Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell ; 159(7): 1603-14, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25525878

RESUMEN

Glioblastomas and brain metastases are highly proliferative brain tumors with short survival times. Previously, using (13)C-NMR analysis of brain tumors resected from patients during infusion of (13)C-glucose, we demonstrated that there is robust oxidation of glucose in the citric acid cycle, yet glucose contributes less than 50% of the carbons to the acetyl-CoA pool. Here, we show that primary and metastatic mouse orthotopic brain tumors have the capacity to oxidize [1,2-(13)C]acetate and can do so while simultaneously oxidizing [1,6-(13)C]glucose. The tumors do not oxidize [U-(13)C]glutamine. In vivo oxidation of [1,2-(13)C]acetate was validated in brain tumor patients and was correlated with expression of acetyl-CoA synthetase enzyme 2, ACSS2. Together, the data demonstrate a strikingly common metabolic phenotype in diverse brain tumors that includes the ability to oxidize acetate in the citric acid cycle. This adaptation may be important for meeting the high biosynthetic and bioenergetic demands of malignant growth.


Asunto(s)
Acetato CoA Ligasa/metabolismo , Acetatos/metabolismo , Neoplasias Encefálicas/metabolismo , Ciclo del Ácido Cítrico , Glioblastoma/metabolismo , Acetato CoA Ligasa/genética , Animales , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Modelos Animales de Enfermedad , Glioblastoma/patología , Ácido Glutámico/metabolismo , Humanos , Ratones , Metástasis de la Neoplasia/patología
2.
Glia ; 68(12): 2613-2630, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32573857

RESUMEN

Glioblastomas (GBMs) are incurable brain tumors with a high degree of cellular heterogeneity and genetic mutations. Transcription factors that normally regulate neural progenitors and glial development are aberrantly coexpressed in GBM, conferring cancer stem-like properties to drive tumor progression and therapeutic resistance. However, the functional role of individual transcription factors in GBMs in vivo remains elusive. Here, we demonstrate that the basic-helix-loop-helix transcription factor ASCL1 regulates transcriptional targets that are central to GBM development, including neural stem cell and glial transcription factors, oncogenic signaling molecules, chromatin modifying genes, and cell cycle and mitotic genes. We also show that the loss of ASCL1 significantly reduces the proliferation of GBMs induced in the brain of a genetically relevant glioma mouse model, resulting in extended survival times. RNA-seq analysis of mouse GBM tumors reveal that the loss of ASCL1 is associated with downregulation of cell cycle genes, illustrating an important role for ASCL1 in controlling the proliferation of GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Genes cdc , Ratones , Factores de Transcripción/metabolismo
3.
Magn Reson Med ; 84(3): 1152-1160, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32003035

RESUMEN

PURPOSE: To generate a preclinical model of isocitrate dehydrogenase (IDH) mutant gliomas from glioma patients and design a MRS method to test the compatibility of 2-hydroxyglutarate (2HG) production between the preclinical model and patients. METHODS: Five patient-derived xenograft (PDX) mice were generated from two glioma patients with IDH1 R132H mutation. A PRESS sequence was tailored at 9.4 T, with computer simulation and phantom analyses, for improving 2HG detection in mice. 2HG and other metabolites in the PDX mice were measured using the optimized MRS at 9.4 T and compared with 3 T MRS measurements of the metabolites in the parental-tumor patients. Spectral fitting was performed with LCModel using in-house basis spectra. Metabolite levels were quantified with reference to water. RESULTS: The PRESS TE was optimized to be 96 ms, at which the 2HG 2.25 ppm signal was narrow and inverted, thereby leading to unequivocal separation of the 2HG resonance from adjacent signals from other metabolites. The optimized MRS provided precise detection of 2HG in mice compared to short-TE MRS at 9.4 T. The 2HG estimates in PDX mice were in excellent agreement with the 2HG measurements in the patients. CONCLUSION: The similarity of 2HG production between PDX models and parental-tumor patients indicates that PDX tumors retain the parental IDH metabolic fingerprint and can serve as a preclinical model for improving our understanding of the IDH-mutation associated metabolic reprogramming.


Asunto(s)
Neoplasias Encefálicas , Glioma , Animales , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Simulación por Computador , Glioma/diagnóstico por imagen , Glioma/genética , Glutaratos , Humanos , Isocitrato Deshidrogenasa/genética , Espectroscopía de Resonancia Magnética , Ratones , Trasplante de Neoplasias
4.
J Neurooncol ; 133(1): 183-192, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28421459

RESUMEN

According to the recently updated World Health Organization (WHO) classification (2016), grade II-III astrocytomas are divided into IDH-wildtype and IDH-mutant groups, the latter being significantly less aggressive in terms of both progression-free and total survival. We identified a small cohort of WHO grade II-III astrocytomas that harbored the IDH1 R132H mutation, as confirmed by both immunohistochemistry and molecular sequence analysis, which nonetheless had unexpectedly rapid recurrence and subsequent progression to glioblastoma. Among these four cases, the mean time to recurrence as glioblastoma was only 16 months and the mean total survival among the three patients who have died during the follow-up was only 31 months. We hypothesized that these tumors had other, unfavorable genetic or epigenetic alterations that negated the favorable effect of the IDH mutation. We applied genome-wide profiling with a methylation array (Illumina Infinium Human Methylation 450k) to screen for genetic and epigenetic alterations in these tumors. As expected, the methylation profiles of all four tumors were found to match most closely with IDH-mutant astrocytomas. Compared with a control group of four indolent, age-similar WHO grade II-III astrocytomas, the tumors showed markedly increased levels of overall copy number changes, but no consistent specific genetic alterations were seen across all of the tumors. While most IDH-mutant WHO grade II-III astrocytomas are relatively indolent, a subset may rapidly recur and progress to glioblastoma. The precise underlying cause of the increased aggressiveness in these gliomas remains unknown, although it may be associated with increased genomic instability.


Asunto(s)
Astrocitoma/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Isocitrato Deshidrogenasa/genética , Mutación , Adulto , Astrocitoma/mortalidad , Astrocitoma/patología , Astrocitoma/fisiopatología , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Variaciones en el Número de Copia de ADN , Metilación de ADN , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Estudio de Asociación del Genoma Completo , Glioblastoma/mortalidad , Glioblastoma/patología , Glioblastoma/fisiopatología , Humanos , Inmunohistoquímica , Isocitrato Deshidrogenasa/metabolismo , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/mortalidad , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(12): 4542-7, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24616497

RESUMEN

Glioblastoma multiforme (GBM), which account for more than 50% of all gliomas, is among the deadliest of all human cancers. Given the dismal prognosis of GBM, it would be advantageous to identify early biomarkers of a response to therapy to avoid continuing ineffective treatments and to initiate other therapeutic strategies. The present in vivo longitudinal study in an orthotopic mouse model demonstrates quantitative assessment of early treatment response during short-term chemotherapy with temozolomide (TMZ) by amide proton transfer (APT) imaging. In a GBM line, only one course of TMZ (3 d exposure and 4 d rest) at a dose of 80 mg/kg resulted in substantial reduction in APT signal compared with untreated control animals, in which the APT signal continued to increase. Although there were no detectable differences in tumor volume, cell density, or apoptosis rate between groups, levels of Ki67 (index of cell proliferation) were substantially reduced in treated tumors. In another TMZ-resistant GBM line, the APT signal and levels of Ki67 increased despite the same course of TMZ treatment. As metabolite changes are known to occur early in the time course of chemotherapy and precede morphologic changes, these results suggest that the APT signal in glioma may be a useful functional biomarker of treatment response or degree of tumor progression. Thus, APT imaging may serve as a sensitive biomarker of early treatment response and could potentially replace invasive biopsies to provide a definitive diagnosis. This would have a major impact on the clinical management of patients with glioma.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Animales , Dacarbazina/uso terapéutico , Humanos , Ratones , Ratones SCID , Pronóstico , Temozolomida , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Magn Reson Med ; 73(6): 2094-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25046359

RESUMEN

PURPOSE: To evaluate the T2 relaxation time of lactate (Lac) in brain tumors and the correlation of the T2 and concentration with tumor grades. METHODS: Eight pairs of the subecho time sets of point-resolved spectroscopy were selected between 58 and 268 ms, with numerical and phantom analyses, for Lac T2 measurement. In vivo spectra were acquired from 24 subjects with gliomas (13 low grade and 11 high grade) and analyzed with LCModel using numerically-calculated basis spectra. The metabolite T2 relaxation time was obtained from monoexponential fitting of the multi-echo time (TE) signal estimates versus TE. The metabolite concentration was estimated from the zero-TE extrapolation of the T2 fits. RESULTS: The Lac T2 was estimated to be approximately 240 ms, without a significant difference between low and high grade tumors. The Lac concentration was estimated to be 4.1 ± 3.4 and 7.0 ± 4.7 mM for low and high grades respectively, but the difference was not significant. CONCLUSION: The Lac T2 was similar among gliomas regardless of their tumor grades. This suggests that the T2 value from this study may be applicable to obtain the T2 relaxation-free estimates of Lac in a subset of brain tumors.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Ácido Láctico/metabolismo , Espectroscopía de Protones por Resonancia Magnética/métodos , Adulto , Anciano , Artefactos , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Química Encefálica , Colina/metabolismo , Creatina/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fantasmas de Imagen
7.
NMR Biomed ; 28(1): 108-15, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25394324

RESUMEN

Glioblastoma (GBM), the most common primary brain tumor, is resistant to currently available treatments. The development of mouse models of human GBM has provided a tool for studying mechanisms involved in tumor initiation and growth as well as a platform for preclinical investigation of new drugs. In this study we used (1) H MR spectroscopy to study the neurochemical profile of a human orthotopic tumor (HOT) mouse model of human GBM. The goal of this study was to evaluate differences in metabolite concentrations in the GBM HOT mice when compared with normal mouse brain in order to determine if MRS could reliably differentiate tumor from normal brain. A TE =19 ms PRESS sequence at 9.4 T was used for measuring metabolite levels in 12 GBM mice and 8 healthy mice. Levels for 12 metabolites and for lipids/macromolecules at 0.9 ppm and at 1.3 ppm were reliably detected in all mouse spectra. The tumors had significantly lower concentrations of total creatine, GABA, glutamate, total N-acetylaspartate, aspartate, lipids/macromolecules at 0.9 ppm, and lipids/macromolecules at 1.3 ppm than did the brains of normal mice. The concentrations of glycine and lactate, however, were significantly higher in tumors than in normal brain.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Metaboloma , Espectroscopía de Protones por Resonancia Magnética/métodos , Animales , Modelos Animales de Enfermedad , Neuronas GABAérgicas/metabolismo , Glioblastoma/metabolismo , Glutamina/metabolismo , Humanos , Ratones
8.
Anal Biochem ; 481: 4-6, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25908561

RESUMEN

(13)C NMR (nuclear magnetic resonance) spectroscopy of extracts from patient tumor samples provides rich information about metabolism. However, in isocitrate dehydrogenase (IDH)-mutant gliomas, (13)C labeling is obscured in oncometabolite 2-hydroxyglutaric acid (2 HG) by glutamate and glutamine, prompting development of a simple method to resolve the metabolites. J-coupled multiplets in 2 HG were similar to glutamate and glutamine and could be clearly resolved at pH 6. A cryogenically cooled (13)C probe, but not J-resolved heteronuclear single quantum coherence spectroscopy, significantly improved detection of 2 HG. These methods enable the monitoring of (13)C-(13)C spin-spin couplings in 2 HG expressing IDH-mutant gliomas.


Asunto(s)
Glioma/genética , Glutaratos/análisis , Isocitrato Deshidrogenasa/genética , Espectroscopía de Resonancia Magnética/métodos , Isótopos de Carbono/análisis , Glioma/patología , Ácido Glutámico/análisis , Glutamina/análisis , Humanos , Mutación
9.
Proc Natl Acad Sci U S A ; 109(35): 14164-9, 2012 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-22891331

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive of the astrocytic malignancies and the most common intracranial tumor in adults. Although the epidermal growth factor receptor (EGFR) is overexpressed and/or mutated in at least 50% of GBM cases and is required for tumor maintenance in animal models, EGFR inhibitors have thus far failed to deliver significant responses in GBM patients. One inherent resistance mechanism in GBM is the coactivation of multiple receptor tyrosine kinases, which generates redundancy in activation of phosphoinositide-3'-kinase (PI3K) signaling. Here we demonstrate that the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor suppressor is frequently phosphorylated at a conserved tyrosine residue, Y240, in GBM clinical samples. Phosphorylation of Y240 is associated with shortened overall survival and resistance to EGFR inhibitor therapy in GBM patients and plays an active role in mediating resistance to EGFR inhibition in vitro. Y240 phosphorylation can be mediated by both fibroblast growth factor receptors and SRC family kinases (SFKs) but does not affect the ability of PTEN to antagonize PI3K signaling. These findings show that, in addition to genetic loss and mutation of PTEN, its modulation by tyrosine phosphorylation has important implications for the development and treatment of GBM.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Fosfohidrolasa PTEN/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Animales , Astrocitos/citología , Astrocitos/fisiología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/fisiología , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Ratones Mutantes , Ratones Desnudos , Fosfohidrolasa PTEN/genética , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Trasplante Heterólogo , Células Tumorales Cultivadas , Tirosina/metabolismo
10.
Magn Reson Med ; 72(2): 316-23, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24123337

RESUMEN

PURPOSE: To test whether citrate is elevated in adult patients with gliomas using (1)H magnetic resonance spectroscopy (MRS) at 3T in vivo. METHODS: Thirty-four adult patients were enrolled in the study, including six subjects with glioblastomas, eight subjects with astrocytomas (World Health Organization grade 3, n = 5; grade 2, n = 3), and 20 subjects with oligodendrogliomas (grade 3, n = 5; grade 2, n = 15). Five healthy volunteers were studied for baseline citrate data. Single-voxel localized spectra were collected with point-resolved spectroscopy (PRESS) echo times of 35 and 97 ms and were analyzed with LCModel software using numerically calculated basis spectra that included the effects of the PRESS radiofrequency and gradient pulses. RESULTS: Citrate was not measurable by MRS in healthy brain but was detected in tumor patients at both echo times. The citrate concentration was estimated to be as high as 1.8 mM with reference to water at 42 M, with Cramér-Rao lower bounds (CRLB) as low as 5%. The mean citrate level was 0.7 ± 0.4 mM (mean ± SD, n = 32) with a median CRLB of ∼12%. No correlation was identified between citrate concentration and tumor grade or histological type. CONCLUSION: Citrate was increased in the majority of gliomas in adult patients. The elevated citrate in our data indicates an altered metabolic state of tumor relative to healthy brain.


Asunto(s)
Biomarcadores de Tumor/análisis , Química Encefálica , Neoplasias Encefálicas/química , Ácido Cítrico/análisis , Glioma/química , Espectroscopía de Protones por Resonancia Magnética/métodos , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Imagen Molecular/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Adulto Joven
11.
J Neurooncol ; 117(1): 183-189, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24519516

RESUMEN

Infiltrating astrocytomas and oligoastrocytomas of low to anaplastic grade (WHO grades II and III), in spite of being associated with a wide range of clinical outcomes, can be difficult to subclassify and grade by the current histopathologic criteria. Unlike oligodendrogliomas and anaplastic oligodendrogliomas that can be identified by the 1p/19q codeletion and the more malignant glioblastomas (WHO grade IV astrocytomas) that can be diagnosed solely based on objective features on routine hematoxylin and eosin sections, no such objective criteria exist for the subclassification of grade II-III astrocytomas and oligoastrocytomas (A+OA II-III). In this study, we evaluated the prognostic and predictive value of the stem cell marker nestin in adult A+OA II-III (n = 50) using immunohistochemistry and computer-assisted analysis on tissue microarrays. In addition, the correlation between nestin mRNA level and total survival was analyzed in the NCI Rembrandt database. The results showed that high nestin expression is a strong adverse prognostic factor for total survival (p = 0.0004). The strength of the correlation was comparable to but independent of the isocitrate dehydrogenase 1/2 (IDH 1/2) mutation status. Histopathological grading and subclassification did not correlate significantly with outcome, although the interpretation of this finding is limited by the fact that grade III tumors were treated more aggressively than grade II tumors. These results suggest that nestin level and IDH 1/2 mutation status are strong prognostic features in A+OA II-III and possibly more helpful for treatment planning than routine histopathological variables such as oligodendroglial component (astrocytoma vs. oligoastrocytoma) and WHO grade (grade II vs. III).


Asunto(s)
Astrocitoma/diagnóstico , Neoplasias Encefálicas/diagnóstico , Glioma/diagnóstico , Nestina/metabolismo , Adulto , Anciano , Astrocitoma/genética , Astrocitoma/metabolismo , Astrocitoma/patología , Biomarcadores/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Femenino , Estudios de Seguimiento , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Isocitrato Deshidrogenasa/genética , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , ARN Mensajero/metabolismo , Adulto Joven
12.
Cancer Cell ; 41(10): 1788-1802.e10, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37816332

RESUMEN

Mitochondria (MT) participate in most metabolic activities of mammalian cells. A near-unidirectional mitochondrial transfer from T cells to cancer cells was recently observed to "metabolically empower" cancer cells while "depleting immune cells," providing new insights into tumor-T cell interaction and immune evasion. Here, we leverage single-cell RNA-seq technology and introduce MERCI, a statistical deconvolution method for tracing and quantifying mitochondrial trafficking between cancer and T cells. Through rigorous benchmarking and validation, MERCI accurately predicts the recipient cells and their relative mitochondrial compositions. Application of MERCI to human cancer samples identifies a reproducible MT transfer phenotype, with its signature genes involved in cytoskeleton remodeling, energy production, and TNF-α signaling pathways. Moreover, MT transfer is associated with increased cell cycle activity and poor clinical outcome across different cancer types. In summary, MERCI enables systematic investigation of an understudied aspect of tumor-T cell interactions that may lead to the development of therapeutic opportunities.


Asunto(s)
ADN Mitocondrial , Neoplasias , Animales , Humanos , ADN Mitocondrial/genética , Linfocitos T/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Mamíferos/genética , Mamíferos/metabolismo
13.
bioRxiv ; 2023 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-37873200

RESUMEN

Glioblastomas (GBMs) are highly aggressive, infiltrative, and heterogeneous brain tumors driven by complex driver mutations and glioma stem cells (GSCs). The neurodevelopmental transcription factors ASCL1 and OLIG2 are co-expressed in GBMs, but their role in regulating the heterogeneity and hierarchy of GBM tumor cells is unclear. Here, we show that oncogenic driver mutations lead to dysregulation of ASCL1 and OLIG2, which function redundantly to initiate brain tumor formation in a mouse model of GBM. Subsequently, the dynamic levels and reciprocal binding of ASCL1 and OLIG2 to each other and to downstream target genes then determine the cell types and degree of migration of tumor cells. Single-cell RNA sequencing (scRNA-seq) reveals that a high level of ASCL1 is key in defining GSCs by upregulating a collection of ribosomal protein, mitochondrial, neural stem cell (NSC), and cancer metastasis genes - all essential for sustaining the high proliferation, migration, and therapeutic resistance of GSCs.

14.
NMR Biomed ; 25(11): 1234-44, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22419606

RESUMEN

Glioblastomas and brain metastases demonstrate avid uptake of 2-[(18) F]fluoro-2-deoxyglucose by positron emission tomography and display perturbations of intracellular metabolite pools by (1) H MRS. These observations suggest that metabolic reprogramming contributes to brain tumor growth in vivo. The Warburg effect, excess metabolism of glucose to lactate in the presence of oxygen, is a hallmark of cancer cells in culture. 2-[(18) F]Fluoro-2-deoxyglucose-positive tumors are assumed to metabolize glucose in a similar manner, with high rates of lactate formation relative to mitochondrial glucose oxidation, but few studies have specifically examined the metabolic fates of glucose in vivo. In particular, the capacity of human brain cancers to oxidize glucose in the tricarboxylic acid cycle is unknown. Here, we studied the metabolism of human brain tumors in situ. [U-(13) C]Glucose (uniformly labeled glucose, i.e. d-glucose labeled with (13) C in all six carbons) was infused during surgical resection, and tumor samples were subsequently subjected to (13) C NMR spectroscopy. The analysis of tumor metabolites revealed lactate production, as expected. We also determined that pyruvate dehydrogenase, turnover of the tricarboxylic acid cycle, anaplerosis and de novo glutamine and glycine synthesis contributed significantly to the ultimate disposition of glucose carbon. Surprisingly, less than 50% of the acetyl-coenzyme A pool was derived from blood-borne glucose, suggesting that additional substrates contribute to tumor bioenergetics. This study illustrates a convenient approach that capitalizes on the high information content of (13) C NMR spectroscopy and enables the analysis of intermediary metabolism in diverse cancers growing in their native microenvironment.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glucosa/metabolismo , Acetilcoenzima A/metabolismo , Glucemia/metabolismo , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Isótopos de Carbono , Carcinoma de Pulmón de Células no Pequeñas/patología , Ciclo del Ácido Cítrico , Femenino , Glioblastoma/sangre , Glioblastoma/metabolismo , Glicina/biosíntesis , Glucólisis , Humanos , Oxidación-Reducción
15.
NMR Biomed ; 25(10): 1177-86, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22383401

RESUMEN

It has been hypothesized that increased flux through the pentose phosphate pathway (PPP) is required to support the metabolic demands of rapid malignant cell growth. Using orthotopic mouse models of human glioblastoma (GBM) and renal cell carcinoma metastatic to brain, we estimated the activity of the PPP relative to glycolysis by infusing [1,2-(13) C(2) ]glucose. The [3-(13) C]lactate/[2,3-(13) C(2) ]lactate ratio was similar for both the GBM and brain metastasis and their respective surrounding brains (GBM, 0.197 ± 0.011 and 0.195 ± 0.033, respectively (p = 1); metastasis: 0.126 and 0.119 ± 0.033, respectively). This suggests that the rate of glycolysis is significantly greater than the PPP flux in these tumors, and that the PPP flux into the lactate pool is similar in both tumors. Remarkably, (13) C-(13) C coupling was observed in molecules derived from Krebs cycle intermediates in both tumor types, denoting glucose oxidation. In the renal cell carcinoma, in contrast with GBM, (13) C multiplets of γ-aminobutyric acid (GABA) differed from its precursor glutamate, suggesting that GABA did not derive from a common glutamate precursor pool. In addition, the orthotopic renal tumor, the patient's primary renal mass and brain metastasis were all strongly immunopositive for the 67-kDa isoform of glutamate decarboxylase, as were 84% of tumors on a renal cell carcinoma tissue microarray of the same histology, suggesting that GABA synthesis is cell autonomous in at least a subset of renal cell carcinomas. Taken together, these data demonstrate that (13) C-labeled glucose can be used in orthotopic mouse models to study tumor metabolism in vivo and to ascertain new metabolic targets for cancer diagnosis and therapy.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Ciclo del Ácido Cítrico , Glucosa/metabolismo , Glucólisis , Vía de Pentosa Fosfato , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/secundario , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Modelos Animales de Enfermedad , Glioblastoma/diagnóstico por imagen , Glioblastoma/metabolismo , Glutamato Descarboxilasa/metabolismo , Ácido Glutámico/metabolismo , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Ácido Láctico/metabolismo , Imagen por Resonancia Magnética , Ratones , Tomografía de Emisión de Positrones , Ácido gamma-Aminobutírico/metabolismo
16.
Cancer Cell ; 1(3): 269-77, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12086863

RESUMEN

Ink4a/Arf inactivation and epidermal growth factor receptor (EGFR) activation are signature lesions in high-grade gliomas. How these mutations mediate the biological features of these tumors is poorly understood. Here, we demonstrate that combined loss of p16(INK4a) and p19(ARF), but not of p53, p16(INK4a), or p19(ARF), enables astrocyte dedifferentiation in response to EGFR activation. Moreover, transduction of Ink4a/Arf(-/-) neural stem cells (NSCs) or astrocytes with constitutively active EGFR induces a common high-grade glioma phenotype. These findings identify NSCs and astrocytes as equally permissive compartments for gliomagenesis and provide evidence that p16(INK4a) and p19(ARF) synergize to maintain terminal astrocyte differentiation. These data support the view that dysregulation of specific genetic pathways, rather than cell-of-origin, dictates the emergence and phenotype of high-grade gliomas.


Asunto(s)
Astrocitos/fisiología , Diferenciación Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Receptores ErbB/fisiología , Neuronas/citología , Células Madre/fisiología , Proteína p14ARF Supresora de Tumor/metabolismo , Animales , Astrocitos/citología , Western Blotting , Células Cultivadas/citología , Proteínas Fluorescentes Verdes , Homocigoto , Humanos , Técnicas para Inmunoenzimas , Bombas de Infusión Implantables , Proteínas Luminiscentes/metabolismo , Imagen por Resonancia Magnética , Ratones , Ratones SCID , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Retroviridae/genética , Células Madre/citología , Transformación Genética/fisiología , Proteína p53 Supresora de Tumor/metabolismo
17.
Differentiation ; 81(4): 233-42, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21419563

RESUMEN

In the central nervous system (CNS), neural stem cells (NSCs) differentiate into neurons, astrocytes, and oligodendrocytes--these cell lineages are considered unidirectional and irreversible under normal conditions. The introduction of a defined set of transcription factors has been shown to directly convert terminally differentiated cells into pluripotent stem cells, reinforcing the notion that preserving cellular identity is an active process. Indeed, recent studies highlight that tumor suppressor genes (TSGs) such as Ink4a/Arf and p53, control the barrier to efficient reprogramming, leaving open the question whether the same TSGs function to maintain the differentiated state. During malignancy or following brain injury, mature astrocytes have been reported to re-express neuronal genes and re-gain neurogenic potential to a certain degree, yet few studies have addressed the underlying mechanisms due to a limited number of cellular models or tools to probe this process. Here, we use a synthetic small-molecule (isoxazole) to demonstrate that highly malignant EGFRvIII-expressing Ink4a/Arf(-/-); Pten(-/-) astrocytes downregulated their astrocyte character, re-entered the cell cycle, and upregulated neuronal gene expression. As a collateral discovery, isoxazole small-molecules blocked tumor cell proliferation in vitro, a phenotype likely coupled to activation of neuronal gene expression. Similarly, histone deacetylase inhibitors induced neuronal gene expression and morphologic changes associated with the neuronal phenotype, suggesting the involvement of epigenetic-mediated gene activation. Our study assesses the contribution of specific genetic pathways underlying the de-differentiation potential of astrocytes and uncovers a novel pharmacological tool to explore astrocyte plasticity, which may bring insight to reprogramming and anti-tumor strategies.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/patología , Desdiferenciación Celular/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/patología , Isoxazoles/farmacología , Neurogénesis/genética , Tiofenos/farmacología , Animales , Desdiferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Reprogramación Celular/genética , Epigénesis Genética/efectos de los fármacos , Receptores ErbB/genética , Glioma/genética , Inhibidores de Histona Desacetilasas/farmacología , Isoxazoles/química , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neuronas/metabolismo , Proteínas Supresoras de Tumor/genética
18.
Neuro Oncol ; 24(4): 556-568, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34515312

RESUMEN

BACKGROUND: We postulate that meningiomas undergo distinct metabolic reprogramming in tumorigenesis and unraveling their metabolic phenotypes provide new therapeutic insights. Glutamine catabolism is key to the growth and proliferation of tumors. Here, we investigated the metabolomics of freshly resected meningiomas and glutamine metabolism in patient-derived meningioma cells. METHODS: 1H NMR spectroscopy of tumor tissues from meningioma patients was used to differentiate the metabolite profiles of grade-I and grade-II meningiomas. Glutamine metabolism was examined using 13C/15N glutamine tracer, in 5 patient-derived meningioma cells. RESULTS: Alanine, lactate, glutamate, glutamine, and glycine were predominantly elevated only in grade-II meningiomas by 74%, 76%, 35%, 75%, and 33%, respectively, with alanine and glutamine levels being statistically significant (P ≤ .02). 13C/15N glutamine tracer experiments revealed that both grade-I and -II meningiomas actively metabolize glutamine to generate various key carbon intermediates including alanine and proline that are necessary for the tumor growth. Also, it is shown that glutaminase (GLS1) inhibitor, CB-839 is highly effective in downregulating glutamine metabolism and decreasing proliferation in meningioma cells. CONCLUSION: Alanine and glutamine/glutamate are mainly elevated in grade-II meningiomas. Grade-I meningiomas possess relatively higher glutamine metabolism providing carbon/nitrogen for the biosynthesis of key nonessential amino acids. GLS1 inhibitor (CB-839) is very effective in downregulating glutamine metabolic pathways in grade-I meningiomas leading to decreased cellular proliferation.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Aminoácidos , Niño , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Espectroscopía de Resonancia Magnética/métodos , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo
19.
Cell Metab ; 34(1): 90-105.e7, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986341

RESUMEN

HER2+ breast cancer patients are presented with either synchronous (S-BM), latent (Lat), or metachronous (M-BM) brain metastases. However, the basis for disparate metastatic fitness among disseminated tumor cells of similar oncotype within a distal organ remains unknown. Here, employing brain metastatic models, we show that metabolic diversity and plasticity within brain-tropic cells determine metastatic fitness. Lactate secreted by aggressive metastatic cells or lactate supplementation to mice bearing Lat cells limits innate immunosurveillance and triggers overt metastasis. Attenuating lactate metabolism in S-BM impedes metastasis, while M-BM adapt and survive as residual disease. In contrast to S-BM, Lat and M-BM survive in equilibrium with innate immunosurveillance, oxidize glutamine, and maintain cellular redox homeostasis through the anionic amino acid transporter xCT. Moreover, xCT expression is significantly higher in matched M-BM brain metastatic samples compared to primary tumors from HER2+ breast cancer patients. Inhibiting xCT function attenuates residual disease and recurrence in these preclinical models.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de la Mama , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Ratones
20.
ACS Chem Neurosci ; 12(15): 2820-2828, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34291630

RESUMEN

Transient disruption of the blood-brain barrier (BBB) with focused ultrasound (FUS) is an emerging clinical method to facilitate targeted drug delivery to the brain. The focal noninvasive disruption of the BBB can be applied to promote the local delivery of hyperpolarized substrates. In this study, we investigated the effects of FUS on imaging brain metabolism using two hyperpolarized 13C-labeled substrates in rodents: [1-13C]pyruvate and [1-13C]glycerate. The BBB is a rate-limiting factor for pyruvate delivery to the brain, and glycerate minimally passes through the BBB. First, cerebral imaging with hyperpolarized [1-13C]pyruvate resulted in an increase in total 13C signals (p = 0.05) after disrupting the BBB with FUS. Significantly higher levels of both [1-13C]lactate (lactate/total 13C signals, p = 0.01) and [13C]bicarbonate (p = 0.008) were detected in the FUS-applied brain region as compared to the contralateral FUS-unaffected normal-appearing brain region. The application of FUS without opening the BBB in a separate group of rodents resulted in comparable lactate and bicarbonate productions between the FUS-applied and the contralateral brain regions. Second, 13C imaging with hyperpolarized [1-13C]glycerate after opening the BBB showed increased [1-13C]glycerate delivery to the FUS-applied region (p = 0.04) relative to the contralateral side, and [1-13C]lactate production was consistently detected from the FUS-applied region. Our findings suggest that FUS accelerates the delivery of hyperpolarized molecules across the BBB and provides enhanced sensitivity to detect metabolic products in the brain; therefore, hyperpolarized 13C imaging with FUS may provide new opportunities to study cerebral metabolic pathways as well as various neurological pathologies.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Animales , Transporte Biológico , Encéfalo/diagnóstico por imagen , Sistemas de Liberación de Medicamentos , Imagen por Resonancia Magnética , Ácido Pirúvico , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA