Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Cell Sci ; 137(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38305737

ABSTRACT

Tight control over transcription factor activity is necessary for a sensible balance between cellular proliferation and differentiation in the embryo and during tissue homeostasis by adult stem cells, but mechanistic details have remained incomplete. The homeodomain transcription factor MEIS2 is an important regulator of neurogenesis in the ventricular-subventricular zone (V-SVZ) adult stem cell niche in mice. We here identify MEIS2 as direct target of the intracellular protease calpain-2 (composed of the catalytic subunit CAPN2 and the regulatory subunit CAPNS1). Phosphorylation at conserved serine and/or threonine residues, or dimerization with PBX1, reduced the sensitivity of MEIS2 towards cleavage by calpain-2. In the adult V-SVZ, calpain-2 activity is high in stem and progenitor cells, but rapidly declines during neuronal differentiation, which is accompanied by increased stability of MEIS2 full-length protein. In accordance with this, blocking calpain-2 activity in stem and progenitor cells, or overexpression of a cleavage-insensitive form of MEIS2, increased the production of neurons, whereas overexpression of a catalytically active CAPN2 reduced it. Collectively, our results support a key role for calpain-2 in controlling the output of adult V-SVZ neural stem and progenitor cells through cleavage of the neuronal fate determinant MEIS2.


Subject(s)
Neural Stem Cells , Transcription Factors , Animals , Mice , Calpain/genetics , Calpain/metabolism , Cell Differentiation , Cell Proliferation , Endopeptidases/metabolism , Lateral Ventricles/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/metabolism , Peptide Hydrolases/metabolism , Transcription Factors/metabolism
2.
Acta Neuropathol ; 140(6): 919-949, 2020 12.
Article in English | MEDLINE | ID: mdl-33009951

ABSTRACT

Patient-based cancer models are essential tools for studying tumor biology and for the assessment of drug responses in a translational context. We report the establishment a large cohort of unique organoids and patient-derived orthotopic xenografts (PDOX) of various glioma subtypes, including gliomas with mutations in IDH1, and paired longitudinal PDOX from primary and recurrent tumors of the same patient. We show that glioma PDOXs enable long-term propagation of patient tumors and represent clinically relevant patient avatars that retain histopathological, genetic, epigenetic, and transcriptomic features of parental tumors. We find no evidence of mouse-specific clonal evolution in glioma PDOXs. Our cohort captures individual molecular genotypes for precision medicine including mutations in IDH1, ATRX, TP53, MDM2/4, amplification of EGFR, PDGFRA, MET, CDK4/6, MDM2/4, and deletion of CDKN2A/B, PTCH, and PTEN. Matched longitudinal PDOX recapitulate the limited genetic evolution of gliomas observed in patients following treatment. At the histological level, we observe increased vascularization in the rat host as compared to mice. PDOX-derived standardized glioma organoids are amenable to high-throughput drug screens that can be validated in mice. We show clinically relevant responses to temozolomide (TMZ) and to targeted treatments, such as EGFR and CDK4/6 inhibitors in (epi)genetically defined subgroups, according to MGMT promoter and EGFR/CDK status, respectively. Dianhydrogalactitol (VAL-083), a promising bifunctional alkylating agent in the current clinical trial, displayed high therapeutic efficacy, and was able to overcome TMZ resistance in glioblastoma. Our work underscores the clinical relevance of glioma organoids and PDOX models for translational research and personalized treatment studies and represents a unique publicly available resource for precision oncology.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Heterografts/immunology , Organoids/pathology , Temozolomide/therapeutic use , Animals , Brain Neoplasms/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioma/genetics , Heterografts/drug effects , Humans , Mice , Neoplasm Recurrence, Local/genetics , Organoids/immunology , Precision Medicine/methods , Rats
3.
Development ; 143(13): 2281-91, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27226325

ABSTRACT

TALE-homeodomain proteins function as components of heteromeric complexes that contain one member each of the PBC and MEIS/PREP subclasses. We recently showed that MEIS2 cooperates with the neurogenic transcription factor PAX6 in the control of adult subventricular zone (SVZ) neurogenesis in rodents. Expression of the PBC protein PBX1 in the SVZ has been reported, but its functional role(s) has not been investigated. Using a genetic loss-of-function mouse model, we now show that Pbx1 is an early regulator of SVZ neurogenesis. Targeted deletion of Pbx1 by retroviral transduction of Cre recombinase into Pbx2-deficient SVZ stem and progenitor cells carrying floxed alleles of Pbx1 significantly reduced the production of neurons and increased the generation of oligodendrocytes. Loss of Pbx1 expression in neuronally committed neuroblasts in the rostral migratory stream in a Pbx2 null background, by contrast, severely compromised cell survival. By chromatin immunoprecipitation from endogenous tissues or isolated cells, we further detected PBX1 binding to known regulatory regions of the neuron-specific genes Dcx and Th days or even weeks before the respective genes are expressed during the normal program of SVZ neurogenesis, suggesting that PBX1 might act as a priming factor to mark these genes for subsequent activation. Collectively, our results establish that PBX1 regulates adult neural cell fate determination in a manner beyond that of its heterodimerization partner MEIS2.


Subject(s)
Aging/metabolism , Homeodomain Proteins/metabolism , Lateral Ventricles/metabolism , Neurogenesis , Transcription Factors/metabolism , Animals , Base Sequence , Cell Differentiation , Cell Lineage , Cell Movement , Cell Survival , Cells, Cultured , Doublecortin Domain Proteins , Doublecortin Protein , Enhancer Elements, Genetic/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Gene Targeting , Homeodomain Proteins/genetics , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurogenesis/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Olfactory Bulb/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Pre-B-Cell Leukemia Transcription Factor 1 , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/genetics , Tyrosine 3-Monooxygenase/metabolism
4.
Development ; 141(1): 28-38, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24284204

ABSTRACT

Meis homeodomain transcription factors control cell proliferation, cell fate specification and differentiation in development and disease. Previous studies have largely focused on Meis contribution to the development of non-neuronal tissues. By contrast, Meis function in the brain is not well understood. Here, we provide evidence for a dual role of the Meis family protein Meis2 in adult olfactory bulb (OB) neurogenesis. Meis2 is strongly expressed in neuroblasts of the subventricular zone (SVZ) and rostral migratory stream (RMS) and in some of the OB interneurons that are continuously replaced during adult life. Targeted manipulations with retroviral vectors expressing function-blocking forms or with small interfering RNAs demonstrated that Meis activity is cell-autonomously required for the acquisition of a general neuronal fate by SVZ-derived progenitors in vivo and in vitro. Additionally, Meis2 activity in the RMS is important for the generation of dopaminergic periglomerular neurons in the OB. Chromatin immunoprecipitation identified doublecortin and tyrosine hydroxylase as direct Meis targets in newly generated neurons and the OB, respectively. Furthermore, biochemical analyses revealed a previously unrecognized complex of Meis2 with Pax6 and Dlx2, two transcription factors involved in OB neurogenesis. The full pro-neurogenic activity of Pax6 in SVZ derived neural stem and progenitor cells requires the presence of Meis. Collectively, these results show that Meis2 cooperates with Pax6 in generic neurogenesis and dopaminergic fate specification in the adult SVZ-OB system.


Subject(s)
Dopaminergic Neurons/cytology , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Neurogenesis/physiology , Olfactory Bulb/embryology , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Base Sequence , Cell Proliferation , Dopaminergic Neurons/metabolism , Doublecortin Domain Proteins , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neuropeptides/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/growth & development , PAX6 Transcription Factor , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism
5.
Heliyon ; 10(5): e27515, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38562501

ABSTRACT

We provide in this paper a comprehensive comparison of various transfer learning strategies and deep learning architectures for computer-aided classification of adult-type diffuse gliomas. We evaluate the generalizability of out-of-domain ImageNet representations for a target domain of histopathological images, and study the impact of in-domain adaptation using self-supervised and multi-task learning approaches for pretraining the models using the medium-to-large scale datasets of histopathological images. A semi-supervised learning approach is furthermore proposed, where the fine-tuned models are utilized to predict the labels of unannotated regions of the whole slide images (WSI). The models are subsequently retrained using the ground-truth labels and weak labels determined in the previous step, providing superior performance in comparison to standard in-domain transfer learning with balanced accuracy of 96.91% and F1-score 97.07%, and minimizing the pathologist's efforts for annotation. Finally, we provide a visualization tool working at WSI level which generates heatmaps that highlight tumor areas; thus, providing insights to pathologists concerning the most informative parts of the WSI.

6.
Cancer Res ; 84(5): 741-756, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38117484

ABSTRACT

Tumor adaptation or selection is thought to underlie therapy resistance in glioma. To investigate longitudinal epigenetic evolution of gliomas in response to therapeutic pressure, we performed an epigenomic analysis of 132 matched initial and recurrent tumors from patients with IDH-wildtype (IDHwt) and IDH-mutant (IDHmut) glioma. IDHwt gliomas showed a stable epigenome over time with relatively low levels of global methylation. The epigenome of IDHmut gliomas showed initial high levels of genome-wide DNA methylation that was progressively reduced to levels similar to those of IDHwt tumors. Integration of epigenomics, gene expression, and functional genomics identified HOXD13 as a master regulator of IDHmut astrocytoma evolution. Furthermore, relapse of IDHmut tumors was accompanied by histologic progression that was associated with survival, as validated in an independent cohort. Finally, the initial cell composition of the tumor microenvironment varied between IDHwt and IDHmut tumors and changed differentially following treatment, suggesting increased neoangiogenesis and T-cell infiltration upon treatment of IDHmut gliomas. This study provides one of the largest cohorts of paired longitudinal glioma samples with epigenomic, transcriptomic, and genomic profiling and suggests that treatment of IDHmut glioma is associated with epigenomic evolution toward an IDHwt-like phenotype. SIGNIFICANCE: Standard treatments are related to loss of DNA methylation in IDHmut glioma, resulting in epigenetic activation of genes associated with tumor progression and alterations in the microenvironment that resemble treatment-naïve IDHwt glioma.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Humans , Brain Neoplasms/pathology , Epigenesis, Genetic , Epigenomics , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Neoplasm Recurrence, Local/genetics , Tumor Microenvironment
7.
Sci Rep ; 11(1): 21013, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34697387

ABSTRACT

PAX6 is a highly conserved transcription factor and key regulator of several neurogenic processes, including the continuous generation of dopaminergic/GABAergic interneurons in the adult ventricular-subventricular (V-SVZ) neurogenic system in mice. Here we report that PAX6 cooperates with the TALE-homeodomain transcription factor PBX1 in this context. Chromatin-immunoprecipitation showed that PBX1 and PAX6 co-occupy shared genomic binding sites in adult V-SVZ stem- and progenitor cell cultures and mouse embryonic stem cells, while depletion of Pbx1 revealed that association of PAX6 with these sites requires the presence of PBX1. Expression profiling together with viral overexpression or knockdown of Pax6 or Pbx1 identified novel PBX1-PAX6 co-regulated genes, including several transcription factors. Computational modeling of genome wide expression identified novel cross-regulatory networks among these very transcription factors. Taken together, the results presented here highlight the intimate link that exists between PAX6 and TALE-HD family proteins and contribute novel insights into how the orchestrated activity of transcription factors shapes adult V-SVZ neurogenesis.


Subject(s)
Epistasis, Genetic , Gene Expression Regulation , Neural Stem Cells/metabolism , PAX6 Transcription Factor/genetics , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Computational Biology/methods , Female , Gene Expression Profiling , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Neural Stem Cells/cytology , Neurogenesis/genetics , PAX6 Transcription Factor/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Protein Binding
8.
Neurooncol Adv ; 3(1): vdab057, 2021.
Article in English | MEDLINE | ID: mdl-34250481

ABSTRACT

BACKGROUND: Mutations in isocitrate dehydrogenase 1 or 2 (IDH1/2) define glioma subtypes and are considered primary events in gliomagenesis, impacting tumor epigenetics and metabolism. IDH enzyme activity is crucial for the generation of reducing potential in normal cells, yet the impact of the mutation on the cellular antioxidant system in glioma is not understood. The aim of this study was to determine how glutathione (GSH), the main antioxidant in the brain, is maintained in IDH1-mutant gliomas, despite an altered NADPH/NADP balance. METHODS: Proteomics, metabolomics, metabolic tracer studies, genetic silencing, and drug targeting approaches in vitro and in vivo were applied. Analyses were done in clinical specimen of different glioma subtypes, in glioma patient-derived cell lines carrying the endogenous IDH1 mutation and corresponding orthotopic xenografts in mice. RESULTS: We find that cystathionine-γ-lyase (CSE), the enzyme responsible for cysteine production upstream of GSH biosynthesis, is specifically upregulated in IDH1-mutant astrocytomas. CSE inhibition sensitized these cells to cysteine depletion, an effect not observed in IDH1 wild-type gliomas. This correlated with an increase in reactive oxygen species and reduced GSH synthesis. Propargylglycine (PAG), a brain-penetrant drug specifically targeting CSE, led to delayed tumor growth in mice. CONCLUSIONS: We show that IDH1-mutant astrocytic gliomas critically rely on NADPH-independent de novo GSH synthesis via CSE to maintain the antioxidant defense, which highlights a novel metabolic vulnerability that may be therapeutically exploited.

9.
STAR Protoc ; 2(2): 100534, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34027491

ABSTRACT

Tumor organoids and patient-derived orthotopic xenografts (PDOXs) are some of the most valuable pre-clinical tools in cancer research. In this protocol, we describe efficient derivation of organoids and PDOX models from glioma patient tumors. We provide detailed steps for organoid culture, intracranial implantation, and detection of tumors in the brain. We further present technical adjustments for standardized functional assays and drug testing. For complete details on the use and execution of this protocol, please refer to Golebiewska et al. (2020).


Subject(s)
Brain Neoplasms/pathology , Drug Screening Assays, Antitumor/methods , Glioma/pathology , Heterografts , Organoids , Animals , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Female , Heterografts/cytology , Heterografts/drug effects , Humans , Male , Mice , Organoids/cytology , Organoids/drug effects , Tumor Cells, Cultured/cytology
10.
Front Oncol ; 10: 604121, 2020.
Article in English | MEDLINE | ID: mdl-33364198

ABSTRACT

Malignant brain tumors remain uniformly fatal, even with the best-to-date treatment. For Glioblastoma (GBM), the most severe form of brain cancer in adults, the median overall survival is roughly over a year. New therapeutic options are urgently needed, yet recent clinical trials in the field have been largely disappointing. This is partially due to inappropriate preclinical model systems, which do not reflect the complexity of patient tumors. Furthermore, clinically relevant patient-derived models recapitulating the immune compartment are lacking, which represents a bottleneck for adequate immunotherapy testing. Emerging 3D organoid cultures offer innovative possibilities for cancer modeling. Here, we review available GBM organoid models amenable to a large variety of pre-clinical applications including functional bioassays such as proliferation and invasion, drug screening, and the generation of patient-derived orthotopic xenografts (PDOX) for validation of biological responses in vivo. We emphasize advantages and technical challenges in establishing immunocompetent ex vivo models based on co-cultures of GBM organoids and human immune cells. The latter can be isolated either from the tumor or from patient or donor blood as peripheral blood mononuclear cells (PBMCs). We also discuss the challenges to generate GBM PDOXs based on humanized mouse models to validate efficacy of immunotherapies in vivo. A detailed characterization of such models at the cellular and molecular level is needed to understand the potential and limitations for various immune activating strategies. Increasing the availability of immunocompetent GBM models will improve research on emerging immune therapeutic approaches against aggressive brain cancer.

11.
Nat Commun ; 11(1): 6366, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33311477

ABSTRACT

The infiltrative nature of Glioblastoma (GBM), the most aggressive primary brain tumor, critically prevents complete surgical resection and masks tumor cells behind the blood brain barrier reducing the efficacy of systemic treatment. Here, we use a genome-wide interference screen to determine invasion-essential genes and identify the AN1/A20 zinc finger domain containing protein 3 (ZFAND3) as a crucial driver of GBM invasion. Using patient-derived cellular models, we show that loss of ZFAND3 hampers the invasive capacity of GBM, whereas ZFAND3 overexpression increases motility in cells that were initially not invasive. At the mechanistic level, we find that ZFAND3 activity requires nuclear localization and integral zinc-finger domains. Our findings indicate that ZFAND3 acts within a nuclear protein complex to activate gene transcription and regulates the promoter of invasion-related genes such as COL6A2, FN1, and NRCAM. Further investigation in ZFAND3 function in GBM and other invasive cancers is warranted.


Subject(s)
Brain Neoplasms/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glioblastoma/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Brain Neoplasms/pathology , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Movement , Collagen Type VI/genetics , Fibronectins/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Mice , Neoplasm Invasiveness/genetics , Protein Domains , Transcriptome
12.
Neurooncol Adv ; 1(1): vdz024, 2019.
Article in English | MEDLINE | ID: mdl-32642659

ABSTRACT

BACKGROUND: Targeted approaches for inhibiting epidermal growth factor receptor (EGFR) and other receptor tyrosine kinases (RTKs) in glioblastoma (GBM) have led to therapeutic resistance and little clinical benefit, raising the need for the development of alternative strategies. Endogenous LRIG1 (Leucine-rich Repeats and ImmunoGlobulin-like domains protein 1) is an RTK inhibitory protein required for stem cell maintenance, and we previously demonstrated the soluble ectodomain of LRIG1 (sLRIG1) to potently inhibit GBM growth in vitro and in vivo. METHODS: Here, we generated a recombinant protein of the ectodomain of LRIG1 (sLRIG1) and determined its activity in various cellular GBM models including patient-derived stem-like cells and patient organoids. We used proliferation, adhesion, and invasion assays, and performed gene and protein expression studies. Proximity ligation assay and NanoBiT complementation technology were applied to assess protein-protein interactions. RESULTS: We show that recombinant sLRIG1 downregulates EGFRvIII but not EGFR, and reduces proliferation in GBM cells, irrespective of their EGFR expression status. We find that sLRIG1 targets and downregulates a wide range of RTKs, including AXL, and alters GBM cell adhesion. Mechanistically, we demonstrate that LRIG1 interferes with AXL but not with EGFR dimerization. CONCLUSIONS: These results identify AXL as a novel sLRIG1 target and show that LRIG1-mediated RTK downregulation depends on direct protein interaction. The pan-RTK inhibitory activity of sLRIG1 warrants further investigation for new GBM treatment approaches.

13.
Stem Cell Reports ; 10(4): 1184-1192, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29641989

ABSTRACT

Adult neurogenesis is regulated by stem cell niche-derived extrinsic factors and cell-intrinsic regulators, yet the mechanisms by which niche signals impinge on the activity of intrinsic neurogenic transcription factors remain poorly defined. Here, we report that MEIS2, an essential regulator of adult SVZ neurogenesis, is subject to posttranslational regulation in the SVZ olfactory bulb neurogenic system. Nuclear accumulation of MEIS2 in adult SVZ-derived progenitor cells follows downregulation of EGFR signaling and is modulated by methylation of MEIS2 on a conserved arginine, which lies in close proximity to nested binding sites for the nuclear export receptor CRM1 and the MEIS dimerization partner PBX1. Methylation impairs interaction with CRM1 without affecting PBX1 dimerization and thereby allows MEIS2 nuclear accumulation, a prerequisite for neuronal differentiation. Our results describe a form of posttranscriptional modulation of adult SVZ neurogenesis whereby an extrinsic signal fine-tunes neurogenesis through posttranslational modification of a transcriptional regulator of cell fate.


Subject(s)
Arginine/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Homeodomain Proteins/metabolism , Lateral Ventricles/cytology , Neural Stem Cells/cytology , Neurons/cytology , Amino Acid Sequence , Animals , Binding, Competitive , ErbB Receptors/metabolism , Homeodomain Proteins/chemistry , Karyopherins/metabolism , Methylation , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurons/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Protein Binding , Protein Stability , Protein Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Exportin 1 Protein
14.
J Cell Biol ; 216(9): 2715-2729, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28739678

ABSTRACT

Pre-B-cell leukemia homeobox (PBX) and myeloid ecotropic viral integration site (MEIS) proteins control cell fate decisions in many physiological and pathophysiological contexts, but how these proteins function mechanistically remains poorly defined. Focusing on the first hours of neuronal differentiation of adult subventricular zone-derived stem/progenitor cells, we describe a sequence of events by which PBX-MEIS facilitates chromatin accessibility of transcriptionally inactive genes: In undifferentiated cells, PBX1 is bound to the H1-compacted promoter/proximal enhancer of the neuron-specific gene doublecortin (Dcx) Once differentiation is induced, MEIS associates with chromatin-bound PBX1, recruits PARP1/ARTD1, and initiates PARP1-mediated eviction of H1 from the chromatin fiber. These results for the first time link MEIS proteins to PARP-regulated chromatin dynamics and provide a mechanistic basis to explain the profound cellular changes elicited by these proteins.


Subject(s)
Adult Stem Cells/enzymology , Cell Lineage , Chromatin/enzymology , Histones/metabolism , Homeodomain Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/enzymology , Neurogenesis , Neuropeptides/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Cell Line, Tumor , Chromatin/genetics , Chromatin Assembly and Disassembly , Doublecortin Domain Proteins , Doublecortin Protein , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Neuropeptides/genetics , Phenotype , Poly (ADP-Ribose) Polymerase-1/genetics , Pre-B-Cell Leukemia Transcription Factor 1 , Promoter Regions, Genetic , Protein Binding , RNA Interference , Spheroids, Cellular , Stem Cell Niche , Time Factors , Transcription Factors/genetics , Transcription, Genetic , Transfection
15.
EMBO Mol Med ; 9(12): 1681-1695, 2017 12.
Article in English | MEDLINE | ID: mdl-29054837

ABSTRACT

Heterozygous mutations in NADP-dependent isocitrate dehydrogenases (IDH) define the large majority of diffuse gliomas and are associated with hypermethylation of DNA and chromatin. The metabolic dysregulations imposed by these mutations, whether dependent or not on the oncometabolite D-2-hydroxyglutarate (D2HG), are less well understood. Here, we applied mass spectrometry imaging on intracranial patient-derived xenografts of IDH-mutant versus IDH wild-type glioma to profile the distribution of metabolites at high anatomical resolution in situ This approach was complemented by in vivo tracing of labeled nutrients followed by liquid chromatography-mass spectrometry (LC-MS) analysis. Selected metabolites were verified on clinical specimen. Our data identify remarkable differences in the phospholipid composition of gliomas harboring the IDH1 mutation. Moreover, we show that these tumors are characterized by reduced glucose turnover and a lower energy potential, correlating with their reduced aggressivity. Despite these differences, our data also show that D2HG overproduction does not result in a global aberration of the central carbon metabolism, indicating strong adaptive mechanisms at hand. Intriguingly, D2HG shows no quantitatively important glucose-derived label in IDH-mutant tumors, which suggests that the synthesis of this oncometabolite may rely on alternative carbon sources. Despite a reduction in NADPH, glutathione levels are maintained. We found that genes coding for key enzymes in de novo glutathione synthesis are highly expressed in IDH-mutant gliomas and the expression of cystathionine-ß-synthase (CBS) correlates with patient survival in the oligodendroglial subtype. This study provides a detailed and clinically relevant insight into the in vivo metabolism of IDH1-mutant gliomas and points to novel metabolic vulnerabilities in these tumors.


Subject(s)
Brain Neoplasms/pathology , Energy Metabolism , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Oxidative Stress , Phospholipids/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Carbon Isotopes/chemistry , Female , Glioma/genetics , Glioma/mortality , Humans , Isotope Labeling , Lipid Metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Nitrogen Isotopes/chemistry , Phospholipids/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Survival Rate , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL