Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Exp Eye Res ; 213: 108845, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34800480

RESUMO

Diabetic retinopathy (DR) is the leading cause of vision impairment in working age adults. In addition to hyperglycemia, retinal inflammation is an important driving factor for DR development. Although DR is clinically described as diabetes-induced damage to the retinal blood vessels, several studies have reported that metabolic dysregulation occurs in the retina prior to the development of microvascular damage. The two most commonly affected metabolic pathways in diabetic conditions are glycolysis and the glutamate pathway. We investigated the role of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and glutamine synthetase (GS) in an in-vitro model of DR incorporating high glucose and pro-inflammatory cytokines. We found that GAPDH and GS enzyme activity were not significantly affected in hyperglycemic conditions or after exposure to cytokines alone, but were significantly decreased in the DR model. This confirmed that pro-inflammatory cytokines IL-1ß and TNFα enhance the hyperglycemic metabolic deficit. We further investigated metabolite and amino acid levels after specific pharmacological inhibition of GAPDH or GS in the absence/presence of pro-inflammatory cytokines. The results indicate that GAPDH inhibition increased glucose and addition of cytokines increased lactate and ATP levels and reduced glutamate levels. GS inhibition did not alter retinal metabolite levels but the addition of cytokines increased ATP levels and caused glutamate accumulation in Müller cells. We conclude that it is the action of pro-inflammatory cytokines concomitantly with the inhibition of the glycolytic or GS mediated glutamate recycling that contribute to metabolic dysregulation in DR. Therefore, in the absence of good glycemic control, therapeutic interventions aimed at regulating inflammation may prevent the onset of early metabolic imbalance in DR.


Assuntos
Retinopatia Diabética/enzimologia , Inibidores Enzimáticos/farmacologia , Glutamato-Amônia Ligase/antagonistas & inibidores , Gliceraldeído-3-Fosfato Desidrogenases/antagonistas & inibidores , Interleucina-1beta/farmacologia , Retina/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Retinopatia Diabética/patologia , Feminino , Glucose/farmacologia , Hiperglicemia/metabolismo , Ácido Iodoacético/farmacologia , L-Lactato Desidrogenase/metabolismo , Metionina Sulfoximina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Retina/enzimologia , Retina/patologia
2.
Oncol Rep ; 45(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33846803

RESUMO

During tumorigenesis, oncogene activation and metabolism rewiring are interconnected. Activated c­Myc upregulates several genes involved in glutamine metabolism, making cancer cells dependent on high levels of this amino acid to survive and proliferate. After studying the response to glutamine deprivation in cancer cells, it was found that glutamine starvation not only blocked cellular proliferation, but also altered c­Myc protein expression, leading to a reduction in the levels of the canonical c­Myc isoform and an increase in the expression of c­Myc 1, a c­Myc isoform translated from an in­frame 5' CUG codon. In an attempt to identify nutrients able to counteract glutamine deprivation effects, it was shown that, in the absence of glutamine, asparagine permitted cell survival and proliferation, and maintained c­Myc expression as in glutamine­fed cells, with high levels of canonical c­Myc and c­Myc 1 almost undetectable. In asparagine­fed cells, global protein translation was higher than in glutamine­starved cells, and there was an increase in the levels of glutamine synthetase (GS), whose activity was essential for cellular viability and proliferation. In glutamine­starved asparagine­fed cells, the inhibition of c­Myc activity led to a decrease in global protein translation and GS synthesis, suggesting an association between c­Myc expression, GS levels and cellular proliferation, mediated by asparagine when exogenous glutamine is absent.


Assuntos
Asparagina/metabolismo , Transformação Celular Neoplásica/metabolismo , Glutamina/deficiência , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proliferação de Células/genética , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Inibidores de Cisteína Proteinase/farmacologia , Regulação Neoplásica da Expressão Gênica , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/metabolismo , Humanos , Leupeptinas/farmacologia , Metionina Sulfoximina/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética
3.
Neurosci Lett ; 755: 135847, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33774150

RESUMO

The development and maintenance of morphine tolerance showed association with neuroinflammation and dysfunction of central glutamatergic system (such as nitration of glutamate transporter). Recent evidence indicated that hydrogen could reduce the levels of neuroinflammation and oxidative stress, but its role in morphine tolerance has not been studied. The rats were intrathecally administered with morphine (10 µg/10 µL each time, twice/day for 5 days). Hydrogen enriched saline (HS) or saline was given intraperitoneally at 1, 3 and 10 mL/kg for 10 min before each dose of morphine administration. The tail-flick latency, mechanical threshold and thermal latency were assessed one day (baseline) before and daily for up to 5 days during morphine injection. The pro-inflammatory cytokine expressions [tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), IL-6)] (by western blotting), astrocyte activation (by immunofluorescence and western blotting), and nitration of glutamate transporter-1 (GLT-1) and glutamine synthetase (GS) (by immunoprecipitation), membrane and total expression of N-methyl-d-aspartic acid (NMDA) receptor NR1 and NR2B subunits were carried out in the spinal dorsal horns. Chronic morphine administration induced antinociceptive tolerance, and together led to increased TNF-α, IL-1ß and IL-6 expression, astrocyte activation, GLT-1 and GS nitration, increased membrane and total NR1, NR2B expression. Injection of HS attenuated morphine tolerance in a dose-dependent manner, decreased proinflammatory cytokine expression, inhibited astrocyte activation, decreased GLT-1 and GS nitration, and inhibited membrane trafficking of NMDA receptor. Our result showed that hydrogen pretreatment prevented morphine tolerance by reducing neuroinflammation, GLT-1, GS nitration, NMDA receptor trafficking in the spinal dorsal horn. Pretreatment with hydrogen might be considered as a novel therapeutic strategy for the prevention of morphine tolerance.


Assuntos
Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Glutamato-Amônia Ligase/antagonistas & inibidores , Mediadores da Inflamação/antagonistas & inibidores , Morfina/administração & dosagem , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Solução Salina/administração & dosagem , Medula Espinal/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Tolerância a Medicamentos/fisiologia , Transportador 2 de Aminoácido Excitatório/metabolismo , Glutamato-Amônia Ligase/metabolismo , Hidrogênio/administração & dosagem , Mediadores da Inflamação/metabolismo , Injeções Intraperitoneais , Injeções Espinhais , Masculino , Nitratos/antagonistas & inibidores , Nitratos/metabolismo , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Medula Espinal/efeitos dos fármacos
4.
Int J Mol Sci ; 22(4)2021 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-33567690

RESUMO

The significance of glutamine in cancer metabolism has been extensively studied. Cancer cells consume an excessive amount of glutamine to facilitate rapid proliferation. Thus, glutamine depletion occurs in various cancer types, especially in poorly vascularized cancers. This makes glutamine synthetase (GS), the only enzyme responsible for de novo synthesizing glutamine, essential in cancer metabolism. In cancer, GS exhibits pro-tumoral features by synthesizing glutamine, supporting nucleotide synthesis. Furthermore, GS is highly expressed in the tumor microenvironment (TME) and provides glutamine to cancer cells, allowing cancer cells to maintain sufficient glutamine level for glutamine catabolism. Glutamine catabolism, the opposite reaction of glutamine synthesis by GS, is well known for supporting cancer cell proliferation via contributing biosynthesis of various essential molecules and energy production. Either glutamine anabolism or catabolism has a critical function in cancer metabolism depending on the complex nature and microenvironment of cancers. In this review, we focus on the role of GS in a variety of cancer types and microenvironments and highlight the mechanism of GS at the transcriptional and post-translational levels. Lastly, we discuss the therapeutic implications of targeting GS in cancer.


Assuntos
Antineoplásicos/farmacologia , Glutamato-Amônia Ligase/antagonistas & inibidores , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Animais , Humanos , Neoplasias/enzimologia , Neoplasias/patologia
5.
BMC Cancer ; 21(1): 174, 2021 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-33596851

RESUMO

BACKGROUND: Cisplatin (CDDP) significantly prolongs survival in various cancers, but many patients also develop resistance that results in treatment failure. Thus, this study aimed to elucidate the underlying mechanisms by which ovarian cancer cells acquire CDDP resistance. METHODS: We evaluated the metabolic profiles in CDDP-sensitive ovarian cancer A2780 cells and CDDP-resistant A2780cis cells using capillary electrophoresis-time-of-flight mass spectrometry (CE-TOFMS). We further examined the expression of glutamine metabolism enzymes using real-time PCR and Western blot analyses. Cell viability was accessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. RESULTS: The results showed that levels of glutamine, glutamate, and glutathione (GSH), a key drug resistance mediator synthesized from glutamate, were significantly elevated in A2780cis cells than those in A2780 cells. Furthermore, glutamine starvation decreased the GSH levels and CDDP resistance in A2780cis cells. Interestingly, the expression of glutamine synthetase (GS/GLUL), which synthesizes glutamine from glutamate and thereby negatively regulates GSH production, was almost completely suppressed in resistant A2780cis cells. In addition, treatment of A2780cis cells with 5-aza-2'-deoxycytidine, a DNA-demethylating agent, restored GS expression and reduced CDDP resistance. In contrast, GS knockdown in CDDP-sensitive A2780 cells induced CDDP resistance. CONCLUSIONS: The results indicate that upregulation of GSH synthesis from glutamine via DNA methylation-mediated silencing of GS causes CDDP resistance in A2780cis cells. Therefore, glutamine metabolism could be a novel therapeutic target against CDDP resistance.


Assuntos
Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Inativação Gênica , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamina/metabolismo , Metaboloma , Neoplasias Ovarianas/tratamento farmacológico , Antineoplásicos/farmacologia , Apoptose , Proliferação de Células , Reprogramação Celular , Feminino , Glutamato-Amônia Ligase/genética , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Células Tumorais Cultivadas
6.
Nature ; 587(7835): 626-631, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33116312

RESUMO

Muscle regeneration is sustained by infiltrating macrophages and the consequent activation of satellite cells1-4. Macrophages and satellite cells communicate in different ways1-5, but their metabolic interplay has not been investigated. Here we show, in a mouse model, that muscle injuries and ageing are characterized by intra-tissue restrictions of glutamine. Low levels of glutamine endow macrophages with the metabolic ability to secrete glutamine via enhanced glutamine synthetase (GS) activity, at the expense of glutamine oxidation mediated by glutamate dehydrogenase 1 (GLUD1). Glud1-knockout macrophages display constitutively high GS activity, which prevents glutamine shortages. The uptake of macrophage-derived glutamine by satellite cells through the glutamine transporter SLC1A5 activates mTOR and promotes the proliferation and differentiation of satellite cells. Consequently, macrophage-specific deletion or pharmacological inhibition of GLUD1 improves muscle regeneration and functional recovery in response to acute injury, ischaemia or ageing. Conversely, SLC1A5 blockade in satellite cells or GS inactivation in macrophages negatively affects satellite cell functions and muscle regeneration. These results highlight the metabolic crosstalk between satellite cells and macrophages, in which macrophage-derived glutamine sustains the functions of satellite cells. Thus, the targeting of GLUD1 may offer therapeutic opportunities for the regeneration of injured or aged muscles.


Assuntos
Glutamina/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Regeneração , Células Satélites de Músculo Esquelético/metabolismo , Envelhecimento/metabolismo , Sistema ASC de Transporte de Aminoácidos/antagonistas & inibidores , Sistema ASC de Transporte de Aminoácidos/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Feminino , Glutamato Desidrogenase/deficiência , Glutamato Desidrogenase/genética , Glutamato Desidrogenase/metabolismo , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/metabolismo , Macrófagos/enzimologia , Masculino , Camundongos , Antígenos de Histocompatibilidade Menor/metabolismo , Músculo Esquelético/citologia , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Oxirredução , Células Satélites de Músculo Esquelético/citologia , Serina-Treonina Quinases TOR
7.
J Nat Prod ; 83(9): 2809-2813, 2020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-32830503

RESUMO

Arsinothricin [AST (1)], a new broad-spectrum organoarsenical antibiotic, is a nonproteinogenic analogue of glutamate that effectively inhibits glutamine synthetase. We report the chemical synthesis of an intermediate in the pathway to 1, hydroxyarsinothricin [AST-OH (2)], which can be converted to 1 by enzymatic methylation catalyzed by the ArsM As(III) S-adenosylmethionine methyltransferase. This is the first report of semisynthesis of 1, providing a source of this novel antibiotic that will be required for future clinical trials.


Assuntos
Antibacterianos/síntese química , Arsenicais/síntese química , Antibacterianos/farmacologia , Arsenicais/farmacologia , Cromatografia Líquida de Alta Pressão , Inibidores Enzimáticos/farmacologia , Glutamato-Amônia Ligase/antagonistas & inibidores , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , S-Adenosilmetionina/antagonistas & inibidores
8.
J Chem Theory Comput ; 16(7): 4694-4705, 2020 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-32551588

RESUMO

Glutamine synthetase (GS) catalyzes an ATP-dependent condensation of glutamate and ammonia to form glutamine. This reaction-and therefore GS-are indispensable for the hepatic nitrogen metabolism. Nitration of tyrosine 336 (Y336) inhibits human GS activity. GS nitration and the consequent loss of GS function are associated with a broad range of neurological diseases. The mechanism by which Y336 nitration inhibits GS, however, is not understood. Here, we show by means of unbiased MD simulations, binding, and configurational free energy computations that Y336 nitration hampers ATP binding but only in the deprotonated and negatively charged state of residue 336. By contrast, for the protonated and neutral state, our computations indicate an increased binding affinity for ATP. pKa computations of nitrated Y336 within GS predict a pKa of ∼5.3. Thus, at physiological pH, nitrated Y336 exists almost exclusively in the deprotonated and negatively charged state. In vitro experiments confirm these predictions, in that, the catalytic activity of nitrated GS is decreased at pH 7 and 6 but not at pH 4. These results indicate a novel, fully reversible, pH-sensitive mechanism for the regulation of GS activity by tyrosine nitration.


Assuntos
Glutamato-Amônia Ligase/metabolismo , Nitratos/química , Tirosina/química , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/genética , Humanos , Concentração de Íons de Hidrogênio , Cinética , Simulação de Dinâmica Molecular , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Termodinâmica
9.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118571, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31706909

RESUMO

The heterotrimeric transcription factor NF-Y binds to CCAAT boxes of genes of glutamine metabolism. We set out to study the role of the regulatory NF-YA subunit in this pathway. We produced U2OS and A549 clones stably overexpressing -OE- the two splicing isoforms of NF-YA. NF-YA OE cells show normal growth and colony formation rates, but they become resistant to cell death upon glutamine deprivation. Increased mRNA and protein expression of the key biosynthetic enzyme GLUL in U2OS entails increased production of endogenous glutamine upon deprivation. The use of GLUL inhibitors dampens the NF-YA-mediated effect. NF-YA OE prevents activation of the pro-apoptotic transcription factor CHOP/DDIT3. Elevated basal levels of SERCA1/2, coding for the molecular target of Thapsigargin, correlate with resistance of NF-YA OE cells to the drug. The work represents a proof-of-principle that elevated levels of NF-YA, as found in some tumor types, helps altering cancer metabolic pathways.


Assuntos
Fator de Ligação a CCAAT/metabolismo , Glutamina/metabolismo , Fator de Ligação a CCAAT/genética , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/metabolismo , Glutamina/deficiência , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Tapsigargina/farmacologia , Fator de Transcrição CHOP/metabolismo
10.
Cell Rep ; 29(5): 1287-1298.e6, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31665640

RESUMO

Glutamine is thought to play an important role in cancer cells by being deaminated via glutaminolysis to α-ketoglutarate (aKG) to fuel the tricarboxylic acid (TCA) cycle. Supporting this notion, aKG supplementation can restore growth/survival of glutamine-deprived cells. However, pancreatic cancers are often poorly vascularized and limited in glutamine supply, in alignment with recent concerns on the significance of glutaminolysis in pancreatic cancer. Here, we show that aKG-mediated rescue of glutamine-deprived pancreatic ductal carcinoma (PDAC) cells requires glutamate ammonia ligase (GLUL), the enzyme responsible for de novo glutamine synthesis. GLUL-deficient PDAC cells are capable of the TCA cycle but defective in aKG-coupled glutamine biosynthesis and subsequent nitrogen anabolic processes. Importantly, GLUL expression is elevated in pancreatic cancer patient samples and in mouse PDAC models. GLUL ablation suppresses the development of KrasG12D-driven murine PDAC. Therefore, GLUL-mediated glutamine biosynthesis couples the TCA cycle with nitrogen anabolism and plays a critical role in PDAC.


Assuntos
Carbono/metabolismo , Glutamina/metabolismo , Nitrogênio/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Carcinoma Ductal Pancreático/enzimologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Deleção de Genes , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/metabolismo , Humanos , Ácidos Cetoglutáricos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia
11.
J Cancer Res Clin Oncol ; 144(5): 821-833, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29435734

RESUMO

PURPOSE: Glutamine (Gln) is essential for the proliferation of most cancer cells, making it an appealing target for cancer therapy. However, the role of Gln in gastric cancer (GC) metabolism is unknown and Gln-targeted therapy against GC remains scarce. The aim of this study was to investigate the relevance of Gln in GC growth and targeting. METHODS: Expression of Gln transporter ASCT2 and glutamine synthetase (GS) in the parental and molecularly engineered GC cells or in human GC specimens was determined by RT-PCR and western blot analysis or immunohistochemistry. Cell proliferation and survival was assessed by CCK-8 assay and colony formation assay. Intracellular Gln content was measured by a HPLC system. Effects of ASCT2 and/or GS inhibitor on tumor growth were investigated in xenograft models. RESULTS: A significant heterogeneity of GC cells was observed with respect to their response to the treatment of ASCT2 inhibitor benzylserine (BenSer). Gln deprivation did not affect the BenSer-resistant cell growth due to endogenous GS expression, whose inhibition remarkably reduced cell proliferation. The differential in vitro sensitivity correlated with overall intracellular Gln content. Combined therapy with both ASCT2 and GS inhibitors produced a greater therapeutic efficacy than the treatment of either inhibitor alone. Furthermore, 77% human GC tissues were found to express moderate and high levels of ASCT2, 12% of which also co-expressed relatively high levels of GS. CONCLUSION: Gln mediates GC growth and the therapeutic efficacy of Gln-targeted treatment relies on distinct ASCT2 and GS expression pattern in specific gastric cancer groups.


Assuntos
Sistema ASC de Transporte de Aminoácidos/genética , Proliferação de Células/genética , Glutamato-Amônia Ligase/genética , Antígenos de Histocompatibilidade Menor/genética , Neoplasias Gástricas/genética , Sistema ASC de Transporte de Aminoácidos/antagonistas & inibidores , Sistema ASC de Transporte de Aminoácidos/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzeno/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/metabolismo , Glutamina/antagonistas & inibidores , Glutamina/metabolismo , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Antígenos de Histocompatibilidade Menor/metabolismo , Terapia de Alvo Molecular/métodos , Serina/administração & dosagem , Serina/química , Serina/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Biochemistry ; 57(1): 117-135, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29039929

RESUMO

Tabtoxinine-ß-lactam (TßL), also known as wildfire toxin, is a time- and ATP-dependent inhibitor of glutamine synthetase produced by plant pathogenic strains of Pseudomonas syringae. Here we demonstrate that recombinant glutamine synthetase from Escherichia coli phosphorylates the C3-hydroxyl group of the TßL 3-(S)-hydroxy-ß-lactam (3-HßL) warhead. Phosphorylation of TßL generates a stable, noncovalent enzyme-ADP-inhibitor complex that resembles the glutamine synthetase tetrahedral transition state. The TßL ß-lactam ring remains intact during enzyme inhibition, making TßL mechanistically distinct from traditional ß-lactam antibiotics such as penicillin. Our findings could enable the design of new 3-HßL transition state inhibitors targeting enzymes in the ATP-dependent carboxylate-amine ligase superfamily with broad therapeutic potential in many disease areas.


Assuntos
Trifosfato de Adenosina/metabolismo , Azetidinas/farmacologia , Toxinas Bacterianas/farmacologia , Proteínas de Escherichia coli/antagonistas & inibidores , Escherichia coli/enzimologia , Glutamato-Amônia Ligase/antagonistas & inibidores , Azetidinas/isolamento & purificação , Azetidinas/metabolismo , Toxinas Bacterianas/biossíntese , Toxinas Bacterianas/isolamento & purificação , Catálise , Cromatografia Líquida , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Espectrometria de Massas , Testes de Sensibilidade Microbiana , Ressonância Magnética Nuclear Biomolecular , Fosforilação , Pseudomonas syringae/metabolismo
13.
PLoS One ; 12(10): e0185068, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28972974

RESUMO

Glutamine synthetase is a ubiquitous central enzyme in nitrogen metabolism that is controlled by up to four regulatory mechanisms, including adenylylation of some or all of the twelve subunits by adenylyl transferase. It is considered a potential therapeutic target for the treatment of tuberculosis, being essential for the growth of Mycobacterium tuberculosis, and is found extracellularly only in the pathogenic Mycobacterium strains. Human glutamine synthetase is not regulated by the adenylylation mechanism, so the adenylylated form of bacterial glutamine synthetase is of particular interest. Previously published reports show that, when M. tuberculosis glutamine synthetase is expressed in Escherichia coli, the E. coli adenylyl transferase does not optimally adenylylate the M. tuberculosis glutamine synthetase. Here, we demonstrate the production of soluble adenylylated M. tuberulosis glutamine synthetase in E. coli by the co-expression of M. tuberculosis glutamine synthetase and M. tuberculosis adenylyl transferase. The differential inhibition of adenylylated M. tuberulosis glutamine synthetase and deadenylylated M. tuberulosis glutamine synthetase by ATP based scaffold inhibitors are reported. Compounds selected on the basis of their enzyme inhibition were also shown to inhibit M. tuberculosis in the BACTEC 460TB™ assay as well as the intracellular inhibition of M. tuberculosis in a mouse bone-marrow derived macrophage assay.


Assuntos
Monofosfato de Adenosina/metabolismo , Descoberta de Drogas , Glutamato-Amônia Ligase/antagonistas & inibidores , Mycobacterium tuberculosis/enzimologia , Animais , Antituberculosos/farmacologia , Relação Dose-Resposta a Droga , Glutamato-Amônia Ligase/metabolismo , Células HeLa , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/efeitos dos fármacos
14.
Appl Microbiol Biotechnol ; 101(9): 3653-3661, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28175947

RESUMO

A glutamine synthetase (GS; 1341 bp) gene with potent L-phosphinothricin (PPT) resistance was isolated and characterized from a marine bacterium Exiguobacterium sp. Molecular docking analysis indicated that the substitution of residues Glu60 and Arg64 may lead to significant changes in binding pocket. To enhance the enzymatic property of GS, variants E60A and R64G were obtained by site-directed mutagenesis. The results revealed a noteworthy change in the thermostability and activity in comparison to the wild type (WT). WT exhibited optimum activity at 35 °C, while E60A and R64G exhibited optimum activity at 45 and 40 °C, respectively. The mutant R64G was 4.3 times more stable at 70 °C in comparison to WT, while E60A was 5.7 times more stable. Kinetic analysis revealed that the k cat value of R64G mutant was 8.10-, 7.25- and 7.63-fold that of WT for ADP, glutamine and hydroxylamine, respectively. The kinetic inhibition (K i, 4.91 ± 0.42 mM) of R64G was 2.02-fold that of WT (2.43 ± 0.14 mM) for L-phosphinothricin. The analysis of structure and function relationship showed that the binding pocket underwent dramatic changes when Arg site of 64 was substituted by Gly, thus promoting the rapid capture of substrates and leading to increase in activity and PPT-resistance of mutant R64G. The rearrangements of the residues at the molecular level formed new hydrogen bonds around the active site, which contributed to the increase of thermostability of enzymes. This study provides new insights into substrate binding mechanism of glutamine synthetase and the improved GS gene also has a potential for application in transgenic crops with L-phosphinothricin tolerance.


Assuntos
Aminobutiratos/metabolismo , Bacillales/enzimologia , Inibidores Enzimáticos/metabolismo , Glutamato-Amônia Ligase/isolamento & purificação , Glutamato-Amônia Ligase/metabolismo , Difosfato de Adenosina/metabolismo , Bacillales/genética , Sítios de Ligação , Estabilidade Enzimática , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/química , Glutamato-Amônia Ligase/genética , Glutamina/metabolismo , Ligação de Hidrogênio , Hidroxilamina/metabolismo , Cinética , Simulação de Acoplamento Molecular , Mutagênese Sítio-Dirigida , Temperatura
15.
J Hazard Mater ; 324(Pt B): 691-700, 2017 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-27899241

RESUMO

Aflatoxins (AFs), produced mainly by Aspergillus flavus and Aspergillus parasiticus, are strongly toxic and carcinogenic. Here, we showed that glutamine is the optimal nitrogen source for AF-production in A. flavus grown in Czapek Dox medium. Additionally, 4mM glutamine was the threshold for high production of aflatoxin B1. However, no significant impact of glutamine synthetase inhibitor was detected for on AF biosynthesis. In contrast, rapamycin could significantly suppress the glutamine inducing effect on AFs production, simultaneously inhibiting the fungal growth and conidiation. To identify the genes and regulatory networks involved in AFs biosynthesis, especially concerning the nitrogen source metabolism pathway and the target of rapamycin (TOR) signaling pathway, we obtained transcriptomes for A. flavus under treatment of three nitrogen sources by RNA-sequencing. We identified 1429 differentially expressed genes. Through GO and KEGG pathway analyses, the relationship between nitrogen metabolism and AFs biosynthesis was revealed, and the effects of TOR inhibitor were confirmed. Additionally, the quantitative real-time PCR results verified the credibility and reliability of the RNA-seq data, and were consistent with the other experimental results. Our research laid the foundation for a primary study on the involvement of the nitrogen regulatory network and TOR signaling pathway in AF biosynthesis.


Assuntos
Aflatoxina B1/biossíntese , Aspergillus flavus/crescimento & desenvolvimento , Glutamina/metabolismo , Nitrogênio/metabolismo , Aspergillus flavus/efeitos dos fármacos , Aspergillus flavus/genética , Aspergillus flavus/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes/efeitos dos fármacos , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/genética , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Transcriptoma/efeitos dos fármacos
16.
J Cell Biochem ; 118(8): 2018-2025, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27791265

RESUMO

Glutamate-ammonia ligase (GLUL) belongs to the glutamine synthetase family. It catalyzes the synthesis of glutamine from glutamate and ammonia in an ATP-dependent reaction. Here, we found higher expression of GLUL in the breast cancer patients was associated with larger tumor size and higher level of HER2 expression. In addition, GLUL was heterogeneously expressed in various breast cancer cells. The mRNA and protein expression levels of GLUL in SK-BR-3 cells were obviously higher than that in the other types of breast cancer cells. Results showed GLUL knockdown in SK-BR-3 cells could significantly decrease the proliferation ability. Furthermore, GLUL knockdown markedly inhibited the p38 MAPK and ERK1/ERK2 signaling pathways in SK-BR-3 cells. Thus, GLUL may represent a novel target for selectively inhibiting p38 MAPK and ERK1/ERK2 signaling pathways and the proliferation potential of breast cancer cells. J. Cell. Biochem. 118: 2018-2025, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Glutamato-Amônia Ligase/genética , RNA Mensageiro/genética , Adulto , Idoso , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/metabolismo , Humanos , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Gradação de Tumores , Estadiamento de Neoplasias , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transdução de Sinais , Análise de Sobrevida , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Metab Brain Dis ; 31(6): 1211-1215, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26521983

RESUMO

The concept of synergistic mechanisms as the pathophysiologic basis of hepatic encephalopathy started with the pioneering work of Les Zieve in Minneapolis some 60 years ago where synergistic actions of the liver-derived toxins ammonia, methanethiol, and octanoic acid were described. More recently, synergistic actions of ammonia and manganese, a toxic metal that is normally eliminated via the hepatobiliary route and shown to accumulate in brain in liver failure, on the glutamatergic neurotransmitter system were described. The current upsurge of interest in brain inflammation (neuroinflammation) in relation to the CNS complications of liver failure has added a third dimension to the synergy debate. The combined actions of ammonia, manganese and pro-inflammatory cytokines in brain in liver failure result in oxidative/nitrosative stress resulting from activation of glutamate (NMDA) receptors and consequent nitration of key brain proteins. One such protein, glutamine synthetase, the sole enzyme responsible for brain ammonia removal is nitrated and inactivated in brain in liver failure. Consequently, brain ammonia levels increase disproportionately resulting in alterations of brain excitability, impaired brain energy metabolism, encephalopathy and brain swelling. Experimental therapeutic approaches for which proof-of-principle has been established include the NMDA receptor antagonist memantine, N-acetyl cysteine (recently shown to have antioxidant properties at both hepatic and cerebral levels) and probiotics.


Assuntos
Glutamato-Amônia Ligase/metabolismo , Encefalopatia Hepática/metabolismo , Mediadores da Inflamação/metabolismo , Cirrose Hepática/metabolismo , Amônia/antagonistas & inibidores , Amônia/metabolismo , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Glutamato-Amônia Ligase/antagonistas & inibidores , Encefalopatia Hepática/tratamento farmacológico , Encefalopatia Hepática/etiologia , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/etiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
19.
Cell Metab ; 22(6): 1068-77, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26603296

RESUMO

c-Myc is known to promote glutamine usage by upregulating glutaminase (GLS), which converts glutamine to glutamate that is catabolized in the TCA cycle. Here we report that in a number of human and murine cells and cancers, Myc induces elevated expression of glutamate-ammonia ligase (GLUL), also termed glutamine synthetase (GS), which catalyzes the de novo synthesis of glutamine from glutamate and ammonia. This is through upregulation of a Myc transcriptional target thymine DNA glycosylase (TDG), which promotes active demethylation of the GS promoter and its increased expression. Elevated expression of GS promotes cell survival under glutamine limitation, while silencing of GS decreases cell proliferation and xenograft tumor growth. Upon GS overexpression, increased glutamine enhances nucleotide synthesis and amino acid transport. These results demonstrate an unexpected role of Myc in inducing glutamine synthesis and suggest a molecular connection between DNA demethylation and glutamine metabolism in Myc-driven cancers.


Assuntos
Glutamato-Amônia Ligase/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Linhagem Celular , DNA/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Feminino , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/genética , Ácido Glutâmico/metabolismo , Humanos , Camundongos , Camundongos Nus , Nucleotídeos/biossíntese , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo
20.
Pharmacol Biochem Behav ; 135: 246-53, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26093193

RESUMO

PURPOSE OF RESEARCH: Since, transient focal cerebral ischaemia exhibits detrimental effect not only during the course of ischaemia but also after the onset of reperfusion, the current study is focussed on identifying the appropriate therapeutic time point at which NG-nitro-l-arginine methyl ester (l-NAME) exerts better neuroprotection. PRINCIPAL RESULTS: Pre-ischaemic administration of l-NAME ameliorated neurological deficits much better than the during ischaemic and post-ischaemic groups. Pre-ischaemic l-NAME has also mitigated glutamate excitotoxicity, increased glutamine synthetase activity, ATP and NAD levels, decreased nitrate/nitrite content, down regulated TNF-α and upregulated IL-10 expressions and reduced the cerebral infarction significantly than the during ischaemic and post-ischaemic groups. MAJOR CONCLUSION: Current study revealed that l-NAME improved neurological deficit at the pre-ischaemic state in transient focal cerebral ischaemia and has also significantly ameliorated glutamate excitotoxicity. Though l-NAME showed neuroprotective effects when administered at during and post-ischaemia (during reperfusion), it exerts considerable neuroprotection when administered pre-ischaemically.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/psicologia , NG-Nitroarginina Metil Éster/uso terapêutico , Doenças do Sistema Nervoso/tratamento farmacológico , Trifosfato de Adenosina/metabolismo , Animais , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/psicologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Glutamato-Amônia Ligase/antagonistas & inibidores , Ácido Glutâmico/toxicidade , Infarto da Artéria Cerebral Média/complicações , Interleucina-10/metabolismo , Masculino , NAD/metabolismo , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/psicologia , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA