27.07.2013 Views

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

manganism. Recent work from our laboratory identified activated microglia expressing<br />

inducible nitric oxide (iNOS) in the basal ganglia <strong>of</strong> mice exposed to manganese<br />

(Mn) before the appearance <strong>of</strong> activated astrocytes; however, the role <strong>of</strong> activated<br />

microglia in manganism is poorly understood. In this study we postulated<br />

that Mn directly activates microglia, resulting in increased expression <strong>of</strong> tumor<br />

necrosis factor α (TNF- α), interleukin 1-beta (IL-1β) and iNOS that ultimately<br />

lead to increased astrocyte activation. Primary microglia and astrocytes were isolated<br />

from C57Bl/6 mice in mixed cultures then purified for experiments.<br />

Immun<strong>of</strong>luorescence staining for microglia and astrocytes was performed using<br />

ionized calcium binding adaptor protein-1 (IBA-1) and glial fibrillary acidic protein<br />

(GFAP), respectively. This method yielded culture purities <strong>of</strong> 97% for microglia<br />

and 99% for astrocytes. Treatment <strong>of</strong> microglia with Mn induced a dose-dependent<br />

expression <strong>of</strong> TNFα, iNOS mRNA and protein. Furthermore, a<br />

quantitative PCR array revealed increased expression <strong>of</strong> proinflammatory mediators<br />

in microglia when treated with Mn. Treatment <strong>of</strong> astrocytes with conditioned<br />

media from Mn-treated microglia or via co-culture with microglia caused an activated<br />

phenotype characterized by increased iNOS, TNFα, and IL-1β expression.<br />

Collectively, these data indicate that Mn activates microglia in a dose-dependent<br />

manner resulting in increased production <strong>of</strong> proinflammatory mediators that enhance<br />

activation <strong>of</strong> astrocytes, suggesting a complex pattern <strong>of</strong> glial-glial interactions<br />

underlying a neuroinflammatory phenotype in this model.<br />

2130 NOVEL NADPH OXIDASE MODULATOR PROTECTS<br />

AGAINST NEUROINFLAMMATION ASSOCIATED<br />

WITH MANGANESE NEUROTOXICITY.<br />

C. Hogan1 , R. Gordon1 , A. Kanthasamy1 , V. Anantharam1 , B. Kalyanaraman2 and A. G. Kanthasamy1 . 1Biomedical Sciences, Iowa Center for Advanced<br />

Neurotoxicology, Iowa State University, Ames, IA and 2Biophys, Medical College <strong>of</strong><br />

Wisconsin, Milwaukee, WI.<br />

Sustained neuroinflammation has become increasingly evident as a causative factor<br />

in the progression <strong>of</strong> many neurodegenerative diseases including Parkinson’s disease<br />

(PD). Exposure to environmental neurotoxicants such as metals and pesticides are<br />

considered as potential risk factors in the pathogenesis <strong>of</strong> PD. Microglia are a large<br />

part <strong>of</strong> the innate immune system <strong>of</strong> the CNS and are capable <strong>of</strong> producing rapid<br />

neuroinflammatory responses when confronted with pathogens or neurotoxic insults.<br />

Metal exposure has been shown to trigger activation <strong>of</strong> both microglia and astroglia<br />

in order to counter neurotoxic insults. Recently, studies have shown that<br />

manganese (Mn) exposure by itself or in combination with other inflammatory<br />

stimuli can produce neuroinflammatory responses in the nigrostriatal dopaminergic<br />

system. We and others have shown that Mn can augment lipopolysaccharide<br />

(LPS)-induced cytokine production, iNOS activation, and ROS generation in microglial<br />

cells. In the present study, we sought to investigate the anti-inflammatory<br />

potential <strong>of</strong> a novel NADPH oxidase inhibitor MIA-4025 against microglial activation<br />

in response to Mn/LPS-induced inflammatory events. MIA-4025 treatment<br />

significantly blocked Mn/LPS-induced iNOS activation and ROS generation in<br />

both primary microglia and BV2 microglia cell models. Measurement <strong>of</strong> cytokine<br />

release in the supernatant <strong>of</strong> MIA-4025 treated BV2 cell lines and primary microglia<br />

cells revealed that the compound effectively suppressed the release <strong>of</strong> TNFα,<br />

IL-1β, IL-6, IL-10 and IL-12. Expression <strong>of</strong> phox proteins, components <strong>of</strong> the<br />

NADPH oxidase complex and a main contributor to ROS production, was also decreased<br />

with MIA-4025 treatment. Collectively, these results suggest that our translational<br />

approach with MIA-4025 can suppress neuroinflammatory events associated<br />

with neurotoxic metal exposure. (Supported by NIH grants ES10586,<br />

NS065167 & NS039958.)<br />

2131 PROTEIN KINASE C IS INVOLVED IN MANGANESE-<br />

MEDIATED DISRUPTION OF GLUTAMINE TURNOVER<br />

IN ASTROCYTES.<br />

M. Sidoryk-Wegrzynowicz1 , E. Lee2 and M. Aschner1 . 1Department <strong>of</strong> Pediatrics,<br />

Vanderbilt University Medical Center, Nashville, TN and 2Department <strong>of</strong> Physiology,<br />

Meharry Medical College, Nashville, TN.<br />

Manganese (Mn) is an essential trace element for normal human development and<br />

is required for proper functioning <strong>of</strong> a variety <strong>of</strong> physiological processes. Chronic<br />

exposure to Mn can cause manganism, a neurodegenerative disorder similar to<br />

Parkinson’s disease. Mn neurotoxicity is characterized by impairment both in the<br />

expression and activity <strong>of</strong> glutamine (Gln) transporters in astrocytes. Since protein<br />

kinase C (PKC) activation leads to downregulation in a number <strong>of</strong> neurotransmitter<br />

transporters and Mn increases PKC activity, we hypothesized that the PKC signaling<br />

pathway may contribute in Mn-mediated disruption <strong>of</strong> glutamine turnover.<br />

We have found that Mn exposure elevates phosphorylation <strong>of</strong> PKCα and PKCδ<br />

is<strong>of</strong>orms. PKCs activity (measured by phosphorylation <strong>of</strong> substrate peptide using<br />

radiolabeled ATP) was found to be increased after Mn treatment. As was previously<br />

observed, Mn caused a decrease in Gln uptake. This effect was blocked by PKCs inhibitor.<br />

Notably, PKC activation by phorbol 12-myristate 13-acetate (PMA) caused<br />

a decrease in Gln uptake mediated by systems ASC and N, and had no effect on<br />

systems A and L. Treatment with PMA significantly decreased SNAT3 (system N)<br />

and ASCT2 (system ASC) protein levels. Additionally, a co-immunoprecipitation<br />

study showed association <strong>of</strong> SNAT3 and ASCT2 with the PKCδ is<strong>of</strong>orm.<br />

Combined, these findings demonstrate that Mn-induced dysregulation <strong>of</strong> Gln<br />

homeostasis in primary astrocytes involves PKC signaling.<br />

Acknowledgements: This study was supported by NIH grant ES-10463.<br />

2132 EXPRESSION OF MUTANT HUNTINGTIN INCREASES<br />

ARGINASE ACTIVITY IN MOUSE STRIATUM, BUT<br />

DECREASES ITS SUSCEPTIBILITY TO MANGANESE<br />

EXPOSURE.<br />

M. Wegrzynowicz 1, 2 , H. K. Holt 1, 2 and A. B. Bowman 1, 2, 3 . 1 Department <strong>of</strong><br />

Neurology, Vanderbilt University Medical Center, Nashville, TN, 2 Vanderbilt<br />

Kennedy Center, Vanderbilt University Medical Center, Nashville, TN and 3 Center<br />

for Molecular <strong>Toxicology</strong>, Vanderbilt University Medical Center, Nashville, TN.<br />

Huntington disease (HD), a neurodegenerative disorder principally affecting the<br />

striatum, is caused by mutation in the huntingtin (Htt) gene, but severity <strong>of</strong> the<br />

disease may be strongly influenced by unidentified environmental factors. We have<br />

reported a gene-environment interaction wherein expression <strong>of</strong> mutant (mt) HTT<br />

impairs striatal manganese (Mn) accumulation. We hypothesize that mtHTT expression<br />

may alter cellular markers <strong>of</strong> Mn toxicity.<br />

When testing potential markers we found that basal activity <strong>of</strong> the Mn-dependent<br />

enzyme, arginase, is elevated in the homogenates <strong>of</strong> striata from mtHTT-expressing<br />

mice (YAC128Q) comparing to wild-type (wt) littermates. Additionally, we observed<br />

that in vivo exposure to Mn (three 50 mg/kg MnCl 2 subcutaneous injections<br />

over one week) strongly elevates enzyme activity in striatal extracts, however this effect<br />

is less pronounced in mutant mice. Cortical activity was lower than striatal, and<br />

no effect <strong>of</strong> genotype was found. In vitro supplementation <strong>of</strong> vehicle-exposed mice<br />

striatum homogenates with 2 μM Mn (a concentration exceeding the values measured<br />

in the homogenates from animals exposed to Mn in vivo) was much less effective<br />

in arginase activation than subcutaneous exposure in vivo. We conclude that increased<br />

activity <strong>of</strong> arginase, an enzyme known to be neuroprotective, may be a<br />

compensatory response to mtHTT cytotoxicity in the striatum. Impaired activation<br />

<strong>of</strong> striatal arginase by Mn administered in vivo in YAC128Q mice is probably<br />

due to a mtHTT-induced deficit <strong>of</strong> Mn accumulation. Interestingly, the effect <strong>of</strong><br />

Mn is most likely only partially mediated by increased availability <strong>of</strong> Mn, a c<strong>of</strong>actor<br />

<strong>of</strong> arginase. Inability <strong>of</strong> in vitro added Mn to replicate the effects <strong>of</strong> in vivo exposure<br />

suggests that Mn may induce some cellular processes contributing in the activation<br />

<strong>of</strong> arginase.<br />

Support: NIH ES016931<br />

2133 NEUROTOXIC EFFECTS OF INTRANASAL<br />

MANGANESE EXPOSURE AND INTERACTION WITH<br />

CADMIUM EXPOSURE IN A MOUSE MODEL.<br />

A. H. Moberly, J. Pottackal, L. Czarnecki, T. Rubinstein, D. J. Turkel and J. P.<br />

McGann. Psychology Department, Rutgers University, Piscataway, NJ.<br />

Exposure to aerosolized manganese occurs in a variety <strong>of</strong> industrial settings, including<br />

mining and welding, but its effects on neurons directly exposed to manganese<br />

has not been explored in vivo. Transgenic mice expressing the fluorescent exocytosis<br />

indicator synaptopHluorin (spH) in olfactory receptor neuron terminals were used<br />

to visualize odorant-evoked neurotransmitter release from the olfactory nerve following<br />

acute toxicant exposure. Mice received a unilateral intranasal instillation <strong>of</strong><br />

2, 20, or 200 μg manganese chloride and a vehicle control on the contralateral side.<br />

Two days later they were anesthetized, implanted with a cranial window overlying<br />

the olfactory bulbs, and odorant-evoked neurotransmitter release was quantified bilaterally<br />

via optical imaging <strong>of</strong> spH signals. At the highest dose, odorant-evoked<br />

sensory input to the olfactory bulb on the Mn-exposed side was reduced to 14% <strong>of</strong><br />

control and while epithelial projections to the olfactory bulb were slightly reduced.<br />

<strong>The</strong> 20 μg dose reduced responses to 58% <strong>of</strong> control, while the lowest dose had no<br />

significant effect. Because we have previously shown similar effects using intranasal<br />

cadmium exposure, we further hypothesized that mixtures <strong>of</strong> the two heavy metals<br />

might demonstrate an interaction. To test this, the above experiment was repeated<br />

using instillations <strong>of</strong> a combination <strong>of</strong> 20 μg Mn and 2 μg Cd, doses found to produce<br />

intermediate reductions in odorant-evoked responding when presented individually.<br />

Mice exposed to this mixture exhibited odorant-evoked transmitter release<br />

from the olfactory nerve equal to only 11% <strong>of</strong> control. This decrease suggests that<br />

peripheral exposure to cadmium and manganese can cause pathopysiology comparable<br />

to the sum <strong>of</strong> their effects individually. We conclude that intranasal Mn exposure<br />

can significantly disrupt olfactory function. Moreover, its neurotoxic effects<br />

can summate with those <strong>of</strong> Cd, suggesting that total exposure to both should be<br />

considered in assessing risk levels.<br />

SOT 2011 ANNUAL MEETING 457

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!