Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 298(2): 101564, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34999118

RESUMEN

The mitochondrial enzyme glutaminase C (GAC) is upregulated in many cancer cells to catalyze the first step in glutamine metabolism, the hydrolysis of glutamine to glutamate. The dependence of cancer cells on this transformed metabolic pathway highlights GAC as a potentially important therapeutic target. GAC acquires maximal catalytic activity upon binding to anionic activators such as inorganic phosphate. To delineate the mechanism of GAC activation, we used the tryptophan substitution of tyrosine 466 in the catalytic site of the enzyme as a fluorescent reporter for glutamine binding in the presence and absence of phosphate. We show that in the absence of phosphate, glutamine binding to the Y466W GAC tetramer exhibits positive cooperativity. A high-resolution X-ray structure of tetrameric Y466W GAC bound to glutamine suggests that cooperativity in substrate binding is coupled to tyrosine 249, located at the edge of the catalytic site (i.e., the "lid"), adopting two distinct conformations. In one dimer within the GAC tetramer, the lids are open and glutamine binds weakly, whereas, in the adjoining dimer, the lids are closed over the substrates, resulting in higher affinity interactions. When crystallized in the presence of glutamine and phosphate, all four subunits of the Y466W GAC tetramer exhibited bound glutamine with closed lids. Glutamine can bind with high affinity to each subunit, which subsequently undergo simultaneous catalysis. These findings explain how the regulated transitioning of GAC between different conformational states ensures that maximal catalytic activity is reached in cancer cells only when an allosteric activator is available.


Asunto(s)
Glutaminasa , Glutamina , Mitocondrias , Dominio Catalítico , Glutaminasa/química , Glutaminasa/metabolismo , Glutamina/química , Glutamina/metabolismo , Mitocondrias/enzimología , Mitocondrias/metabolismo , Fosfatos/química , Fosfatos/metabolismo , Conformación Proteica , Tirosina/química , Tirosina/metabolismo
2.
J Biol Chem ; 298(2): 101535, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34954143

RESUMEN

Cancer cells frequently exhibit uncoupling of the glycolytic pathway from the TCA cycle (i.e., the "Warburg effect") and as a result, often become dependent on their ability to increase glutamine catabolism. The mitochondrial enzyme Glutaminase C (GAC) helps to satisfy this 'glutamine addiction' of cancer cells by catalyzing the hydrolysis of glutamine to glutamate, which is then converted to the TCA-cycle intermediate α-ketoglutarate. This makes GAC an intriguing drug target and spurred the molecules derived from bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide (the so-called BPTES class of allosteric GAC inhibitors), including CB-839, which is currently in clinical trials. However, none of the drugs targeting GAC are yet approved for cancer treatment and their mechanism of action is not well understood. Here, we shed new light on the underlying basis for the differential potencies exhibited by members of the BPTES/CB-839 family of compounds, which could not previously be explained with standard cryo-cooled X-ray crystal structures of GAC bound to CB-839 or its analogs. Using an emerging technique known as serial room temperature crystallography, we were able to observe clear differences between the binding conformations of inhibitors with significantly different potencies. We also developed a computational model to further elucidate the molecular basis of differential inhibitor potency. We then corroborated the results from our modeling efforts using recently established fluorescence assays that directly read out inhibitor binding to GAC. Together, these findings should aid in future design of more potent GAC inhibitors with better clinical outlook.


Asunto(s)
Inhibidores Enzimáticos , Glutaminasa , Neoplasias , Sulfuros , Tiadiazoles , Cristalografía , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glutaminasa/antagonistas & inhibidores , Glutaminasa/química , Glutaminasa/metabolismo , Glutamina/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Sulfuros/química , Sulfuros/farmacología , Temperatura , Tiadiazoles/química , Tiadiazoles/farmacología
3.
Haematologica ; 108(2): 409-419, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35979719

RESUMEN

Asparagine is a non-essential amino acid since it can either be taken up via the diet or synthesized by asparagine synthetase. Acute lymphoblastic leukemia (ALL) cells do not express asparagine synthetase or express it only minimally, which makes them completely dependent on extracellular asparagine for their growth and survival. This dependency makes ALL cells vulnerable to treatment with L-asparaginase, an enzyme that hydrolyzes asparagine. To date, all clinically approved L-asparaginases have significant L-glutaminase co-activity, associated with non-immune related toxic side effects observed during therapy. Therefore, reduction of L-glutaminase co-activity with concomitant maintenance of its anticancer L-asparaginase effect may effectively improve the tolerability of this unique drug. Previously, we designed a new alternative variant of Erwinia chrysanthemi (ErA; Erwinaze) with decreased L-glutaminase co-activity, while maintaining its L-asparaginase activity, by the introduction of three key mutations around the active site (ErA-TM). However, Erwinaze and our ErA-TM variant have very short half-lives in vivo. Here, we show that the fusion of ErA-TM with an albumin binding domain (ABD)-tag significantly increases its in vivo persistence. In addition, we evaluated the in vivo therapeutic efficacy of ABD-ErA-TM in a B-ALL xenograft model of SUP-B15. Our results show a comparable long-lasting durable antileukemic effect between the standard-of-care pegylated-asparaginase and ABD-ErA-TM L-asparaginase, but with fewer co-glutaminase-related acute side effects. Since the toxic side effects of current L-asparaginases often result in treatment discontinuation in ALL patients, this novel ErA-TM variant with ultra-low L-glutaminase co-activity and long in vivo persistence may have great clinical potential.


Asunto(s)
Aspartatoamoníaco Ligasa , Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Asparaginasa/farmacología , Asparaginasa/uso terapéutico , Glutaminasa/química , Glutaminasa/genética , Glutaminasa/metabolismo , Asparagina , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia Mieloide Aguda/tratamiento farmacológico
4.
J Sci Food Agric ; 103(9): 4562-4572, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36853147

RESUMEN

BACKGROUND: Protein glutaminase (PG) is a novel protein modification biotechnology that is increasingly being used in the food industry. However, the current level of fermentation of PG-producing strains still does not meet the requirements of industrial production. To obtain the mutant strains with high PG production, the atmospheric and room temperature plasma (ARTP) combined with LiCl chemical mutagen were used in mutagenesis of a PG producing Chryseobacterium proteolyticum 1003. RESULTS: A mutant strain (WG15) was successfully obtained based on malonic acid resistance screening after compound mutagenesis of the starting strain C. proteolyticum 1003 using ARTP with LiCl, and it was confirmed to be genetically stable in PG synthesis after 15 generations. The protein glutaminase production of WG15 was 2.91 U mL-1 after optimization of fermentation conditions, which is 48.69% higher than the original strain C. proteolyticum 1003. The PG obtained from fermentation showed good activities in deamidation of soy protein isolate. The solubility and foaming properties of the PG-treated soy protein isolate were significantly increased by 36.50% and 10.03%, respectively, when PG was added at the amount of 100 U mL-1 . In addition, the emulsifying activity and emulsion stability of the treated soy protein isolate were improved by 12.44% and 10.34%, respectively, on the addition of 10 U mL-1 PG. The secondary structure of the soy protein isolate changed after PG treatment, with an increased proportion of glutamate. CONCLUSION: The results of the present study indicate that the PG produced by this mutant strain could improve the functional properties of soybean protein isolate and the C. proteolyticum mutant WG15 has great potential in food industry. © 2023 Society of Chemical Industry.


Asunto(s)
Chryseobacterium , Glutaminasa , Glutaminasa/química , Proteínas de Soja/química , Chryseobacterium/metabolismo , Mutagénesis
5.
Chembiochem ; 23(9): e202100555, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-34762772

RESUMEN

The catalytic mechanism of Pdx2 was studied with atomic detail employing the computational ONIOM hybrid QM/MM methodology. Pdx2 employs a Cys-His-Glu catalytic triad to deaminate glutamine to glutamate and ammonia - the source of the nitrogen of pyridoxal 5'-phosphate (PLP). This enzyme is, therefore, a rate-limiting step in the PLP biosynthetic pathway of Malaria and Tuberculosis pathogens that rely on this mechanism to obtain PLP. For this reason, Pdx2 is considered a novel and promising drug target to treat these diseases. The results obtained show that the catalytic mechanism of Pdx2 occurs in six steps that can be divided into four stages: (i) activation of Cys87 , (ii) deamination of glutamine with the formation of the glutamyl-thioester intermediate, (iii) hydrolysis of the formed intermediate, and (iv) enzymatic turnover. The kinetic data available in the literature (19.1-19.5 kcal mol-1 ) agree very well with the calculated free energy barrier of the hydrolytic step (18.2 kcal.mol-11 ), which is the rate-limiting step of the catalytic process when substrate is readily available in the active site. This catalytic mechanism differs from other known amidases in three main points: i) it requires the activation of the nucleophile Cys87 to a thiolate; ii) the hydrolysis occurs in a single step and therefore does not require the formation of a second tetrahedral reaction intermediate, as it is proposed, and iii) Glu198 does not have a direct role in the catalytic process. Together, these results can be used for the synthesis of new transition state analogue inhibitors capable of inhibiting Pdx2 and impair diseases like Malaria and Tuberculosis.


Asunto(s)
Glutaminasa , Malaria , Catálisis , Ácido Glutámico , Glutaminasa/química , Glutaminasa/metabolismo , Glutamina/metabolismo , Humanos , Fosfato de Piridoxal/química
6.
Protein Expr Purif ; 190: 106006, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34742913

RESUMEN

l-asparaginase is a chemotherapeutic drug used in the treatment of acute lymphoblastic leukemia, a malignant disorder in children. l-asparaginase helps in removing acrylamide found in fried and baked foods which is carcinogenic in nature. The search for new therapeutic enzymes is of great interest in both medical and food applications. The present work aims to isolate the intracellular l-asparaginase from endophytic fungi Chaetomium sp. The intracellular enzyme was partially purified by chromatographic techniques. Molecular weight of enzyme was found to be ~66 kDa by SDS PAGE analysis. The enzyme is highly specific for l-asparagine and did not show glutaminase and urease activity. Maximum enzyme activity was found to be 58 ± 5 U/mL at 40 °C, pH 7.0 with 2 µg of protein. Intracellular l-asparaginase from Chaetomium sp. exhibited anticancer activity on human blood cancer (MOLT-4) cells.


Asunto(s)
Antineoplásicos , Asparaginasa , Chaetomium/enzimología , Proteínas Fúngicas , Glutaminasa/química , Ureasa/química , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Asparaginasa/química , Asparaginasa/aislamiento & purificación , Asparaginasa/farmacología , Línea Celular Tumoral , Proteínas Fúngicas/química , Proteínas Fúngicas/aislamiento & purificación , Proteínas Fúngicas/farmacología , Humanos
7.
World J Microbiol Biotechnol ; 38(11): 204, 2022 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-36002753

RESUMEN

L-Glutaminases are enzymes that catalyze the cleavage of the gamma-amido bond of L-glutamine residues, producing ammonia and L-glutamate. These enzymes have several applications in food and pharmaceutical industries. However, the L-glutaminases that hydrolyze free L-glutamine (L-glutamine glutaminases, EC 3.5.1.2) have different structures and properties with respect to the L-glutaminases that hydrolyze the same amino acid covalently bound in peptides (peptidyl glutaminases, EC 3.5.1.43) and proteins (protein-glutamine glutaminase, EC 3.5.1.44). In the food industry, L-glutamine glutaminases are applied to enhance the flavor of foods, whereas protein glutaminases are useful to improve the functional properties of proteins. This review will focus on structural backgrounds and differences between these enzymes, the methodology available to measure the activity as well as strengths and limitations. Production methods, applications, and challenges in the food industry will be also discussed. This review will provide useful information to search and identify the suitable L-glutaminase that best fits to the intended application.


Asunto(s)
Glutaminasa , Glutamina , Catálisis , Industria de Alimentos , Ácido Glutámico/metabolismo , Glutaminasa/química , Glutaminasa/metabolismo , Glutamina/metabolismo
8.
J Biol Chem ; 295(5): 1328-1337, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31871054

RESUMEN

The glutaminase C (GAC) isoform of mitochondrial glutaminase is overexpressed in many cancer cells and therefore represents a potential therapeutic target. Understanding the regulation of GAC activity has been guided by the development of spectroscopic approaches that measure glutaminase activity in real time. Previously, we engineered a GAC protein (GAC(F327W)) in which a tryptophan residue is substituted for phenylalanine in an activation loop to explore the role of this loop in enzyme activity. We showed that the fluorescence emission of Trp-327 is enhanced in response to activator binding, but quenched by inhibitors of the BPTES class that bind to the GAC tetramer and contact the activation loop, thereby constraining it in an inactive conformation. In the present work, we took advantage of a tryptophan substitution at position 471, proximal to the GAC catalytic site, to examine the conformational coupling between the activation loop and the substrate-binding cleft, separated by ∼16 Å. Comparison of glutamine binding in the presence or absence of the BPTES analog CB-839 revealed a reciprocal relationship between the constraints imposed on the activation loop position and the affinity of GAC for substrate. Binding of the inhibitor weakened the affinity of GAC for glutamine, whereas activating anions such as Pi increased this affinity. These results indicate that the conformations of the activation loop and the substrate-binding cleft in GAC are allosterically coupled and that this coupling determines substrate affinity and enzymatic activity and explains the activities of CB-839, which is currently in clinical trials.


Asunto(s)
Bencenoacetamidas/farmacología , Glutaminasa/química , Glutamina/metabolismo , Mitocondrias/enzimología , Tiadiazoles/farmacología , Regulación Alostérica/genética , Sitio Alostérico/genética , Sustitución de Aminoácidos/genética , Animales , Ingeniería Biomédica , Dominio Catalítico/genética , Glutaminasa/metabolismo , Cinética , Ratones , Mitocondrias/química , Modelos Moleculares , Mutación , Isoformas de Proteínas , Estructura Terciaria de Proteína/genética , Proteínas Recombinantes , Sulfuros/farmacología
9.
Int Microbiol ; 24(2): 169-181, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33389217

RESUMEN

L-Glutaminase is an amidohydrolase which can act as a vital chemotherapeutic agent against various malignancies. In the present work, L-glutaminase productivity from Aspergillus versicolor Faesay4 was significantly increased by 7.72-fold (from 12.33 ± 0.47 to 95.15 ± 0.89 U/mL) by optimizing submerged fermentation parameters in Czapek's Dox (CZD) medium including an incubation period from 3 (12.33 ± 0.47 U/mL) to 6 days (23.36 ± 0.58 U/mL), an incubation temperature from 30 °C (23.36 ± 0.49 U/mL) to 25 °C (31.08 ± 0.60 U/mL), initial pH from pH 5.0 (8.49 ± 0.21 U/mL)  to pH 7.0 (32.18 ± 0.57 U/mL), replacement of glucose (30.19 ± 0.52 U/mL) by sucrose (48.97 ± 0.67 U/mL) as the carbon source at a concentration of 2.0% (w/v), increasing glutamine concentration as the nitrogen source from 1.0% (w/v, 48.54 ± 0.48 U/mL) to 1.5% (w/v, 63.01 ± 0.60 U/mL), and addition of a mixture of KH2PO4 and NaCl (0.5% w/v for both) to SZD as the metal supplementation (95.15 ± 0.89 U/mL). Faesay4 L-glutaminase was purified to yield total activity 13,160 ± 22.76 (U), specific activity 398.79 ± 9.81 (U/mg of protein), and purification fold 2.1 ± 3.18 with final enzyme recovery 57.22 ± 2.17%. The pure enzyme showed a molecular weight of 61.80 kDa, and it was stable and retained 100.0% of its activity at a temperature ranged from 10 to 40 °C and pH 7.0. In our trials, to increase the enzyme activity by optimizing the assay conditions (which were temperature 60 °C, pH 7.0, substrate glutamine, substrate concentration 1.0%, and reaction time 60 min), the enzyme activity increased by 358.8% after changing the assay temperature from 60 to 30 °C and then increased by 138% after decreasing the reaction time from 60 to 40 min. However, both pH 7.0 and glutamine as the substrate remain the best assay parameters for the L-glutaminase activity. When the glutamine in the assay as the reaction substrate was replaced by asparagine, lysine, proline, methionine, cysteine, glycine, valine, phenylalanine, L-alanine, aspartic acid, tyrosine, and serine, the enzyme lost 23.86%, 29.0%, 31.0%, 48.3%, 50.0%, 73.6%, 74.51%, 80.42%, 82.5%, 83.43%, 88.36%, and 89.78% of its activity with glutamine, respectively. Furthermore, Mn2+, K+, Na+, and Fe3+ were enzymatic activators that increased the L-glutaminase activity by 25.0%, 18.05%, 10.97%, and 8.0%, respectively. Faesay4 L-glutaminase was characterized as a serine protease enzyme as a result of complete inhibition by all serine protease inhibitors (PMSF, benzamidine, and TLCK). Purified L-glutaminase isolated from Aspergillus versicolor Faesay4 showed potent DPPH scavenging activities with IC50 = 50 µg/mL and anticancer activities against human liver (HepG-2), colon (HCT-116), breast (MCF-7), lung (A-549), and cervical (Hela) cancer cell lines with IC50 39.61, 12.8, 6.18, 11.48, and 7.25 µg/mL, respectively.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Antioxidantes/aislamiento & purificación , Aspergillus/enzimología , Proteínas Fúngicas/química , Proteínas Fúngicas/aislamiento & purificación , Glutaminasa/química , Glutaminasa/aislamiento & purificación , Antineoplásicos/farmacología , Antioxidantes/química , Antioxidantes/farmacología , Aspergillus/química , Aspergillus/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Estabilidad de Enzimas , Proteínas Fúngicas/farmacología , Glutaminasa/farmacología , Humanos , Concentración de Iones de Hidrógeno , Cinética , Peso Molecular , Especificidad por Sustrato
10.
Mol Divers ; 25(2): 1077-1089, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-32328963

RESUMEN

Glutaminase plays an important role in carcinogenesis and cancer cell growth. This biological target is interesting against cancer cells. Therefore, in this work, in silico [docking and molecular dynamics (MD) simulations] and in vitro methods (antiproliferative and LC-MS metabolomics) were employed to assay a hybrid compound derived from glutamine and valproic acid (Gln-VPA), which was compared with 6-diazo-5-oxo-L-norleucine (DON, a glutaminase inhibitor) and VPA (contained in Gln-VPA structure). Docking results from some snapshots retrieved from MD simulations show that glutaminase recognized Gln-VPA and DON. Additionally, Gln-VPA showed antiproliferative effects in HeLa cells and inhibited glutaminase activity. Finally, the LC-MS-based metabolomics studies on HeLa cells treated with either Gln-VPA (IC60 = 8 mM) or DON (IC50 = 3.5 mM) show different metabolomics behaviors, suggesting that they modulate different biological targets of the cell death mechanism. In conclusion, Gln-VPA is capable of interfering with more than one pharmacological target of cancer, making it an interesting drug that can be used to avoid multitherapy of classic anticancer drugs.


Asunto(s)
Antineoplásicos , Glutamina , Ácido Valproico , Antineoplásicos/química , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromatografía Liquida , Glutaminasa/antagonistas & inhibidores , Glutaminasa/química , Glutamina/química , Glutamina/farmacología , Células HeLa , Humanos , Espectrometría de Masas , Metaboloma/efectos de los fármacos , Metabolómica , Modelos Moleculares , Ácido Valproico/química , Ácido Valproico/farmacología
11.
Prep Biochem Biotechnol ; 51(3): 277-288, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32921254

RESUMEN

L-asparaginase (ASNase) is an essential drug in the treatment of acute lymphoblastic leukemia (ALL). Commercial bacterial ASNases increase patient survival, but the consequent immunological reactions remain a challenge. Yeasts ASNase is closer to human congeners and could lead to lower side effects. Among 134 yeast strains isolated from marine-sediments in King George Island, Antarctica, nine were L-asparaginase producing yeasts and glutaminase-free. Leucosporidium muscorum CRM 1648 yielded the highest ASNase activity (490.41 U.L-1) and volumetric productivity (5.12 U.L-1 h-1). Sucrose, yeast extract and proline were the best carbon and nitrogen sources to support growth and ASNase production. A full factorial design analysis pointed the optimum media condition for yeast growth and ASNase yield: 20 g L-1 sucrose, 15 g L-1 yeast extract and 20 g L-1 proline, which resulted in 4582.5 U L-1 and 63.64 U L-1 h-1 of ASNase and volumetric productivity, respectively. Analysis of temperature, pH, inoculum and addition of seawater indicated the best condition for ASNase production by this yeast: 12-15 °C, pH 5.5-6.5 and seawater >25% (v/v). Inoculum concentration seems not to interfere. This work is pioneer on the production of ASNase by cold-adapted yeasts, highlighting the potential of these microbial resources as a source of glutaminase-free L-asparaginase for commercial purposes.


Asunto(s)
Asparaginasa/química , Basidiomycota/metabolismo , Biotecnología/métodos , Sedimentos Geológicos/química , Glutaminasa/química , Regiones Antárticas , Antineoplásicos/farmacología , Biomasa , Carbono/química , Geografía , Concentración de Iones de Hidrógeno , Prolina/química , Análisis de Regresión , Agua de Mar , Sacarosa/química , Temperatura
12.
J Sci Food Agric ; 101(4): 1301-1306, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-32790072

RESUMEN

BACKGROUND: L-Glutaminase is considered to be an important industrial enzyme in both the pharmaceutical and food industries, especially for producing functional glutamyl compounds, such as l-theanine. Pseudomonas nitroreducens SP.001 with intracellular l-glutaminase activity has been screened previously. In the present study, three physical permeabilization methods were used to improve l-glutaminase activity. Then, the whole-cell immobilization conditions of permeabilized cells using sodium alginate as an embedding agent were optimized to enhance the enzyme's stability and reusability. The characteristics of the immobilized cells were investigated in comparison with those of permeabilized cells. RESULTS: The results obtained showed that cell permeabilization using osmotic shock with 155 g L-1 sucrose markedly improved enzyme activity. Then, an effective procedure for immobilization of permeabilized P. nitroreducens cells was established. The optimum conditions for cell immobilization were: sodium alginate 40 g L-1 , calcium chloride 30 g L-1 , cell mass 100 g L-1 and a curing time of 3 h. After successful immobilization, characterization studies revealed that the thermostability and pH resistance of l-glutaminase from immobilized cells were enhanced compared to those from permeabilized cells. Moreover, the immobilized biocatalyst could be reused up to 10 times and retained 80% of its activity. CONCLUSION: The stability and reusability of the permeabilized cells were improved through the immobilization. These findings indicated that immobilized whole-cell l-glutaminase from P. nitroreducens SP.001 possesses more potential for various industrial biotechnological applications than free cells. © 2020 Society of Chemical Industry.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glutaminasa/metabolismo , Pseudomonas/enzimología , Alginatos/química , Proteínas Bacterianas/química , Biocatálisis , Células Inmovilizadas/química , Células Inmovilizadas/enzimología , Glutamatos/metabolismo , Glutaminasa/química , Pseudomonas/química , Pseudomonas/crecimiento & desarrollo
13.
Bioorg Med Chem Lett ; 30(11): 127134, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32253062

RESUMEN

The rate of drug-induced proliferation (DIP) has been proposed as an unbiased alternative drug effect metric. However, current assays are not easy and precise enough to track minor changes in cell growth. Here, we report the optimized EZMTT based detection method which can continuously measure time-dependent growth after drug treatment and reliably detect partial drug resistance for cancer cells. Importantly, tracking time-dependent growth after drug treatment demonstrated that a KGA allosteric inhibitor alone failed to completely inhibit cancer cell growth, but a drug combination was able to provide complete inhibition in cell-based assays that translated well in in vivo animal experiments. In conclusion, this simple EZMTT method provided precise measurement of loss of susceptibility after drug treatment and has great potential to be developed for drug efficacy and drug combination studies to solve the unmet medical need.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Regulación Alostérica/efectos de los fármacos , Antineoplásicos/química , Línea Celular Tumoral , Sinergismo Farmacológico , Glutaminasa/química , Glutaminasa/metabolismo , Humanos , Concentración 50 Inhibidora , Paclitaxel/química , Paclitaxel/farmacología , Sirolimus/química , Sirolimus/farmacología
14.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-32825141

RESUMEN

Plasmodium species are protozoan parasites causing the deadly malaria disease. They have developed effective resistance mechanisms against most antimalarial medication, causing an urgent need to identify new antimalarial drug targets. Ideally, new drugs would be generated to specifically target the parasite with minimal or no toxicity to humans, requiring these drug targets to be distinctly different from the host's metabolic processes or even absent in the host. In this context, the essential presence of vitamin B6 biosynthesis enzymes in Plasmodium, the pyridoxal phosphate (PLP) biosynthesis enzyme complex, and its absence in humans is recognized as a potential drug target. To characterize the PLP enzyme complex in terms of initial drug discovery investigations, we performed structural analysis of the Plasmodium vivax PLP synthase domain (Pdx1), glutaminase domain (Pdx2), and Pdx1-Pdx2 (Pdx) complex (PLP synthase complex) by utilizing complementary bioanalytical techniques, such as dynamic light scattering (DLS), X-ray solution scattering (SAXS), and electron microscopy (EM). Our investigations revealed a dodecameric Pdx1 and a monodispersed Pdx complex. Pdx2 was identified in monomeric and in different oligomeric states in solution. Interestingly, mixing oligomeric and polydisperse Pdx2 with dodecameric monodisperse Pdx1 resulted in a monodispersed Pdx complex. SAXS measurements revealed the low-resolution dodecameric structure of Pdx1, different oligomeric structures for Pdx2, and a ring-shaped dodecameric Pdx1 decorated with Pdx2, forming a heteromeric 24-meric Pdx complex.


Asunto(s)
Glutaminasa/química , Simulación de Dinámica Molecular , Plasmodium vivax/enzimología , Multimerización de Proteína , Proteínas Protozoarias/química , Sitios de Unión , Glutaminasa/metabolismo , Unión Proteica , Proteínas Protozoarias/metabolismo , Fosfato de Piridoxal/biosíntesis , Vitamina B 6/biosíntesis
15.
Prep Biochem Biotechnol ; 50(4): 408-418, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31846380

RESUMEN

L-glutaminase from bacterial sources has been proven to be effective and economical agents in cancer therapy, food industry and high-value chemicals like threonine. In the present study, a newly isolated bacterial strain was potentially producing extracellular L-glutaminase, it identified as Bacillus subtilis OHEM11 (MK389501) using the 16S rRNA gene. L-glutaminase production optimized and the optimum factors for production under submerged fermentation were at pH 6.5-7.0 and 35 °C after 28 hr using rhamnose and glutamine as carbon and nitrogen sources, respectively, while bagasse was the best inducer for the production under solid-state fermentation. Ethanol precipitation and ion-exchange chromatography using QFF are the purification steps. L-glutaminase was purified to 2-fold with specific activity 89.78 U/mg and its molecular weight about 54.8 kDa with the alkaline property of the enzyme makes it clear having carcinostatic property; maximum enzyme activity at pH 8.2 and 40 °C and retained about 90% activity for 1 hr. The cytotoxicity effect of L-glutaminase indicated a significant safety on Vero cells with high anticancer activity against NFS-60, HepG-2, and MCF-7 cancer cell lines. The outcomes demonstrated that L-glutaminase could be applied in many biotechnological applications such as pharmaceutical and food processing.


Asunto(s)
Antineoplásicos/farmacología , Glutaminasa/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Bacillus subtilis/enzimología , Bacillus subtilis/aislamiento & purificación , Línea Celular Tumoral , Chlorocebus aethiops , Ensayos de Selección de Medicamentos Antitumorales , Pruebas de Enzimas , Estabilidad de Enzimas , Glutaminasa/química , Glutaminasa/aislamiento & purificación , Humanos , Ratones , Temperatura , Células Vero
16.
Biochemistry ; 58(22): 2584-2588, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31117390

RESUMEN

The members of the glutamine amidotransferase (GATase) family catalyze the incorporation of ammonia within numerous metabolic pathways and can be categorized in two classes. Here, we concentrated on class I GATases, which are heteromeric enzyme complexes consisting of synthase subunits and glutaminase subunits with a catalytic Cys-His-Glu triad. Glutamine hydrolysis at the glutaminase subunit is (i) dependent on the formation of tight synthase-glutaminase complexes and (ii) allosterically coupled to the presence of the substrate at the synthase subunit. The structural basis of both complex formation and allostery is poorly understood. However, previous work on 4-amino-4-deoxychorismate synthase and imidazole glycerol phosphate synthase suggested that a conserved aspartate residue in their synthase subunits, which is located at the subunit interface close to the glutaminase catalytic triad, might be important for both features. We performed a computational screen of class I GATases from the Protein Data Bank and identified conserved and similarly located aspartate residues. We then generated alanine and glutamate mutants of these residues and characterized them by analytical gel filtration and steady-state enzyme kinetics. The results confirmed the important role of the wild-type aspartate residues for the formation of stable synthase-glutaminase complexes (in three of four cases) and the stimulation of glutaminase activity in the analyzed GATases (in all four cases). We present a model for rationalizing the dual role of the conserved aspartate residue toward a unifying regulation mechanism in the entire class I GATase family.


Asunto(s)
Ácido Aspártico/química , Glutaminasa/química , Complejos Multienzimáticos/química , Regulación Alostérica/genética , Dominio Catalítico , Cristalografía por Rayos X , Escherichia coli/química , Glutaminasa/genética , Cinética , Complejos Multienzimáticos/genética , Mutagénesis Sitio-Dirigida , Mutación , Multimerización de Proteína/genética
17.
J Biol Chem ; 293(10): 3535-3545, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29317493

RESUMEN

Altered glycolytic flux in cancer cells (the "Warburg effect") causes their proliferation to rely upon elevated glutamine metabolism ("glutamine addiction"). This requirement is met by the overexpression of glutaminase C (GAC), which catalyzes the first step in glutamine metabolism and therefore represents a potential therapeutic target. The small molecule CB-839 was reported to be more potent than other allosteric GAC inhibitors, including the parent compound bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl (BPTES), and is in clinical trials. Recently, we described the synthesis of BPTES analogs having distinct saturated heterocyclic cores as a replacement for the flexible chain moiety, with improved microsomal stability relative to CB-839 and BPTES. Here, we show that one of these new compounds, UPGL00004, like CB-839, more potently inhibits the enzymatic activity of GAC, compared with BPTES. We also compare the abilities of UPGL00004, CB-839, and BPTES to directly bind to recombinant GAC and demonstrate that UPGL00004 has a similar binding affinity as CB-839 for GAC. We also show that UPGL00004 potently inhibits the growth of triple-negative breast cancer cells, as well as tumor growth when combined with the anti-vascular endothelial growth factor antibody bevacizumab. Finally, we compare the X-ray crystal structures for UPGL00004 and CB-839 bound to GAC, verifying that UPGL00004 occupies the same binding site as CB-839 or BPTES and that all three inhibitors regulate the enzymatic activity of GAC via a similar allosteric mechanism. These results provide insights regarding the potency of these inhibitors that will be useful in designing novel small-molecules that target a key enzyme in cancer cell metabolism.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Glutaminasa/antagonistas & inhibidores , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Sitio Alostérico/efectos de los fármacos , Sustitución de Aminoácidos , Antineoplásicos/química , Antineoplásicos/metabolismo , Bencenoacetamidas/química , Bencenoacetamidas/metabolismo , Bencenoacetamidas/farmacología , Unión Competitiva , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Glutaminasa/química , Glutaminasa/genética , Glutaminasa/metabolismo , Glutamina/antagonistas & inhibidores , Glutamina/química , Glutamina/metabolismo , Humanos , Enlace de Hidrógeno , Conformación Molecular , Mutación , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Sulfuros/química , Sulfuros/metabolismo , Sulfuros/farmacología , Tiadiazoles/química , Tiadiazoles/metabolismo , Tiadiazoles/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
18.
Crit Rev Biotechnol ; 39(7): 944-963, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31327254

RESUMEN

This article focuses on significant advances in the production and applications of microbial glutaminases and provides insight into the structures of different glutaminases. Glutaminases catalyze the deamidation of glutamine to glutamic acid, and this unique ability forms the basis of their applications in various industries such as pharmaceutical and food organizations. Microbial glutaminases from bacteria, actinomycetes, yeast, and fungi are of greater significance than animal glutaminases because of their stability, affordability, and ease of production. Owing to these notable benefits, they are considered to possess considerable potential in anticancer and antiviral therapy, flavor enhancers in oriental foods, biosensors and in the production of a nutraceutical theanine. This review also aims to fully explore the potential of microbial glutaminases and to set the pace for future prospects.


Asunto(s)
Glutaminasa/biosíntesis , Microbiología Industrial/métodos , Animales , Clonación Molecular , Glutaminasa/química , Glutaminasa/genética , Glutaminasa/farmacología , Humanos , Conformación Proteica , Tolerancia a la Sal
19.
Bioorg Med Chem Lett ; 29(13): 1673-1676, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31076348

RESUMEN

Organotellurium compounds have been reported as an immune-modulator sensitizing chemotherapeutics. Herein, we report the design and synthesis of a series of novel tellurodibenzoic acids as mimics of diphenylarsenic acid (DPAA) and potential selective KGA inhibitors. Representative compound 3B exhibited potent inhibition of KGA and glutamine-dependent HCT-116 cells. Stability experiments indicated that 3B has excellent stability under acidic (HCOOH), basic (NH3·H2O) and oxidative (H2O2) conditions, but reacts with ß-ME, DTT and lysine which suggested that compound 3B may interact with cysteine or lysine residues. Moreover, molecular docking disclosed that compound 3B binds to the allosteric site of the GAC tetramer containing Arg317-Lys320-Leu321-Phe322-Tyr394-Glu325, which helped to rationalize the SAR and further design and optimization. Taken together, compound 3B could be used as a starting point for the development of new KGA inhibitors.


Asunto(s)
Benzoatos/química , Inhibidores Enzimáticos/química , Glutaminasa/antagonistas & inhibidores , Compuestos Organometálicos/química , Telurio/química , Benzoatos/síntesis química , Benzoatos/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Glutaminasa/química , Células HCT116 , Humanos , Riñón/enzimología , Simulación del Acoplamiento Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/farmacología
20.
J Biol Chem ; 292(27): 11572-11585, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28526749

RESUMEN

On the basis of tissue-specific enzyme activity and inhibition by catalytic products, Hans Krebs first demonstrated the existence of multiple glutaminases in mammals. Currently, two human genes are known to encode at least four glutaminase isoforms. However, the phylogeny of these medically relevant enzymes remains unclear, prompting us to investigate their origin and evolution. Using prokaryotic and eukaryotic glutaminase sequences, we built a phylogenetic tree whose topology suggested that the multidomain architecture was inherited from bacterial ancestors, probably simultaneously with the hosting of the proto-mitochondrion endosymbiont. We propose an evolutionary model wherein the appearance of the most active enzyme isoform, glutaminase C (GAC), which is expressed in many cancers, was a late retrotransposition event that occurred in fishes from the Chondrichthyes class. The ankyrin (ANK) repeats in the glutaminases were acquired early in their evolution. To obtain information on ANK folding, we solved two high-resolution structures of the ANK repeat-containing C termini of both kidney-type glutaminase (KGA) and GLS2 isoforms (glutaminase B and liver-type glutaminase). We found that the glutaminase ANK repeats form unique intramolecular contacts through two highly conserved motifs; curiously, this arrangement occludes a region usually involved in ANK-mediated protein-protein interactions. We also solved the crystal structure of full-length KGA and present a small-angle X-ray scattering model for full-length GLS2. These structures explain these proteins' compromised ability to assemble into catalytically active supra-tetrameric filaments, as previously shown for GAC. Collectively, these results provide information about glutaminases that may aid in the design of isoform-specific glutaminase inhibitors.


Asunto(s)
Evolución Molecular , Glutaminasa , Modelos Genéticos , Modelos Moleculares , Repetición de Anquirina , Cristalografía por Rayos X , Glutaminasa/química , Glutaminasa/genética , Humanos , Isoenzimas/química , Isoenzimas/genética , Dominios Proteicos , Estructura Cuaternaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA