Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mar Drugs ; 19(11)2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34822459

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a devastating digestive system carcinoma with high incidence and death rates. PDAC cells are dependent on the Gln metabolism, which can preferentially utilize glutamic oxaloacetate transaminase 1 (GOT1) to maintain the redox homeostasis of cancer cells. Therefore, small molecule inhibitors targeting GOT1 can be used as a new strategy for developing cancer therapies. In this study, 18 butyrolactone derivatives (1-18) were isolated from a marine-derived Aspergillus terreus, and asperteretone B (5), aspulvinone H (AH, 6), and (+)-3',3'-di-(dimethylallyl)-butyrolactone II (12) were discovered to possess significant GOT1-inhibitory activities in vitro, with IC50 values of (19.16 ± 0.15), (5.91 ± 0.04), and (26.38 ± 0.1) µM, respectively. Significantly, the molecular mechanism of the crystal structure of GOT1-AH was elucidated, wherein AH and the cofactor pyrido-aldehyde 5-phosphate competitively bound to the active sites of GOT1. More importantly, although the crystal structure of GOT1 has been discovered, the complex structure of GOT1 and its inhibitors has never been obtained, and the crystal structure of GOT1-AH is the first reported complex structure of GOT1/inhibitor. Further in vitro biological study indicated that AH could suppress glutamine metabolism, making PDAC cells sensitive to oxidative stress and inhibiting cell proliferation. More significantly, AH exhibited potent in vivo antitumor activity in an SW1990-cell-induced xenograft model. These findings suggest that AH could be considered as a promising lead molecule for the development of anti-PDAC agents.


Subject(s)
Antineoplastic Agents/pharmacology , Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Aspergillus , Enzyme Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Aquatic Organisms , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Inhibitory Concentration 50 , Pancreatic Neoplasms/drug therapy , Xenograft Model Antitumor Assays
2.
Nat Commun ; 12(1): 4860, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381026

ABSTRACT

Cancer metabolism is rewired to support cell survival in response to intrinsic and environmental stressors. Identification of strategies to target these adaptions is an area of active research. We previously described a cytosolic aspartate aminotransaminase (GOT1)-driven pathway in pancreatic cancer used to maintain redox balance. Here, we sought to identify metabolic dependencies following GOT1 inhibition to exploit this feature of pancreatic cancer and to provide additional insight into regulation of redox metabolism. Using pharmacological methods, we identify cysteine, glutathione, and lipid antioxidant function as metabolic vulnerabilities following GOT1 withdrawal. We demonstrate that targeting any of these pathways triggers ferroptosis, an oxidative, iron-dependent form of cell death, in GOT1 knockdown cells. Mechanistically, we reveal that GOT1 inhibition represses mitochondrial metabolism and promotes a catabolic state. Consequently, we find that this enhances labile iron availability through autophagy, which potentiates the activity of ferroptotic stimuli. Overall, our study identifies a biochemical connection between GOT1, iron regulation, and ferroptosis.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Ferroptosis , Pancreatic Neoplasms/metabolism , Animals , Antioxidants/pharmacology , Aspartate Aminotransferase, Cytoplasmic/genetics , Aspartate Aminotransferase, Cytoplasmic/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Cystine/metabolism , Ferroptosis/drug effects , Glutathione/biosynthesis , Humans , Iron/metabolism , Mice , Mitochondria/metabolism , Pancreatic Neoplasms/pathology
3.
Transl Res ; 230: 68-81, 2021 04.
Article in English | MEDLINE | ID: mdl-33132087

ABSTRACT

Glutamate oxaloacetate transaminase 1 (GOT1) enzyme plays a critical role in the cell metabolism by participating in the carbohydrate and amino acid metabolism. In ischemic stroke, we have demonstrated that recombinant GOT1 acts as a novel neuroprotective treatment against the excess of extracellular glutamate that accumulates in the brain following ischemic stroke. In this study, we investigated the inhibitory effect of GOT1 on brain metabolism and on the ischemic damage in a rat model of ischemic stroke by means of a specific antibody developed against this enzyme. Inhibition of GOT1 caused higher brain glutamate and lactate levels and this response was associated with larger ischemic lesion. This study represents the first demonstration that the inhibition of the blood GOT1 activity leads to more severe ischemic damage and poorer outcome and supports the protective role of GOT1 against ischemic insults.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Aspartate Aminotransferase, Cytoplasmic/metabolism , Brain Ischemia/enzymology , Brain Ischemia/pathology , Animals , Antibodies , Aspartate Aminotransferase, Cytoplasmic/cerebrospinal fluid , Brain/enzymology , Cloning, Molecular , Dose-Response Relationship, Immunologic , Glutamic Acid/blood , Hep G2 Cells , Humans , Immunoglobulin G , Lactic Acid/blood , Male , Rats , Rats, Sprague-Dawley
4.
Biochem Biophys Res Commun ; 522(3): 633-638, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31787239

ABSTRACT

Metabolic programs are rewired in cancer cells to support survival and tumor growth. Among these, recent studies have demonstrated that glutamate-oxaloacetate transaminase 1 (GOT1) plays key roles in maintaining redox homeostasis and proliferation of pancreatic ductal adenocarcinomas (PDA). This suggests that small molecule inhibitors of GOT1 could have utility for the treatment of PDA. However, the development of GOT1 inhibitors has been challenging, and no compound has yet demonstrated selectivity for GOT1-dependent cell metabolism or selective growth inhibition of PDA cell lines. In contrast, potent inhibitors that covalently bind to the transaminase cofactor pyridoxal-5'-phosphate (PLP), within the active site of the enzyme, have been reported for kynurenine aminotransferase (KAT) and gamma-aminobutyric acid aminotransferase (GABA-AT). Given the drug discovery successes with these transaminases, we aimed to identify PLP-dependent suicide substrate-type GOT1 inhibitors. Here, we demonstrate that PF-04859989, a known KAT2 inhibitor, has PLP-dependent inhibitory activity against GOT1 and shows selective growth inhibition of PDA cell lines.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Carcinoma, Pancreatic Ductal/drug therapy , Enzyme Inhibitors/pharmacology , Pancreatic Neoplasms/drug therapy , Pyrazoles/pharmacology , Aspartate Aminotransferase, Cytoplasmic/metabolism , Carcinoma, Pancreatic Ductal/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Humans , Pancreatic Neoplasms/enzymology
5.
Bioorg Chem ; 93: 103315, 2019 12.
Article in English | MEDLINE | ID: mdl-31605927

ABSTRACT

Glutamic-oxaloacetic transaminase 1 (GOT1) regulates cellular metabolism through coordinating the utilization of carbohydrates and amino acids to meet nutrient requirements for sustained proliferation. As such, the GOT1 inhibitor may provide a new strategy for the treatment of various cancers. Adapalene has been approved by FDA for the treatment of acne, pimples and pustules, and it may also contribute to the adjunctive therapy for advanced stages of liver and colorectal cancers. In this work, we first examined the enzyme inhibition of over 500 compounds against GOT1 in vitro. As a result, Adapalene effectively inhibited GOT1 enzyme in a non-competitive manner. MST and DARTS assay further confirmed the high affinity between Adapalene and GOT1. Furthermore, the growth and migration of ovarian cancer ES-2 cells were obviously inhibited by the treatment of Adapalene. And it induced the apoptosis of ES-2 cells according to Western blot and Hoechst 33258 straining. In addition, molecular docking demonstrated that Adapalene coordinated in an allosteric site of GOT1 with low binding energy. Furthermore, knockdown of GOT1 in ES-2 cells decreased their anti-proliferative sensitivity to Adapalene. Together, our data strongly suggest Adapalene, as a GOT1 inhibitor, could be regarded as a potential drug candidate for ovarian cancer therapy.


Subject(s)
Adapalene/chemistry , Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Adapalene/metabolism , Adapalene/pharmacology , Allosteric Site , Aspartate Aminotransferase, Cytoplasmic/genetics , Aspartate Aminotransferase, Cytoplasmic/metabolism , Binding Sites , Catalytic Domain , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Kinetics , Molecular Docking Simulation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism
6.
Biochemistry ; 57(47): 6604-6614, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30365304

ABSTRACT

Pancreatic cancer cells are characterized by deregulated metabolic programs that facilitate growth and resistance to oxidative stress. Among these programs, pancreatic cancers preferentially utilize a metabolic pathway through the enzyme aspartate aminotransferase 1 [also known as glutamate oxaloacetate transaminase 1 (GOT1)] to support cellular redox homeostasis. As such, small molecule inhibitors that target GOT1 could serve as starting points for the development of new therapies for pancreatic cancer. We ran a high-throughput screen for inhibitors of GOT1 and identified a small molecule, iGOT1-01, with in vitro GOT1 inhibitor activity. Application in pancreatic cancer cells revealed metabolic and growth inhibitory activity reflecting a promiscuous inhibitory profile. We then performed an in silico docking analysis to study inhibitor-GOT1 interactions with iGOT1-01 analogues that possess improved solubility and potency properties. These results suggested that the GOT1 inhibitor competed for binding to the pyridoxal 5-phosphate (PLP) cofactor site of GOT1. To analyze how the GOT1 inhibitor bound to GOT1, a series of GOT1 mutant enzymes that abolished PLP binding were generated. Application of the mutants in X-ray crystallography and thermal shift assays again suggested but were unable to formally conclude that the GOT1 inhibitor bound to the PLP site. Mutational studies revealed the relationship between PLP binding and the thermal stability of GOT1 while highlighting the essential nature of several residues for GOT1 catalytic activity. Insight into the mode of action of GOT1 inhibitors may provide leads to the development of drugs that target redox balance in pancreatic cancer.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Mutation , Pancreatic Neoplasms/pathology , Aspartate Aminotransferase, Cytoplasmic/genetics , Aspartate Aminotransferase, Cytoplasmic/metabolism , Binding Sites , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Humans , Metabolomics , Models, Molecular , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Protein Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Cancer Chemother Pharmacol ; 79(4): 835-840, 2017 04.
Article in English | MEDLINE | ID: mdl-28314989

ABSTRACT

PURPOSE: Almost all colorectal cancer (CRC) cell lines are known to overexpress aspartate aminotransferase (GOT1), which potentially regulates the intracellular levels of reactive oxygen species (ROS) via the production of NADPH, and supports tumor growth. In our study, the role of GOT1 in the anticancer efficacy of 5-fluorouracil (5-FU) was examined. METHODS: HCT116, SW480, and HT-29 cells were transfected with lentiviral vectors expressing short hairpin RNA (shRNA) against GOT1. Following 5-FU treatment, cellular proliferation was evaluated, the NADP+/NADPH ratio was monitored, ROS was measured, and intracellular levels of glutamine (Gln), Aspartate (Asp), oxaloacetate (OAA), malate, and pyruvate were investigated using liquid chromatography-mass spectrometry (LC-MS). A CRC subcutaneous tumor model was performed to determine the impact of GOT1 inhibition on 5-FU efficacy in vivo. RESULTS: In response to 5-FU administration, CRC cells undergo metabolic adaptation, resulting in increased glutamine flux for the synthesis of aspartate. GOT1 is responsible for the conversion of glutamine-derived aspartate into OAA, which subsequently can be converted into malate and pyruvate. The GOT1-mediated metabolic process is able to maintain the NADP+/NADPH ratio, which counteracts 5-FU-induced oxidative stress. Inhibition of GOT1 impaired the defense against 5-FU-induced ROS, thereby sensitizing cells to 5-FU. The importance of GOT1 in supporting tumor growth during 5-FU treatment was also indicated in an in vivo tumor model of CRC. CONCLUSION: These findings show that GOT1 could serve as a promising target for increasing the anticancer efficacy of the conventional therapy in patients with CRC.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , Fluorouracil/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , HT29 Cells , Humans , Lentivirus/genetics , NADP/metabolism , Oxidative Stress/drug effects , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Transfection
8.
J Neurosurg Anesthesiol ; 21(3): 235-41, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19543002

ABSTRACT

INTRODUCTION: Treatment with oxaloacetate after traumatic brain injury has been shown to decrease blood glutamate levels and protect against the neurotoxic effects of glutamate on the brain. A number of potential mechanisms have been suggested to explain oxaloacetate-induced neuroprotection. We hypothesize that the primary mechanism by which intravenous oxaloacetate provides neuroprotection is by activation of the blood glutamate-scavenging enzyme glutamate-oxaloacetate transaminase, increasing thereby the driving force for the efflux of excess glutamate from brain interstitial fluids into blood. If so, coadministration of maleate, a glutamate-oxaloacetate transaminase-blocker is expected to prevent the neuroprotective effects of oxaloacetate. MATERIALS AND METHODS: A neurological severity score (NSS) was measured 1 hour after closed head injury (CHI) in rats. Then, rats received 30 microL/min/100 g infusion of saline, or 1 mmol/100 g solution of oxaloacetate, maleate, or a mixture of oxaloacetate and maleate. NSS was reassessed at 24 and 48 hour after CHI. Blood glutamate and glucose levels were measured at 0, 60, 90, and 120 minutes. RESULTS: NSS improved significantly at 24 hour (P<0.001) and 48 hour (P<0.001) only in the rats treated with oxaloacetate. Blood glutamate decreased significantly in the oxaloacetate-treated group at 90 minute (at the conclusion of oxaloacetate administration) (P<0.00001), but not in the control, maleate or oxaloacetate+maleate groups. A strong correlation r2=0.86 was found to exist between the percent decrease in blood glutamate levels and percent improvement in NSS. DISCUSSION: The results of this study demonstrate that the primary mechanism by which oxaloacetate provides neuroprotective activity after CHI is related to its blood glutamate scavenging activity. Management of blood glutamate concentration may have important implications in the treatment of acute brain conditions, including CHI and stroke.


Subject(s)
Enzyme Inhibitors/pharmacology , Glutamic Acid/blood , Head Injuries, Closed/drug therapy , Maleates/pharmacology , Neuroprotective Agents , Oxaloacetic Acid/pharmacology , Animals , Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Aspartate Aminotransferase, Cytoplasmic/metabolism , Behavior, Animal/drug effects , Blood Gas Analysis , Head Injuries, Closed/psychology , Male , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley
9.
Biol Reprod ; 80(2): 295-301, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18971426

ABSTRACT

The nutrient requirements and metabolic pathways used by the developing embryo transition from predominantly pyruvate during early cleavage stages to glucose at the blastocyst; however, the complexities involved in the regulation of metabolism at different developmental stages are not clear. The aims of this study were to examine the role of the malate-aspartate shuttle (MAS) in nutrient metabolism pathways in the developing mouse blastocyst and the consequences of impaired metabolism on embryo viability and fetal and placental growth. Eight-cell-stage mouse embryos were cultured in the presence of the MAS inhibitor amino-oxyacetate, with or without pyruvate as an energy substrate in the media. When the MAS was inhibited, the rate of glycolysis and lactate production was significantly elevated and glucose uptake reduced, relative to control cultured embryos in the presence of pyruvate. Despite these changes in embryo metabolism, this did not influence development to the blastocyst stage, but it did reduce the number of inner cell mass and trophectoderm cells. When these embryos were transferred to psuedopregnant females, inhibition of the MAS significantly reduced the proportion of embryos that implanted and developed into fetuses on Day 18 of pregnancy. Finally, fetal growth was reduced while placental weight was maintained, leading to a decreased fetal:placental weight ratio relative to control embryos. These results suggest that impaired metabolism of glucose in the blastocyst via the MAS alters the ability of the embryos to implant and form a pregnancy and leads to reduced fetal weight, likely via altered placental development and function.


Subject(s)
Aminooxyacetic Acid/pharmacology , Blastocyst/metabolism , Fetal Development/drug effects , Fetal Viability/drug effects , Mitochondria/metabolism , Animals , Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Aspartate Aminotransferase, Cytoplasmic/metabolism , Aspartic Acid/metabolism , Biological Transport/drug effects , Blastocyst/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Female , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/metabolism , Malates/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mitochondria/drug effects , Mitochondria/genetics , Pregnancy
10.
J Enzyme Inhib Med Chem ; 17(1): 19-23, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12365456

ABSTRACT

Acrolein is a reactive lipid peroxidation byproduct, which is found in ischemic tissue. We examined the effects of acrolein on cytosolic aspartate aminotransferase (cAAT), which is an enzyme that was previously shown to be inhibited by glycating agents. cAAT is thought to protect against ischemic injury. We observed that acrolein cross-linked cAAT subunits as evidenced by the presence of high molecular weight bands following SDS-PAGE. Acrolein-modified cAAT resisted thermal denaturation when compared with native cAAT. We also observed a decrease in intrinsic fluorescence (290 nm, ex; 380 nm, em). These observations are consistent with an acrolein-induced change in conformation that is more rigid and compact than native cAAT, suggesting that intramolecular cross-links occurred. Acrolein also inhibited activity, and the inhibition of enzyme activity correlated with the acrolein-induced formation of cAAT cross-links.


Subject(s)
Acrolein/pharmacology , Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Acrolein/chemistry , Animals , Aspartate Aminotransferase, Cytoplasmic/chemistry , Cross-Linking Reagents , Ischemia/metabolism , Protein Conformation/drug effects , Protein Denaturation/drug effects , Protein Subunits , Swine , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...