Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 1368, 2023 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-36914647

RESUMO

Inhibition of AMPK is tightly associated with metabolic perturbations upon over nutrition, yet the molecular mechanisms underlying are not clear. Here, we demonstrate the serine/threonine-protein phosphatase 6 regulatory subunit 3, SAPS3, is a negative regulator of AMPK. SAPS3 is induced under high fat diet (HFD) and recruits the PP6 catalytic subunit to deactivate phosphorylated-AMPK, thereby inhibiting AMPK-controlled metabolic pathways. Either whole-body or liver-specific deletion of SAPS3 protects male mice against HFD-induced detrimental consequences and reverses HFD-induced metabolic and transcriptional alterations while loss of SAPS3 has no effects on mice under balanced diets. Furthermore, genetic inhibition of AMPK is sufficient to block the protective phenotype in SAPS3 knockout mice under HFD. Together, our results reveal that SAPS3 is a negative regulator of AMPK and suppression of SAPS3 functions as a guardian when metabolism is perturbed and represents a potential therapeutic strategy to treat metabolic syndromes.


Assuntos
Proteínas Quinases Ativadas por AMP , Dieta Hiperlipídica , Fosfoproteínas Fosfatases , Animais , Masculino , Camundongos , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Dieta Hiperlipídica/efeitos adversos , Homeostase , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas Fosfatases/metabolismo
2.
J Biol Chem ; 299(4): 103039, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36803962

RESUMO

The small molecule erastin inhibits the cystine-glutamate antiporter, system xc-, which leads to intracellular cysteine and glutathione depletion. This can cause ferroptosis, which is an oxidative cell death process characterized by uncontrolled lipid peroxidation. Erastin and other ferroptosis inducers have been shown to affect metabolism but the metabolic effects of these drugs have not been systematically studied. To this end, we investigated how erastin impacts global metabolism in cultured cells and compared this metabolic profile to that caused by the ferroptosis inducer RAS-selective lethal 3 or in vivo cysteine deprivation. Common among the metabolic profiles were alterations in nucleotide and central carbon metabolism. Supplementing nucleosides to cysteine-deprived cells rescued cell proliferation in certain contexts, showing that these alterations to nucleotide metabolism can affect cellular fitness. While inhibition of the glutathione peroxidase GPX4 caused a similar metabolic profile as cysteine deprivation, nucleoside treatment did not rescue cell viability or proliferation under RAS-selective lethal 3 treatment, suggesting that these metabolic changes have varying importance in different scenarios of ferroptosis. Together, our study shows how global metabolism is affected during ferroptosis and points to nucleotide metabolism as an important target of cysteine deprivation.


Assuntos
Cisteína , Ferroptose , Nucleotídeos , Piperazinas , Morte Celular , Cisteína/metabolismo , Glutationa Peroxidase/metabolismo , Peroxidação de Lipídeos , Piperazinas/farmacologia , Nucleotídeos/metabolismo
4.
Nature ; 608(7921): 209-216, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35859173

RESUMO

Mechanistic target of rapamycin complex 1 (mTORC1) regulates cell growth and metabolism in response to multiple nutrients, including the essential amino acid leucine1. Recent work in cultured mammalian cells established the Sestrins as leucine-binding proteins that inhibit mTORC1 signalling during leucine deprivation2,3, but their role in the organismal response to dietary leucine remains elusive. Here we find that Sestrin-null flies (Sesn-/-) fail to inhibit mTORC1 or activate autophagy after acute leucine starvation and have impaired development and a shortened lifespan on a low-leucine diet. Knock-in flies expressing a leucine-binding-deficient Sestrin mutant (SesnL431E) have reduced, leucine-insensitive mTORC1 activity. Notably, we find that flies can discriminate between food with or without leucine, and preferentially feed and lay progeny on leucine-containing food. This preference depends on Sestrin and its capacity to bind leucine. Leucine regulates mTORC1 activity in glial cells, and knockdown of Sesn in these cells reduces the ability of flies to detect leucine-free food. Thus, nutrient sensing by mTORC1 is necessary for flies not only to adapt to, but also to detect, a diet deficient in an essential nutrient.


Assuntos
Adaptação Fisiológica , Dieta , Proteínas de Drosophila , Drosophila melanogaster , Leucina , Sestrinas , Adaptação Fisiológica/genética , Ração Animal , Animais , Autofagia , Dieta/veterinária , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Preferências Alimentares , Leucina/deficiência , Leucina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neuroglia/metabolismo , Sestrinas/deficiência , Sestrinas/genética , Sestrinas/metabolismo , Transdução de Sinais
5.
Nature ; 572(7769): 397-401, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31367041

RESUMO

Nutrition exerts considerable effects on health, and dietary interventions are commonly used to treat diseases of metabolic aetiology. Although cancer has a substantial metabolic component1, the principles that define whether nutrition may be used to influence outcomes of cancer are unclear2. Nevertheless, it is established that targeting metabolic pathways with pharmacological agents or radiation can sometimes lead to controlled therapeutic outcomes. By contrast, whether specific dietary interventions can influence the metabolic pathways that are targeted in standard cancer therapies is not known. Here we show that dietary restriction of the essential amino acid methionine-the reduction of which has anti-ageing and anti-obesogenic properties-influences cancer outcome, through controlled and reproducible changes to one-carbon metabolism. This pathway metabolizes methionine and is the target of a variety of cancer interventions that involve chemotherapy and radiation. Methionine restriction produced therapeutic responses in two patient-derived xenograft models of chemotherapy-resistant RAS-driven colorectal cancer, and in a mouse model of autochthonous soft-tissue sarcoma driven by a G12D mutation in KRAS and knockout of p53 (KrasG12D/+;Trp53-/-) that is resistant to radiation. Metabolomics revealed that the therapeutic mechanisms operate via tumour-cell-autonomous effects on flux through one-carbon metabolism that affects redox and nucleotide metabolism-and thus interact with the antimetabolite or radiation intervention. In a controlled and tolerated feeding study in humans, methionine restriction resulted in effects on systemic metabolism that were similar to those obtained in mice. These findings provide evidence that a targeted dietary manipulation can specifically affect tumour-cell metabolism to mediate broad aspects of cancer outcome.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Modelos Animais de Doenças , Metabolômica , Metionina/administração & dosagem , Metionina/farmacologia , Sarcoma/tratamento farmacológico , Neoplasias de Tecidos Moles/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Dieta , Feminino , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Genes p53 , Genes ras , Voluntários Saudáveis , Humanos , Masculino , Metionina/metabolismo , Camundongos , Pessoa de Meia-Idade , Mutação , Estudo de Prova de Conceito , Sarcoma/genética , Sarcoma/metabolismo , Neoplasias de Tecidos Moles/genética , Neoplasias de Tecidos Moles/metabolismo , Enxofre/metabolismo , Resultado do Tratamento
6.
Cancer Cell ; 35(3): 339-341, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30889375

RESUMO

Altered metabolism is a common feature of new and recurring malignancy. In this issue of Cancer Cell, Reina-Campos and colleagues report upregulation of the serine, glycine, one-carbon (SGOC) metabolic network is required for neuroendocrine prostate cancer, a castration-resistant aggressive form of the disease, and presents a targetable vulnerability.


Assuntos
Neoplasias da Próstata , Serina , Carbono , Humanos , Masculino , Metionina , Recidiva Local de Neoplasia
7.
Cell Rep ; 26(6): 1544-1556.e8, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30726737

RESUMO

The tripeptide glutathione suppresses the iron-dependent, non-apoptotic cell death process of ferroptosis. How glutathione abundance is regulated in the cell and how this regulation alters ferroptosis sensitivity is poorly understood. Using genome-wide human haploid genetic screening technology coupled to fluorescence-activated cell sorting (FACS), we directly identify genes that regulate intracellular glutathione abundance and characterize their role in ferroptosis regulation. Disruption of the ATP binding cassette (ABC)-family transporter multidrug resistance protein 1 (MRP1) prevents glutathione efflux from the cell and strongly inhibits ferroptosis. High levels of MRP1 expression decrease sensitivity to certain pro-apoptotic chemotherapeutic drugs, while collaterally sensitizing to all tested pro-ferroptotic agents. By contrast, disruption of KEAP1 and NAA38, leading to the stabilization of the transcription factor NRF2, increases glutathione levels but only weakly protects from ferroptosis. This is due in part to concomitant NRF2-mediated upregulation of MRP1. These results pinpoint glutathione efflux as an unanticipated regulator of ferroptosis sensitivity.


Assuntos
Ferroptose/genética , Citometria de Fluxo/métodos , Glutationa/metabolismo , Haploidia , Linhagem Celular Tumoral , Feminino , Genoma Humano , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Masculino , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Acetiltransferase N-Terminal C/genética , Acetiltransferase N-Terminal C/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/genética , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo
8.
Nat Commun ; 10(1): 809, 2019 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-30778058

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human cancers. It thrives in a nutrient-poor environment; however, the mechanisms by which PDAC cells undergo metabolic reprogramming to adapt to metabolic stress are still poorly understood. Here, we show that microRNA-135 is significantly increased in PDAC patient samples compared to adjacent normal tissue. Mechanistically, miR-135 accumulates specifically in response to glutamine deprivation and requires ROS-dependent activation of mutant p53, which directly promotes miR-135 expression. Functionally, we found miR-135 targets phosphofructokinase-1 (PFK1) and inhibits aerobic glycolysis, thereby promoting the utilization of glucose to support the tricarboxylic acid (TCA) cycle. Consistently, miR-135 silencing sensitizes PDAC cells to glutamine deprivation and represses tumor growth in vivo. Together, these results identify a mechanism used by PDAC cells to survive the nutrient-poor tumor microenvironment, and also provide insight regarding the role of mutant p53 and miRNA in pancreatic cancer cell adaptation to metabolic stresses.


Assuntos
Carcinoma Ductal Pancreático/genética , Glicólise/genética , MicroRNAs/genética , Neoplasias Pancreáticas/genética , Fosfofrutoquinase-1 Tipo C/genética , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Regulação Neoplásica da Expressão Gênica , Glutamina/genética , Glutamina/metabolismo , Humanos , Masculino , Camundongos Nus , MicroRNAs/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fosfofrutoquinase-1 Tipo C/metabolismo , Estresse Fisiológico/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nat Commun ; 9(1): 5442, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30575741

RESUMO

Phosphoglycerate dehydrogenase (PHGDH) catalyzes the committed step in de novo serine biosynthesis. Paradoxically, PHGDH and serine synthesis are required in the presence of abundant environmental serine even when serine uptake exceeds the requirements for nucleotide synthesis. Here, we establish a mechanism for how PHGDH maintains nucleotide metabolism. We show that inhibition of PHGDH induces alterations in nucleotide metabolism independent of serine utilization. These changes are not attributable to defects in serine-derived nucleotide synthesis and redox maintenance, another key aspect of serine metabolism, but result from disruption of mass balance within central carbon metabolism. Mechanistically, this leads to simultaneous alterations in both the pentose phosphate pathway and the tri-carboxylic acid cycle, as we demonstrate based on a quantitative model. These findings define a mechanism whereby disruption of one metabolic pathway induces toxicity by simultaneously affecting the activity of multiple related pathways.


Assuntos
Ciclo do Ácido Cítrico , Nucleotídeos/biossíntese , Via de Pentose Fosfato , Fosfoglicerato Desidrogenase/metabolismo , Células HCT116 , Humanos , Células MCF-7 , Análise do Fluxo Metabólico , Serina/biossíntese
10.
Oncogenesis ; 7(11): 93, 2018 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-30478303

RESUMO

One of the hallmarks of cancer is the ability to reprogram cellular metabolism to increase the uptake of necessary nutrients such as glucose and glutamine. Driven by oncogenes, cancer cells have increased glutamine uptake to support their highly proliferative nature. However, as cancer cells continue to replicate and grow, they lose access to vascular tissues and deplete local supply of nutrients and oxygen. We previously showed that many tumor cells situate in a low glutamine microenvironment in vivo, yet the mechanisms of how they are able to adapt to this metabolic stress are still not fully understood. Here, we report that IκB-kinase ß (IKKß) is needed to promote survival and its activation is accompanied by phosphorylation of the metabolic sensor, p53, in response to glutamine deprivation. Knockdown of IKKß decreases the level of wild-type and mutant p53 phosphorylation and its transcriptional activity, indicating a novel relationship between IKKß and p53 in mediating cancer cell survival in response to glutamine withdrawal. Phosphopeptide mass spectrometry analysis further reveals that IKKß phosphorylates p53 on Ser392 to facilitate its activation upon glutamine deprivation, independent of the NF-κB pathway. The results of this study offer an insight into the metabolic reprogramming in cancer cells that is dependent on a previously unidentified IKKß-p53 signaling axis in response to glutamine depletion. More importantly, this study highlights a new therapeutic strategy for cancer treatment and advances our understanding of adaptive mechanisms that could lead to resistance to current glutamine targeting therapies.

11.
Cell ; 175(2): 502-513.e13, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30245009

RESUMO

Acetate is a major nutrient that supports acetyl-coenzyme A (Ac-CoA) metabolism and thus lipogenesis and protein acetylation. However, its source is unclear. Here, we report that pyruvate, the end product of glycolysis and key node in central carbon metabolism, quantitatively generates acetate in mammals. This phenomenon becomes more pronounced in the context of nutritional excess, such as during hyperactive glucose metabolism. Conversion of pyruvate to acetate occurs through two mechanisms: (1) coupling to reactive oxygen species (ROS) and (2) neomorphic enzyme activity from keto acid dehydrogenases that enable function as pyruvate decarboxylases. Further, we demonstrate that de novo acetate production sustains Ac-CoA pools and cell proliferation in limited metabolic environments, such as during mitochondrial dysfunction or ATP citrate lyase (ACLY) deficiency. By virtue of de novo acetate production being coupled to mitochondrial metabolism, there are numerous possible regulatory mechanisms and links to pathophysiology.


Assuntos
Acetatos/metabolismo , Glucose/metabolismo , Ácido Pirúvico/metabolismo , ATP Citrato (pro-S)-Liase/fisiologia , Acetilcoenzima A/biossíntese , Acetilcoenzima A/metabolismo , Acetilação , Animais , Feminino , Glicólise/fisiologia , Lipogênese/fisiologia , Masculino , Mamíferos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Oxirredutases , Piruvato Descarboxilase/fisiologia , Espécies Reativas de Oxigênio/metabolismo
12.
Cell Rep ; 22(13): 3507-3520, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590619

RESUMO

Cell proliferation can be dependent on the non-essential amino acid serine, and dietary restriction of serine inhibits tumor growth, but the underlying mechanisms remain incompletely understood. Using a metabolomics approach, we found that serine deprivation most predominantly impacts cellular acylcarnitine levels, a signature of altered mitochondrial function. Fuel utilization from fatty acid, glucose, and glutamine is affected by serine deprivation, as are mitochondrial morphological dynamics leading to increased fragmentation. Interestingly, these changes can occur independently of nucleotide and redox metabolism, two known major functions of serine. A lipidomics analysis revealed an overall decrease in ceramide levels. Importantly, supplementation of the lipid component of bovine serum or C16:0-ceramide could partially restore defects in cell proliferation and mitochondrial fragmentation induced by serine deprivation. Together, these data define a role for serine in supporting mitochondrial function and cell proliferation through ceramide metabolism.


Assuntos
Metabolismo dos Lipídeos/fisiologia , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/fisiologia , Serina/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Ciclo do Ácido Cítrico , Ácidos Graxos/metabolismo , Glucose/metabolismo , Glutamina/metabolismo , Glicerofosfolipídeos/metabolismo , Células HCT116 , Células HT29 , Humanos , Nucleotídeos/metabolismo , Oxirredução , Serina/deficiência , Esfingolipídeos/metabolismo
13.
PLoS Biol ; 15(11): e2002810, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29107960

RESUMO

Driven by oncogenic signaling, glutamine addiction exhibited by cancer cells often leads to severe glutamine depletion in solid tumors. Despite this nutritional environment that tumor cells often experience, the effect of glutamine deficiency on cellular responses to DNA damage and chemotherapeutic treatment remains unclear. Here, we show that glutamine deficiency, through the reduction of alpha-ketoglutarate, inhibits the AlkB homolog (ALKBH) enzymes activity and induces DNA alkylation damage. As a result, glutamine deprivation or glutaminase inhibitor treatment triggers DNA damage accumulation independent of cell death. In addition, low glutamine-induced DNA damage is abolished in ALKBH deficient cells. Importantly, we show that glutaminase inhibitors, 6-Diazo-5-oxo-L-norleucine (DON) or CB-839, hypersensitize cancer cells to alkylating agents both in vitro and in vivo. Together, the crosstalk between glutamine metabolism and the DNA repair pathway identified in this study highlights a potential role of metabolic stress in genomic instability and therapeutic response in cancer.


Assuntos
Enzimas AlkB/antagonistas & inibidores , Antineoplásicos Alquilantes/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Glutaminase/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Enzimas AlkB/genética , Enzimas AlkB/metabolismo , Homólogo AlkB 3 da Dioxigenase Dependente de alfa-Cetoglutarato/antagonistas & inibidores , Homólogo AlkB 3 da Dioxigenase Dependente de alfa-Cetoglutarato/genética , Homólogo AlkB 3 da Dioxigenase Dependente de alfa-Cetoglutarato/metabolismo , Alquilação/efeitos dos fármacos , Animais , Antineoplásicos Alquilantes/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Inibidores Enzimáticos/farmacologia , Glutaminase/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Interferência de RNA , Distribuição Aleatória , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cell ; 171(4): 736-737, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29100069

RESUMO

Nearly 3% of the human population carries bi-allelic loss-of-function variants in the gene encoding CLYBL. While largely healthy, these individuals exhibit reduced circulating vitamin B12 levels. In this issue of Cell, Shen and colleagues uncover the metabolic role of CLYBL, linking its function to B12 metabolism and the immunomodulatory metabolite, itaconate.


Assuntos
Succinatos , Vitamina B 12 , Técnicas de Inativação de Genes , Humanos , Vitaminas
15.
Nat Cell Biol ; 19(11): 1298-1306, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29058720

RESUMO

The substrates used to modify nucleic acids and chromatin are affected by nutrient availability and the activity of metabolic pathways. Thus, cellular metabolism constitutes a fundamental component of chromatin status and thereby of genome regulation. Here we describe the biochemical and genetic principles of how metabolism can influence chromatin biology and epigenetics, discuss the functional roles of this interplay in developmental and cancer biology, and present future directions in this rapidly emerging area.


Assuntos
Cromatina/genética , Epigênese Genética/genética , Redes e Vias Metabólicas/genética , Animais , Epigenômica/métodos , Humanos , Neoplasias/genética
16.
Mol Aspects Med ; 54: 50-57, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27620316

RESUMO

Cancer cells have epigenetic alterations that are known to drive cancer progression. The reversibility of the epigenetic posttranslational modifications on chromatin and DNA renders targeting these modifications an attractive means for cancer therapy. Cellular epigenetic status interacts with cell metabolism, and we are now beginning to understand the nature of how this interaction occurs and the biological contexts that mediate its function. Given the tremendous interest in understanding and targeting metabolic reprogramming in cancer, this nexus also provides opportunities for exploring the liabilities of cancers. This review summarizes recent developments in our understanding of the interaction of cancer metabolism and epigenetics.


Assuntos
Epigênese Genética , Neoplasias/genética , Animais , Metilação de DNA , Histonas/metabolismo , Humanos , Metaboloma , Modelos Biológicos
17.
Nat Cell Biol ; 18(10): 1090-101, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27617932

RESUMO

Poorly organized tumour vasculature often results in areas of limited nutrient supply and hypoxia. Despite our understanding of solid tumour responses to hypoxia, how nutrient deprivation regionally affects tumour growth and therapeutic response is poorly understood. Here, we show that the core region of solid tumours displayed glutamine deficiency compared with other amino acids. Low glutamine in tumour core regions led to dramatic histone hypermethylation due to decreased α-ketoglutarate levels, a key cofactor for the Jumonji-domain-containing histone demethylases. Using patient-derived (V600E)BRAF melanoma cells, we found that low-glutamine-induced histone hypermethylation resulted in cancer cell dedifferentiation and resistance to BRAF inhibitor treatment, which was largely mediated by methylation on H3K27, as knockdown of the H3K27-specific demethylase KDM6B and the methyltransferase EZH2 respectively reproduced and attenuated the low-glutamine effects in vitro and in vivo. Thus, intratumoral regional variation in the nutritional microenvironment contributes to tumour heterogeneity and therapeutic response.


Assuntos
Metilação de DNA/fisiologia , Histona Desmetilases/metabolismo , Histonas/metabolismo , Metiltransferases/metabolismo , Neoplasias/metabolismo , Animais , Glutamina/deficiência , Glutamina/metabolismo , Histonas/genética , Humanos , Ácidos Cetoglutáricos/metabolismo , Metilação
18.
Genes Dev ; 30(16): 1837-51, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27585591

RESUMO

Glutamine is an essential nutrient for cancer cell survival and proliferation. Enhanced utilization of glutamine often depletes its local supply, yet how cancer cells adapt to low glutamine conditions is largely unknown. Here, we report that IκB kinase ß (IKKß) is activated upon glutamine deprivation and is required for cell survival independently of NF-κB transcription. We demonstrate that IKKß directly interacts with and phosphorylates 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase isoform 3 (PFKFB3), a major driver of aerobic glycolysis, at Ser269 upon glutamine deprivation to inhibit its activity, thereby down-regulating aerobic glycolysis when glutamine levels are low. Thus, due to lack of inhibition of PFKFB3, IKKß-deficient cells exhibit elevated aerobic glycolysis and lactate production, leading to less glucose carbons contributing to tricarboxylic acid (TCA) cycle intermediates and the pentose phosphate pathway, which results in increased glutamine dependence for both TCA cycle intermediates and reactive oxygen species suppression. Therefore, coinhibition of IKKß and glutamine metabolism results in dramatic synergistic killing of cancer cells both in vitro and in vivo. In all, our results uncover a previously unidentified role of IKKß in regulating glycolysis, sensing low-glutamine-induced metabolic stress, and promoting cellular adaptation to nutrient availability.


Assuntos
Glutamina/metabolismo , Quinase I-kappa B/metabolismo , Fosfofrutoquinase-2/metabolismo , Adaptação Fisiológica/genética , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Técnicas de Silenciamento de Genes , Glicólise/genética , Células HEK293 , Células HeLa , Humanos , Quinase I-kappa B/genética , Células MCF-7 , Camundongos , NF-kappa B/metabolismo , Fosforilação
19.
Sci Rep ; 6: 21520, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26861754

RESUMO

Changes in cellular oxygen tension play important roles in physiological processes including development and pathological processes such as tumor promotion. The cellular adaptations to sustained hypoxia are mediated by hypoxia-inducible factors (HIFs) to regulate downstream target gene expression. With hypoxia, the stabilized HIF-α and aryl hydrocarbon receptor nuclear translocator (ARNT, also known as HIF-ß) heterodimer bind to hypoxia response elements (HREs) and regulate expression of target genes. Here, we report that WNT11 is induced by hypoxia in many cell types, and that transcription of WNT11 is regulated primarily by HIF-1α. We observed induced WNT11 expression in the hypoxic area of allograft tumors. In addition, in mice bearing orthotopic malignant gliomas, inhibition with bevacizumab of vascular endothelial growth factor, which is an important stimulus for angiogenesis, increased nuclear HIF-1α and HIF-2α, and expression of WNT11. Gain- and loss-of-function approaches revealed that WNT11 stimulates proliferation, migration and invasion of cancer-derived cells, and increases activity of matrix metalloproteinase (MMP)-2 and 9. Since tumor hypoxia has been proposed to increase tumor aggressiveness, these data suggest WNT11 as a possible target for cancer therapies, especially for tumors treated with antiangiogenic therapy.


Assuntos
Hipóxia Celular/fisiologia , Movimento Celular/genética , Proliferação de Células/genética , Glioma/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Invasividade Neoplásica/genética , Proteínas Wnt/biossíntese , Inibidores da Angiogênese/farmacologia , Animais , Translocador Nuclear Receptor Aril Hidrocarboneto/biossíntese , Bevacizumab/farmacologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/tratamento farmacológico , Oxigênio/metabolismo , Biossíntese de Proteínas/genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteínas Wnt/genética
20.
PLoS One ; 10(12): e0145938, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26717153

RESUMO

Despite advances in our understanding of protein kinase regulation in the DNA damage response, the mechanism that controls protein phosphatase activity in this pathway is unclear. Unlike kinases, the activity and specificity of serine/threonine phosphatases is governed largely by their associated proteins. Here we show that Tip41-like protein (TIPRL), an evolutionarily conserved binding protein for PP2A-family phosphatases, is a negative regulator of protein phosphatase 4 (PP4). Knockdown of TIPRL resulted in increased PP4 phosphatase activity and formation of the active PP4-C/PP4R2 complex known to dephosphorylate γ-H2AX. Thus, overexpression of TIPRL promotes phosphorylation of H2AX, and increases γ-H2AX positive foci in response to DNA damage, whereas knockdown of TIPRL inhibits γ-H2AX phosphorylation. In correlation with γ-H2AX levels, we found that TIPRL overexpression promotes cell death in response to genotoxic stress, and knockdown of TIPRL protects cells from genotoxic agents. Taken together, these data demonstrate that TIPRL inhibits PP4 activity to allow for H2AX phosphorylation and the subsequent DNA damage response.


Assuntos
Dano ao DNA , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfoproteínas Fosfatases/antagonistas & inibidores , Células 3T3 , Animais , Morte Celular , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Fosfoproteínas Fosfatases/metabolismo , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...