Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Chemosphere ; 177: 65-76, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28284117

ABSTRACT

Organochlorine pesticides (OCPs) in sediment were a potential damage for humans and ecosystems. The aim of this work was to determine the effectiveness of carbon materials remedy hexachlorocyclohexane (HCH) and dichlorodiphenyltrichloroethanes (DDTs) in sediment. Two different carbon materials including activated carbon (AC) and multi-walled carbon nanotubes (MWCNTs) were used in the present research. Sediment treated with 2 wt% AC and MWCNTs after 150 d contact showed 97%, and 75% reduction for HCH, and 93% and 59% decrease for DDTs in aqueous equilibrium concentration, respectively. Similarly, the reduction efficiencies of DDT and HCH uptake by semipermeable membrane devices (SPMDs) treated with AC (MWCNTs) were 97% (75%) and 92% (63%), respectively under the identical conditions. Furthermore, for 2 wt% AC (MWCNTs) system, a reduction of XAD beads uptake up to 87% (52%) and 73% (67%) was obtained in HCH and DDT flux to overlying water in quiescent system. Adding MWCNTs to contaminated sediment did not significantly decrease aqueous equilibrium concentration and DDTs and HCH availability in SPMDs compared to AC treatment. A series of results indicated that AC had significantly higher remediation efficiency towards HCH and DDTs in sediment than MWCNTs. Additionally, the removal efficiencies of two organic pollutants improved with increasing material doses and contact times. The greater effectiveness of AC was attributed to its greater specific surface area, which was favorable for binding contaminants. These results highlighted the potential for using AC as in-situ sorbent amendments for sediment remediation.


Subject(s)
Charcoal/chemistry , Environmental Restoration and Remediation/methods , Hydrocarbons, Chlorinated/analysis , Nanotubes, Carbon/chemistry , Water Pollutants, Chemical/analysis , Adsorption , China , DDT/analysis , Geography , Geologic Sediments/chemistry , Hexachlorocyclohexane/analysis , Lakes/chemistry , Pesticides/analysis , Solubility , Water/chemistry
2.
Alcohol Clin Exp Res ; 40(11): 2320-2328, 2016 11.
Article in English | MEDLINE | ID: mdl-27647657

ABSTRACT

BACKGROUND: Ethanol (EtOH) neurotoxicity can result in devastating effects on brain and behavior by disrupting homeostatic signaling cascades and inducing cell death. One such mechanism involves double-stranded RNA activated protein kinase (PKR), a primary regulator of protein translation and cell viability in the presence of a virus or other external stimuli. EtOH-mediated up-regulation of interferon-gamma (IFN-γ; the oxidative stress-inducible regulator of PKR), PKR, and its target, p53, are still being fully elucidated. METHODS: Using Western blot analysis, immunofluorescence, and linear regression analyses, changes in the IFN-γ-PKR-p53 pathway following chronic EtOH treatment in the frontal cortex of rodents were examined. The role of PKR on cell viability was also assessed in EtOH-treated cells using PKR overexpression vector and PKR inhibitor (PKRI). RESULTS: In rats chronically fed EtOH, PKR, phosphorylated PKR (p-PKR), IFN-γ, and p53 were significantly increased following chronic EtOH exposure. Linear regression revealed a significant correlation between IFN-γ and p-PKR protein levels, as well as p-PKR expression and age of EtOH exposure. Overexpression of PKR resulted in greater cell death, while use of PKRI enhanced cell viability in EtOH-treated cells. CONCLUSIONS: Chronic EtOH exposure activates the IFN-γ-PKR-p53 pathway in the frontal cortex of rodents. p-PKR expression is greater in brains of rodents exposed to EtOH at earlier ages compared to later life, suggesting a mechanism by which young brains could be more susceptible to EtOH-related brain injury. PKR and p-PKR were also colocalized in neurons and astrocytes of rats. This study provides additional insight into biochemical mechanisms underlying alcohol use disorder related neuropathology and warrants further investigation of PKR as a potential pharmacotherapeutic target to combat EtOH-related neurotoxicity, loss of protein translation and brain injury.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Interferon-gamma/metabolism , Prefrontal Cortex/drug effects , Tumor Suppressor Protein p53/metabolism , eIF-2 Kinase/metabolism , Age of Onset , Animals , Cell Death/drug effects , Cell Line, Tumor , Humans , Male , Prefrontal Cortex/metabolism , Random Allocation , Rats, Wistar , Signal Transduction/drug effects , Up-Regulation/drug effects
3.
J Colloid Interface Sci ; 471: 94-102, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26994349

ABSTRACT

Silver nanoparticle-decorated magnetic graphene oxide (MGO-Ag) was synthesized by doping silver and Fe3O4 nanoparticles on the surface of GO, which was used as an antibacterial agent. MGO-Ag was characterized by scanning electron microscopy (SEM), transmission electron microscopy (TEM), Energy dispersive X-ray (EDS), X-ray diffraction (XRD), Raman spectroscopy and magnetic property tests. It can be found that magnetic iron oxide nanoparticles and nano-Ag was well dispersed on graphene oxide; and MGO-Ag exhibited excellent antibacterial activity against Escherichia coli and Staphylococcus aureus. Several factors were investigated to study the antibacterial effect of MGO-Ag, such as temperature, time, pH and bacterial concentration. We also found that MGO-Ag maintained high inactivation rates after use six times and can be separated easily after antibacterial process. Moreover, the antibacterial mechanism is discussed and the synergistic effect of GO, Fe3O4 nanoparticles and nano-Ag accounted for high inactivation of MGO-Ag.


Subject(s)
Anti-Bacterial Agents , Escherichia coli/growth & development , Graphite/chemistry , Metal Nanoparticles/chemistry , Silver/chemistry , Staphylococcus aureus/growth & development , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Metal Nanoparticles/ultrastructure
4.
Neuropharmacology ; 105: 329-340, 2016 06.
Article in English | MEDLINE | ID: mdl-26805422

ABSTRACT

Binge drinking induces several neurotoxic consequences including oxidative stress and neurodegeneration. Because of these effects, drugs which prevent ethanol-induced damage to the brain may be clinically beneficial. In this study, we investigated the ethanol-mediated KLF11-MAO cell death cascade in the frontal cortex of Sprague-Dawley rats exposed to a modified Majchowicz 4-day binge ethanol model and control rats. Moreover, MAO inhibitors (MAOIs) were investigated for neuroprotective activity against binge ethanol. Binge ethanol-treated rats demonstrated a significant increase in KLF11, both MAO isoforms, protein oxidation and caspase-3, as well as a reduction in BDNF expression in the frontal cortex compared to control rats. MAOIs prevented these binge ethanol-induced changes, suggesting a neuroprotective benefit. Neither binge ethanol nor MAOI treatment significantly affected protein expression levels of the oxidative stress enzymes, SOD2 or catalase. Furthermore, ethanol-induced antinociception was enhanced following exposure to the 4-day ethanol binge. These results demonstrate that the KLF11-MAO pathway is activated by binge ethanol exposure and MAOIs are neuroprotective by preventing the binge ethanol-induced changes associated with this cell death cascade. This study supports KLF11-MAO as a mechanism of ethanol-induced neurotoxicity and cell death that could be targeted with MAOI drug therapy to alleviate alcohol-related brain injury. Further examination of MAOIs to reduce alcohol use disorder-related brain injury could provide pivotal insight to future pharmacotherapeutic opportunities.


Subject(s)
Binge Drinking/enzymology , Brain Diseases/prevention & control , Central Nervous System Depressants/toxicity , Ethanol/toxicity , Monoamine Oxidase Inhibitors/therapeutic use , Monoamine Oxidase/genetics , Signal Transduction/drug effects , Trans-Activators/drug effects , Trans-Activators/genetics , Animals , Brain Diseases/chemically induced , Brain-Derived Neurotrophic Factor/metabolism , Caspase 3/metabolism , Cell Death , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/antagonists & inhibitors , Ethanol/administration & dosage , Ethanol/antagonists & inhibitors , Male , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Oxidative Stress/drug effects , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley
5.
Int J Biol Macromol ; 82: 702-10, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26582339

ABSTRACT

Magnetic chitosan-graphene oxide (MCGO) nanocomposite was prepared as a multi-functional nanomaterial for the applications of antibacterial and dye removal. The nanocomposite was characterized by scanning electronic microscope (SEM), transmission electron microscopy (TEM), X-ray diffraction (XRD) and Fourier transform infrared spectrometer (FTIR). The antibacterial performance for MCGO against Escherichia coli was varied depending on the concentration of MCGO. SEM images of E. coli cells demonstrated that the antimicrobial performance of MCGO nanocomposite was possibly due to the damage of cell membrane. This work also explored MCGO's adsorption performance for methyl orange (MO). The experimental parameters including adsorbent mass, pH value, contact time and concentration of MO on the adsorption capacity were investigated. The maximum adsorption capacity of MCGO for MO was 398.08 mg/g. This study showed that the MCGO offered enormous potential applications for water treatment.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Chitosan/chemistry , Coloring Agents/chemistry , Graphite/chemistry , Magnetite Nanoparticles/chemistry , Nanocomposites/chemistry , Oxides/chemistry , Adsorption , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Escherichia coli/drug effects , Escherichia coli/ultrastructure , Hydrogen-Ion Concentration , Magnetite Nanoparticles/ultrastructure , Microbial Sensitivity Tests , Nanocomposites/ultrastructure , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
6.
Neurotox Res ; 28(1): 18-31, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25739536

ABSTRACT

Major depressive disorder and alcoholism are significant health burdens that can affect executive functioning, cognitive ability, job responsibilities, and personal relationships. Studies in animal models related to depression or alcoholism reveal that the expression of Krüppel-like factor 11 (KLF11, also called TIEG2) is elevated in frontal cortex, which suggests that KLF11 may play a role in stress- or ethanol-induced psychiatric conditions. KLF11 is a transcriptional activator of monoamine oxidase A and B, but also serves other functions in cell cycle regulation and apoptotic cell death. In the present study, immunohistochemistry was used to quantify intensity of nuclear KLF11, combined with an unbiased stereological approach to assess nuclei in fronto-limbic, limbic, and other brain regions of rats exposed chronically to social defeat or ethanol. KLF11 immunoreactivity was increased significantly in the medial prefrontal cortex, frontal cortex, and hippocampus of both stressed rats and rats fed ethanol. However, expression of KLF11 protein was not significantly affected in the thalamus, hypothalamus, or amygdala in either treatment group compared to respective control rats. Triple-label immunofluorescence revealed that KLF11 protein was localized in nuclei of neurons and astrocytes. KLF11 was also co-localized with the immunoreactivity of cleaved caspase-3. In addition, Western blot analysis revealed a significant reduction in anti-apoptotic protein, Bcl-xL, but an increase of caspase-3 expression in the frontal cortex of ethanol-treated rats compared to ethanol-preferring controls. Thus, KLF11 protein is up-regulated following chronic exposure to stress or ethanol in a region-specific manner and may contribute to pro-apoptotic signaling in ethanol-treated rats. Further investigation into the KLF11 signaling cascade as a mechanism for neurotoxicity and cell death in depression and alcoholism may provide novel pharmacological targets to lessen brain damage and maximize neuroprotection in these disorders.


Subject(s)
Apoptosis , Brain/metabolism , Ethanol/administration & dosage , Stress, Psychological/metabolism , Trans-Activators/metabolism , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Brain/drug effects , Caspase 3/metabolism , Dominance-Subordination , Ethanol/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , bcl-X Protein/metabolism
7.
Alcohol Clin Exp Res ; 39(3): 476-84, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25704249

ABSTRACT

BACKGROUND: Brain cell death is a major pathological consequence of alcohol neurotoxicity. However, the molecular cascades in alcohol-induced brain tissue injury are unclear. METHODS: Using Western blot and double immunofluorescence, we examined the expression of interferon (IFN)-induced protein kinase R (PKR), phosphorylated-PKR (p-PKR), and IFN gamma (IFNγ) in the prefrontal cortex (PFC) of postmortem brains from subjects with alcohol use disorders (AUD). RESULTS: The protein levels of PKR, p-PKR, and IFNγ were significantly increased in subjects with AUD compared with control subjects without AUD, and a younger age of onset of AUD was significantly correlated with higher protein levels of p-PKR. In addition, elevated PKR- and p-PKR-IR were observed in both neurons and astrocytes in the PFC of subjects with AUD compared to subjects without AUD. CONCLUSIONS: The activation of the IFNγ-PKR pathway in PFC of humans is associated with chronic excessive ethanol use with an age of onset dependent manner, and activation of this pathway may play a pivotal role in AUD-related brain tissue injury. This study provides insight into neurodegenerative key factors related to AUD and identifies potential targets for the treatment of alcohol-induced neurotoxicity.


Subject(s)
Alcohol-Related Disorders/metabolism , Interferon-gamma/biosynthesis , Prefrontal Cortex/metabolism , Signal Transduction , eIF-2 Kinase/biosynthesis , Adult , Alcohol-Related Disorders/pathology , Female , Humans , Male , Middle Aged , Prefrontal Cortex/pathology , Signal Transduction/physiology
8.
Neuropsychopharmacology ; 40(6): 1373-82, 2015 May.
Article in English | MEDLINE | ID: mdl-25502632

ABSTRACT

The biochemical pathways underlying major depressive disorder (MDD) and chronic stress are not well understood. However, it has been reported that monoamine oxidase A (MAO A, a major neurotransmitter-degrading enzyme) is significantly increased in the brains of human subjects affected with MDD and rats exposed to chronic social defeat (CSD) stress, which is used to model depression. In the current study, we compared the protein levels of a MAO A-transcriptional activator, Kruppel-like factor 11 (KLF11 , also recognized as transforming growth factor-beta-inducible early gene 2) between the brains of 18 human subjects with MDD and 18 control subjects. We found that, indeed, the expression of KLF11 is increased by 36% (p<0.02) in the postmortem prefrontal cortex of human subjects with MDD compared with controls. We also observed a positive correlation between KLF11 levels and those of its target gene, MAO A, both in association with MDD. KLF11 protein expression was also increased by 44% (p<0.02) in the frontal cortex of KLF11 wild-type mice (Klf11(+/+)) vs Klf11(-/-) when both exposed to CSD stress. In contrast, locomotor activities, central box duration and sucrose preference were significantly reduced in the stressed Klf11(+/+) mice, suggesting that Klf11(+/+) mice are more severely affected by the stress model compared with Klf11(-/-) mice. These results serve to assign an important role of KLF11 in upregulating MAO A in MDD and chronic social stress, suggesting that inhibition of the pathways regulated by this transcription factor may aid in the therapeutics of neuropsychiatric illnesses. Thus, the new knowledge derived from the current study extends our understanding of transcriptional mechanisms that are operational in the pathophysiology of common human diseases and thus bears significant biomedical relevance.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Depressive Disorder, Major/metabolism , Frontal Lobe/metabolism , Monoamine Oxidase/metabolism , Repressor Proteins/metabolism , Stress, Psychological/metabolism , Transcription Factors/metabolism , Animals , Apoptosis Regulatory Proteins , Chronic Disease , DNA-Binding Proteins/genetics , Disease Models, Animal , Dominance-Subordination , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Motor Activity/physiology , Severity of Illness Index , Transcription Factors/genetics , Up-Regulation
9.
Alcohol Clin Exp Res ; 38(2): 401-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24428663

ABSTRACT

BACKGROUND: Alcohol (EtOH [ethanol]) is an antinociceptive agent, working in part, by reducing sensitivity to painful stimuli. The transcription factor Kruppel-like factor 11 (KLF11), a human diabetes-causing gene that also regulates the neurotransmitter metabolic enzymes monoamine oxidase (MAO), has recently been identified as an EtOH-inducible gene. However, its role in antinociception remains unknown. Consequently, we investigated the function of KLF11 in chronic EtOH-induced antinociception using a genetically engineered knockout mouse model. METHODS: Wild-type (Klf11(+/+) ) and KLF11 knockout (Klf11(-/-) ) mice were fed a liquid diet containing EtOH for 28 days with increasing amounts of EtOH from 0% up to a final concentration of 6.4%, representing a final diet containing 36% of calories primarily from EtOH. Control mice from both genotypes were fed liquid diet without EtOH for 28 days. The EtOH-induced antinociceptive effect was determined using the tail-flick test before and after EtOH exposure (on day 29). In addition, the enzyme activity and mRNA levels of MAO A and MAO B were measured by real-time RT-PCR and enzyme assays, respectively. RESULTS: EtOH produced an antinociceptive response to thermal pain in Klf11(+/+) mice, as expected. In contrast, deletion of KLF11 in the Klf11(-/-) mice abolished the EtOH-induced antinociceptive effect. The mRNA and protein levels of KLF11 were significantly increased in the brain prefrontal cortex of Klf11(+/+) mice exposed to EtOH compared with control Klf11(+/+) mice. Furthermore, MAO enzyme activities were affected differently in Klf11 wild-type versus Klf11 knockout mice exposed to chronic EtOH. Chronic EtOH intake significantly increased MAO B activity in Klf11(+/+) mice. CONCLUSIONS: The data show KLF11 modulation of EtOH-induced antinociception. The KLF11-targeted MAO B enzyme may contribute more significantly to EtOH-induced antinociception. Thus, this study revealed a new role for the KLF11 gene in the mechanisms underlying the antinociceptive effects of chronic EtOH exposure.


Subject(s)
Alcoholism/genetics , Alcoholism/psychology , Analgesics , Central Nervous System Depressants/pharmacology , DNA-Binding Proteins/physiology , Diabetes Mellitus/genetics , Ethanol/pharmacology , Nociception/drug effects , Transcription Factors/physiology , Animals , Apoptosis Regulatory Proteins , Blotting, Western , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Male , Mice , Mice, Knockout , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Pain Measurement/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/enzymology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reaction Time/drug effects , Real-Time Polymerase Chain Reaction , Repressor Proteins , Transcription Factors/biosynthesis , Transcription Factors/genetics
10.
Alcohol Clin Exp Res ; 38(1): 144-51, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23915421

ABSTRACT

BACKGROUND: The biochemical pathways underlying alcohol abuse and dependence are not well understood, although brain cell loss and neurotoxicity have been reported in subjects with alcohol dependence. Monoamine oxidase B (MAO B; an enzyme that catabolizes neurotransmitters such as dopamine) is consistently increased in this psychiatric illness. MAO B has been implicated in the pathogenesis of alcohol dependence and alcohol-induced brain neurotoxicity. Recently, the cell growth inhibitor protein, Kruppel-like factor 11 (KLF11), has been reported to be an MAO transcriptional activator. KLF11 is also known as TIEG2 (transforming growth factor-beta-inducible early gene 2) and mediates apoptotic cell death. This study investigates the protein expression of KLF11 and its relationship with MAO B using human postmortem prefrontal cortex from subjects with alcohol dependence. METHODS: Twelve subjects with alcohol dependence and the respective psychiatrically normal control subjects were investigated. Expression of KLF11 and MAO B proteins in the prefrontal cortex was measured by Western blot analysis. Correlation studies involving KLF11 and MAO B protein expression were performed. Localization of KLF11 in the human prefrontal cortex was also determined by immunohistochemistry. RESULTS: Levels of KLF11 protein were significantly increased by 44% (p < 0.03) in the postmortem prefrontal cortex of subjects with alcohol dependence as compared to age- and gender-matched, psychiatrically normal control subjects. Furthermore, KLF11 levels were significantly and positively correlated with both the increased MAO B protein levels and blood alcohol content in alcohol-dependent subjects. In addition, KLF11 protein expression was visualized in both neuronal and glial cells. CONCLUSIONS: This novel study shows the important role of KLF11, an MAO transcriptional activator, in human alcohol dependence. It further supports that the KLF11-MAO B cell death cascade may contribute to chronic alcohol-induced brain damage. This argues a case for KLF11-MAO B inhibition as a novel therapeutic strategy that may impact this highly prevalent illness.


Subject(s)
Alcoholism/metabolism , Cell Cycle Proteins/biosynthesis , Gene Expression Regulation , Monoamine Oxidase/biosynthesis , Prefrontal Cortex/metabolism , Repressor Proteins/biosynthesis , Transcriptional Activation/physiology , Alcoholism/pathology , Apoptosis Regulatory Proteins , Female , Humans , Male , Middle Aged , Prefrontal Cortex/pathology , Retrospective Studies
11.
J Chromatogr A ; 1318: 251-6, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24354006

ABSTRACT

A microchip electrophoresis-mass spectrometric (MCE-MS) method was developed for fast chiral analysis. The proposed MCE-MS platform deployed a glass/PDMS hybrid microchip with an easy-to-fabricate monolithic nanoelectrospray emitter. Enantiomeric MCE separation was achieved by means of the partial filling technique. A novel chip design with an arm channel connecting to the middle of the MCE separation channel for delivering the chiral selector was tested and proven valid. Enantiomeric separation of3.4-dihydroxyphenylalanine (DOPA), glutamic acid (Glu), and serine (Ser), the selected test compounds,were achieved within 130 s with resolution values (R(s)) of 2.4, 1.1, and 1.0, respectively. The proposed chiral MCE-MS assay was sensitive and had detection limits of 43 nM for l-DOPA and 47 nM for d-DOPA.The analytical platform was well suited for studies of stereochemical preference in living cells because it integrated cell culture, sample injection, chiral separation, and MS detection into a single platform.Metabolism of DOPA in human SH-SY5Y neuronal cells was studied as a model system. On-chip incubation of SH-SY5Y cells with racemic DOPA was carried out, and the incubation solution was injected and in-line assayed at time intervals. It was found that l-DOPA concentration decreased gradually as incubation time increased while the concentration of coexisting d-DOPA remained constant. The results firmly indicated that SH-SY5Y cells metabolized l-DOPA effectively while left d-DOPA intact.


Subject(s)
Dihydroxyphenylalanine/chemistry , Electrophoresis, Microchip/methods , Glutamic Acid/chemistry , Mass Spectrometry/methods , Serine/chemistry , Cell Line , Cells/chemistry , Cells/metabolism , Dihydroxyphenylalanine/metabolism , Humans , Stereoisomerism
12.
J Pain ; 14(6): 549-57, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23602266

ABSTRACT

UNLABELLED: Epigenetic regulation of gene expression is a rapidly growing area of research. Considering the longevity and plasticity of neurons, the studies on epigenetic pathways in the nervous system should be of special interest for both epigeneticists and neuroscientists. Activation or inactivation of different epigenetic pathways becomes more pronounced when the cells experience rapid changes in their environment, and such changes can be easily caused by injury and inflammation, resulting in pain perception or distortion of pain perception (eg, hyperalgesia). Therefore, in this regard, the field of pain is at an advantage to study the epigenetic pathways. More importantly, understanding pain from an epigenetics point of view would provide a new paradigm for developing drugs or strategies for pain management. In this review, we introduce basic concepts of epigenetics, including chromatin dynamics, histone modifications, DNA methylation, and RNA-induced gene silencing. In addition, we provide evidence from published studies suggesting wide implication of different epigenetic pathways within pain pathways. PERSPECTIVE: This article provides a brief overview of epigenetic pathways for gene regulation and highlights their involvement in pain. Our goal is to expose the readers to these concepts so that pain-related phenotypes can be investigated from the epigenetic point of view.


Subject(s)
Epigenesis, Genetic , Pain Management , Pain , Humans , Pain/diagnosis , Pain/genetics , Pain/pathology
13.
J Neurosci ; 32(48): 17120-7, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197705

ABSTRACT

Monoamine oxidase-A (MAO-A), a key brain enzyme which metabolizes monoamines, is implicated in the pathophysiology of stress-related illnesses, including major depressive disorder, addiction, and violent behavior. Chronic stressors and glucocorticoid-administration typically associate with elevated MAO-A levels/activity. However, the relationship of shorter stress or glucocorticoid exposures and MAO-A levels/activity is not well established. Our objectives are to assess effects of acute stress upon MAO-A V(T,) an index of MAO-A density, in human brain and acute glucocorticoid exposure upon MAO-A levels in human neuronal and glial cell lines. Twelve healthy, non-smoking participants aged 18-50 underwent [(11)C]harmine positron emission tomography to measure brain MAO-A V(T) on two different days: One under acute psychosocial stress (via Trier Social Stress and Montreal Imaging Stress Tasks) and one under a non-stress condition. MAO-A density (by Western blot) and activity (by [(14)C]-5-HT metabolism and liquid scintillation spectroscopy) were measured in human neuronal and glial cell lines after 4 h exposure to dexamethasone. We observed a significant reduction in whole-brain MAO-A binding as reflected by reductions in 10 of 11 brain regions. Acute dexamethasone exposure in neuronal and glial cells significantly decreased MAO-A activity and protein levels. We observed a highly consistent relationship between acute stressors and glucocorticoid administration and decreased MAO-A binding, activity and protein levels. Since MAO-A metabolizes monoamines, this phenomenon may explain why acute stressors benefit healthy animals even though chronic stress is associated with illness.


Subject(s)
Brain/metabolism , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Monoamine Oxidase/metabolism , Stress, Psychological/metabolism , Adult , Brain/diagnostic imaging , Brain/drug effects , Cell Line, Tumor , Female , Humans , Hydrocortisone/blood , Male , Middle Aged , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Radionuclide Imaging , Stress, Psychological/diagnostic imaging
14.
Drug Discov Ther ; 6(3): 112-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22890201

ABSTRACT

Monoamine oxidases play an integral role in brain function. Both monoamine oxidase A (MAO-A) and monoamine oxidase B (MAO-B) regulate neurochemistry by degrading monoamine neurotransmitters (serotonin, dopamine, and norepinephrine). Any alteration in MAO levels can have devastating effects on the brain and behavior by lowering or raising neurotransmitter levels and producing toxic reactive oxygen species. In this review article, MAO is examined in terms of function and genetic organization, with special focus on recent discoveries related to the transcriptional regulation of MAO. In recent studies, transcriptional regulation involves a repressor protein, R1, for MAO-A and an activator protein, KLF11 (a Krüppel-like factor; also referred to as transforming growth factor-beta early inducible gene 2, TIEG2), for both MAO-A and MAO-B, by binding to Sp/KLF sites in the core promoters of MAO and regulating MAO gene expression. Furthermore, KLF11 may influence MAO-B expression and augment glyceraldehyde-3 phosphate dehydrogenase (GAPDH) to upregulate MAO-B transcription upon exposure to ethanol. Finally, we review recent progress in MAO research and highlight the roles that MAOs play in several psychiatric conditions, including chronic stress, major depressive disorder and alcohol dependence. Further research in this area is needed to better understand MAOs, their transcription factors and signaling pathways in psychiatric illnesses in order to develop new strategies for pharmacological advancement.


Subject(s)
Alcoholism/enzymology , Depressive Disorder, Major/enzymology , Monoamine Oxidase/physiology , Alcoholism/etiology , Apoptosis Regulatory Proteins , Cell Cycle Proteins/physiology , Depressive Disorder, Major/etiology , Humans , Monoamine Oxidase/genetics , Repressor Proteins/physiology , Stress, Psychological/enzymology , Transcription, Genetic
15.
J Biol Chem ; 287(29): 24195-206, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22628545

ABSTRACT

Chronic stress is a risk factor for psychiatric illnesses, including depressive disorders, and is characterized by increased blood glucocorticoids and brain monoamine oxidase A (MAO A, which degrades monoamine neurotransmitters). This study elucidates the relationship between stress-induced MAO A and the transcription factor Kruppel-like factor 11 (KLF11, also called TIEG2, a member of the Sp/KLF- family), which inhibits cell growth. We report that 1) a glucocorticoid (dexamethasone) increases KLF11 mRNA and protein levels in cultured neuronal cells; 2) overexpressing KLF11 increases levels of MAO A mRNA and enzymatic activity, which is further enhanced by glucocorticoids; in contrast, siRNA-mediated KLF11 knockdown reduces glucocorticoid-induced MAO A expression in cultured neurons; 3) induction of KLF11 and translocation of KLF11 from the cytoplasm to the nucleus are key regulatory mechanisms leading to increased MAO A catalytic activity and mRNA levels because of direct activation of the MAO A promoter via Sp/KLF-binding sites; 4) KLF11 knockout mice show reduced MAO A mRNA and catalytic activity in the brain cortex compared with wild-type mice; and 5) exposure to chronic social defeat stress induces blood glucocorticoids and activates the KLF11 pathway in the rat brain, which results in increased MAO A mRNA and enzymatic activity. Thus, this study reveals for the first time that KLF11 is an MAO A regulator and is produced in response to neuronal stress, which transcriptionally activates MAO A. The novel glucocorticoid-KLF11-MAO A pathway may play a crucial role in modulating distinct pathophysiological steps in stress-related disorders.


Subject(s)
Cell Cycle Proteins/metabolism , Monoamine Oxidase/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins , Blotting, Western , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cells, Cultured , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Corticosterone/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dexamethasone/metabolism , Fluorescent Antibody Technique , Gene Expression/drug effects , Humans , Male , Mice , Mice, Knockout , Monoamine Oxidase/genetics , Radioimmunoassay , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Serotonin/metabolism , Stress, Physiological/genetics , Stress, Physiological/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
16.
PLoS One ; 7(3): e33493, 2012.
Article in English | MEDLINE | ID: mdl-22428062

ABSTRACT

The molecular mechanisms for the discrepancy in outcome of initiating estrogen therapy (ET) around peri-menopause or several years after menopause in women are unknown. We hypothesize that the level of expression of a dominant negative estrogen receptor (ER) ß variant, ERß2, may be a key factor determining the effectiveness of ET in post-menopausal women. We tested this hypothesis in ovariectomized nine month-old (an age when irregular estrous cycles occur) female Sprague Dawley rats. Estradiol treatment was initiated either 6 days (Early ET, analogous to 4 months post-menopause in humans), or 180 days (Late ET, analogous to 11 years post-menopause in humans) after ovariectomy. Although ERß2 expression increased in all OVX rats, neurogenic and neuroprotective responses to estradiol differed in Early and Late ET. Early ET reduced ERß2 expression in both hippocampus and white blood cells, increased the hippocampal cell proliferation as assessed by Ki-67 expression, and improved mobility in the forced swim test. Late ET resulted in either no or modest effects on these parameters. There was a close correlation between the degree of ERß2 expression and the preservation of neural effects by ET after OVX in rats, supporting the hypothesis that persistent elevated levels of ERß2 are a molecular basis for the diminished effectiveness of ET in late post-menopausal women. The correlation between the expression of ERß2 in circulating white blood cells and brain cells suggests that ERß2 expression in peripheral blood cells may be an easily accessible marker to predict the effective window for ET in the brain.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Estrogen Replacement Therapy/methods , Ovariectomy/adverse effects , Protein Isoforms/metabolism , Analysis of Variance , Animals , Blotting, Western , Estrogen Receptor beta/genetics , Female , Flow Cytometry , Hippocampus/cytology , Hippocampus/metabolism , Leukocytes/metabolism , Motor Activity/drug effects , Protein Isoforms/genetics , Rats , Rats, Sprague-Dawley
17.
Neuropsychopharmacology ; 36(10): 2139-48, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21654740

ABSTRACT

The novel transcriptional repressor protein, R1 (JPO2/CDCA7L/RAM2), inhibits monoamine oxidase A (MAO A) gene expression and influences cell proliferation and survival. MAO A is implicated in several neuropsychiatric illnesses and highly elevated in major depressive disorder (MDD); however, whether R1 is involved in these disorders is unknown. This study evaluates the role of R1 in depressed subjects either untreated or treated with antidepressant drugs. R1 protein levels were determined in the postmortem prefrontal cortex of 18 untreated MDD subjects and 12 medicated MDD subjects compared with 18 matched psychiatrically normal control subjects. Western blot analysis showed that R1 was significantly decreased by 37.5% (p<0.005) in untreated MDD subjects. The R1 level in medicated MDD subjects was also significantly lower (by 30%; p<0.05) compared with control subjects, but was not significantly different compared with untreated MDD subjects. Interestingly, the reduction in R1 was significantly correlated with an increase (approximately 40%; p<0.05) in MAO A protein levels within the MDD groups compared with controls. Consistent with the change in MAO A protein expression, the MAO A catalytic activity was significantly greater in both MDD groups compared with controls. These results suggest that reduced R1 may lead to elevated MAO A levels in untreated and treated MDD subjects; moreover, the reduction of R1 has been implicated in apoptotic cell death and apoptosis has also been observed in the brains of MDD subjects. Therefore, modulation of R1 levels may provide a new therapeutic target in the development of more effective strategies to treat MDD.


Subject(s)
Depressive Disorder, Major/metabolism , Monoamine Oxidase/deficiency , Monoamine Oxidase/genetics , Repressor Proteins/antagonists & inhibitors , Apoptosis/genetics , Apoptosis/physiology , Depressive Disorder, Major/enzymology , Depressive Disorder, Major/genetics , Female , Humans , Male , Middle Aged , Monoamine Oxidase/biosynthesis , Repressor Proteins/physiology , Retrospective Studies
18.
Mol Pharmacol ; 79(2): 308-17, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20980443

ABSTRACT

Glucocorticoid steroid hormones play important roles in many neurophysiological processes such as responses to stress, behavioral adaption, and mood. One mechanism by which glucocorticoids exert functions in the brain is via the modulation of neurotransmission systems. Glucocorticoids are capable of inducing the activities of monoamine oxidases (MAOs), which degrade monoamine neurotransmitters including serotonin, norepinephrine, phenylethylamine, and dopamine. However, the molecular mechanisms for such induction are not yet fully understood. Here, we report that dexamethasone, a synthetic glucocorticoid hormone, stimulates MAO B (an isoform of MAOs) promoter and catalytic activities via both the fourth glucocorticoid response element (GRE) and simian virus 40 promoter factor 1 (Sp1) binding sites in MAO B promoter. Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation analysis demonstrated that glucocorticoid receptor binds to the fourth GRE in vitro and in vivo. Using Sp1-binding motifs as bait in a yeast one-hybrid system, we identified two novel transcriptional repressors of MAO B, E2F-associated phosphoprotein (EAPP) and R1 (RAM2/CDCA7L/JPO2), that down-regulate MAO B via MAO B core promoter, which contains Sp1 sites. EMSA suggested that EAPP and R1 competed with Sp1 for binding to the Sp1 site in vitro. Moreover, EAPP and R1 reduced Sp1-activated glucocorticoid activation of MAO B promoter. In response to dexamethasone, lower occupancy by EAPP and R1 and higher occupancy by Sp1 were shown at the natural MAO B core promoter. Together, this study uncovers for the first time the molecular mechanisms for glucocorticoid activation of MAO B gene and provides new insights into the hormonal regulation of MAO.


Subject(s)
Dexamethasone/pharmacology , E2F Transcription Factors/metabolism , Monoamine Oxidase/metabolism , Phosphoproteins/metabolism , Sp1 Transcription Factor/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers , Enzyme Activation , Humans , Monoamine Oxidase/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic
19.
Neurotox Res ; 19(4): 511-8, 2011 May.
Article in English | MEDLINE | ID: mdl-20204567

ABSTRACT

Brain cell loss has been reported in subjects with alcoholism. However, the molecular mechanisms are unclear. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and monoamine oxidase B (MAO B) reportedly play a role in cellular dysfunction with regards to ethanol exposure. We have recently reported that GAPDH protein expression was increased in the brains of rats fed with ethanol. Furthermore, GAPDH interacts with the transcriptional activator, transforming growth factor-beta-inducible early gene 2 (TIEG2), to augment TIEG2-mediated MAO B activation, resulting in neuronal cell damage due to ethanol exposure. The current study investigates whether the TIEG2-MAO B cascade is also active in the brains of rats fed with ethanol. Ten ethanol-preferring rats were fed with a liquid diet containing ethanol, with increasing amounts of ethanol up to a final concentration of 6.4% representing a final diet containing 36% of calories for 28 days. Ten control rats were fed the liquid diet without ethanol. The expression of TIEG2 protein, MAO B mRNA levels, MAO B catalytic activity, and the levels of anti-apoptotic protein Bcl 2 and apoptotic protein caspase 3 were determined in the prefrontal cortex of the rats. Ethanol significantly increased protein levels of TIEG2, active caspase 3, MAO B mRNA and enzyme activity, but significantly decreased Bcl 2 protein expression compared to control rats. In summary, ethanol increases the TIEG2-MAO B brain cell death cascade in rat brains, suggesting that the TIEG2-MAO B pathway is a novel pathway for brain cell damage resulting from ethanol exposure, and may contribute to chronic alcohol-induced brain damage.


Subject(s)
Alcohol Drinking/metabolism , Ethanol/administration & dosage , Monoamine Oxidase/biosynthesis , Prefrontal Cortex/enzymology , Signal Transduction/physiology , Trans-Activators/biosynthesis , Alcohol Drinking/pathology , Animals , Cell Death/drug effects , Cell Death/physiology , Ethanol/pharmacology , Male , Prefrontal Cortex/drug effects , Random Allocation , Rats , Rats, Wistar , Signal Transduction/drug effects , Trans-Activators/genetics
20.
Front Neurosci ; 4: 180, 2010.
Article in English | MEDLINE | ID: mdl-21103012

ABSTRACT

Stress detrimentally affects the brain and body and can lead to or be accompanied by depression. Although stress and depression may contribute to each other, the exact molecular mechanism underlying the effects is unclear. However, there is a correlation between stress and an increase in glucocorticoid secretion which causes a subsequent increase in monoamine oxidase (MAO) activity during stress. Consequently, MAO inhibitors have been used as traditional antidepressant drugs. Cellular treatment with the synthetic glucocorticoid, dexamethasone (a cellular stressor), has been reported to markedly increase both MAO A and MAO B catalytic activities, as well as apoptosis. This study compares the neuroprotective abilities of M30 (a new generation inhibitor of both MAO A and MAO B) with rasagiline (Azilect(®), another new MAO B inhibitor) and selegiline (Deprenyl(®), a traditional MAO B inhibitor) in the prevention of dexamethasone-induced brain cell death and MAO activity in human neuroblastoma cells, SH-SY5Y. M30 demonstrated the highest inhibitory effect on MAO A; however, M30 showed the lowest inhibitory effect on MAO B enzymatic activity in comparison to rasagiline and selegiline. Although, M30 exhibited the greatest neuroprotective effect by decreasing cell death rates and apoptotic DNA damage compared to rasagiline and selegiline, these neuroprotective effects of M30 were, overall, similar to rasagiline. Summarily, M30 has a generally greater impact on neuroprotection than the MAO B inhibitors, selegiline and rasagiline. Our results suggest that M30 may have great potential in alleviating disorders involving increases in both MAO A and MAO B, such as stress-induced disorders.

SELECTION OF CITATIONS
SEARCH DETAIL
...