Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 314
Filter
1.
Diabetes Metab Syndr ; 15(6): 102311, 2021.
Article in English | MEDLINE | ID: mdl-34678576

ABSTRACT

BACKGROUND AND AIMS: Possible protective effects of saffron (Crocus sativus L) have been reported in several randomized clinical trials (RCTs). Current systematic review was performed to summarize the efficacy of saffron intake on liver enzymes. METHODS: An electronic database search was conducted on PubMed/Medline, Scopus, Web of Science, and Cochrane for RCTs comparing effect of saffron and placebo on liver enzymes from inception to July 2021. There was no restriction in language of included studies and we calculated the standardized mean difference (SMD) and 95% Confidence Intervals (CI) for each variable. Random-effect model was used to calculate effect size. RESULTS: Eight studies (n = 463 participants) were included in the systematic review. The saffron intake was associated with a statistically significant decrease in aspartate aminotransferase (AST) (SMD: -0.18; 95% CI: -0.34, -0.02; I2 = 0%) in comparison to placebo intake. Our results also indicated that saffron consumption did not have a significant effect on alanine aminotransferase (ALT) (SMD: -0.14; 95% CI: -0.36, 0.09; I2 = 47.0%) and alkaline phosphatase (ALP) levels (SMD: 0.14; 95% CI: -0.18, 0.46; I2 = 42.9%) compared to placebo. CONCLUSIONS: Saffron intake showed beneficial impacts on circulating AST levels. However, larger well-designed RCTs are still needed to clarify the effect of saffron intake on these and other liver enzymes.


Subject(s)
Aspartate Aminotransferases/antagonists & inhibitors , Crocus , Dietary Supplements , Liver/drug effects , Liver/enzymology , Randomized Controlled Trials as Topic/methods , Alanine Transaminase/antagonists & inhibitors , Alanine Transaminase/blood , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Aspartate Aminotransferases/blood , Humans
2.
Drug Des Devel Ther ; 14: 5515-5520, 2020.
Article in English | MEDLINE | ID: mdl-33364747

ABSTRACT

OBJECTIVE: We aimed to investigate the effect of switching from tenofovir disoproxil fumarate (TDF) to tenofovir alafenamide (TAF) on the hepatic safety and metabolic profile. METHODS: Consecutive HIV patients, enrolled in the Surveillance Cohort Long-term Toxicity Antiretrovirals/Antivirals (SCOLTA) project, switching from TDF to TAF were included. Changes from baseline (T0) to 6-month follow-up (T1) were evaluated using paired t-test and signed rank test. RESULTS: A total of 190 patients switched from TDF to TAF and had one 6-month follow-up visit. They were 80% male, 74.2% at CDC stage A-B, 93.7% with undetectable HIV-viral load. Mean age was 46.7±10.7 years, body mass index was 25.0±3.9 kg/m2, median CD4 cell count was 634 cell/µL (interquartile range [IQR]=439-900), aspartate aminotransferase (AST) was 23 (IQR=19-30) IU/L, and alanine aminotransferase (ALT) was 24 (IQR=17-34) IU/L. At T1, both AST (median=-1, IQR=-5-2 IU/L, P=0.004) and ALT (median=-2, IQR=-7-3 IU/L, P=0.0004) showed a significant decrease. Among 28 patients with ALT >40 at baseline, reduction was significant both clinically (-17, IQR=-32--1) and statistically (P=0.0003). Total cholesterol levels (TC) increased (+13.4±3.8 mg/dL, P=0.0006), as well as HDL-cholesterol (HDL-C) (+3.8±1.2 mg/dL, P=0.02), LDL Cholesterol (LDL-C) (+7.6±3.4, P=0.03) and glucose (+4.0±1.8 mg/dL, P=0.02). D:A:D: and Framingham risk score did not change at 6 months after switch. CONCLUSION: A significant reduction of liver enzymes was observed after switching from TDF to TAF, especially in subjects with initial level of ALT >40 IU/L. Glucose, TC, HDL-C, and LDL-C increased, with no effect on cardiovascular risk scores.


Subject(s)
Alanine Transaminase/antagonists & inhibitors , Alanine/pharmacology , Aspartate Aminotransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Tenofovir/analogs & derivatives , Adult , Aged , Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , Female , Glucose/analysis , Humans , Lipids/analysis , Male , Middle Aged , Tenofovir/pharmacology
3.
Molecules ; 25(20)2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33092276

ABSTRACT

The insecticidal activity is the result of a series of complex interactions between toxic substances as ligands and insect's enzymes as targets. Actually, synthetic insecticides used in pest control programs are harmful to the environment and may affect non-target organisms; thus, the use of natural products as pest control agents can be very attractive. In the present work, the toxic effect of aniseed (Pimpinella anisum L.) essential oil (EO) and its nanoemulsion (NE) against the red flour beetle Tribolium castaneum, has been evaluated. To assess the EO mode of action, the impact of sub-lethal concentrations of aniseed EO and NE was evaluated on enzymatic and macromolecular parameters of the beetles, including aspartate aminotransferase (AST), alanine aminotransferase (ALT), total protein, total lipids and glucose. Finally, a molecular docking study was conducted to predict the mode of action of the major EO and NE components namely E-anethole, Limonene, alpha-himalachalene, trans-Verbenol and Linalool at binding site of the enzymes AST and ALT. Herein, the binding location of the main compounds in both proteins are discussed suggesting the possible interactions between the considered enzymes and ligands. The obtained results open new horizons to understand the evolution and response of insect-plant compounds interactions and their effect predicted at the molecular levels and side effects of both animal and human.


Subject(s)
Monoterpenes/chemistry , Oils, Volatile/chemistry , Pimpinella/chemistry , Tribolium/drug effects , Alanine Transaminase/antagonists & inhibitors , Alanine Transaminase/chemistry , Animals , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/chemistry , Emulsions/chemistry , Emulsions/pharmacology , Herb-Drug Interactions , Humans , Molecular Docking Simulation , Monoterpenes/pharmacology , Nanostructures/chemistry , Oils, Volatile/pharmacology
4.
J Enzyme Inhib Med Chem ; 35(1): 1137-1144, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32367744

ABSTRACT

We report here in silico repurposing studies on 52 new pyridazinone-based small-molecules through inverse virtual screening (iVS) methodologies. These analogues were originally designed as formyl peptide receptor (FPR) ligands. As it is sometimes the case in drug discovery programmes, subsequent biological screening demonstrated the inefficacy of the molecules in binding FPRs, failing in the identification of new hits. Through a focussed drug-repurposing approach we have defined a variety of potential targets that are suitable to interact with this library of pyridazinone-based analogues. A two-step approach has been conducted for computational analysis. Specifically, the molecules were initially processed through a pharmacophore-based screening. Secondly, the resulting features of binding were investigated by docking studies and following molecular dynamic simulations, in order to univocally confirm "pyridazinone-based ligand-target protein" interactions. Our findings propose aspartate aminotransferase as the most favourable repurposed target for this small-molecule series, worth of additional medicinal chemistry investigations in the field.


Subject(s)
Aspartate Aminotransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pyridazines/pharmacology , Small Molecule Libraries/pharmacology , Aspartate Aminotransferases/metabolism , Chemistry, Pharmaceutical , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Ligands , Models, Molecular , Molecular Structure , Pyridazines/chemistry , Small Molecule Libraries/chemistry
5.
Biochem Biophys Res Commun ; 524(4): 916-922, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32057367

ABSTRACT

Macrophage-mediated inflammation is a key pathophysiological component of cardiovascular diseases, but the underlying mechanisms by which the macrophage regulates inflammation have been unclear. In our study, we, for the first time, showed an endogenous sulfur dioxide (SO2) production in RAW267.4 macrophages by using HPLC and SO2-specific fluorescent probe assays. Moreover, the endogenous SO2 generating enzyme aspartate aminotransferase (AAT) was found to be expressed by the macrophages. Furthermore, we showed that AAT2 knockdown triggered spontaneous macrophage-mediated inflammation, as represented by the increased TNF-α and IL-6 levels and the enhanced macrophage chemotaxis; these effects could be reversed by the treatment with a SO2 donor. Mechanistically, AAT2 knockdown activated the NF-κB signaling pathway in macrophages, while SO2 successfully rescued NF-κB activation. In contrast, forced AAT2 expression reversed AngII-induced NF-κB activation and subsequent macrophage inflammation. Moreover, treatment with a SO2 donor also alleviated macrophage infiltration in AngII-treated mouse hearts. Collectively, our data suggest that macrophage-derived SO2 is an important regulator of macrophage activation and it acts as an endogenous "on-off switch" in the control of macrophage activation. This knowledge might enable a new therapeutic strategy for cardiovascular diseases.


Subject(s)
Aspartate Aminotransferases/genetics , Myocytes, Cardiac/drug effects , Myocytes, Smooth Muscle/drug effects , NF-kappa B/genetics , Sulfur Dioxide/pharmacology , Angiotensin II/pharmacology , Animals , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/immunology , Cell Line , Chemotaxis/drug effects , Gene Expression Regulation , Inflammation , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Myocytes, Cardiac/immunology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/immunology , NF-kappa B/immunology , RAW 264.7 Cells , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Sulfites/chemistry , Sulfites/pharmacology , Sulfur Dioxide/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
6.
Cell Commun Signal ; 17(1): 111, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31470862

ABSTRACT

BACKGROUND: Distinctive from their normal counterparts, cancer cells exhibit unique metabolic dependencies on glutamine to fuel anabolic processes. Specifically, pancreatic ductal adenocarcinoma (PDAC) cells rely on an unconventional metabolic pathway catalyzed by aspartate transaminase 1 (GOT1) to rewire glutamine metabolism and support nicotinamide adenine dinucleotide phosphate (NADPH) production. Thus, the important role of GOT1 in energy metabolism and Reactive Oxygen Species (ROS) balance demonstrates that targeting GOT1 may serve as an important therapeutic target in PDAC. METHODS: To assay the binding affinity between Aspulvinone O (AO) and GOT1 proteins, the virtual docking, microscale thermophoresis (MST), cellular thermal shift assay (CETSA) and drug affinity responsive target stability (DARTS) methods were employed. GOT1 was silenced in several PDAC cell lines. The level of OCR and ECR were assayed by seahorse. To evaluate the in vivo anti-tumor efficacy of AO, the xenograft model was built in CB17/scid mouse. RESULTS: Screening of an in-house natural compound library identified the AO as a novel inhibitor of GOT1 and repressed glutamine metabolism, which sensitizes PDAC cells to oxidative stress and suppresses cell proliferation. Virtual docking analysis suggested that AO could bind to the active site of GOT1 and form obvious hydrophobic interaction with Trp141 together with hydrogen bonds with Thr110 and Ser256. Further in vitro validation, including MST, CETSA and DARTS, further demonstrated the specific combining capacity of AO. We also show that the selective inhibition of GOT1 by AO significantly reduces proliferation of PDAC in vitro and in vivo. CONCLUSIONS: Taken together, our findings identify AO as a potent bioactive inhibitor of GOT1 and a novel anti-tumour agent for PDAC therapy.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Aspartate Aminotransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Furans/pharmacology , Glutamine/metabolism , Pancreatic Neoplasms/pathology , Antineoplastic Agents/metabolism , Aspartate Aminotransferases/chemistry , Aspartate Aminotransferases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/metabolism , Humans , Molecular Docking Simulation , Protein Conformation , Reactive Oxygen Species/metabolism
7.
Int J Biol Macromol ; 124: 788-795, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30502438

ABSTRACT

Mesona blumes polysaccharide (MBP), a primary active component extracted from Mesona blumes, has a number of bioactivities. Nevertheless, hepatoprotective activity of MBP has been rarely reported. The purpose of this study is to investigate hepatoprotective effects of MBP on acute liver injury in mice. Results indicated that the MBP could remarkably decrease the increased levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in the serum caused by tetrachloride (CCl4) treatment (P < 0.05). Medium and high dose of MBP treatment (200 mg/kg body weight, 300 mg/kg body weight) not only prominently enhanced the levels of antioxidant enzymes (superoxide dismutase, SOD) and non-enzyme antioxidants (glutathione, GSH) compared with CCl4-induced, but also dramatically decreased lipid peroxidation levels of liver tissues (P < 0.05). In addition, medium and high doses of MBP significantly enhanced the serum levels of IL-1ß and TNF-α (P < 0.05). This study showed that MBP had hepatoprotective activity against acute liver injury caused by CCl4.


Subject(s)
Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Gene Expression Regulation/drug effects , Lamiaceae/chemistry , Polysaccharides/pharmacology , Alanine Transaminase/antagonists & inhibitors , Alanine Transaminase/blood , Alanine Transaminase/genetics , Animals , Antioxidants/chemistry , Antioxidants/isolation & purification , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/genetics , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Glutathione/agonists , Glutathione/blood , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/blood , Interleukin-1beta/genetics , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Oxidative Stress/drug effects , Plant Extracts/chemistry , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Superoxide Dismutase/blood , Superoxide Dismutase/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
8.
PLoS One ; 13(12): e0208025, 2018.
Article in English | MEDLINE | ID: mdl-30532126

ABSTRACT

An emerging method to help elucidate the mode of action of experimental drugs is to use untargeted metabolomics of cell-systems. The interpretations of such screens are however complex and more examples with inhibitors of known targets are needed. Here two T-cell lines were treated with an inhibitor of aspartate aminotransferase and analyzed with untargeted GC-MS. The interpretation of the data was enhanced by the use of two different cell-lines and supports aspartate aminotransferase as a target. In addition, the data suggest an unexpected off-target effect on glutamate decarboxylase. The results exemplify the potency of metabolomics to provide insight into both mode of action and off-target effects of drug candidates.


Subject(s)
Aspartate Aminotransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hydrazines/pharmacology , Metabolome/drug effects , Succinates/pharmacology , T-Lymphocytes/drug effects , Aspartate Aminotransferases/metabolism , Drug Evaluation, Preclinical/methods , Gas Chromatography-Mass Spectrometry , Glutamate Decarboxylase/metabolism , Humans , Jurkat Cells , Metabolomics , T-Lymphocytes/metabolism
9.
Bioorg Med Chem Lett ; 28(16): 2675-2678, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29731362

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy that is extremely refractory to the therapeutic approaches that have been evaluated to date. Recently, it has been demonstrated that PDAC tumors are dependent upon a metabolic pathway involving aspartate aminotransferase 1, also known as glutamate-oxaloacetate transaminase 1 (GOT1), for the maintenance of redox homeostasis and sustained proliferation. As such, small molecule inhibitors targeting this metabolic pathway may provide a novel therapeutic approach for the treatment of this devastating disease. To this end, from a high throughput screen of ∼800,000 molecules, 4-(1H-indol-4-yl)-N-phenylpiperazine-1-carboxamide was identified as an inhibitor of GOT1. Mouse pharmacokinetic studies revealed that potency, rather than inherent metabolic instability, would limit immediate cell- and rodent xenograft-based experiments aimed at validating this potential cancer metabolism-related target. Medicinal chemistry-based optimization resulted in the identification of multiple derivatives with >10-fold improvements in potency, as well as the identification of a tryptamine-based series of GOT1 inhibitors.


Subject(s)
Aspartate Aminotransferases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Indoles/therapeutic use , Phenylurea Compounds/therapeutic use , Piperazines/therapeutic use , Transaminases/antagonists & inhibitors , Animals , Aspartate Aminotransferase, Cytoplasmic , Carcinoma, Pancreatic Ductal/drug therapy , Drug Discovery , Drug Stability , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Female , Indoles/chemical synthesis , Indoles/chemistry , Indoles/pharmacokinetics , Mice , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacokinetics , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacokinetics , Structure-Activity Relationship
10.
Pol J Vet Sci ; 21(1): 39-46, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29624020

ABSTRACT

This study examined the effect of chlorpyrifos and/or enrofloxacin on the activity of acetylcholinesterase (AChE) in the blood and brain, and the activity of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in serum. The experiment was conducted on Wistar strain rats. Chlorpyrifos was administered with a stomach tube at a dose of 0.04 LD50 for 28 days and enrofloxacin at a dose of 5 mg/kg bw for 5 consecutive days. The experiment found that enrofloxacin changed the activity of the enzymes under study only to a small extent. At the dose applied in the experiment, chlorpyrifos decreased the activity of AChE significantly, both in blood and in the brain, and increased the activity of ALT and AST in rat serum. The administration of chlorpyrifos in combination with enrofloxacin changed the activity of the enzymes under study only slightly. A weaker, but longer, inhibition of AChE activity in both blood and the brain was observed in this group compared to the animals exposed only to chlorpyrifos. However, although enrofloxacin, like chlorpyrifos, increases the activity of ALT and AST in serum, their combined administration did not increase the hepatotoxic effect.


Subject(s)
Acetylcholinesterase/metabolism , Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , Brain/enzymology , Chlorpyrifos/pharmacokinetics , Enrofloxacin/pharmacokinetics , Acetylcholinesterase/blood , Alanine Transaminase/antagonists & inhibitors , Alanine Transaminase/blood , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/blood , Chlorpyrifos/administration & dosage , Cholinesterase Inhibitors/administration & dosage , Cholinesterase Inhibitors/pharmacokinetics , Drug Interactions , Drug Therapy, Combination , Enrofloxacin/administration & dosage , Male , Random Allocation , Rats , Rats, Wistar
11.
Molecules ; 23(2)2018 Feb 12.
Article in English | MEDLINE | ID: mdl-29439513

ABSTRACT

Phospholipase A2s (PLA2) is a major component of snake venom with diverse pathologic toxicities and, therefore, a potential target for antivenom therapy. Varespladib was initially designed as an inhibitor of mammal PLA2s, and was recently repurposed to a broad-spectrum inhibitor of PLA2 in snake venom. To evaluate the protective abilities of varespladib to hemorrhage, myonecrosis, and systemic toxicities that are inflicted by different crude snake venoms, subcutaneous ecchymosis, muscle damage, and biochemical variation in serum enzymes derived from the envenomed mice were determined, respectively. Varespladib treatment showed a significant inhibitory effect to snake venom PLA2, which was estimated by IC50 in vitro and ED50 in vivo. In animal models, the severely hemorrhagic toxicity of D. acutus and A. halys venom was almost fully inhibited after administration of varespladib. Moreover, signs of edema in gastrocnemius muscle were remarkably attenuated by administration of varespladib, with a reduced loss of myonecrosis and desmin. Serum levels of creatine kinase, lactate dehydrogenase isoenzyme 1, aspartate transaminase, and alanine transaminase were down-regulated after treatment with varespladib, which indicated the protection to viscera injury. In conclusion, varespladib may be a potential first-line drug candidate in snakebite envenomation first aid or clinical therapy.


Subject(s)
Acetates/pharmacology , Antivenins/pharmacology , Crotalid Venoms/toxicity , Indoles/pharmacology , Phospholipase A2 Inhibitors/pharmacology , Phospholipases A2/metabolism , Snake Bites/drug therapy , Alanine Transaminase/antagonists & inhibitors , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/blood , Creatine Kinase/blood , Crotalinae/physiology , Ecchymosis/prevention & control , Edema/prevention & control , Female , Isoenzymes/antagonists & inhibitors , Isoenzymes/blood , Keto Acids , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/blood , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Snake Bites/metabolism , Snake Bites/physiopathology
12.
Cell Death Dis ; 9(2): 55, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29352139

ABSTRACT

Cellular senescence, which leads to a cell cycle arrest of damaged or dysfunctional cells, is an important mechanism to restrain the malignant progression of cancer cells. Because metabolic changes underlie many cell-fate decisions, it has been suggested that cell metabolism might play key roles in senescence pathways. Here, we show that mitochondrial glutamine metabolism regulates senescence in human pancreatic ductal adenocarcinoma (PDAC) cells. Glutamine deprivation or inhibition of mitochondrial aspartate transaminase (GOT2) results in a profound induction of senescence and a suppression of PDAC growth. Glutamine carbon flow through GOT2 is required to create NADPH and to maintain the cellular redox state. We found that elevated reactive oxygen species levels by GOT2 knockdown lead to the cyclin-dependent kinase inhibitor p27-mediated senescence. Importantly, PDAC cells exhibit distinct dependence on this pathway, whereas knockdown of GOT2 did not induce senescence in non-transformed cells. The essentiality of GOT2 in senescence regulation of PDAC, which is dispensable in their normal counterparts, may have profound implications for the development of strategies to treat these refractory cancers.


Subject(s)
Aspartate Aminotransferases/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Glutamine/metabolism , Mitochondria/metabolism , Pancreatic Neoplasms/metabolism , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/deficiency , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Cellular Senescence/physiology , Glutamine/deficiency , HEK293 Cells , Humans , Pancreatic Neoplasms/pathology
13.
Neuroreport ; 29(2): 99-105, 2018 Jan 17.
Article in English | MEDLINE | ID: mdl-29215467

ABSTRACT

Both mitochondrial aspartate aminotransferase (mAST) and cytosolic aspartate aminotransferase (cAST) are important components in the malate-aspartate shuttle - one of the two types of NADH shuttles in cells. A major goal of our current study was to determine specifically the roles of cAST in maintaining the [Ca]i, mitochondrial membrane potential and the survival of BV2 microglia by applying molecular approach to modulate the cAST levels. Our study found that decreased cAST by cAST siRNA can lead to significant increases in the [Ca]i, mitochondrial depolarization and apoptosis of BV2 microglia. The cAST siRNA-induced mitochondrial depolarization can be significantly attenuated by an inhibitor of calpain. We further found that the cAST siRNA-induced apoptosis can be prevented by the calpain inhibitor. Collectively, our study suggests that decreased cAST induces calpain activation by increasing the [Ca]i of BV2 microglia, resulting in mitochondrial depolarization and cell death. Moreover, our data suggest that decreased cAST may produce these pathological effects by malate-aspartate shuttle-independent pathways.


Subject(s)
Aspartate Aminotransferases/metabolism , Calcium/metabolism , Cell Survival/physiology , Cytosol/enzymology , Membrane Potential, Mitochondrial/physiology , Microglia/enzymology , Animals , Apoptosis/physiology , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/genetics , Calpain/antagonists & inhibitors , Calpain/metabolism , Cations, Divalent/metabolism , Cells, Cultured , Homeostasis/physiology , Mice , Mitochondria/metabolism , RNA, Small Interfering
14.
Biomed Pharmacother ; 97: 19-25, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29080454

ABSTRACT

Most traditional Chinese medicine prescription dosages are imprecise. This study analyzes the toxicities and adverse effects of a combination the active ingredients of licorice and Kushen medicine: oxymatrine (OMT) and diammonium glycyrrhizinate (DG). The median lethal dose (LD50) and mortality were analyzed in single-dose OMT (or DG) intraperitoneally injected mice with or without combination DG (or OMT). Body weight changes as well as levels of serum sodium and potassium, alanine transaminase (ALT), aspartate transaminase (AST), creatinine, and urea were measured in mice treated with a daily dose of OMT and/or DG for 14days. This study showed that the LD50 of OMT for males and females were 347.44 and 429.15mg/kg, respectively. The LD50 of DG were 525.10 and 997.26mg/kg for males and females, respectively. DG significantly decreased the mice LD50-induced mortality of the OMT, however OMT did not succeed in reducing the LD50-induced mortality rate of DG. The combination of OMT and DG obviously attenuated the changes of the body weight, serum sodium, and potassium induced by DG or OMT alone. These results suggested that toxicity and adverse effects of the OMT was significantly attenuated by DG. The OMT neutralized the adverse effects of the DG, but not the toxicity.


Subject(s)
Alkaloids/administration & dosage , Alkaloids/toxicity , Anti-Inflammatory Agents/administration & dosage , Glycyrrhizic Acid/administration & dosage , Quinolizines/administration & dosage , Quinolizines/toxicity , Alanine Transaminase/antagonists & inhibitors , Alanine Transaminase/blood , Alkaloids/antagonists & inhibitors , Animals , Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/toxicity , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/blood , Body Weight/drug effects , Body Weight/physiology , Female , Male , Mice , Mice, Inbred ICR , Mortality/trends , Quinolizines/antagonists & inhibitors , Random Allocation
15.
Biochemistry ; 56(37): 4951-4961, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28816437

ABSTRACT

Potent mechanism-based inactivators can be rationally designed against pyridoxal 5'-phosphate (PLP)-dependent drug targets, such as ornithine aminotransferase (OAT) or γ-aminobutyric acid aminotransferase (GABA-AT). An important challenge, however, is the lack of selectivity toward other PLP-dependent, off-target enzymes, because of similarities in mechanisms of all PLP-dependent aminotransferase reactions. On the basis of complex crystal structures, we investigate the inactivation mechanism of OAT, a hepatocellular carcinoma target, by (1R,3S,4S)-3-amino-4-fluorocyclopentane-1-carboxylic acid (FCP), a known inactivator of GABA-AT. A crystal structure of OAT and FCP showed the formation of a ternary adduct. This adduct can be rationalized as occurring via an enamine mechanism of inactivation, similar to that reported for GABA-AT. However, the crystal structure of an off-target, PLP-dependent enzyme, aspartate aminotransferase (Asp-AT), in complex with FCP, along with the results of attempted inhibition assays, suggests that FCP is not an inactivator of Asp-AT, but rather an alternate substrate. Turnover of FCP by Asp-AT is also supported by high-resolution mass spectrometry. Amid existing difficulties in achieving selectivity of inactivation among a large number of PLP-dependent enzymes, the obtained results provide evidence that a desirable selectivity could be achieved, taking advantage of subtle structural and mechanistic differences between a drug-target enzyme and an off-target enzyme, despite their largely similar substrate binding sites and catalytic mechanisms.


Subject(s)
4-Aminobutyrate Transaminase/antagonists & inhibitors , Aspartate Aminotransferases/antagonists & inhibitors , Cycloleucine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Models, Molecular , Ornithine-Oxo-Acid Transaminase/antagonists & inhibitors , Pyridoxal Phosphate/metabolism , 4-Aminobutyrate Transaminase/chemistry , 4-Aminobutyrate Transaminase/metabolism , Aspartate Aminotransferases/chemistry , Aspartate Aminotransferases/genetics , Aspartate Aminotransferases/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cycloleucine/chemistry , Cycloleucine/metabolism , Cycloleucine/pharmacology , Databases, Chemical , Databases, Protein , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Ligands , Molecular Conformation , Ornithine-Oxo-Acid Transaminase/chemistry , Ornithine-Oxo-Acid Transaminase/genetics , Ornithine-Oxo-Acid Transaminase/metabolism , Protein Conformation , Pyridoxal Phosphate/chemistry , Pyridoxamine/chemistry , Pyridoxamine/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structural Homology, Protein , Substrate Specificity
16.
Life Sci ; 178: 49-55, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28385612

ABSTRACT

AIMS: This study aimed to investigate the inhibitory effects of two natural flavonoids, hesperetin (HT) and hesperidin (HD), on two gluconeogenesis enzymes, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities and their possible mechanisms of action. MAIN METHODS: Rat liver incubated with different concentrations of HT and HD was used to measure enzyme activities spectrophotometrically, based on monitoring the oxidation of NADH to NAD+ at 340nm. Molecular docking simulation was also applied to reveal the molecular mechanism of the inhibition caused by HT and HD. KEY FINDINGS: Both flavonoids demonstrated inhibitory effects against the enzyme activities, with IC50 values of 153.9 and 68.88µM for HT-ALT and HD-ALT treatment respectively. Likewise, the IC50 values of 85.29µM for HT-AST and 110.3µM for HD-AST were obtained from spectrophotometric results. CONCLUSION: The docking simulation revealed that HT and HD block the enzyme entrance channel and prevent the substrates from accessing the enzyme active sites. Having prevented production of pyruvate, α-ketoglutarate, and the oxaloacetate, these two compounds inhibit hepatic gluconeogenesis and consequently, hinder the progression of diabetes. SIGNIFICANCE: This study suggests that HT and HD may be considered as leading compounds for designing safe and effective drugs in management of increased ALT and AST-related disorders specially diabetes.


Subject(s)
Alanine Transaminase/pharmacology , Aspartate Aminotransferases/antagonists & inhibitors , Hesperidin/pharmacology , Liver/drug effects , Animals , Diabetes Mellitus/drug therapy , Diabetes Mellitus/enzymology , Drug Design , Gluconeogenesis/drug effects , Hesperidin/administration & dosage , Inhibitory Concentration 50 , Liver/enzymology , Molecular Docking Simulation , Rats
17.
Molecules ; 22(1)2017 Jan 06.
Article in English | MEDLINE | ID: mdl-28067837

ABSTRACT

Previously, we reported the role of synergy between two flavonoids-namely, chrysin and kaempferol-in inhibiting the secretion of a few major proinflammatory mediators such as tumor necrosis factor-alpha (TNF-α), prostaglandin E2 (PGE2), and nitric oxide (NO) from lipopolysaccharide (LPS)-induced RAW 264.7 cells. The present study aims to evaluate the effects of this combination on a murine model of polymicrobial sepsis induced by cecal ligation and puncture (CLP). Severe sepsis was induced in male ICR mice (n = 7) via the CLP procedure. The effects of chrysin and kaempferol combination treatment on septic mice were investigated using a 7-day survival study. The levels of key proinflammatory mediators and markers-such as aspartate aminotransferase (AST), TNF-α, and NO-in the sera samples of the septic mice were determined via ELISA and fluorescence determination at different time point intervals post-CLP challenge. Liver tissue samples from septic mice were harvested to measure myeloperoxidase (MPO) levels using a spectrophotometer. Moreover, intraperitoneal fluid (IPF) bacterial clearance and total leukocyte count were also assessed to detect any antibacterial effects exerted by chrysin and kaempferol, individually and in combination. Kaempferol treatment improved the survival rate of CLP-challenged mice by up to 16%. During this treatment, kaempferol expressed antibacterial, antiapoptotic and antioxidant activities through the attenuation of bacterial forming units, AST and NO levels, and increased polymorphonuclear leukocyte (PMN) count in the IPF. On the other hand, the chrysin treatment significantly reduced serum TNF-α levels. However, it failed to significantly improve the survival rate of the CLP-challenged mice. Subsequently, the kaempferol/chrysin combination treatment significantly improved the overall 7-day survival rate by 2-fold-up to 29%. Kaempferol and chrysin revealed some synergistic effects by acting individually upon multiple pathophysiological factors involved during sepsis. Although the kaempferol/chrysin combination did not exhibit significant antibacterial effects, it did exhibit anti-inflammatory and antioxidant activities, which translate to significant improvement in the survival rate of septic animals. These findings suggest the potential application of this combination treatment as a beneficial adjuvant supplement strategy in sepsis control.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Flavonoids/pharmacology , Kaempferols/pharmacology , Sepsis/drug therapy , Animals , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/genetics , Biomarkers/blood , Colony Count, Microbial , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Gene Expression , Leukocyte Count , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred ICR , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/blood , Peroxidase/genetics , Peroxidase/metabolism , Sepsis/blood , Sepsis/genetics , Sepsis/pathology , Survival Analysis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
18.
Sci Rep ; 6: 27026, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27246393

ABSTRACT

The present study was designed to determine whether sulfur dioxide (SO2) could be endogenously produced in adipocyte and served as a novel adipocyte-derived inflammatory inhibitor. SO2 was detected in adipose tissue using high-performance liquid chromatography with fluorescence detection. SO2 synthase aspartate aminotransferase (AAT1 and AAT2) mRNA and protein expressions in adipose tissues were measured. For in vitro study, 3T3-L1 adipocytes were cultured, infected with adenovirus carrying AAT1 gene or lentivirus carrying shRNA to AAT1, and then treated with tumor necrosis factor-α (TNF-α). We found that endogenous SO2/AAT pathway existed in adipose tissues including perivascular, perirenal, epididymal, subcutaneous and brown adipose tissue. AAT1 overexpression significantly increased SO2 production and inhibited TNF-α-induced inflammatory factors, monocyte chemoattractant protein-1 (MCP-1) and interleukin-8 (IL-8) secretion from 3T3-L1 adipocytes. By contrast, AAT1 knockdown decreased SO2 production and exacerbated TNF-α-stimulated MCP-1 and IL-8 secretion. Mechanistically, AAT1 overexpression attenuated TNF-α-induced IκBα phosphorylation and degradation, and nuclear factor-κB (NF-κB) p65 phosphorylation, while AAT1 knockdown aggravated TNF-α-activated NF-κB pathway, which was blocked by SO2. NF-κB inhibitors, PDTC or Bay 11-7082, abolished excessive p65 phosphorylation and adipocyte inflammation induced by AAT1 knockdown. This is the first report to suggest that endogenous SO2 is a novel adipocyte-derived inflammatory inhibitor.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Anti-Inflammatory Agents/metabolism , Sulfur Dioxide/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Aspartate Aminotransferases/antagonists & inhibitors , Aspartate Aminotransferases/genetics , Aspartate Aminotransferases/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression Regulation , Interleukin-8/genetics , Interleukin-8/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , NF-KappaB Inhibitor alpha/genetics , NF-KappaB Inhibitor alpha/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Proline/analogs & derivatives , Proline/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Sulfones/pharmacology , Thiocarbamates/pharmacology , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
19.
Biochem Biophys Res Commun ; 473(4): 1295-1300, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27086848

ABSTRACT

Triple-negative breast cancer (TNBC) cell lines are identified to overexpress aspartate transaminase (GOT1), which can potentially control the intracellular levels of reactive oxygen species (ROS) through NADPH synthesis and enhances tumor growth. In this study, the impact of GOT1 on the efficacy of doxorubicin was investigated. Following doxorubicin administration, TNBC cells acquire metabolic alteration, causing increased glutamine flux for the synthesis of aspartate which can be converted into OAA by GOT1. Subsequently, this OAA is converted into malate and then pyruvate, maintaining the NADP(+)/NADPH ratio which neutralize doxorubicin-induced oxidative stress. Repression of GOT1 using the shRNAs for GOT1 resulted in doxorubicin-induced formation of ROS, thereby increasing doxorubicin sensitivity. The enhanced efficacy of doxorubicin by simultaneous repression of GOT1 was also indicated in an in vivo tumor model of TNBC. These results demonstrate that targeting GOT1 in TNBCs may provide a novel therapeutic approach for improving the efficacy of chemotherapy in patients with these refractory tumors.


Subject(s)
Aspartate Aminotransferases/antagonists & inhibitors , Doxorubicin/administration & dosage , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Treatment Outcome , Triple Negative Breast Neoplasms/enzymology
20.
Oncotarget ; 6(31): 31151-63, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26439804

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) cells utilize a novel non-canonical pathway of glutamine metabolism that is essential for tumor growth and redox balance. Inhibition of this metabolic pathway in PDAC can potentially synergize with therapies that increase intracellular reactive oxygen species (ROS) such as radiation. Here, we evaluated the dependence of pancreatic cancer stem cells (PCSCs) on this non-canonical glutamine metabolism pathway and researched whether inhibiting this pathway can enhance radiosensitivity of PCSCs. We showed that glutamine deprivation significantly inhibited self-renewal, decreased expression of stemness-related genes, increased intracellular ROS, and induced apoptosis in PCSCs. These effects were countered by oxaloacetate, but not α-ketoglutarate. Knockdown of glutamic-oxaloacetic transaminase dramatically impaired PCSCs properties, while glutamate dehydrogenase knockdown had a limited effect, suggesting a dependence of PCSCs on non-canonical glutamine metabolism. Additionally, glutamine deprivation significantly increased radiation-induced ROS and sensitized PCSCs to fractionated radiation. Moreover, transaminase inhibitors effectively enhanced ROS generation, promoted radiation sensitivity, and attenuated tumor growth in nude mice following radiation exposure. Our findings reveal that inhibiting the non-canonical pathway of glutamine metabolism enhances the PCSC radiosensitivity and may be an effective adjunct in cancer radiotherapy.


Subject(s)
Aspartate Aminotransferases/antagonists & inhibitors , Gamma Rays/adverse effects , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamine/metabolism , Neoplastic Stem Cells/radiation effects , Pancreatic Neoplasms/radiotherapy , Radiation Tolerance/genetics , Animals , Apoptosis , Aspartate Aminotransferases/genetics , Aspartate Aminotransferases/metabolism , Blotting, Western , Cell Proliferation , Flow Cytometry , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...