Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 199: 106570, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38885850

ABSTRACT

BACKGROUND: Hepatic lipoprotein receptor-related protein 1 (LRP-1) plays a central role in peripheral amyloid beta (Aß) clearance, but its importance in Alzheimer's disease (AD) pathology is understudied. Our previous work showed that intragastric alcohol feeding to C57BL/6 J mice reduced hepatic LRP-1 expression which correlated with significant AD-relevant brain changes. Herein, we examined the role of hepatic LRP-1 in AD pathogenesis in APP/PS1 AD mice using two approaches to modulate hepatic LRP-1, intragastric alcohol feeding to model chronic heavy drinking shown by us to reduce hepatic LRP-1, and hepato-specific LRP-1 silencing. METHODS: Eight-month-old male APP/PS1 mice were fed ethanol or control diet intragastrically for 5 weeks (n = 7-11/group). Brain and liver Aß were assessed using immunoassays. Three important mechanisms of brain amyloidosis were investigated: hepatic LRP-1 (major peripheral Aß regulator), blood-brain barrier (BBB) function (vascular Aß regulator), and microglia (major brain Aß regulator) using immunoassays. Spatial LRP-1 gene expression in the periportal versus pericentral hepatic regions was confirmed using NanoString GeoMx Digital Spatial Profiler. Further, hepatic LRP-1 was silenced by injecting LRP-1 microRNA delivered by the adeno-associated virus 8 (AAV8) and the hepato-specific thyroxine-binding globulin (TBG) promoter to 4-month-old male APP/PS1 mice (n = 6). Control male APP/PS1 mice received control AAV8 (n = 6). Spatial memory and locomotion were assessed 12 weeks after LRP-1 silencing using Y-maze and open-field test, respectively, and brain and liver Aß were measured. RESULTS: Alcohol feeding reduced plaque-associated microglia in APP/PS1 mice brains and increased aggregated Aß (p < 0.05) by ELISA and 6E10-positive Aß load by immunostaining (p < 0.05). Increased brain Aß corresponded with a significant downregulation of hepatic LRP-1 (p < 0.01) at the protein and transcript level, primarily in pericentral hepatocytes (zone 3) where alcohol-induced injury occurs. Hepato-specific LRP-1 silencing significantly increased brain Aß and locomotion hyperactivity (p < 0.05) in APP/PS1 mice. CONCLUSION: Chronic heavy alcohol intake reduced hepatic LRP-1 expression and increased brain Aß. The hepato-specific LRP-1 silencing similarly increased brain Aß which was associated with behavioral deficits in APP/PS1 mice. Collectively, our results suggest that hepatic LRP-1 is a key regulator of brain amyloidosis in alcohol-dependent AD.

2.
Front Physiol ; 13: 930402, 2022.
Article in English | MEDLINE | ID: mdl-36187787

ABSTRACT

Heavy alcohol consumption is a known risk factor for various forms of dementia and the development of Alzheimer's disease (AD). In this work, we investigated how intragastric alcohol feeding may alter the liver-to-brain axis to induce and/or promote AD pathology. Four weeks of intragastric alcohol feeding to mice, which causes significant fatty liver (steatosis) and liver injury, caused no changes in AD pathology markers in the brain [amyloid precursor protein (APP), presenilin], except for a decrease in microglial cell number in the cortex of the brain. Interestingly, the decline in microglial numbers correlated with serum alanine transaminase (ALT) levels, suggesting a potential link between liver injury and microglial loss in the brain. Intragastric alcohol feeding significantly affected two hepatic proteins important in amyloid-beta (Aß) processing by the liver: 1) alcohol feeding downregulated lipoprotein receptor-related protein 1 (LRP1, ∼46%), the major receptor in the liver that removes Aß from blood and peripheral organs, and 2) alcohol significantly upregulated APP (∼2-fold), a potentially important source of Aß in the periphery and brain. The decrease in hepatic LRP1 and increase in hepatic APP likely switches the liver from being a remover or low producer of Aß to an important source of Aß in the periphery, which can impact the brain. The downregulation of LRP1 and upregulation of APP in the liver was observed in the first week of intragastric alcohol feeding, and also occurred in other alcohol feeding models (NIAAA binge alcohol model and intragastric alcohol feeding to rats). Modulation of hepatic LRP1 and APP does not seem alcohol-specific, as ob/ob mice with significant steatosis also had declines in LRP1 and increases in APP expression in the liver. These findings suggest that liver steatosis rather than alcohol-induced liver injury is likely responsible for regulation of hepatic LRP1 and APP. Both obesity and alcohol intake have been linked to AD and our data suggests that liver steatosis associated with these two conditions modulates hepatic LRP1 and APP to disrupt Aß processing by the liver to promote AD.

3.
FEBS Open Bio ; 12(1): 203-210, 2022 01.
Article in English | MEDLINE | ID: mdl-34738322

ABSTRACT

Tunneling nanotubes (TNTs) are F-actin-based open-ended tubular extensions that form following stresses, such as nutritional deprivation and oxidative stress. The chemotherapy agent 5-fluorouracil (5-FU) represents a significant stressor to cancer cells and induces thymidine deficiency, a state similar to nutritional deprivation. However, the ability of 5-FU to induce TNT formation in cancer cells and potentially enhance survival has not been explored. In this study, we examined whether 5-FU can induce TNT formation in MCF-7 breast cancer cells. Cytotoxic doses of 5-FU (150-350 µm) were observed to significantly induce TNT formation beginning at 24 h after exposure. TNTs formed following 5-FU treatment probably originated as extensions of gap junctions as MCF-7 cells detach from cell clusters. TNTs act as conduits for exchange of cellular components and we observed mitochondrial exchange through TNTs following 5-FU treatment. 5-FU-induced TNT formation was inhibited by over 80% following treatment with the F-actin-depolymerizing agent, cytochalasin B (cytoB). The inhibition of TNTs by cytoB corresponded with increased 5-FU-induced cytotoxicity by 30-62% starting at 48 h, suggesting TNT formation aides in MCF-7 cell survival against 5-FU. Two other widely used chemotherapy agents, docetaxel and doxorubicin induced TNT formation at much lower levels than 5-FU. Our work suggests that the therapeutic targeting of TNTs may increase 5-FU chemotherapy efficacy and decrease drug resistance in cancer cells, and these findings merits further investigation.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Cell Communication , Cell Membrane Structures , Female , Fluorouracil/pharmacology , Humans , MCF-7 Cells , Nanotubes
4.
J Clin Transl Res ; 7(3): 289-296, 2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34179543

ABSTRACT

BACKGROUND AND AIMS: Physical exercise may help combat disease and elicits a possible "protective" anti-inflammatory effect on the body. Inflammatory cytokines, C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-α (TNFα), along with transcription factor, nuclear factor-kappa B (NFκB) in young (n=16, 21.1±2.1 years) individuals were examined in a cross-sectional descriptive study, to assess the effects of chronic stimulation on their expression and relationship with health parameters. METHODS: Fasting venous whole blood and lipid levels along with body composition measurements were obtained from young, healthy, endurance-trained NCAA Division III student-athletes and untrained individuals. Assays (ELISA) were conducted to analyze fasting plasma (CRP, IL-6, and TNFα) and isolated lymphocyte NF-κB activation (lymphocytes were isolated from whole blood samples through differential centrifugation and Ficoll-Paque). A Spearman's rank order correlation coefficient was used for associations between variables and a regression analysis was performed to determine which measurement accounted for the inflammation in this young and apparently healthy population. RESULTS: While the inflammatory markers were not associated with each other, CRP levels were associated with body composition and following regression analyses, body fat percentage (P>0.05) was a significant factor for elevated CRP. CONCLUSIONS: Chronic physical exercise eliciting lower body fat percentages in young adults may have a positive protective impact through anti-inflammatory status, minimizing disease risk in a young population. RELEVANCE FOR PATIENTS: Chronic physically active young adult patients may exhibit less inflammation and lower body fat levels which may decrease their risk for chronic disease.

5.
Biochem Biophys Rep ; 24: 100824, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33204855

ABSTRACT

Mitochondria oscillate along a morphological continuum from fragmented individual units to hyperfused tubular networks. Their position at the junction of catabolic and anabolic metabolism couples this morphological plasticity, called mitochondrial dynamics, to larger cellular metabolic programs, which in turn implicate mitochondria in a number of disease states. In many cancers, fragmented mitochondria engage the cell with the biosynthetic capacity of aerobic glycolysis in service of proliferation and progression. Chemo-resistant cancers, however, favor remodeling dynamics that yield fused mitochondrial assemblies utilizing oxidative phosphorylation (OXPHOS) through the electron transport chain (ETC). In this study, expression of Mitofusin-2 (MFN-2), a GTPase protein mediator of mitochondrial fusion, was found to closely correlate to Jurkat leukemia cell survival post doxorubicin (DxR) assault. Moreover, this was accompanied by dramatically increased expression of OXPHOS respiratory complexes and ATP Synthase, as well as a commensurate escalation of state III respiration and respiratory control ratio (RCR). Importantly, CRISPR knockout of MFN-2 resulted in a considerable decrease of doxorubicin (DxR) median lethal dose compared to a treated wildtype control, suggesting an important role of mitochondrial fusion in chemotherapy sensitivity and acute resistance.

6.
Int J Pharm ; 577: 118995, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-31935471

ABSTRACT

Drug repurposing is on the rise as an atypical strategy for discovery of new molecules, involving use of pre-existing molecules for a different therapeutic application than the approved indication. Using this strategy, the current study aims to leverage effects of quinacrine (QA), a well-known anti-malarial drug, for treatment of non-small cell lung cancer (NSCLC). For respiratory diseases, designing a QA loaded inhalable delivery system has multiple advantages over invasive delivery. QA-loaded nanoparticles (NPs) were thus prepared using polyethyleneimine (PEI) as a cationic stabilizer. While the use of PEI provided cationic charge on the particles, it also mediated a burst release of QA and demonstrated potential particle toxicity. These concerns were circumvented by coating nanoparticles with bovine serum albumin (BSA), which retained the cationic charge, reduced NP toxicity and modulated QA release. Prepared nanoparticles were characterized for physicochemical properties along with their aerosolization potential. Therapeutic efficacy of the formulations was tested in different NSCLC cells. Mechanism of higher anti-proliferation was evaluated by studying cell cycle profile, apoptosis and molecular markers involved in the progression of lung cancer. BSA coated QA nanoparticles demonstrated good aerosolization potential with a mass median aerodynamic diameter of significantly less than 5 µm. Nanoparticles also demonstrated improved therapeutic efficacy against NSCLC cells in terms of low IC50 values, cell cycle arrest at G2/M phase and autophagy inhibition leading to increased apoptosis. BSA coated QA NPs also demonstrated enhanced therapeutic efficacy in a 3D cell culture model. The present study thus lays solid groundwork for pre-clinical and eventual clinical studies as a standalone therapy and in combination with existing chemotherapeutics.


Subject(s)
Drug Compounding/methods , Drug Delivery Systems/methods , Drug Repositioning/methods , Nanoparticles/chemistry , Quinacrine/chemistry , Serum Albumin, Bovine/chemistry , Administration, Inhalation , Aerosols/chemistry , Aerosols/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Liberation , Humans , Lung Neoplasms/drug therapy , Nanoparticles/administration & dosage , Particle Size , Polyethyleneimine/chemistry , Quinacrine/administration & dosage , Quinacrine/pharmacology
7.
Int J Biol Macromol ; 122: 338-347, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30401652

ABSTRACT

This study was aimed at developing a nanoparticle strategy to overcome acquired resistance against erlotinib in non-small cell lung cancer (NSCLC). To load erlotinib on biodegradable PLGA nanoparticles, erlotinib-cyclodextrin (Erlo-CD) complex was prepared using ß-cyclodextrin sulfobutyl ether, which was in turn loaded in the core of PLGA nanoparticles using multiple emulsion solvent evaporation. Nanoparticles were characterized for size distribution, entrapment and loading efficiency, in-vitro release, and therapeutic efficacy against different lung cancer cells. Effect of formulation on cell cycle, apoptosis, and other markers was evaluated using flow cytometry and western blotting studies. The efficacy of optimized nanoformulation was evaluated using a clinically relevant in-vitro 3D-spheroid model. Results showed that Erlo-CD loaded nanoparticles (210 ±â€¯8 nm in size) demonstrated 3-fold higher entrapment (61.5 ±â€¯3.2% vs 21.9 ±â€¯3.7% of plain erlotinib loaded nanoparticles) with ~5% loading efficiency and sustained release characteristics. Developed nanoparticles demonstrated significantly improved therapeutic efficacy against NSCLC cells in terms of low IC50 values and suppressed colony forming ability of cancer cells, increased apoptosis, and autophagy inhibition. Interestingly, 3D spheroid study demonstrated better anticancer activity of Erlo-CD nanoparticles compared to plain erlotinib. Present study has shown a premise to improve therapeutic efficacy against erlotinib-resistant lung cancer using modified nanoErlo formulations.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cyclodextrins/chemistry , Drug Carriers/chemistry , Erlotinib Hydrochloride/pharmacology , Lung Neoplasms/pathology , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Liberation , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/therapeutic use , Humans , Lung Neoplasms/drug therapy , Particle Size
8.
FEBS Lett ; 592(6): 916-927, 2018 03.
Article in English | MEDLINE | ID: mdl-29430655

ABSTRACT

We investigated if obesity/steatosis promotes mitochondrial remodeling in the liver of ob/ob mice (an obesity model). Liver mitochondria from ob/ob mice (21 weeks with significant steatosis) had ~ 2-fold increases in state III respiration compared with control (C57BL/6J, C57BL/6NJ) for all respiratory substrates examined (glutamate/malate, succinate, octanoate, and glycerol 3-phosphate). A corresponding 2-fold increase in the expression of respiratory complexes (I, IV, and V) and other respiratory proteins (glycerol phosphate dehydrogenase-2 and medium-chain acyl-coenzyme A dehydrogenase) occur in liver mitochondria of mature ob/ob mice. Conversely, respiration in liver mitochondria from young ob/ob mice (6 weeks) does not differ from control with any respiratory substrates examined. Overall, mitochondrial remodeling that enhances respiration increases with obesity/steatosis in the liver of ob/ob mice.


Subject(s)
Fatty Liver/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Obesity/metabolism , Oxygen Consumption , Animals , Electron Transport , Electron Transport Chain Complex Proteins/biosynthesis , Fatty Liver/pathology , Liver/pathology , Mice , Mice, Obese , Mitochondria, Liver/pathology , Mitochondrial Proteins/biosynthesis , Obesity/pathology , Species Specificity
9.
J Med Entomol ; 55(2): 477-480, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29228245

ABSTRACT

Wyeomyia (Wyeomyia) mitchellii (Theobald) (Diptera: Culicidae) was recovered for the first time on Guam, United States of America, in 2017. Larval specimens were collected from water-filled axils of bromeliads during a larval survey carried out in a residential neighborhood of the Chalan Pago/Ordot area. Native to the New World, Wy. mitchellii has likely made its way to the Pacific Islands through the possibly illegal import of ornamental bromeliads. While this mosquito does not represent a significant threat to public health, this finding highlights the vulnerability of the Pacific Islands to the introduction of exotic species, including mosquito species that may increase public health risks.


Subject(s)
Animal Distribution , Culicidae/physiology , Animals , Culicidae/growth & development , Guam , Introduced Species , Larva/growth & development , Larva/physiology
10.
Biol Open ; 5(8): 1093-101, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27402966

ABSTRACT

A fundamental question in biology is how an organism's morphology and physiology are shaped by its environment. Here, we evaluate the effects of a hypersaline environment on the morphology and physiology of a population of livebearing fish in the genus Limia (Poeciliidae). We sampled from two populations of Limia perugiae (one freshwater and one hypersaline) in the southwest Dominican Republic. We evaluated relative abundance of osmoregulatory proteins using western blot analyses and used a geometric morphometric approach to evaluate fine-scale changes to size and shape. Our data show that gill tissue isolated from hypersaline fish contained approximately two and a half times higher expression of Na(+)/K(+) ATPase proteins. We also show evidence for mitochondrial changes within the gills, with eight times more complex I and four times higher expression of ATP synthase within the gill tissue from the hypersaline population. The energetic consequences to Limia living in saline and hypersaline environments may be a driver for phenotypic diversity, reducing the overall body size and changing the relative size and shape of the head, as well as impeding the growth of secondary sex features among the males.

11.
Chem Res Toxicol ; 27(5): 794-804, 2014 May 19.
Article in English | MEDLINE | ID: mdl-24716714

ABSTRACT

S-Nitrosylation is a reversible post-translational modification on cysteinyl thiols that can modulate the function of redox-sensitive proteins. The S-nitrosylation of mitochondrial proteins has been shown to regulate various mitochondrial activities involved in energy-transducing systems and mitochondrion-driven apoptosis. In isolated rat brain mitochondria, we demonstrate that mitochondrial protein S-nitrosylation is regulated by respiratory substrates (glutamate/malate) through a thiol-dependent pathway. Mitochondrial proteins become susceptible to S-nitrosoglutathione (GSNO)-induced S-nitrosylation in mitochondria with an oxidized environment (low glutathione (GSH), NADH, and NADPH, and high GSSG, NAD(+), and NADP(+)) caused by isolation of mitochondria using a discontinuous Percoll gradient. Activation of mitochondrial respiration by respiratory substrates leads to increased NAD(P)H and GSH levels, which in turn reduces mitochondrial S-nitrosylated proteins. 1-Chloro-2,4-dinitrobenzene (CDNB), which depletes mitochondrial GSH and inhibits the thioredoxin-thioredoxin reductase system, prevented the denitrosylation of mitochondrial proteins caused by respiratory substrate treatment. Using biotin-switch coupled with LC-MS/MS, several mitochondrial proteins were identified as targets of S-nitrosylation including adenine nucleotide translocase (ANT) and voltage-dependent anion channel (VDAC), important components of the mitochondria permeability transition pore (MPTP), as well as ATP synthase. The S-nitrosylation of ATP synthase by GSNO was found to inhibit its activity. These findings emphasize the importance of respiratory substrates in regulating S-nitrosylation through a thiol-dependent (GSH and/or thioredoxin) pathway, with implications for mitochondrial bioenergetics and mitochondrion-driven apoptosis.


Subject(s)
Mitochondrial Proteins/metabolism , S-Nitrosoglutathione/metabolism , Animals , Cell Respiration , Glutamic Acid/metabolism , Malates/metabolism , Male , Oxidation-Reduction , Rats , Rats, Wistar , Signal Transduction , Sulfhydryl Compounds/metabolism
12.
J Biol Chem ; 285(51): 39646-54, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20937819

ABSTRACT

Brain and liver mitochondria isolated by a discontinuous Percoll gradient show an oxidized redox environment, which is reflected by low GSH levels and high GSSG levels and significant glutathionylation of mitochondrial proteins as well as by low NAD(P)H/NAD(P) values. The redox potential of brain mitochondria isolated by a discontinuous Percoll gradient method was calculated to be -171 mV based on GSH and GSSG concentrations. Immunoblotting and LC/MS/MS analysis revealed that succinyl-CoA transferase and ATP synthase (F(1) complex, α-subunit) were extensively glutathionylated; S-glutathionylation of these proteins resulted in a substantial decrease of activity. Supplementation of mitochondria with complex I or complex II respiratory substrates (malate/glutamate or succinate, respectively) increased NADH and NADPH levels, resulting in the restoration of GSH levels through reduction of GSSG and deglutathionylation of mitochondrial proteins. Under these conditions, the redox potential of brain mitochondria was calculated to be -291 mV. Supplementation of mitochondria with respiratory substrates prevented GSSG formation and, consequently, ATP synthase glutathionylation in response to H(2)O(2) challenges. ATP synthase appears to be the major mitochondrial protein that becomes glutathionylated under oxidative stress conditions. Glutathionylation of mitochondrial proteins is a major consequence of oxidative stress, and respiratory substrates are key regulators of mitochondrial redox status (as reflected by thiol/disulfide exchange) by maintaining mitochondrial NADPH levels.


Subject(s)
Glutathione Disulfide/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , NADP/metabolism , Oxidative Stress/physiology , Protein Processing, Post-Translational/physiology , Acyltransferases/metabolism , Animals , Brain/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Protein Processing, Post-Translational/drug effects , Proton-Translocating ATPases/metabolism , Rats
13.
Methods Enzymol ; 473: 137-47, 2010.
Article in English | MEDLINE | ID: mdl-20513475

ABSTRACT

GSNO is an important intermediate in nitric oxide metabolism and mediates many ()NO-mediated signaling pathways through the post-translational modification of redox-sensitive proteins. The detection of GSNO in biological samples has been hampered by a lack of sensitive and simple assays. In this work, we describe the utilization of HPLC with electrochemical detection for the identification and quantification of GSNO in biological samples. GSNO requires a high potential (>700 mV) for its electrochemical detection, similar to that of GSSG. A simple isocratic HPLC system can be used to separate and simultaneously detect GSH, GSSG, and GSNO electrochemically. This HPLC system can be utilized to measure the redox profile of biological samples and applied for the measurement of GSNO reductase activity in cells. Proper sample preparation is essential in GSNO measurements, because artifactual formation of GSNO occurs in acidic conditions due to the reaction between GSH and nitrite. Treatment of samples with ammonium sulfamate or N-ethylmaleimide (NEM) can prevent the artifactual formation of GSNO and accurately detect GSNO in biological samples. Overall, the HPLC with electrochemical detection is a powerful tool to measure redox status in cells and tissues.


Subject(s)
Electrochemical Techniques/methods , Glutathione Disulfide/analysis , Glutathione/analysis , S-Nitrosoglutathione/analysis , Aldehyde Oxidoreductases/analysis , Aldehyde Oxidoreductases/metabolism , Animals , Chromatography, High Pressure Liquid/methods , Glutathione/chemistry , Glutathione Disulfide/chemistry , Humans , S-Nitrosoglutathione/chemistry
14.
Biochem J ; 428(1): 85-93, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20210787

ABSTRACT

Excessive generation of nitric oxide radical (NO*) in neuroinflammation, excitotoxicity and during age-related neurodegenerative disorders entails the localized and concerted increase in nitric oxide synthase(s) expression in glial cells and neurons. The aim of the present study was to assess the biological significance of the impact of NO* on the cell's thiol status with emphasis on S-glutathionylation of targeted proteins. Exposure of primary cortical neurons or astrocytes to increasing flow rates of NO* (0.061-0.25 microM/s) resulted in the following. (i) A decrease in GSH (glutathione) in neurons accompanied by formation of GSNO (S-nitrosoglutathione) and GSSG (glutathione disulfide); neurons were far more sensitive to NO* exposure than astrocytes. (ii) A dose-dependent oxidation of the cellular redox status: the neuron's redox potential increased approximately 42 mV and that of astrocytes approximately 23 mV. A good correlation was observed between cell viability and the cellular redox potential. The higher susceptibility of neurons to NO* can be partly explained by a reduced capacity to recover GSH through lower activities of GSNO and GSSG reductases. (iii) S-glutathionylation of a small subset of proteins, among them GAPDH (glyceraldehyde-3-phosphate dehydrogenase), the S-glutathionylation of which resulted in inhibition of enzyme activity. The quantitative analyses of changes in the cell's thiol potential upon NO* exposure and their consequences for S-glutathionylation are discussed in terms of the distinct redox environment of astrocytes and neurons.


Subject(s)
Glutathione/metabolism , Neurons/metabolism , Nitric Oxide/metabolism , Animals , Female , Glutathione Disulfide/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Rats , Rats, Inbred F344 , S-Nitrosoglutathione/metabolism
15.
Adv Drug Deliv Rev ; 61(14): 1283-98, 2009 Nov 30.
Article in English | MEDLINE | ID: mdl-19716388

ABSTRACT

Decrease in mitochondrial energy-transducing capacity is a feature of the aging process that accompanies redox alterations, such as increased generation of mitochondrial oxidants, altered GSH status, and increased protein oxidation. The decrease in mitochondrial energy-transducing capacity and altered redox status should be viewed as a concerted process that embodies the mitochondrial energy-redox axis and is linked through various mechanisms including: (a) an inter-convertible reducing equivalents pool (i.e., NAD(P)(+)/NAD(P)H) and (b) redox-mediated protein post-translational modifications involved in energy metabolism. The energy-redox axis provides the rationale for therapeutic approaches targeted to each or both component(s) of the axis that effectively preserves or improve mitochondrial function and that have implications for aging and age-related neurodegenerative disorders.


Subject(s)
Aging/metabolism , Energy Metabolism/physiology , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Oxidation-Reduction/drug effects , Drug Delivery Systems/methods , Energy Metabolism/drug effects , Humans , Models, Biological , Neurons/metabolism
16.
Biochemistry ; 44(36): 11986-96, 2005 Sep 13.
Article in English | MEDLINE | ID: mdl-16142896

ABSTRACT

Aconitases are iron-sulfur cluster-containing proteins present both in mitochondria and cytosol of cells; the cubane iron-sulfur (Fe-S) cluster in the active site is essential for catalytic activity, but it also renders aconitase highly vulnerable to reactive oxygen and nitrogen species. This study examined the sites and mechanisms of aconitase inactivation by peroxynitrite (ONOO-), a strong oxidant and nitrating agent readily formed from superoxide anion and nitric oxide generated by mitochondria. ONOO- inactivated aconitase in a dose-dependent manner (half-maximal inhibition was observed with approximately 3 microM ONOO-). Low levels of ONOO- caused the conversion of the Fe-S cluster from the [4Fe-4S]2+ form to the inactive [3Fe-4S]1+ form with the loss of labile iron, as confirmed by low-temperature EPR analysis. In the presence of the substrate, citrate, 66-fold higher concentrations of ONOO- were required for half-maximal inhibition. The protective effects of citrate corresponded to its binding to the active site. The inactivation of aconitase in the presence of citrate was due to ONOO--mediated cysteine thiol loss and tyrosine nitration in the enzyme as shown by Western blot analyses. LC/MS/MS analyses revealed that ONOO- treatment to aconitase resulted in nitration of tyrosines 151 and 472 and oxidation to sulfonic acid of cysteines 126 and 385. The latter is one of the three cysteine residues in aconitase that binds to the Fe-S cluster. All other modified tyrosine and cysteine residues were adjacent to the binding site, thus suggesting that these modifications caused conformational changes leading to active-site disruption. Aconitase cysteine thiol modifications other than oxidation to sulfonic acid, such as S-glutathionylation, also decreased aconitase activity, thus indicating that glutathionylation may be an important means of modulating aconitase activity under oxidative and nitrative stress. Taken together, these results demonstrate that the Fe-S cluster in the active site, cysteine 385 bound to the Fe-S cluster, and tyrosine and cysteine residues in the vicinity of the active site are important targets of oxidative and/or nitrative attack, which is selectively controlled by the mitochondrial matrix citrate levels. The mechanisms inherent in aconitase inactivation by ONOO- are discussed in terms of the mitochondrial matrix metabolic and thiol redox state.


Subject(s)
Aconitate Hydratase/chemistry , Aconitate Hydratase/metabolism , Citric Acid/pharmacology , Glutathione/pharmacology , Peroxynitrous Acid/pharmacology , Swine , Aconitate Hydratase/antagonists & inhibitors , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Cysteine/metabolism , Enzyme Activation/drug effects , Mass Spectrometry , Molecular Sequence Data , Myocardium/enzymology , Peroxynitrous Acid/chemistry , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...