG all showed statistically significant correlations with glioma grade, with correlation coefficients ( r ) of 0.667 ( P < 0.001), 0.693 ( P < 0.001), and 0.487 ( P = 0.021), respectively. Additionally, the SUV max , SUV mean , and TLG exhibited higher distinguishing performance for glioma grade by receiver operating characteristic curve analysis. The areas under the receiver operating characteristic curve of SUV max , SUV mean , and TLG were 0.976 (95% confidence interval [CI], 0.918-1) ( P = 0.002), 0.976 (95% CI, 0.918-1) ( P = 0.002), and 0.835 (95% CI, 0.628-1.000) ( P = 0.026), respectively. For glioma isocitrate dehydrogenase (IDH) mutation status, the SUV max of IDH wildtype and mutant glioma were 2.95 ± 1.99 and 6.13 ± 2.16 ( P = 0.005), respectively. The SUV mean and SUV max had good-to-satisfactory performance for IDH status with the area under the receiver operating characteristic curve of SUV max and SUV mean of 0.885 (95% CI, 0.734-1.000) ( P = 0.009) and 0.942 (95% CI, 0.828-1) ( P = 0.002). CONCLUSIONS Although we do not assert that 18 F-FGln PET/CT imaging is satisfactory in the differential diagnosis of glioma, we revealed its potential for identifying the stage of gliomas and the IDH mutation status and propose that glutamine-based PET imaging enables the assessment of metabolic nutrient uptake of gliomas to assist clinical diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Xiaoxia Xu
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Heqian Zhao
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Futao Liu
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qing Xie
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jianhua Yang
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zhi Yang
- From the State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
2
|
De los Santos-Jiménez J, Campos-Sandoval JA, Rosales T, Ko B, Alonso FJ, Márquez J, DeBerardinis RJ, Matés JM. Glutaminase-2 Expression Induces Metabolic Changes and Regulates Pyruvate Dehydrogenase Activity in Glioblastoma Cells. Int J Mol Sci 2025; 26:427. [PMID: 39796278 PMCID: PMC11721818 DOI: 10.3390/ijms26010427] [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: 12/16/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Glutaminase controls the first step in glutaminolysis, impacting bioenergetics, biosynthesis and oxidative stress. Two isoenzymes exist in humans, GLS and GLS2. GLS is considered prooncogenic and overexpressed in many tumours, while GLS2 may act as prooncogenic or as a tumour suppressor. Glioblastoma cells usually lack GLS2 while they express high GLS. We investigated how GLS2 expression modifies the metabolism of glioblastoma cells, looking for changes that may explain GLS2's potential tumour suppressive role. We developed LN-229 glioblastoma cells stably expressing GLS2 and performed isotope tracing using U-13C-glutamine and metabolomic quantification to analyze metabolic changes. Treatment with GLS inhibitor CB-839 was also included to concomitantly inhibit endogenous GLS. GLS2 overexpression resulted in extensive metabolic changes, altering the TCA cycle by upregulating part of the cycle but blocking the synthesis of the 6-carbon intermediates from acetyl-CoA. Expression of GLS2 caused downregulation of PDH activity through phosphorylation of S293 of PDHA1. GLS2 also altered nucleotide levels and induced the accumulation of methylated metabolites and S-adenosyl methionine. These changes suggest that GLS2 may be a key regulator linking glutamine and glucose metabolism, also impacting nucleotides and epigenetics. Future research should ascertain the mechanisms involved and the generalizability of these findings in cancer or physiological conditions.
Collapse
Affiliation(s)
- Juan De los Santos-Jiménez
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| | - José A. Campos-Sandoval
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| | - Tracy Rosales
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA; (T.R.); (B.K.); (R.J.D.)
| | - Bookyung Ko
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA; (T.R.); (B.K.); (R.J.D.)
| | - Francisco J. Alonso
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| | - Javier Márquez
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA; (T.R.); (B.K.); (R.J.D.)
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA
| | - José M. Matés
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| |
Collapse
|
3
|
Bafiti V, Thanou E, Ouzounis S, Kotsakis A, Georgoulias V, Lianidou E, Katsila T, Markou A. Profiling Plasma Extracellular Vesicle Metabotypes and miRNAs: An Unobserved Clue for Predicting Relapse in Patients with Early-Stage NSCLC. Cancers (Basel) 2024; 16:3729. [PMID: 39594687 PMCID: PMC11592109 DOI: 10.3390/cancers16223729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Lung cancer, the second most prevalent cancer globally, poses significant challenges in early detection and prognostic assessment. Despite advancements in targeted therapies and immunotherapy, the timely identification of relapse remains elusive. Blood-based liquid biopsy biomarkers, including circulating tumor cells (CTCs), cell-free DNA (cfDNA), circulating tumor DNA (ctDNA), circulating-free RNAs (cfRNAs), and extracellular vesicles (EVs)/exosomes, offer promise for non-invasive monitoring. METHODS We employ a comprehensive approach integrating miRNA/lncRNA/metabolomic datasets, following a mixed-methods content analysis, to identify candidate biomarkers in NSCLC. NSCLC-associated miRNA/gene/lncRNA associations were linked to in silico-derived molecular pathways. RESULTS For data validation, mass spectrometry-based untargeted metabolomics of plasma EVs highlighted miRNA/lncRNA/metabotypes, linking "glycerophospholipid metabolism" to lncRNA H19 and "alanine, aspartate and glutamate metabolism" to miR-29a-3p. Prognostic significance was established for miR-29a-3p, showing lower expression in NSCLC patients with disease progression compared to stable disease (p = 0.004). Kaplan-Meier survival analysis indicated that patients with miR-29a-3p under-expression had significantly shorter overall survival (OS) (p = 0.038). Despite the expression of lncRNA H19 in plasma EVs being undetected, its expression in plasma cfRNAs correlated significantly with disease progression (p = 0.035). CONCLUSIONS Herein, we showcase the potential of plasma EV-derived miR-29a-3p as a prognostic biomarker and underscore the intricate interplay of miRNAs, lncRNAs, and metabolites in NSCLC biology. Our findings offer new insights and avenues for further exploration, contributing to the ongoing quest for effective biomarkers in early-stage NSCLC.
Collapse
Affiliation(s)
- Vivi Bafiti
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (V.B.); (S.O.)
| | - Eleni Thanou
- Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (E.T.); (E.L.)
| | - Sotiris Ouzounis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (V.B.); (S.O.)
| | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Larissa, 41334 Larissa, Greece;
| | - Vasilis Georgoulias
- First Department of Medical Oncology, Metropolitan General Hospital of Athens, 15562 Cholargos, Greece;
| | - Evi Lianidou
- Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (E.T.); (E.L.)
| | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (V.B.); (S.O.)
| | - Athina Markou
- Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (E.T.); (E.L.)
| |
Collapse
|
4
|
Pan S, Yin L, Liu J, Tong J, Wang Z, Zhao J, Liu X, Chen Y, Miao J, Zhou Y, Zeng S, Xu T. Metabolomics-driven approaches for identifying therapeutic targets in drug discovery. MedComm (Beijing) 2024; 5:e792. [PMID: 39534557 PMCID: PMC11555024 DOI: 10.1002/mco2.792] [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: 07/07/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Identification of therapeutic targets can directly elucidate the mechanism and effect of drug therapy, which is a central step in drug development. The disconnect between protein targets and phenotypes under complex mechanisms hampers comprehensive target understanding. Metabolomics, as a systems biology tool that captures phenotypic changes induced by exogenous compounds, has emerged as a valuable approach for target identification. A comprehensive overview was provided in this review to illustrate the principles and advantages of metabolomics, delving into the application of metabolomics in target identification. This review outlines various metabolomics-based methods, such as dose-response metabolomics, stable isotope-resolved metabolomics, and multiomics, which identify key enzymes and metabolic pathways affected by exogenous substances through dose-dependent metabolite-drug interactions. Emerging techniques, including single-cell metabolomics, artificial intelligence, and mass spectrometry imaging, are also explored for their potential to enhance target discovery. The review emphasizes metabolomics' critical role in advancing our understanding of disease mechanisms and accelerating targeted drug development, while acknowledging current challenges in the field.
Collapse
Affiliation(s)
- Shanshan Pan
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Luan Yin
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jie Liu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jie Tong
- Department of Radiology and Biomedical ImagingPET CenterYale School of MedicineNew HavenConnecticutUSA
| | - Zichuan Wang
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Jiahui Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Xuesong Liu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouZhejiangChina
| | - Yong Chen
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouZhejiangChina
| | - Jing Miao
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Yuan Zhou
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Su Zeng
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| | - Tengfei Xu
- Research Center for Clinical PharmacyCollege of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiangChina
| |
Collapse
|
5
|
Trejo-Solís C, Serrano-García N, Castillo-Rodríguez RA, Robledo-Cadena DX, Jimenez-Farfan D, Marín-Hernández Á, Silva-Adaya D, Rodríguez-Pérez CE, Gallardo-Pérez JC. Metabolic dysregulation of tricarboxylic acid cycle and oxidative phosphorylation in glioblastoma. Rev Neurosci 2024; 35:813-838. [PMID: 38841811 DOI: 10.1515/revneuro-2024-0054] [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: 04/16/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
Glioblastoma multiforme (GBM) exhibits genetic alterations that induce the deregulation of oncogenic pathways, thus promoting metabolic adaptation. The modulation of metabolic enzyme activities is necessary to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates essential for fulfilling the biosynthetic needs of glioma cells. Moreover, the TCA cycle produces intermediates that play important roles in the metabolism of glucose, fatty acids, or non-essential amino acids, and act as signaling molecules associated with the activation of oncogenic pathways, transcriptional changes, and epigenetic modifications. In this review, we aim to explore how dysregulated metabolic enzymes from the TCA cycle and oxidative phosphorylation, along with their metabolites, modulate both catabolic and anabolic metabolic pathways, as well as pro-oncogenic signaling pathways, transcriptional changes, and epigenetic modifications in GBM cells, contributing to the formation, survival, growth, and invasion of glioma cells. Additionally, we discuss promising therapeutic strategies targeting key players in metabolic regulation. Therefore, understanding metabolic reprogramming is necessary to fully comprehend the biology of malignant gliomas and significantly improve patient survival.
Collapse
Affiliation(s)
- Cristina Trejo-Solís
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Norma Serrano-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Rosa Angelica Castillo-Rodríguez
- CICATA Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya, Xochitepec 62790, Mexico
| | - Diana Xochiquetzal Robledo-Cadena
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico
| | - Álvaro Marín-Hernández
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Citlali Ekaterina Rodríguez-Pérez
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Neurobiología Molecular y Celular, Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico
| | - Juan Carlos Gallardo-Pérez
- Departamento de Fisiopatología Cardio-Renal, Departamento de Bioquímica, Instituto Nacional de Cardiología, Ciudad de México 14080, Mexico
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Coyoacán, 04510, Ciudad de México, Mexico
| |
Collapse
|
6
|
Moosa NY, Azeem SA, Lodge JK, Cheung W, Ahmed SU. Vitamin B6 Pathway Maintains Glioblastoma Cell Survival in 3D Spheroid Cultures. Int J Mol Sci 2024; 25:10428. [PMID: 39408757 PMCID: PMC11476381 DOI: 10.3390/ijms251910428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is a deadly brain cancer. The prognosis of GBM patients has marginally improved over the last three decades. The response of GBMs to initial treatment is inevitably followed by relapse. Thus, there is an urgent need to identify and develop new therapeutics to target this cancer and improve both patient outcomes and long-term survival. Metabolic reprogramming is considered one of the hallmarks of cancers. However, cell-based studies fail to accurately recapitulate the in vivo tumour microenvironment that influences metabolic signalling and rewiring. Against this backdrop, we conducted global, untargeted metabolomics analysis of the G7 and R24 GBM 2D monolayers and 3D spheroid cultures under identical cell culture conditions. Our studies revealed that the levels of multiple metabolites associated with the vitamin B6 pathway were significantly altered in 3D spheroids compared to the 2D monolayer cultures. Importantly, we show that pharmacological intervention with hydralazine, a small molecule that reduces vitamin B6 levels, resulted in the cell death of 3D GBM spheroid cultures. Thus, our study shows that inhibition of the vitamin B6 pathway is a novel therapeutic strategy for the development of targeted therapies in GBMs.
Collapse
Affiliation(s)
- Najla Yussuf Moosa
- School of Medicine, Murray Health, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK; (N.Y.M.); (S.A.A.)
| | - Sara Abdullah Azeem
- School of Medicine, Murray Health, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK; (N.Y.M.); (S.A.A.)
| | - John K. Lodge
- School of Human Sciences, London Metropolitan University, Tower Building, Holloway Road, London N7 8DB, UK;
| | - William Cheung
- Department of Applied Sciences, Northumbria University, Ellison Building, Northumberland Road, Newcastle Upon Tyne NE1 8ST, UK;
| | - Shafiq Uddin Ahmed
- School of Medicine, Murray Health, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK; (N.Y.M.); (S.A.A.)
| |
Collapse
|
7
|
Fu H, Wu S, Shen H, Luo K, Huang Z, Lu N, Li Y, Lan Q, Xian Y. Glutamine Metabolism Heterogeneity in Glioblastoma Unveils an Innovative Combination Therapy Strategy. J Mol Neurosci 2024; 74:52. [PMID: 38724832 DOI: 10.1007/s12031-024-02201-x] [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: 08/07/2023] [Accepted: 02/12/2024] [Indexed: 07/20/2024]
Abstract
Treatment of glioblastoma multiforme (GBM) remains challenging. Unraveling the orchestration of glutamine metabolism may provide a novel viewpoint on GBM therapy. The study presented a full and comprehensive comprehending of the glutamine metabolism atlas and heterogeneity in GBM for facilitating the development of a more effective therapeutic choice. Transcriptome data from large GBM cohorts were integrated in this study. A glutamine metabolism-based classification was established through consensus clustering approach, and a classifier by LASSO analysis was defined for differentiating the classification. Prognosis, signaling pathway activity, tumor microenvironment, and responses to immune checkpoint blockade (ICB) and small molecular drugs were characterized in each cluster. A combinational therapy of glutaminase inhibitor CB839 with dihydroartemisinin (DHA) was proposed, and the influence on glutamine metabolism, apoptosis, reactive oxygen species (ROS), and migration was measured in U251 and U373 cells. We discovered that GBM presented heterogeneous glutamine metabolism-based clusters, with unique survival outcomes, activity of signaling pathways, tumor microenvironment, and responses to ICB and small molecular compounds. In addition, the classifier could accurately differentiate the two clusters. Strikingly, the combinational therapy of CB839 with DHA synergistically attenuated glutamine metabolism, triggered apoptosis and ROS accumulation, and impaired migrative capacity in GBM cells, demonstrating the excellent preclinical efficacy. Altogether, our findings unveil the glutamine metabolism heterogeneity in GBM and propose an innovative combination therapy of CB839 with DHA for this malignant disease.
Collapse
Affiliation(s)
- Huangde Fu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China.
- Department of Neurosurgery, The Third Nanning People's Hospital, Nanning, Guangxi, 530005, China.
| | - Shengtian Wu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Hechun Shen
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Kai Luo
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Zhongxiang Huang
- Department of Pathology, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Nankun Lu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Yaolin Li
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Qian Lan
- Department of Laboratory, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Yishun Xian
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| |
Collapse
|
8
|
van Noorden CJ, Yetkin-Arik B, Serrano Martinez P, Bakker N, van Breest Smallenburg ME, Schlingemann RO, Klaassen I, Majc B, Habic A, Bogataj U, Galun SK, Vittori M, Erdani Kreft M, Novak M, Breznik B, Hira VV. New Insights in ATP Synthesis as Therapeutic Target in Cancer and Angiogenic Ocular Diseases. J Histochem Cytochem 2024; 72:329-352. [PMID: 38733294 PMCID: PMC11107438 DOI: 10.1369/00221554241249515] [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: 12/05/2023] [Accepted: 04/01/2024] [Indexed: 05/13/2024] Open
Abstract
Lactate and ATP formation by aerobic glycolysis, the Warburg effect, is considered a hallmark of cancer. During angiogenesis in non-cancerous tissue, proliferating stalk endothelial cells (ECs) also produce lactate and ATP by aerobic glycolysis. In fact, all proliferating cells, both non-cancer and cancer cells, need lactate for the biosynthesis of building blocks for cell growth and tissue expansion. Moreover, both non-proliferating cancer stem cells in tumors and leader tip ECs during angiogenesis rely on glycolysis for pyruvate production, which is used for ATP synthesis in mitochondria through oxidative phosphorylation (OXPHOS). Therefore, aerobic glycolysis is not a specific hallmark of cancer but rather a hallmark of proliferating cells and limits its utility in cancer therapy. However, local treatment of angiogenic eye conditions with inhibitors of glycolysis may be a safe therapeutic option that warrants experimental investigation. Most types of cells in the eye such as photoreceptors and pericytes use OXPHOS for ATP production, whereas proliferating angiogenic stalk ECs rely on glycolysis for lactate and ATP production. (J Histochem Cytochem XX.XXX-XXX, XXXX).
Collapse
Affiliation(s)
- Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | - Bahar Yetkin-Arik
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
- Centre for Living Technologies, Alliance TU/e, WUR, UU, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paola Serrano Martinez
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | - Noëlle Bakker
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | | | - Reinier O. Schlingemann
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | - Bernarda Majc
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Anamarija Habic
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
- Jozef Stefan Postgraduate School, Ljubljana, Slovenia
| | - Urban Bogataj
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - S. Katrin Galun
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Milos Vittori
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Metka Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Vashendriya V.V. Hira
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| |
Collapse
|
9
|
Li HY, Feng YH, Lin CL, Hsu TI. Mitochondrial Mechanisms in Temozolomide Resistance: Unraveling the Complex Interplay and Therapeutic Strategies in Glioblastoma. Mitochondrion 2024; 75:101836. [PMID: 38158149 DOI: 10.1016/j.mito.2023.101836] [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: 03/28/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive and lethal brain tumor, with temozolomide (TMZ) being the standard chemotherapeutic agent for its treatment. However, TMZ resistance often develops, limiting its therapeutic efficacy and contributing to poor patient outcomes. Recent evidence highlights the crucial role of mitochondria in the development of TMZ resistance through various mechanisms, including alterations in reactive oxygen species (ROS) production, metabolic reprogramming, apoptosis regulation, biogenesis, dynamics, stress response, and mtDNA mutations. This review article aims to provide a comprehensive overview of the mitochondrial mechanisms involved in TMZ resistance and discuss potential therapeutic strategies targeting these mechanisms to overcome resistance in GBM. We explore the current state of clinical trials targeting mitochondria or related pathways in primary GBM or recurrent GBM, as well as the challenges and future perspectives in this field. Understanding the complex interplay between mitochondria and TMZ resistance will facilitate the development of more effective therapeutic strategies and ultimately improve the prognosis for GBM patients.
Collapse
Affiliation(s)
- Hao-Yi Li
- Department of Biochemistry, Ludwig-Maximilians-University, Munich 81377, Germany; Gene Center, Ludwig-Maximilians-University, Munich 81377, Germany
| | | | | | - Tsung-I Hsu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan.
| |
Collapse
|
10
|
Yalaz C, Bridges E, Alham NK, Zois CE, Chen J, Bensaad K, Miar A, Pires E, Muschel RJ, McCullagh JSO, Harris AL. Cone photoreceptor phosphodiesterase PDE6H inhibition regulates cancer cell growth and metabolism, replicating the dark retina response. Cancer Metab 2024; 12:5. [PMID: 38350962 PMCID: PMC10863171 DOI: 10.1186/s40170-023-00326-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/24/2023] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND PDE6H encodes PDE6γ', the inhibitory subunit of the cGMP-specific phosphodiesterase 6 in cone photoreceptors. Inhibition of PDE6, which has been widely studied for its role in light transduction, increases cGMP levels. The purpose of this study is to characterise the role of PDE6H in cancer cell growth. METHODS From an siRNA screen for 487 genes involved in metabolism, PDE6H was identified as a controller of cell cycle progression in HCT116 cells. Role of PDE6H in cancer cell growth and metabolism was studied through the effects of its depletion on levels of cell cycle controllers, mTOR effectors, metabolite levels, and metabolic energy assays. Effect of PDE6H deletion on tumour growth was also studied in a xenograft model. RESULTS PDE6H knockout resulted in an increase of intracellular cGMP levels, as well as changes to the levels of nucleotides and key energy metabolism intermediates. PDE6H knockdown induced G1 cell cycle arrest and cell death and reduced mTORC1 signalling in cancer cell lines. Both knockdown and knockout of PDE6H resulted in the suppression of mitochondrial function. HCT116 xenografts revealed that PDE6H deletion, as well as treatment with the PDE5/6 inhibitor sildenafil, slowed down tumour growth and improved survival, while sildenafil treatment did not have an additive effect on slowing the growth of PDE6γ'-deficient tumours. CONCLUSIONS Our results indicate that the changes in cGMP and purine pools, as well as mitochondrial function which is observed upon PDE6γ' depletion, are independent of the PKG pathway. We show that in HCT116, PDE6H deletion replicates many effects of the dark retina response and identify PDE6H as a new target in preventing cancer cell proliferation and tumour growth.
Collapse
Affiliation(s)
- Ceren Yalaz
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Esther Bridges
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Nasullah K Alham
- Department of Engineering Science, Institute of Biomedical Engineering (IBME), University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Christos E Zois
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Jianzhou Chen
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Karim Bensaad
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Ana Miar
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Elisabete Pires
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Ruth J Muschel
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - James S O McCullagh
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Adrian L Harris
- Molecular Oncology Laboratories, Department of Medical Oncology, John Radcliffe Hospital, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| |
Collapse
|
11
|
Shin E, Kim B, Kang H, Lee H, Park J, Kang J, Park E, Jo S, Kim HY, Lee JS, Lee JM, Youn H, Youn B. Mitochondrial glutamate transporter SLC25A22 uni-directionally export glutamate for metabolic rewiring in radioresistant glioblastoma. Int J Biol Macromol 2023; 253:127511. [PMID: 37866557 DOI: 10.1016/j.ijbiomac.2023.127511] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/23/2023] [Accepted: 10/11/2023] [Indexed: 10/24/2023]
Abstract
Glioblastoma Multiforme (GBM) is a malignant primary brain tumor. Radiotherapy, one of the standard treatments for GBM patients, could induce GBM radioresistance via rewiring cellular metabolism. However, the precise mechanism attributing to GBM radioresistance or targeting strategies to overcome GBM radioresistance are lacking. Here, we demonstrate that SLC25A22, a mitochondrial bi-directional glutamate transporter, is upregulated and showed uni-directionality from mitochondria to cytosol in radioresistant GBM cells, resulting in accumulating cytosolic glutamate. However, mitochondrial glutaminolysis-mediated TCA cycle metabolites and OCR are maintained constantly. The accumulated cytosolic glutamate enhances the glutathione (GSH) production and proline synthesis in radioresistant GBM cells. Increased GSH protects cells against ionizing radiation (IR)-induced reactive oxygen species (ROS) whereas increased proline, a rate-limiting substrate for collagen biosynthesis, induces extracellular matrix (ECM) remodeling, leading to GBM invasive phenotypes. Finally, we discover that genetic inhibition of SLC25A22 using miR-184 mimic decreases GBM radioresistance and aggressiveness both in vitro and in vivo. Collectively, our study suggests that SLC25A22 upregulation confers GBM radioresistance by rewiring glutamate metabolism, and SLC25A22 could be a significant therapeutic target to overcome GBM radioresistance.
Collapse
Affiliation(s)
- Eunguk Shin
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Byeongsoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Haksoo Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - Junhyung Park
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea
| | - JiHoon Kang
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Sunmi Jo
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan 48108, Republic of Korea
| | - Hae Yu Kim
- Department of Neurosurgery, Haeundae Paik Hospital, Inje University College of Medicine, Busan 48108, Republic of Korea
| | - Jung Sub Lee
- Department of Orthopaedic Surgery, Biomedical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan 49241, Republic of Korea
| | - Jae-Myung Lee
- Department of Naval Architecture and Ocean Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Republic of Korea; Nuclear Science Research Institute, Pusan National University, Busan 46241, Republic of Korea; Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
12
|
Trejo-Solis C, Silva-Adaya D, Serrano-García N, Magaña-Maldonado R, Jimenez-Farfan D, Ferreira-Guerrero E, Cruz-Salgado A, Castillo-Rodriguez RA. Role of Glycolytic and Glutamine Metabolism Reprogramming on the Proliferation, Invasion, and Apoptosis Resistance through Modulation of Signaling Pathways in Glioblastoma. Int J Mol Sci 2023; 24:17633. [PMID: 38139462 PMCID: PMC10744281 DOI: 10.3390/ijms242417633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Glioma cells exhibit genetic and metabolic alterations that affect the deregulation of several cellular signal transduction pathways, including those related to glucose metabolism. Moreover, oncogenic signaling pathways induce the expression of metabolic genes, increasing the metabolic enzyme activities and thus the critical biosynthetic pathways to generate nucleotides, amino acids, and fatty acids, which provide energy and metabolic intermediates that are essential to accomplish the biosynthetic needs of glioma cells. In this review, we aim to explore how dysregulated metabolic enzymes and their metabolites from primary metabolism pathways in glioblastoma (GBM) such as glycolysis and glutaminolysis modulate anabolic and catabolic metabolic pathways as well as pro-oncogenic signaling and contribute to the formation, survival, growth, and malignancy of glioma cells. Also, we discuss promising therapeutic strategies by targeting the key players in metabolic regulation. Therefore, the knowledge of metabolic reprogramming is necessary to fully understand the biology of malignant gliomas to improve patient survival significantly.
Collapse
Affiliation(s)
- Cristina Trejo-Solis
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Norma Serrano-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Roxana Magaña-Maldonado
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Laboratorio de Reprogramación Celular, Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (D.S.-A.); (N.S.-G.); (R.M.-M.)
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico;
| | - Elizabeth Ferreira-Guerrero
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (E.F.-G.); (A.C.-S.)
| | - Arturo Cruz-Salgado
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico; (E.F.-G.); (A.C.-S.)
| | | |
Collapse
|
13
|
Mehjardi NZ, Kessler J, Sanin AY, Picard D, Westhoff P, Nickel AC, Uhlmann C, Shi W, Steiger HJ, Remke M, Fischer I, Vordermark D, Croner RS, Kahlert UD. The development of a hiPSC-based platform to identify tissue-dependencies of IDH1 R132H. Cell Death Discov 2023; 9:452. [PMID: 38086797 PMCID: PMC10716401 DOI: 10.1038/s41420-023-01747-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/13/2023] [Accepted: 11/27/2023] [Indexed: 07/29/2024] Open
Abstract
The application of patient-derived (PD) in vitro tumor models represents the classical strategy for clinical translational oncology research. Using these cellular heterogeneous cultures for the isolation of cancer stem cells (CSCs), suggested to be the main driver for disease malignancy, relies on the use of surrogate biomarkers or is based on CSC-enriching culture conditions. However, the ability of those strategies to exclusively and efficiently enrich for CSC pool has been questioned. Here we present an alternative in vitro CSC model based on the oncogenic transformation of single clone-derived human induced pluripotent stem cells (hiPSC). Hotspot mutations in the DNA encoding for the R132 codon of the enzyme isocitrate dehydrogenase 1 (IDH1) and codon R175 of p53 are commonly occurring molecular features of different tumors and were selected for our transformation strategy. By choosing p53 mutant glial tumors as our model disease, we show that in vitro therapy discovery tests on IDH1-engineered synthetic CSCs (sCSCs) can identify kinases-targeting chemotherapeutics that preferentially target tumor cells expressing corresponding genetic alteration. In contrast, neural stem cells (NSCs) derived from the IDH1R132H overexpressing hiPSCs increase their resistance to the tested interventions indicating glial-to-neural tissue-dependent differences of IDH1R132H. Taken together, we provide proof for the potential of our sCSC technology as a potent addition to biomarker-driven drug development projects or studies on tumor therapy resistance. Moreover, follow-up projects such as comparing in vitro drug sensitivity profiles of hiPSC-derived tissue progenitors of different lineages, might help to understand a variety of tissue-related functions of IDH1 mutations.
Collapse
Affiliation(s)
- N Z Mehjardi
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
| | - J Kessler
- Clinic for Radiation Therapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - A Y Sanin
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular, and Transplant Surgery, Medical Faculty and University Hospital Magdeburg, Magdeburg, Germany
| | - D Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Hospital Düsseldorf and Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - P Westhoff
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Science, Heinrich Heine University, Düsseldorf, Germany
| | - Ann-Christin Nickel
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
- Diaceutics PLC, Düsseldorf, Germany
| | - C Uhlmann
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
- Charles River, Wuppertal, Germany
| | - W Shi
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular, and Transplant Surgery, Medical Faculty and University Hospital Magdeburg, Magdeburg, Germany
| | - H J Steiger
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
| | - M Remke
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Hospital Düsseldorf and Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Pediatric Oncology and Hematology, University Hospital Saarbrücken, Saarbrücken, Germany
| | - I Fischer
- Clinic for Neurosurgery, Medical Faculty Heinrich-Heine University and University Medical Center Düsseldorf, Düsseldorf, Germany
| | - D Vordermark
- Clinic for Radiation Therapy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - R S Croner
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular, and Transplant Surgery, Medical Faculty and University Hospital Magdeburg, Magdeburg, Germany
| | - U D Kahlert
- Department of Molecular and Experimental Surgery, Clinic for General, Visceral, Vascular, and Transplant Surgery, Medical Faculty and University Hospital Magdeburg, Magdeburg, Germany.
| |
Collapse
|
14
|
Zheng Y, Yao Y, Ge T, Ge S, Jia R, Song X, Zhuang A. Amino acid metabolism reprogramming: shedding new light on T cell anti-tumor immunity. J Exp Clin Cancer Res 2023; 42:291. [PMID: 37924140 PMCID: PMC10623764 DOI: 10.1186/s13046-023-02845-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 09/28/2023] [Indexed: 11/06/2023] Open
Abstract
Metabolic reprogramming of amino acids has been increasingly recognized to initiate and fuel tumorigenesis and survival. Therefore, there is emerging interest in the application of amino acid metabolic strategies in antitumor therapy. Tremendous efforts have been made to develop amino acid metabolic node interventions such as amino acid antagonists and targeting amino acid transporters, key enzymes of amino acid metabolism, and common downstream pathways of amino acid metabolism. In addition to playing an essential role in sustaining tumor growth, new technologies and studies has revealed amino acid metabolic reprograming to have wide implications in the regulation of antitumor immune responses. Specifically, extensive crosstalk between amino acid metabolism and T cell immunity has been reported. Tumor cells can inhibit T cell immunity by depleting amino acids in the microenvironment through nutrient competition, and toxic metabolites of amino acids can also inhibit T cell function. In addition, amino acids can interfere with T cells by regulating glucose and lipid metabolism. This crucial crosstalk inspires the exploitation of novel strategies of immunotherapy enhancement and combination, owing to the unprecedented benefits of immunotherapy and the limited population it can benefit. Herein, we review recent findings related to the crosstalk between amino acid metabolism and T cell immunity. We also describe possible approaches to intervene in amino acid metabolic pathways by targeting various signaling nodes. Novel efforts to combine with and unleash potential immunotherapy are also discussed. Hopefully, some strategies that take the lead in the pipeline may soon be used for the common good.
Collapse
Affiliation(s)
- Yue Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China
| | - Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China.
| | - Xin Song
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 20025, P. R. China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, P. R. China.
| |
Collapse
|
15
|
Timofeeva N, Ayres ML, Baran N, Santiago-O’Farrill JM, Bildik G, Lu Z, Konopleva M, Gandhi V. Preclinical investigations of the efficacy of the glutaminase inhibitor CB-839 alone and in combinations in chronic lymphocytic leukemia. Front Oncol 2023; 13:1161254. [PMID: 37228498 PMCID: PMC10203524 DOI: 10.3389/fonc.2023.1161254] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/18/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Chronic lymphocytic leukemia (CLL) cells are metabolically flexible and adapt to modern anticancer treatments. Bruton tyrosine kinase (BTK) and B-cell lymphoma-2 (BCL-2) inhibitors have been widely used to treat CLL, but CLL cells become resistant to these treatments over time. CB-839 is a small-molecule glutaminase-1 (GLS-1) inhibitor that impairs glutamine use, disrupts downstream energy metabolism, and impedes the elimination of reactive oxygen species. Methods To investigate the in vitro effects of CB-839 on CLL cells, we tested CB-839 alone and in combination with ibrutinib, venetoclax, or AZD-5991 on the HG-3 and MEC-1 CLL cell lines and on primary CLL lymphocytes. Results We found that CB-839 caused dose-dependent decreases in GLS-1 activity and glutathione synthesis. CB-839-treated cells also showed increased mitochondrial superoxide metabolism and impaired energy metabolism, which were reflected in decreases in the oxygen consumption rate and depletion of the adenosine triphosphate pool and led to the inhibition of cell proliferation. In the cell lines, CB-839 combined with venetoclax or AZD-5991, but not with ibrutinib, demonstrated synergism with an increased apoptosis rate and cell proliferation inhibition. In the primary lymphocytes, no significant effects of CB-839 alone or in combination with venetoclax, ibrutinib, or AZD-5991 were observed. Discussion Our findings suggest that CB-839 has limited efficacy in CLL treatment and shows limited synergy in combination with widely used CLL drugs.
Collapse
Affiliation(s)
- Natalia Timofeeva
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mary L. Ayres
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Janice M. Santiago-O’Farrill
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gamze Bildik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
16
|
Park JW. Metabolic Rewiring in Adult-Type Diffuse Gliomas. Int J Mol Sci 2023; 24:ijms24087348. [PMID: 37108511 PMCID: PMC10138713 DOI: 10.3390/ijms24087348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Multiple metabolic pathways are utilized to maintain cellular homeostasis. Given the evidence that altered cell metabolism significantly contributes to glioma biology, the current research efforts aim to improve our understanding of metabolic rewiring between glioma's complex genotype and tissue context. In addition, extensive molecular profiling has revealed activated oncogenes and inactivated tumor suppressors that directly or indirectly impact the cellular metabolism that is associated with the pathogenesis of gliomas. The mutation status of isocitrate dehydrogenases (IDHs) is one of the most important prognostic factors in adult-type diffuse gliomas. This review presents an overview of the metabolic alterations in IDH-mutant gliomas and IDH-wildtype glioblastoma (GBM). A particular focus is placed on targeting metabolic vulnerabilities to identify new therapeutic strategies for glioma.
Collapse
Affiliation(s)
- Jong-Whi Park
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
- Neuroscience Research Institute, Gachon University, Incheon 21565, Republic of Korea
| |
Collapse
|
17
|
Glutamine Metabolism in Cancer Stem Cells: A Complex Liaison in the Tumor Microenvironment. Int J Mol Sci 2023; 24:ijms24032337. [PMID: 36768660 PMCID: PMC9916789 DOI: 10.3390/ijms24032337] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
In this review we focus on the role of glutamine in control of cancer stem cell (CSC) fate. We first provide an overview of glutamine metabolism, and then summarize relevant studies investigating how glutamine metabolism modulates the CSC compartment, concentrating on solid tumors. We schematically describe how glutamine in CSC contributes to several metabolic pathways, such as redox metabolic pathways, ATP production, non-essential aminoacids and nucleotides biosynthesis, and ammonia production. Furthermore, we show that glutamine metabolism is a key regulator of epigenetic modifications in CSC. Finally, we briefly discuss how cancer-associated fibroblasts, adipocytes, and senescent cells in the tumor microenvironment may indirectly influence CSC fate by modulating glutamine availability. We aim to highlight the complexity of glutamine's role in CSC, which supports our knowledge about metabolic heterogeneity within the CSC population.
Collapse
|
18
|
De los Santos-Jiménez J, Rosales T, Ko B, Campos-Sandoval JA, Alonso FJ, Márquez J, DeBerardinis RJ, Matés JM. Metabolic Adjustments following Glutaminase Inhibition by CB-839 in Glioblastoma Cell Lines. Cancers (Basel) 2023; 15:531. [PMID: 36672480 PMCID: PMC9856342 DOI: 10.3390/cancers15020531] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Most tumor cells can use glutamine (Gln) for energy generation and biosynthetic purposes. Glutaminases (GAs) convert Gln into glutamate and ammonium. In humans, GAs are encoded by two genes: GLS and GLS2. In glioblastoma, GLS is commonly overexpressed and considered pro-oncogenic. We studied the metabolic effects of inhibiting GLS activity in T98G, LN229, and U87MG human glioblastoma cell lines by using the inhibitor CB-839. We performed metabolomics and isotope tracing experiments using U-13C-labeled Gln, as well as 15N-labeled Gln in the amide group, to determine the metabolic fates of Gln carbon and nitrogen atoms. In the presence of the inhibitor, the results showed an accumulation of Gln and lower levels of tricarboxylic acid cycle intermediates, and aspartate, along with a decreased oxidative labeling and diminished reductive carboxylation-related labeling of these metabolites. Additionally, CB-839 treatment caused decreased levels of metabolites from pyrimidine biosynthesis and an accumulation of intermediate metabolites in the de novo purine nucleotide biosynthesis pathway. The levels of some acetylated and methylated metabolites were significantly increased, including acetyl-carnitine, trimethyl-lysine, and 5-methylcytosine. In conclusion, we analyzed the metabolic landscape caused by the GLS inhibition of CB-839 in human glioma cells, which might lead to the future development of new combination therapies with CB-839.
Collapse
Affiliation(s)
- Juan De los Santos-Jiménez
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29010 Málaga, Spain
| | - Tracy Rosales
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bookyung Ko
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - José A. Campos-Sandoval
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29010 Málaga, Spain
| | - Francisco J. Alonso
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29010 Málaga, Spain
| | - Javier Márquez
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29010 Málaga, Spain
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - José M. Matés
- Canceromics Laboratory, Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29010 Málaga, Spain
| |
Collapse
|
19
|
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
|
20
|
Mao H, Wen Y, Yu Y, Li H, Wang J, Sun B. Bioinspired nanocatalytic tumor therapy by simultaneous reactive oxygen species generation enhancement and glutamine pathway-mediated glutathione depletion. J Mater Chem B 2022; 11:131-143. [PMID: 36484247 DOI: 10.1039/d2tb02194c] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
An insufficient intracellular H2O2 level and overexpressed glutathione (GSH) are still the major challenges for effective chemodynamic therapy (CDT). Inspired by the unique glutamine metabolism pathway in cancer cells, herein, intelligent nanocatalytic theranostics is used to enhance intracellular reactive oxygen species (ROS) accumulation via the production of H2O2 by a biomimetic nanozyme, and simultaneously reduce ROS consumption via the depression of GSH synthesis by the glutamine metabolic inhibitor. In this reactor, nano-sized Au and Fe3O4 coloaded dendritic mesoporous silica nanoparticles (DMSN-Au-Fe3O4) serve as the bifunctional nanozyme, where intracellular glucose is catalyzed into H2O2 by the glucose oxidase-mimicking Au nanoparticles and then immediately transformed into ˙OH by the peroxidase-like Fe3O4 nanoparticles. Then, CB839, the glutaminase (GLS) inhibitor, is grafted on the nanozyme, blocking the glutamine pathway and GSH biosynthesis. As a result, the as-designed nanoplatform with a three-pronged integration of Au-mediated H2O2 self-supply, Fe3O4-triggered Fenton-like reaction, and glutamine pathway-mediated GSH depletion significantly boosts the CDT efficacy, achieving remarkable and specific antitumor properties both in vitro and in vivo. This work not only paves a new way for rationally designing multi-functional nanozymes for achieving high therapeutic efficacy, but also provides new insights into the construction of bioinspired synergetic therapy by combining CDT and a key anticancer pathway.
Collapse
Affiliation(s)
- Huijia Mao
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing 100048, China.
| | - Yangyang Wen
- College of Chemistry and Materials Engineering, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing 100048, China
| | - Yonghui Yu
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing 100048, China.
| | - Hongyan Li
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing 100048, China.
| | - Jing Wang
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing 100048, China.
| | - Baoguo Sun
- China-Canada Joint Lab of Food Nutrition and Health (Beijing), School of Food and Health, Beijing Technology and Business University (BTBU), 11 Fucheng Road, Beijing 100048, China.
| |
Collapse
|
21
|
Kao TW, Chuang YC, Lee HL, Kuo CC, Shen YA. Therapeutic Targeting of Glutaminolysis as a Novel Strategy to Combat Cancer Stem Cells. Int J Mol Sci 2022; 23:ijms232315296. [PMID: 36499623 PMCID: PMC9737183 DOI: 10.3390/ijms232315296] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Rare subpopulations of cancer stem cells (CSCs) have the ability to self-renew and are the primary driving force behind cancer metastatic dissemination and the preeminent hurdle to cancer treatment. As opposed to differentiated, non-malignant tumor offspring, CSCs have sophisticated metabolic patterns that, depending on the kind of cancer, rely mostly on the oxidation of major fuel substrates such as glucose, glutamine, and fatty acids for survival. Glutaminolysis is a series of metabolic reactions that convert glutamine to glutamate and, eventually, α-ketoglutarate, an intermediate in the tricarboxylic acid (TCA) cycle that provides biosynthetic building blocks. These building blocks are mostly utilized in the synthesis of macromolecules and antioxidants for redox homeostasis. A recent study revealed the cellular and molecular interconnections between glutamine and cancer stemness in the cell. Researchers have increasingly focused on glutamine catabolism in their attempt to discover an effective therapy for cancer stem cells. Targeting catalytic enzymes in glutaminolysis, such as glutaminase (GLS), is achievable with small molecule inhibitors, some of which are in early-phase clinical trials and have promising safety profiles. This review summarizes the current findings in glutaminolysis of CSCs and focuses on novel cancer therapies that target glutaminolysis in CSCs.
Collapse
Affiliation(s)
- Ting-Wan Kao
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Yao-Chen Chuang
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan
| | - Hsin-Lun Lee
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei 110301, Taiwan
| | - Chia-Chun Kuo
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 110301, Taiwan
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei 110301, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
| | - Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Correspondence:
| |
Collapse
|
22
|
Bafiti V, Katsila T. Pharmacometabolomics-Based Translational Biomarkers: How to Navigate the Data Ocean. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:542-551. [PMID: 36149303 DOI: 10.1089/omi.2022.0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Metabolome is the end point of the genome-environment interplay, and enables an important holistic overview of individual adaptability and host responses to environmental, ecological, as well as endogenous changes such as disease. Pharmacometabolomics is the application of metabolome knowledge to decipher the mechanisms of interindividual and intraindividual variations in drug efficacy and safety. Pharmacometabolomics also contributes to prediction of drug treatment outcomes on the basis of baseline (predose) and postdose metabotypes through mathematical modeling. Thus, pharmacometabolomics is a strong asset for a diverse community of stakeholders interested in theory and practice of evidence-based and precision/personalized medicine: academic researchers, public health scholars, health professionals, pharmaceutical, diagnostics, and biotechnology industries, among others. In this expert review, we discuss pharmacometabolomics in four contexts: (1) an interdisciplinary omics tool and field to map the mechanisms and scale of interindividual variability in drug effects, (2) discovery and development of translational biomarkers, (3) advance digital biomarkers, and (4) empower drug repurposing, a field that is increasingly proving useful in the current era of Covid-19. As the applications of pharmacometabolomics are growing rapidly in the current postgenome era, next-generation proteomics and metabolomics follow the example of next-generation sequencing analyses. Pharmacometabolomics can also empower data reliability and reproducibility through multiomics integration strategies, which use each data layer to correct, connect with, and inform each other. Finally, we underscore here that contextual data remain crucial for precision medicine and drug development that stand the test of time and clinical relevance.
Collapse
Affiliation(s)
- Vivi Bafiti
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Theodora Katsila
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| |
Collapse
|
23
|
McCarthy L, Verma G, Hangel G, Neal A, Moffat BA, Stockmann JP, Andronesi OC, Balchandani P, Hadjipanayis CG. Application of 7T MRS to High-Grade Gliomas. AJNR Am J Neuroradiol 2022; 43:1378-1395. [PMID: 35618424 PMCID: PMC9575545 DOI: 10.3174/ajnr.a7502] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/11/2022] [Indexed: 01/26/2023]
Abstract
MRS, including single-voxel spectroscopy and MR spectroscopic imaging, captures metabolites in high-grade gliomas. Emerging evidence indicates that 7T MRS may be more sensitive to aberrant metabolic activity than lower-field strength MRS. However, the literature on the use of 7T MRS to visualize high-grade gliomas has not been summarized. We aimed to identify metabolic information provided by 7T MRS, optimal spectroscopic sequences, and areas for improvement in and new applications for 7T MRS. Literature was found on PubMed using "high-grade glioma," "malignant glioma," "glioblastoma," "anaplastic astrocytoma," "7T," "MR spectroscopy," and "MR spectroscopic imaging." 7T MRS offers higher SNR, modestly improved spatial resolution, and better resolution of overlapping resonances. 7T MRS also yields reduced Cramér-Rao lower bound values. These features help to quantify D-2-hydroxyglutarate in isocitrate dehydrogenase 1 and 2 gliomas and to isolate variable glutamate, increased glutamine, and increased glycine with higher sensitivity and specificity. 7T MRS may better characterize tumor infiltration and treatment effect in high-grade gliomas, though further study is necessary. 7T MRS will benefit from increased sample size; reductions in field inhomogeneity, specific absorption rate, and acquisition time; and advanced editing techniques. These findings suggest that 7T MRS may advance understanding of high-grade glioma metabolism, with reduced Cramér-Rao lower bound values and better measurement of smaller metabolite signals. Nevertheless, 7T is not widely used clinically, and technical improvements are necessary. 7T MRS isolates metabolites that may be valuable therapeutic targets in high-grade gliomas, potentially resulting in wider ranging neuro-oncologic applications.
Collapse
Affiliation(s)
- L McCarthy
- From the Department of Neurosurgery (L.M., C.G.H.), Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York
| | - G Verma
- BioMedical Engineering and Imaging Institute (G.V., P.B.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - G Hangel
- Department of Neurosurgery (G.H.)
- High-field MR Center (G.H.), Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - A Neal
- Department of Medicine (A.N.), Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
- Department of Neurology (A.N.), Royal Melbourne Hospital, Melbourne, Australia
| | - B A Moffat
- The Melbourne Brain Centre Imaging Unit (B.A.M.), Department of Radiology, The University of Melbourne, Melbourne, Australia
| | - J P Stockmann
- A. A. Martinos Center for Biomedical Imaging (J.P.S., O.C.A.), Massachusetts General Hospital, Charlestown, Massachusetts
- Harvard Medical School (J.P.S., O.C.A.), Boston, Massachusetts
| | - O C Andronesi
- A. A. Martinos Center for Biomedical Imaging (J.P.S., O.C.A.), Massachusetts General Hospital, Charlestown, Massachusetts
- Harvard Medical School (J.P.S., O.C.A.), Boston, Massachusetts
| | - P Balchandani
- BioMedical Engineering and Imaging Institute (G.V., P.B.), Icahn School of Medicine at Mount Sinai, New York, New York
| | - C G Hadjipanayis
- From the Department of Neurosurgery (L.M., C.G.H.), Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York
| |
Collapse
|
24
|
Banik A, Sharma R, Chauhan A, Singh S. Cutting the umbilical cord: Cancer stem cell-targeted therapeutics. Life Sci 2022; 299:120502. [PMID: 35351466 DOI: 10.1016/j.lfs.2022.120502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
Abstract
Cancer Stem Cells (CSCs) are a notoriously quiescent subpopulation of cells within heterogeneous tumors exhibiting self-renewal, differentiation and drug-resistant capabilities leading to tumor relapse. Heterogeneous cell populations in tumor microenvironment develop an elaborate network of signalling and factors supporting the CSC population within a niche. Identification of specific biomarkers for CSCs facilitates their isolation. CSCs demonstrate abilities that bypass immune surveillance, exhibit resistance to therapy, and induce cancer recurrence while promoting altered metabolism of the bulk tumor, thereby encouraging metastasis. The fight against cancer is prone to relapse without discussing the issue of CSCs, making it imperative for encapsulation of current studies. In this review, we provide extensive knowledge of recent therapeutics developed that target CSCs via multiple signalling cascades, altered metabolism and the tumor microenvironment. Thorough understanding of the functioning of CSCs, their interaction with different cells in the tumor microenvironment as well as current gaps in knowledge are addressed. We present possible strategies to disrupt the cellular and molecular interplay within the tumor microenvironment and make it less conducive for CSCs, which may aid in their eradication with subsequently better treatment outcomes. In conclusion, we discuss a brief yet functional idea of emerging concepts in CSC biology to develop efficient therapeutics acting on cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Ankit Banik
- Department of Biotechnology, Pondicherry University, Chinna Kalapet, Puducherry 605014, India
| | - Rishika Sharma
- Department of Biotechnology, Indian Institute of Technology, Roorkee, Roorkee 247667, India
| | - Akansha Chauhan
- Amity Institute of Physiology and Allied Sciences, Amity University, Noida, India
| | - Sandhya Singh
- Amity Institute of Physiology and Allied Sciences, Amity University, Noida, India.
| |
Collapse
|
25
|
Poonaki E, Nickel AC, Shafiee Ardestani M, Rademacher L, Kaul M, Apartsin E, Meuth SG, Gorji A, Janiak C, Kahlert UD. CD133-Functionalized Gold Nanoparticles as a Carrier Platform for Telaglenastat (CB-839) against Tumor Stem Cells. Int J Mol Sci 2022; 23:5479. [PMID: 35628289 PMCID: PMC9141725 DOI: 10.3390/ijms23105479] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 01/01/2023] Open
Abstract
The failure of a long-lasting curative therapeutic benefit of currently applied chemotherapies against malignant cancers is suggested to be caused by the ineffectiveness of such interventions on cancer stem cells (CSCs). CD133/AC133 is a cell surface protein previously shown to have potential to identify CSCs in various tumors, including brain tumors. Moreover, an increase in the rate of cellular metabolism of glutamine and glucose are contributors to the fast cellular proliferation of some high-grade malignancies. Inhibition of glutaminolysis by utilizing pharmacological inhibitors of the enzyme glutaminase 1 (GLS1) can be an effective anti-CSC strategy. In this study, the clinical-stage GLS1 inhibitor Telaglenastat (CB-839) was loaded into PEGylated gold nanoparticles equipped with the covalently conjugated CD133 aptamer (Au-PEG-CD133-CB-839) and exposed to a collection of CD133-positive brain tumor models in vitro. Our results show that Au-PEG-CD133-CB-839 significantly decreased the viability of CD133-postive cancer cells in a dose-dependent manner, which was higher as compared to the effects of treatment of the cells with the individual components of the assembled nanodrug. Interestingly, the treatment effect was observed in glioblastoma stem cells modeling different transcriptomic subtypes of the disease. The presented platform is the fundament for subsequent target specificity characterization and in vivo application.
Collapse
Affiliation(s)
- Elham Poonaki
- Department of Neurology, Faculty of Medicine, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (E.P.); (S.G.M.)
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-University, 40204 Düsseldorf, Germany; (L.R.); (M.K.)
- Molecular and Experimental Surgery, University Clinic for General-, Visceral-, Vascular- and Transplantation Surgery, Faculty of Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Ann-Christin Nickel
- Clinic for Neurosurgery, Heinrich-Heine-University, 40225 Düsseldorf, Germany;
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1416634793, Iran;
| | - Lars Rademacher
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-University, 40204 Düsseldorf, Germany; (L.R.); (M.K.)
| | - Marilyn Kaul
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-University, 40204 Düsseldorf, Germany; (L.R.); (M.K.)
| | - Evgeny Apartsin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia;
- Laboratoire de Chimie de Coordination CNRS, 31400 Toulouse, France
| | - Sven G. Meuth
- Department of Neurology, Faculty of Medicine, Heinrich-Heine-University, 40225 Düsseldorf, Germany; (E.P.); (S.G.M.)
| | - Ali Gorji
- Epilepsy Research Center, Department of Neurosurgery and Department of Neurology, Westfälische Wilhelms-Universität, 48149 Münster, Germany;
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 9815733169, Iran
| | - Christoph Janiak
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-University, 40204 Düsseldorf, Germany; (L.R.); (M.K.)
| | - Ulf Dietrich Kahlert
- Molecular and Experimental Surgery, University Clinic for General-, Visceral-, Vascular- and Transplantation Surgery, Faculty of Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| |
Collapse
|
26
|
Zhang Q, Li W. Correlation between amino acid metabolism and self-renewal of cancer stem cells: Perspectives in cancer therapy. World J Stem Cells 2022; 14:267-286. [PMID: 35662861 PMCID: PMC9136564 DOI: 10.4252/wjsc.v14.i4.267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/19/2022] [Accepted: 04/25/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) possess self-renewal and differentiation potential, which may be related to recurrence, metastasis, and radiochemotherapy resistance during tumor treatment. Understanding the mechanisms via which CSCs maintain self-renewal may reveal new therapeutic targets for attenuating CSC resistance and extending patient life-span. Recent studies have shown that amino acid metabolism plays an important role in maintaining the self-renewal of CSCs and is involved in regulating their tumorigenicity characteristics. This review summarizes the relationship between CSCs and amino acid metabolism, and discusses the possible mechanisms by which amino acid metabolism regulates CSC characteristics particularly self-renewal, survival and stemness. The ultimate goal is to identify new targets and research directions for elimination of CSCs.
Collapse
Affiliation(s)
- Qi Zhang
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Wei Li
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| |
Collapse
|
27
|
Uhlmann C, Nickel AC, Picard D, Rossi A, Li G, Hildebrandt B, Brockerhoff G, Bendt F, Hübenthal U, Hewera M, Steiger HJ, Wieczorek D, Perrakis A, Zhang W, Remke M, Koch K, Tigges J, Croner RS, Fritsche E, Kahlert UD. Progenitor cells derived from gene-engineered human induced pluripotent stem cells as synthetic cancer cell alternatives for in vitro pharmacology. Biotechnol J 2022; 17:e2100693. [PMID: 35334498 DOI: 10.1002/biot.202100693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 11/08/2022]
Abstract
Limitations in genetic stability and recapitulating accurate physiological disease properties challenge the utility of patient-derived (PD) cancer models for reproducible and translational research. We have genetically engineered a portfolio of isogenic human induced pluripotent stem cells (hiPSCs) with different pan-cancer relevant oncoprotein signatures followed by differentiation into lineage-committed progenitor cells. Characterization on molecular and biological level validated successful stable genetic alterations in pluripotency state as well as upon differentiation to prove the functionality of our approach Meanwhile proposing core molecular networks possibly involved in early dysregulation of stem cell homeostasis, the application of our cell systems in comparative substance testing indicates the potential for cancer research such as identification of augmented therapy resistance of stem cells in response to activation of distinct oncogenic signatures. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Constanze Uhlmann
- Department for Neurosurgery, Medical Faculty and University Medical Center Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ann-Christin Nickel
- Department for Neurosurgery, Medical Faculty and University Medical Center Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Daniel Picard
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.,Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Andrea Rossi
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Guanzhang Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, P. R. China
| | - Barbara Hildebrandt
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany
| | | | - Farina Bendt
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Ulrike Hübenthal
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Michael Hewera
- Department for Neurosurgery, Medical Faculty and University Medical Center Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Jakob Steiger
- Department for Neurosurgery, Medical Faculty and University Medical Center Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Dagmar Wieczorek
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Germany
| | - Aristoteles Perrakis
- Molecular and Experimental Surgery, University Clinic for General, Visceral and Vascular Surgery, University Medical Center Magdeburg and Faculty of Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Wei Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, P. R. China
| | - Marc Remke
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.,Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany.,Department of Neuropathology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Katharina Koch
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Julia Tigges
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Roland S Croner
- Molecular and Experimental Surgery, University Clinic for General, Visceral and Vascular Surgery, University Medical Center Magdeburg and Faculty of Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.,Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Ulf D Kahlert
- Molecular and Experimental Surgery, University Clinic for General, Visceral and Vascular Surgery, University Medical Center Magdeburg and Faculty of Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
28
|
Wang S, Yan Y, Xu WJ, Gong SG, Zhong XJ, An QY, Zhao YL, Liu JM, Wang L, Yuan P, Jiang R. The Role of Glutamine and Glutaminase in Pulmonary Hypertension. Front Cardiovasc Med 2022; 9:838657. [PMID: 35310969 PMCID: PMC8924297 DOI: 10.3389/fcvm.2022.838657] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/14/2022] [Indexed: 01/07/2023] Open
Abstract
Pulmonary hypertension (PH) refers to a clinical and pathophysiological syndrome in which pulmonary vascular resistance and pulmonary arterial pressure are increased due to structural or functional changes in pulmonary vasculature caused by a variety of etiologies and different pathogenic mechanisms. It is followed by the development of right heart failure and even death. In recent years, most studies have found that PH and cancer shared a complex common pathological metabolic disturbance, such as the shift from oxidative phosphorylation to glycolysis. During the shifting process, there is an upregulation of glutamine decomposition driven by glutaminase. However, the relationship between PH and glutamine hydrolysis, especially by glutaminase is yet unclear. This review aims to explore the special linking among glutamine hydrolysis, glutaminase and PH, so as to provide theoretical basis for clinical precision treatment in PH.
Collapse
Affiliation(s)
- Shang Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Yan
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Wei-Jie Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Su-Gang Gong
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiu-Jun Zhong
- Department of Respiratory Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qin-Yan An
- Department of Respiratory, Sijing Hospital of Songjiang District, Shanghai, China
| | - Ya-Lin Zhao
- Department of Respiratory and Critical Care Medicine, The First Hospital of Kunming, Kunming, China
| | - Jin-Ming Liu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lan Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Ping Yuan,
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Rong Jiang,
| |
Collapse
|
29
|
Gonzalez-Covarrubias V, Martínez-Martínez E, del Bosque-Plata L. The Potential of Metabolomics in Biomedical Applications. Metabolites 2022; 12:metabo12020194. [PMID: 35208267 PMCID: PMC8880031 DOI: 10.3390/metabo12020194] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 12/12/2022] Open
Abstract
The metabolome offers a dynamic, comprehensive, and precise picture of the phenotype. Current high-throughput technologies have allowed the discovery of relevant metabolites that characterize a wide variety of human phenotypes with respect to health, disease, drug monitoring, and even aging. Metabolomics, parallel to genomics, has led to the discovery of biomarkers and has aided in the understanding of a diversity of molecular mechanisms, highlighting its application in precision medicine. This review focuses on the metabolomics that can be applied to improve human health, as well as its trends and impacts in metabolic and neurodegenerative diseases, cancer, longevity, the exposome, liquid biopsy development, and pharmacometabolomics. The identification of distinct metabolomic profiles will help in the discovery and improvement of clinical strategies to treat human disease. In the years to come, metabolomics will become a tool routinely applied to diagnose and monitor health and disease, aging, or drug development. Biomedical applications of metabolomics can already be foreseen to monitor the progression of metabolic diseases, such as obesity and diabetes, using branched-chain amino acids, acylcarnitines, certain phospholipids, and genomics; these can assess disease severity and predict a potential treatment. Future endeavors should focus on determining the applicability and clinical utility of metabolomic-derived markers and their appropriate implementation in large-scale clinical settings.
Collapse
Affiliation(s)
| | - Eduardo Martínez-Martínez
- Laboratory of Cell Communication and Extracellular Vesicles, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Laura del Bosque-Plata
- Laboratory of Nutrigenetics and Nutrigenomics, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
- Correspondence: ; Tel.: +52-55-53-50-1974
| |
Collapse
|
30
|
Sharma S, Agnihotri N, Kumar S. Targeting fuel pocket of cancer cell metabolism: A focus on glutaminolysis. Biochem Pharmacol 2022; 198:114943. [DOI: 10.1016/j.bcp.2022.114943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022]
|
31
|
Li Y, Sharma A, Maciaczyk J, Schmidt-Wolf IGH. Recent Development in NKT-Based Immunotherapy of Glioblastoma: From Bench to Bedside. Int J Mol Sci 2022; 23:ijms23031311. [PMID: 35163235 PMCID: PMC8835986 DOI: 10.3390/ijms23031311] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive and dismal disease with a median overall survival of around 15 months and a 5-year survival rate of 7.2%. Owing to genetic mutations, drug resistance, disruption to the blood–brain barrier (BBB)/blood–brain tumor barrier (BBTB), and the complexity of the immunosuppressive environment, the therapeutic approaches to GBM represent still major challenges. Conventional therapies, including surgery, radiotherapy, and standard chemotherapy with temozolomide, have not resulted in satisfactory improvements in the overall survival of GBM patients. Among cancer immunotherapeutic approaches, we propose that adjuvant NKT immunotherapy with invariant NKT (iNKT) and cytokine-induced killer (CIK) cells may improve the clinical scenario of this devastating disease. Considering this, herein, we discuss the current strategies of NKT therapy for GBM based primarily on in vitro/in vivo experiments, clinical trials, and the combinatorial approaches with future therapeutic potential.
Collapse
Affiliation(s)
- Yutao Li
- Center for Integrated Oncology (CIO), Department of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Amit Sharma
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany; (A.S.); (J.M.)
| | - Jarek Maciaczyk
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany; (A.S.); (J.M.)
- Department of Surgical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ingo G. H. Schmidt-Wolf
- Center for Integrated Oncology (CIO), Department of Integrated Oncology, University Hospital Bonn, 53127 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-2871-7050
| |
Collapse
|
32
|
Halama A, Suhre K. Advancing Cancer Treatment by Targeting Glutamine Metabolism-A Roadmap. Cancers (Basel) 2022; 14:553. [PMID: 35158820 PMCID: PMC8833671 DOI: 10.3390/cancers14030553] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Tumor growth and metastasis strongly depend on adapted cell metabolism. Cancer cells adjust their metabolic program to their specific energy needs and in response to an often challenging tumor microenvironment. Glutamine metabolism is one of the metabolic pathways that can be successfully targeted in cancer treatment. The dependence of many hematological and solid tumors on glutamine is associated with mitochondrial glutaminase (GLS) activity that enables channeling of glutamine into the tricarboxylic acid (TCA) cycle, generation of ATP and NADPH, and regulation of glutathione homeostasis and reactive oxygen species (ROS). Small molecules that target glutamine metabolism through inhibition of GLS therefore simultaneously limit energy availability and increase oxidative stress. However, some cancers can reprogram their metabolism to evade this metabolic trap. Therefore, the effectiveness of treatment strategies that rely solely on glutamine inhibition is limited. In this review, we discuss the metabolic and molecular pathways that are linked to dysregulated glutamine metabolism in multiple cancer types. We further summarize and review current clinical trials of glutaminolysis inhibition in cancer patients. Finally, we put into perspective strategies that deploy a combined treatment targeting glutamine metabolism along with other molecular or metabolic pathways and discuss their potential for clinical applications.
Collapse
Affiliation(s)
- Anna Halama
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Doha 24144, Qatar
| |
Collapse
|
33
|
Glutamine-Derived Aspartate Biosynthesis in Cancer Cells: Role of Mitochondrial Transporters and New Therapeutic Perspectives. Cancers (Basel) 2022; 14:cancers14010245. [PMID: 35008407 PMCID: PMC8750728 DOI: 10.3390/cancers14010245] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary In recent years, aspartate has been increasingly acknowledged as a critical player in the metabolism of cancer cells which use this metabolite for nucleotide and protein synthesis and for redox homeostasis. Most intracellular aspartate derives from the mitochondrial catabolism of glutamine. To date at least four mitochondrial transporters have been involved in this metabolic pathway. Their involvement appears to be cancer type-specific and dependent on glutamine availability. Targeting these mitochondrial transporters may represent a new attractive strategy to fight cancer. The aim of this review is to dissect the role of each of these transporters in relation to the type of cancer and the availability of nutrients in the tumoral microenvironment. Abstract Aspartate has a central role in cancer cell metabolism. Aspartate cytosolic availability is crucial for protein and nucleotide biosynthesis as well as for redox homeostasis. Since tumor cells display poor aspartate uptake from the external environment, most of the cellular pool of aspartate derives from mitochondrial catabolism of glutamine. At least four transporters are involved in this metabolic pathway: the glutamine (SLC1A5_var), the aspartate/glutamate (AGC), the aspartate/phosphate (uncoupling protein 2, UCP2), and the glutamate (GC) carriers, the last three belonging to the mitochondrial carrier family (MCF). The loss of one of these transporters causes a paucity of cytosolic aspartate and an arrest of cell proliferation in many different cancer types. The aim of this review is to clarify why different cancers have varying dependencies on metabolite transporters to support cytosolic glutamine-derived aspartate availability. Dissecting the precise metabolic routes that glutamine undergoes in specific tumor types is of upmost importance as it promises to unveil the best metabolic target for therapeutic intervention.
Collapse
|
34
|
van Noorden CJ, Breznik B, Novak M, van Dijck AJ, Tanan S, Vittori M, Bogataj U, Bakker N, Khoury JD, Molenaar RJ, Hira VV. Cell Biology Meets Cell Metabolism: Energy Production Is Similar in Stem Cells and in Cancer Stem Cells in Brain and Bone Marrow. J Histochem Cytochem 2022; 70:29-51. [PMID: 34714696 PMCID: PMC8721571 DOI: 10.1369/00221554211054585] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Energy production by means of ATP synthesis in cancer cells has been investigated frequently as a potential therapeutic target in this century. Both (an)aerobic glycolysis and oxidative phosphorylation (OXPHOS) have been studied. Here, we review recent literature on energy production in glioblastoma stem cells (GSCs) and leukemic stem cells (LSCs) versus their normal counterparts, neural stem cells (NSCs) and hematopoietic stem cells (HSCs), respectively. These two cancer stem cell types were compared because their niches in glioblastoma tumors and in bone marrow are similar. In this study, it became apparent that (1) ATP is produced in NSCs and HSCs by anaerobic glycolysis, whereas fatty acid oxidation (FAO) is essential for their stem cell fate and (2) ATP is produced in GSCs and LSCs by OXPHOS despite the hypoxic conditions in their niches with FAO and amino acids providing its substrate. These metabolic processes appeared to be under tight control of cellular regulation mechanisms which are discussed in depth. However, our conclusion is that systemic therapeutic targeting of ATP production via glycolysis or OXPHOS is not an attractive option because of its unwanted side effects in cancer patients.
Collapse
Affiliation(s)
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Metka Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | | | | | - Miloš Vittori
- Amsterdam UMC Location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Urban Bogataj
- Amsterdam UMC Location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | - Joseph D. Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Remco J. Molenaar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia,Department of Medical Oncology
| | - Vashendriya V.V. Hira
- Vashendriya V.V. Hira, Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia. E-mail:
| |
Collapse
|
35
|
Ekici S, Nye J, Neill S, Allen J, Shu HK, Fleischer C. Glutamine Imaging: A New Avenue for Glioma Management. AJNR Am J Neuroradiol 2022; 43:11-18. [PMID: 34737183 PMCID: PMC8757564 DOI: 10.3174/ajnr.a7333] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/04/2021] [Indexed: 01/03/2023]
Abstract
The glutamine pathway is emerging as an important marker of cancer prognosis and a target for new treatments. In gliomas, the most common type of brain tumors, metabolic reprogramming leads to abnormal consumption of glutamine as an energy source, and increased glutamine concentrations are associated with treatment resistance and proliferation. A key challenge in the development of glutamine-based biomarkers and therapies is the limited number of in vivo tools to noninvasively assess local glutamine metabolism and monitor its changes. In this review, we describe the importance of glutamine metabolism in gliomas and review the current landscape of translational and emerging imaging techniques to measure glutamine in the brain. These techniques include MRS, PET, SPECT, and preclinical methods such as fluorescence and mass spectrometry imaging. Finally, we discuss the roadblocks that must be overcome before incorporating glutamine into a personalized approach for glioma management.
Collapse
Affiliation(s)
- S. Ekici
- From the Departments of Radiology and Imaging Sciences (S.E., J.A.N., J.W.A., C.C.F.)
| | - J.A. Nye
- From the Departments of Radiology and Imaging Sciences (S.E., J.A.N., J.W.A., C.C.F.)
| | - S.G. Neill
- Pathology and Laboratory Medicine (S.G.N.), Emory University School of Medicine, Atlanta, Georgia
| | - J.W. Allen
- From the Departments of Radiology and Imaging Sciences (S.E., J.A.N., J.W.A., C.C.F.),Neurology (J.W.A.), Emory University School of Medicine, Atlanta, Georgia
| | - H.-K. Shu
- Radiation Oncology (H.-K.S.), Emory University School of Medicine, Atlanta, Georgia
| | - C.C. Fleischer
- From the Departments of Radiology and Imaging Sciences (S.E., J.A.N., J.W.A., C.C.F.),Wallace H. Coulter Department of Biomedical Engineering (C.C.F.), Geogria Institute of Technology and Emory University, Atlanta, Georgia
| |
Collapse
|
36
|
Basheer HA, Elsalem L, Salem A, Tailor A, Hunter K, Afarinkia K. The Expression of Glutaminases and their Association with Clinicopathological Parameters in the Head and Neck Cancers. Curr Cancer Drug Targets 2021; 22:169-179. [PMID: 34951574 DOI: 10.2174/1568009622666211224111425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/11/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The increased glutamine metabolism is a characteristic feature of cancer cells. The interconversion between glutamine and glutamate is catalyzed by two glutaminase isoforms, GLS1 and GLS2, which appear to have different roles in different types of cancer. We investigated for the first time the protein expression of GLS1 and GLS2, and their correlation with advanced clinicopathological parameters in head and neck cancers. METHOD Consecutive slides from a tissue microarray comprised of 80 samples ranging from normal to metastatic, were stained immunohistochemically for GLS1, GLS2, HIF-1α or CD147. Following analysis by two expert pathologists we carried out statistical analysis of the scores. RESULTS GLS1 and GLS2 are upregulated at protein level in head and neck tumours compared to normal tissues and this increased expression correlated positively (GLS1) and negatively (GLS2) with tumor grade, indicating a shift of expression between GLS enzyme isoforms based on tumor differentiation. Increased expression of GLS1 was associated with high CD147 expression; and elevated GLS2 expression was associated with both high CD147 and high HIF-1α expressions. The correlation of the GLS1 and GLS2 with HIF-1α or CD147 was strongly associated with more advanced clinicopathological parameters. CONCLUSION The increased expression of GLS1 and GLS2 may be explored as a new treatment for head and neck cancers.
Collapse
Affiliation(s)
- Haneen A Basheer
- Faculty of Pharmacy, Zarqa University, PO Box 132222, Zarqa 13132, Jordan
| | - Lina Elsalem
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, PO Box 3030, Irbid 22110, Jordan
| | - Anwar Salem
- Institute of Cancer Therapeutics, University of Bradford, Richmond Road, BD7 1DP, United Kingdom. 4School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Artysha Tailor
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Keith Hunter
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Kamyar Afarinkia
- Institute of Cancer Therapeutics, University of Bradford, Richmond Road, BD7 1DP, United Kingdom
| |
Collapse
|
37
|
Koch K, Hartmann R, Suwala AK, Rios DH, Kamp MA, Sabel M, Steiger HJ, Willbold D, Sharma A, Kahlert UD, Maciaczyk J. Overexpression of Cystine/Glutamate Antiporter xCT Correlates with Nutrient Flexibility and ZEB1 Expression in Highly Clonogenic Glioblastoma Stem-like Cells (GSCs). Cancers (Basel) 2021; 13:cancers13236001. [PMID: 34885110 PMCID: PMC8672273 DOI: 10.3390/cancers13236001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) is the most aggressive form of glioma (WHO grade IV), and mounting evidence suggests that glioblastoma stem-like cells (GSCs) play an important role in tumor growth and response to therapy. In the current study, we sought to understand the metabolic dependencies of GSCs using high-resolution proton magnetic resonance spectroscopy (1H-NMR). In a defined experimental setting, we stratified in vitro GSC models into two subtypes (Gln/GluHigh, Gln/GluLow) and used diverse molecular approaches to perform comprehensive analyses in GSC neurosphere cultures and primary GBM samples. Abstract Cancer stem-like cells mediate tumor initiation, progression, and therapy resistance; however, their identification and selective eradication remain challenging. Herein, we analyze the metabolic dependencies of glioblastoma stem-like cells (GSCs) with high-resolution proton nuclear magnetic resonance (1H-NMR) spectroscopy. We stratify our in vitro GSC models into two subtypes primarily based on their relative amount of glutamine in relationship to glutamate (Gln/Glu). Gln/GluHigh GSCs were found to be resistant to glutamine deprivation, whereas Gln/GluLow GSCs respond with significantly decreased in vitro clonogenicity and impaired cell growth. The starvation resistance appeared to be mediated by an increased expression of the glutamate/cystine antiporter SLC7A11/xCT and efficient cellular clearance of reactive oxygen species (ROS). Moreover, we were able to directly correlate xCT-dependent starvation resistance and high Gln/Glu ratios with in vitro clonogenicity, since targeted differentiation of GSCs with bone morphogenic protein 4 (BMP4) impaired xCT expression, decreased the Gln/Glu ratio, and restored the sensitivity to glutamine starvation. Moreover, significantly reduced levels of the oncometabolites lactate (Lac), phosphocholine (PC), total choline (tCho), myo-inositol (Myo-I), and glycine (Gly) were observed in differentiated GSCs. Furthermore, we found a strong association between high Gln/Glu ratios and increased expression of Zinc finger E-box-binding homeobox 1 (ZEB1) and xCT in primary GBM tumor tissues. Our analyses suggest that the inhibition of xCT represents a potential therapeutic target in glioblastoma; thus, we could further extend its importance in GSC biology and stress responses. We also propose that monitoring of the intracellular Gln/Glu ratio can be used to predict nutrient stress resistance.
Collapse
Affiliation(s)
- Katharina Koch
- Department of Neurosurgery, University Hospital Duesseldorf, 40225 Duesseldorf, Germany; (K.K.); (D.H.R.); (M.S.); (H.-J.S.); (U.D.K.)
- IUF—Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany
| | - Rudolf Hartmann
- Institute of Biological Information Processing (IBI-7) Structural Biochemistry, Forschungszentrum Juelich, 52425 Juelich, Germany; (R.H.); (D.W.)
| | - Abigail Kora Suwala
- Department of Neurological Surgery, Helen Diller Research Center, University of California San Francisco, San Francisco, CA 94158, USA;
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - Dayana Herrera Rios
- Department of Neurosurgery, University Hospital Duesseldorf, 40225 Duesseldorf, Germany; (K.K.); (D.H.R.); (M.S.); (H.-J.S.); (U.D.K.)
- Skin Cancer Unit of the Dermatology Department, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany
| | - Marcel Alexander Kamp
- Department of Neurosurgery, Centre of Neuro-Oncology, Jena University Hospital, Friedrich-Schiller-University Jena, 07747 Jena, Germany;
| | - Michael Sabel
- Department of Neurosurgery, University Hospital Duesseldorf, 40225 Duesseldorf, Germany; (K.K.); (D.H.R.); (M.S.); (H.-J.S.); (U.D.K.)
| | - Hans-Jakob Steiger
- Department of Neurosurgery, University Hospital Duesseldorf, 40225 Duesseldorf, Germany; (K.K.); (D.H.R.); (M.S.); (H.-J.S.); (U.D.K.)
| | - Dieter Willbold
- Institute of Biological Information Processing (IBI-7) Structural Biochemistry, Forschungszentrum Juelich, 52425 Juelich, Germany; (R.H.); (D.W.)
- Institut für Physikalische Biologie, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Amit Sharma
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany;
| | - Ulf Dietrich Kahlert
- Department of Neurosurgery, University Hospital Duesseldorf, 40225 Duesseldorf, Germany; (K.K.); (D.H.R.); (M.S.); (H.-J.S.); (U.D.K.)
- Department of Molecular and Experimental Surgery, Clinic of General, Visceral, Vascular and Transplant Surgery, Faculty of Medicine, Otto-von-Guericke University and University Hospital Magdeburg, 39120 Magdeburg, Germany
| | - Jarek Maciaczyk
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany;
- Department of Surgical Sciences, University of Otago, Dunedin 9054, New Zealand
- Correspondence: ; Tel.: +49-(0)228-287-16531
| |
Collapse
|
38
|
Choudhury FK. Mitochondrial Redox Metabolism: The Epicenter of Metabolism during Cancer Progression. Antioxidants (Basel) 2021; 10:antiox10111838. [PMID: 34829708 PMCID: PMC8615124 DOI: 10.3390/antiox10111838] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial redox metabolism is the central component in the cellular metabolic landscape, where anabolic and catabolic pathways are reprogrammed to maintain optimum redox homeostasis. During different stages of cancer, the mitochondrial redox status plays an active role in navigating cancer cells’ progression and regulating metabolic adaptation according to the constraints of each stage. Mitochondrial reactive oxygen species (ROS) accumulation induces malignant transformation. Once vigorous cell proliferation renders the core of the solid tumor hypoxic, the mitochondrial electron transport chain mediates ROS signaling for bringing about cellular adaptation to hypoxia. Highly aggressive cells are selected in this process, which are capable of progressing through the enhanced oxidative stress encountered during different stages of metastasis for distant colonization. Mitochondrial oxidative metabolism is suppressed to lower ROS generation, and the overall cellular metabolism is reprogrammed to maintain the optimum NADPH level in the mitochondria required for redox homeostasis. After reaching the distant organ, the intrinsic metabolic limitations of that organ dictate the success of colonization and flexibility of the mitochondrial metabolism of cancer cells plays a pivotal role in their adaptation to the new environment.
Collapse
Affiliation(s)
- Feroza K Choudhury
- Drug Metabolism and Pharmacokinetics Department, Genentech Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
39
|
The expression of glutamate metabolism modulators in the intracranial tumors and glioblastoma cell line. Mol Biol Rep 2021; 49:1077-1083. [PMID: 34773180 DOI: 10.1007/s11033-021-06931-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND The accumulation of excess glutamate in the synapse leads to excitotoxicity, which is the underlying reason of neuronal death in intracranial tumors. METHODS AND RESULTS We identified the expression levels of glutamate dehydrogenase, glutamine synthetase and sirtuin 4 in U87 cell line and various intracranial tumors. mRNA expressions of glutamate dehydrogenase (GDH), glutamine synthetase (GS) and sirtuin 4 (SIRT4) were analyzed in various intracranial tumors using qPCR. GDH, GS and SIRT4 protein expressions were analyzed in glioblastoma (U87) and glial (IHA-immortalized human astrocytes) cell lines via western blotting. The protein expressions of SIRT4 and GS were shown to be elevated and GDH protein expression was reduced in U87 cells in comparison to IHA cells. All types of intracranial tumors displayed lower GS mRNA expressions compared to controls. SIRT4 mRNA expressions were also shown to be lower in all the tumors and grades, although not significantly. GDH mRNA expression was found to be similar in all groups. CONCLUSION The molecular mechanisms of glutamate metabolism and excitotoxicity should be discovered to develop therapies against intracranial tumors.
Collapse
|
40
|
Deshmukh R, Allega MF, Tardito S. A map of the altered glioma metabolism. Trends Mol Med 2021; 27:1045-1059. [PMID: 34489164 DOI: 10.1016/j.molmed.2021.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022]
Abstract
The frequent occurrence of neomorphic isocitrate dehydrogenase 1 (IDH1) mutations in low-grade glioma led to an IDH-centric classification of these tumors. However, exploiting metabolic alterations of glioma for diagnostic imaging and treatment has marginally improved patients' prognosis. Here we discuss the nutritional microenvironment of glioma, shaped by the distinctive dependence of the brain on glucose and ketone bodies for energy, and on amino acids for neurotransmission. We highlight the progress in metabolic applications for glioma diagnosis and therapy, and present a map that streamlines the rewired glioma metabolism. The map illustrates the altered reactions in central carbon and nitrogen metabolism that drive glioma biology, and represent metabolic vulnerabilities with translational potential.
Collapse
Affiliation(s)
- Ruhi Deshmukh
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Maria Francesca Allega
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Saverio Tardito
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
41
|
Ganini C, Amelio I, Bertolo R, Bove P, Buonomo OC, Candi E, Cipriani C, Di Daniele N, Juhl H, Mauriello A, Marani C, Marshall J, Melino S, Marchetti P, Montanaro M, Natale ME, Novelli F, Palmieri G, Piacentini M, Rendina EA, Roselli M, Sica G, Tesauro M, Rovella V, Tisone G, Shi Y, Wang Y, Melino G. Global mapping of cancers: The Cancer Genome Atlas and beyond. Mol Oncol 2021; 15:2823-2840. [PMID: 34245122 PMCID: PMC8564642 DOI: 10.1002/1878-0261.13056] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/04/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer genomes have been explored from the early 2000s through massive exome sequencing efforts, leading to the publication of The Cancer Genome Atlas in 2013. Sequencing techniques have been developed alongside this project and have allowed scientists to bypass the limitation of costs for whole-genome sequencing (WGS) of single specimens by developing more accurate and extensive cancer sequencing projects, such as deep sequencing of whole genomes and transcriptomic analysis. The Pan-Cancer Analysis of Whole Genomes recently published WGS data from more than 2600 human cancers together with almost 1200 related transcriptomes. The application of WGS on a large database allowed, for the first time in history, a global analysis of features such as molecular signatures, large structural variations and noncoding regions of the genome, as well as the evaluation of RNA alterations in the absence of underlying DNA mutations. The vast amount of data generated still needs to be thoroughly deciphered, and the advent of machine-learning approaches will be the next step towards the generation of personalized approaches for cancer medicine. The present manuscript wants to give a broad perspective on some of the biological evidence derived from the largest sequencing attempts on human cancers so far, discussing advantages and limitations of this approach and its power in the era of machine learning.
Collapse
Affiliation(s)
- Carlo Ganini
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- IDI‐IRCCSRomeItaly
| | - Ivano Amelio
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Riccardo Bertolo
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - Pierluigi Bove
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - Oreste Claudio Buonomo
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Eleonora Candi
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- IDI‐IRCCSRomeItaly
| | - Chiara Cipriani
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - Nicola Di Daniele
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | | | - Alessandro Mauriello
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Carla Marani
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - John Marshall
- Medstar Georgetown University HospitalGeorgetown UniversityWashingtonDCUSA
| | - Sonia Melino
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | | | - Manuela Montanaro
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Maria Emanuela Natale
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- San Carlo di Nancy HospitalRomeItaly
| | - Flavia Novelli
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Giampiero Palmieri
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Mauro Piacentini
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | | | - Mario Roselli
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Giuseppe Sica
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Manfredi Tesauro
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Valentina Rovella
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Giuseppe Tisone
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| | - Yufang Shi
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthShanghai Institutes for Biological SciencesUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
- The First Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSoochow UniversityChina
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and TumorShanghai Institute of Nutrition and HealthShanghai Institutes for Biological SciencesUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Gerry Melino
- Department of Experimental MedicineTorvergata Oncoscience Research Centre of Excellence, TORUniversity of Rome Tor VergataItaly
| |
Collapse
|
42
|
Chisari A, Golán I, Campisano S, Gélabert C, Moustakas A, Sancho P, Caja L. Glucose and Amino Acid Metabolic Dependencies Linked to Stemness and Metastasis in Different Aggressive Cancer Types. Front Pharmacol 2021; 12:723798. [PMID: 34588983 PMCID: PMC8473699 DOI: 10.3389/fphar.2021.723798] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022] Open
Abstract
Malignant cells are commonly characterised by being capable of invading tissue, growing self-sufficiently and uncontrollably, being insensitive to apoptosis induction and controlling their environment, for example inducing angiogenesis. Amongst them, a subpopulation of cancer cells, called cancer stem cells (CSCs) shows sustained replicative potential, tumor-initiating properties and chemoresistance. These characteristics make CSCs responsible for therapy resistance, tumor relapse and growth in distant organs, causing metastatic dissemination. For these reasons, eliminating CSCs is necessary in order to achieve long-term survival of cancer patients. New insights in cancer metabolism have revealed that cellular metabolism in tumors is highly heterogeneous and that CSCs show specific metabolic traits supporting their unique functionality. Indeed, CSCs adapt differently to the deprivation of specific nutrients that represent potentially targetable vulnerabilities. This review focuses on three of the most aggressive tumor types: pancreatic ductal adenocarcinoma (PDAC), hepatocellular carcinoma (HCC) and glioblastoma (GBM). The aim is to prove whether CSCs from different tumour types share common metabolic requirements and responses to nutrient starvation, by outlining the diverse roles of glucose and amino acids within tumour cells and in the tumour microenvironment, as well as the consequences of their deprivation. Beyond their role in biosynthesis, they serve as energy sources and help maintain redox balance. In addition, glucose and amino acid derivatives contribute to immune responses linked to tumourigenesis and metastasis. Furthermore, potential metabolic liabilities are identified and discussed as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Andrea Chisari
- Department of Chemistry, School of Sciences, National University of Mar del Plata, Mar del Plata, Argentina
| | - Irene Golán
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Sabrina Campisano
- Department of Chemistry, School of Sciences, National University of Mar del Plata, Mar del Plata, Argentina
| | - Caroline Gélabert
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Patricia Sancho
- Translational Research Unit, Hospital Universitario Miguel Servet, IIS Aragon, Zaragoza, Spain
| | - Laia Caja
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Biomedical Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
Shen YA, Chen CC, Chen BJ, Wu YT, Juan JR, Chen LY, Teng YC, Wei YH. Potential Therapies Targeting Metabolic Pathways in Cancer Stem Cells. Cells 2021; 10:1772. [PMID: 34359941 PMCID: PMC8304173 DOI: 10.3390/cells10071772] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) are heterogeneous cells with stem cell-like properties that are responsible for therapeutic resistance, recurrence, and metastasis, and are the major cause for cancer treatment failure. Since CSCs have distinct metabolic characteristics that plays an important role in cancer development and progression, targeting metabolic pathways of CSCs appears to be a promising therapeutic approach for cancer treatment. Here we classify and discuss the unique metabolisms that CSCs rely on for energy production and survival, including mitochondrial respiration, glycolysis, glutaminolysis, and fatty acid metabolism. Because of metabolic plasticity, CSCs can switch between these metabolisms to acquire energy for tumor progression in different microenvironments compare to the rest of tumor bulk. Thus, we highlight the specific conditions and factors that promote or suppress CSCs properties to portray distinct metabolic phenotypes that attribute to CSCs in common cancers. Identification and characterization of the features in these metabolisms can offer new anticancer opportunities and improve the prognosis of cancer. However, the therapeutic window of metabolic inhibitors used alone or in combination may be rather narrow due to cytotoxicity to normal cells. In this review, we present current findings of potential targets in these four metabolic pathways for the development of more effective and alternative strategies to eradicate CSCs and treat cancer more effectively in the future.
Collapse
Affiliation(s)
- Yao-An Shen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chang-Cyuan Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Bo-Jung Chen
- Department of Pathology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan;
| | - Yu-Ting Wu
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 50046, Taiwan;
| | - Jiun-Ru Juan
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Liang-Yun Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Yueh-Chun Teng
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-A.S.); (C.-C.C.); (J.-R.J.); (L.-Y.C.); (Y.-C.T.)
| | - Yau-Huei Wei
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 50046, Taiwan;
| |
Collapse
|
44
|
Abstract
Cancer stem cells (CSCs) are heterogeneous cells with stem cell-like properties that are responsible for therapeutic resistance, recurrence, and metastasis, and are the major cause for cancer treatment failure. Since CSCs have distinct metabolic characteristics that plays an important role in cancer development and progression, targeting metabolic pathways of CSCs appears to be a promising therapeutic approach for cancer treatment. Here we classify and discuss the unique metabolisms that CSCs rely on for energy production and survival, including mitochondrial respiration, glycolysis, glutaminolysis, and fatty acid metabolism. Because of metabolic plasticity, CSCs can switch between these metabolisms to acquire energy for tumor progression in different microenvironments compare to the rest of tumor bulk. Thus, we highlight the specific conditions and factors that promote or suppress CSCs properties to portray distinct metabolic phenotypes that attribute to CSCs in common cancers. Identification and characterization of the features in these metabolisms can offer new anticancer opportunities and improve the prognosis of cancer. However, the therapeutic window of metabolic inhibitors used alone or in combination may be rather narrow due to cytotoxicity to normal cells. In this review, we present current findings of potential targets in these four metabolic pathways for the development of more effective and alternative strategies to eradicate CSCs and treat cancer more effectively in the future.
Collapse
|
45
|
Moldovan OL, Rusu A, Tanase C, Vari CE. Glutamate - A multifaceted molecule: Endogenous neurotransmitter, controversial food additive, design compound for anti-cancer drugs. A critical appraisal. Food Chem Toxicol 2021; 153:112290. [PMID: 34023459 DOI: 10.1016/j.fct.2021.112290] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022]
Abstract
One of the most widely used flavour enhancers in the food industry is monosodium glutamate (MSG). MSG consumption has been on an upward trend, worrying in terms of potential toxic effects. This review is focused on the long-term toxicity of MSG and the experimental evidence that supports it. The article's primary purpose was to survey recently published data regarding the consumption of MSG within safe limits. The administered doses in animal models are very varied and have given rise to controversy. Also, the paper comprises pathways to lower MSG toxicity and highlight other underexploited biological effects, as anti-cancer potential. The administration of MSG, combined with various compounds, has been shown benefit against toxic effects. Several recent studies have identified a possible mechanism that recommends MSG and some derivatives as potential anti-cancer agents. New anti-cancer compounds based on the glutamic acid structure must be studied and further exploited. International regulations require harmonization of safe doses of MSG based on current scientific studies. Replacing MSG with other umami flavour enhancers may be a safer alternative for human health in the future. The biological consequences of MSG consumption or therapeutical administration have not been fully deciphered yet.
Collapse
Affiliation(s)
- Octavia-Laura Moldovan
- Medicine and Pharmacy Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142, Târgu Mureș, Romania.
| | - Aura Rusu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142, Târgu Mureș, Romania.
| | - Corneliu Tanase
- Pharmaceutical Botany Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142, Târgu Mureș, Romania.
| | - Camil-Eugen Vari
- Pharmacy and Clinical Pharmacy Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142, Târgu Mureș, Romania.
| |
Collapse
|
46
|
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
|
47
|
van Noorden CJ, Hira VV, van Dijck AJ, Novak M, Breznik B, Molenaar RJ. Energy Metabolism in IDH1 Wild-Type and IDH1-Mutated Glioblastoma Stem Cells: A Novel Target for Therapy? Cells 2021; 10:cells10030705. [PMID: 33810170 PMCID: PMC8005124 DOI: 10.3390/cells10030705] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is a redox disease. Low levels of reactive oxygen species (ROS) are beneficial for cells and have anti-cancer effects. ROS are produced in the mitochondria during ATP production by oxidative phosphorylation (OXPHOS). In the present review, we describe ATP production in primary brain tumors, glioblastoma, in relation to ROS production. Differentiated glioblastoma cells mainly use glycolysis for ATP production (aerobic glycolysis) without ROS production, whereas glioblastoma stem cells (GSCs) in hypoxic periarteriolar niches use OXPHOS for ATP and ROS production, which is modest because of the hypoxia and quiescence of GSCs. In a significant proportion of glioblastoma, isocitrate dehydrogenase 1 (IDH1) is mutated, causing metabolic rewiring, and all cancer cells use OXPHOS for ATP and ROS production. Systemic therapeutic inhibition of glycolysis is not an option as clinical trials have shown ineffectiveness or unwanted side effects. We argue that systemic therapeutic inhibition of OXPHOS is not an option either because the anti-cancer effects of ROS production in healthy cells is inhibited as well. Therefore, we advocate to remove GSCs out of their hypoxic niches by the inhibition of their binding to niches to enable their differentiation and thus increase their sensitivity to radiotherapy and/or chemotherapy.
Collapse
Affiliation(s)
- Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
- Department of Medical Biology, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Correspondence: ; Tel.: +31-638-639-561
| | - Vashendriya V.V. Hira
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
| | - Amber J. van Dijck
- Department of Medical Biology, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Metka Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
| | - Remco J. Molenaar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna Pot 111, 1000 Ljubljana, Slovenia; (V.V.V.H.); (M.N.); (B.B.); (R.J.M.)
- Department of Medical Oncology, Amsterdam UMC Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
48
|
Li G, Chen TW, Nickel AC, Muhammad S, Steiger HJ, Tzaridis T, Hänggi D, Zeidler R, Zhang W, Kahlert UD. Carbonic Anhydrase XII is a Clinically Significant, Molecular Tumor-Subtype Specific Therapeutic Target in Glioma with the Potential to Combat Invasion of Brain Tumor Cells. Onco Targets Ther 2021; 14:1707-1718. [PMID: 33692626 PMCID: PMC7939492 DOI: 10.2147/ott.s300623] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background The metabolic enzyme carbonic anhydrase 12 (CA12/CAXII) emerges as a promising cancer therapeutic target with drug development projects underway. Previous reports proposed the relevance of CA12 in the context of glioma but are limited in patient data quantity, ignore ethnic diversity of patients or rely on semi-quantitative, thereby out of date, methodology. Moreover, little is known on the association of CA12 to brain tumor stemness or on the effect of anti-CAXII-directed monotherapies on glioma stem cells (GSCs), in particular their response regarding mesenchymal differentiation status. Methods We performed in silico analysis on three independent, large-scale patient datasets interrogating state of the art molecular diagnostics alongside clinical outcomes. We analyzed CAXII abundance on a collection of GSCs and functionally tested their response to exposure to CAXII blocking antibody 6A10. Results CA12 is highly expressed in glial tumors compared with normal tissue and predicts for poor clinical course of tumor patients. CA12 expression in glioblastoma significantly correlates with clinically established, molecular markers of IDH1WT DNA, WHO grade IV or absence of 1p/19q chromosome arm co-deletion. Furthermore, tumors with elevated CA12 cluster into the mesenchymal transcription subclass of the disease. CAXII abundance in different GSCs ranges from almost absent to high levels and does not correlate to stem cell marker CD133/AC133 cell surface expression. Moreover, aiming to pharmacologically block CAXII in our cells with antibody 6A10 caused significant functional response only in one of the tested GSCs models, featuring suppression of cell invasion accompanied by reduction of ZEB1 protein and other stem cell markers. Conclusion CA12 represents a clinically relevant and molecular brain tumor-subtype specific therapeutic target. Our correlative data from experimental and clinical samples does not support CA12/CAXII to be GSC specific. 6A10 possesses promising potential to impede the invasive capacity of glioma cells and supports the emerging concept that CAXII interacts with cancer EMT programs. However, further mechanistic studies are required to comprehensively assess the therapeutic potential of 6A10 and to identify different resistance mechanisms of GSCs.
Collapse
Affiliation(s)
- Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| | - Ting-Wei Chen
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Ann-Christin Nickel
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Sajjad Muhammad
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hans-Jakob Steiger
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Theophilos Tzaridis
- Division of Clinical Neurooncology, Department of Neurology and Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, 53127, Germany.,Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Daniel Hänggi
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Reinhard Zeidler
- Department for Otorhinolaryngology, Klinikum der Universität München (LMU), Munich, Germany
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, People's Republic of China
| | - Ulf Dietrich Kahlert
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China.,Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
49
|
Jones CL, Inguva A, Jordan CT. Targeting Energy Metabolism in Cancer Stem Cells: Progress and Challenges in Leukemia and Solid Tumors. Cell Stem Cell 2021; 28:378-393. [PMID: 33667359 PMCID: PMC7951949 DOI: 10.1016/j.stem.2021.02.013] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malignant stem cells have long been considered a key therapeutic target in leukemia. Therapeutic strategies designed to target the fundamental biology of leukemia stem cells while sparing normal hematopoietic cells may provide better outcomes for leukemia patients. One process in leukemia stem cell biology that has intriguing therapeutic potential is energy metabolism. In this article we discuss the metabolic properties of leukemia stem cells and how targeting energy metabolism may provide more effective therapeutic regimens for leukemia patients. In addition, we highlight the similarities and differences in energy metabolism between leukemia stem cells and malignant stem cells from solid tumors.
Collapse
Affiliation(s)
- Courtney L Jones
- Princess Margaret Cancer Centre, 101 College St. Toronto, ON M5G 1L7, Canada
| | - Anagha Inguva
- Division of Hematology, University of Colorado, 12700 East 19th Ave., Aurora, CO 80045, USA
| | - Craig T Jordan
- Division of Hematology, University of Colorado, 12700 East 19th Ave., Aurora, CO 80045, USA.
| |
Collapse
|
50
|
Giesen B, Nickel AC, Barthel J, Kahlert UD, Janiak C. Augmented Therapeutic Potential of Glutaminase Inhibitor CB839 in Glioblastoma Stem Cells Using Gold Nanoparticle Delivery. Pharmaceutics 2021; 13:295. [PMID: 33672398 PMCID: PMC7926460 DOI: 10.3390/pharmaceutics13020295] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Gold nanoparticles (Au NPs) are studied as delivery systems to enhance the effect of the glutaminase1 inhibitor CB839, a promising drug candidate already in clinical trials for tumor treatments. Au NPs were synthesized using a bottom-up approach and covered with polymers able to bind CB839 as a Au-polymer-CB839 conjugate. The drug loading efficiency (DLE) was determined using high-performance liquid chromatography and characterization of the CB839-loaded NPs was done with various microscopic and spectroscopic methods. Despite the chemical inertness of CB839, Au NPs were efficient carriers with a DLE of up to 12%, depending on the polymer used. The therapeutic effect of CB839 with and without Au was assessed in vitro in 2D and 3D glioblastoma (GBM) cell models using different assays based on the colony formation ability of GBM stem cells (GSCs). To avoid readout disturbances from the Au metal, viability methods which do not require optical detection were hereby optimized. These showed that Au NP delivery increased the efficacy of CB839 in GSCs, compared to CB839 alone. Fluorescent microscopy proved successful NP penetration into the GSCs. With this first attempt to combine CB839 with Au nanotechnology, we hope to overcome delivery hurdles of this pharmacotherapy and increase bioavailability in target sites.
Collapse
Affiliation(s)
- Beatriz Giesen
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-Universität Düsseldorf, 40204 Düsseldorf, Germany;
| | - Ann-Christin Nickel
- Klinik für Neurochirurgie, Medizinische Fakultät, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany;
| | - Juri Barthel
- Ernst Ruska-Centrum für Mikroskopie und Spektroskopie mit Elektronen (ER-C 2), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany;
| | - Ulf Dietrich Kahlert
- Klinik für Neurochirurgie, Medizinische Fakultät, Universitätsklinikum Düsseldorf, 40225 Düsseldorf, Germany;
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), 40225 Düsseldorf, Germany
| | - Christoph Janiak
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine-Universität Düsseldorf, 40204 Düsseldorf, Germany;
| |
Collapse
|