Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 396
Filtrar
1.
Nutr Neurosci ; 25(1): 64-69, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31900092

RESUMEN

Background: Glutamine synthetase (GS) is the only enzyme known to synthesize significant amounts of glutamine in mammals, and loss of GS in the hippocampus has been implicated in the pathophysiology of medication refractory mesial temporal lobe epilepsy (MTLE). Moreover, loss-of-function mutations of the GS gene causes severe epileptic encephalopathy, and supplementation with glutamine has been shown to normalize EEG and possibly improve the outcome in these patients. Here we examined whether oral glutamine supplementation is an effective treatment for MTLE by assessing the frequency and severity of seizures after supplementation in a translationally relevant model of the disease.Methods: Male Sprague Dawley rats (380-400 g) were allowed to drink unlimited amounts of glutamine in water (3.6% w/v; n = 8) or pure water (n = 8) for several weeks. Ten days after the start of glutamine supplementation, GS was chronically inhibited in the hippocampus to induce MTLE. Continuous video-intracranial EEG was collected for 21 days to determine the frequency and severity of seizures.Results: While there was no change in seizure frequency between the groups, the proportion of convulsive seizures was significantly higher in glutamine treated animals during the first three days of GS inhibition.Conclusion: The results suggest that oral glutamine supplementation transiently increases seizure severity in the initial stages of an epilepsy model, indicating a potential role of the amino acid in seizure propagation and epileptogenesis.


Asunto(s)
Epilepsia del Lóbulo Temporal/fisiopatología , Glutamina/administración & dosificación , Convulsiones/inducido químicamente , Índice de Severidad de la Enfermedad , Animales , Suplementos Dietéticos , Modelos Animales de Enfermedad , Epilepsia del Lóbulo Temporal/etiología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Hipocampo/enzimología , Masculino , Ratas , Ratas Sprague-Dawley
2.
Exp Eye Res ; 213: 108845, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34800480

RESUMEN

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.


Asunto(s)
Retinopatía Diabética/enzimología , Inhibidores Enzimáticos/farmacología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Gliceraldehído-3-Fosfato Deshidrogenasas/antagonistas & inhibidores , Interleucina-1beta/farmacología , Retina/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Adenosina Trifosfato/metabolismo , Animales , Western Blotting , Retinopatía Diabética/patología , Femenino , Glucosa/farmacología , Hiperglucemia/metabolismo , Ácido Yodoacético/farmacología , L-Lactato Deshidrogenasa/metabolismo , Metionina Sulfoximina/farmacología , Ratones , Ratones Endogámicos C57BL , Retina/enzimología , Retina/patología
3.
Oncol Rep ; 45(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33846803

RESUMEN

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.


Asunto(s)
Asparagina/metabolismo , Transformación Celular Neoplásica/metabolismo , Glutamina/deficiencia , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proliferación Celular/genética , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Inhibidores de Cisteína Proteinasa/farmacología , Regulación Neoplásica de la Expresión Génica , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Humanos , Leupeptinas/farmacología , Metionina Sulfoximina/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética
4.
Neurosci Lett ; 755: 135847, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33774150

RESUMEN

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.


Asunto(s)
Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Mediadores de Inflamación/antagonistas & inhibidores , Morfina/administración & dosificación , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Solución Salina/administración & dosificación , Médula Espinal/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos/fisiología , Transportador 2 de Aminoácidos Excitadores/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Hidrógeno/administración & dosificación , Mediadores de Inflamación/metabolismo , Inyecciones Intraperitoneales , Inyecciones Espinales , Masculino , Nitratos/antagonistas & inhibidores , Nitratos/metabolismo , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Médula Espinal/efectos de los fármacos
5.
Int J Mol Sci ; 22(4)2021 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-33567690

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Animales , Humanos , Neoplasias/enzimología , Neoplasias/patología
6.
BMC Cancer ; 21(1): 174, 2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33596851

RESUMEN

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.


Asunto(s)
Cisplatino/farmacología , Resistencia a Antineoplásicos , Silenciador del Gen , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamina/metabolismo , Metaboloma , Neoplasias Ováricas/tratamiento farmacológico , Antineoplásicos/farmacología , Apoptosis , Proliferación Celular , Reprogramación Celular , Femenino , Glutamato-Amoníaco Ligasa/genética , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Células Tumorales Cultivadas
7.
EMBO J ; 40(6): e106583, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33459428

RESUMEN

Plasmodium falciparum (Pf) is a major cause of human malaria and is transmitted by infected Anopheles mosquitoes. The initial asymptomatic infection is characterized by parasite invasion of hepatocytes, followed by massive replication generating schizonts with blood-infective merozoites. Hepatocytes can be categorized by their zonal location and metabolic functions within a liver lobule. To understand specific host conditions that affect infectivity, we studied Pf parasite liver stage development in relation to the metabolic heterogeneity of fresh human hepatocytes. We found selective preference of different Pf strains for a minority of hepatocytes, which are characterized by the particular presence of glutamine synthetase (hGS). Schizont growth is significantly enhanced by hGS uptake early in development, showcasing a novel import system. In conclusion, Pf development is strongly determined by the differential metabolic status in hepatocyte subtypes. These findings underscore the importance of detailed understanding of hepatocyte host-Pf interactions and may delineate novel pathways for intervention strategies.


Asunto(s)
Glutamato-Amoníaco Ligasa/metabolismo , Hepatocitos/parasitología , Malaria Falciparum/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Transporte Biológico/fisiología , Proliferación Celular/fisiología , Glucosa/metabolismo , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Humanos , Hígado/parasitología , Hígado/patología
8.
Microb Genom ; 6(11)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33074087

RESUMEN

We report on the combination of chemical mutagenesis, azithromycin selection and next-generation sequencing (Mut-Seq) for the identification of small nucleotide variants that decrease the susceptibility of Streptococcus pneumoniae to the macrolide antibiotic azithromycin. Mutations in the 23S ribosomal RNA or in ribosomal proteins can confer resistance to macrolides and these were detected by Mut-Seq. By concentrating on recurrent variants, we could associate mutations in genes implicated in the metabolism of glutamine with decreased azithromycin susceptibility among S. pneumoniae mutants. Glutamine synthetase catalyses the transformation of glutamate and ammonium into glutamine and its chemical inhibition is shown to sensitize S. pneumoniae to antibiotics. A mutation affecting the ribosomal-binding site of a putative ribonuclease J2 is also shown to confer low-level resistance. Mut-Seq has the potential to reveal chromosomal changes enabling high resistance as well as novel events conferring more subtle phenotypes.


Asunto(s)
Antibacterianos/farmacología , Azitromicina/farmacología , Farmacorresistencia Bacteriana/genética , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/genética , Endorribonucleasas/metabolismo , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Secuenciación de Nucleótidos de Alto Rendimiento , Pruebas de Sensibilidad Microbiana , Mutagénesis/genética , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Ribosómico 23S/genética
9.
Nature ; 587(7835): 626-631, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33116312

RESUMEN

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.


Asunto(s)
Glutamina/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Envejecimiento/metabolismo , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Glutamato Deshidrogenasa/deficiencia , Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/metabolismo , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Macrófagos/enzimología , Masculino , Ratones , Antígenos de Histocompatibilidad Menor/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Oxidación-Reducción , Células Satélite del Músculo Esquelético/citología , Serina-Treonina Quinasas TOR
10.
J Nat Prod ; 83(9): 2809-2813, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32830503

RESUMEN

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.


Asunto(s)
Antibacterianos/síntesis química , Arsenicales/síntesis química , Antibacterianos/farmacología , Arsenicales/farmacología , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos/farmacología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , S-Adenosilmetionina/antagonistas & inhibidores
11.
J Chem Theory Comput ; 16(7): 4694-4705, 2020 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-32551588

RESUMEN

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.


Asunto(s)
Glutamato-Amoníaco Ligasa/metabolismo , Nitratos/química , Tirosina/química , Adenosina Trifosfato/metabolismo , Sitios de Unión , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/genética , Humanos , Concentración de Iones de Hidrógeno , Cinética , Simulación de Dinámica Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Termodinámica
12.
Biochim Biophys Acta Mol Cell Res ; 1867(2): 118571, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31706909

RESUMEN

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.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Glutamina/metabolismo , Factor de Unión a CCAAT/genética , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Regulación de la Expresión Génica/efectos de los fármacos , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Glutamina/deficiencia , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Tapsigargina/farmacología , Factor de Transcripción CHOP/metabolismo
13.
BMC Plant Biol ; 19(1): 425, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31615403

RESUMEN

BACKGROUND: Nitrogen (N) nutrition significantly affected metabolism and accumulation of quality-related compounds in tea plant (Camellia sinensis L.). Little is known about the physiological and molecular mechanisms underlying the effects of short-term repression of N metabolism on tea roots and leaves for a short time. RESULTS: In this study, we subjected tea plants to a specific inhibitor of glutamine synthetase (GS), methionine sulfoximine (MSX), for a short time (30 min) and investigated the effect of the inhibition of N metabolism on the transcriptome and metabolome of quality-related compounds. Our results showed that GS activities in tea roots and leaves were significantly inhibited upon MSX treatment, and both tissue types showed a sensitive metabolic response to GS inhibition. In tea leaves, the hydrolysis of theanine decreased with the increase in theanine and free ammonium content. The biosynthesis of all other amino acids was repressed, and the content of N-containing lipids declined, suggesting that short-term inhibition of GS reduces the level of N reutilization in tea leaves. Metabolites related to glycolysis and the tricarboxylic acid (TCA) cycle accumulated after GS repression, whereas the content of amino acids such as glycine, serine, isoleucine, threonine, leucine, and valine declined in the MXS treated group. We speculate that the biosynthesis of amino acids is affected by glycolysis and the TCA cycle in a feedback loop. CONCLUSIONS: Overall, our data suggest that GS repression in tea plant leads to the reprogramming of amino acid and lipid metabolic pathways.


Asunto(s)
Aminoácidos/metabolismo , Camellia sinensis/metabolismo , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Metabolismo de los Lípidos , Metionina Sulfoximina/farmacología , Proteínas de Plantas/antagonistas & inhibidores , Camellia sinensis/efectos de los fármacos , Camellia sinensis/enzimología , Metabolismo de los Lípidos/efectos de los fármacos , Hojas de la Planta/efectos de los fármacos , Hojas de la Planta/metabolismo , Raíces de Plantas/efectos de los fármacos , Raíces de Plantas/metabolismo
14.
Cell Rep ; 29(5): 1287-1298.e6, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31665640

RESUMEN

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.


Asunto(s)
Carbono/metabolismo , Glutamina/metabolismo , Nitrógeno/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Carcinoma Ductal Pancreático/enzimología , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular , Femenino , Eliminación de Gen , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Humanos , Ácidos Cetoglutáricos/metabolismo , Masculino , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología
15.
Biotechnol Prog ; 35(5): e2856, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31148368

RESUMEN

Passaging and expansion of animal cells in lean maintenance medium could result in periods of limitation of some nutrients. Over time, such stresses could possibly result in selection of cells with metabolic changes and contribute to heterogeneity. Here, we investigate whether selection of Chinese Hamster Ovary (CHO) cells under glutamine limitation results in changes in growth under glutamine-replete conditions. In glutamine-limiting medium, compared to control cells passaged in glutamine-rich medium, the selected cells showed higher glutamine synthetase (GS) activity and attained a higher peak viable cell density (PVCD). Surprisingly, in glutamine-replete conditions, selected cells still showed a higher GS activity but a lower PVCD. We show that in glutamine-replete medium, PVCD of selected cells was restored on (a) inhibition of GS activity with methionine sulfoximine, (b) supplementation of aspartate-without affecting GS activity, and (c) supplementation of serine, which is reported to inhibit GS in vitro. Consistent with the reported effect of serine, inhibition of GS activity was observed upon serine supplementation along with reduced growth of cells under glutamine-limiting conditions. The latter observation is important for the design of glutamine-free culture medium and feed used for GS-CHO and GS-NS0. In summary, we show that CHO cells selected under glutamine limitation have superfluous GS activity in glutamine-replete medium, which negatively affects their PVCD. This may be due to its effect on availability of aspartate which was the limiting nutrient for the growth of selected cells in glutamine-replete conditions.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Glutamato-Amoníaco Ligasa , Glutamina/metabolismo , Serina/metabolismo , Animales , Células CHO , Isótopos de Carbono/análisis , Isótopos de Carbono/metabolismo , Cricetinae , Cricetulus , Medios de Cultivo/química , Medios de Cultivo/metabolismo , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Glutamina/análisis , Metionina Sulfoximina
16.
Planta ; 249(6): 1837-1849, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30850862

RESUMEN

MAIN CONCLUSION: Glufosinate is primarily toxic to plants due to a massive light-dependent generation of reactive oxygen species rather than ammonia accumulation or carbon assimilation inhibition. Glutamine synthetase (GS) plays a key role in plant nitrogen metabolism and photorespiration. Glufosinate (C5H12NO4P) targets GS and causes catastrophic consequences leading to rapid plant cell death, and the causes for phytoxicity have been attributed to ammonia accumulation and carbon assimilation restriction. This study aimed to examine the biochemical and physiological consequences of GS inhibition to identify the actual cause for rapid phytotoxicity. Monocotyledonous and dicotyledonous species with different forms of carbon assimilation (C3 versus C4) were selected as model plants. Glufosinate sensitivity was proportional to the uptake of herbicide between species. Herbicide uptake also correlated with the level of GS inhibition and ammonia accumulation in planta even with all species having the same levels of enzyme sensitivity in vitro. Depletion of both glutamine and glutamate occurred in glufosinate-treated leaves; however, amino acid starvation would be expected to cause a slow plant response. Ammonia accumulation in response to GS inhibition, often reported as the driver of glufosinate phytotoxicity, occurred in all species, but did not correlate with either reductions in carbon assimilation or cell death. This is supported by the fact that plants can accumulate high levels of ammonia but show low inhibition of carbon assimilation and absence of phytotoxicity. Glufosinate-treated plants showed a massive light-dependent generation of reactive oxygen species, followed by malondialdehyde accumulation. Consequently, we propose that glufosinate is toxic to plants not because of ammonia accumulation nor carbon assimilation inhibition, but the production of reactive oxygen species driving the catastrophic lipid peroxidation of the cell membranes and rapid cell death.


Asunto(s)
Amaranthus/efectos de los fármacos , Aminobutiratos/toxicidad , Bassia scoparia/efectos de los fármacos , Herbicidas/toxicidad , Poaceae/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Amaranthus/metabolismo , Amaranthus/efectos de la radiación , Aminobutiratos/efectos de la radiación , Amoníaco/metabolismo , Bassia scoparia/metabolismo , Bassia scoparia/efectos de la radiación , Carbono/metabolismo , Muerte Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Herbicidas/efectos de la radiación , Luz , Peroxidación de Lípido/efectos de los fármacos , Hojas de la Planta/efectos de los fármacos , Hojas de la Planta/metabolismo , Hojas de la Planta/efectos de la radiación , Proteínas de Plantas/antagonistas & inhibidores , Poaceae/metabolismo , Poaceae/efectos de la radiación , Especies Reactivas de Oxígeno/efectos de la radiación
17.
Sci Rep ; 9(1): 252, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30670758

RESUMEN

Glutamatergic synapses constitute a major excitatory neurotransmission system and are regulated by glutamate/glutamine (Gln) cycling between neurons and astrocytes. Gln synthetase (GS) produced by astrocytes plays an important role in maintaining the cycle. However, the significance of GS during synaptogenesis has not been clarified. GS activity and expression significantly increase from postnatal day (PD) 7 to 21, and GS is expressed prior to glial fibrillary acidic protein (GFAP) and is more abundant than GFAP throughout synaptogenesis. These observations suggest that GS plays an important role in synaptogenesis. We investigated this by inhibiting GS activity in neonatal mice and assessed the consequences in adult animals. Lower expression levels of GS and GFAP were found in the CA3 region of the hippocampus but not in the CA1 region. Moreover, synaptic puncta and glutamatergic neurotransmission were also decreased in CA3. Behaviorally, mice with inhibited GS during synaptogenesis showed spatial memory-related impairment as adults. These results suggest that postnatal GS activity is important for glutamatergic synapse development in CA3.


Asunto(s)
Glutamato-Amoníaco Ligasa/metabolismo , Neurogénesis/fisiología , Memoria Espacial/fisiología , Transmisión Sináptica/fisiología , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Técnicas de Observación Conductual , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Región CA3 Hipocampal/crecimiento & desarrollo , Región CA3 Hipocampal/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Masculino , Metionina Sulfoximina/farmacología , Ratones , Modelos Animales , Neurogénesis/efectos de los fármacos , Neuronas/metabolismo , Memoria Espacial/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
18.
Gut ; 68(10): 1872-1883, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30580251

RESUMEN

OBJECTIVE: Acute-on-chronic liver failure (ACLF) is associated with dysfunctional circulating monocytes whereby patients become highly susceptible to bacterial infections. Here, we identify the pathways underlying monocyte dysfunction in ACLF and we investigate whether metabolic rewiring reinstates their phagocytic and inflammatory capacity. DESIGN: Following phenotypic characterisation, we performed RNA sequencing on CD14+CD16- monocytes from patients with ACLF and decompensated alcoholic cirrhosis. Additionally, an in vitro model mimicking ACLF patient-derived features was implemented to investigate the efficacy of metabolic regulators on monocyte function. RESULTS: Monocytes from patients with ACLF featured elevated frequencies of interleukin (IL)-10-producing cells, reduced human leucocyte antigen DR isotype (HLA-DR) expression and impaired phagocytic and oxidative burst capacity. Transcriptional profiling of isolated CD14+CD16- monocytes in ACLF revealed upregulation of an array of immunosuppressive parameters and compromised antibacterial and antigen presentation machinery. In contrast, monocytes in decompensated cirrhosis showed intact capacity to respond to inflammatory triggers. Culturing healthy monocytes in ACLF plasma mimicked the immunosuppressive characteristics observed in patients, inducing a blunted phagocytic response and metabolic program associated with a tolerant state. Metabolic rewiring of the cells using a pharmacological inhibitor of glutamine synthetase, partially restored the phagocytic and inflammatory capacity of in vitro generated- as well as ACLF patient-derived monocytes. Highlighting its biological relevance, the glutamine synthetase/glutaminase ratio of ACLF patient-derived monocytes positively correlated with disease severity scores. CONCLUSION: In ACLF, monocytes feature a distinct transcriptional profile, polarised towards an immunotolerant state and altered metabolism. We demonstrated that metabolic rewiring of ACLF monocytes partially revives their function, opening up new options for therapeutic targeting in these patients.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada/tratamiento farmacológico , Infecciones Bacterianas/tratamiento farmacológico , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Inmunosupresores/uso terapéutico , Monocitos/enzimología , Insuficiencia Hepática Crónica Agudizada/inmunología , Insuficiencia Hepática Crónica Agudizada/metabolismo , Adulto , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Células Cultivadas , Citocinas/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Fagocitosis , Estudios Retrospectivos
19.
Tuberculosis (Edinb) ; 108: 106-113, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29523309

RESUMEN

Tuberculosis remains a major health problem accentuated by the rise of resistance to all available drugs. Therefore, this study was launched to discover a novel antituberculosis agent from wild Egyptian Sahara plants. Twelve such plants were screened, in vitro, for their activity against various Mycobacterium species. The most active plant, Euphorbia paralias, was further fractionated with different organic solvents, and the activity of the obtained fractions was determined by the agar diffusion and broth microdilution methods. The methanol fraction was the most active against Mycobacterium spp., and was non-toxic in doses up to 10 g/kg of animal weight. Its main component was separated by column chromatography, and then identified by ultraviolet spectroscopy and nuclear magnetic resonance analysis as quercetin-3-O-ß-D-glucoside. Docking analysis suggested that quercetin-3-O-ß-D-glucoside inhibits the glutamine synthetase enzyme, a promising target for the development of antituberculosis drugs. This prediction was confirmed by an in vitro glutamine synthetase biosynthetic assay. To the best of our knowledge, and based on bioinformatics mining of the BioPhytMol database, this is the first report on the antimycobacterial activity of Euphorbia paralias plant. It is also the first report on the inhibition of mycobacterial glutamine synthetase by the flavonoid quercetin.


Asunto(s)
Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Euphorbia , Glucósidos/farmacología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Mycobacterium/efectos de los fármacos , Quercetina/análogos & derivados , Antituberculosos/química , Antituberculosos/aislamiento & purificación , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Egipto , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Euphorbia/química , Glucósidos/química , Glucósidos/aislamiento & purificación , Glutamato-Amoníaco Ligasa/química , Glutamato-Amoníaco Ligasa/metabolismo , Espectroscopía de Resonancia Magnética , Metanol/química , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Mycobacterium/enzimología , Fitoterapia , Plantas Medicinales , Conformación Proteica , Quercetina/química , Quercetina/aislamiento & purificación , Quercetina/farmacología , Solventes/química , Espectrofotometría Ultravioleta , Relación Estructura-Actividad
20.
J Cancer Res Clin Oncol ; 144(5): 821-833, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29435734

RESUMEN

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.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/genética , Proliferación Celular/genética , Glutamato-Amoníaco Ligasa/genética , Antígenos de Histocompatibilidad Menor/genética , Neoplasias Gástricas/genética , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Benceno/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Glutamina/antagonistas & inhibidores , Glutamina/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Antígenos de Histocompatibilidad Menor/metabolismo , Terapia Molecular Dirigida/métodos , Serina/administración & dosificación , Serina/química , Serina/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...