Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Br J Cancer ; 120(9): 957, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30867565

RESUMO

This article was originally published under a CC BY NC SA License, but has now been made available under a CC BY 4.0 License.

2.
Br J Cancer ; 121(1): 51-64, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31114017

RESUMO

BACKGROUND: Previous studies suggested that the metabolism is differently reprogrammed in the major subtypes of non-small cell lung cancer (NSCLC), squamous cell carcinomas (SCC) and adenocarcinomas (AdC). However, a comprehensive analysis of this differential metabolic reprogramming is lacking. METHODS: Publicly available gene expression data from human lung cancer samples and cell lines were analysed. Stable isotope resolved metabolomics were performed on SCC and ADC tumours in human patients and in freshly resected tumour slices. RESULTS: Analysis of multiple transcriptomics data from human samples identified a SCC-distinguishing enzyme gene signature. SCC tumours from patients infused with [U-13C]-glucose and SCC tissue slices incubated with stable isotope tracers demonstrated differential glucose and glutamine catabolism compared to AdCs or non-cancerous lung, confirming increased activity through pathways defined by the SCC metabolic gene signature. Furthermore, the upregulation of Notch target genes was a distinguishing feature of SCCs, which correlated with the metabolic signature. Notch and MYC-driven murine lung tumours recapitulated the SCC-distinguishing metabolic reprogramming. However, the differences between SCCs and AdCs disappear in established cell lines in 2D culture. CONCLUSIONS: Our data emphasise the importance of studying lung cancer metabolism in vivo. They also highlight potential targets for therapeutic intervention in SCC patients including differentially expressed enzymes that catalyse reactions in glycolysis, glutamine catabolism, serine, nucleotide and glutathione biosynthesis.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores Notch/fisiologia , Adenocarcinoma de Pulmão/metabolismo , Animais , Carcinoma de Células Escamosas/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-myc/fisiologia , Transcriptoma , Microambiente Tumoral
3.
Br J Cancer ; 116(11): 1375-1381, 2017 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-28441384

RESUMO

Altered cell metabolism enables tumours to sustain their increased energetic and biosynthetic needs. Although tumour metabolism has long been considered a promising discipline in the development of cancer therapeutics, the majority of work has focused on changes in glucose metabolism. However, the complexity of cellular metabolism means that very rarely is an individual metabolite required for a single purpose, and thus understanding the overall metabolic requirements of tumours is vital. Over the past 30 years, increasing evidence has shown that many tumours require glutamine as well as glucose for their proliferation and survival. In this minireview, we explore the complexity of glutamine metabolism in tumour cells, discussing how the overall context of the tumour dictates the requirement for glutamine and how this can affect the design of effective therapeutic strategies.


Assuntos
Glutamina/metabolismo , Redes e Vias Metabólicas , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Autofagia , Descoberta de Drogas , Glutamina/química , Glicosilação , Humanos , Serina-Treonina Quinases TOR/metabolismo , Microambiente Tumoral
4.
Nat Metab ; 5(8): 1303-1318, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37580540

RESUMO

The genomic landscape of colorectal cancer (CRC) is shaped by inactivating mutations in tumour suppressors such as APC, and oncogenic mutations such as mutant KRAS. Here we used genetically engineered mouse models, and multimodal mass spectrometry-based metabolomics to study the impact of common genetic drivers of CRC on the metabolic landscape of the intestine. We show that untargeted metabolic profiling can be applied to stratify intestinal tissues according to underlying genetic alterations, and use mass spectrometry imaging to identify tumour, stromal and normal adjacent tissues. By identifying ions that drive variation between normal and transformed tissues, we found dysregulation of the methionine cycle to be a hallmark of APC-deficient CRC. Loss of Apc in the mouse intestine was found to be sufficient to drive expression of one of its enzymes, adenosylhomocysteinase (AHCY), which was also found to be transcriptionally upregulated in human CRC. Targeting of AHCY function impaired growth of APC-deficient organoids in vitro, and prevented the characteristic hyperproliferative/crypt progenitor phenotype driven by acute deletion of Apc in vivo, even in the context of mutant Kras. Finally, pharmacological inhibition of AHCY reduced intestinal tumour burden in ApcMin/+ mice indicating its potential as a metabolic drug target in CRC.


Assuntos
Neoplasias Colorretais , Animais , Humanos , Camundongos , Adenosil-Homocisteinase/genética , Adenosil-Homocisteinase/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Metabolômica , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética
5.
J Clin Invest ; 125(2): 687-98, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25607840

RESUMO

Anabolic biosynthesis requires precursors supplied by the Krebs cycle, which in turn requires anaplerosis to replenish precursor intermediates. The major anaplerotic sources are pyruvate and glutamine, which require the activity of pyruvate carboxylase (PC) and glutaminase 1 (GLS1), respectively. Due to their rapid proliferation, cancer cells have increased anabolic and energy demands; however, different cancer cell types exhibit differential requirements for PC- and GLS-mediated pathways for anaplerosis and cell proliferation. Here, we infused patients with early-stage non-small-cell lung cancer (NSCLC) with uniformly 13C-labeled glucose before tissue resection and determined that the cancerous tissues in these patients had enhanced PC activity. Freshly resected paired lung tissue slices cultured in 13C6-glucose or 13C5,15N2-glutamine tracers confirmed selective activation of PC over GLS in NSCLC. Compared with noncancerous tissues, PC expression was greatly enhanced in cancerous tissues, whereas GLS1 expression showed no trend. Moreover, immunohistochemical analysis of paired lung tissues showed PC overexpression in cancer cells rather than in stromal cells of tumor tissues. PC knockdown induced multinucleation, decreased cell proliferation and colony formation in human NSCLC cells, and reduced tumor growth in a mouse xenograft model. Growth inhibition was accompanied by perturbed Krebs cycle activity, inhibition of lipid and nucleotide biosynthesis, and altered glutathione homeostasis. These findings indicate that PC-mediated anaplerosis in early-stage NSCLC is required for tumor survival and proliferation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/enzimologia , Proliferação de Células , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/enzimologia , Proteínas de Neoplasias/biossíntese , Piruvato Carboxilase/biossíntese , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Ciclo do Ácido Cítrico/genética , Feminino , Glucose/metabolismo , Glutationa/biossíntese , Glutationa/genética , Células HEK293 , Humanos , Metabolismo dos Lipídeos/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Proteínas de Neoplasias/genética , Nucleotídeos/biossíntese , Nucleotídeos/genética , Piruvato Carboxilase/genética , Traçadores Radioativos
6.
Cell Metab ; 15(2): 157-70, 2012 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-22326218

RESUMO

The altered metabolism of tumors has been considered a target for anticancer therapy. However, the relationship between distinct tumor-initiating lesions and anomalies of tumor metabolism in vivo has not been addressed. We report that MYC-induced mouse liver tumors significantly increase both glucose and glutamine catabolism, whereas MET-induced liver tumors use glucose to produce glutamine. Increased glutamine catabolism in MYC-induced liver tumors is associated with decreased levels of glutamine synthetase (Glul) and the switch from Gls2 to Gls1 glutaminase. In contrast to liver tumors, MYC-induced lung tumors display increased expression of both Glul and Gls1 and accumulate glutamine. We also show that inhibition of Gls1 kills cells that overexpress MYC and catabolize glutamine. Our results suggest that the metabolic profiles of tumors are likely to depend on both the genotype and tissue of origin and have implications regarding the design of therapies targeting tumor metabolism.


Assuntos
Transformação Celular Neoplásica/metabolismo , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Pulmonares/metabolismo , Metaboloma/fisiologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico/fisiologia , Primers do DNA/genética , Glucoquinase/metabolismo , Glucose/metabolismo , Glutamina/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Marcação por Isótopo , Ácido Láctico/metabolismo , Neoplasias Hepáticas Experimentais/etiologia , Neoplasias Pulmonares/etiologia , Metaboloma/genética , Camundongos , Ressonância Magnética Nuclear Biomolecular , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa