Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Enzyme Microb Technol ; 150: 109863, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34489022

ABSTRACT

Protein-glutaminase (EC 3.5.1.44, PG) converts protein glutamine residues in proteins and peptides into glutamic acid residue, and markedly improves the solubility, emulsification, and foaming properties of food proteins. However, the source bacteria, Chryseobacterium proteolyticum, have low enzyme production ability, inefficient genetic operation, and high production cost. Therefore, it is critical to establish an efficient expression system for active PG. Here, combinatorial engineering was developed for high-yield production of PG in Bacillus subtilis. First, we evaluated different B. subtilis strains for PG self-activation. Then, combinatorial optimization involving promoters, signal peptides, and culture medium was applied to produce active recombinant PG in a B. subtilis expression system. Through combinatorial engineering, PG enzyme activity reached 3.23 U/mL in shaken-flask cultures. Active PG with the yield of 7.07 U/mL was obtained at 40 h by the PSecA-YdeJ combination in fed-batch fermentation, which is the highest yield of PG in existing reports.


Subject(s)
Bacillus subtilis , Bacterial Proteins/biosynthesis , Chryseobacterium , Glutaminase/biosynthesis , Bacillus subtilis/metabolism , Chryseobacterium/enzymology , Fermentation , Protein Engineering , Protein Sorting Signals
2.
Curr Drug Metab ; 21(3): 211-220, 2020.
Article in English | MEDLINE | ID: mdl-32316889

ABSTRACT

AIM AND OBJECTIVE: To review the applications and production studies of reported antileukemic drug L-glutaminase under Solid-state Fermentation (SSF). OVERVIEW: An amidohydrolase that gained economic importance because of its wide range of applications in the pharmaceutical industry, as well as the food industry, is L-glutaminase. The medical applications utilized it as an anti-tumor agent as well as an antiretroviral agent. L-glutaminase is employed in the food industry as an acrylamide degradation agent, as a flavor enhancer and for the synthesis of theanine. Another application includes its use in hybridoma technology as a biosensing agent. Because of its diverse applications, scientists are now focusing on enhancing the production and optimization of L-glutaminase from various sources by both Solid-state Fermentation (SSF) and submerged fermentation studies. Of both types of fermentation processes, SSF has gained importance because of its minimal cost and energy requirement. L-glutaminase can be produced by SSF from both bacteria and fungi. Single-factor studies, as well as multi-level optimization studies, were employed to enhance L-glutaminase production. It was concluded that L-glutaminase activity achieved by SSF was 1690 U/g using wheat bran and Bengal gram husk by applying feed-forward artificial neural network and genetic algorithm. The highest L-glutaminase activity achieved under SSF was 3300 U/gds from Bacillus sp., by mixture design. Purification and kinetics studies were also reported to find the molecular weight as well as the stability of L-glutaminase. CONCLUSION: The current review is focused on the production of L-glutaminase by SSF from both bacteria and fungi. It was concluded from reported literature that optimization studies enhanced L-glutaminase production. Researchers have also confirmed antileukemic and anti-tumor properties of the purified L-glutaminase on various cell lines.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Fermentation , Glutaminase/biosynthesis , Glutaminase/pharmacology , Bacteria , Fungi , Humans , Kinetics
3.
FASEB J ; 33(11): 12200-12212, 2019 11.
Article in English | MEDLINE | ID: mdl-31373848

ABSTRACT

In hypertrophic scar (HS) formation, the type 2 immune response induces the alternatively activated macrophages (M2), which manipulate fibroblasts to differentiate into myofibroblasts with active biologic functions and proliferation. Myofibroblasts express α-smooth muscle actin (α-SMA) and synthesize and produce additional collagen type I and collagen type III, inducing HS formation. However, studies on the mechanism of M2 macrophage modulation are only based on the recognition of profibrotic factors such as TGF-ß1 secreted by macrophages. The influence of exosomes from M2 macrophages on scar formation is still unknown. Both M2 macrophages and myofibroblasts highly express glutaminases (GLSs). GLS is a critical enzyme in glutaminolysis and is important for M2 macrophage and fibroblast polarization. In this study, we found that in a TGF-ß1-stimulated coculture system, a long noncoding RNA (lncRNA) named lncRNA-ASLNCS5088 was enriched in M2 macrophage-derived exosomes. This lncRNA could be transferred with high efficiency to fibroblasts and acted as an endogenous sponge to adsorb microRNA-200c-3p, resulting in increased GLS and α-SMA expression. Pretreatment with GW4869, which impairs M2 macrophage exosome synthesis, ameliorated these pathologic changes in fibroblasts in vitro. Local injection in the late scar formation period with GW4869 reduced α-SMA+ fibroblasts and alleviated the fibrosis of tissue after wound healing in vivo.-Chen, J., Zhou, R., Liang, Y., Fu, X., Wang, D., Wang, C. Blockade of lncRNA-ASLNCS5088-enriched exosome generation in M2 macrophages by GW4869 dampens the effect of M2 macrophages on orchestrating fibroblast activation.


Subject(s)
Aniline Compounds/pharmacology , Benzylidene Compounds/pharmacology , Cicatrix, Hypertrophic/etiology , Exosomes/physiology , Fibroblasts/physiology , Macrophages/physiology , RNA, Long Noncoding/physiology , Actins/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Glutaminase/biosynthesis , Humans , THP-1 Cells , Transforming Growth Factor beta1/physiology
4.
Crit Rev Biotechnol ; 39(7): 944-963, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31327254

ABSTRACT

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.


Subject(s)
Glutaminase/biosynthesis , Industrial Microbiology/methods , Animals , Cloning, Molecular , Glutaminase/chemistry , Glutaminase/genetics , Glutaminase/pharmacology , Humans , Protein Conformation , Salt Tolerance
5.
Molecules ; 24(3)2019 Feb 07.
Article in English | MEDLINE | ID: mdl-30736411

ABSTRACT

In this study, the Micrococcus luteus K-3 glutaminase was successfully over-expressed in the GRAS (Generally Recognized as Safe) Bacillus subtilis strain 168 by integration of the Mglu gene in the 16S rDNA locus. This was done in order to screen a strain producing high levels of recombinant glutaminase from the selected candidates. The transcription of the glutaminase genes in the B. subtilis 168 chromosome and the expression of glutaminase protein was further assessed by qPCR, SDS-PAGE analysis and an enzyme activity assay. To further increase the production of glutaminase, the nprB and nprE genes, which encode specific proteases, were disrupted by integration of the Mglu gene. After continuous cell culturing without the addition of antibiotics, the integrated recombinant strains showed excellent genetic stability, demonstrating favorable industrialization potential. After the fermentation temperature was optimized, a 5-L bioreactor was used for fed-batch fermentation of the recombinant glutaminase producing strain at 24 °C, and the highest enzyme activity achieved was approximately 357.6 U/mL.


Subject(s)
Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/genetics , DNA, Ribosomal/genetics , Endopeptidases/genetics , Fermentation , Glutaminase/biosynthesis , Gene Expression Regulation, Bacterial , Glutaminase/metabolism , Temperature
6.
Proteomics Clin Appl ; 13(4): e1800038, 2019 07.
Article in English | MEDLINE | ID: mdl-30485682

ABSTRACT

PURPOSE: To screen the novel biomarkers for gastric cancer and to determine the values of glutaminase 1 (GLS1) and gamma-glutamylcyclotransferase (GGCT) for detecting gastric cancer. EXPERIMENTAL DESIGN: A discovery group of four paired gastric cancer tissue samples are labeled with Isobaric tag for relative and absolute quantitation agents and identified with LC-ESI-MS/MS. A validation group of 168 gastric cancer samples and 30 healthy controls are used to validate the expression of GLS1 and GGCT. RESULTS: Four hundred and thirty-one proteins are found differentially expressed in gastric cancer tissues. Of these proteins, GLS1 and GGCT are found overexpressed in gastric cancer patients, with sensitivity of 75.6% (95% CI: 69-82.2%) and specificity of 81% (95% CI: 75-87%) for GLS1, and with sensitivity of 63.1% (95% CI: 55.7-71.5%) and specificity of 60.7% (95% CI: 53.3-68.2%) for GGCT. The co-expression of GLS1 and GGCT in gastric cancer tissues has sensitivity of 78.1% (95% CI: 70.1-86.1%) and specificity of 86.5% (95% CI: 79.5-93.4%). Moreover, both GLS1 and GGCT present higher expression of 82.6% (95% CI: 68.5-99.4%) and 73.9% (95% CI: 54.5-93.3%) in lymph node metastasis specimen than those in non-lymph node metastasis specimen. The areas under ROC curves are up to 0.734 for the co-expression of GLS1 and GGCT in gastric cancer. The co-expression of GLS1 and GGCT is strongly associated with histological grade, lymph node metastasis, and TNM stage Ⅲ/Ⅳ. CONCLUSIONS AND CLINICAL RELEVANCE: The present study provides the quantitative proteomic analysis of gastric cancer tissues to identify prognostic biomarkers of gastric cancer. The co-expression level of GLS1 and GGCT is of great clinical value to serve as diagnostic and therapeutic biomarkers for early gastric cancer.


Subject(s)
Biomarkers, Tumor/biosynthesis , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Glutaminase/biosynthesis , Glutamine/metabolism , Neoplasm Proteins/biosynthesis , Stomach Neoplasms/metabolism , gamma-Glutamylcyclotransferase/biosynthesis , Chromatography, Liquid , Female , Humans , Male , Mass Spectrometry , Proteomics , Stomach Neoplasms/pathology
7.
Histol Histopathol ; 34(2): 167-174, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30191947

ABSTRACT

PURPOSE: We evaluated the expression of glutaminolysis-related proteins in Hurthle cell neoplasms (HCN) and follicular neoplasms (FN) of the thyroid, and investigated its clinical implication. METHODS: Tissue microarrays were constructed from 264 FNs (112 follicular carcinomas [FCs] and 152 follicular adenomas [FAs]) and 108 HCNs (27 Hurthle cell carcinomas [HCCs] and 81 Hurthle cell adenomas [HCAs]. The immunohistochemical staining result of 3 glutaminolysis-related proteins (Glutaminase 1 [GLS1], glutaminate dehydrogenase [GDH] and alanine- serine, cysteine-preferring transporter 2 [ASCT2]) was analyzed. RESULTS: GLS1 and GDH showed significantly higher expression rates in HCN compared to FN (P<0.001). More HCN cases showed co-positivity of multiple glutaminolysis-related proteins than those of FN cases (P<0.001). In silico analysis, both GLUD1 and GLUD2 showed higher expression rate in HCA compared to FA (P=0.027 and P=0.018, respectively). SLC1A5 expression was highest in HCA, followed by FC and FA (HCA vs FC, P=0.023; FC vs FA, P=0.002). CONCLUSION: FN and HCN exhibit a different expression pattern for glutaminolysis-related proteins, and GLS1 and GDH have higher expression rates in HCN and FN.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Adenoma, Oxyphilic/metabolism , Thyroid Neoplasms/metabolism , Adult , Amino Acid Transport System ASC/analysis , Amino Acid Transport System ASC/biosynthesis , Female , Glutamate Dehydrogenase/analysis , Glutamate Dehydrogenase/biosynthesis , Glutaminase/analysis , Glutaminase/biosynthesis , Glutamine/metabolism , Humans , Male , Middle Aged , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/biosynthesis
8.
Prostate ; 78(15): 1181-1195, 2018 11.
Article in English | MEDLINE | ID: mdl-30009389

ABSTRACT

In the course of studies aimed at the role of oxidative stress in the development of metastatic potential in the LNCaP-C4-2B prostate cancer progression model system, we found a relative decrease in the level of expression of the cytoplasmic nicotinamide riboside: quinone oxidoreductase (NQO2) and an increase in the oxidative stress in C4-2B cells compared to that in LNCaP or its derivatives C4 and C4-2. It was also found that C4-2B cells specifically shed large extracellular vesicles (LEVs) suggesting that these LEVs and their cargo could participate in the establishment of the osseous metastases. The level of expression of caveolin-1 increased as the system progresses from LNCaP to C4-2B. Since NQO2 RNA levels were not changed in LNCaP, C4, C4-2, and C4-2B, we tested an altered cellular distribution hypothesis of NQO2 being compartmentalized in the membrane fractions of C4-2B cells which are rich in lipid rafts and caveolae. This was confirmed when the detergent resistant membrane fractions were probed on immunoblots. Moreover, when the LEVs were analyzed for membrane associated caveolin-1 as possible cargo, we noticed that the enzyme NQO2 was also a component of the cargo along with caveolin-1 as seen in double immunofluorescence studies. Molecular modeling studies showed that a caveolin-1 accessible site is present in NQO2. Specific interaction between NQO2 and caveolin-1 was confirmed using deletion constructs of caveolin-1 fused with glutathione S-transferase (GST). Interestingly, whole cell lysate and mitochondrial preparations of LNCaP, C4, C4-2, and C4-2B showed an increasing expression of glutaminase (GLS, kidney type). The extrusion of LEVs appears to be a specific property of the bone metastatic C4-2B cells and this process could be inhibited by a GLS specific inhibitor BPTES, suggesting the critical role of a functioning glutamine metabolism. Our results indicate that a high level of expression of caveolin-1 in C4-2B cells contributes to an interaction between caveolin-1 and NQO2 and to their packaging as cargo in the shed LEVs. These results suggest an important role of membrane associated oxidoreductases in the establishment of osseous metastases in prostate cancer.


Subject(s)
Extracellular Vesicles/enzymology , Glutaminase/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Quinone Reductases/metabolism , Amino Acid Sequence , Binding Sites , Caveolin 1/metabolism , Cell Line, Tumor , Disease Progression , Extracellular Vesicles/metabolism , Glutaminase/biosynthesis , Glutamine/metabolism , Humans , Immunoblotting , Male , Models, Molecular , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasm Metastasis , Oxidative Stress , Prostatic Neoplasms/metabolism , Quinone Reductases/biosynthesis , Quinone Reductases/chemistry
9.
Rev Esp Enferm Dig ; 110(7): 427-433, 2018 07.
Article in English | MEDLINE | ID: mdl-29542325

ABSTRACT

AIM: to analyze the effect of metformin on ammonia production derived from glutamine metabolism in vitro and in vivo. METHODS: twenty male Wistar rats were studied for 28 days after a porto-caval anastomosis (n = 16) or a sham operation (n = 4). Porto-caval shunted animals were randomized into two groups (n = 8) and either received 30 mg/kg/day of metformin for two weeks or were control animals. Plasma ammonia concentration, Gls gene expression and K-type glutaminase activity were measured in the small intestine, muscle and kidney. Furthermore, Caco2 were grown in different culture media containing glucose/glutamine as the main carbon source and exposed to different concentrations of the drug. The expression of genes implicated in glutamine metabolism were analyzed. RESULTS: metformin was associated with a significant inhibition of glutaminase activity levels in the small intestine of porto-caval shunted rats (0.277 ± 0.07 IU/mg vs 0.142 ± 0.04 IU/mg) and a significant decrease in plasma ammonia (204.3 ± 24.4 µg/dl vs 129.6 ± 16.1 µg/dl). Glucose withdrawal induced the expression of the glutamine transporter SLC1A5 (2.54 ± 0.33 fold change; p < 0.05). Metformin use reduced MYC levels in Caco2 and consequently, SLC1A5 and GLS expression, with a greater effect in cells dependent on glutaminolytic metabolism. CONCLUSION: metformin regulates ammonia homeostasis by modulating glutamine metabolism in the enterocyte, exerting an indirect control of both the uptake and degradation of glutamine. This entails a reduction in the production of metabolites and energy through this pathway and indirectly causes a decrease in ammonia production that could be related to a decreased risk of HE development.


Subject(s)
Glutamine/metabolism , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Ammonia/metabolism , Animals , Caco-2 Cells , Child, Preschool , Glutaminase/antagonists & inhibitors , Glutaminase/biosynthesis , Glutaminase/genetics , Humans , Male , Rats , Rats, Wistar
10.
PLoS One ; 12(8): e0181745, 2017.
Article in English | MEDLINE | ID: mdl-28813436

ABSTRACT

Microbial anti-cancer enzymes have been proven to be effective and economical agents for cancer treatment. Aeromonas veronii has been identified as a microorganism with the potential to produce L-glutaminase, an anticancer agent effective against acute lymphocytic leukaemia. In this study, a selective medium of Aeromonas veronii was used to culture the microorganism. Strain improvement was done by adaptive and induced mutational techniques. A selective minimal agar media was incorporated for the growth of the strain which further supports adaptive mutation. Strains were also UV-irradiated and successively treated with N-methyl-N'-nitro-N-nitrosoguanidine to find a resilient strain capable of producing L-glutaminase efficiently. The Plackett-Burman design and central composite designs were used to screen and optimize additional carbon and nitrogen sources. Adaptive mutation resulted in promising yield improvements compared to native strain (P<0.001). The mean yield of 30 treated colonies from the induced mutation was significantly increased compared to the non-induced strain (P< 0.001). The economically feasible statistical designs were found to reinforce each other in order to maximize the yield of the enzyme. The interactions of nutrient factors were understood from the 3D response surface plots. The model was found to be a perfect fit in terms of maximizing enzyme yield, with the productivity improving at every stage to a fourfold output of enzyme (591.11 ±7.97 IU/mL) compared to the native strain (135±3.51 IU/mL).


Subject(s)
Adaptation, Biological , Aeromonas veronii/enzymology , Aeromonas veronii/genetics , Antineoplastic Agents/metabolism , Glutaminase/biosynthesis , Glutaminase/genetics , Mutation , Analysis of Variance , Base Sequence , DNA Mutational Analysis
11.
Virchows Arch ; 470(4): 381-389, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28185053

ABSTRACT

Glutamine metabolism is emerging as one aspect of dysregulated metabolism of tumors. Triple-negative breast cancer (TNBC) cells are glutamine dependent, whereas luminal-type cells tend to be glutamine independent. Therefore, TNBC patients might benefit from therapies targeting glutamine metabolism. To investigate the clinical significance of glutamine metabolism, we examined expression and prognostic significance of glutaminase in tumor cells and tumor-infiltrating lymphocytes (TILs) in TNBC. We retrieved 658 surgically resected TNBCs and analyzed glutaminase expression in tumor cells and TILs by immunohistochemical staining. Glutaminase expression was observed in 237 cases (36.0%) in tumor cells and 104 cases (15.5%) in TILs. Although glutaminase expression in tumor cells was significantly associated with a low level of TILs (p = 0.018), glutaminase expression in TILs was significantly higher in cases with a high level of TILs (p = 0.031). Glutaminase expression in tumor cells was significantly associated with poor disease-free survival in patients with lymph node metastasis and high levels of TILs (p = 0.020). In addition, it was an independent poor prognostic factor (hazard ratio = 10.643, 95% confidence interval = 1.999-56.668; p = 0.006). Glutaminase expression in tumor cells was observed in a subset of TNBC patients. It was significantly associated with a low level of TILs and poor disease-free survival in TNBCs presenting with lymph node metastasis and high levels of TILs.


Subject(s)
Biomarkers, Tumor/analysis , Glutaminase/biosynthesis , Lymphocytes, Tumor-Infiltrating/pathology , Triple Negative Breast Neoplasms/pathology , Adult , Aged , Disease-Free Survival , Female , Glutaminase/analysis , Humans , Immunohistochemistry , Lymphatic Metastasis/pathology , Middle Aged , Prognosis , Proportional Hazards Models , Tissue Array Analysis , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/mortality
12.
Appl Biochem Biotechnol ; 183(1): 30-50, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28205049

ABSTRACT

Among all fungal endophytes isolates derived from different ethno-medical plants, the hyper-yield L-asparaginase and L-glutaminase wild strains Trichoderma sp. Gen 9 and Cladosporium sp. Gen 20 using rice straw under solid-state fermentation (SSF) were selected. The selected strains were used as parents for the intergeneric protoplast fusion program to construct recombinant strain for prompt improvement production of these enzymes in one recombinant strain. Among 21 fusants obtained, the recombinant strain AYA 20-1, with 2.11-fold and 2.58-fold increase in L-asparaginase and L-glutaminase activities more than the parental isolates Trichoderma sp. Gen 9 and Cladosporium sp. Gen 20, respectively, was achieved using rice straw under SSF. Both therapeutic enzymes L-asparaginase and L-glutaminase were purified and characterized from the culture supernatant of the recombinant AYA 20-1 strain with molecular weights of 50.6 and 83.2 kDa, respectively. Both enzymes were not metalloenzymes. Whereas thiol group blocking reagents such as p-chloromercurybenzoate and iodoacetamide totally inhibited L-asparaginase activity, which refer to sulfhydryl groups and cysteine residues involved in its catalytic activity, they have no effect toward L-glutaminase activity. Interestingly, potent anticancer, antioxidant, and antimicrobial activities were detected for both enzymes.


Subject(s)
Antineoplastic Agents/metabolism , Asparaginase , Glutaminase , Oryza/chemistry , Trichoderma , Asparaginase/biosynthesis , Asparaginase/genetics , Glutaminase/biosynthesis , Glutaminase/genetics , Humans , Protoplasts/enzymology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Trichoderma/enzymology , Trichoderma/genetics
13.
Oncotarget ; 7(33): 53628-53641, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27447554

ABSTRACT

PURPOSE: This study aimed to investigate the expression of glutamine metabolism-related protein in tumor and stromal compartments among the histologic subtypes of thyroid cancer. RESULTS: GLS1 and GDH expression in tumor and stromal compartments were the highest in AC than in other subtypes. Tumoral ASCT2 expression was higher in MC but lower in FC (p < 0.001). In PTC, tumoral GLS1 and tumoral GDH expression was higher in the conventional type than in the follicular variant (p = 0.043 and 0.001, respectively), and in PTC with BRAF V600E mutation than in PTC without BRAF V600E mutation (p<0.001). Stromal GDH positivity was the independent factor associated with short overall survival (hazard ratio: 21.48, 95% confidence interval: 2.178-211.8, p = 0.009). METHODS: We performed tissue microarrays with 557 thyroid cancer cases (papillary thyroid carcinoma [PTC]: 344, follicular carcinoma [FC]: 112, medullary carcinoma [MC]: 70, poorly differentiated carcinoma [PDC]: 23, and anaplastic carcinoma [AC]: 8) and 152 follicular adenoma (FA) cases. We performed immunohistochemical staining of glutaminolysis-related proteins (glutaminase 1 [GLS1], glutamate dehydrogenase [GDH], and amino acid transporter-2 [ASCT-2]). CONCLUSION: Glutamine metabolism-related protein expression differed among the histologic subtypes of thyroid cancer.


Subject(s)
Glutaminase/biosynthesis , Glutamine/metabolism , Sugar Alcohol Dehydrogenases/biosynthesis , Thyroid Neoplasms/metabolism , Adult , Aged , Amino Acid Transport System ASC/biosynthesis , Female , Humans , Male , Middle Aged , Minor Histocompatibility Antigens/biosynthesis
14.
Adv Food Nutr Res ; 78: 95-114, 2016.
Article in English | MEDLINE | ID: mdl-27452167

ABSTRACT

There is an increased l-glutaminase market worldwide due to its relevant industrial applications. Salt tolerance l-glutaminases play a vital role in the increase of flavor of different types of foods like soya sauce and tofu. This chapter is presenting the economically viable l-glutaminases production in solid-state fermentation (SSF) by Aspergillus flavus MTCC 9972 as a case study. The enzyme production was improved following a three step optimization process. Initially mixture design (MD) (augmented simplex lattice design) was employed to optimize the solid substrate mixture. Such solid substrate mixture consisted of 59:41 of wheat bran and Bengal gram husk has given higher amounts of l-glutaminase. Glucose and l-glutamine were screened as a finest additional carbon and nitrogen sources for l-glutaminase production with help of Plackett-Burman Design (PBD). l-Glutamine also acting as a nitrogen source as well as inducer for secretion of l-glutaminase from A. flavus MTCC 9972. In the final step of optimization various environmental and nutritive parameters such as pH, temperature, moisture content, inoculum concentration, glucose, and l-glutamine levels were optimized through the use of hybrid feed forward neural networks (FFNNs) and genetic algorithm (GA). Through sequential optimization methods MD-PBD-FFNN-GA, the l-glutaminase production in SSF could be improved by 2.7-fold (453-1690U/g).


Subject(s)
Aquatic Organisms/enzymology , Fermentation , Glutaminase/biosynthesis , Aquatic Organisms/microbiology , Aspergillus flavus/enzymology , Aspergillus flavus/growth & development , Costs and Cost Analysis , Glutaminase/economics
15.
Mol Med Rep ; 13(1): 925-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26647830

ABSTRACT

Indoleamine 2,3­dioxygenase (IDO), through L­tryptophan depletion, activates general control non­derepressible (GCN) 2 kinase and suppresses T­cell proliferation, in addition to suppressing aerobic glycolysis and glutaminolysis, which are required for these rapidly proliferating cells. A number of, however not all of these alterations, are partially mediated through IDO­induced p53 upregulation. In two­way mixed lymphocyte reactions (MLRs), IDO reduced cellular proliferation. In MLR­derived T­cells, IDO induced the expression levels of p53 and p21, however concurrently reduced the levels of ζ­chain, c­Myc, lactate dehydrogenase A (LDH­A) and glutaminase (GLS)2. However, p53 had no effect on the expression of the above proteins. These results were recapitulated in T­cells activated with anti­CD2, anti­CD3 and anti­CD28 by direct activation of the GCN2 kinase with tryptophanol. In conclusion, IDO, through GCN2 kinase activation, downregulates the levels of TCR­complex ζ­chain and c­Myc, resulting in the suppression of T­cell proliferation and a reduction in the levels of LDH­A and GLS2, which are key enzymes involved in aerobic glycolysis and glutaminolysis, respectively.


Subject(s)
Glutaminase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , L-Lactate Dehydrogenase/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , Cell Proliferation/genetics , Glutaminase/genetics , Glycolysis/genetics , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , L-Lactate Dehydrogenase/genetics , Mitochondria/genetics , Mitochondria/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Suppressor Protein p53/biosynthesis , p21-Activated Kinases/biosynthesis
16.
Eur Arch Otorhinolaryngol ; 273(2): 495-503, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25663193

ABSTRACT

Glutaminolysis is a crucial factor for tumor metabolism in the carcinogenesis of several tumors but has not been clarified for oral squamous cell carcinoma (OSCC) yet. Expression of glutaminolysis-related solute carrier family 1, member 5 (SLC1A5)/neutral amino acid transporter (ASCT2), glutaminase (GLS), and glutamate dehydrogenase (GLDH) was analyzed in normal oral mucosa (n = 5), oral precursor lesions (simple hyperplasia, n = 11; squamous intraepithelial neoplasia, SIN I-III, n = 35), and OSCC specimen (n = 42) by immunohistochemistry. SLC1A5/ASCT2 and GLS were significantly overexpressed in the carcinogenesis of OSCC compared with normal tissue, while GLDH was weakly detected. Compared with SIN I-III SLC1A5/ASCT2 and GLS expression were significantly increased in OSCC. GLDH expression did not significantly differ from SIN I-III compared with OSCC. This study shows the first evidence of glutaminolysis-related SLC1A5/ASCT2, GLS, and GLDH expression in OSCC. The very weak GLDH expression indicates that glutamine metabolism is rather related to nucleotide or protein/hexosamine biosynthesis or to the function as an antioxidant (glutathione) than to energy production or generation of lactate through entering the tricarboxylic acid cycle. Overcoming glutaminolysis by targeting c-Myc oncogene (e.g. by natural compounds) and thereby cross-activation of mammalian target of rapamycin complex 1 or SLC1A5/ASCT2, GLS inhibitors may be a useful strategy to sensitize cancer cells to common OSCC cancer therapies.


Subject(s)
Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Glutamine/genetics , Mouth Neoplasms/metabolism , RNA, Neoplasm/genetics , Amino Acid Transport System ASC/biosynthesis , Animals , Biomarkers, Tumor/biosynthesis , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Female , Glutaminase/biosynthesis , Glutamine/biosynthesis , Humans , Immunohistochemistry , Middle Aged , Minor Histocompatibility Antigens , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Mouth Neoplasms/pathology , Oxidoreductases Acting on CH-CH Group Donors/biosynthesis , Real-Time Polymerase Chain Reaction
17.
Biomed Res Int ; 2015: 627031, 2015.
Article in English | MEDLINE | ID: mdl-26180806

ABSTRACT

Among the antitumor drugs, bacterial enzyme L-asparaginase has been employed as the most effective chemotherapeutic agent in pediatric oncotherapy especially for acute lymphoblastic leukemia. Glutaminase free L-asparaginase producing actinomycetes were isolated from soil samples collected from Egypt. Among them, a potential culture, strain NEAE-119, was selected and identified on the basis of morphological, cultural, physiological, and biochemical properties together with 16S rRNA sequence as Streptomyces olivaceus NEAE-119 and sequencing product (1509 bp) was deposited in the GenBank database under accession number KJ200342. The optimization of different process parameters for L-asparaginase production by Streptomyces olivaceus NEAE-119 using Plackett-Burman experimental design and response surface methodology was carried out. Fifteen variables (temperature, pH, incubation time, inoculum size, inoculum age, agitation speed, dextrose, starch, L-asparagine, KNO3, yeast extract, K2HPO4, MgSO4·7H2O, NaCl, and FeSO4·7H2O) were screened using Plackett-Burman experimental design. The most positive significant independent variables affecting enzyme production (temperature, inoculum age, and agitation speed) were further optimized by the face-centered central composite design-response surface methodology.


Subject(s)
Asparaginase , Hypolipidemic Agents , Asparaginase/biosynthesis , Asparaginase/genetics , Asparaginase/isolation & purification , Cell Culture Techniques , Glutaminase/biosynthesis , Glutaminase/genetics , Humans , Hypolipidemic Agents/isolation & purification , Hypolipidemic Agents/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Streptomyces/enzymology , Streptomyces/genetics , Streptomyces/growth & development
18.
Oncol Rep ; 34(3): 1533-42, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26134042

ABSTRACT

There is a strong rationale for targeting the metabolic alterations of cancer cells. The most studied of these are the higher rates of glycolysis, glutaminolysis and de novo synthesis of fatty acids (FAs). Despite the availability of pharmacological inhibitors of these pathways, no preclinical studies targeting them simultaneously have been performed. In the present study it was determined whether three key enzymes for glycolysis, glutaminolysis and de novo synthesis of FAs, hexokinase-2, glutaminase and fatty acid synthase, respectively, were overexpressed as compared to primary fibroblasts. In addition, we showed that at clinically relevant concentrations lonidamine, 6-diazo-5-oxo-L-norleucine and orlistat, known inhibitors of the mentioned enzymes, exerted a cell viability inhibitory effect. Genetic downregulation of the three enzymes also reduced cell viability. The three drugs were highly synergistic when administered as a triple combination. Of note, the cytotoxicity of the triple combination was low in primary fibroblasts and was well tolerated when administered into healthy BALB/c mice. The results suggest the feasibility and potential clinical utility of the triple metabolic targeting which merits to be further studied by using either repositioned old drugs or newer, more selective inhibitors.


Subject(s)
Fatty Acid Synthases/biosynthesis , Glutaminase/biosynthesis , Hexokinase/biosynthesis , Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Diazooxonorleucine/administration & dosage , Drug Synergism , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acids/metabolism , Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glutaminase/antagonists & inhibitors , Glycolysis/drug effects , Hexokinase/antagonists & inhibitors , Humans , Indazoles/administration & dosage , Lactones/administration & dosage , Metabolic Networks and Pathways/drug effects , Mice , Neoplasms/enzymology , Neoplasms/pathology , Orlistat
19.
J Clin Invest ; 125(6): 2293-306, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25915584

ABSTRACT

Glutaminase (GLS), which converts glutamine to glutamate, plays a key role in cancer cell metabolism, growth, and proliferation. GLS is being explored as a cancer therapeutic target, but whether GLS inhibitors affect cancer cell-autonomous growth or the host microenvironment or have off-target effects is unknown. Here, we report that loss of one copy of Gls blunted tumor progression in an immune-competent MYC-mediated mouse model of hepatocellular carcinoma. Compared with results in untreated animals with MYC-induced hepatocellular carcinoma, administration of the GLS-specific inhibitor bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl)ethyl sulfide (BPTES) prolonged survival without any apparent toxicities. BPTES also inhibited growth of a MYC-dependent human B cell lymphoma cell line (P493) by blocking DNA replication, leading to cell death and fragmentation. In mice harboring P493 tumor xenografts, BPTES treatment inhibited tumor cell growth; however, P493 xenografts expressing a BPTES-resistant GLS mutant (GLS-K325A) or overexpressing GLS were not affected by BPTES treatment. Moreover, a customized Vivo-Morpholino that targets human GLS mRNA markedly inhibited P493 xenograft growth without affecting mouse Gls expression. Conversely, a Vivo-Morpholino directed at mouse Gls had no antitumor activity in vivo. Collectively, our studies demonstrate that GLS is required for tumorigenesis and support small molecule and genetic inhibition of GLS as potential approaches for targeting the tumor cell-autonomous dependence on GLS for cancer therapy.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glutaminase/biosynthesis , Liver Neoplasms, Experimental/enzymology , Amino Acid Substitution , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Heterografts , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mutation, Missense , Neoplasm Transplantation , Sulfides/pharmacology , Thiadiazoles/pharmacology
20.
J Clin Invest ; 125(4): 1591-602, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25798620

ABSTRACT

The mechanistic target of rapamycin (mTOR) is hyperactivated in many types of cancer, rendering it a compelling drug target; however, the impact of mTOR inhibition on metabolic reprogramming in cancer is incompletely understood. Here, by integrating metabolic and functional studies in glioblastoma multiforme (GBM) cell lines, preclinical models, and clinical samples, we demonstrate that the compensatory upregulation of glutamine metabolism promotes resistance to mTOR kinase inhibitors. Metabolomic studies in GBM cells revealed that glutaminase (GLS) and glutamate levels are elevated following mTOR kinase inhibitor treatment. Moreover, these mTOR inhibitor-dependent metabolic alterations were confirmed in a GBM xenograft model. Expression of GLS following mTOR inhibitor treatment promoted GBM survival in an α-ketoglutarate-dependent (αKG-dependent) manner. Combined genetic and/or pharmacological inhibition of mTOR kinase and GLS resulted in massive synergistic tumor cell death and growth inhibition in tumor-bearing mice. These results highlight a critical role for compensatory glutamine metabolism in promoting mTOR inhibitor resistance and suggest that rational combination therapy has the potential to suppress resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzophenanthridines/therapeutic use , Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/physiology , Glioblastoma/drug therapy , Glutaminase/physiology , Glutamine/metabolism , Indoles/pharmacology , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aged , Animals , Benzophenanthridines/administration & dosage , Benzophenanthridines/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Citric Acid Cycle , Drug Synergism , Energy Metabolism , Gas Chromatography-Mass Spectrometry , Glioblastoma/metabolism , Glioblastoma/pathology , Glutamic Acid/metabolism , Glutaminase/antagonists & inhibitors , Glutaminase/biosynthesis , Glutaminase/genetics , Glycolysis , Humans , Indoles/administration & dosage , Indoles/therapeutic use , Ketoglutaric Acids/metabolism , Magnetic Resonance Spectroscopy , Male , Metabolome/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/therapeutic use , Purines/administration & dosage , Purines/therapeutic use , RNA, Small Interfering/pharmacology , Rotarod Performance Test , Signal Transduction/drug effects , Signal Transduction/physiology , Temporal Lobe/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...