1
|
Šofranko J, Gondáš E, Murín R. Application of the Hydrophilic Interaction Liquid Chromatography (HILIC-MS) Novel Protocol to Study the Metabolic Heterogeneity of Glioblastoma Cells. Metabolites 2024; 14:297. [PMID: 38921432 PMCID: PMC11205371 DOI: 10.3390/metabo14060297] [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: 04/09/2024] [Revised: 05/13/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Glioblastoma is a highly malignant brain tumor consisting of a heterogeneous cellular population. The transformed metabolism of glioblastoma cells supports their growth and division on the background of their milieu. One might hypothesize that the transformed metabolism of a primary glioblastoma could be well adapted to limitations in the variety and number of substrates imported into the brain parenchyma and present it their microenvironment. Additionally, the phenotypic heterogeneity of cancer cells could promote the variations among their metabolic capabilities regarding the utilization of available substrates and release of metabolic intermediates. With the aim to identify the putative metabolic footprint of different types of glioblastoma cells, we exploited the possibility for separation of polar and ionic molecules present in culture media or cell lysates by hydrophilic interaction liquid chromatography (HILIC). The mass spectrometry (MS) was then used to identify and quantify the eluted compounds. The introduced method allows the detection and quantification of more than 150 polar and ionic metabolites in a single run, which may be present either in culture media or cell lysates and provide data for polaromic studies within metabolomics. The method was applied to analyze the culture media and cell lysates derived from two types of glioblastoma cells, T98G and U118. The analysis revealed that even both types of glioblastoma cells share several common metabolic aspects, and they also exhibit differences in their metabolic capability. This finding agrees with the hypothesis about metabolic heterogeneity of glioblastoma cells. Furthermore, the combination of both analytical methods, HILIC-MS, provides a valuable tool for metabolomic studies based on the simultaneous identification and quantification of a wide range of polar and ionic metabolites-polaromics.
Collapse
Affiliation(s)
- Jakub Šofranko
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia
| | - Eduard Gondáš
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia
| | - Radovan Murín
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovakia
| |
Collapse
|
2
|
Effectiveness of Flattening-Filter-Free versus Flattened Beams in V79 and Glioblastoma Patient-Derived Stem-like Cells. Int J Mol Sci 2023; 24:ijms24021107. [PMID: 36674623 PMCID: PMC9861147 DOI: 10.3390/ijms24021107] [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: 11/03/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Literature data on the administration of conventional high-dose beams with (FF) or without flattening filters (FFF) show conflicting results on biological effects at the cellular level. To contribute to this field, we irradiated V79 Chinese hamster lung fibroblasts and two patient-derived glioblastoma stem-like cell lines (GSCs-named #1 and #83) using a clinical 10 MV accelerator with FF (at 4 Gy/min) and FFF (at two dose rates 4 and 24 Gy/min). Cell killing and DNA damage induction, determined using the γ-H2AX assay, and gene expression were studied. No significant differences in the early survival of V79 cells were observed as a function of dose rates and FF or FFF beams, while a trend of reduction in late survival was observed at the highest dose rate with the FFF beam. GSCs showed similar survival levels as a function of dose rates, both delivered in the FFF regimen. The amount of DNA damage measured for both dose rates after 2 h was much higher in line #1 than in line #83, with statistically significant differences between the two dose rates only in line #83. The gene expression analysis of the two GSC lines indicates gene signatures mimicking the prognosis of glioblastoma (GBM) patients derived from a public database. Overall, the results support the current use of FFF and highlight the possibility of identifying patients with candidate gene signatures that could benefit from irradiation with FFF beams at a high dose rate.
Collapse
|
3
|
Characterization of Glioblastoma Cells Response to Regorafenib. Cancers (Basel) 2022; 14:cancers14246193. [PMID: 36551679 PMCID: PMC9777191 DOI: 10.3390/cancers14246193] [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: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma (GBM), the most malignant primary brain tumor in adults. Although not frequent, it has a relevant social impact because the peak incidence coincides with the age of professional maturity. A number of novel treatments have been proposed, yet clinical trials have been disappointing. Recently, a phase II clinical trial (REGOMA) demonstrated that the multikinase inhibitor regorafenib significantly increased the median overall survival (OS) of GBM patients when compared to lomustine-treated patients. On this basis, the National Comprehensive Cancer Network (NCCN) 2020 Guidelines included regorafenib as a preferred regimen in relapsed GBM treatment. Despite the use in GBM patients' therapy, little is known about the molecular mechanisms governing regorafenib effectiveness on the GBM tumor. Here we report an in vitro characterization of GBM tumor cells' response to regorafenib, performed both on cell lines and on patient-derived glioma stem cells (GSCs). Overall, regorafenib significantly reduced cell growth of 2D tumor cell cultures and of 3D tumor spheroids. Strikingly, this effect was accompanied by transcriptional regulation of epithelial to mesenchymal transition (EMT) genes and by an increased ability of surviving tumor cells to invade the surrounding matrix. Taken together, our data suggest that regorafenib limits cell growth, however, it might induce an invasive phenotype.
Collapse
|
4
|
Glioblastoma Stem-Like Cells (GSCs) with Mesenchymal Signature: Lipid Profiles of Mobile Lipids Obtained with MRS before and after Radio/Chemical Treatments. Biomolecules 2022; 12:biom12081051. [PMID: 36008944 PMCID: PMC9405836 DOI: 10.3390/biom12081051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/15/2022] [Accepted: 07/26/2022] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma is the most common and lethal primary malignant brain tumor in adults. Glioblastoma stem cells (GSCs) promote and are responsible for glioblastoma intratumoral heterogeneity and therapy resistance, due to their two main features: self-renewal and differentiation. Lipids have important biological and physiological functions that are critical for understanding the regulation and control of stem cell fate; lipid metabolism and related unsaturation levels play a possible role as the target of therapeutics to overcome glioblastoma radioresistance. This paper aimed at an in-depth analysis of 13 GSC mesenchymal (MES) lines, two subclones, and a stabilized glioblastoma line (T98G) by magnetic resonance spectroscopy (MRS). Particularly, 2D MRS was used to investigate lipid unsaturation behavior during growth in culture and after treatment with etomoxir and photon beams. MES lines, although belonging to the same genetic and metabolic cluster, showed metabolic heterogeneity when observed by MRS, focusing on lipid signals. Nonetheless, the observed unsaturation level stability for two representative lines after stressful treatments suggests unusual robustness of the unsaturation levels for each line, as a peculiar and intrinsic characteristic of GSCs.
Collapse
|
5
|
Effects of the Combined Treatment with a G-Quadruplex-Stabilizing Ligand and Photon Beams on Glioblastoma Stem-like Cells: A Magnetic Resonance Study. Int J Mol Sci 2021; 22:ijms222312709. [PMID: 34884511 PMCID: PMC8657890 DOI: 10.3390/ijms222312709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiforme is a malignant primary brain tumor with a poor prognosis and high rates of chemo-radiotherapy failure, mainly due to a small cell fraction with stem-like properties (GSCs). The mechanisms underlying GSC response to radiation need to be elucidated to enhance sensitivity to treatments and to develop new therapeutic strategies. In a previous study, two GSC lines, named line #1 and line #83, responded differently to carbon ions and photon beams, with the differences likely attributable to their own different metabolic fingerprint rather than to radiation type. Data from the literature showed the capability of RHPS4, a G-quadruplex stabilizing ligand, to sensitize the glioblastoma radioresistant U251MG cells to X-rays. The combined metabolic effect of ligand #190, a new RHPS4-derivative showing reduced cardiotoxicity, and a photon beam has been monitored by magnetic resonance (MR) spectroscopy for the two GSC lines, #1 and #83, to reveal whether a synergistic response occurs. MR spectra from both lines were affected by single and combined treatments, but the variations of the analysed metabolites were statistically significant mainly in line #1, without synergistic effects due to combination. The multivariate analysis of ten metabolites shows a separation between control and treated samples in line #1 regardless of treatment type, while separation was not detected in line #83.
Collapse
|
6
|
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: 8] [Impact Index Per Article: 2.7] [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
|
7
|
Palma A, Grande S, Ricci-Vitiani L, Luciani AM, Buccarelli M, Biffoni M, Dini V, Cirrone GAP, Ciocca M, Guidoni L, Pallini R, Viti V, Rosi A. Different Mechanisms Underlie the Metabolic Response of GBM Stem-Like Cells to Ionizing Radiation: Biological and MRS Studies on Effects of Photons and Carbon Ions. Int J Mol Sci 2020; 21:ijms21145167. [PMID: 32708312 PMCID: PMC7404344 DOI: 10.3390/ijms21145167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a malignant primary brain tumor with very poor prognosis, high recurrence rate, and failure of chemo-radiotherapy, mainly due to a small fraction of cells with stem-like properties (GSCs). To study the mechanisms of GSCs resistance to radiation, two GSC lines, named line #1 and line #83, with different metabolic patterns and clinical outcome, were irradiated with photon beams and carbon ions and assessed by 1H Magnetic Resonance Spectroscopy (MRS). Both irradiation modalities induced early cytotoxic effects in line #1 with small effects on cell cycle, whereas a proliferative G2/M cytostatic block was observed in line #83. MR spectroscopy signals from mobile lipids (ML) increased in spectra of line #1 after photon and C-ion irradiation with effects on lipid unsaturation level, whereas no effects were detected in line #83 spectra. Gamma-Aminobutyric Acid (GABA), glutamic acid (glu) and Phosphocreatine (pCr) signals showed a significant variation only for line #1 after carbon ion irradiation. Glucose (glc) level and lactate (Lac) extrusion behaved differently in the two lines. Our findings suggest that the differences in irradiation response of GSCs #1 and #83 lines are likely attributable to their different metabolic fingerprint rather than to the different radiation types.
Collapse
Affiliation(s)
- Alessandra Palma
- National Centre for Innovative Technologies in Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.P.); (S.G.); (A.M.L.); (V.D.); (L.G.); (V.V.)
| | - Sveva Grande
- National Centre for Innovative Technologies in Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.P.); (S.G.); (A.M.L.); (V.D.); (L.G.); (V.V.)
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (L.R.-V.); (M.B.); (M.B.)
| | - Anna Maria Luciani
- National Centre for Innovative Technologies in Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.P.); (S.G.); (A.M.L.); (V.D.); (L.G.); (V.V.)
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (L.R.-V.); (M.B.); (M.B.)
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (L.R.-V.); (M.B.); (M.B.)
| | - Valentina Dini
- National Centre for Innovative Technologies in Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.P.); (S.G.); (A.M.L.); (V.D.); (L.G.); (V.V.)
- Istituto Nazionale di Fisica Nucleare INFN Sez. di Roma, 00185 Rome, Italy
| | - Giuseppe A. P. Cirrone
- National Institute for Nuclear Physics, Laboratori Nazionali del Sud, INFN-LNS, 95123 Catania, Italy;
| | - Mario Ciocca
- Centro Nazionale di Adroterapia Oncologica (CNAO)-National Center for Oncological Hadrontherapy, 27100 Pavia, Italy;
| | - Laura Guidoni
- National Centre for Innovative Technologies in Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.P.); (S.G.); (A.M.L.); (V.D.); (L.G.); (V.V.)
| | - Roberto Pallini
- Department of Neuroscience, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Vincenza Viti
- National Centre for Innovative Technologies in Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.P.); (S.G.); (A.M.L.); (V.D.); (L.G.); (V.V.)
| | - Antonella Rosi
- National Centre for Innovative Technologies in Public Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.P.); (S.G.); (A.M.L.); (V.D.); (L.G.); (V.V.)
- Correspondence: ; Tel.: +39-06-49903159
| |
Collapse
|
8
|
Papale M, Buccarelli M, Mollinari C, Russo MA, Pallini R, Ricci-Vitiani L, Tafani M. Hypoxia, Inflammation and Necrosis as Determinants of Glioblastoma Cancer Stem Cells Progression. Int J Mol Sci 2020; 21:ijms21082660. [PMID: 32290386 PMCID: PMC7215563 DOI: 10.3390/ijms21082660] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor hypoxic microenvironment causes hypoxia inducible factor 1 alpha (HIF-1α) activation and necrosis with alarmins release. Importantly, HIF-1α also controls the expression of alarmin receptors in tumor cells that can bind to and be activated by alarmins. Human tumor tissues possess 1–2% of cancer stem cells (CSCs) residing in hypoxic niches and responsible for the metastatic potential of tumors. Our hypothesis is that hypoxic CSCs express alarmin receptors that can bind alarmins released during necrosis, an event favoring CSCs migration. To investigate this aspect, glioblastoma stem-like cell (GSC) lines were kept under hypoxia to determine the expression of hypoxic markers as well as receptor for advanced glycation end products (RAGE). The presence of necrotic extracts increased migration, invasion and cellular adhesion. Importantly, HIF-1α inhibition by digoxin or acriflavine prevented the response of GSCs to hypoxia alone or plus necrotic extracts. In vivo, GSCs injected in one brain hemisphere of NOD/SCID mice were induced to migrate to the other one in which a necrotic extract was previously injected. In conclusion, our results show that hypoxia is important not only for GSCs maintenance but also for guiding their response to external necrosis. Inhibition of hypoxic pathway may therefore represent a target for preventing brain invasion by glioblastoma stem cells (GSCs).
Collapse
Affiliation(s)
- Marco Papale
- Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy;
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome Italy; (M.B.); (L.R.-V.)
| | - Cristiana Mollinari
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy;
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Matteo A. Russo
- IRCCS San Raffaele Pisana, 00163 Rome, Italy;
- MEBIC Consortium, San Raffaele Open University, 00166 Rome, Italy
| | - Roberto Pallini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Institute of Neurosurgery, Catholic University School of Medicine, 00168 Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome Italy; (M.B.); (L.R.-V.)
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy;
- Correspondence: ; Tel.: +39-06-49918234
| |
Collapse
|
9
|
Gandía-González ML, Cerdán S, Barrios L, López-Larrubia P, Feijoó PG, Palpan A, Roda JM, Solivera J. Assessment of Overall Survival in Glioma Patients as Predicted by Metabolomic Criteria. Front Oncol 2019; 9:328. [PMID: 31134147 PMCID: PMC6524167 DOI: 10.3389/fonc.2019.00328] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/11/2019] [Indexed: 11/17/2022] Open
Abstract
Objective: We assess the efficacy of the metabolomic profile from glioma biopsies in providing estimates of postsurgical Overall Survival in glioma patients. Methods: Tumor biopsies from 46 patients bearing gliomas, obtained neurosurgically in the period 1992–1998, were analyzed by high resolution 1H magnetic resonance spectroscopy (HR- 1H MRS), following retrospectively individual postsurgical Overall Survival up to 720 weeks. Results: The Overall Survival profile could be resolved in three groups; Short (shorter than 52 weeks, n = 19), Intermediate (between 53 and 364 weeks, n = 19) or Long (longer than 365 weeks, n = 8), respectively. Classical histopathological analysis assigned WHO grades II–IV to every biopsy but notably, some patients with low grade glioma depicted unexpectedly Short Overall Survival, while some patients with high grade glioma, presented unpredictably Long Overall Survival. To explore the reasons underlying these different responses, we analyzed HR-1H MRS spectra from acid extracts of the same biopsies, to characterize the metabolite patterns associated to OS predictions. Poor prognosis was found in biopsies with higher contents of alanine, acetate, glutamate, total choline, phosphorylcholine, and glycine, while more favorable prognosis was achieved in biopsies with larger contents of total creatine, glycerol-phosphorylcholine, and myo-inositol. We then implemented a multivariate analysis to identify hierarchically the influence of metabolomic biomarkers on OS predictions, using a Classification Regression Tree (CRT) approach. The CRT based in metabolomic biomarkers grew up to three branches and split into eight nodes, predicting correctly the outcome of 94.7% of the patients in the Short Overall Survival group, 78.9% of the patients in the Intermediate Overall Survival group, and 75% of the patients in the Long Overall Survival group, respectively. Conclusion: Present results indicate that metabolic profiling by HR-1H MRS improves the Overall Survival predictions derived exclusively from classical histopathological gradings, thus favoring more precise therapeutic decisions.
Collapse
Affiliation(s)
| | - Sebastián Cerdán
- Institute of Biomedical Research "Alberto Sols" CSIC/UAM, Madrid, Spain
| | | | | | - Pablo G Feijoó
- Department of Neurosurgery, Hospital Universitario La Paz, Madrid, Spain
| | - Alexis Palpan
- Department of Neurosurgery, Hospital Universitario La Paz, Madrid, Spain
| | - José M Roda
- Department of Neurosurgery, Hospital Universitario La Paz, Madrid, Spain
| | - Juan Solivera
- Department of Neurosurgery, University Hospital Reina Sofía, Córdoba, Spain
| |
Collapse
|
10
|
Palma A, Grande S, Ricci-Vitiani L, Luciani A, Buccarelli M, Biffoni M, Molinari A, Calcabrini A, D’Amore E, Guidoni L, Pallini R, Viti V, Rosi A. 307. Metabolic heterogeneity among Glioblastoma stem-like cells reflects differences in response to drug treatments. Phys Med 2018. [DOI: 10.1016/j.ejmp.2018.04.316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
11
|
Metabolic Heterogeneity Evidenced by MRS among Patient-Derived Glioblastoma Multiforme Stem-Like Cells Accounts for Cell Clustering and Different Responses to Drugs. Stem Cells Int 2018. [PMID: 29531533 PMCID: PMC5835274 DOI: 10.1155/2018/3292704] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Clustering of patient-derived glioma stem-like cells (GSCs) through unsupervised analysis of metabolites detected by magnetic resonance spectroscopy (MRS) evidenced three subgroups, namely clusters 1a and 1b, with high intergroup similarity and neural fingerprints, and cluster 2, with a metabolism typical of commercial tumor lines. In addition, subclones generated by the same GSC line showed different metabolic phenotypes. Aerobic glycolysis prevailed in cluster 2 cells as demonstrated by higher lactate production compared to cluster 1 cells. Oligomycin, a mitochondrial ATPase inhibitor, induced high lactate extrusion only in cluster 1 cells, where it produced neutral lipid accumulation detected as mobile lipid signals by MRS and lipid droplets by confocal microscopy. These results indicate a relevant role of mitochondrial fatty acid oxidation for energy production in GSCs. On the other hand, further metabolic differences, likely accounting for different therapy responsiveness observed after etomoxir treatment, suggest that caution must be used in considering patient treatment with mitochondria FAO blockers. Metabolomics and metabolic profiling may contribute to discover new diagnostic or prognostic biomarkers to be used for personalized therapies.
Collapse
|
12
|
Abstract
Cell metabolism is a key determinant factor for the pluripotency and fate commitment of Stem Cells (SCs) during development, ageing, pathological onset and progression. We derived and cultured selected subpopulations of rodent fetal, postnatal, adult Neural SCs (NSCs) and postnatal glial progenitors, Olfactory Ensheathing Cells (OECs), respectively from the subventricular zone (SVZ) and the olfactory bulb (OB). Cell lysates were analyzed by proton Nuclear Magnetic Resonance (1H-NMR) spectroscopy leading to metabolites identification and quantitation. Subsequent multivariate analysis of NMR data by Principal Component Analysis (PCA), and Partial Least Square Discriminant Analysis (PLS-DA) allowed data reduction and cluster analysis. This strategy ensures the definition of specific features in the metabolic content of phenotypically similar SCs sharing a common developmental origin. The metabolic fingerprints for selective metabolites or for the whole spectra demonstrated enhanced peculiarities among cell types. The key result of our work is a neat divergence between OECs and the remaining NSC cells. We also show that statistically significant differences for selective metabolites characterizes NSCs of different ages. Finally, the retrived metabolome in cell cultures correlates to the physiological SC features, thus allowing an integrated bioengineering approach for biologic fingerprints able to dissect the (neural) SC molecular specificities.
Collapse
|
13
|
Adamo A, Fiore D, De Martino F, Roscigno G, Affinito A, Donnarumma E, Puoti I, Ricci Vitiani L, Pallini R, Quintavalle C, Condorelli G. RYK promotes the stemness of glioblastoma cells via the WNT/ β-catenin pathway. Oncotarget 2017; 8:13476-13487. [PMID: 28086236 PMCID: PMC5355113 DOI: 10.18632/oncotarget.14564] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/27/2016] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is characterized by a strong self-renewal potential and a poor differentiation state. Since receptor-like tyrosine kinase (RYK) activates the WNT/β-catenin pathway essential for cancer stem cell maintenance, we evaluated its contribution in conferring stemness to GBM cells. Here, we report that Ryk (related-to-receptor tyrosine kinase), an atypical tyrosine kinase receptor, is upregulated in samples from GBM patients as well as in GSCs. Ryk overexpression confers stemness properties to GBM cells through the modulation of the canonical Wnt signaling and by promoting the activation of pluripotency-related transcription factor circuitry and neurosphere formation ability. In contrast, siRNA-mediated knockdown of Ryk expression suppresses this stem-like phenotype. Rescue experiments reveal that stemness-promoting activity of Ryk is attributable, at least in part, to β-catenin stabilization. Furthermore, Ryk overexpression improves cell motility and anchorage independent cell growth. Taken together, our findings demonstrate that Ryk promotes stem cell-like and tumorigenic features to glioma cells its essential for the maintenance of GSCs and could be a target of novel therapies.
Collapse
Affiliation(s)
- Assunta Adamo
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Danilo Fiore
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Fabio De Martino
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Giuseppina Roscigno
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy.,IEOS, CNR, Naples, Italy
| | - Alessandra Affinito
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | | | - Ilaria Puoti
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Lucia Ricci Vitiani
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberto Pallini
- Institute of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cristina Quintavalle
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Gerolama Condorelli
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy.,IEOS, CNR, Naples, Italy
| |
Collapse
|
14
|
Dong BW, Qin GM, Luo Y, Mao JS. Metabolic enzymes: key modulators of functionality in cancer stem-like cells. Oncotarget 2017; 8:14251-14267. [PMID: 28009990 PMCID: PMC5355174 DOI: 10.18632/oncotarget.14041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/07/2016] [Indexed: 12/22/2022] Open
Abstract
Cancer Stem-like Cells (CSCs) are a subpopulation of cancer cells with self-renewal capacity and are important for the initiation, progression and recurrence of cancer diseases. The metabolic profile of CSCs is consistent with their stem-like properties. Studies have indicated that enzymes, the main regulators of cellular metabolism, dictate functionalities of CSCs in both catalysis-dependent and catalysis-independent manners. This paper reviews diverse studies of metabolic enzymes, and describes the effects of these enzymes on metabolic adaptation, gene transcription and signal transduction, in CSCs.
Collapse
Affiliation(s)
- Bo-Wen Dong
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Guang-Ming Qin
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Luo
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Jian-Shan Mao
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
Fontana F, Ge X, Su X, Hathi D, Xiang J, Cenci S, Civitelli R, Shoghi KI, Akers WJ, D'avignon A, Weilbaecher KN, Shokeen M. Evaluating Acetate Metabolism for Imaging and Targeting in Multiple Myeloma. Clin Cancer Res 2016; 23:416-429. [PMID: 27486177 DOI: 10.1158/1078-0432.ccr-15-2134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE We hypothesized that in multiple myeloma cells (MMC), high membrane biosynthesis will induce acetate uptake in vitro and in vivo Here, we studied acetate metabolism and targeting in MMC in vitro and tested the efficacy of 11C-acetate-positron emission tomography (PET) to detect and quantitatively image myeloma treatment response in vivo EXPERIMENTAL DESIGN: Acetate fate tracking using 13C-edited-1H NMR (nuclear magnetic resonance) was performed to study in vitro acetate uptake and metabolism in MMC. Effects of pharmacological modulation of acetate transport or acetate incorporation into lipids on MMC cell survival and viability were assessed. Preclinical mouse MM models of subcutaneous and bone tumors were evaluated using 11C-acetate-PET/CT imaging and tissue biodistribution. RESULTS In vitro, NMR showed significant uptake of acetate by MMC and acetate incorporation into intracellular metabolites and membrane lipids. Inhibition of lipid synthesis and acetate transport was toxic to MMC, while sparing resident bone cells or normal B cells. In vivo, 11C-acetate uptake by PET imaging was significantly enhanced in subcutaneous and bone MMC tumors compared with unaffected bone or muscle tissue. Likewise, 11C-acetate uptake was significantly reduced in MM tumors after treatment. CONCLUSIONS Uptake of acetate from the extracellular environment was enhanced in MMC and was critical to cellular viability. 11C-Acetate-PET detected the presence of myeloma cells in vivo, including uptake in intramedullary bone disease. 11C-Acetate-PET also detected response to therapy in vivo Our data suggested that acetate metabolism and incorporation into lipids was crucial to MM cell biology and that 11C-acetate-PET is a promising imaging modality for MM. Clin Cancer Res; 23(2); 416-29. ©2016 AACR.
Collapse
Affiliation(s)
- Francesca Fontana
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Xia Ge
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Xinming Su
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Deep Hathi
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Jingyu Xiang
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Simone Cenci
- San Raffaele Scientific Institute (SRSI), Division of Genetics and Cell Biology, and Università Vita-Salute San Raffaele, Milano, Italy
| | - Roberto Civitelli
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, Missouri
| | - Kooresh I Shoghi
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Walter J Akers
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Andre D'avignon
- Department of Chemistry, Washington University, St. Louis, Missouri
| | - Katherine N Weilbaecher
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, Missouri.
| | - Monica Shokeen
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
16
|
Kahlert UD, Mooney SM, Natsumeda M, Steiger HJ, Maciaczyk J. Targeting cancer stem-like cells in glioblastoma and colorectal cancer through metabolic pathways. Int J Cancer 2016; 140:10-22. [PMID: 27389307 DOI: 10.1002/ijc.30259] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/29/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Cancer stem-like cells (CSCs) are thought to be the main cause of tumor occurrence, progression and therapeutic resistance. Strong research efforts in the last decade have led to the development of several tailored approaches to target CSCs with some very promising clinical trials underway; however, until now no anti-CSC therapy has been approved for clinical use. Given the recent improvement in our understanding of how onco-proteins can manipulate cellular metabolic networks to promote tumorigenesis, cancer metabolism research may well lead to innovative strategies to identify novel regulators and downstream mediators of CSC maintenance. Interfering with distinct stages of CSC-associated metabolics may elucidate novel, more efficient strategies to target this highly malignant cell population. Here recent discoveries regarding the metabolic properties attributed to CSCs in glioblastoma (GBM) and malignant colorectal cancer (CRC) were summarized. The association between stem cell markers, the response to hypoxia and other environmental stresses including therapeutic insults as well as developmentally conserved signaling pathways with alterations in cellular bioenergetic networks were also discussed. The recent developments in metabolic imaging to identify CSCs were also summarized. This summary should comprehensively update basic and clinical scientists on the metabolic traits of CSCs in GBM and malignant CRC.
Collapse
Affiliation(s)
- U D Kahlert
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| | - S M Mooney
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - M Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - H-J Steiger
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| | - J Maciaczyk
- Department of Neurosurgery, Heinrich-Heine University Medical Center, Düsseldorf, Germany
| |
Collapse
|
17
|
Palma A, Grande S, Luciani AM, Mlynárik V, Guidoni L, Viti V, Rosi A. Metabolic Study of Breast MCF-7 Tumor Spheroids after Gamma Irradiation by (1)H NMR Spectroscopy and Microimaging. Front Oncol 2016; 6:105. [PMID: 27200293 PMCID: PMC4848320 DOI: 10.3389/fonc.2016.00105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/13/2016] [Indexed: 12/14/2022] Open
Abstract
Multicellular tumor spheroids are an important model system to investigate the response of tumor cells to radio- and chemotherapy. They share more properties with the original tumor than cells cultured as 2D monolayers do, which helps distinguish the intrinsic properties of monolayer cells from those induced during cell aggregation in 3D spheroids. The paper investigates some metabolic aspects of small tumor spheroids of breast cancer and their originating MCF-7 cells, grown as monolayer, by means of high-resolution (HR) (1)H NMR spectroscopy and MR microimaging before and after gamma irradiation. The spectra of spheroids were characterized by higher intensity of mobile lipids, mostly neutral lipids, and glutamine (Gln) signals with respect to their monolayer cells counterpart, mainly owing to the lower oxygen supply in spheroids. Morphological changes of small spheroids after gamma-ray irradiation, such as loss of their regular shape, were observed by MR microimaging. Lipid signal intensity increased after irradiation, as evidenced in both MR localized spectra of the single spheroid and in HR NMR spectra of spheroid suspensions. Furthermore, the intense Gln signal from spectra of irradiated spheroids remained unchanged, while the low Gln signal observed in monolayer cells increased after irradiation. Similar results were observed in cells grown in hypoxic conditions. The different behavior of Gln in 2D monolayers and in 3D spheroids supports the hypothesis that a lower oxygen supply induces both an upregulation of Gln synthetase and a downregulation of glutaminases with the consequent increase in Gln content, as already observed under hypoxic conditions. The data herein indicate that (1)H NMR spectroscopy can be a useful tool for monitoring cell response to different constraints. The use of spheroid suspensions seems to be a feasible alternative to localized spectroscopy since similar effects were found after radiation treatment.
Collapse
Affiliation(s)
- Alessandra Palma
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy; INFN Sezione di Roma, Rome, Italy
| | - Sveva Grande
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy; INFN Sezione di Roma, Rome, Italy
| | - Anna Maria Luciani
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy; INFN Sezione di Roma, Rome, Italy
| | - Vladimír Mlynárik
- Department of Biomedical Imaging and Image-Guided Therapy, High-Field MR Center, Medical University of Vienna , Vienna , Austria
| | | | | | - Antonella Rosi
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy; INFN Sezione di Roma, Rome, Italy
| |
Collapse
|
18
|
Marziali G, Signore M, Buccarelli M, Grande S, Palma A, Biffoni M, Rosi A, D'Alessandris QG, Martini M, Larocca LM, De Maria R, Pallini R, Ricci-Vitiani L. Metabolic/Proteomic Signature Defines Two Glioblastoma Subtypes With Different Clinical Outcome. Sci Rep 2016; 6:21557. [PMID: 26857460 PMCID: PMC4746700 DOI: 10.1038/srep21557] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/22/2016] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma (GBM) is one of the deadliest human cancers. Because of the extremely unfavorable prognosis of GBM, it is important to develop more effective diagnostic and therapeutic strategies based on biologically and clinically relevant subclassification systems. Analyzing a collection of seventeen patient-derived glioblastoma stem-like cells (GSCs) by gene expression profiling, NMR spectroscopy and signal transduction pathway activation, we identified two GSC clusters, one characterized by a pro-neural-like phenotype and the other showing a mesenchymal-like phenotype. Evaluating the levels of proteins differentially expressed by the two GSC clusters in the TCGA GBM sample collection, we found that SRC activation is associated with a GBM subgroup showing better prognosis whereas activation of RPS6, an effector of mTOR pathway, identifies a subgroup with a worse prognosis. The two clusters are also differentiated by NMR spectroscopy profiles suggesting a potential prognostic stratification based on metabolic evaluation. Our data show that the metabolic/proteomic profile of GSCs is informative of the genomic/proteomic GBM landscape, which differs among tumor subtypes and is associated with clinical outcome.
Collapse
Affiliation(s)
- G Marziali
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - M Signore
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - M Buccarelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - S Grande
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy
| | - A Palma
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy
| | - M Biffoni
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - A Rosi
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Q G D'Alessandris
- nstitute of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - M Martini
- Institute of Anatomic Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - L M Larocca
- Institute of Anatomic Pathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - R De Maria
- Regina Elena National Cancer Institute, Rome, Italy
| | - R Pallini
- nstitute of Neurosurgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - L Ricci-Vitiani
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
19
|
Kahlert UD, Cheng M, Koch K, Marchionni L, Fan X, Raabe EH, Maciaczyk J, Glunde K, Eberhart CG. Alterations in cellular metabolome after pharmacological inhibition of Notch in glioblastoma cells. Int J Cancer 2015; 138:1246-55. [PMID: 26422827 DOI: 10.1002/ijc.29873] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 09/04/2015] [Accepted: 09/17/2015] [Indexed: 12/18/2022]
Abstract
Notch signaling can promote tumorigenesis in the nervous system and plays important roles in stem-like cancer cells. However, little is known about how Notch inhibition might alter tumor metabolism, particularly in lesions arising in the brain. The gamma-secretase inhibitor MRK003 was used to treat glioblastoma neurospheres, and they were subdivided into sensitive and insensitive groups in terms of canonical Notch target response. Global metabolomes were then examined using proton magnetic resonance spectroscopy, and changes in intracellular concentration of various metabolites identified which correlate with Notch inhibition. Reductions in glutamate were verified by oxidation-based colorimetric assays. Interestingly, the alkylating chemotherapeutic agent temozolomide, the mTOR-inhibitor MLN0128, and the WNT inhibitor LGK974 did not reduce glutamate levels, suggesting that changes to this metabolite might reflect specific downstream effects of Notch blockade in gliomas rather than general sequelae of tumor growth inhibition. Global and targeted expression analyses revealed that multiple genes important in glutamate homeostasis, including glutaminase, are dysregulated after Notch inhibition. Treatment with an allosteric inhibitor of glutaminase, compound 968, could slow glioblastoma growth, and Notch inhibition may act at least in part by regulating glutaminase and glutamate.
Collapse
Affiliation(s)
- Ulf D Kahlert
- Department of Pathology, Division of Neuropathology, Johns Hopkins Hospital, Baltimore, MD.,Department of Neurosurgery, University Medical Center, Forschungsgebaeude Pathologie, Düsseldorf, Germany
| | - Menglin Cheng
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD
| | - Katharina Koch
- Department of Neurosurgery, University Medical Center, Forschungsgebaeude Pathologie, Düsseldorf, Germany
| | - Luigi Marchionni
- Department of Oncology, Johns Hopkins Hospital, Baltimore, MD.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Xing Fan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Eric H Raabe
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD.,Department of Pediatric Oncology, Johns Hopkins Hospital, Baltimore, MD
| | - Jarek Maciaczyk
- Department of Neurosurgery, University Medical Center, Forschungsgebaeude Pathologie, Düsseldorf, Germany
| | - Kristine Glunde
- Division of Cancer Imaging Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Charles G Eberhart
- Department of Pathology, Division of Neuropathology, Johns Hopkins Hospital, Baltimore, MD
| |
Collapse
|
20
|
Pecchia I, Dini V, Ricci-Vitiani L, Biffoni M, Balduzzi M, Fratini E, Belli M, Campa A, Esposito G, Cirrone G, Romano F, Stancampiano C, Pelacchi F, Pallini R, Tabocchini MA. Glioblastoma stem cells: radiobiological response to ionising radiations of different qualities. RADIATION PROTECTION DOSIMETRY 2015; 166:374-378. [PMID: 25969527 DOI: 10.1093/rpd/ncv299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant primary brain tumour, with very poor prognosis. The high recurrence rate and failure of conventional treatments are expected to be related to the presence of radio-resistant cancer stem cells (CSCs) inside the tumour mass. CSCs can both self-renew and differentiate into the heterogeneous lineages of cancer cells. Recent evidence showed a higher effectiveness of C-ions and protons in inactivating CSCs, suggesting a potential advantage of Hadrontherapy compared with conventional radiotherapy for GBM treatment. To investigate the mechanisms involved in the molecular and cellular responses of CSCs to ionising radiations, two GBM stem cell (GSC) lines, named lines 1 and 83, which were derived from patients with different clinical outcomes and having different metabolic profiles (as shown by NMR spectroscopy), were irradiated with (137)Cs photons and with protons or C-ions of 62 MeV u(-1) in the dose range of 5-40 Gy. The biological effects investigated were: cell death, cell cycle progression, and DNA damage induction and repair. Preliminary results show a different response to ionising radiation between the two GSC lines for the different end points investigated. Further experiments are in progress to consolidate the data and to get more insights on the influence of radiation quality.
Collapse
Affiliation(s)
- I Pecchia
- Department of Technology and Health, Istituto Superiore di Sanità, viale Regina Elena 299, Rome 00161, Italy
| | - V Dini
- Department of Technology and Health, Istituto Superiore di Sanità, viale Regina Elena 299, Rome 00161, Italy Sez. Roma1-Gruppo collegato Sanità, INFN, Rome, Italy
| | - L Ricci-Vitiani
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - M Biffoni
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - M Balduzzi
- Sez. Roma1-Gruppo collegato Sanità, INFN, Rome, Italy ENEA-CASACCIA, Rome, Italy
| | - E Fratini
- Sez. Roma1-Gruppo collegato Sanità, INFN, Rome, Italy Museo storico della fisica e Centro Studi e Ricerche 'E. Fermi', Rome, Italy
| | - M Belli
- Sez. Roma1-Gruppo collegato Sanità, INFN, Rome, Italy
| | - A Campa
- Department of Technology and Health, Istituto Superiore di Sanità, viale Regina Elena 299, Rome 00161, Italy Sez. Roma1-Gruppo collegato Sanità, INFN, Rome, Italy
| | - G Esposito
- Department of Technology and Health, Istituto Superiore di Sanità, viale Regina Elena 299, Rome 00161, Italy Sez. Roma1-Gruppo collegato Sanità, INFN, Rome, Italy
| | - G Cirrone
- Laboratori Nazionali del Sud (LNS) - INFN, Catania, Italy
| | - F Romano
- Laboratori Nazionali del Sud (LNS) - INFN, Catania, Italy
| | - C Stancampiano
- Laboratori Nazionali del Sud (LNS) - INFN, Catania, Italy
| | - F Pelacchi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - R Pallini
- Department of Neurosurgery, Catholic University of Rome, Rome, Italy
| | - M A Tabocchini
- Department of Technology and Health, Istituto Superiore di Sanità, viale Regina Elena 299, Rome 00161, Italy Sez. Roma1-Gruppo collegato Sanità, INFN, Rome, Italy
| |
Collapse
|
21
|
Arnold JM, Choi WT, Sreekumar A, Maletić-Savatić M. Analytical strategies for studying stem cell metabolism. ACTA ACUST UNITED AC 2015. [PMID: 26213533 DOI: 10.1007/s11515-015-1357-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Owing to their capacity for self-renewal and pluripotency, stem cells possess untold potential for revolutionizing the field of regenerative medicine through the development of novel therapeutic strategies for treating cancer, diabetes, cardiovascular and neurodegenerative diseases. Central to developing these strategies is improving our understanding of biological mechanisms responsible for governing stem cell fate and self-renewal. Increasing attention is being given to the significance of metabolism, through the production of energy and generation of small molecules, as a critical regulator of stem cell functioning. Rapid advances in the field of metabolomics now allow for in-depth profiling of stem cells both in vitro and in vivo, providing a systems perspective on key metabolic and molecular pathways which influence stem cell biology. Understanding the analytical platforms and techniques that are currently used to study stem cell metabolomics, as well as how new insights can be derived from this knowledge, will accelerate new research in the field and improve future efforts to expand our understanding of the interplay between metabolism and stem cell biology.
Collapse
Affiliation(s)
- James M Arnold
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - William T Choi
- Program in Developmental Biology and Medical Scientist Training Program, Baylor College of Medicine; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Arun Sreekumar
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mirjana Maletić-Savatić
- Program in Developmental Biology and Medical Scientist Training Program, Baylor College of Medicine; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA ; Departments of Pediatrics-Neurology and Neuroscience, and Program in Structural and Computational Biology and Molecular Biophysics Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
22
|
CORAZZA ANDREA, MANCO LUIGI, SGHEDONI ROBERTO, IORI MAURO, NITROSI ANDREA, BERTOLINI MARCO, TURRA ALESSANDRO, FAINARDI ENRICO, GIAMPIERI ENRICO, REMONDINI DANIEL, CASTELLANI GASTONE. MODELING GLIOBLASTOMA RESPONSE TO RADIOTHERAPY BY COMBINING A TWO-COMPARTMENT KINETIC MODEL AND MULTIPARAMETRIC NMR DATA. J MECH MED BIOL 2015. [DOI: 10.1142/s0219519415400175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Glioblastoma are the most common and malignant primary brain tumor, and actual treatments consist of surgery (when possible), radiotherapy and chemotherapy. Recent discoveries in biology revealed the important role of radioresistant cancer stem cell in the tumor proliferation and also showed that differentiated tumor cells can revert to a stem-like state because of radiation. These discoveries can be used to create mathematical models to study and plan new optimized radiotherapy schedules. In literature, some models have already been developed on murine population. The aim of this study was to reproduce these models, to perform a sensitivity analysis to find the most sensitive parameters and to adapt them to standard schedules used with human patients. We found that the most sensitive parameters are those involving tumor cell proliferation, radio-sensibility and quiescence times of both stem and tumor cells.
Collapse
Affiliation(s)
- ANDREA CORAZZA
- Post-Graduate School in Medical Physics, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - LUIGI MANCO
- Post-Graduate School in Medical Physics, University of Bologna, via Massarenti 9, 40138 Bologna, Italy
| | - ROBERTO SGHEDONI
- Medical Physics Unit, Arcispedale Santa Maria Nuova Hospital IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - MAURO IORI
- Medical Physics Unit, Arcispedale Santa Maria Nuova Hospital IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - ANDREA NITROSI
- Medical Physics Unit, Arcispedale Santa Maria Nuova Hospital IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - MARCO BERTOLINI
- Medical Physics Unit, Arcispedale Santa Maria Nuova Hospital IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - ALESSANDRO TURRA
- Medical Physics Unit, Arcispedale Sant'Anna Hospital, Via Aldo Moro 8, 44124 Cona, Ferrara, Italy
| | - ENRICO FAINARDI
- Neuroradiology Unit, Department of Neurosciences and Rehabilitation, Arcispedale Sant'Anna Hospital, Via Aldo Moro 8, 44124 Cona, Ferrara, Italy
| | - ENRICO GIAMPIERI
- Physics and Astronomy Department, University of Bologna and INFN Sez. Bologna, viale Berti Pichat 6/2, 40138 Bologna, Italy
| | - DANIEL REMONDINI
- Physics and Astronomy Department, University of Bologna and INFN Sez. Bologna, viale Berti Pichat 6/2, 40138 Bologna, Italy
| | - GASTONE CASTELLANI
- Physics and Astronomy Department, University of Bologna and INFN Sez. Bologna, viale Berti Pichat 6/2, 40138 Bologna, Italy
| |
Collapse
|
23
|
Rosi A, Ricci-Vitiani L, Biffoni M, Grande S, Luciani AM, Palma A, Runci D, Cappellari M, De Maria R, Guidoni L, Pallini R, Viti V. (1) H NMR spectroscopy of glioblastoma stem-like cells identifies alpha-aminoadipate as a marker of tumor aggressiveness. NMR IN BIOMEDICINE 2015; 28:317-26. [PMID: 25581615 DOI: 10.1002/nbm.3254] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 11/12/2014] [Accepted: 12/03/2014] [Indexed: 05/24/2023]
Abstract
Patients suffering from glioblastoma multiforme (GBM) face a poor prognosis with median survival of about 14 months. High recurrence rate and failure of conventional treatments are attributed to the presence of GBM cells with stem-like properties (GSCs). Metabolite profiles of 42 GSC lines established from the tumor tissue of adult GBM patients were screened with (1) H NMR spectroscopy and compared with human neural progenitor cells from human adult olfactory bulb (OB-NPCs) and from the developing human brain (HNPCs). A first subset (n=12) of GSCs exhibited a dramatic accumulation of the metabolite α-aminoadipate (αAAD), product of the oxidation of α-aminoadipic semialdehyde catalyzed by the ALDH7A1 aldehyde dehydrogenase (ALDH) family in lysine catabolism. αAAD was low/not detectable in a second GSC subset (n=13) with the same neural metabolic profile as well as in a third GSC subset (n=17) characterized by intense lipid signals. Likewise, αAAD was not detected in the spectra of OB-NPCs or HNPCs. Inhibition of mitochondrial ATP synthase by oligomycin treatment revealed that the lysine degradative pathway leading to αAAD formation proceeds through saccharopine, as usually observed in developing brain. Survival curves indicated that high αAAD levels in GSCs significantly correlated with poor patient survival, similarly to prostate and non-small-cell-lung cancers, where activity of ALDH7A1 correlates with tumor aggressiveness.
Collapse
Affiliation(s)
- Antonella Rosi
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy; INFN Sezione di Roma, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Applying metabolomics to understand the aggressive phenotype and identify novel therapeutic targets in glioblastoma. Metabolites 2014; 4:740-50. [PMID: 25167383 PMCID: PMC4192690 DOI: 10.3390/metabo4030740] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 08/20/2014] [Accepted: 08/21/2014] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma continues to be an invariably fatal malignancy. The established approach for understanding the biology of these aggressive tumors in an effort to identify novel molecular targets has largely been genotype-based. Unfortunately, clinical gains offered by this level of understanding have been limited, largely based on the complex nature of signaling networks associated with tumorigenesis and the inability to delineate the key “functional” signaling pathways actually driving growth in an individual tumor. Metabolomics is the global quantitative assessment of endogenous metabolites within a biological system, taking into account genetic regulation, altered kinetic activity of enzymes, and changes in metabolic reactions. Thus, compared to genomics and proteomics, metabolomics reflects changes in phenotype and therefore function. In this review, we highlight some of the key advancements that have been made in applying metabolomics to understand the aggressive phenotype of glioblastoma. Collectively, these studies have provided a previously unrecognized window into the underlying biology of these tumors. Current and future efforts are designed to determine how this technology may be applied to improve diagnosis and predict the aggressiveness of glioblastoma, and more importantly, identify novel, therapeutic strategies designed to improve clinical outcomes.
Collapse
|