Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 473
Filter
1.
Angew Chem Int Ed Engl ; 62(28): e202304646, 2023 07 10.
Article in English | MEDLINE | ID: mdl-37151182

ABSTRACT

Azaserine is a bacterial metabolite containing a biologically unusual and synthetically enabling α-diazoester functional group. Herein, we report the discovery of the azaserine (aza) biosynthetic gene cluster from Glycomyces harbinensis. Discovery of related gene clusters reveals previously unappreciated azaserine producers, and heterologous expression of the aza gene cluster confirms its role in azaserine assembly. Notably, this gene cluster encodes homologues of hydrazonoacetic acid (HYAA)-producing enzymes, implicating HYAA in α-diazoester biosynthesis. Isotope feeding and biochemical experiments support this hypothesis. These discoveries indicate that a 2-electron oxidation of a hydrazonoacetyl intermediate is required for α-diazoester formation, constituting a distinct logic for diazo biosynthesis. Uncovering this biological route for α-diazoester synthesis now enables the production of a highly versatile carbene precursor in cells, facilitating approaches for engineering complete carbene-mediated biosynthetic transformations in vivo.


Subject(s)
Azaserine , Biosynthetic Pathways , Biosynthetic Pathways/genetics , Methane , Oxidation-Reduction , Multigene Family
2.
Org Lett ; 25(22): 4061-4065, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37235858

ABSTRACT

Azaserine (1) is a natural product and nonproteinogenic amino acid containing a diazo group. Here we report the biosynthetic gene cluster for 1 from Glycomyces harbinensis. We then use isotopic feeding, gene deletion, and biochemical experiments to support a pathway whereby hydrazinoacetic acid (2) and a peptidyl carrier protein-loaded serine (3) are intermediates on route to the final natural product 1.


Subject(s)
Azaserine , Biological Products , Serine , Multigene Family , Hydrazines
3.
Nature ; 617(7960): 403-408, 2023 05.
Article in English | MEDLINE | ID: mdl-37138074

ABSTRACT

Biosynthesis is an environmentally benign and renewable approach that can be used to produce a broad range of natural and, in some cases, new-to-nature products. However, biology lacks many of the reactions that are available to synthetic chemists, resulting in a narrower scope of accessible products when using biosynthesis rather than synthetic chemistry. A prime example of such chemistry is carbene-transfer reactions1. Although it was recently shown that carbene-transfer reactions can be performed in a cell and used for biosynthesis2,3, carbene donors and unnatural cofactors needed to be added exogenously and transported into cells to effect the desired reactions, precluding cost-effective scale-up of the biosynthesis process with these reactions. Here we report the access to a diazo ester carbene precursor by cellular metabolism and a microbial platform for introducing unnatural carbene-transfer reactions into biosynthesis. The α-diazoester azaserine was produced by expressing a biosynthetic gene cluster in Streptomyces albus. The intracellularly produced azaserine was used as a carbene donor to cyclopropanate another intracellularly produced molecule-styrene. The reaction was catalysed by engineered P450 mutants containing a native cofactor with excellent diastereoselectivity and a moderate yield. Our study establishes a scalable, microbial platform for conducting intracellular abiological carbene-transfer reactions to functionalize a range of natural and new-to-nature products and expands the scope of organic products that can be produced by cellular metabolism.


Subject(s)
Azaserine , Azaserine/biosynthesis , Azaserine/chemistry , Biological Products/chemistry , Biological Products/metabolism , Multigene Family/genetics , Styrene/chemistry , Cyclopropanes/chemistry , Coenzymes/chemistry , Coenzymes/metabolism , Biocatalysis , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism
4.
Asian Pac J Cancer Prev ; 24(4): 1307-1312, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37116153

ABSTRACT

Hydatid cyst is a zoonotic infestation caused by Echinococcus granulosus, and it is known that some parasites found in humans cause cancer in humans or some may have a protective effect against cancer. Cancer is one of the most serious health problems of today and it has been shown in some studies that parasites such as Echinococcus granulosus can have an inhibitory effect. The aim of this study was determined as whether Echinococcus granulosus has an inhibitory effect on exocrine pancreatic cancer with the help of the azaserine-rat model used in different cancer studies.  Material and Methods: During experimental process a total of 45 male Wistar rats used, 14-day-old male Wistar rats were divided into groups according to the experimental protocol, administered azaserine injection protocol or kept as a control group without azaserine injection. Animals are grouped as Group 1, Control Group (group not treated with Azaserine and not injected with protoscolex.) (E-A-) (n=7); Group 2, Group injected with (IP) Azaserine only (30mg/kg) (E-A+)  (n=8);Group 3, Group injected (IP) with protoscolex suspension of 1 cc only (E+A-) (n=15);Group 4, Group injected both Azaserine (IP) and protoscolex suspension (IP) (E+A+) (n=15). Atypical Acinar Cell Foci (AACF) load in the exocrine pancreas of each rat was measured quantitatively with the help of a video image analyzer and the AACF load was calculated with the help of a mathematical model. Results: Findings showed that the Atypical Acinar Cell Foci (AACF) burden was statistically significantly lower in the Azaserine+ protoscolex (Azaserine-injected-protoscolex-implanted) rat group compared to the other groups, suggesting that Echinococcosis in the azaserine-rat model could inhibit the development of precursor foci of neoplastic changes in the exocrine pancreas. Conclusion: The most significant aspect of our study is that it contributes new insights into the controversy that Echinococcosis suppresses pancreatic cancer.


Subject(s)
Echinococcosis , Echinococcus granulosus , Pancreatic Neoplasms , Humans , Rats , Male , Animals , Rats, Wistar , Azaserine/pharmacology , Pancreatic Neoplasms/prevention & control , Pancreas , Pancreatic Neoplasms
5.
Chem Res Toxicol ; 34(6): 1518-1529, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34061515

ABSTRACT

The O6-alkylguanosine adduct O6-carboxymethyldeoxyguanosine (O6-CMdG) has been detected at elevated levels in blood and tissue samples from colorectal cancer patients and from healthy volunteers after consuming red meat. The diazo compound l-azaserine leads to the formation of O6-CMdG as well as the corresponding methyl adduct O6-methyldeoxyguanosine (O6-MedG) in cells and is therefore in wide use as a chemical probe in cellular studies concerning DNA damage and mutation. However, there remain knowledge gaps concerning the chemical basis of DNA adduct formation by l-azaserine. To characterize O6-CMdG formation by l-azaserine, we carried out a combination of chemical and enzymatic stability and reactivity studies supported by liquid chromatography tandem mass spectrometry for the simultaneous quantification of O6-CMdG and O6-MedG. We found that l-azaserine is stable under physiological and alkaline conditions as well as in active biological matrices but undergoes acid-catalyzed hydrolysis. We show, for the first time, that l-azaserine reacts directly with guanosine (dG) and oligonucleotides to form an O6-serine-CMdG (O6-Ser-CMdG) adduct. Moreover, by characterizing the reaction of dG with l-azaserine, we demonstrate that O6-Ser-CMdG forms as an intermediate that spontaneously decomposes to form O6-CMdG. Finally, we quantified levels of O6-CMdG and O6-MedG in a human cell line exposed to l-azaserine and found maximal adduct levels after 48 h. The findings of this work elucidate the chemical basis of how l-azaserine reacts with deoxyguanosine and support its use as a chemical probe for N-nitroso compound exposure in carcinogenesis research, particularly concerning the identification of pathways and factors that promote adduct formation.


Subject(s)
Azaserine/chemistry , Deoxyguanosine/chemical synthesis , Alkylation , Animals , Cells, Cultured , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/chemistry , Humans , Hydrogen-Ion Concentration , Molecular Structure , Swine
6.
Pancreas ; 50(4): 564-570, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33939670

ABSTRACT

OBJECTIVES: High-fat diet has been considered a risk factor for the development of pancreatic cancer. It is also shown to significantly impact composition and dysbiosis of gut microbiota in both humans and animals. However, there is little information on the effect of high-fat diet on the development of pancreatic cancer or upon the gut microbiota of patients with pancreatic cancer in humans or animal models. METHODS: In this study, the effect of high-fat diet on cancer pathology and the gut microbiota was investigated by a carcinogen-induced pancreatic cancer mouse model. RESULTS: Compared with carcinogen alone, mice with high-fat diet and carcinogen showed more obvious pathological changes in pancreatic tissue; increased levels of proinflammatory cytokine tumor necrosis factor-α, interleukin-6, interleukin-10, and carbohydrate antigen 242; and increased expression of cancer-associated biomarkers mucin-4 and claudin-4 in pancreatic tissue. Moreover, there is a significant change in the gut microbiota between the carcinogen group and the carcinogen with high-fat diet group. We identified that Johnsonella ignava especially existed in the carcinogen with high-fat diet group, which may contribute to pancreatic cancer development. CONCLUSIONS: Our results revealed that high-fat diet changed the composition of the gut microbiota and was involved in carcinogen-induced pancreatic cancer progression.


Subject(s)
Carcinogens/pharmacology , Diet, High-Fat/adverse effects , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/drug effects , Pancreatic Neoplasms/metabolism , Animals , Azaserine/pharmacology , Bacteria/classification , Bacteria/genetics , Claudin-4/metabolism , Cytokines/blood , Cytokines/metabolism , Drug Synergism , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Humans , Mice, Inbred C57BL , Mucin-4/metabolism , Pancreatic Neoplasms/etiology , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA/methods
7.
Nat Metab ; 2(12): 1401-1412, 2020 12.
Article in English | MEDLINE | ID: mdl-33257855

ABSTRACT

In non-small-cell lung cancer (NSCLC), concurrent mutations in the oncogene KRAS and the tumour suppressor STK11 (also known as LKB1) encoding the kinase LKB1 result in aggressive tumours prone to metastasis but with liabilities arising from reprogrammed metabolism. We previously demonstrated perturbed nitrogen metabolism and addiction to an unconventional pathway of pyrimidine synthesis in KRAS/LKB1 co-mutant cancer cells. To gain broader insight into metabolic reprogramming in NSCLC, we analysed tumour metabolomes in a series of genetically engineered mouse models with oncogenic KRAS combined with mutations in LKB1 or p53. Metabolomics and gene expression profiling pointed towards activation of the hexosamine biosynthesis pathway (HBP), another nitrogen-related metabolic pathway, in both mouse and human KRAS/LKB1 co-mutant tumours. KRAS/LKB1 co-mutant cells contain high levels of HBP metabolites, higher flux through the HBP pathway and elevated dependence on the HBP enzyme glutamine-fructose-6-phosphate transaminase [isomerizing] 2 (GFPT2). GFPT2 inhibition selectively reduced KRAS/LKB1 co-mutant tumour cell growth in culture, xenografts and genetically modified mice. Our results define a new metabolic vulnerability in KRAS/LKB1 co-mutant tumours and provide a rationale for targeting GFPT2 in this aggressive NSCLC subtype.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Hexosamines/biosynthesis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Metabolic Networks and Pathways , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , AMP-Activated Protein Kinase Kinases , Animals , Azaserine/therapeutic use , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/antagonists & inhibitors , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Humans , Lung Neoplasms/mortality , Metabolomics , Mice , Mutation , Survival Analysis , Tumor Stem Cell Assay
8.
Molecules ; 25(7)2020 Apr 02.
Article in English | MEDLINE | ID: mdl-32252252

ABSTRACT

Trypanosoma cruzi is the aetiologic agent of Chagas disease, which affects people in the Americas and worldwide. The parasite has a complex life cycle that alternates among mammalian hosts and insect vectors. During its life cycle, T. cruzi passes through different environments and faces nutrient shortages. It has been established that amino acids, such as proline, histidine, alanine, and glutamate, are crucial to T. cruzi survival. Recently, we described that T. cruzi can biosynthesize glutamine from glutamate and/or obtain it from the extracellular environment, and the role of glutamine in energetic metabolism and metacyclogenesis was demonstrated. In this study, we analysed the effect of glutamine analogues on the parasite life cycle. Here, we show that glutamine analogues impair cell proliferation, the developmental cycle during the infection of mammalian host cells and metacyclogenesis. Taken together, these results show that glutamine is an important metabolite for T. cruzi survival and suggest that glutamine analogues can be used as scaffolds for the development of new trypanocidal drugs. These data also reinforce the supposition that glutamine metabolism is an unexplored possible therapeutic target.


Subject(s)
Glutamine/analogs & derivatives , Life Cycle Stages/drug effects , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/growth & development , Animals , Azaserine/chemistry , Azaserine/pharmacology , CHO Cells , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cricetulus , Energy Metabolism/drug effects , Glutamic Acid/metabolism , Glutamine/metabolism , Isoxazoles/chemistry , Isoxazoles/pharmacology , Molecular Structure , Trypanocidal Agents/chemistry , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/metabolism
9.
Am J Physiol Endocrinol Metab ; 318(3): E405-E416, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31935112

ABSTRACT

The extent of glucose metabolism during oocyte maturation is closely related to oocyte developmental potential. Thioredoxin-interacting protein (TXNIP) is an α-arrestin family protein that negatively regulates glucose uptake into cells. However, little information is available regarding the function of TXNIP in bovine oocytes. Accordingly, the present study was performed to investigate the influence of TXNIP on glucose metabolism in bovine oocytes during in vitro maturation. Pharmacological inhibition of TXNIP by azaserine enhanced glucose uptake and imparted a specific metabolic effect on glycolysis and pentose phosphate pathway (PPP). RNA interference (RNAi) was adopted to further determine the biological significance of TXNIP in regulating glucose metabolism. The maturation rate and the developmental competence of TXNIP siRNA-treated oocytes were significantly improved. Knockdown of TXNIP in bovine oocytes significantly increased glycolysis by increasing the activities of phosphofructokinase (PFK), pyruvate kinase, and lactate dehydrogenase; pyruvate and lactate production; and intracellular ATP level, as well as mitochondrial activity. Furthermore, glucose metabolism through PPP was also enhanced by TXNIP depletion, as TXNIP siRNA treatment promoted glucose-6-phosphate dehydrogenase (G6PDH) activity and NADPH content, and helped maintain a high level of glutathione and a low level of reactive oxygen species within the oocytes. Further studies revealed that inhibition of TXNIP resulted increases in glucose transporter 1 (GLUT1) expression, as well as PFK1 platelet isoform (PFKP) and G6PDH mRNA levels. These results reveal that TXNIP depletion promotes oocyte maturation by enhancing both glycolysis and the PPP. During in vitro maturation of bovine oocytes, TXNIP serves as a key regulator of glucose uptake by controlling GLUT1 expression.


Subject(s)
Carrier Proteins/metabolism , Glucose/metabolism , Oocytes/metabolism , Adenosine Triphosphate/metabolism , Animals , Azaserine/pharmacology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cattle , Female , Gene Knockdown Techniques , Glycolysis , In Vitro Oocyte Maturation Techniques , Infertility, Female/genetics , Infertility, Female/metabolism , Intracellular Space/metabolism , Mitochondria/metabolism , Oxidation-Reduction , RNA Interference , RNA, Small Interfering/pharmacology
10.
Article in English | MEDLINE | ID: mdl-31655299

ABSTRACT

Cells under stress generate reactive oxygen species (ROS) in excess, which causes mitochondrial dysfunction and stimulates the apoptotic cascade. However, mild stress or pre-conditioning lead to the evasion of apoptosis by activating mitogenic signaling, including the signaling of inhibitors of apoptosis proteins (IAPs), or by inactivating certain apoptotic molecules. The silkworm (Bombyx mori) is an important economic insect which serves as a model organism in biological research. Bombyx mori apoptotic protease inducing factor (BmApaf1), a death-related ced-3/Nedd2-like protein (BmDredd), and BmSurvivin-2 (BmSvv2) are known to play significant roles in metamorphosis. Azaserine is an analogue of glutamine and irreversibly inhibits glutamine-utilizing enzymes and cysteine-glutamate transporter genes EAAT2. In the present study, we experimentally demonstrated stress induced by azaserine along with the capacity of antioxidants to modulate apoptotic/anti-apoptotic gene expression in determining the fate of the larvae. We observed higher larval survival with higher azaserine dosages and attributed this to the quantum of ROS generated and AOEs response, which favoured the BmSvv2 expression. Meanwhile higher levels of ROS with concomitant changes in AOEs were found to be responsible for BmApaf1 and BmDredd expression, which reflected a higher mortality rate.


Subject(s)
Azaserine/pharmacology , Bombyx/drug effects , Bombyx/physiology , Oxidative Stress , Animals , Apoptosis/physiology , Apoptotic Protease-Activating Factor 1/metabolism , Caspases/metabolism , Drosophila Proteins/metabolism , Insect Proteins/metabolism , Reactive Oxygen Species/metabolism , Survivin/metabolism
11.
Am J Chin Med ; 47(6): 1325-1343, 2019.
Article in English | MEDLINE | ID: mdl-31488031

ABSTRACT

Artemisinin and its analogues (ARTs) are currently the most effective anti-malarial drugs, but the precise mechanism of action is still highly controversial. Effects of ARTs on Plasmodium genes expression are studied in our Lab. The overexpression of an interesting amidotransferase, NADH-dependent glutamate synthase (NADH-GltS) was found in treated by dihydroartemisinin (DHA). The increased expression occurred not only from global transcriptomics analysis on the human malaria parasite Plasmodium falciparum (P. falciparum) 3D7 and gene expression screening on all of iron-sulphur cluster proteins from P.f. 3D7 in vitro but also from Plasmodium berghei (P. berghei) ANKA in mice. Influence of DHA on NADH-GltS was specifically at trophozoite stage of P. falciparum and in a dose-dependent manner below the effective doses. L-glutamine (Gln) and L-glutamate (Glu) are the substrate and product of NADH-GltS respectively. Azaserine (Aza) is specific inhibitor for NADH-GltS. Experimental data showed that Glu levels were significantly decreasing with DHA dose increasing but NADH-GltS enzyme activities were still remained at higher levels in parasites, and appropriate amount of exogenous Glu could significantly reduce anti-malarial action of DHA but excessive amount lost the above effect. Aza alone could inhibit proliferation of P. falciparum and had an additive effect in combination with DHA. Those results could suggest that: Glutamate depletion is one of the anti-malarial actions of DHA; overexpression of NADH-GltS would be a feedback pattern of parasite itself due to glutamate depletion, but not a direct action of DHA; the "feedback pattern" is one of protective strategies of Plasmodium to interfere with the anti-malarial actions of DHA; and specific inhibitor for NADH-GltS as a new type of anti-malarial agents or new partner in ACT might provide a potential.


Subject(s)
Antimalarials , Artemisinins/pharmacology , Artemisinins/therapeutic use , Gene Expression/drug effects , Glutamate Synthase (NADH)/genetics , Glutamate Synthase (NADH)/metabolism , Malaria/drug therapy , Phytotherapy , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Animals , Azaserine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Glutamate Synthase (NADH)/antagonists & inhibitors , Glutamic Acid/metabolism , Humans , Mice, Inbred C57BL , Plasmodium falciparum/physiology
12.
J Cancer Res Ther ; 15(1): 231-236, 2019.
Article in English | MEDLINE | ID: mdl-30880783

ABSTRACT

AIM: The effect of acetylsalicylic acid (ASA) on thiol levels was studied in a rat model of azaserine carcinogenesis. MATERIALS AND METHODS: ASA and azaserine were applied to the animals to research changes in cellular sulfhydryl (-SH) content and variations in free and protein-bound molecules containing the -SH group. Such effects in rats injected with azaserine were investigated at low (200 ppm) and high (400 ppm) concentrations of ASA over a relatively short (6 months) and a relatively long (12 months) period. RESULTS: Changes in the hepatic, pancreatic, and renal -SH contents were also determined. CONCLUSION: Compared to the other tissues studied, the liver contained the highest levels of both free and protein-bound -SH.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aspirin/administration & dosage , Azaserine/toxicity , Carcinogens/toxicity , Neoplasms/prevention & control , Animals , Dose-Response Relationship, Drug , Humans , Kidney/chemistry , Kidney/drug effects , Kidney/pathology , Liver/chemistry , Liver/drug effects , Liver/pathology , Male , Neoplasms/chemically induced , Neoplasms/pathology , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Pancreas/chemistry , Pancreas/drug effects , Pancreas/pathology , Rats , Rats, Wistar , Sulfhydryl Compounds/analysis , Treatment Outcome
13.
Anal Chem ; 90(8): 5201-5208, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29533603

ABSTRACT

The structures and amounts of glycosaminoglycan (GAG) produced by cells have attracted much interest because GAG biosynthesis activity can change in cellular processes such as disease and differentiation. ß-Xylosides, also called saccharide primers, have been used as artificial acceptors not only to generate GAG oligosaccharides in cells and tissues but also to investigate their biosynthetic pathways. Various analytical methods have been applied to confirm the structure and amounts of GAG oligosaccharides elongated using saccharide primers, yet sample preparation processes such as solid-phase extraction in analysis can cause experimental error and disrupt accurate comparative quantification of glycosylated products. In this study, we developed a new quantification method using a deuterium-labeled saccharide primer. The "heavy" and "light" primers were chemically synthesized, and priming abilities were confirmed by liquid chromatography-tandem mass spectrometry. Relative peak areas of light/heavy products showed good linearity and were well correlated with the theoretical amounts of glycosylated products. Then, as a validation study, we carried out a biosynthesis inhibition assay using known GAG biosynthesis inhibitors. According to the relative quantification using saccharide primers, differences in the mode-of-action among the four GAG biosynthesis inhibitors were dependent on the GAG biosynthetic pathway. Our results indicate that the method will likely forge a new path for comparative glycosaminoglycomics using cultured cells and tissues.


Subject(s)
Glycosaminoglycans/analysis , Glycosides/chemistry , Isotope Labeling , Oligosaccharides/chemistry , Azaserine/pharmacology , Brefeldin A/pharmacology , Cell Line , Genistein/pharmacology , Glycosaminoglycans/antagonists & inhibitors , Glycosaminoglycans/biosynthesis , Glycosylation , Humans , Molecular Structure , Rhodamines/pharmacology
14.
Cell Rep ; 22(8): 1945-1955, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466723

ABSTRACT

Sirtuin 3 (SIRT3) is a NAD+-dependent deacetylase downregulated in aging and age-associated diseases such as cancer and neurodegeneration and in high-fat diet (HFD)-induced metabolic disorders. Here, we performed a small-molecule screen and identified an unexpected metabolic vulnerability associated with SIRT3 loss. Azaserine, a glutamine analog, was the top compound that inhibited growth and proliferation of cells lacking SIRT3. Using stable isotope tracing of glutamine, we observed its increased incorporation into de novo nucleotide synthesis in SIRT3 knockout (KO) cells. Furthermore, we found that SIRT3 KO cells upregulated the diversion of glutamine into de novo nucleotide synthesis through hyperactive mTORC1 signaling. Overexpression of SIRT3 suppressed mTORC1 and growth in vivo in a xenograft tumor model of breast cancer. Thus, we have uncovered a metabolic vulnerability of cells with SIRT3 loss by using an unbiased small-molecule screen.


Subject(s)
Nucleotides/biosynthesis , Sirtuin 3/deficiency , Small Molecule Libraries/pharmacology , Amino Acid Sequence , Animals , Azaserine/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Glutamine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout , Mice, Nude , Promoter Regions, Genetic/genetics , Signal Transduction/drug effects , Sirtuin 3/metabolism , Up-Regulation/drug effects
15.
Nat Commun ; 9(1): 374, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371602

ABSTRACT

AMP-activated protein kinase (AMPK) has been shown to inhibit cardiac hypertrophy. Here, we show that submaximal AMPK activation blocks cardiomyocyte hypertrophy without affecting downstream targets previously suggested to be involved, such as p70 ribosomal S6 protein kinase, calcineurin/nuclear factor of activated T cells (NFAT) and extracellular signal-regulated kinases. Instead, cardiomyocyte hypertrophy is accompanied by increased protein O-GlcNAcylation, which is reversed by AMPK activation. Decreasing O-GlcNAcylation by inhibitors of the glutamine:fructose-6-phosphate aminotransferase (GFAT), blocks cardiomyocyte hypertrophy, mimicking AMPK activation. Conversely, O-GlcNAcylation-inducing agents counteract the anti-hypertrophic effect of AMPK. In vivo, AMPK activation prevents myocardial hypertrophy and the concomitant rise of O-GlcNAcylation in wild-type but not in AMPKα2-deficient mice. Treatment of wild-type mice with O-GlcNAcylation-inducing agents reverses AMPK action. Finally, we demonstrate that AMPK inhibits O-GlcNAcylation by mainly controlling GFAT phosphorylation, thereby reducing O-GlcNAcylation of proteins such as troponin T. We conclude that AMPK activation prevents cardiac hypertrophy predominantly by inhibiting O-GlcNAcylation.


Subject(s)
AMP-Activated Protein Kinases/genetics , Acetylglucosamine/metabolism , Cardiomegaly/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nitrogenous Group Transferases/genetics , AMP-Activated Protein Kinases/deficiency , Acetylglucosamine/pharmacology , Acylation/drug effects , Animals , Animals, Newborn , Azaserine/pharmacology , Azo Compounds/pharmacology , Biphenyl Compounds , Cardiomegaly/metabolism , Cardiomegaly/pathology , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Gene Expression Regulation , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) , Glycosylation/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/pathology , Male , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Nitrogenous Group Transferases/antagonists & inhibitors , Nitrogenous Group Transferases/metabolism , Norleucine/analogs & derivatives , Norleucine/pharmacology , Phosphorylation/drug effects , Primary Cell Culture , Pyrones/pharmacology , Rats , Rats, Wistar , Signal Transduction , Thiophenes/pharmacology , Troponin T/genetics , Troponin T/metabolism
16.
Oncotarget ; 7(49): 80599-80611, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27716624

ABSTRACT

The hexosamine biosynthetic pathway (HBP) requires two key nutrients glucose and glutamine for O-linked N-acetylglucosamine (O-GlcNAc) cycling, a post-translational protein modification that adds GlcNAc to nuclear and cytoplasmic proteins. Increased GlcNAc has been linked to regulatory factors involved in cancer cell growth and survival. However, the biological significance of GlcNAc in diffuse large B-cell lymphoma (DLBCL) is not well defined. This study is the first to show that both the substrate and the endpoint O-GlcNAc transferase (OGT) enzyme of the HBP were highly expressed in DLBCL cell lines and in patient tumors compared with normal B-lymphocytes. Notably, high OGT mRNA levels were associated with poor survival of DLBCL patients. Targeting OGT via small interference RNA in DLBCL cells inhibited activation of GlcNAc, nuclear factor kappa B (NF-κB), and nuclear factor of activated T-cells 1 (NFATc1), as well as cell growth. Depleting both glucose and glutamine in DLBCL cells or treating them with an HBP inhibitor (azaserine) diminished O-GlcNAc protein substrate, inhibited constitutive NF-κB and NFATc1 activation, and induced G0/G1 cell-cycle arrest and apoptosis. Replenishing glucose-and glutamine-deprived DLBCL cells with a synthetic glucose analog (ethylenedicysteine-N-acetylglucosamine [ECG]) reversed these phenotypes. Finally, we showed in both in vitro and in vivo murine models that DLBCL cells easily take up radiolabeled technetium-99m-ECG conjugate. These findings suggest that targeting the HBP has therapeutic relevance for DLBCL and underscores the imaging potential of the glucosamine analog ECG in DLBCL.


Subject(s)
Acetylglucosamine/administration & dosage , Antineoplastic Agents/pharmacology , Azaserine/pharmacology , Contrast Media/administration & dosage , Cysteine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Hexosamines/biosynthesis , Lymphoma, Large B-Cell, Diffuse/diagnostic imaging , Lymphoma, Large B-Cell, Diffuse/therapy , N-Acetylglucosaminyltransferases/metabolism , Organotechnetium Compounds/administration & dosage , RNAi Therapeutics , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine/administration & dosage , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glycosylation , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice, Inbred NOD , Mice, SCID , N-Acetylglucosaminyltransferases/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transfection
17.
Anal Chem ; 88(16): 8036-42, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27441891

ABSTRACT

Humans are exposed to N-nitroso compounds through environmental exposure and endogenous metabolism. Some N-nitroso compounds can be metabolically activated to yield diazoacetate, which is known to induce DNA carboxymethylation. DNA lesion measurement remains one of the core tasks in toxicology and in evaluating human health risks associated with carcinogen exposure. In this study, we developed a highly sensitive nanoflow liquid chromatography-nanoelectrospray ionization-multistage tandem mass spectrometry (nLC-nESI-MS(3)) method for the simultaneous quantification of O(6)-carboxymethyl-2'-deoxyguanosine (O(6)-CMdG), O(6)-methyl-2'-deoxyguanosine (O(6)-MedG), and N(6)-carboxymethyl-2'-deoxyadenosine (N(6)-CMdA). We were able to measure the levels of these three lesions with the use of low-microgram quantities of DNA from cultured human skin fibroblasts and human colorectal carcinoma cells treated with azaserine, a DNA carboxymethylating agent. Our results revealed that the levels of O(6)-CMdG and O(6)-MedG increased when the dose of azaserine was increased from 0 to 450 µM. We, however, did not observe an apparent dose-dependent induction of N(6)-CMdA, suggesting the presence of repair mechanism(s) for the rapid clearance of this lesion in cells. This is the first report about the application of nLC-nESI-MS(3) technique for the simultaneous quantification of O(6)-CMdG, O(6)-MedG, and N(6)-CMdA. The method reported here will be useful for future investigations about the repair of the carboxymethylated DNA lesions and about the implications of these lesions in carcinogenesis.


Subject(s)
Azaserine/analysis , Nanotechnology , Azaserine/pharmacology , Cells, Cultured , Chromatography, Liquid , DNA Damage , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Humans , Indicator Dilution Techniques , Methylation/drug effects , Molecular Structure , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Tandem Mass Spectrometry
18.
Med Sci Monit ; 22: 1460-6, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27131835

ABSTRACT

BACKGROUND Diabetic retinopathy (DR) is a progressive neurodegenerative disease with early-stage symptoms such as dysfunction of Muller cells, which leads to ganglion cell death. Its pathogenesis is probably associated with oxidative stress and a recently discovered protein, thioredoxin-interacting protein (TXNIP). MATERIAL AND METHODS To explore the role of TXNIP in DR, we cultured Muller cells under diabetic conditions, and then used immunohistochemistry, Western blot, and RT-PCR to detect the expression level of TXNIP under diabetic conditions. We demonstrated the expression level of glutamine synthetase (GS) when TXNIP was inhibited. To explore the potential pathway of TXNIP-induced cell damage in DR, we confirmed the role of IL-1ß under diabetic conditions. RESULTS Diabetes induces TXNIP expressions at mRNA levels, but shows the opposite effect on GS. IL-1ß plays an important role in this pathway. Azaserine effectively increased the expression of GS via attenuating the expression of TXNIP. CONCLUSIONS This study demonstrates the role of TXNIP and its mechanism in DR, provides a possible treatment for DR, and lays a new theoretical foundation for the clinical treatment of DR and other diabetic microvascular changes.


Subject(s)
Carrier Proteins/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Ependymoglial Cells/pathology , Glutamate-Ammonia Ligase/metabolism , Oxidative Stress , Retina/pathology , Thioredoxins/metabolism , Animals , Azaserine/metabolism , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Fluorescent Antibody Technique , Gene Expression Regulation , Glucose/toxicity , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Sci Rep ; 6: 21184, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26878908

ABSTRACT

Cancer was hypothesized to be driven by cancer stem cells (CSCs), but the metabolic determinants of CSC-like phenotype still remain elusive. Here, we present that hexosamine biosynthetic pathway (HBP) at least in part rescues cancer cell fate with inactivation of glycolysis. Firstly, metabolomic analysis profiled cellular metabolome in CSCs of hepatocellular carcinoma using CD133 cell-surface marker. The metabolic signatures of CD133-positive subpopulation compared to CD133-negative cells highlighted HBP as one of the distinct metabolic pathways, prompting us to uncover the role of HBP in maintenance of CSC-like phenotype. To address this, CSC-like phenotypes and cell survival were investigated in cancer cells under low glucose conditions. As a result, HBP inhibitor azaserine reduced CD133-positive subpopulation and CD133 expression under high glucose condition. Furthermore, treatment of N-Acetylglucosamine in part restores CD133-positive subpopulation when either 2.5 mM glucose in culture media or glycolytic inhibitor 2-deoxy-D-glucose in HCC cell lines was applied, enhancing CD133 expression as well as promoting cancer cell survival. Together, HBP might be a key metabolic determinant in the functions of hepatic CSC marker CD133.


Subject(s)
AC133 Antigen/metabolism , Biosynthetic Pathways , Carcinoma, Hepatocellular/metabolism , Glucose/metabolism , Hexosamines/biosynthesis , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Azaserine/pharmacology , Biomarkers , Biosynthetic Pathways/drug effects , Cell Line, Tumor , Cell Survival , Glycolysis , Humans , Metabolomics/methods , Phenotype
20.
Lipids Health Dis ; 14: 110, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26377577

ABSTRACT

BACKGROUND: We previously demonstrated that hyperglycemia could suppress apolipoprotein M (apoM) synthesis both in vivo and in vitro; however, the mechanism of hyperglycemia-induced downregulation of apoM expression is unknown yet. METHODS: In the present study we further examined if hexosamine pathway, one of the most important pathways of glucose turnover, being involved in modulating apoM expression in the hyperglycemia condition. We examined the effect of glucosamine, a prominent component of hexosamine pathway and intracellular mediator of insulin resistance, on apoM expression in HepG2 cells and in rat's models. In the present study we also determined apolipoprotein A1 (apoA1) as a control gene. RESULTS: Our results demonstrated that glucosamine could even up-regulate both apoM and apoA1 expressions in HepG2 cell cultures. The glucosamine induced upregulation of apoM expression could be blocked by addition of azaserine, an inhibitor of hexosamine pathway. Moreover, intravenous infusion of glucosamine could enhance hepatic apoM expression in rats, although serum apoM levels were not significantly influences. CONCLUSIONS: It is concluded that both exogenous and endogenous glucosamine were essential for the over-expression of apoM, which may suggest that the increased intracellular content of glucosamine does not be responsible for the depressed apoM expression at hyperglycemia condition.


Subject(s)
Apolipoprotein A-I/genetics , Apolipoproteins/genetics , Hyperglycemia/genetics , Lipocalins/genetics , Liver/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Apolipoprotein A-I/metabolism , Apolipoproteins/metabolism , Apolipoproteins M , Azaserine/pharmacology , Gene Expression Regulation , Glucosamine/administration & dosage , Glucosamine/metabolism , Hep G2 Cells , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Infusions, Intravenous , Lipocalins/metabolism , Liver/pathology , Male , Rats , Rats, Sprague-Dawley , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...