Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pharmacogenomics J ; 12(4): 328-41, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21383773

ABSTRACT

The overall neurobiological mechanisms by which lithium and valproate stabilize mood in bipolar disorder patients have yet to be fully defined. The therapeutic efficacy and dissimilar chemical structures of these medications suggest that they perturb both shared and disparate cellular processes. To investigate key pathways and functional clusters involved in the global action of lithium and valproate, we generated interaction networks formed by well-supported drug targets. Striking functional similarities emerged. Intersecting nodes in lithium and valproate networks highlighted a strong enrichment of apoptosis clusters and neurotrophin signaling. Other enriched pathways included MAPK, ErbB, insulin, VEGF, Wnt and long-term potentiation indicating a widespread effect of both drugs on diverse signaling systems. MAPK1/3 and AKT1/2 were the most preponderant nodes across pathways suggesting a central role in mediating pathway interactions. The convergence of biological responses unveils a functional signature for lithium and valproate that could be key modulators of their therapeutic efficacy.


Subject(s)
Apoptosis/drug effects , Bipolar Disorder/drug therapy , Lithium Compounds/therapeutic use , Nerve Growth Factors/drug effects , Valproic Acid/therapeutic use , Animals , Humans , Intercellular Signaling Peptides and Proteins/therapeutic use , Mice , Phosphoric Monoester Hydrolases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Rats , Signal Transduction/drug effects , Transcriptome/drug effects
2.
Ecotoxicology ; 21(1): 56-65, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21842397

ABSTRACT

Tetrahydrofuran (THF) is a toxic and carcinogenic compound that is commonly released from pharmaceutical, chemical and related industry wastewater. Currently, the effects of THF contamination on wastewater are unknown and a better understanding of THF toxicity toward biological processes in wastewater treatment is critical. In this study, we firstly investigated the toxic effects of THF on enzymatic activity and the microbial diversity in activated sludge from a sequencing batch reactor during long-term exposure to 10 mM THF. The activity of five enzymes (catalase, dehydrogenase, urease, phosphatase and protease) was remarkably decreased in the presence of 10 mM THF during a period of 85 days. Of these five affected enzymes, dehydrogenase activity was close to detection level limits and was nearly completely inhibited. Analysis of the microbial community demonstrated that THF, at a concentration of 10 mM, altered the distribution of microbes within the community and significantly decreased microbial diversity during long-term contamination, according to denaturing gradient gel electrophoresis (DGGE) analysis. The fraction of Actinobacteria increased in the community, while the fraction of Proteobacteria significantly decreased after THF exposure.


Subject(s)
Actinobacteria/drug effects , Bioreactors , Furans/toxicity , Proteobacteria/drug effects , Sewage/microbiology , Actinobacteria/growth & development , Catalase/antagonists & inhibitors , Catalase/drug effects , Catalase/metabolism , Cloning, Molecular , DNA, Bacterial/genetics , Environmental Exposure , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/drug effects , Oxidoreductases/metabolism , Peptide Hydrolases/drug effects , Peptide Hydrolases/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/metabolism , Proteobacteria/growth & development , Sequence Analysis, DNA , Urease/antagonists & inhibitors , Urease/drug effects , Urease/metabolism
3.
Hum Exp Toxicol ; 41: 9603271211065972, 2022.
Article in English | MEDLINE | ID: mdl-35238236

ABSTRACT

Propofol is a commonly used drug for sedation and general anesthesia during cancer surgery. Previous studies indicate that propofol exerts anti-tumor effect in various cancers. The aim of this study was to investigate the underlying molecular mechanism of propofol in liver cancer. The effects of propofol on liver cancer cells were evaluated by cell viability assay, colony formation assay, and tumor xenograft model. Dysregulated lncRNAs of propofol-treated liver cancer cells were evaluated by transcriptome RNA sequencing. The underlying molecular mechanisms of lncRNA cancer susceptibility candidate 9 (CASC9) in propofol-induced anti-tumor effects were evaluated by western blot, quantitative real-time polymerase chain reaction (qRT-PCR), wound scratch healing assay, transwell cell migration and invasion assay, TUNEL staining, fluorescence in situ hybridization, RNA immunoprecipitation (RIP), and chromatin immunoprecipitation (ChIP). We found that propofol suppressed proliferation, migration, invasion, and tumor xenograft growth of liver cancer cells in a dose-dependent manner. Exosomes transfer from propofol-treated cells inhibited proliferation, migration, and invasion and promoted apoptosis of liver cancer cells. Transcriptional profiling of propofol-treated liver cancer cells identified CASC9 as significantly downregulated lncRNA in cells and exosomes. Enforced CASC9 expression partially rescued the inhibitory effects of propofol on liver cancer cells. Furthermore, CASC9 was found to interact directly with EZH2 and epigenetically regulated PTEN expression. Restoration of CASC9 partially abrogated the inhibition of propofol on Akt/mTOR signaling. Our results indicated that propofol exerted anti-tumor effects by downregulating CASC9, and subsequently suppressed Akt/mTOR signaling. Our findings provided a novel insight into propofol-induced anti-tumor effects in liver cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Liver Neoplasms/drug therapy , Phosphoric Monoester Hydrolases/metabolism , Propofol/therapeutic use , Tacrolimus Binding Proteins/metabolism , Antineoplastic Agents/pharmacology , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/physiopathology , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/genetics , Propofol/metabolism , Propofol/pharmacology , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tacrolimus Binding Proteins/drug effects , Tacrolimus Binding Proteins/genetics
4.
J Nat Prod ; 74(5): 1093-9, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21539394

ABSTRACT

Two new cyclic depsipeptides, turnagainolides A (1) and B (2), have been isolated from laboratory cultures of a marine isolate of Bacillus sp. The structures of 1 and 2, which are simply epimers at the site of macrolactonization, were elucidated by analysis of NMR data and chemical degradation. A total synthesis of the turnagainolides confirmed their structures. Turnagainolide B (2) showed activity in a SHIP1 activation assay.


Subject(s)
Bacillus/chemistry , Depsipeptides/chemistry , Depsipeptides/isolation & purification , Phosphoric Monoester Hydrolases/metabolism , British Columbia , Depsipeptides/chemical synthesis , Depsipeptides/pharmacology , Inositol Polyphosphate 5-Phosphatases , Marine Biology , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Phosphoric Monoester Hydrolases/drug effects
5.
J Appl Toxicol ; 31(8): 752-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21404308

ABSTRACT

Among the numerous chemicals discharged into aquatic ecosystems, nonylphenol (NP) and octylphenol (OP) have been shown to have a potent effect on the endocrine system of fish; this issue has been clearly dealt with in several studies. The objective of this study was to assess and compare the general toxicity of these estrogenic chemicals individually on Clarias gariepinus. Fish were exposed to different concentrations of both NP and OP (250, 500, 750 and 1000 µg l(-1) ) under semi-static conditions for a period of 7 days. The adverse effect was evaluated with use of blood cell counting, hemoglobin (Hb), hematocrit (HCT), hematimetric indices, bilirubin, protein, glucose, serum transaminases, serum phosphatases, lactate dehydrogenase and cortisol. The results showed a clear indication of anemia, increases in leukocyte count and bilirubin content and a reduction in plasma protein levels with higher concentrations of both the toxicants compared with controls. Furthermore, with all the concentrations the inevitable increase in serum cortisol and plasma glucose showed primary and secondary stress responses. Moreover, probable tissue damage gave rise to a series of fluctuations of enzyme levels at lower concentrations, but a decrease with higher concentrations showed the severity of the effect. Depending on the parameters examined, OP had a relatively greater effect than NP. Overall, these two chemicals seemingly affected hematology and the activity of some enzymes, leading to serious impairment of the metabolism and physiology of C. gariepinus.


Subject(s)
Catfishes/metabolism , Endocrine Disruptors/toxicity , Phenols/toxicity , Animals , Bilirubin/metabolism , Blood Cell Count/methods , Dose-Response Relationship, Drug , Hematocrit , Hemoglobins/drug effects , Hydrocortisone/blood , L-Lactate Dehydrogenase/blood , L-Lactate Dehydrogenase/drug effects , Phosphoric Monoester Hydrolases/blood , Phosphoric Monoester Hydrolases/drug effects , Transaminases/blood , Transaminases/drug effects
6.
Neurochem Int ; 148: 105081, 2021 09.
Article in English | MEDLINE | ID: mdl-34082063

ABSTRACT

Cerebral ischemia is a disease of ischemic necrosis of brain tissue caused by intracranial artery stenosis or occlusion and cerebral artery embolization. Neuroinflammation plays an important role in the pathophysiology of cerebral ischemia. Microglia, astrocytes, leukocytes and other cells that release a variety of inflammatory factors involved in neuroinflammation may play a damaging or protective role during the process of cerebral ischemia. TP53-induced glycolysis and apoptotic regulators (TIGAR) may facilitate the production of nicotinamide adenine dinucleotide phosphoric acid (NADPH) via the pentose phosphate pathway (PPP) to inhibit oxidative stress and neuroinflammation. TIGAR can also directly inhibit NF-κB to inhibit neuroinflammation. TIGAR thus protect against cerebral ischemic injury. Exogenous NADPH can inhibit neuroinflammation by inhibiting oxidative stress and regulating a variety of signals. However, since NADPH oxidase (NOX) may use NADPH as a substrate to generate reactive oxygen species (ROS) to mediate neuroinflammation, the combination of NADPH and NOX inhibitors may produce more powerful anti-neuroinflammatory effects. Here, we review the cells and regulatory signals involved in neuroinflammation during cerebral ischemia, and discuss the possible mechanisms of targeting neuroinflammation in the treatment of cerebral ischemia with TIGAR/NADPH axis, so as to provide new ideas for the prevention and treatment of cerebral ischemia.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Brain Ischemia/drug therapy , NADP/genetics , Neuroinflammatory Diseases/drug therapy , Phosphoric Monoester Hydrolases/genetics , Animals , Apoptosis Regulatory Proteins/drug effects , Brain Ischemia/pathology , Humans , NADP/drug effects , Phosphoric Monoester Hydrolases/drug effects , Signal Transduction/drug effects
7.
Clin Transl Med ; 11(11): e592, 2021 11.
Article in English | MEDLINE | ID: mdl-34841698

ABSTRACT

BACKGROUND: The key role of hypoxia-inducible factor 2alpha (HIF2α) in the process of renal cancer has been confirmed. In the field of tumour research, oxidative stress is also considered to be an important influencing factor. However, the relationship and biological benefits of oxidative stress and HIF2α in ccRCC remain unclear. This research attempts to explore the effect of oxidative stress on the cancer-promoting effect of HIF2α in ccRCC and reveal its mechanism of action. METHODS: The bioinformatics analysis for ccRCC is based on whole transcriptome sequencing and TCGA database. The detection of the expression level of related molecules is realised by western blot and PCR. The expression of Nucleoside diphosphate-linked moiety X-type motif 1 (NUDT1) was knocked down by lentiviral infection technology. The functional role of NUDT1 were further investigated by CCK8 assays, transwell assays and cell oxidative stress indicator detection. The exploration of related molecular mechanisms is realised by Luciferase assays and Chromatin immunoprecipitation (ChIP) assays. RESULTS: Molecular screening based on knockdown HIF2α sequencing data and oxidative stress related data sets showed that NUDT1 is considered to be an important molecule for the interaction of HIF2α with oxidative stress. Subsequent experimental results showed that NUDT1 can cooperate with HIF2α to promote the progression of ccRCC. And this biological effect was found to be caused by the oxidative stress regulated by NUDT1. Mechanistically, HIF2α transcription activates the expression of NUDT1, thereby inhibiting oxidative stress and promoting the progression of ccRCC. CONCLUSIONS: This research clarified a novel mechanism by which HIF2α stabilises sirtuin 3 (SIRT3) through direct transcriptional activation of NUDT1, thereby inhibiting oxidative stress to promote the development of ccRCC. It provided the possibility for the selection of new therapeutic targets for ccRCC and the study of combination medication regimens.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/adverse effects , Carcinoma, Renal Cell/genetics , DNA Repair Enzymes/drug effects , Oxidative Stress/genetics , Phosphoric Monoester Hydrolases/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/physiopathology , Cell Line, Tumor/metabolism , DNA Repair Enzymes/genetics , Humans , Neoplasms , Oxidative Stress/drug effects , Phosphoric Monoester Hydrolases/genetics
8.
J Clin Endocrinol Metab ; 106(2): e875-e890, 2021 01 23.
Article in English | MEDLINE | ID: mdl-33165596

ABSTRACT

CONTEXT: Perturbed inositol physiology in insulin-resistant conditions has led to proposals of inositol supplementation for gestational diabetes (GDM) prevention, but placental inositol biology is poorly understood. OBJECTIVE: Investigate associations of maternal glycemia with placental inositol content, determine glucose effects on placental expression of inositol enzymes and transporters, and examine relations with birthweight. DESIGN AND PARTICIPANTS: Case-control study of placentae from term singleton pregnancies (GDM n = 24, non-GDM n = 26), and culture of another 9 placentae in different concentrations of glucose and myo-inositol for 48 hours. MAIN OUTCOME MEASURES: Placental inositol was quantified by the Megazyme assay. Relative expression of enzymes involved in myo-inositol metabolism and plasma membrane inositol transport was determined by quantitative RT-PCR and immunoblotting. Linear regression analyses were adjusted for maternal age, body mass index, ethnicity, gestational age, and sex. RESULTS: Placental inositol content was 17% lower in GDM compared with non-GDM. Higher maternal mid-gestation glycemia were associated with lower placental inositol. Increasing fasting glycemia was associated with lower protein levels of the myo-inositol synthesis enzyme, IMPA1, and the inositol transporters, SLC5A11 and SLC2A13, the expression of which also correlated with placental inositol content. In vitro, higher glucose concentrations reduced IMPA1 and SLC5A11 mRNA expression. Increasing fasting glycemia positively associated with customized birthweight percentile as expected in cases with low placental inositol, but this association was attenuated with high placental inositol. CONCLUSION: Glycemia-induced dysregulation of placental inositol synthesis and transport may be implicated in reduced placental inositol content in GDM, and this may in turn be permissive to accelerated fetal growth.


Subject(s)
Diabetes, Gestational/metabolism , Glucose/pharmacology , Inositol/metabolism , Phosphoric Monoester Hydrolases/genetics , Placenta/metabolism , Adult , Blood Glucose/physiology , Case-Control Studies , Cells, Cultured , Diabetes, Gestational/blood , Diabetes, Gestational/genetics , Down-Regulation , Female , Gene Expression Regulation/drug effects , Glucose/metabolism , Glucose Transport Proteins, Facilitative/drug effects , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Humans , Infant, Newborn , Male , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/metabolism , Placenta/pathology , Pregnancy , Signal Transduction/drug effects , Signal Transduction/genetics , Sodium-Glucose Transport Proteins/drug effects , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism
9.
Behav Pharmacol ; 21(5-6): 473-92, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20700048

ABSTRACT

This is a short overview focusing on the biochemical interactions underlying the protective effects of lithium at the neuronal level. These include lithium modulation of autophagy, growth factors, excitotoxicity, and a variety of mechanisms underlying cell death, neurogenesis, and neuronal differentiation. All these effects represent the result of a multifaceted pharmacology, which is becoming more and more complex. Nonetheless, when trying to dissect the various mechanisms of action of lithium, two primary targets emerge: glycogen synthase kinase 3beta and phosphatidylinositol phosphatase. The numerous lithium effects on biochemical systems are placed downstream of these two main mechanisms. At several steps, these mechanisms interconnect to each other, thus making it difficult to keep distinct the biochemical cascades promoted by lithium. In this way, it is not surprising that, despite being described as different phenomena at the behavioral level, molecular mechanisms underlying the effects of lithium on mood, motor activity, and sensitization overlap with those responsible for neuroprotection and neurorestoration. It is likely that the ancestral role of this ion as a modulator of cell survival, cell growth, movement, and mood is the consequence of a few molecular mechanisms operating in different neuronal networks, where a variety of cascade events take place. This review is an attempt to elucidate the primary effects of lithium to interconnect the simpler targets to the most complex pharmacological effects.


Subject(s)
Antimanic Agents/pharmacology , Drug Delivery Systems , Lithium Compounds/pharmacology , Affect/drug effects , Animals , Cell Survival/drug effects , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/metabolism
10.
J Environ Sci Health B ; 45(3): 249-53, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20390958

ABSTRACT

Metolachlor [2-chloro-N-(2-ethyl-6-methylphenyl)-N-(2-methoxy-1-methylethyl) acetamide] is a chiral acetanilide herbicide. We investigated its enantioselective interactions, and that of its (S)-isomer, with Penicillium expansum alkaline lipase and phosphatase. UV differential spectroscopy and fluorescence spectrophotometry studies were conducted in phosphate buffered solution at pH 7. Chiral differences in the UV absorption and fluorescence spectra of lipase and phosphatase with metolachlor and its (S)-isomer were detected. The results showed that the interactions of metolachlor and its (S)-isomer with lipase and phosphatase occur statically through complex formation, and enantioselectivity was clearly observed. In addition, both UV absorption and fluorescence spectrophotometry showed that the (S)-isomer interacted more strongly with lipase and phosphatase than metolachlor.


Subject(s)
Acetamides/chemistry , Herbicides/chemistry , Lipase/drug effects , Penicillium/enzymology , Phosphoric Monoester Hydrolases/drug effects , Circular Dichroism , Hydrogen-Ion Concentration , Isomerism , Lipase/chemistry , Lipase/metabolism , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet
11.
J Ment Health ; 19(2): 142-56, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20433322

ABSTRACT

BACKGROUND: It is difficult to reliably identify clinical predictors of lithium response. In recent years a number of pharmacogenetic studies have attempted to identify genetic variants which might be associated with response to lithium in bipolar patients. AIMS: To critically review pharmacogenetic studies of lithium response in bipolar disorder and suggest strategies for future work in this field. METHOD: Computerized searches of electronic databases for studies published between 1966 and October 2007. Studies were selected for detailed review according to the following criteria: (i) contained original data; (ii) included bipolar patients (including DSM-IV BP-I and BP-II); and (iii) lithium was the prophylactic agent under study. RESULTS: One hundred and fifty studies of interest were identified but only 15 relevant papers were selected for detailed review based on the selection criteria. Despite some interesting preliminary findings, there are substantial methodological limitations with almost all of the studies published to date. These issues are critically reviewed. CONCLUSIONS: The pharmacogenetics of lithium response in bipolar disorder remains a field in its infancy. There is a need for large-scale, prospective studies of biologically plausible candidate gene genes. The advent of genome-wide association studies holds particular promise for future studies.


Subject(s)
Antimanic Agents/pharmacology , Antimanic Agents/therapeutic use , Bipolar Disorder , Lithium Carbonate/pharmacology , Lithium Carbonate/therapeutic use , Pharmacogenetics , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/genetics , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/genetics , Bipolar Disorder/diagnosis , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Diagnostic and Statistical Manual of Mental Disorders , Humans , Predictive Value of Tests , Prospective Studies , Treatment Outcome
12.
Theranostics ; 10(8): 3488-3502, 2020.
Article in English | MEDLINE | ID: mdl-32206103

ABSTRACT

Rationale: Whole-genome sequencing has identified many amplified genes in esophageal squamous-cell carcinoma (ESCC). This study investigated the role and clinical relevance of these genes in ESCC. Methods: We collected ESCC and non-tumor esophageal tissues from 225 individuals who underwent surgery. Clinical data were collected and survival time was measured from the date of diagnosis to the date of last follow-up or death. Patient survival was compared with immunohistochemical staining score using Kaplan-Meier methods and hazard ratios were calculated by Cox models. Cells with gene overexpression and knockout were analyzed in proliferation, migration and invasion assays. Cells were also analyzed for levels of intracellular lactate, NADPH, ATP and mRNA and protein expression patterns. Protein levels in cell line and tissue samples were measured by immunoblotting or immunohistochemistry. ESCC cell were grown as xenograft tumors in nude mice. Primary ESCC in genetically engineered mice and patient-derived xenograft mouse models were established for test of therapeutic effects. Results: We show that TP53-induced glycolysis and apoptosis regulator (TIGAR) is a major player in ESCC progression and chemoresistance. TIGAR reprograms glucose metabolism from glycolysis to the glutamine pathway through AMP-activated kinase, and its overexpression is correlated with poor disease outcomes. Tigar knockout mice have reduced ESCC tumor burden and growth rates. Treatment of TIGAR-overexpressing ESCC cell xenografts and patient-derived tumor xenografts in mice with combination of glutaminase inhibitor and chemotherapeutic agents achieves significant more efficacy than chemotherapy alone. Conclusion: These findings shed light on an important role of TIGAR in ESCC and might provide evidence for targeted treatment of TIGAR-overexpressing ESCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/genetics , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Neoplasm Proteins/genetics , Phosphoric Monoester Hydrolases/genetics , Animals , Apoptosis Regulatory Proteins/drug effects , Cell Line, Tumor , Cell Proliferation , Disease Progression , Drug Delivery Systems , Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/mortality , Female , Glutaminase/antagonists & inhibitors , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/drug effects , Oncogenes , Phosphoric Monoester Hydrolases/drug effects , Survival Rate , Xenograft Model Antitumor Assays
13.
J Environ Sci Health B ; 44(5): 461-71, 2009 Jun.
Article in English | MEDLINE | ID: mdl-20183051

ABSTRACT

A pot trial was carried out to investigate the adverse effects of tetracycline (TC) on soil microbial communities, microbial activities, and the growth of ryegrass (Lolium perenne L). The results showed that the presence of TC significantly disturbed the structure of microbial communities and inhibited soil microbial activities in terms of urease, acid phosphatase and dehydrogenase (p < 0.05). Plant biomass was adversely influenced by TC, especially the roots with a reduction of 40% when compared with the control. Furthermore, TC decreased the assimilation of phosphorus by the plant although the concentration of phosphorus was increased by 20% due to decreased plant biomass. TC seemed to increase the concentration of dissolved organic carbon (by 20%) in soil. The findings implied that the agricultural use of animal manure or fishpond sediment containing considerable amounts of antibiotics may give rise to ecological risks.


Subject(s)
Drug Residues/toxicity , Enzyme Activation/drug effects , Lolium/growth & development , Soil Microbiology , Tetracycline/toxicity , Animals , Bacteria/drug effects , Bacteria/enzymology , Biomass , Carbon/metabolism , Drug Residues/metabolism , Lolium/drug effects , Manure , Oxidoreductases/drug effects , Oxidoreductases/metabolism , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/metabolism , Phosphorus/metabolism , Plant Roots/drug effects , Plant Roots/growth & development , Soil , Soil Pollutants/metabolism , Soil Pollutants/toxicity , Tetracycline/metabolism , Urease/drug effects , Urease/metabolism
14.
Neurobiol Learn Mem ; 90(3): 544-52, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18625329

ABSTRACT

In Lymnaea, aerial respiration can be operantly conditioned and depending on the training procedure employed two forms of memory can result: intermediate-term (ITM) and long-term memory (LTM). ITM, which persists for 3h, is dependent on de novo protein synthesis whilst LTM, which persists for at least 24 h, is dependent on both de novo protein synthesis and altered gene activity. A single 0.5 h training session (i.e. ITM-training) leaves behind a residual molecular memory trace, which a second bout of ITM-training can activate and boost it to a LTM. Here we extend this finding to show that either inhibiting protein phosphatase activity with okadaic acid (1 microM), or increasing protein kinase C (PKC) activity and therefore protein phosphorylation with bryostatin (0.25 ng/mL) treatment prior to ITM-training, results in a LTM. However, following right pedal dorsal 1 (RPeD1) soma ablation neither of these treatments are effective in producing LTM following ITM-training, indicating transcription is a necessity. These findings suggest that the balance between phosphorylation and dephosphorylation in neurons is a key factor for LTM formation.


Subject(s)
Conditioning, Operant/physiology , Ganglia, Invertebrate/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Retention, Psychology/physiology , Analysis of Variance , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Bryostatins/pharmacology , Conditioning, Operant/drug effects , Drug Administration Schedule , Enzyme Inhibitors/pharmacology , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/drug effects , Gene Expression Regulation/physiology , Lymnaea , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/drug effects , Neurons/drug effects , Okadaic Acid/pharmacology , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Retention, Psychology/drug effects
15.
Acta Trop ; 107(2): 153-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18599005

ABSTRACT

In this work, we characterized a Mg(2+)-dependent ecto-phosphatase activity present in live Trypanosoma rangeli epimastigotes. This enzyme showed capacity to hydrolyze the artificial substrate for phosphatases, p-nitrophenylphosphate (p-NPP). At saturating concentration of p-NPP, half-maximal p-NPP hydrolysis was obtained with 0.23mM Mg(2+). Ca(2+) had no effect on the basal phosphatase activity, could not substitute Mg(2+) as an activator and in contrast inhibited the p-NPP hydrolysis stimulated by Mg(2+). The dependence on p-NPP concentration showed a normal Michaelis-Menten kinetics for this phosphatase activity with values of V(max) of 8.94+/-0.36 nmol p-NP x h(-1) x 10(-7) cells and apparent K(m) of 1.04+/-0.16 mM p-NPP. Mg(2+)-dependent ecto-phosphatase activity was stimulated by the alkaline pH range. Experiments using inhibitors, such as, sodium fluoride, sodium orthovanadate and ammonium molybdate, inhibited the Mg(2+)-dependent ecto-phosphatase activity. Inorganic phosphate (Pi), a product of phosphatases, inhibited reversibly in 50% this activity. Okadaic acid and microcystin-LR, specific phosphoserine/threonine phosphatase inhibitors, inhibited significantly the Mg(2+)-dependent ecto-phosphatase activity. In addition, this phosphatase activity was able to recognize as substrates only o-phosphoserine and o-phosphothreonine, while o-phosphotyrosine was not a good substrate for this phosphatase. Epimastigote forms of T. rangeli exhibit a typical growth curve, achieving the stationary phase around fifth or sixth day and the Mg(2+)-dependent ecto-phosphatase activity decreased around 10-fold with the cell growth progression. Cells maintained at Pi-deprived medium (2 mM Pi) present Mg(2+)-dependent ecto-phosphatase activity approximately threefold higher than that maintained at Pi-supplemented medium (50 mM Pi).


Subject(s)
Gene Expression Regulation, Enzymologic , Inorganic Chemicals/pharmacology , Magnesium/metabolism , Phosphates/pharmacology , Phosphoric Monoester Hydrolases/drug effects , Trypanosoma/enzymology , Animals , Calcium/metabolism , Cell Membrane/enzymology , Culture Media , Phosphoric Monoester Hydrolases/metabolism , Substrate Specificity , Trypanosoma/drug effects , Trypanosoma/growth & development
16.
Circulation ; 114(11): 1159-68, 2006 Sep 12.
Article in English | MEDLINE | ID: mdl-16952979

ABSTRACT

BACKGROUND: Cellular hypertrophy requires coordinated regulation of progrowth and antigrowth mechanisms. In cultured neonatal cardiomyocytes, Foxo transcription factors trigger an atrophy-related gene program that counters hypertrophic growth. However, downstream molecular events are not yet well defined. METHODS AND RESULTS: Here, we report that expression of either Foxo1 or Foxo3 in cardiomyocytes attenuates calcineurin phosphatase activity and inhibits agonist-induced hypertrophic growth. Consistent with these results, Foxo proteins decrease calcineurin phosphatase activity and repress both basal and hypertrophic agonist-induced expression of MCIP1.4, a direct downstream target of the calcineurin/NFAT pathway. Furthermore, hearts from Foxo3-null mice exhibit increased MCIP1.4 abundance and a hypertrophic phenotype with normal systolic function at baseline. Together, these results suggest that Foxo proteins repress cardiac growth at least in part through inhibition of the calcineurin/NFAT pathway. Given that hypertrophic growth of the heart occurs in multiple contexts, our findings also suggest that certain hypertrophic signals are capable of overriding the antigrowth program induced by Foxo. Consistent with this, multiple hypertrophic agonists triggered inactivation of Foxo proteins in cardiomyocytes through a mechanism requiring the PI3K/Akt pathway. In addition, both Foxo1 and Foxo3 are phosphorylated and consequently inactivated in hearts undergoing hypertrophic growth induced by hemodynamic stress. CONCLUSIONS: This study suggests that inhibition of the calcineurin/NFAT signaling cascade by Foxo and release of this repressive action by the PI3K/Akt pathway are important mechanisms whereby Foxo factors govern cell growth in the heart.


Subject(s)
Cardiomegaly/physiopathology , Forkhead Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Angiotensin II/pharmacology , Animals , Calcineurin/physiology , Calcium-Binding Proteins , Cardiomegaly/genetics , Cardiomegaly/pathology , Cells, Cultured , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Pharmacol Ther ; 112(3): 799-809, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16842857

ABSTRACT

Phosphatidyl inositol 3-kinase (PI3-kinase) functions as a lipid kinase to produce PI(3,4,5)P(3) from PI(4,5)P(2) in vivo. PI(3,4,5)P(3) is crucial as a lipid second messenger in various metabolic effects of insulin. Lipid phosphatases, src homology 2 domain containing inositol 5'-phosphatase 2 (SHIP2) and skeletal muscle and kidney-enriched inositol phosphatase (SKIP) hydrolyze PI(3,4,5)P(3) to PI(3,4)P(2) and phosphatase and tensin homolog deleted on chromosome ten (PTEN) hydrolyzes PI(3,4,5)P(3) to PI(4,5)P(2). SHIP2 negatively regulates insulin signaling relatively specifically via its 5'-phosphatase activity. Targeted disruption of the SHIP2 gene in mice resulted in increased insulin sensitivity and conferred protection from obesity induced by a high-fat diet. Polymorphisms in the human SHIP2 gene are associated, at least in part, with the insulin resistance of type 2 diabetes. Importantly, inhibition of endogenous SHIP2 through the liver-specific expression of a dominant-negative SHIP2 improves glucose metabolism and insulin resistance in diabetic db/db mice. Overexpression of PTEN and SKIP also inhibited insulin-induced phosphorylation of Akt and the uptake of glucose in cultured cells. Although a homozygous disruption of the PTEN gene in mice results in embryonic lethality, either skeletal muscle or adipose tissue-specific disruption of PTEN ameliorated glucose metabolism without formation of tumors in animal models of diabetes. The role of SKIP in glucose metabolism remains to be further clarified in vivo. Taken together, inhibition of endogenous SHIP2 in the whole body appears to be effective at improving the insulin resistance associated with type 2 diabetes and/or obesity. Inhibition of PTEN in the tissues specifically targeted, including skeletal muscle and fat, may result in an amelioration of insulin resistance in type 2 diabetes, although caution against the formation of tumors is needed.


Subject(s)
Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/enzymology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Lipid Metabolism/physiology , Obesity/drug therapy , Obesity/enzymology , Phosphoric Monoester Hydrolases/physiology , Animals , Atherosclerosis/enzymology , Brain/enzymology , Brain/physiology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/physiology , Lipid Metabolism/drug effects , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/physiology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology
18.
Cell Signal ; 18(11): 2022-32, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16682172

ABSTRACT

Phosphatidylinositol(3,4,5)triphosphate (PtdIns(3,4,5)P(3)) plays important signaling roles in immune cells, particularly in the control of activating pathways and of survival. It is formed by a family of phosphatidylinositol 3'-kinases (PI3Ks) which phosphorylate PtdIns(4,5)P(2) in vivo. In human neutrophils, the levels of PtdIns(3,4,5)P(3) increase rapidly at the leading edge of locomoting cells and at the base of the phagocytic cup during FcgammaR-mediated particle ingestion. Even though these, and other, data indicate that PtdIns(3,4,5)P(3) is involved in the control of chemotaxis and phagocytosis in human neutrophils, the mechanisms that regulate its levels have yet to be fully elucidated in these cells. We evaluated the potential implication of SHIP1 and PTEN, two lipid phosphatases that utilize PtdIns(3,4,5)P(3) as substrate, in the signaling pathways called upon in response to CD32a cross-linking. We observed that the cross-linking of CD32a resulted in a transient accumulation of PtdIns(3,4,5)P(3). CD32a cross-linking also induced the tyrosine phosphorylation of SHIP1, its translocation to the plasma membrane and its co-immunoprecipitation with CD32a. CD32a cross-linking had no effect on the level of serine/threonine phosphorylation of PTEN and did not stimulate its translocation to the plasma membrane. PP2, a Src kinase inhibitor, inhibited the tyrosine phosphorylation of SHIP1 as well as its translocation to the plasma membrane. Wortmannin, a PI3K inhibitor, had no effect on either of these two indices of activation of SHIP1. Our results indicate that SHIP1 is involved, in a Src kinase-dependent manner, in the early signaling events observed upon the cross-linking of CD32a in human neutrophils.


Subject(s)
Antigens, CD/metabolism , Neutrophils/metabolism , Phosphoric Monoester Hydrolases/physiology , Receptors, IgG/metabolism , Signal Transduction , Androstadienes/pharmacology , Cell Membrane/metabolism , Enzyme Inhibitors/pharmacology , Humans , Inositol Polyphosphate 5-Phosphatases , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/drug effects , Phosphorylation/drug effects , Protein Transport/drug effects , Wortmannin , src Homology Domains , src-Family Kinases/metabolism
19.
Mol Cell Biol ; 23(17): 6027-36, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12917327

ABSTRACT

Lithium has been used as an effective mood-stabilizing drug for the treatment of manic episodes and depression for 50 years. More recently, lithium has been found to protect neurons from death induced by a wide array of neurotoxic insults. However, the molecular basis for the prophylactic effects of lithium have remained obscure. A target of lithium, glycogen synthase kinase 3 (GSK-3), is implicated in neuronal death after trophic deprivation. The mechanism whereby GSK-3 exerts its neurotoxic effects is also unknown. Here we show that lithium blocks the canonical c-Jun apoptotic pathway in cerebellar granule neurons deprived of trophic support. This effect is mimicked by the structurally independent inhibitors of GSK-3, FRAT1, and indirubin. Like lithium, these prevent the stress induced c-Jun protein increase and subsequent apoptosis. These events are downstream of c-Jun transactivation, since GSK-3 inhibitors block neuronal death induced by constitutively active c-Jun (Ser/Thr-->Asp) and FRAT1 expression inhibits AP1 reporter activity. Consistent with this, AP1-dependent expression of proapoptotic Bim requires GSK-3-like activity. These data suggest that a GSK-3-like kinase acts in tandem with c-Jun N-terminal kinase to coordinate the full execution of the c-Jun stress response and neuronal death in response to trophic deprivation.


Subject(s)
Glycogen Synthase Kinase 3/drug effects , Lithium/pharmacology , Membrane Proteins , Mitogen-Activated Protein Kinases/drug effects , Neoplasm Proteins , Neurons/drug effects , Neuroprotective Agents/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Cerebellum/cytology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Genes, Reporter , Glycogen Synthase Kinase 3/metabolism , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Oximes/pharmacology , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats , Stress, Physiological , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Up-Regulation
20.
Exp Hematol ; 34(11): 1532-41, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17046573

ABSTRACT

OBJECTIVE: Mast cells (MCs) play central roles for the onset and development of immediate-type and inflammatory allergic reactions. Since the inverse relationship between atopic disorders and diabetes mellitus has been observed in animals and humans, we investigated the effects of insulin (Ins) on MC signaling and biological function. METHODS: In bone marrow-derived MCs (BMMCs) from wild-type as well as SHIP-deficient mice Ins as well as insulin-like growth factor-1 (IGF-I)-triggered intracellular signaling events and MC effector functions were studied. RESULTS: We found that the addition of either Ins or IGF-1 to BMMCs triggers the phosphorylation of protein kinase B (PKB) and p38 kinase but not extracellular signal-regulated kinase (Erk). We also found that Ins/IGF-1 stimulates the tyrosine phosphorylation of SHIP1 and, in keeping with this, Ins/IGF-1-induced PKB phosphorylation is higher in SHIP1-/- BMMCs and is inhibited in SHIP+/+ as well as SHIP1-/- BMMCs with inhibitors of phosphatidylinositol-3-kinase (PI3K). Ins/IGF-1, like antigen (Ag), also stimulates the Rac-dependent activation of PAK as well as the production of hydrogen peroxide (H2O2). To elucidate the role of Ins and IGF-1 in MC biology, we studied their effects on Ag-mediated degranulation and MC survival. Although both only slightly enhanced Ag-mediated degranulation, they significantly promoted MC survival in the absence of IL-3 in a PI3K-dependent manner. CONCLUSION: The promotion of BMMC survival by induction of Ins/IGF-1 signaling may, in part, be responsible for the inverse correlation observed between atopic disorders and diabetes mellitus.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Mast Cells/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Degranulation/drug effects , Cell Survival/drug effects , Cells, Cultured , Hydrogen Peroxide/metabolism , Inositol Polyphosphate 5-Phosphatases , Male , Mast Cells/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/drug effects , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL