Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cancers (Basel) ; 14(4)2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35205709

RESUMEN

An elevated neutrophil-lymphocyte ratio negatively predicts the outcome of patients with cancer and is associated with cachexia, the terminal wasting syndrome. Here, using murine model systems of colorectal and pancreatic cancer we show that neutrophilia in the circulation and multiple organs, accompanied by extramedullary hematopoiesis, is an early event during cancer progression. Transcriptomic and metabolic assessment reveals that neutrophils in tumor-bearing animals utilize aerobic glycolysis, similar to cancer cells. Although pharmacological inhibition of aerobic glycolysis slows down tumor growth in C26 tumor-bearing mice, it precipitates cachexia, thereby shortening the overall survival. This negative effect may be explained by our observation that acute depletion of neutrophils in pre-cachectic mice impairs systemic glucose homeostasis secondary to altered hepatic lipid processing. Thus, changes in neutrophil number, distribution, and metabolism play an adaptive role in host metabolic homeostasis during cancer progression. Our findings provide insight into early events during cancer progression to cachexia, with implications for therapy.

2.
Br J Cancer ; 122(2): 258-265, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819193

RESUMEN

BACKGROUND: Epidemiological studies suggest that metformin may reduce the incidence of cancer in patients with diabetes and multiple late phase clinical trials assessing the potential of repurposing this drug are underway. Transcriptomic profiling of tumour samples is an excellent tool to understand drug bioactivity, identify candidate biomarkers and assess for mechanisms of resistance to therapy. METHODS: Thirty-six patients with untreated primary breast cancer were recruited to a window study and transcriptomic profiling of tumour samples carried out before and after metformin treatment. RESULTS: Multiple genes that regulate fatty acid oxidation were upregulated at the transcriptomic level and there was a differential change in expression between two previously identified cohorts of patients with distinct metabolic responses. Increase in expression of a mitochondrial fatty oxidation gene composite signature correlated with change in a proliferation gene signature. In vitro assays showed that, in contrast to previous studies in models of normal cells, metformin reduces fatty acid oxidation with a subsequent accumulation of intracellular triglyceride, independent of AMPK activation. CONCLUSIONS: We propose that metformin at clinical doses targets fatty acid oxidation in cancer cells with implications for patient selection and drug combinations. CLINICAL TRIAL REGISTRATION: NCT01266486.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Ácidos Grasos/metabolismo , Metformina/farmacología , Proteínas Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Transcriptoma/efectos de los fármacos
3.
Cell Metab ; 28(5): 679-688.e4, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30244975

RESUMEN

Late-phase clinical trials investigating metformin as a cancer therapy are underway. However, there remains controversy as to the mode of action of metformin in tumors at clinical doses. We conducted a clinical study integrating measurement of markers of systemic metabolism, dynamic FDG-PET-CT, transcriptomics, and metabolomics at paired time points to profile the bioactivity of metformin in primary breast cancer. We show metformin reduces the levels of mitochondrial metabolites, activates multiple mitochondrial metabolic pathways, and increases 18-FDG flux in tumors. Two tumor groups are identified with distinct metabolic responses, an OXPHOS transcriptional response (OTR) group for which there is an increase in OXPHOS gene transcription and an FDG response group with increased 18-FDG uptake. Increase in proliferation, as measured by a validated proliferation signature, suggested that patients in the OTR group were resistant to metformin treatment. We conclude that mitochondrial response to metformin in primary breast cancer may define anti-tumor effect.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Hipoglucemiantes/farmacología , Redes y Vías Metabólicas/efectos de los fármacos , Metformina/farmacología , Adulto , Anciano , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucosa/análogos & derivados , Glucosa/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Metformina/uso terapéutico , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Transcriptoma/efectos de los fármacos
4.
Nat Med ; 24(11): 1691-1695, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30250142

RESUMEN

Mutations of the mitochondrial genome (mtDNA) underlie a substantial portion of mitochondrial disease burden. These disorders are currently incurable and effectively untreatable, with heterogeneous penetrance, presentation and prognosis. To address the lack of effective treatment for these disorders, we exploited a recently developed mouse model that recapitulates common molecular features of heteroplasmic mtDNA disease in cardiac tissue: the m.5024C>T tRNAAla mouse. Through application of a programmable nuclease therapy approach, using systemically administered, mitochondrially targeted zinc-finger nucleases (mtZFN) delivered by adeno-associated virus, we induced specific elimination of mutant mtDNA across the heart, coupled to a reversion of molecular and biochemical phenotypes. These findings constitute proof of principle that mtDNA heteroplasmy correction using programmable nucleases could provide a therapeutic route for heteroplasmic mitochondrial diseases of diverse genetic origin.


Asunto(s)
Edición Génica , Mitocondrias Cardíacas/genética , Enfermedades Mitocondriales/genética , Nucleasas con Dedos de Zinc/genética , Animales , ADN Mitocondrial/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Mitocondrias Cardíacas/patología , Enfermedades Mitocondriales/patología , Enfermedades Mitocondriales/terapia , Mutación/genética , Pronóstico , ARN de Transferencia/genética , Nucleasas con Dedos de Zinc/uso terapéutico
5.
Cell Rep ; 24(5): 1093-1104.e6, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30067967

RESUMEN

Renal cell carcinomas (RCCs) are common cancers diagnosed in more than 350,000 people each year worldwide. Several pathways are de-regulated in RCCs, including mTORC1. However, how mTOR drives tumorigenesis in this context is unknown. The lack of faithful animal models has limited progress in understanding and targeting RCCs. Here, we generated a mouse model harboring the kidney-specific inactivation of Tsc1. These animals develop cysts that evolve into papillae, cystadenomas, and papillary carcinomas. Global profiling confirmed several metabolic derangements previously attributed to mTORC1. Notably, Tsc1 inactivation results in the accumulation of fumarate and in mTOR-dependent downregulation of the TCA cycle enzyme fumarate hydratase (FH). The re-expression of FH in cellular systems lacking Tsc1 partially rescued renal epithelial transformation. Importantly, the mTORC1-FH axis is likely conserved in human RCC specimens. We reveal a role of mTORC1 in renal tumorigenesis, which depends on the oncometabolite fumarate.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Fumaratos/metabolismo , Neoplasias Renales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Animales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Células Cultivadas , Femenino , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba
6.
Ecancermedicalscience ; 12: ed84, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30093919

RESUMEN

Breath biopsy enables the non-invasive collection and analysis of volatile organic compounds (VOCs) in exhaled breath, providing valuable information about disease processes occurring in the body. Metabolic changes occur in cancer cells at the earliest stages of disease. We discuss progress in the use of breath biopsy for discovery of breath-based biomarkers for early detection of cancer, and potential applications for breath biopsy in enabling precision medicine in cancer.

7.
Mol Cell ; 69(4): 581-593.e7, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29452638

RESUMEN

The bioenergetics and molecular determinants of the metabolic response to mitochondrial dysfunction are incompletely understood, in part due to a lack of appropriate isogenic cellular models of primary mitochondrial defects. Here, we capitalize on a recently developed cell model with defined levels of m.8993T>G mutation heteroplasmy, mTUNE, to investigate the metabolic underpinnings of mitochondrial dysfunction. We found that impaired utilization of reduced nicotinamide adenine dinucleotide (NADH) by the mitochondrial respiratory chain leads to cytosolic reductive carboxylation of glutamine as a new mechanism for cytosol-confined NADH recycling supported by malate dehydrogenase 1 (MDH1). We also observed that increased glycolysis in cells with mitochondrial dysfunction is associated with increased cell migration in an MDH1-dependent fashion. Our results describe a novel link between glycolysis and mitochondrial dysfunction mediated by reductive carboxylation of glutamine.


Asunto(s)
Citosol/metabolismo , Glutamina/metabolismo , Malato Deshidrogenasa/metabolismo , Mitocondrias/patología , NAD/metabolismo , Osteosarcoma/patología , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Movimiento Celular , Ciclo del Ácido Cítrico , ADN Mitocondrial/genética , Metabolismo Energético , Femenino , Glucosa/metabolismo , Glucólisis , Humanos , Mitocondrias/metabolismo , Osteosarcoma/genética , Osteosarcoma/metabolismo , Oxidación-Reducción , Células Tumorales Cultivadas
8.
Cell Rep ; 21(4): 1036-1047, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29069586

RESUMEN

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid (TCA) cycle mutated in hereditary and sporadic cancers. Despite recent advances in understanding its role in tumorigenesis, the effects of FH loss on mitochondrial metabolism are still unclear. Here, we used mouse and human cell lines to assess mitochondrial function of FH-deficient cells. We found that human and mouse FH-deficient cells exhibit decreased respiration, accompanied by a varying degree of dysfunction of respiratory chain (RC) complex I and II. Moreover, we show that fumarate induces succination of key components of the iron-sulfur cluster biogenesis family of proteins, leading to defects in the biogenesis of iron-sulfur clusters that affect complex I function. We also demonstrate that suppression of complex II activity is caused by product inhibition due to fumarate accumulation. Overall, our work provides evidence that the loss of a single TCA cycle enzyme is sufficient to cause combined RC activity dysfunction.


Asunto(s)
Complejo II de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Fumarato Hidratasa/metabolismo , Animales , Línea Celular Tumoral , Respiración de la Célula , Fumarato Hidratasa/deficiencia , Fumarato Hidratasa/genética , Fumaratos/metabolismo , Humanos , Proteínas Hierro-Azufre/metabolismo , Ratones
9.
Trends Endocrinol Metab ; 28(10): 748-757, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28938972

RESUMEN

Altered metabolism is a distinct feature of cancer cells. During transformation, the entire metabolic network is rewired to efficiently convert nutrients to biosynthetic precursors to sustain cancer cell growth and proliferation. Whilst the molecular underpinnings of this metabolic reprogramming have been described, its role in tumor progression is still under investigation. Importantly, the mitochondrion is a central actor in many of the metabolic processes that are altered in tumors. Yet, we have only begun to understand the dualities of mitochondrial function during cancer metastasis and therapy resistance. Paradoxically, mitochondrial metabolism can be both advantageous and detrimental to these processes, highlighting the need for a better understanding of the molecular and microenvironmental cues that define the role of this fascinating organelle. In this review article, we present an updated view on the different mitochondrial metabolic strategies adopted by cancer cells to overcome the many hurdles faced during tumor progression.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Mitocondrias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Progresión de la Enfermedad , Metabolismo Energético/fisiología , Humanos , Redes y Vías Metabólicas/fisiología
10.
Nat Commun ; 7: 13041, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27721378

RESUMEN

Cancer cells undergo a multifaceted rewiring of cellular metabolism to support their biosynthetic needs. Although the major determinants of this metabolic transformation have been elucidated, their broad biological implications and clinical relevance are unclear. Here we systematically analyse the expression of metabolic genes across 20 different cancer types and investigate their impact on clinical outcome. We find that cancers undergo a tissue-specific metabolic rewiring, which converges towards a common metabolic landscape. Of note, downregulation of mitochondrial genes is associated with the worst clinical outcome across all cancer types and correlates with the expression of epithelial-to-mesenchymal transition gene signature, a feature of invasive and metastatic cancers. Consistently, suppression of mitochondrial genes is identified as a key metabolic signature of metastatic melanoma and renal cancer, and metastatic cell lines. This comprehensive analysis reveals unexpected facets of cancer metabolism, with important implications for cancer patients' stratification, prognosis and therapy.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Neoplasias/genética , Especificidad de Órganos , Fosforilación Oxidativa , Análisis de Supervivencia , Resultado del Tratamiento
11.
Nature ; 537(7621): 544-547, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27580029

RESUMEN

Mutations of the tricarboxylic acid cycle enzyme fumarate hydratase cause hereditary leiomyomatosis and renal cell cancer. Fumarate hydratase-deficient renal cancers are highly aggressive and metastasize even when small, leading to a very poor clinical outcome. Fumarate, a small molecule metabolite that accumulates in fumarate hydratase-deficient cells, plays a key role in cell transformation, making it a bona fide oncometabolite. Fumarate has been shown to inhibit α-ketoglutarate-dependent dioxygenases that are involved in DNA and histone demethylation. However, the link between fumarate accumulation, epigenetic changes, and tumorigenesis is unclear. Here we show that loss of fumarate hydratase and the subsequent accumulation of fumarate in mouse and human cells elicits an epithelial-to-mesenchymal-transition (EMT), a phenotypic switch associated with cancer initiation, invasion, and metastasis. We demonstrate that fumarate inhibits Tet-mediated demethylation of a regulatory region of the antimetastatic miRNA cluster mir-200ba429, leading to the expression of EMT-related transcription factors and enhanced migratory properties. These epigenetic and phenotypic changes are recapitulated by the incubation of fumarate hydratase-proficient cells with cell-permeable fumarate. Loss of fumarate hydratase is associated with suppression of miR-200 and the EMT signature in renal cancer and is associated with poor clinical outcome. These results imply that loss of fumarate hydratase and fumarate accumulation contribute to the aggressive features of fumarate hydratase-deficient tumours.


Asunto(s)
Epigénesis Genética , Transición Epitelial-Mesenquimal , Fumaratos/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Fumarato Hidratasa/deficiencia , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Células HEK293 , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Mesodermo/metabolismo , Ratones , MicroARNs/genética , Factores de Transcripción/metabolismo , Transcriptoma
12.
J Neuroinflammation ; 13(1): 232, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27590826

RESUMEN

BACKGROUND: Neural stem cells (NSCs) display tissue trophic and immune modulatory therapeutic activities after transplantation in central nervous system disorders. The intercellular interplay between stem cells and target immune cells is increased in NSCs exposed to inflammatory cues. Here, we hypothesize that inflammatory cytokine signalling leads to metabolic reprogramming of NSCs regulating some of their immune modulatory effects. METHODS: NSC lines were prepared from the subventricular zone (SVZ) of 7-12-week-old mice. Whole secretome-based screening and analysis of intracellular small metabolites was performed in NSCs exposed to cocktails of either Th1-like (IFN-γ, 500 U/ml; TNF-α, 200 U/ml; IL-1ß, 100 U/ml) or Th2-like (IL-4, IL-5 and IL-13; 10 ng/ml) inflammatory cytokines for 16 h in vitro. Isotopologues distribution of arginine and downstream metabolites was assessed by liquid chromatography/mass spectrometry in NSCs incubated with U-(13)C6 L-arginine in the presence or absence of Th1 or Th2 cocktails (Th1 NSCs or Th2 NSCs). The expression of arginase I and II was investigated in vitro in Th1 NSCs and Th2 NSCs and in vivo in the SVZ of mice with experimental autoimmune encephalomyelitis, as prototypical model of Th1 cell-driven brain inflammatory disease. The effects of the inflammatory cytokine signalling were studied in NSC-lymph node cells (LNC) co-cultures by flow cytometry-based analysis of cell proliferation following pan-arginase inhibition with N(ω)-hydroxy-nor-arginine (nor-NOHA). RESULTS: Cytokine-primed NSCs showed significantly higher anti-proliferative effect in co-cultures vs. control NSCs. Metabolomic analysis of intracellular metabolites revealed alteration of arginine metabolism and increased extracellular arginase I activity in cytokine-primed NSCs. Arginase inhibition by nor-NOHA partly rescued the anti-proliferative effects of cytokine-primed NSCs. CONCLUSIONS: Our work underlines the use of metabolic profiling as hypothesis-generating tools that helps unravelling how stem cell-mediated mechanisms of tissue restoration become affected by local inflammatory responses. Among different therapeutic candidates, we identify arginase signalling as novel metabolic determinant of the NSC-to-immune system communication.


Asunto(s)
Arginina/metabolismo , Citocinas/metabolismo , Factores Inmunológicos/metabolismo , Células-Madre Neurales/inmunología , Células-Madre Neurales/metabolismo , Animales , Arginasa/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Colorimetría , Citocinas/farmacología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ventrículos Laterales/citología , Metabolómica , Ratones , Células-Madre Neurales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
13.
Nucleic Acids Res ; 44(16): 7804-16, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27466392

RESUMEN

Mitochondrial diseases are frequently associated with mutations in mitochondrial DNA (mtDNA). In most cases, mutant and wild-type mtDNAs coexist, resulting in heteroplasmy. The selective elimination of mutant mtDNA, and consequent enrichment of wild-type mtDNA, can rescue pathological phenotypes in heteroplasmic cells. Use of the mitochondrially targeted zinc finger-nuclease (mtZFN) results in degradation of mutant mtDNA through site-specific DNA cleavage. Here, we describe a substantial enhancement of our previous mtZFN-based approaches to targeting mtDNA, allowing near-complete directional shifts of mtDNA heteroplasmy, either by iterative treatment or through finely controlled expression of mtZFN, which limits off-target catalysis and undesired mtDNA copy number depletion. To demonstrate the utility of this improved approach, we generated an isogenic distribution of heteroplasmic cells with variable mtDNA mutant level from the same parental source without clonal selection. Analysis of these populations demonstrated an altered metabolic signature in cells harbouring decreased levels of mutant m.8993T>G mtDNA, associated with neuropathy, ataxia, and retinitis pigmentosa (NARP). We conclude that mtZFN-based approaches offer means for mtDNA heteroplasmy manipulation in basic research, and may provide a strategy for therapeutic intervention in selected mitochondrial diseases.


Asunto(s)
ADN Mitocondrial/genética , Endonucleasas/metabolismo , Mitocondrias/metabolismo , Mutación/genética , Dedos de Zinc , Línea Celular Tumoral , Citometría de Flujo , Dosificación de Gen , Humanos , ARN Catalítico/metabolismo
14.
Sci Rep ; 6: 22950, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26972697

RESUMEN

The hypoxic tumour microenvironment represents an aggressive, therapy-resistant compartment. As arginine is required for specific hypoxia-induced processes, we hypothesised that arginine-deprivation therapy may be useful in targeting hypoxic cancer cells. We explored the effects of the arginine-degrading agent ADI-PEG20 on hypoxia-inducible factor (HIF) activation, the hypoxia-induced nitric oxide (NO) pathway and proliferation using HCT116 and UMUC3 cells and xenografts. The latter lack argininosuccinate synthetase (ASS1) making them auxotrophic for arginine. In HCT116 cells, ADI-PEG20 inhibited hypoxic-activation of HIF-1α and HIF-2α, leading to decreased inducible-nitric oxide synthase (iNOS), NO-production, and VEGF. Interestingly, combining hypoxia and ADI-PEG20 synergistically inhibited ASS1. ADI-PEG20 inhibited mTORC1 and activated the unfolded protein response providing a mechanism for inhibition of HIF and ASS1. ADI-PEG20 inhibited tumour growth, impaired hypoxia-associated NO-production, and decreased vascular perfusion. Expression of HIF-1α/HIF-2α/iNOS and VEGF were reduced, despite an increased hypoxic tumour fraction. Similar effects were observed in UMUC3 xenografts. In summary, ADI-PEG20 inhibits HIF-activated processes in two tumour models with widely different arginine biology. Thus, ADI-PEG20 may be useful in the clinic to target therapy-resistant hypoxic cells in ASS1-proficient tumours and ASS1-deficient tumours.


Asunto(s)
Hidrolasas/farmacología , Neoplasias/tratamiento farmacológico , Óxido Nítrico/biosíntesis , Polietilenglicoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Arginina/metabolismo , Argininosuccinato Sintasa/antagonistas & inhibidores , Argininosuccinato Sintasa/genética , Argininosuccinato Sintasa/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Western Blotting , Hipoxia de la Célula , Línea Celular Tumoral , Células HCT116 , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones SCID , Complejos Multiproteicos/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Perfusión , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Nature ; 531(7592): 110-3, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26909577

RESUMEN

The RAS/MAPK (mitogen-activated protein kinase) signalling pathway is frequently deregulated in non-small-cell lung cancer, often through KRAS activating mutations. A single endogenous mutant Kras allele is sufficient to promote lung tumour formation in mice but malignant progression requires additional genetic alterations. We recently showed that advanced lung tumours from Kras(G12D/+);p53-null mice frequently exhibit Kras(G12D) allelic enrichment (Kras(G12D)/Kras(wild-type) > 1) (ref. 7), implying that mutant Kras copy gains are positively selected during progression. Here we show, through a comprehensive analysis of mutant Kras homozygous and heterozygous mouse embryonic fibroblasts and lung cancer cells, that these genotypes are phenotypically distinct. In particular, Kras(G12D/G12D) cells exhibit a glycolytic switch coupled to increased channelling of glucose-derived metabolites into the tricarboxylic acid cycle and glutathione biosynthesis, resulting in enhanced glutathione-mediated detoxification. This metabolic rewiring is recapitulated in mutant KRAS homozygous non-small-cell lung cancer cells and in vivo, in spontaneous advanced murine lung tumours (which display a high frequency of Kras(G12D) copy gain), but not in the corresponding early tumours (Kras(G12D) heterozygous). Finally, we demonstrate that mutant Kras copy gain creates unique metabolic dependences that can be exploited to selectively target these aggressive mutant Kras tumours. Our data demonstrate that mutant Kras lung tumours are not a single disease but rather a heterogeneous group comprising two classes of tumours with distinct metabolic profiles, prognosis and therapeutic susceptibility, which can be discriminated on the basis of their relative mutant allelic content. We also provide the first, to our knowledge, in vivo evidence of metabolic rewiring during lung cancer malignant progression.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Genes ras/genética , Glucosa/metabolismo , Glucólisis , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Mutación/genética , Alelos , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ciclo del Ácido Cítrico , Progresión de la Enfermedad , Femenino , Fibroblastos/metabolismo , Genotipo , Glutatión/biosíntesis , Glutatión/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Oxidación-Reducción , Fenotipo , Pronóstico
17.
Nat Mater ; 15(2): 227-34, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26595121

RESUMEN

The profound metabolic reprogramming that occurs in cancer cells has been investigated primarily in two-dimensional cell cultures, which fail to recapitulate spatial aspects of cell-to-cell interactions as well as tissue gradients present in three-dimensional tumours. Here, we describe an engineered model to assemble three-dimensional tumours by rolling a scaffold-tumour composite strip. By unrolling the strip, the model can be rapidly disassembled for snapshot analysis, allowing spatial mapping of cell metabolism in concert with cell phenotype. We also show that the establishment of oxygen gradients within samples that are shaped by oxygen-dependent signalling pathways, as well as the consequential variations in cell growth, response to hypoxic gradients extending from normoxia to severe hypoxia, and therapy responsiveness, are consistent with those of tumours in vivo. Moreover, by using liquid chromatography tandem mass spectrometry, we mapped cellular metabolism and identified spatially defined metabolic signatures of cancer cells to reveal both known and novel metabolic responses to hypoxia.


Asunto(s)
Neoplasias/metabolismo , Oxígeno/metabolismo , Ingeniería de Tejidos , Andamios del Tejido , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Transducción de Señal
18.
Oncotarget ; 6(30): 30102-14, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26337086

RESUMEN

The mechanisms of cisplatin resistance, one of the major limitations of current chemotherapy, has only partially been described. We previously demonstrated that cisplatin-resistant ovarian cancer cells (C13), are characterized by reduced mitochondrial activity and higher glucose-dependency when compared to the cisplatin-sensitive counterpart (2008). In this work we further characterized the role of metabolic transformation in cisplatin resistance. By using transmitochondrial hybrids we show that metabolic reprogramming of cisplatin-resistant cell is not caused by inherent mtDNA mutations. We also found that C13 cells not only present an increased glucose-uptake and consumption, but also exhibit increased expression and enzymatic activity of the Pentose Phosphate pathway (PPP) enzyme Glucose-6-Phosphate Dehydrogenase (G6PDH). Moreover, we show that cisplatin-resistant cells are more sensitive to G6PDH inhibition. Even if the metabolomic fingerprint of ovarian cancer cells remains to be further elucidated, these findings indicate that PPP offers innovative potential targets to overcome cisplatin resistance.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glucosafosfato Deshidrogenasa/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Línea Celular Tumoral , ADN Mitocondrial/genética , Relación Dosis-Respuesta a Droga , Femenino , Glucosa/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Glutamina/metabolismo , Humanos , Mitocondrias/enzimología , Mitocondrias/patología , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
19.
Nat Commun ; 6: 6001, 2015 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-25613188

RESUMEN

Mutations in the tricarboxylic acid (TCA) cycle enzyme fumarate hydratase (FH) are associated with a highly malignant form of renal cancer. We combined analytical chemistry and metabolic computational modelling to investigate the metabolic implications of FH loss in immortalized and primary mouse kidney cells. Here, we show that the accumulation of fumarate caused by the inactivation of FH leads to oxidative stress that is mediated by the formation of succinicGSH, a covalent adduct between fumarate and glutathione. Chronic succination of GSH, caused by the loss of FH, or by exogenous fumarate, leads to persistent oxidative stress and cellular senescence in vitro and in vivo. Importantly, the ablation of p21, a key mediator of senescence, in Fh1-deficient mice resulted in the transformation of benign renal cysts into a hyperplastic lesion, suggesting that fumarate-induced senescence needs to be bypassed for the initiation of renal cancers.


Asunto(s)
Fumaratos/química , Glutatión/metabolismo , Animales , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica , Senescencia Celular , Cromatografía Liquida , Biología Computacional , Femenino , Fibroblastos/metabolismo , Fumarato Hidratasa/química , Glutamina/química , Inmunohistoquímica , Riñón/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Espectrometría de Masas , Metabolómica , Ratones , Ratones Endogámicos C57BL , Mutación , Oxidación-Reducción , Estrés Oxidativo , Transcriptoma
20.
Nature ; 515(7527): 431-435, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25383517

RESUMEN

Ischaemia-reperfusion injury occurs when the blood supply to an organ is disrupted and then restored, and underlies many disorders, notably heart attack and stroke. While reperfusion of ischaemic tissue is essential for survival, it also initiates oxidative damage, cell death and aberrant immune responses through the generation of mitochondrial reactive oxygen species (ROS). Although mitochondrial ROS production in ischaemia reperfusion is established, it has generally been considered a nonspecific response to reperfusion. Here we develop a comparative in vivo metabolomic analysis, and unexpectedly identify widely conserved metabolic pathways responsible for mitochondrial ROS production during ischaemia reperfusion. We show that selective accumulation of the citric acid cycle intermediate succinate is a universal metabolic signature of ischaemia in a range of tissues and is responsible for mitochondrial ROS production during reperfusion. Ischaemic succinate accumulation arises from reversal of succinate dehydrogenase, which in turn is driven by fumarate overflow from purine nucleotide breakdown and partial reversal of the malate/aspartate shuttle. After reperfusion, the accumulated succinate is rapidly re-oxidized by succinate dehydrogenase, driving extensive ROS generation by reverse electron transport at mitochondrial complex I. Decreasing ischaemic succinate accumulation by pharmacological inhibition is sufficient to ameliorate in vivo ischaemia-reperfusion injury in murine models of heart attack and stroke. Thus, we have identified a conserved metabolic response of tissues to ischaemia and reperfusion that unifies many hitherto unconnected aspects of ischaemia-reperfusion injury. Furthermore, these findings reveal a new pathway for metabolic control of ROS production in vivo, while demonstrating that inhibition of ischaemic succinate accumulation and its oxidation after subsequent reperfusion is a potential therapeutic target to decrease ischaemia-reperfusion injury in a range of pathologies.


Asunto(s)
Isquemia/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Ácido Succínico/metabolismo , Adenosina Monofosfato/metabolismo , Animales , Ácido Aspártico/metabolismo , Ciclo del Ácido Cítrico , Modelos Animales de Enfermedad , Transporte de Electrón , Complejo I de Transporte de Electrón/metabolismo , Fumaratos/metabolismo , Isquemia/enzimología , Malatos/metabolismo , Masculino , Metabolómica , Ratones , Mitocondrias/enzimología , Infarto del Miocardio/enzimología , Infarto del Miocardio/metabolismo , Miocardio/citología , Miocardio/enzimología , Miocardio/metabolismo , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , NAD/metabolismo , Daño por Reperfusión/enzimología , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/metabolismo , Succinato Deshidrogenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA