Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Cell ; 169(2): 258-272.e17, 2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28388410

RESUMO

A complex interplay of environmental factors impacts the metabolism of human cells, but neither traditional culture media nor mouse plasma mimic the metabolite composition of human plasma. Here, we developed a culture medium with polar metabolite concentrations comparable to those of human plasma (human plasma-like medium [HPLM]). Culture in HPLM, relative to that in traditional media, had widespread effects on cellular metabolism, including on the metabolome, redox state, and glucose utilization. Among the most prominent was an inhibition of de novo pyrimidine synthesis-an effect traced to uric acid, which is 10-fold higher in the blood of humans than of mice and other non-primates. We find that uric acid directly inhibits uridine monophosphate synthase (UMPS) and consequently reduces the sensitivity of cancer cells to the chemotherapeutic agent 5-fluorouracil. Thus, media that better recapitulates the composition of human plasma reveals unforeseen metabolic wiring and regulation, suggesting that HPLM should be of broad utility.


Assuntos
Meios de Cultura/química , Complexos Multienzimáticos/antagonistas & inibidores , Orotato Fosforribosiltransferase/antagonistas & inibidores , Orotidina-5'-Fosfato Descarboxilase/antagonistas & inibidores , Ácido Úrico/metabolismo , Idoso , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Fluoruracila/farmacologia , Glucose/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Complexos Multienzimáticos/química , Orotato Fosforribosiltransferase/química , Orotidina-5'-Fosfato Descarboxilase/química , Domínios Proteicos , Pirimidinas/biossíntese
2.
Cell ; 171(3): 642-654.e12, 2017 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-29053970

RESUMO

The mTORC1 kinase is a master growth regulator that senses many environmental cues, including amino acids. Activation of mTORC1 by arginine requires SLC38A9, a poorly understood lysosomal membrane protein with homology to amino acid transporters. Here, we validate that SLC38A9 is an arginine sensor for the mTORC1 pathway, and we uncover an unexpectedly central role for SLC38A9 in amino acid homeostasis. SLC38A9 mediates the transport, in an arginine-regulated fashion, of many essential amino acids out of lysosomes, including leucine, which mTORC1 senses through the cytosolic Sestrin proteins. SLC38A9 is necessary for leucine generated via lysosomal proteolysis to exit lysosomes and activate mTORC1. Pancreatic cancer cells, which use macropinocytosed protein as a nutrient source, require SLC38A9 to form tumors. Thus, through SLC38A9, arginine serves as a lysosomal messenger that couples mTORC1 activation to the release from lysosomes of the essential amino acids needed to drive cell growth.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Aminoácidos Essenciais/metabolismo , Lisossomos/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/genética , Animais , Arginina/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Alinhamento de Sequência
3.
Cell ; 166(5): 1324-1337.e11, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27565352

RESUMO

Mitochondria house metabolic pathways that impact most aspects of cellular physiology. While metabolite profiling by mass spectrometry is widely applied at the whole-cell level, it is not routinely possible to measure the concentrations of small molecules in mammalian organelles. We describe a method for the rapid and specific isolation of mitochondria and use it in tandem with a database of predicted mitochondrial metabolites ("MITObolome") to measure the matrix concentrations of more than 100 metabolites across various states of respiratory chain (RC) function. Disruption of the RC reveals extensive compartmentalization of mitochondrial metabolism and signatures unique to the inhibition of each RC complex. Pyruvate enables the proliferation of RC-deficient cells but has surprisingly limited effects on matrix contents. Interestingly, despite failing to restore matrix NADH/NAD balance, pyruvate does increase aspartate, likely through the exchange of matrix glutamate for cytosolic aspartate. We demonstrate the value of mitochondrial metabolite profiling and describe a strategy applicable to other organelles.


Assuntos
Redes e Vias Metabólicas , Metaboloma , Mitocôndrias/metabolismo , Transporte de Elétrons/genética , Células HeLa , Humanos , Ácido Pirúvico/metabolismo , Ácido Pirúvico/farmacologia
4.
Cell ; 162(3): 540-51, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26232224

RESUMO

The mitochondrial electron transport chain (ETC) enables many metabolic processes, but why its inhibition suppresses cell proliferation is unclear. It is also not well understood why pyruvate supplementation allows cells lacking ETC function to proliferate. We used a CRISPR-based genetic screen to identify genes whose loss sensitizes human cells to phenformin, a complex I inhibitor. The screen yielded GOT1, the cytosolic aspartate aminotransferase, loss of which kills cells upon ETC inhibition. GOT1 normally consumes aspartate to transfer electrons into mitochondria, but, upon ETC inhibition, it reverses to generate aspartate in the cytosol, which partially compensates for the loss of mitochondrial aspartate synthesis. Pyruvate stimulates aspartate synthesis in a GOT1-dependent fashion, which is required for pyruvate to rescue proliferation of cells with ETC dysfunction. Aspartate supplementation or overexpression of an aspartate transporter allows cells without ETC activity to proliferate. Thus, enabling aspartate synthesis is an essential role of the ETC in cell proliferation.


Assuntos
Ácido Aspártico/biossíntese , Proliferação de Células , Transporte de Elétrons , Mitocôndrias/metabolismo , Aspartato Aminotransferase Citoplasmática/metabolismo , Ácido Aspártico/metabolismo , DNA Mitocondrial/genética , Humanos , Células Jurkat , Mutação , Fenformin/farmacologia , Ácido Pirúvico/metabolismo
5.
Cell ; 162(3): 552-63, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-26232225

RESUMO

Mitochondrial respiration is important for cell proliferation; however, the specific metabolic requirements fulfilled by respiration to support proliferation have not been defined. Here, we show that a major role of respiration in proliferating cells is to provide electron acceptors for aspartate synthesis. This finding is consistent with the observation that cells lacking a functional respiratory chain are auxotrophic for pyruvate, which serves as an exogenous electron acceptor. Further, the pyruvate requirement can be fulfilled with an alternative electron acceptor, alpha-ketobutyrate, which provides cells neither carbon nor ATP. Alpha-ketobutyrate restores proliferation when respiration is inhibited, suggesting that an alternative electron acceptor can substitute for respiration to support proliferation. We find that electron acceptors are limiting for producing aspartate, and supplying aspartate enables proliferation of respiration deficient cells in the absence of exogenous electron acceptors. Together, these data argue a major function of respiration in proliferating cells is to support aspartate synthesis.


Assuntos
Ácido Aspártico/biossíntese , Proliferação de Células , Respiração Celular , Trifosfato de Adenosina/metabolismo , Butiratos/metabolismo , Linhagem Celular Tumoral , Elétrons , Humanos , Mitocôndrias/metabolismo , Nucleotídeos/biossíntese , Ácido Pirúvico
6.
Cell ; 158(5): 1094-1109, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25171410

RESUMO

It is increasingly appreciated that oncogenic transformation alters cellular metabolism to facilitate cell proliferation, but less is known about the metabolic changes that promote cancer cell aggressiveness. Here, we analyzed metabolic gene expression in cancer cell lines and found that a set of high-grade carcinoma lines expressing mesenchymal markers share a unique 44 gene signature, designated the "mesenchymal metabolic signature" (MMS). A FACS-based shRNA screen identified several MMS genes as essential for the epithelial-mesenchymal transition (EMT), but not for cell proliferation. Dihydropyrimidine dehydrogenase (DPYD), a pyrimidine-degrading enzyme, was highly expressed upon EMT induction and was necessary for cells to acquire mesenchymal characteristics in vitro and for tumorigenic cells to extravasate into the mouse lung. This role of DPYD was mediated through its catalytic activity and enzymatic products, the dihydropyrimidines. Thus, we identify metabolic processes essential for the EMT, a program associated with the acquisition of metastatic and aggressive cancer cell traits.


Assuntos
Transição Epitelial-Mesenquimal , Pirimidinas/metabolismo , Animais , Carcinoma/metabolismo , Linhagem Celular Tumoral , Di-Hidrouracila Desidrogenase (NADP)/genética , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , RNA Interferente Pequeno/metabolismo
7.
Immunity ; 47(4): 664-679.e6, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29030115

RESUMO

Store-operated Ca2+ entry (SOCE) is the main Ca2+ influx pathway in lymphocytes and is essential for T cell function and adaptive immunity. SOCE is mediated by Ca2+ release-activated Ca2+ (CRAC) channels that are activated by stromal interaction molecule (STIM) 1 and STIM2. SOCE regulates many Ca2+-dependent signaling molecules, including calcineurin, and inhibition of SOCE or calcineurin impairs antigen-dependent T cell proliferation. We here report that SOCE and calcineurin regulate cell cycle entry of quiescent T cells by controlling glycolysis and oxidative phosphorylation. SOCE directs the metabolic reprogramming of naive T cells by regulating the expression of glucose transporters, glycolytic enzymes, and metabolic regulators through the activation of nuclear factor of activated T cells (NFAT) and the PI3K-AKT kinase-mTOR nutrient-sensing pathway. We propose that SOCE controls a critical "metabolic checkpoint" at which T cells assess adequate nutrient supply to support clonal expansion and adaptive immune responses.


Assuntos
Canais de Cálcio/imunologia , Sinalização do Cálcio/imunologia , Cálcio/imunologia , Linfócitos T/imunologia , Animais , Calcineurina/imunologia , Calcineurina/metabolismo , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Divisão Celular/imunologia , Células Cultivadas , Feminino , Glicólise/imunologia , Células HEK293 , Humanos , Immunoblotting , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/imunologia , Fatores de Transcrição NFATC/metabolismo , Fosfatidilinositol 3-Quinases/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/imunologia , Molécula 1 de Interação Estromal/metabolismo , Molécula 2 de Interação Estromal/genética , Molécula 2 de Interação Estromal/imunologia , Molécula 2 de Interação Estromal/metabolismo , Linfócitos T/metabolismo
8.
Nature ; 567(7746): 118-122, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30760928

RESUMO

Cholesterol is essential for cells to grow and proliferate. Normal mammalian cells meet their need for cholesterol through its uptake or de novo synthesis1, but the extent to which cancer cells rely on each of these pathways remains poorly understood. Here, using a competitive proliferation assay on a pooled collection of DNA-barcoded cell lines, we identify a subset of cancer cells that is auxotrophic for cholesterol and thus highly dependent on its uptake. Through metabolic gene expression analysis, we pinpoint the loss of squalene monooxygenase expression as a cause of cholesterol auxotrophy, particularly in ALK+ anaplastic large cell lymphoma (ALCL) cell lines and primary tumours. Squalene monooxygenase catalyses the oxidation of squalene to 2,3-oxidosqualene in the cholesterol synthesis pathway and its loss results in accumulation of the upstream metabolite squalene, which is normally undetectable. In ALK+ ALCLs, squalene alters the cellular lipid profile and protects cancer cells from ferroptotic cell death, providing a growth advantage under conditions of oxidative stress and in tumour xenografts. Finally, a CRISPR-based genetic screen identified cholesterol uptake by the low-density lipoprotein receptor as essential for the growth of ALCL cells in culture and as patient-derived xenografts. This work reveals that the cholesterol auxotrophy of ALCLs is a targetable liability and, more broadly, that systematic approaches can be used to identify nutrient dependencies unique to individual cancer types.


Assuntos
Apoptose , Colesterol/metabolismo , Linfoma Anaplásico de Células Grandes/metabolismo , Linfoma Anaplásico de Células Grandes/patologia , Estresse Oxidativo , Esqualeno/metabolismo , Idoso , Animais , Linhagem Celular Tumoral , Proliferação de Células , Colesterol/biossíntese , Código de Barras de DNA Taxonômico , Farnesil-Difosfato Farnesiltransferase/genética , Farnesil-Difosfato Farnesiltransferase/metabolismo , Feminino , Humanos , Ferro/metabolismo , Linfoma Anaplásico de Células Grandes/enzimologia , Masculino , Lipídeos de Membrana/química , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Receptores de LDL/genética , Receptores de LDL/metabolismo , Esqualeno Mono-Oxigenase/genética , Esqualeno Mono-Oxigenase/metabolismo , Adulto Jovem
9.
Nature ; 559(7715): 632-636, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29995852

RESUMO

The chemotherapeutic drug methotrexate inhibits the enzyme dihydrofolate reductase1, which generates tetrahydrofolate, an essential cofactor in nucleotide synthesis2. Depletion of tetrahydrofolate causes cell death by suppressing DNA and RNA production3. Although methotrexate is widely used as an anticancer agent and is the subject of over a thousand ongoing clinical trials4, its high toxicity often leads to the premature termination of its use, which reduces its potential efficacy5. To identify genes that modulate the response of cancer cells to methotrexate, we performed a CRISPR-Cas9-based screen6,7. This screen yielded FTCD, which encodes an enzyme-formimidoyltransferase cyclodeaminase-that is required for the catabolism of the amino acid histidine8, a process that has not previously been linked to methotrexate sensitivity. In cultured cancer cells, depletion of several genes in the histidine degradation pathway markedly decreased sensitivity to methotrexate. Mechanistically, histidine catabolism drains the cellular pool of tetrahydrofolate, which is particularly detrimental to methotrexate-treated cells. Moreover, expression of the rate-limiting enzyme in histidine catabolism is associated with methotrexate sensitivity in cancer cell lines and with survival rate in patients. In vivo dietary supplementation of histidine increased flux through the histidine degradation pathway and enhanced the sensitivity of leukaemia xenografts to methotrexate. The histidine degradation pathway markedly influences the sensitivity of cancer cells to methotrexate and may be exploited to improve methotrexate efficacy through a simple dietary intervention.


Assuntos
Histidina/metabolismo , Metotrexato/farmacologia , Metotrexato/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Amônia-Liases/deficiência , Amônia-Liases/genética , Amônia-Liases/metabolismo , Animais , Sistemas CRISPR-Cas/genética , Linhagem Celular Tumoral , Feminino , Antagonistas do Ácido Fólico/farmacologia , Antagonistas do Ácido Fólico/uso terapêutico , Glutamato Formimidoiltransferase/deficiência , Glutamato Formimidoiltransferase/genética , Glutamato Formimidoiltransferase/metabolismo , Histidina/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Enzimas Multifuncionais , Nucleotídeos/biossíntese , Proteína Carregadora de Folato Reduzido/genética , Proteína Carregadora de Folato Reduzido/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Tetra-Hidrofolatos/deficiência , Tetra-Hidrofolatos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nature ; 543(7647): 681-686, 2017 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-28329758

RESUMO

Post-mitotic, differentiated cells exhibit a variety of characteristics that contrast with those of actively growing neoplastic cells, such as the expression of cell-cycle inhibitors and differentiation factors. We hypothesized that the gene expression profiles of these differentiated cells could reveal the identities of genes that may function as tumour suppressors. Here we show, using in vitro and in vivo studies in mice and humans, that the mitochondrial protein LACTB potently inhibits the proliferation of breast cancer cells. Its mechanism of action involves alteration of mitochondrial lipid metabolism and differentiation of breast cancer cells. This is achieved, at least in part, through reduction of the levels of mitochondrial phosphatidylserine decarboxylase, which is involved in the synthesis of mitochondrial phosphatidylethanolamine. These observations uncover a novel mitochondrial tumour suppressor and demonstrate a connection between mitochondrial lipid metabolism and the differentiation program of breast cancer cells, thereby revealing a previously undescribed mechanism of tumour suppression.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Diferenciação Celular , Metabolismo dos Lipídeos , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Supressoras de Tumor/metabolismo , beta-Lactamases/metabolismo , Animais , Neoplasias da Mama/genética , Carboxiliases/metabolismo , Diferenciação Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metabolismo dos Lipídeos/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Fosfatidiletanolaminas/metabolismo , Proteínas Supressoras de Tumor/genética , beta-Lactamases/genética
12.
BMC Microbiol ; 21(1): 59, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33618670

RESUMO

BACKGROUND: Stool metabolites provide essential insights into the function of the gut microbiome. The current gold standard for storage of stool samples for metabolomics is flash-freezing at - 80 °C which can be inconvenient and expensive. Ambient temperature storage of stool is more practical, however no available methodologies adequately preserve the metabolomic profile of stool. A novel sampling kit (OMNImet.GUT; DNA Genotek, Inc.) was introduced for ambient temperature storage and stabilization of feces for metabolomics; we aimed to test the performance of this kit vs. flash-freezing. To do this stool was collected from an infant's diaper was divided into two aliquots: 1) flash-frozen and 2) stored in an OMNImet.GUT tube at ambient temperature for 3-4 days. Samples from the same infant were collected at 2 different time points to assess metabolite changes over time. Subsequently, all samples underwent metabolomic analysis by liquid chromatography - tandem mass spectrometry (LC-MS/MS). RESULTS: Paired fecal samples (flash-frozen and ambient temperature) from 16 infants were collected at 2 time points (32 individual samples, 64 aliquots). Similar numbers of metabolites were detected in both the frozen and ambient temperature samples (1126 in frozen, 1107 in ambient temperature, 1064 shared between sample types). Metabolite abundances were strongly correlated between storage methods (median Spearman correlation Rs = 0.785 across metabolites). Hierarchical clustering analysis and principal component analysis showed that samples from the same individuals at a given time point clustered closely, regardless of the storage method. Repeat samples from the same individual were compared by paired t-test, separately for the frozen and OMNImet.GUT. The number of metabolites in each biochemical class that significantly changed (p < 0.05) at timepoint 2 relative to timepoint 1 was similar in flash-frozen versus ambient temperature storage. Changes in microbiota modified metabolites over time were also consistent across both methodologies. CONCLUSION: Ambient temperature storage and stabilization of stool in the OMNImet.GUT device yielded comparable metabolomic results to flash freezing in terms of 1) the identity and abundance of detected biochemicals 2) the distinct metabolomic profiles of subjects and 3) changes in metabolites over time that are plausibly microbiota-induced. This method potentially provides a more convenient, less expensive home collection and storage option for stool metabolomic analysis.


Assuntos
Fezes/microbiologia , Congelamento , Metabolômica/métodos , Preservação Biológica/instrumentação , Preservação Biológica/métodos , Manejo de Espécimes/instrumentação , Temperatura , Cromatografia Líquida , DNA Bacteriano/genética , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiologia , Humanos , Lactente , Metabolômica/instrumentação , RNA Ribossômico 16S/genética , Manejo de Espécimes/métodos , Espectrometria de Massas em Tandem
13.
Nature ; 520(7547): 363-7, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25855294

RESUMO

Cancer cells adapt their metabolic processes to support rapid proliferation, but less is known about how cancer cells alter metabolism to promote cell survival in a poorly vascularized tumour microenvironment. Here we identify a key role for serine and glycine metabolism in the survival of brain cancer cells within the ischaemic zones of gliomas. In human glioblastoma multiforme, mitochondrial serine hydroxymethyltransferase (SHMT2) and glycine decarboxylase (GLDC) are highly expressed in the pseudopalisading cells that surround necrotic foci. We find that SHMT2 activity limits that of pyruvate kinase (PKM2) and reduces oxygen consumption, eliciting a metabolic state that confers a profound survival advantage to cells in poorly vascularized tumour regions. GLDC inhibition impairs cells with high SHMT2 levels as the excess glycine not metabolized by GLDC can be converted to the toxic molecules aminoacetone and methylglyoxal. Thus, SHMT2 is required for cancer cells to adapt to the tumour environment, but also renders these cells sensitive to glycine cleavage system inhibition.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Glicina Hidroximetiltransferase/metabolismo , Glicina/metabolismo , Isquemia/metabolismo , Acetona/análogos & derivados , Acetona/metabolismo , Acetona/toxicidade , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/enzimologia , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Glioblastoma/irrigação sanguínea , Glioblastoma/enzimologia , Glicina Desidrogenase (Descarboxilante)/antagonistas & inibidores , Glicina Desidrogenase (Descarboxilante)/metabolismo , Humanos , Isquemia/enzimologia , Isquemia/patologia , Camundongos , Necrose , Consumo de Oxigênio , Aldeído Pirúvico/metabolismo , Aldeído Pirúvico/toxicidade , Piruvato Quinase/metabolismo , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Proc Natl Acad Sci U S A ; 115(34): E7978-E7986, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30082411

RESUMO

The metabolic and redox state changes during the transition from an arrested oocyte to a totipotent embryo remain uncharacterized. Here, we applied state-of-the-art, integrated methodologies to dissect these changes in Drosophila We demonstrate that early embryos have a more oxidized state than mature oocytes. We identified specific alterations in reactive cysteines at a proteome-wide scale as a result of this metabolic and developmental transition. Consistent with a requirement for redox change, we demonstrate a role for the ovary-specific thioredoxin Deadhead (DHD). dhd-mutant oocytes are prematurely oxidized and exhibit meiotic defects. Epistatic analyses with redox regulators link dhd function to the distinctive redox-state balance set at the oocyte-to-embryo transition. Crucially, global thiol-redox profiling identified proteins whose cysteines became differentially modified in the absence of DHD. We validated these potential DHD substrates by recovering DHD-interaction partners using multiple approaches. One such target, NO66, is a conserved protein that genetically interacts with DHD, revealing parallel functions. As redox changes also have been observed in mammalian oocytes, we hypothesize a link between developmental control of this cell-cycle transition and regulation by metabolic cues. This link likely operates both by general redox state and by changes in the redox state of specific proteins. The redox proteome defined here is a valuable resource for future investigation of the mechanisms of redox-modulated control at the oocyte-to-embryo transition.


Assuntos
Ciclo Celular/fisiologia , Cisteína/metabolismo , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/fisiologia , Oócitos/metabolismo , Animais , Cisteína/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Embrião não Mamífero/citologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Oócitos/citologia , Oxirredução , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
15.
Proc Natl Acad Sci U S A ; 115(16): 4228-4233, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610318

RESUMO

Non-small-cell lung cancer (NSCLC) is a leading cause of cancer death worldwide, with 25% of cases harboring oncogenic Kirsten rat sarcoma (KRAS). Although KRAS direct binding to and activation of PI3K is required for KRAS-driven lung tumorigenesis, the contribution of insulin receptor (IR) and insulin-like growth factor 1 receptor (IGF1R) in the context of mutant KRAS remains controversial. Here, we provide genetic evidence that lung-specific dual ablation of insulin receptor substrates 1/2 (Irs1/Irs2), which mediate insulin and IGF1 signaling, strongly suppresses tumor initiation and dramatically extends the survival of a mouse model of lung cancer with Kras activation and p53 loss. Mice with Irs1/Irs2 loss eventually succumb to tumor burden, with tumor cells displaying suppressed Akt activation and strikingly diminished intracellular levels of essential amino acids. Acute loss of IRS1/IRS2 or inhibition of IR/IGF1R in KRAS-mutant human NSCLC cells decreases the uptake and lowers the intracellular levels of amino acids, while enhancing basal autophagy and sensitivity to autophagy and proteasome inhibitors. These findings demonstrate that insulin/IGF1 signaling is required for KRAS-mutant lung cancer initiation, and identify decreased amino acid levels as a metabolic vulnerability in tumor cells with IR/IGF1R inhibition. Consequently, combinatorial targeting of IR/IGF1R with autophagy or proteasome inhibitors may represent an effective therapeutic strategy in KRAS-mutant NSCLC.


Assuntos
Carcinogênese/metabolismo , Carcinoma Pulmonar de Células não Pequenas/prevenção & controle , Genes ras , Proteínas Substratos do Receptor de Insulina/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Insulina/farmacologia , Neoplasias Pulmonares/prevenção & controle , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Células A549 , Aminoácidos/metabolismo , Animais , Autofagia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/fisiopatologia , Códon de Terminação , Humanos , Proteínas Substratos do Receptor de Insulina/deficiência , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/fisiopatologia , Camundongos , Proteínas de Neoplasias/fisiologia , Proteólise , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia
16.
Blood ; 131(26): 2955-2966, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29712634

RESUMO

Red cells contain a unique constellation of membrane lipids. Although much is known about regulated protein expression, the regulation of lipid metabolism during erythropoiesis is poorly studied. Here, we show that transcription of PHOSPHO1, a phosphoethanolamine and phosphocholine phosphatase that mediates the hydrolysis of phosphocholine to choline, is strongly upregulated during the terminal stages of erythropoiesis of both human and mouse erythropoiesis, concomitant with increased catabolism of phosphatidylcholine (PC) and phosphocholine as shown by global lipidomic analyses of mouse and human terminal erythropoiesis. Depletion of PHOSPHO1 impaired differentiation of fetal mouse and human erythroblasts, and, in adult mice, depletion impaired phenylhydrazine-induced stress erythropoiesis. Loss of PHOSPHO1 also impaired phosphocholine catabolism in mouse fetal liver progenitors and resulted in accumulation of several lipids; adenosine triphosphate (ATP) production was reduced as a result of decreased oxidative phosphorylation. Glycolysis replaced oxidative phosphorylation in PHOSPHO1-knockout erythroblasts and the increased glycolysis was used for the production of serine or glycine. Our study elucidates the dynamic changes in lipid metabolism during terminal erythropoiesis and reveals the key roles of PC and phosphocholine metabolism in energy balance and amino acid supply.


Assuntos
Eritroblastos/metabolismo , Eritropoese , Fosforilcolina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Eritroblastos/citologia , Deleção de Genes , Glicólise , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação Oxidativa , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo
17.
Proc Natl Acad Sci U S A ; 114(47): E10132-E10141, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29109271

RESUMO

Mammalian spermatogenesis is an elaborately organized differentiation process, starting with diploid spermatogonia, which include germ-line stem cells, and ending with haploid spermatozoa. The process involves four pivotal transitions occurring in physical proximity: spermatogonial differentiation, meiotic initiation, initiation of spermatid elongation, and release of spermatozoa. We report how the four transitions are coordinated in mice. Two premeiotic transitions, spermatogonial differentiation and meiotic initiation, were known to be coregulated by an extrinsic signal, retinoic acid (RA). Our chemical manipulations of RA levels in mouse testes now reveal that RA also regulates the two postmeiotic transitions: initiation of spermatid elongation and spermatozoa release. We measured RA concentrations and found that they changed periodically, as also reflected in the expression patterns of an RA-responsive gene, STRA8; RA levels were low before the four transitions, increased when the transitions occurred, and remained elevated thereafter. We found that pachytene spermatocytes, which express an RA-synthesizing enzyme, Aldh1a2, contribute directly and significantly to RA production in testes. Indeed, chemical and genetic depletion of pachytene spermatocytes revealed that RA from pachytene spermatocytes was required for the two postmeiotic transitions, but not for the two premeiotic transitions. We conclude that the premeiotic transitions are coordinated by RA from Sertoli (somatic) cells. Once germ cells enter meiosis, pachytene spermatocytes produce RA to coordinate the two postmeiotic transitions. In combination, these elements underpin the spatiotemporal coordination of spermatogenesis and ensure its prodigious output in adult males.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Aldeído Desidrogenase/genética , Regulação da Expressão Gênica no Desenvolvimento , Espermatogênese/genética , Espermatozoides/metabolismo , Tretinoína/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Diferenciação Celular , Masculino , Meiose , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estágio Paquíteno , Retinal Desidrogenase , Transdução de Sinais , Espermátides/citologia , Espermátides/crescimento & desenvolvimento , Espermátides/metabolismo , Espermatócitos/citologia , Espermatócitos/crescimento & desenvolvimento , Espermatócitos/metabolismo , Espermatogônias/citologia , Espermatogônias/crescimento & desenvolvimento , Espermatogônias/imunologia , Espermatozoides/citologia , Espermatozoides/crescimento & desenvolvimento , Testículo/citologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
18.
Proc Natl Acad Sci U S A ; 114(17): E3434-E3443, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28396387

RESUMO

Oncogenic PIK3CA mutations are found in a significant fraction of human cancers, but therapeutic inhibition of PI3K has only shown limited success in clinical trials. To understand how mutant PIK3CA contributes to cancer cell proliferation, we used genome scale loss-of-function screening in a large number of genomically annotated cancer cell lines. As expected, we found that PIK3CA mutant cancer cells require PIK3CA but also require the expression of the TCA cycle enzyme 2-oxoglutarate dehydrogenase (OGDH). To understand the relationship between oncogenic PIK3CA and OGDH function, we interrogated metabolic requirements and found an increased reliance on glucose metabolism to sustain PIK3CA mutant cell proliferation. Functional metabolic studies revealed that OGDH suppression increased levels of the metabolite 2-oxoglutarate (2OG). We found that this increase in 2OG levels, either by OGDH suppression or exogenous 2OG treatment, resulted in aspartate depletion that was specifically manifested as auxotrophy within PIK3CA mutant cells. Reduced levels of aspartate deregulated the malate-aspartate shuttle, which is important for cytoplasmic NAD+ regeneration that sustains rapid glucose breakdown through glycolysis. Consequently, because PIK3CA mutant cells exhibit a profound reliance on glucose metabolism, malate-aspartate shuttle deregulation leads to a specific proliferative block due to the inability to maintain NAD+/NADH homeostasis. Together these observations define a precise metabolic vulnerability imposed by a recurrently mutated oncogene.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases , Complexo Cetoglutarato Desidrogenase , Mutação , Proteínas de Neoplasias , Neoplasias , Animais , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico/genética , Classe I de Fosfatidilinositol 3-Quinases/genética , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Glicólise/genética , Humanos , Complexo Cetoglutarato Desidrogenase/biossíntese , Complexo Cetoglutarato Desidrogenase/genética , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia
19.
Proc Natl Acad Sci U S A ; 113(28): 7894-9, 2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27354530

RESUMO

In choanoflagellates, the closest living relatives of animals, multicellular rosette development is regulated by environmental bacteria. The simplicity of this evolutionarily relevant interaction provides an opportunity to identify the molecules and regulatory logic underpinning bacterial regulation of development. We find that the rosette-inducing bacterium Algoriphagus machipongonensis produces three structurally divergent classes of bioactive lipids that, together, activate, enhance, and inhibit rosette development in the choanoflagellate Salpingoeca rosetta. One class of molecules, the lysophosphatidylethanolamines (LPEs), elicits no response on its own but synergizes with activating sulfonolipid rosette-inducing factors (RIFs) to recapitulate the full bioactivity of live Algoriphagus. LPEs, although ubiquitous in bacteria and eukaryotes, have not previously been implicated in the regulation of a host-microbe interaction. This study reveals that multiple bacterially produced lipids converge to activate, enhance, and inhibit multicellular development in a choanoflagellate.


Assuntos
Proteínas de Bactérias/fisiologia , Bacteroidetes/fisiologia , Coanoflagelados/fisiologia , Proteínas de Bactérias/isolamento & purificação , Lipídeos/fisiologia
20.
Nat Chem Biol ; 12(6): 452-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27110680

RESUMO

Serine is both a proteinogenic amino acid and the source of one-carbon units essential for de novo purine and deoxythymidine synthesis. In the canonical pathway of glucose-derived serine synthesis, Homo sapiens phosphoglycerate dehydrogenase (PHGDH) catalyzes the first, rate-limiting step. Genetic loss of PHGDH is toxic toward PHGDH-overexpressing breast cancer cell lines even in the presence of exogenous serine. Here, we used a quantitative high-throughput screen to identify small-molecule PHGDH inhibitors. These compounds reduce the production of glucose-derived serine in cells and suppress the growth of PHGDH-dependent cancer cells in culture and in orthotopic xenograft tumors. Surprisingly, PHGDH inhibition reduced the incorporation into nucleotides of one-carbon units from glucose-derived and exogenous serine. We conclude that glycolytic serine synthesis coordinates the use of one-carbon units from endogenous and exogenous serine in nucleotide synthesis, and we suggest that one-carbon unit wasting thus may contribute to the efficacy of PHGDH inhibitors in vitro and in vivo.


Assuntos
Carbono/metabolismo , Inibidores Enzimáticos/farmacologia , Fosfoglicerato Desidrogenase/antagonistas & inibidores , Serina/biossíntese , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Carbono/química , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Feminino , Glicólise/efeitos dos fármacos , Humanos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Estrutura Molecular , Fosfoglicerato Desidrogenase/metabolismo , Purinas/biossíntese , Serina/química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Timidina/biossíntese , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa