Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Front Aging Neurosci ; 13: 674688, 2021.
Article in English | MEDLINE | ID: mdl-34276338

ABSTRACT

In mouse models of Alzheimer's disease (AD), normobaric intermittent hypoxia training (IHT) can preserve neurobehavioral function when applied before deficits develop, but IHT's effectiveness after onset of amyloid-ß (Aß) accumulation is unclear. This study tested the hypothesis that IHT improves learning-memory behavior, diminishes Aß accumulation in cerebral cortex and hippocampus, and enhances cerebrocortical contents of the neuroprotective trophic factors erythropoietin and brain-derived neurotrophic factor (BDNF) in mice manifesting AD traits. Twelve-month-old female 3xTg-AD mice were assigned to untreated 3xTg-AD (n = 6), AD+IHT (n = 6), and AD+sham-IHT (n = 6) groups; 8 untreated wild-type (WT) mice also were studied. AD+IHT mice alternately breathed 10% O2 for 6 min and room air for 4 min, 10 cycles/day for 21 days; AD+sham-IHT mice breathed room air. Spatial learning-memory was assessed by Morris water maze. Cerebrocortical and hippocampal Aß40 and Aß42 contents were determined by ELISA, and cerebrocortical erythropoietin and BDNF were analyzed by immunoblotting and ELISA. The significance of time (12 vs. 12 months + 21 days) and treatment (IHT vs. sham-IHT) was evaluated by two-factor ANOVA. The change in swimming distance to find the water maze platform after 21 d IHT (-1.6 ± 1.8 m) differed from that after sham-IHT (+5.8 ± 2.6 m). Cerebrocortical and hippocampal Aß42 contents were greater in 3xTg-AD than WT mice, but neither time nor treatment significantly affected Aß40 or Aß42 contents in the 3xTg-AD mice. Cerebrocortical erythropoietin and BDNF contents increased appreciably after IHT as compared to untreated 3xTg-AD and AD+sham-IHT mice. In conclusion, moderate, normobaric IHT prevented spatial learning-memory decline and restored cerebrocortical erythropoietin and BDNF contents despite ongoing Aß accumulation in 3xTg-AD mice.

2.
Pharmacol Biochem Behav ; 191: 172873, 2020 04.
Article in English | MEDLINE | ID: mdl-32105662

ABSTRACT

Many elderly American women use CNS depressant benzodiazepine (BZD) to ameliorate anxiety or insomnia. However, the chronic use of BZD (cBZD) is prevalent, causing adverse effects of BZD that include movement deficit. We previously reported that cBZD upregulates neurotoxic amyloid ß42 (Aß42) and downregulates neuroprotective translocator protein (TSPO) in the cerebellum, the brain area of movement and balance. The aim of the current study is two-fold: 1) to determine a direct effect of TSPO (inhibition) on cBZD-induced Aß42 and Aß-associated molecules; Aß-producing-protein presenilin-1 (PS1) and Aß-degrading-enzyme neprilysin and 2) to determine whether Aß42 upregulation and motoric deficit occur upon a long-term (cBZD) rather than a short-term BZD (sBZD) treatment. Old female mice received BZD (lorazepam) for 20 days (cBZD) or 3 days (sBZD) with or without prototype TSPO ligand PK11195 and were tested for motoric performance for 3 days using Rotarod. ELISA was conducted to measure Aß42 level and neprilysin activity in cerebellum. RT-PCR and immunoblot were conducted to measure the mRNA and protein levels of TSPO, PS1, and neprilysin. cBZD treatment decreased TSPO and neprilysin but increased Aß42 accompanied by motoric deficit. Chronic PK11195 treatment acted as a TSPO inhibitor by suppressing TSPO expression and mimicked or exacerbated the effects of cBZD on all parameters measured except for PS1. None of the molecular and behavioral changes induced by cBZD were reproduced by sBZD treatment. These data suggest that cBZD upregulates Aß42 and downregulates neprilysin in part through TSPO inhibition, the mechanisms distinct from sBZD, collectively contributing to motoric deficit.


Subject(s)
Amyloid beta-Peptides/metabolism , Benzodiazepines/pharmacology , Central Nervous System Depressants/pharmacology , Motor Activity/drug effects , Peptide Fragments/metabolism , Receptors, GABA/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Animals , Cerebellum/metabolism , Down-Regulation/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Isoquinolines/pharmacology , Mice , Mice, Inbred C57BL , Neprilysin/metabolism , Rotarod Performance Test
3.
Curr Alzheimer Res ; 17(1): 3-15, 2020.
Article in English | MEDLINE | ID: mdl-32065102

ABSTRACT

Translocator Protein (18 kDa) (TSPO) is a mitochondrial protein that locates cytosol cholesterol to mitochondrial membranes to begin the synthesis of steroids including neurotrophic neurosteroids. TSPO is abundantly present in glial cells that support neurons and respond to neuroinflammation. Located at the outer membrane of mitochondria, TSPO regulates the opening of mitochondrial permeability transition pore (mPTP) that controls the entry of molecules necessary for mitochondrial function. TSPO is linked to neurodegenerative Alzheimer's Disease (AD) such that TSPO is upregulated in the brain of AD patients and signals AD-induced adverse changes in brain. The initial increase in TSPO in response to brain insults remains elevated to repair cellular damages and perhaps to prevent further neuronal degeneration as AD progresses. To exert such protective activities, TSPO increases the synthesis of neuroprotective steroids, decreases neuroinflammation, limits the opening of mPTP, and reduces the generation of reactive oxygen species. The beneficial effects of TSPO on AD brain are manifested as the attenuation of neurotoxic amyloid ß and mitochondrial dysfunction accompanied by the improvement of memory and cognition. However, the protective activities of TSPO appear to be temporary and eventually diminish as the severity of AD becomes profound. Timely treatment with TSPO agonists/ligands before the loss of endogenous TSPO's activity may promote the protective functions and may extend neuronal survival.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Receptors, GABA/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Neurosteroids/metabolism , Reactive Oxygen Species/metabolism
4.
Pharmacol Biochem Behav ; 172: 59-67, 2018 09.
Article in English | MEDLINE | ID: mdl-30030126

ABSTRACT

Benzodiazepine (BZD) is a commonly prescribed anxiolytic and sedation aid medication, especially in elderly women. However, long-term use of BZD provokes adverse nontherapeutic effects that include movement deficit. Here, we investigated motoric deficit and molecular changes in cerebellum associated with the chronic use of BZD (cBZD) in female mice. We measured neuroprotective translocator protein (TSPO), neurotoxic amyloid ß (Aß), Aß-producing presenilin-1 (PS1), and Aß-degrading neprilysin. We also tested whether cBZD treatment damages mitochondrial membranes by measuring mitochondrial membrane swelling and mitochondrial respiration. Young and old mice received BZD (lorazepam) for 20 days, were tested for motoric function using Rotarod, and then euthanized to collect cerebellum. The major methods were immunoblot and RT-PCR for TSPO, PS1, and neprilysin expressions; ELISA for Aß level; spectrometry for mitochondrial membrane swelling; XF-respirometry for mitochondrial respiration. cBZD-treated old mice showed poorer motoric function than old control or young cBZD-treated mice. Old mice treated with cBZD showed a decrease in TSPO and neprilysin and an increase in Aß and PS1 production compared to old control mice. Old cBZD-mice also showed an increase in mitochondrial membrane swelling and a decrease in mitochondrial respiration. These data suggest that cBZD exacerbates motoric aging in a manner that involves diminished TSPO, elevated Aß, and mitochondrial damage.


Subject(s)
Amyloid beta-Peptides/metabolism , Lorazepam/administration & dosage , Receptors, GABA/metabolism , Age Factors , Amyloid beta-Peptides/biosynthesis , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Lorazepam/pharmacology , Mice, Inbred C57BL , Mitochondrial Membranes/drug effects , Mitochondrial Swelling , Models, Animal , Motor Activity/drug effects , Neprilysin/biosynthesis , Neprilysin/metabolism , Respiration/drug effects , Reverse Transcriptase Polymerase Chain Reaction
5.
Respir Physiol Neurobiol ; 256: 67-78, 2018 10.
Article in English | MEDLINE | ID: mdl-28811138

ABSTRACT

Ethanol intoxication and withdrawal exact a devastating toll on the central nervous system. Abrupt ethanol withdrawal provokes massive release of the excitatory neurotransmitter glutamate, which over-activates its postsynaptic receptors, causing intense Ca2+ loading, p38 mitogen activated protein kinase activation and oxidative stress, culminating in ATP depletion, mitochondrial injury, amyloid ß deposition and neuronal death. Collectively, these mechanisms produce neurocognitive and sensorimotor dysfunction that discourages continued abstinence. Although the brain is heavily dependent on blood-borne O2 to sustain its aerobic ATP production, brief, cyclic episodes of moderate hypoxia and reoxygenation, when judiciously applied over the course of days or weeks, evoke adaptations that protect the brain from ethanol withdrawal-induced glutamate excitotoxicity, mitochondrial damage, oxidative stress and amyloid ß accumulation. This review summarizes evidence from ongoing preclinical research that demonstrates intermittent hypoxia training to be a potentially powerful yet non-invasive intervention capable of affording robust, sustained neuroprotection during ethanol withdrawal.


Subject(s)
Alcoholism/complications , Hypoxia , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/therapy , Animals , Brain Injuries/etiology , Humans , Substance Withdrawal Syndrome/pathology
6.
Am J Physiol Regul Integr Comp Physiol ; 313(1): R10-R18, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28490448

ABSTRACT

Abrupt cessation of chronic alcohol consumption triggers signaling cascades that harm vulnerable brain regions and produce neurobehavioral deficits. We have demonstrated that a program of intermittent, normobaric hypoxia training (IHT) in rats prevents brain damage and neurobehavioral impairment resulting from abrupt ethanol withdrawal (EW). Moreover, EW induced expression of stress-activated protein kinase p38 and presenilin 1 (PS1), the catalytic subunit of γ-secretase that produces the neurotoxic amyloid-ß (Aß) peptides Aß40 and Aß42. We tested the hypotheses that 1) IHT limits EW-induced activation of the p38-PS1 axis, thereby attenuating γ-secretase activation and Aß accumulation, and 2) EW disables heat shock protein 25 (HSP25), a p38 substrate, molecular chaperone, and antioxidant, and provokes protein carbonylation in a manner suppressed by IHT. Adult male rats completed two cycles of a 4-wk ethanol diet (6.5% wt/vol) and a 3-wk EW or an isocaloric, dextrin-based control diet. A 20-day IHT program (5-8 daily cycles of 5-10 min of 9.5-10% fractional inspired O2 + 4 min of 21% fractional inspired O2) was administered during the first EW phase. After the second EW phase, the brain was excised and the prefrontal cortex extracted. PS1, phosphorylated p38 (p-p38), and HSP25 were analyzed by immunoblot, PS1 messenger RNA by quantitative polymerase chain reaction, protein carbonyl content by spectrometry, and Aß40 and Aß42 contents by enzyme-linked immunosorbent assay. IHT attenuated the EW-associated increases in PS1, p-p38, Aß40, Aß42, and protein carbonyl contents, but not that of PS1 messenger RNA, while preserving functionally competent HSP25 dimers in EW rats. Collectively, these findings suggest that IHT may attenuate EW-induced γ-secretase overactivation by suppressing activation of the p38-PS1 axis and by preventing oxidative protein damage.


Subject(s)
Amyloid beta-Peptides/metabolism , Cerebral Cortex/metabolism , Ethanol/toxicity , Hypoxia/metabolism , Presenilin-1/metabolism , Animals , Cerebral Cortex/drug effects , Gene Expression Regulation , HSP27 Heat-Shock Proteins/metabolism , Ischemic Preconditioning , Male , Oxygen , Presenilin-1/genetics , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/prevention & control , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Behav Brain Res ; 314: 199-214, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27503149

ABSTRACT

Ethanol withdrawal (EW) is referred to the abrupt termination of long-term heavy drinking, and provokes oxidative brain damage. Here, we investigated whether the cerebellum and hippocampus of female rats are less affected by prooxidant EW than male rats due to the antioxidant effect of 17ß-estradiol (E2). Female and male rats received a four-week ethanol diet and three-week withdrawal per cycle for two cycles. Some female rats were ovariectomized with E2 or antioxidant (Vitamin E+Co-Q10) treatment. Measurements were cerebellum (Rotarod) and hippocampus (water-maze)-related behaviors, oxidative markers (O2(-), malondialdehyde, protein carbonyls), mitochondrial membrane swelling, and a key mitochondrial enzyme, cytochrome c oxidase (CcO). Separately, HT22 (hippocampal) cells were subjected to ethanol-exposure and withdrawal for two cycles to assess the effect of a CcO inhibitor on E2's protection for mitochondrial respiration and cell viability. Ethanol-withdrawn female rats showed a smaller increase in oxidative markers in cerebellum and hippocampus than male rats, and E2 treatment decreased the oxidative markers. Compared to male counterparts, ethanol-withdrawn female rats showed better Rotarod but poorer water-maze performance, accompanied by more severe mitochondrial membrane swelling and CcO suppression in hippocampus. E2 or antioxidant treatment improved Rotarod but not water-maze performance. In the presence of a CcO inhibitor, E2 treatment failed to protect mitochondrial respiration and cell viability from EW. These data suggest that antioxidant E2 contributes to smaller oxidative stress in ethanol-withdrawn female than male rats. They also suggest that EW-induced severe mitochondrial damage in hippocampus may blunt E2's antioxidant protection for hippocampus-related behavior.


Subject(s)
Ethanol/pharmacology , Oxidative Stress/drug effects , Sex Characteristics , Substance Withdrawal Syndrome/metabolism , Animals , Antioxidants/pharmacology , Estradiol/pharmacology , Female , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
8.
J Pharmacol Exp Ther ; 358(3): 516-26, 2016 09.
Article in English | MEDLINE | ID: mdl-27278235

ABSTRACT

Presenilin-1 (PS1) is a core component of γ-secretase that is involved in neurodegeneration. We have previously shown that PS1 interacts with a mitogen-activated protein kinase [(MAPK) jun-NH2-terminal-kinase], and another MAPK (p38) is activated by ethanol withdrawal (EW), abrupt termination from chronic ethanol exposure. EW is excitotoxic in nature, induces glutamate upregulation, and provokes neuronal damage. Here, we explored a potential mechanistic pathway involving glutamate, p38 (p38α isozyme), and PS1 that may mediate EW-induced excitotoxic stress. We used the prefrontal cortex of male rats withdrawn from a chronic ethanol diet. Additionally, we used ethanol-withdrawn HT22 cells (mouse hippocampal) treated with the inhibitor of glutamate receptors [dizocilpine (MK-801)], p38α (SB203580; 4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine), or γ-secretase [N-[N- (3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT)] during EW. Separately, ethanol-free HT22 cells were exposed to glutamate with or without SB203580 or DAPT. Protein levels, mRNA levels, and cell viability were assessed using immunoblotting, qualitative polymerase chain reaction, and calcein assay, respectively. The prefrontal cortex of ethanol-withdrawn rats or HT22 cells showed an increase in PS1 and p38α, which was attenuated by MK-801 and SB203580, but mimicked by glutamate treatment to ethanol-free HT22 cells. DAPT attenuated the toxic effect of EW or glutamate on HT22 cells. These results suggest that PS1 expression is triggered by glutamate through p38α, contributing to the excitotoxic stimulus of EW.


Subject(s)
Ethanol/pharmacology , Presenilin-1/metabolism , Substance Withdrawal Syndrome/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Body Weight/drug effects , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Ethanol/blood , Gene Expression Regulation, Enzymologic/drug effects , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mitogen-Activated Protein Kinase 14/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/pathology
9.
Int J Med Sci ; 12(5): 432-40, 2015.
Article in English | MEDLINE | ID: mdl-26078703

ABSTRACT

Intermittent hypoxia preconditioning (IHP) has been shown to protect neurons against ischemic stroke injury. Studying how proteins respond to IHP may identify targets that can help fight stroke. The objective of the present study was to investigate whether mitochondrial dihydrolipoamide dehydrogenase (DLDH) would respond to IHP and if so, whether such a response could be linked to neuroprotection in ischemic stroke injury. To do this, we subjected male rats to IHP for 20 days and measured the content and activity of DLDH as well as the three α-keto acid dehydrogenase complexes that contain DLDH. We also measured mitochondrial electron transport chain enzyme activities. Results show that DLDH content was indeed upregulated by IHP and this upregulation did not alter the activities of the three α-keto acid dehydrogenase complexes. Results also show that the activities of the five mitochondrial complexes (I-V) were not altered either by IHP. To investigate whether IHP-induced DLDH upregulation is linked to neuroprotection against ischemic stroke injury, we subjected both DLDH deficient mouse and DLDH transgenic mouse to stroke surgery followed by measurement of brain infarction volume. Results indicate that while mouse deficient in DLDH had exacerbated brain injury after stroke, mouse overexpressing human DLDH also showed increased brain injury after stroke. Therefore, the physiological significance of IHP-induced DLDH upregulation remains to be further investigated.


Subject(s)
Brain Ischemia/metabolism , Dihydrolipoamide Dehydrogenase/metabolism , Mitochondria/metabolism , Animals , Brain Ischemia/pathology , Cell Hypoxia , Dihydrolipoamide Dehydrogenase/genetics , Disease Models, Animal , Electron Transport Chain Complex Proteins/metabolism , Humans , Ischemic Preconditioning , Mice, Transgenic , Rats , Up-Regulation
10.
Cerebellum ; 14(4): 421-37, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25195804

ABSTRACT

Cerebellar disorders trigger the symptoms of movement problems, imbalance, incoordination, and frequent fall. Cerebellar disorders are shown in various CNS illnesses including a drinking disorder called alcoholism. Alcoholism is manifested as an inability to control drinking in spite of adverse consequences. Human and animal studies have shown that cerebellar symptoms persist even after complete abstinence from drinking. In particular, the abrupt termination (ethanol withdrawal) of long-term excessive ethanol consumption has shown to provoke a variety of neuronal and mitochondrial damage to the cerebellum. Upon ethanol withdrawal, excitatory neurotransmitter molecules such as glutamate are overly released in brain areas including cerebellum. This is particularly relevant to the cerebellar neuronal network as glutamate signals are projected to Purkinje neurons through granular cells that are the most populated neuronal type in CNS. This excitatory neuronal signal may be elevated by ethanol withdrawal stress, which promotes an increase in intracellular Ca(2+) level and a decrease in a Ca(2+)-binding protein, both of which result in the excessive entry of Ca(2+) to the mitochondria. Subsequently, mitochondria undergo a prolonged opening of mitochondrial permeability transition pore and the overproduction of harmful free radicals, impeding adenosine triphosphate (ATP)-generating function. This in turn provokes the leakage of mitochondrial molecule cytochrome c to the cytosol, which triggers a cascade of adverse cytosol reactions. Upstream to this pathway, cerebellum under the condition of ethanol withdrawal has shown aberrant gene modifications through altered DNA methylation, histone acetylation, or microRNA expression. Interplay between these events and molecules may result in functional damage to cerebellar mitochondria and consequent neuronal degeneration, thereby contributing to motoric deficit. Mitochondria-targeting research may help develop a powerful new therapy to manage cerebellar disorders associated with hyperexcitatory CNS disorders like ethanol withdrawal.


Subject(s)
Cerebellum/pathology , Cerebellum/ultrastructure , Mitochondria/pathology , Substance Withdrawal Syndrome/pathology , Alcoholism/complications , Animals , Humans , Mitochondria/ultrastructure
11.
J Pharmacol Exp Ther ; 352(2): 258-66, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406171

ABSTRACT

The acetylation of histone proteins in the core of DNA regulates gene expression, including those affecting mitochondria. Both histone acetylation and mitochondrial deficit have been implicated in neuronal damage associated with drinking problems. Many alcoholics will repeat unsuccessful attempts at abstaining, developing a pattern of repeated drinking and withdrawal. We investigated whether aberrant histone acetylation contributes to mitochondrial and cellular damage induced by repeated ethanol withdrawal (EW). We also investigated whether this effect of histone acetylation involves let-7f, a small noncoding RNA (microRNA). Male rats received two cycles of an ethanol/control diet (7.5%, 4 weeks) and withdrawal. Their prefrontal cortex was collected to measure the mitochondrial respiration and histone acetylation using extracellular flux (XF) real-time respirometry and gold immunostaining, respectively. Separately, HT22 (mouse hippocampal) cells received two cycles of ethanol exposure (100 mM, 20 hours) and withdrawal. Trichostatin A (TSA) as a histone acetylation promoter and let-7f antagomir were applied during withdrawal. The mitochondrial respiration, let-7f level, and cell viability were assessed using XF respirometry, quantitative polymerase chain reaction, TaqMan let-7f primers, and a calcein-acetoxymethyl assay, respectively. Repeated ethanol withdrawn rats showed a more than 2-fold increase in histone acetylation, accompanied by mitochondrial respiratory suppression. EW-induced mitochondrial respiratory suppression was exacerbated by TSA treatment in a manner that was attenuated by let-7f antagomir cotreatment. TSA treatment did not alter the increasing effect of EW on the let-7f level but dramatically exacerbated the cell death induced by EW. These data suggest that the multiple episodes of withdrawal from chronic ethanol impede mitochondrial and cellular integrity through upregulating histone acetylation, independent of or additively with let-7f.


Subject(s)
Alcoholism/metabolism , Histones/metabolism , Mitochondria/metabolism , Oxygen Consumption/physiology , Prefrontal Cortex/metabolism , Substance Withdrawal Syndrome/metabolism , Acetylation , Alcoholism/pathology , Animals , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Ethanol/pharmacology , Male , Mice , MicroRNAs/metabolism , Mitochondria/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/pathology , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/pathology
12.
J Appl Physiol (1985) ; 112(10): 1706-14, 2012 May.
Article in English | MEDLINE | ID: mdl-22403345

ABSTRACT

Intermittent hypoxia (IH) conditioning minimizes neurocognitive impairment and stabilizes brain mitochondrial integrity during ethanol withdrawal (EW) in rats, but the mitoprotective mechanism is unclear. We investigated whether IH conditioning protects a key mitochondrial enzyme, cytochrome c oxidase (COX), from EW stress by inhibiting mitochondrially directed apoptotic pathways involving cytochrome c, Bax, or phosphor-P38 (pP38). Male rats completed two cycles of a 4-wk ethanol diet (6.5%) and 3 wk of EW. An IH program consisting of 5-10 bouts of 5-8 min of mild hypoxia (9.5-10% inspired O(2)) and 4 min of reoxygenation for 20 consecutive days began 3 days before the first EW period. For some animals, vitamin E replaced IH conditioning to test the contributions of antioxidant mechanisms to IH's mitoprotection. During the second EW, cerebellar-related motor function was evaluated by measuring latency of fall from a rotating rod (Rotarod test). After the second EW, COX activity in cerebellar mitochondria was measured by spectrophotometry, and COX, cytochrome c, Bax, and pP38 content were analyzed by immunoblot. Mitochondrial protein oxidation was detected by measuring carbonyl contents and by immunochemistry. Earlier IH conditioning prevented motor impairment, COX inactivation, depletion of COX subunit 4, protein carbonylation, and P38 phosphorylation during EW. IH did not prevent cytochrome c depletion during EW, and Bax content was unaffected by EW ± IH. Vitamin E treatment recapitulated IH protection of COX, and P38 inhibition attenuated protein oxidation during EW. Thus IH protects COX and improves cerebellar function during EW by limiting P38-dependent oxidative damage.


Subject(s)
Alcohol Drinking/adverse effects , Cerebellum/enzymology , Electron Transport Complex IV/metabolism , Ethanol/adverse effects , Hypoxia/enzymology , Mitochondria/enzymology , Oxidative Stress , Substance Withdrawal Syndrome/prevention & control , Animals , Antioxidants/pharmacology , Apoptosis , Behavior, Animal , Blotting, Western , Cerebellum/drug effects , Cerebellum/pathology , Cerebellum/physiopathology , Cytochromes c/metabolism , Disease Models, Animal , Hypoxia/pathology , Hypoxia/physiopathology , Hypoxia/psychology , Imidazoles/pharmacology , Male , Mitochondria/drug effects , Mitochondria/pathology , Motor Activity , Oxidative Stress/drug effects , Phosphorylation , Protein Carbonylation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Reaction Time , Spectrophotometry , Substance Withdrawal Syndrome/enzymology , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/psychology , Time Factors , Vitamin E/pharmacology , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Neurobiol Aging ; 33(3): 618.e21-32, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21439684

ABSTRACT

We investigated whether abrupt ethanol withdrawal (EW) age-specifically inhibits a key mitochondrial enzyme, cytochrome c oxidase (COX), and whether estrogen mitigates this problem. We also tested whether this possible effect of EW involves a substrate (cytochrome c) deficiency that is associated with proapoptotic Bcl2-associated X protein (BAX) and mitochondrial membrane swelling. Ovariectomized young, middle age, and older rats, with or without 17ß-estradiol (E2) implantation, underwent repeated EW. Cerebelli were collected to measure COX activity and the mitochondrial membrane swelling using spectrophotometry and the mitochondrial levels of cytochrome c and BAX using an immunoblot method. The loss of COX activity and the mitochondrial membrane swelling occurred only in older rats under control diet conditions but occurred earlier, starting in the young rats under EW conditions. E2 treatment mitigated these EW effects. EW increased mitochondrial BAX particularly in middle age rats but did not alter cytochrome c. Collectively EW hastens but E2 delays the age-associated loss of COX activity. This EW effect is independent of cytochrome c but may involve the mitochondrial overload of BAX and membrane vulnerability.


Subject(s)
Aging/drug effects , Aging/metabolism , Central Nervous System Depressants/adverse effects , Electron Transport Complex IV/physiology , Ethanol/adverse effects , Substance Withdrawal Syndrome/metabolism , Animals , Electron Transport Complex IV/chemistry , Female , Protein Stability , Rats , Rats, Inbred F344 , Substance Withdrawal Syndrome/etiology , Substrate Specificity/genetics , Up-Regulation/genetics , bcl-2-Associated X Protein/metabolism
14.
Neurobiol Aging ; 32(12): 2266-78, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20122756

ABSTRACT

We investigated whether protein kinase p38 plays a role in the brain-aging changes associated with repeated ethanol withdrawal (EW). Ovariectomized young, middle-age and older rats, with or without 17ß-estradiol (E2) implantation, received a 90-day ethanol with repeated withdrawal. They were tested for active pP38 expression in cerebellar Purkinje neurons and whole-cerebellar lysates using immunohistochemistry and enzyme-linked immunosorbent assay, respectively. They were also tested for the Rotarod task to determine the behavioral manifestation of cerebellar neuronal stress and for reactive oxygen species (ROS) and mitochondrial protein carbonyls to determine oxidative mechanisms. Middle-age EW rats showed higher levels of pP38-positive Purkinje neurons/cerebellar lysates, which coincided with increased mitochondrial protein oxidation than other diet/age groups. Exacerbated motor deficit due to age-EW combination also began at the middle-age. In comparison, ROS contents peaked in older EW rats. E2 treatment mitigated each of the EW effects to a different extent. Collectively, pP38 may mediate the brain-aging changes associated with pro-oxidant EW at vulnerable ages and in vulnerable neurons in a manner protected by estrogen.


Subject(s)
Aging/metabolism , Cerebellum/enzymology , Ethanol/adverse effects , Oxidative Stress/physiology , Substance Withdrawal Syndrome/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Aging/drug effects , Animals , Cerebellum/drug effects , Enzyme Activation/physiology , Ethanol/administration & dosage , Female , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
15.
Molecules ; 15(7): 4984-5011, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20657404

ABSTRACT

Unmanaged sudden withdrawal from the excessive consumption of alcohol (ethanol) adversely alters neuronal integrity in vulnerable brain regions such as the cerebellum, hippocampus, or cortex. In addition to well known hyperexcitatory neurotransmissions, ethanol withdrawal (EW) provokes the intense generation of reactive oxygen species (ROS) and the activation of stress-responding protein kinases, which are the focus of this review article. EW also inflicts mitochondrial membranes/membrane potential, perturbs redox balance, and suppresses mitochondrial enzymes, all of which impair a fundamental function of mitochondria. Moreover, EW acts as an age-provoking stressor. The vulnerable age to EW stress is not necessarily the oldest age and varies depending upon the target molecule of EW. A major female sex steroid, 17beta-estradiol (E2), interferes with the EW-induced alteration of oxidative signaling pathways and thereby protects neurons, mitochondria, and behaviors. The current review attempts to provide integrated information at the levels of oxidative signaling mechanisms by which EW provokes brain injuries and E2 protects against it. Unmanaged sudden withdrawal from the excessive consumption of alcohol (ethanol) adversely alters neuronal integrity in vulnerable brain regions such as the cerebellum, hippocampus, or cortex. In addition to well known hyperexcitatory neurotransmissions, ethanol withdrawal (EW) provokes the intense generation of reactive oxygen species (ROS) and the activation of stress-responding protein kinases, which are the focus of this review article. EW also inflicts mitochondrial membranes/membrane potential, perturbs redox balance, and suppresses mitochondrial enzymes, all of which impair a fundamental function of mitochondria. Moreover, EW acts as an age-provoking stressor. The vulnerable age to EW stress is not necessarily the oldest age and varies depending upon the target molecule of EW. A major female sex steroid, 17beta-estradiol (E2), interferes with the EW-induced alteration of oxidative signaling pathways and thereby protects neurons, mitochondria, and behaviors. The current review attempts to provide integrated information at the levels of oxidative signaling mechanisms by which EW provokes brain injuries and E2 protects against it.


Subject(s)
Alcohol-Induced Disorders/metabolism , Brain Diseases, Metabolic/chemically induced , Substance Withdrawal Syndrome/metabolism , Animals , Ethanol/pharmacology , Humans , Oxidative Stress , Reactive Oxygen Species/metabolism , Substance Withdrawal Syndrome/physiopathology
16.
Int J Mol Sci ; 10(4): 1773-1787, 2009 Apr 20.
Article in English | MEDLINE | ID: mdl-19468338

ABSTRACT

Ethanol withdrawal is linked to elevated oxidative damage to neurons. Here we report our findings on the contribution of phenolic antioxidants (17beta-estradiol, p-octyl-phenol and 2,6-di-tert-butyl-4-methylphenol) to counterbalance sudden ethanol withdrawal-initiated oxidative events in hippocampus-derived cultured HT-22 cells. We showed that ethanol withdrawal for 4 h after 24-h ethanol treatment provoked greater levels of oxidative damage than the preceding ethanol exposure. Phenolic antioxidant treatment either during ethanol exposure or ethanol withdrawal only, however, dose-dependently reversed cellular oxidative damage, as demonstrated by the significantly enhanced cell viability, reduced malondialdehyde production and protein carbonylation, compared to untreated cells. Interestingly, the antioxidant treatment schedule had no significant impact on the observed neuroprotection. In addition, the efficacy of the three phenolic compounds was practically equipotent in protecting HT-22 cells in spite of predictions based on an in silico study and a cell free assay of lipid peroxidation. This finding implies that free-radical scavenging may not be the sole factor responsible for the observed neuroprotection and warrants further studies to establish, whether the HT-22 line is indeed a suitable model for in vitro screening of antioxidants against EW-related neuronal damage.


Subject(s)
Antioxidants/pharmacology , Hippocampus/cytology , Oxidative Stress/drug effects , Phenols/pharmacology , Animals , Antioxidants/chemistry , Cell Line , Cell Survival/drug effects , Ethanol/pharmacology , Hippocampus/metabolism , Lipid Peroxidation/drug effects , Mice , Neurons/metabolism , Phenols/chemistry , Protein Carbonylation/drug effects
17.
J Pharmacol Exp Ther ; 328(3): 692-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19050172

ABSTRACT

We have reported that the major endogenous estrogen, 17beta-estradiol (E2), protects against oxidative injury during ethanol withdrawal (EW) in a cultured hippocampal cell line (HT22). Here, we investigated whether the pro-oxidant nature of EW mediates opening of the mitochondrial membrane permeability transition pore (PTP) in a manner protected by E2. Excess PTP opening provokes mitochondrial membrane swelling (MMS) and the collapse of membrane potential (DeltaPsim). HT22 cells were collected at the end of ethanol exposure (100 mM) for 24 h or at 4 h of EW to assess MMS by monitoring absorbance decline at 540 nm and to assess DeltaPsim using flow cytometry. Protective effects of E2 on PTP were compared with an antioxidant butylated hydroxytoluene (BHT) and an E2 analog, ZYC26 [(3-hydroxy-2-adamantyl(1)-4-methyl-estra-1,3,5(10)-17-one], with higher antioxidant potency than E2. To assess cellular consequences of PTP opening, effects of a PTP inhibitor (cyclosporin A) on EW-induced cell death were assessed using the calcein assay. Major findings were that: 1) EW resulted in rapid MMS and DeltaPsim collapse; 2) cyclosporin A attenuated EW-induced cell death; and 3) E2 treatment restricted to the EW phase protected against the PTP opening more prominently than BHT and to a similar degree to ZYC26. These findings suggest that EW provokes PTP opening partly but not entirely through the pro-oxidant nature and that E2 counteracts EW-associated factors to protect against the PTP opening.


Subject(s)
Estrogens/pharmacology , Ethanol/pharmacology , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Membranes/physiology , Neurons/physiology , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Estrone/analogs & derivatives , Estrone/pharmacology , Female , Hippocampus/cytology , Hippocampus/physiology , Humans , Mice , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Permeability Transition Pore , Neurons/drug effects , Ovariectomy , Permeability
18.
J Appl Physiol (1985) ; 105(2): 510-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18499779

ABSTRACT

Intermittent hypoxia (IH) has been found to protect brain from ischemic injury. We investigated whether IH mitigates brain oxidative stress and behavioral deficits in rats subjected to ethanol intoxication and abrupt ethanol withdrawal (EW). The effects of IH on overt EW behavioral signs, superoxide generation, protein oxidation, and mitochondrial permeability transition pore (PTP) opening were examined. Male rats consumed dextrin or 6.5% (wt/vol) ethanol for 35 days. During the last 20 days, rats were treated with repetitive (5-8 per day), brief (5-10 min) cycles of hypoxia (9.5-10% inspired O2) separated by 4-min normoxia exposures. Cerebellum, cortex, and hippocampus were biopsied on day 35 of the diet or at 24 h of EW. Superoxide and protein carbonyl contents in tissue homogenates and absorbance decline at 540 nm in mitochondrial suspensions served as indicators of oxidative stress, protein oxidation, and PTP opening, respectively. Although IH altered neither ethanol consumption nor blood ethanol concentration, it sharply lowered the severity of EW signs including tremor, tail rigidity, and startle response. Compared with dextrin and ethanol per se, in the three brain regions, EW increased superoxide and protein carbonyl contents and accelerated PTP opening in a manner ameliorated by IH. Administration of antioxidant N-acetylcysteine throughout the IH program abrogated the reductions in EW signs and superoxide content, implicating IH-induced ROS as mediators of the salutary adaptations. We conclude that IH conditioning during chronic ethanol consumption attenuates oxidative damage to the brain and mitigates behavioral abnormalities during subsequent EW. IH-induced ROS may evoke this powerful protection.


Subject(s)
Behavior, Animal/drug effects , Brain Chemistry/physiology , Hypoxia/physiopathology , Ischemic Preconditioning , Oxidative Stress/physiology , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Central Nervous System Depressants/adverse effects , Central Nervous System Depressants/blood , Dextrins/pharmacology , Diet , Ethanol/adverse effects , Ethanol/blood , Male , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Membranes/drug effects , Mitochondrial Swelling/physiology , Oxidants/metabolism , Protein Carbonylation , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/physiopathology , Substance Withdrawal Syndrome/psychology , Superoxides/metabolism
19.
J Bioenerg Biomembr ; 40(1): 35-44, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18210193

ABSTRACT

We investigated whether ethanol withdrawal (EW) oxidizes mitochondrial proteins and provokes mitochondrial membrane swelling and whether estrogen deprivation contributes to this problem. Ovariectomized female rats with or without 17beta-estradiol (E2)-implantation received a control diet or a liquid ethanol diet (6.5%) for 5 weeks and were sacrificed during EW. Protein oxidation was assessed by measuring carbonyl contents and was visualized by immunochemistry. Mitochondrial membrane swelling as an indicator of mitochondrial membrane fragility was assessed by monitoring absorbance at 540 nm and was compared with that of male rats. Compared to the control diet group and ovariectomized rats with E2-implantation, ovariectomized rats without E2-implantation showed higher carbonylation of mitochondrial proteins and more rapid mitochondrial membrane swelling during EW. Such rapid mitochondrial membrane swelling was comparable to that of male rats undergoing EW. These findings demonstrate that EW provokes oxidative injury to mitochondrial membranes in a manner that is exacerbated by estrogen deprivation.


Subject(s)
Alcohol-Induced Disorders/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Ethanol , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Alcohol-Induced Disorders/pathology , Animals , Female , Mitochondria/pathology , Mitochondrial Membranes/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Sprague-Dawley , Time Factors
20.
Neurosci Lett ; 416(2): 160-4, 2007 Apr 12.
Article in English | MEDLINE | ID: mdl-17320290

ABSTRACT

Cytochrome c oxidase (COX) is a key mitochondrial enzyme that catalyzes electron transfer at the terminal stage of respiratory chain and is composed of multisubunits. We hypothesize that ethanol withdrawal (EW) impairs the activity of COX and estrogen deprivation exacerbates this problem. Five-month-old ovariectomized rats with or without 17beta-estradiol (E2) replacement received a control dextrin or a liquid ethanol diet (6.5%, 5 weeks). They were then sacrificed either during ethanol exposure or at 24h of EW (EW group). Mitochondria of the cerebellum and cortex were processed to measure the activities of total COX, COX subunit I, and IV. The effects of EW and E2 on the protein levels of these subunits were also assessed using an immunoblotting method. As compared to the control dextrin and ethanol exposure, EW decreased the activities of total COX, COX I, and COX IV. E2 treatment prevented the effects of EW on the activities of total COX and COX IV but not COX I. Neither EW nor E2 altered the protein levels of the subunits. These findings suggest that a counteracting relationship exists between the effects of EW and E2 on the activity of COX in a subunit specific manner.


Subject(s)
Central Nervous System Depressants/toxicity , Electron Transport Complex IV/drug effects , Electron Transport Complex IV/metabolism , Enzyme Activation/drug effects , Estradiol/pharmacology , Ethanol/toxicity , Animals , Brain/drug effects , Brain/metabolism , Female , Immunoblotting , Mitochondria/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Ovariectomy , Polymerase Chain Reaction , Rats , Substance Withdrawal Syndrome/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL