Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Clin Endocrinol (Oxf) ; 93(5): 528-538, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32686200

RESUMEN

The citric acid cycle, also known as the Krebs cycle, plays an integral role in cellular metabolism and aerobic respiration. Mutations in genes encoding the citric acid cycle enzymes succinate dehydrogenase, fumarate hydratase and malate dehydrogenase all predispose to hereditary tumour syndromes. The succinate dehydrogenase enzyme complex (SDH) couples the oxidation of succinate to fumarate in the citric acid cycle and the reduction of ubiquinone to ubiquinol in the electron transport chain. A loss of function in the succinate dehydrogenase (SDH) enzyme complex is most commonly caused by an inherited mutation in one of the four SDHx genes (SDHA, SDHB, SDHC and SDHD). This mechanism was first implicated in familial phaeochromocytoma and paraganglioma. However, over the past two decades the spectrum of tumours associated with SDH deficiency has been extended to include gastrointestinal stromal tumours (GIST), renal cell carcinoma (RCC) and pituitary adenomas. The aim of this review is to describe the extended tumour spectrum associated with SDHx gene mutations and to consider how functional tests may help to establish the role of SDHx mutations in new or unexpected tumour phenotypes.


Asunto(s)
Paraganglioma , Feocromocitoma , Mutación de Línea Germinal/genética , Humanos , Mutación , Paraganglioma/genética , Feocromocitoma/genética , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo
2.
EMBO J ; 33(12): 1365-82, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24837709

RESUMEN

Tumour cells sustain their high proliferation rate through metabolic reprogramming, whereby cellular metabolism shifts from oxidative phosphorylation to aerobic glycolysis, even under normal oxygen levels. Hypoxia-inducible factor 1A (HIF1A) is a major regulator of this process, but its activation under normoxic conditions, termed pseudohypoxia, is not well documented. Here, using an integrative approach combining the first genome-wide mapping of chromatin binding for an endocytic adaptor, ARRB1, both in vitro and in vivo with gene expression profiling, we demonstrate that nuclear ARRB1 contributes to this metabolic shift in prostate cancer cells via regulation of HIF1A transcriptional activity under normoxic conditions through regulation of succinate dehydrogenase A (SDHA) and fumarate hydratase (FH) expression. ARRB1-induced pseudohypoxia may facilitate adaptation of cancer cells to growth in the harsh conditions that are frequently encountered within solid tumours. Our study is the first example of an endocytic adaptor protein regulating metabolic pathways. It implicates ARRB1 as a potential tumour promoter in prostate cancer and highlights the importance of metabolic alterations in prostate cancer.


Asunto(s)
Arrestinas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Redes y Vías Metabólicas/fisiología , Modelos Biológicos , Neoplasias de la Próstata/fisiopatología , Inmunoprecipitación de Cromatina , Técnica del Anticuerpo Fluorescente , Fumarato Hidratasa/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Inmunohistoquímica , Espectroscopía de Resonancia Magnética , Masculino , Metabolómica , Neoplasias de la Próstata/metabolismo , Interferencia de ARN , Succinato Deshidrogenasa/metabolismo , Análisis de Matrices Tisulares , beta-Arrestina 1 , beta-Arrestinas
3.
EMBO J ; 30(13): 2719-33, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21602788

RESUMEN

The androgen receptor (AR) is a key regulator of prostate growth and the principal drug target for the treatment of prostate cancer. Previous studies have mapped AR targets and identified some candidates which may contribute to cancer progression, but did not characterize AR biology in an integrated manner. In this study, we took an interdisciplinary approach, integrating detailed genomic studies with metabolomic profiling and identify an anabolic transcriptional network involving AR as the core regulator. Restricting flux through anabolic pathways is an attractive approach to deprive tumours of the building blocks needed to sustain tumour growth. Therefore, we searched for targets of the AR that may contribute to these anabolic processes and could be amenable to therapeutic intervention by virtue of differential expression in prostate tumours. This highlighted calcium/calmodulin-dependent protein kinase kinase 2, which we show is overexpressed in prostate cancer and regulates cancer cell growth via its unexpected role as a hormone-dependent modulator of anabolic metabolism. In conclusion, it is possible to progress from transcriptional studies to a promising therapeutic target by taking an unbiased interdisciplinary approach.


Asunto(s)
Carcinoma/genética , Carcinoma/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/fisiología , Animales , Secuencia de Bases , Sitios de Unión/genética , Vías Biosintéticas/genética , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular , Análisis por Conglomerados , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Metabolismo/genética , Metabolismo/fisiología , Ratones , Modelos Biológicos , Neoplasias de la Próstata/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Elementos de Respuesta/genética , Trasplante Heterólogo
4.
Bioinformatics ; 30(15): 2155-61, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24711654

RESUMEN

MOTIVATION: In metabolomics, the goal is to identify and measure the concentrations of different metabolites (small molecules) in a cell or a biological system. The metabolites form an important layer in the complex metabolic network, and the interactions between different metabolites are often of interest. It is crucial to perform proper normalization of metabolomics data, but current methods may not be applicable when estimating interactions in the form of correlations between metabolites. We propose a normalization approach based on a mixed model, with simultaneous estimation of a correlation matrix. We also investigate how the common use of a calibration standard in nuclear magnetic resonance (NMR) experiments affects the estimation of correlations. RESULTS: We show with both real and simulated data that our proposed normalization method is robust and has good performance when discovering true correlations between metabolites. The standardization of NMR data is shown in simulation studies to affect our ability to discover true correlations to a small extent. However, comparing standardized and non-standardized real data does not result in any large differences in correlation estimates. AVAILABILITY AND IMPLEMENTATION: Source code is freely available at https://sourceforge.net/projects/metabnorm/ CONTACT: alexandra.jauhiainen@ki.se SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Metabolómica/métodos , Interpretación Estadística de Datos , Humanos , Espectroscopía de Resonancia Magnética , Redes y Vías Metabólicas , Metabolómica/normas , Estándares de Referencia
5.
MAGMA ; 28(2): 161-71, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25108704

RESUMEN

OBJECT: Metabolomic studies on cultured cells involve assays of cell extracts and culture medium, both of which are often performed by (1)H NMR. Cell culture is nowadays performed in plastic dishes or flasks, and the extraction of metabolites from the cells is typically performed with perchloric acid, methanol-chloroform, or acetonitrile, ideally while the cells are still adherent to the culture dish. We conducted this investigation to identify contaminants from cell culture plasticware in metabolomic studies. MATERIALS AND METHODS: Human diploid fibroblasts (IMR90) (n = 6), HeLa cells (n = 6), and transformed astrocytes with HIF-1 knockout (Astro-KO) (n = 6) were cultured. Cells were seeded in 100 mm Petri dishes with 10 ml complete growth medium (Dulbecco's minimum essential medium) containing 10 % foetal bovine serum (FBS). Cell cultures were incubated at 37 °C in 5 % CO2 for approximately 3 days. Metabolites were extracted by use of a perchloric acid procedure. (1)H NMR spectroscopy was used for metabolite analysis. "Null sample" (i.e. cell-free) experiments were performed by either rinsing dishes with medium or incubating the medium in Petri dishes from five different manufacturers for 72 h and then by performing a dummy "extraction" of each Petri dish by the perchloric acid, methanol-chloroform, or acetonitrile procedures. Principal components analysis was used for classification of samples and to determine the contaminants arising from plasticware. RESULTS: We found that even brief rinsing of cell culture plasticware with culture medium elutes artefactual chemicals, the (1)H NMR signals of which could confound assays of acetate, succinate, and glycolate. Incubation of culture medium in cell-culture dishes for 72 h (as in a typical cell-culture experiment) followed by perchloric extraction in the dishes enhanced elution of the artefacts. These artefacts were present, but somewhat less pronounced, in the (1)H NMR spectra of null samples extracted with methanol and acetonitrile. Ethanol, lactate, alanine, fructose, and fumarate signals that appear in the (1)H NMR spectrum of the unused (pure) medium originate from FBS. CONCLUSIONS: Plastic Petri dishes from five different manufacturers gave rise to essentially identical artefactual peaks. Use of a pH indicator to assist neutralisation introduced still more artefactual signals in the aromatic region, as well as methanol and ethanol signals. Methanol and acetonitrile extracts also contained artefacts arising from the plasticware, although the amounts were less than in the perchloric acid extracts. Finally, we provide suggestions for minimizing these artefacts. The best practice would be to run a "null" extraction with every batch of cellular metabolomics experiments to test for contamination and to provide a "background" spectrum.


Asunto(s)
Artefactos , Astrocitos/citología , Fibroblastos/metabolismo , Metaboloma/fisiología , Plásticos/química , Espectroscopía de Protones por Resonancia Magnética/métodos , Algoritmos , Técnicas de Cultivo de Célula/instrumentación , Línea Celular , Cultura , Células HeLa , Humanos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Manejo de Especímenes/instrumentación
6.
J Mol Cell Cardiol ; 55: 27-30, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23277191

RESUMEN

Perhexiline is a potent anti-anginal drug used for treatment of refractory angina and other forms of heart disease. It provides an oxygen sparing effect in the myocardium by creating a switch from fatty acid to glucose metabolism through partial inhibition of carnitine palmitoyltransferase 1 and 2. However, the precise molecular mechanisms underlying the cardioprotective effects elicited by perhexiline are not fully understood. The present study employed a combined proteomics, metabolomics and computational approach to characterise changes in murine hearts upon treatment with perhexiline. According to results based on difference in-gel electrophoresis, the most profound change in the cardiac proteome related to the activation of the pyruvate dehydrogenase complex. Metabolomic analysis by high-resolution nuclear magnetic resonance spectroscopy showed lower levels of total creatine and taurine in hearts of perhexiline-treated mice. Creatine and taurine levels were also significantly correlated in a cross-correlation analysis of all metabolites. Computational modelling suggested that far from inducing a simple shift from fatty acid to glucose oxidation, perhexiline may cause complex rebalancing of carbon and nucleotide phosphate fluxes, fuelled by increased lactate and amino acid uptake, to increase metabolic flexibility and to maintain cardiac output. This article is part of a Special Issue entitled "Focus on Cardiac Metabolism".


Asunto(s)
Fármacos Cardiovasculares/farmacología , Corazón/efectos de los fármacos , Metaboloma , Miocardio/metabolismo , Perhexilina/farmacología , Proteoma , Animales , Análisis por Conglomerados , Simulación por Computador , Masculino , Metabolómica , Ratones , Modelos Biológicos , Proteómica
7.
Metabolites ; 13(8)2023 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-37623883

RESUMEN

The results of publications in PubMed with the MeSH terms "cancer", "biology", "imaging and cancer", "metabolism" and "spectroscopy" are shown in Figure 1 in the form of a Venn diagram [...].

8.
J Mol Cell Cardiol ; 50(6): 982-90, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21354174

RESUMEN

A transgenic mouse model for conditional induction of long-term hibernation via myocardium-specific expression of a VEGF-sequestering soluble receptor allowed the dissection of the hibernation process into an initiation and a maintenance phase. The hypoxic initiation phase was characterized by peak levels of K(ATP) channel and glucose transporter 1 (GLUT1) expression. Glibenclamide, an inhibitor of K(ATP) channels, blocked GLUT1 induction. In the maintenance phase, tissue hypoxia and GLUT1 expression were reduced. Thus, we employed a combined "-omics" approach to resolve this cardioprotective adaptation process. Unguided bioinformatics analysis on the transcriptomic, proteomic and metabolomic datasets confirmed that anaerobic glycolysis was affected and that the observed enzymatic changes in cardiac metabolism were directly linked to hypoxia-inducible factor (HIF)-1 activation. Although metabolite concentrations were kept relatively constant, the combination of the proteomic and transcriptomic dataset improved the statistical confidence of the pathway analysis by 2 orders of magnitude. Importantly, proteomics revealed a reduced phosphorylation state of myosin light chain 2 and cardiac troponin I within the contractile apparatus of hibernating hearts in the absence of changes in protein abundance. Our study demonstrates how combining different "-omics" datasets aids in the identification of key biological pathways: chronic hypoxia resulted in a pronounced adaptive response at the transcript and the protein level to keep metabolite levels steady. This preservation of metabolic homeostasis is likely to contribute to the long-term survival of the hibernating myocardium.


Asunto(s)
Adaptación Fisiológica , Perfilación de la Expresión Génica , Homeostasis/fisiología , Aturdimiento Miocárdico/metabolismo , Proteoma , Animales , Biología Computacional , Regulación de la Expresión Génica/fisiología , Hipoxia/metabolismo , Hipoxia/patología , Redes y Vías Metabólicas/fisiología , Ratones , Ratones Transgénicos , Aturdimiento Miocárdico/patología , Procesamiento Proteico-Postraduccional/fisiología , Proteómica , Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
9.
Circ Res ; 102(9): 1046-56, 2008 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-18388323

RESUMEN

We have recently demonstrated that stem cell antigen 1-positive (Sca-1(+)) progenitors exist in the vascular adventitia of apolipoprotein E-deficient (apoE(-/-)) mice and contribute to smooth muscle cell (SMC) accumulation in vein graft atherosclerosis. Using a combined proteomic and metabolomic approach, we now characterize these local progenitors, which participate in the formation of native atherosclerotic lesions in chow-fed apoE(-/-) mice. Unlike Sca-1(+) progenitors from embryonic stem cells, the resident Sca-1(+) stem cell population from the vasculature acquired a mature aortic SMC phenotype after platelet-derived growth factor-BB stimulation. It shared proteomic and metabolomic characteristics of apoE(-/-) SMCs, which were clearly distinct from wild-type SMCs under normoxic and hypoxic conditions. Among the differentially expressed proteins were key enzymes in glucose metabolism, resulting in faster glucose consumption and a compensatory reduction in baseline interleukin-6 secretion. The latter was associated with a marked upregulation of insulin-like growth factor binding proteins (IGFBPs) 3 and 6. Notably, reconstitution of interleukin-6 to levels measured in the conditioned medium of wild-type SMCs attenuated the elevated IGFBP expression in apoE(-/-) SMCs and their vascular progenitors. This coregulation of apoE, interleukin-6, and IGFBPs was replicated in wild-type SMCs from hypercholesterolemic mice and confirmed by silencing apoE expression in SMCs from normocholesterolemic mice. In summary, we provide evidence that Sca-1(+) progenitors contribute to native atherosclerosis in apoE(-/-) mice, that apoE deficiency and hypercholesterolemia alter progenitor cell behavior, and that inflammatory cytokines such as interleukin-6 act as metabolic regulators in SMCs of hyperlipidemic mice.


Asunto(s)
Apolipoproteínas E/metabolismo , Arterias/metabolismo , Aterosclerosis/metabolismo , Tejido Conectivo/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteómica , Células Madre/metabolismo , Túnica Media/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Arterias/enzimología , Arterias/patología , Ataxina-1 , Ataxinas , Aterosclerosis/genética , Aterosclerosis/patología , Becaplermina , Bioensayo , Hipoxia de la Célula , Células Cultivadas , Tejido Conectivo/enzimología , Tejido Conectivo/patología , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Glucosa/metabolismo , Hipercolesterolemia/metabolismo , Immunoblotting , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Interleucina-6/metabolismo , Espectroscopía de Resonancia Magnética , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteómica/métodos , Proteínas Proto-Oncogénicas c-sis , Células Madre/enzimología , Células Madre/patología , Espectrometría de Masas en Tándem , Túnica Media/enzimología , Túnica Media/patología
10.
Sci Rep ; 10(1): 13616, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32788746

RESUMEN

Topographical variations of metabolite concentrations have been reported in the duodenum, jejunum and ileum of the small intestine, and in human intestinal tumours from those regions, but there are no published metabolite concentrations measurements correlated with linear position in the mouse small intestine or intestinal tumours. Since DNA methylation dynamics are influenced by metabolite concentrations, they too could show linear anatomical variation. We measured metabolites by HR-MAS 1H NMR spectroscopy and DNA cytosine modifications by LC/MS, in normal small intestines of C57BL/6J wild-type mice, and in normal and tumour samples from ApcMin/+ mice. Wild-type mouse intestines showed approximately linear, negative concentration gradations from the pylorus (i.e. the junction with the stomach) of alanine, choline compounds, creatine, leucine and valine. ApcMin/+ mouse tumours showed negative choline and valine gradients, but a positive glycine gradient. 5-Hydroxymethylcytosine showed a positive gradient in the tumours. The linear gradients we found along the length of the mouse small intestine and in tumours contrast with previous reports of discrete concentration changes in the duodenum, jejunum and ileum. To our knowledge, this is also the first report of a systematic measurement of global levels of DNA cytosine modification in wild-type and ApcMin/+ mouse small intestine.


Asunto(s)
5-Metilcitosina/análogos & derivados , Proteína de la Poliposis Adenomatosa del Colon/genética , Colon/química , Neoplasias Intestinales/metabolismo , Intestino Delgado/química , Píloro/química , 5-Metilcitosina/química , Animales , Cromatografía Liquida , Femenino , Neoplasias Intestinales/genética , Masculino , Espectrometría de Masas , Metabolómica , Ratones , Ratones Endogámicos C57BL , Espectroscopía de Protones por Resonancia Magnética
11.
J Mol Cell Cardiol ; 46(2): 268-77, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19027023

RESUMEN

We applied a combined proteomic and metabolomic approach to obtain novel mechanistic insights in PKCvarepsilon-mediated cardioprotection. Mitochondrial and cytosolic proteins from control and transgenic hearts with constitutively active or dominant negative PKCvarepsilon were analyzed using difference in-gel electrophoresis (DIGE). Among the differentially expressed proteins were creatine kinase, pyruvate kinase, lactate dehydrogenase, and the cytosolic isoforms of aspartate amino transferase and malate dehydrogenase, the two enzymatic components of the malate aspartate shuttle, which are required for the import of reducing equivalents from glycolysis across the inner mitochondrial membrane. These enzymatic changes appeared to be dependent on PKCvarepsilon activity, as they were not observed in mice expressing inactive PKCvarepsilon. High-resolution proton nuclear magnetic resonance ((1)H-NMR) spectroscopy confirmed a pronounced effect of PKCvarepsilon activity on cardiac glucose and energy metabolism: normoxic hearts with constitutively active PKCvarepsilon had significantly lower concentrations of glucose, lactate, glutamine and creatine, but higher levels of choline, glutamate and total adenosine nucleotides. Moreover, the depletion of cardiac energy metabolites was slower during ischemia/reperfusion injury and glucose metabolism recovered faster upon reperfusion in transgenic hearts with active PKCvarepsilon. Notably, inhibition of PKCvarepsilon resulted in compensatory phosphorylation and mitochondrial translocation of PKCdelta. Taken together, our findings are the first evidence that PKCvarepsilon activity modulates cardiac glucose metabolism and provide a possible explanation for the synergistic effect of PKCdelta and PKCvarepsilon in cardioprotection.


Asunto(s)
Glucosa/metabolismo , Metabolómica/métodos , Miocardio/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Proteómica/métodos , Animales , Modelos Animales de Enfermedad , Ratones , Mitocondrias/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Unión Proteica
12.
Sci Rep ; 9(1): 10244, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31308404

RESUMEN

The enzyme succinate dehydrogenase (SDH) functions in the citric acid cycle and loss of function predisposes to the development of phaeochromocytoma/paraganglioma (PPGL), wild type gastrointestinal stromal tumour (wtGIST) and renal cell carcinoma. SDH-deficient tumours are most commonly associated with a germline SDH subunit gene (SDHA/B/C/D) mutation but can also be associated with epigenetic silencing of the SDHC gene. However, clinical diagnostic testing for an SDHC epimutation is not widely available. The objective of this study was to investigate the indications for and the optimum diagnostic pathways for the detection of SDHC epimutations in clinical practice. SDHC promoter methylation analysis of 32 paraffin embedded tumours (including 15 GIST and 17 PPGL) was performed using a pyrosequencing technique and correlated with SDHC gene expression. SDHC promoter methylation was identified in 6 (18.7%) tumours. All 6 SDHC epimutation cases presented with SDH deficient wtGIST and 3/6 cases had multiple primary tumours. No case of constitutional SDHC promoter hypermethylation was detected. Whole genome sequencing of germline DNA from three wtGIST cases with an SDHC epimutation, did not reveal any causative sequence anomalies. Herein, we recommend a diagnostic workflow for the detection of an SDHC epimutation in a service setting.


Asunto(s)
Epigénesis Genética/genética , Tumores del Estroma Gastrointestinal/genética , Succinato Deshidrogenasa/genética , Adolescente , Neoplasias de las Glándulas Suprarrenales/genética , Adulto , Anciano , Metilación de ADN/genética , Epigenómica/métodos , Femenino , Tumores del Estroma Gastrointestinal/metabolismo , Genes Reguladores/genética , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Mutación , Paraganglioma/genética , Feocromocitoma/genética , Regiones Promotoras Genéticas/genética , Succinato Deshidrogenasa/metabolismo , Transcriptoma/genética
13.
Trends Cardiovasc Med ; 17(2): 43-8, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17292045

RESUMEN

Proteomics and metabolomics offer a nonbiased suite of tools to address pathophysiologic mechanisms from various levels by integrating signal transduction, cellular metabolism, and phenotype analysis. To link alterations of cellular proteins to metabolism and function, we have recently combined proteomic and metabolomic techniques. Examples, including genetic manipulation, ischemic preconditioning, atherosclerosis, and stem cell differentiation, are discussed to illustrate how the combination of these updated technologies may advance our understanding of cardiovascular biology.


Asunto(s)
Investigación Biomédica/tendencias , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Proteómica/métodos , Humanos , Precondicionamiento Isquémico , Redes y Vías Metabólicas/fisiología , Fenotipo , Transducción de Señal , Trasplante de Células Madre
14.
Cancer Res ; 66(1): 427-34, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16397258

RESUMEN

MN58b is a novel anticancer drug that inhibits choline kinase, resulting in inhibition of phosphocholine synthesis. The aim of this work was to develop a noninvasive and robust pharmacodynamic biomarker for target inhibition and, potentially, tumor response following MN58b treatment. Human HT29 (colon) and MDA-MB-231 (breast) carcinoma cells were examined by proton (1H) and phosphorus (31P) magnetic resonance spectroscopy (MRS) before and after treatment with MN58b both in culture and in xenografts. An in vitro time course study of MN58b treatment was also carried out in MDA-MB-231 cells. In addition, enzymatic assays of choline kinase activity in cells were done. A decrease in phosphocholine and total choline levels (P < 0.05) was observed in vitro in both cell lines after MN58b treatment, whereas the inactive analogue ACG20b had no effect. In MDA-MB-231 cells, phosphocholine fell significantly as early as 4 hours following MN58b treatment, whereas a drop in cell number was observed at 48 hours. Significant correlation was also found between phosphocholine levels (measured by MRS) and choline kinase activities (r2 = 0.95, P = 0.0008) following MN58b treatment. Phosphomonoesters also decreased significantly (P < 0.05) in both HT29 and MDA-MB-231 xenografts with no significant changes in controls. 31P-MRS and 1H-MRS of tumor extracts showed a significant decrease in phosphocholine (P < or = 0.05). Inhibition of choline kinase by MN58b resulted in altered phospholipid metabolism both in cultured tumor cells and in vivo. Phosphocholine levels were found to correlate with choline kinase activities. The decrease in phosphocholine, total choline, and phosphomonoesters may have potential as noninvasive pharmacodynamic biomarkers for determining tumor response following treatment with choline kinase inhibitors.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Butanos/farmacología , Carcinoma/tratamiento farmacológico , Colina Quinasa/antagonistas & inhibidores , Neoplasias del Colon/tratamiento farmacológico , Compuestos de Piridinio/farmacología , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Carcinoma/enzimología , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Inhibidores Enzimáticos/farmacología , Células HT29 , Humanos , Ratones , Ratones Desnudos , Resonancia Magnética Nuclear Biomolecular/métodos , Fósforo , Fosforilcolina/metabolismo , Protones , Ensayos Antitumor por Modelo de Xenoinjerto
15.
JCO Precis Oncol ; 2: 1-12, 2018 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-30949620

RESUMEN

PURPOSE: Mutations in the mitochondrial enzyme succinate dehydrogenase (SDH) subunit genes are associated with a wide spectrum of tumours including phaeochromocytoma and paraganglioma (PPGL) 1, 2, gastrointestinal stromal tumours (GIST) 3, renal cell carcinoma (RCC) 4 and pituitary adenomas5. SDH-related tumorigenesis is believed to be secondary to accumulation of the oncometabolite succinate. Our aim was to investigate the potential clinical applications of MRI spectroscopy (1H-MRS) in a range of suspected SDH-related tumours. PATIENTS AND METHODS: Fifteen patients were recruited to this study. Respiratory-gated single-voxel 1H-MRS was performed at 3T to quantify the content of succinate at 2.4 ppm and choline at 3.22 ppm. RESULTS: A succinate peak was seen in six patients, all of whom had a germline SDHx mutation or loss of SDHB by immunohistochemistry. A succinate peak was also detected in two patients with a metastatic wild-type GIST (wtGIST) and no detectable germline SDHx mutation but a somatic epimutation in SDHC. Three patients without a tumour succinate peak retained SDHB expression, consistent with SDH functionality. In six cases with a borderline or absent peak, technical difficulties such as motion artefact rendered 1H-MRS difficult to interpret. Sequential imaging in a patient with a metastatic abdominal paraganglioma demonstrated loss of the succinate peak after four cycles of [177Lu]-DOTATATE, with a corresponding biochemical response in normetanephrine. CONCLUSIONS: This study has demonstrated the translation into clinical practice of in vivo metabolomic analysis using 1H-MRS in patients with SDH-deficient tumours. Potential applications include non-invasive diagnosis and disease stratification, as well as monitoring of tumour response to targeted treatments.

16.
Cell Metab ; 27(4): 898-913.e7, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29617647

RESUMEN

Animals require an immediate response to oxygen availability to allow rapid shifts between oxidative and glycolytic metabolism. These metabolic shifts are highly regulated by the HIF transcription factor. The factor inhibiting HIF (FIH) is an asparaginyl hydroxylase that controls HIF transcriptional activity in an oxygen-dependent manner. We show here that FIH loss increases oxidative metabolism, while also increasing glycolytic capacity, and that this gives rise to an increase in oxygen consumption. We further show that the loss of FIH acts to accelerate the cellular metabolic response to hypoxia. Skeletal muscle expresses 50-fold higher levels of FIH than other tissues: we analyzed skeletal muscle FIH mutants and found a decreased metabolic efficiency, correlated with an increased oxidative rate and an increased rate of hypoxic response. We find that FIH, through its regulation of oxidation, acts in concert with the PHD/vHL pathway to accelerate HIF-mediated metabolic responses to hypoxia.


Asunto(s)
Adaptación Fisiológica , Oxigenasas de Función Mixta/metabolismo , Oxígeno/metabolismo , Animales , Hipoxia de la Célula , Regulación de la Expresión Génica , Glucólisis/fisiología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno , Procolágeno-Prolina Dioxigenasa/metabolismo , Transducción de Señal , Transcripción Genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
17.
PLoS One ; 12(10): e0185980, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29069098

RESUMEN

METHODS: We quantified 378 HRMAS 1H NMR spectra of human brain tumours (132 glioblastomas, 101 astrocytomas, 75 meningiomas, 37 oligodendrogliomas and 33 metastases) from the eTumour database and looked for metabolic interactions by metabolite-metabolite correlation analysis (MMCA). RESULTS: All tumour types showed remarkably similar metabolic correlations. Lactate correlated positively with alanine, glutamate with glutamine; creatine + phosphocreatine (tCr) correlated positively with lactate, alanine and choline + phosphocholine + glycerophosphocholine (tCho), and tCho correlated positively with lactate; fatty acids correlated negatively with lactate, glutamate + glutamine (tGlut), tCr and tCho. Oligodendrogliomas had fewer correlations but they still fitted that pattern. CONCLUSIONS: Possible explanations include (i) glycolytic tumour cells (the Warburg effect) generating pyruvate which is converted to lactate, alanine, glutamate and then glutamine; (ii) an association between elevated glycolysis and increased choline turnover in membranes; (iii) an increase in the tCr pool to facilitate phosphocreatine-driven glutamate uptake; (iv) lipid signals come from cytosolic lipid droplets in necrotic or pre-necrotic tumour tissue that has lower concentrations of anabolic and catabolic metabolites. Additional metabolite exchanges with host cells may also be involved. If tumours co-opt a standard set of biochemical mechanisms to grow in the brain, then drugs might be developed to disrupt those mechanisms.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Espectroscopía de Protones por Resonancia Magnética/métodos , Neoplasias Encefálicas/clasificación , Neoplasias Encefálicas/patología , Humanos
18.
Cancer Metab ; 5: 4, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28261475

RESUMEN

BACKGROUND: Ketone bodies have both metabolic and epigenetic roles in cancer. In several studies, they showed an anti-cancer effect via inhibition of histone deacetylases; however, other studies observed faster tumour growth. The related molecule butyrate also inhibits growth of some cancer cells and accelerates it in others. This "butyrate paradox" is thought to be due to butyrate mediating histone acetylation and thus inhibiting cell proliferation in cancers that preferentially utilise glucose (the Warburg effect); whereas in cells that oxidise butyrate as a fuel, it fails to reach inhibitory concentrations and can stimulate growth. METHODS: We treated transgenic mice bearing spontaneous MMTV-NEU-NT mammary tumours with the ketone body ß-hydroxybutyrate (ß-OHB) and monitored tumour growth, metabolite concentrations and histone acetylation. In a cell line derived from these tumours, we also measured uptake of ß-OHB and glucose, and lactate production, in the absence and presence of ß-OHB. RESULTS: ß-OHB administration accelerated growth of MMTV-NEU-NT tumours, and their metabolic profile showed significant increases in ATP, glutamine, serine and choline-related metabolites. The ß-OHB concentration within the treated tumours, 0.46 ± 0.05 µmol/g, had no effect on histone acetylation as shown by western blots. Cultured tumour cells incubated with 0.5 mM ß-OHB showed ß-OHB uptake that would be equivalent to 54% of glycolytic ATP phosphorylation and no significant change in glucose consumption or lactate production. CONCLUSIONS: These results suggest that a ß-OHB paradox may occur in these mammary tumours in a manner analogous to the butyrate paradox. At low ß-OHB concentrations (<1 mM, as observed in our tumour model post-treatment), and in the absence of a Warburg effect, ß-OHB is consumed and thus acts as an oxidative energy source and not as an epigenetic factor. This would explain the increase in tumour growth after treatment, the metabolic profiles and the absence of an effect on histone H3 acetylation.

19.
Clin Cancer Res ; 11(10): 3705-13, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15897567

RESUMEN

PURPOSE: To use (31)P and (1)H magnetic resonance spectroscopy (MRS) to assess changes in tumor metabolic profile in vivo in response to 5,6-dimethylxanthenone-4-acetic acid (DMXAA) with a view to identifying biomarkers associated with tumor dose response. EXPERIMENTAL DESIGN: In vivo (31)P and (1)H MRS measurements of (a) tumor bioenergetics [beta-nucleoside triphosphate/inorganic phosphate (beta-NTP/Pi)], (b) the membrane-associated phosphodiesters and phosphomonoesters (PDE/PME), (c) choline (mmol/L), and (d) lactate/water ratio were made on murine HT29 colon carcinoma xenografts pretreatment and 6 or 24 hours posttreatment with increasing doses of DMXAA. Following in vivo MRS, the tumors were excised and used for high-resolution (31)P and (1)H MRS of extracts to provide validation of the in vivo MRS data, histologic analysis of necrosis, and high-performance liquid chromatography. RESULTS: Both beta-NTP/Pi and PDE/PME decreased in a dose-dependent manner 6 hours posttreatment with DMXAA, with significant decreases in beta-NTP/Pi with 15 mg/kg (P < 0.001) and 21 mg/kg (P < 0.01). A significant decrease in total choline in vivo was found 24 hours posttreatment with 21 mg/kg DMXAA (P < 0.05); this was associated with a significant reduction in the concentration of the membrane degradation products glycerophosphoethanolamine and glycerophosphocholine measured in tissue extracts (P < 0.05). CONCLUSIONS: The reduction in tumor energetics and membrane turnover is consistent with the vascular-disrupting activity of DMXAA. (31)P MRS revealed tumor response to DMXAA at doses below the maximum tolerated dose for mice. Both (31)P and (1)H MRS provide biomarkers of tumor response to DMXAA that could be used in clinical trials.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Xantonas/farmacología , Animales , Carcinoma/metabolismo , Membrana Celular/metabolismo , Neoplasias del Colon/metabolismo , Relación Dosis-Respuesta a Droga , Metabolismo Energético , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica , Trasplante Heterólogo
20.
Metabolomics ; 12: 120, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27429605

RESUMEN

INTRODUCTION: The androgen receptor (AR) is the master regulator of prostate cancer cell metabolism. Degarelix is a novel gonadotrophin-releasing hormone blocker, used to decrease serum androgen levels in order to treat advanced human prostate cancer. Little is known of the rapid metabolic response of the human prostate cancer tissue samples to the decreased androgen levels. OBJECTIVES: To investigate the metabolic responses in benign and cancerous tissue samples from patients after treatment with Degarelix by using HRMAS 1H NMR spectroscopy. METHODS: Using non-destructive HR-MAS 1H NMR spectroscopy we analysed the metabolic changes induced by decreased AR signalling in human prostate cancer tissue samples. Absolute concentrations of the metabolites alanine, lactate, glutamine, glutamate, citrate, choline compounds [t-choline = choline + phosphocholine (PC) + glycerophosphocholine (GPC)], creatine compounds [t-creatine = creatine (Cr) + phosphocreatine (PCr)], taurine, myo-inositol and polyamines were measured in benign prostate tissue samples (n = 10), in prostate cancer specimens from untreated patients (n = 7) and prostate cancer specimens from patients treated with Degarelix (n = 6). RESULTS: Lactate, alanine and t-choline concentrations were significantly elevated in high-grade prostate cancer samples when compared to benign samples in untreated patients. Decreased androgen levels resulted in significant decreases of lactate and t-choline concentrations in human prostate cancer biopsies. CONCLUSIONS: The reduced concentrations of lactate and t-choline metabolites due to Degarelix could in principle be monitored by in vivo 1H MRS, which suggests that it would be possible to monitor the effects of physical or chemical castration in patients by that non-invasive method.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA