TY - JOUR A1 - Peres, Tanara Vieira A1 - Arantes, Leticia P. A1 - Miah, Mahfuzur R. A1 - Bornhorst, Julia A1 - Schwerdtle, Tanja A1 - Bowman, Aaron B. A1 - Leal, Rodrigo B. A1 - Aschner, Michael T1 - Role of Caenorhabditis elegans AKT-1/2 and SGK-1 in Manganese Toxicity JF - Neurotoxicity Research N2 - Excessive levels of the essential metal manganese (Mn) may cause a syndrome similar to Parkinson’s disease. The model organism Caenorhabditis elegans mimics some of Mn effects in mammals, including dopaminergic neurodegeneration, oxidative stress, and increased levels of AKT. The evolutionarily conserved insulin/insulin-like growth factor-1 signaling pathway (IIS) modulates worm longevity, metabolism, and antioxidant responses by antagonizing the transcription factors DAF-16/FOXO and SKN-1/Nrf-2. AKT-1, AKT-2, and SGK-1 act upstream of these transcription factors. To study the role of these proteins in C. elegans response to Mn intoxication, wild-type N2 and loss-of-function mutants were exposed to Mn (2.5 to 100 mM) for 1 h at the L1 larval stage. Strains with loss-of-function in akt-1, akt-2, and sgk-1 had higher resistance to Mn compared to N2 in the survival test. All strains tested accumulated Mn similarly, as shown by ICP-MS. DAF-16 nuclear translocation was observed by fluorescence microscopy in WT and loss-of-function strains exposed to Mn. qRT-PCR data indicate increased expression of γ-glutamyl cysteine synthetase (GCS-1) antioxidant enzyme in akt-1 mutants. The expression of sod-3 (superoxide dismutase homologue) was increased in the akt-1 mutant worms, independent of Mn treatment. However, dopaminergic neurons degenerated even in the more resistant strains. Dopaminergic function was evaluated with the basal slowing response behavioral test and dopaminergic neuron integrity was evaluated using worms expressing green fluorescent protein (GFP) under the dopamine transporter (DAT-1) promoter. These results suggest that AKT-1/2 and SGK-1 play a role in C. elegans response to Mn intoxication. However, tissue-specific responses may occur in dopaminergic neurons, contributing to degeneration. KW - Manganese . C. elegans KW - Signaling pathways KW - DAF-16 KW - Akt/PKB KW - SGK-1 Y1 - 2018 U6 - https://doi.org/10.1007/s12640-018-9915-1 SN - 1029-8428 SN - 1476-3524 VL - 34 IS - 3 SP - 584 EP - 596 PB - Springer CY - New York ER - TY - JOUR A1 - Gubert, Priscila A1 - Puntel, Bruna A1 - Lehmen, Tassia A1 - Fessel, Joshua P. A1 - Cheng, Pan A1 - Bornhorst, Julia A1 - Trindade, Lucas Siqueira A1 - Avila, Daiana S. A1 - Aschner, Michael A1 - Soares, Felix A. A. T1 - Metabolic effects of manganese in the nematode Caenorhabditis elegans through DAergic pathway and transcription factors activation JF - Neurotoxicology : the interdisciplinary journal of effects to toxic substances on the nervous system N2 - Manganese (Mn) is an essential trace element for physiological functions since it acts as an enzymatic co-factor. Nevertheless, overexposure to Mn has been associated with a pathologic condition called manganism. Furthermore, Mn has been reported to affect lipid metabolism by mechanisms which have yet to be established. Herein, we used the nematode Caenorhabditis elegans to examine Mn’s effects on the dopaminergic (DAergic) system and determine which transcription factors that regulate with lipid metabolism are affected by it. Worms were exposed to Mn for four hours in the presence of bacteria and in a liquid medium (85 mM NaCl). Mn increased fat storage as evidenced both by Oil Red O accumulation and triglyceride levels. In addition, metabolic activity was reduced as a reflection of decreased oxygen consumption caused by Mn. Mn also affected feeding behavior as evidenced by decreased pharyngeal pumping rate. DAergic neurons viability were not altered by Mn, however the dopamine levels were significantly reduced following Mn exposure. Furthermore, the expression of sbp-1 transcription factor and let-363 protein kinase responsible for lipid accumulation control was increased and decreased, respectively, by Mn. Altogether, our data suggest that Mn increases the fat storage in C. elegans, secondary to DAergic system alterations, under the control of SBP-1 and LET-363 proteins. KW - Manganese KW - Caenorhabditis elegans KW - Lipid metabolism KW - Dopaminergic system KW - Manganism Y1 - 2018 U6 - https://doi.org/10.1016/j.neuro.2018.04.008 SN - 0161-813X SN - 1872-9711 VL - 67 SP - 65 EP - 72 PB - Elsevier CY - Amsterdam ER - TY - JOUR A1 - Rohn, Isabelle A1 - Marschall, Talke Anu A1 - Kröpfl, Nina A1 - Jensen, Kenneth Bendix A1 - Aschner, Michael A1 - Tuck, Simon A1 - Kuehnelt, Doris A1 - Schwerdtle, Tanja A1 - Bornhorst, Julia T1 - Selenium species-dependent toxicity, bioavailability and metabolic transformations in Caenorhabditis elegans JF - Metallomics : integrated biometal science N2 - The essential micronutrient selenium (Se) is required for various systemic functions, but its beneficial range is narrow and overexposure may result in adverse health effects. Additionally, the chemical form of the ingested selenium contributes crucially to its health effects. While small Se species play a major role in Se metabolism, their toxicological effects, bioavailability and metabolic transformations following elevated uptake are poorly understood. Utilizing the tractable invertebrate Caenorhabditis elegans allowed for an alternative approach to study species-specific characteristics of organic and inorganic Se forms in vivo, revealing remarkable species-dependent differences in the toxicity and bioavailability of selenite, selenomethionine (SeMet) and Se-methylselenocysteine (MeSeCys). An inverse relationship was found between toxicity and bioavailability of the Se species, with the organic species displaying a higher bioavailability than the inorganic form, yet being less toxic. Quantitative Se speciation analysis with HPLC/mass spectrometry revealed a partial metabolism of SeMet and MeSeCys. In SeMet exposed worms, identified metabolites were Se-adenosylselenomethionine (AdoSeMet) and Se-adenosylselenohomocysteine (AdoSeHcy), while worms exposed to MeSeCys produced Se-methylselenoglutathione (MeSeGSH) and -glutamyl-MeSeCys (-Glu-MeSeCys). Moreover, the possible role of the sole selenoprotein in the nematode, thioredoxin reductase-1 (TrxR-1), was studied comparing wildtype and trxr-1 deletion mutants. Although a lower basal Se level was detected in trxr-1 mutants, Se toxicity and bioavailability following acute exposure was indistinguishable from wildtype worms. Altogether, the current study demonstrates the suitability of C. elegans as a model for Se species dependent toxicity and metabolism, while further research is needed to elucidate TrxR-1 function in the nematode. Y1 - 2018 U6 - https://doi.org/10.1039/c8mt00066b SN - 1756-5901 SN - 1756-591X VL - 10 IS - 6 SP - 818 EP - 827 PB - Royal Society of Chemistry CY - Cambridge ER - TY - JOUR A1 - Rohn, Isabelle A1 - Raschke, Stefanie A1 - Aschner, Michael A1 - Tuck, Simon A1 - Kuehnelt, Doris A1 - Kipp, Anna Patricia A1 - Schwerdtle, Tanja A1 - Bornhorst, Julia T1 - Treatment of caenorhabditis elegans with small selenium species enhances antioxidant defense systems JF - Molecular nutrition & food research : bioactivity, chemistry, immunology, microbiology, safety, technology N2 - ScopeSmall selenium (Se) species play a key role in Se metabolism and act as dietary sources of the essential trace element. However, they are redox-active and trigger pro- and antioxidant responses. As health outcomes are strongly species-dependent, species-specific characteristics of Se compounds are tested in vivo. Methods and resultsIn the model organism Caenorhabditis elegans (C. elegans), immediate and sustained effects of selenite, selenomethionine (SeMet), and Se-methylselenocysteine (MeSeCys) are studied regarding their bioavailability, incorporation into proteins, as well as modulation of the cellular redox status. While all tested Se compounds are bioavailable, only SeMet persistently accumulates and is non-specifically incorporated into proteins. However, the protection toward chemically-induced formation of reactive species is independent of the applied Se compound. Increased thioredoxin reductase (TXNRD) activity and changes in mRNA expression levels of antioxidant proteins indicate the activation of cellular defense mechanisms. However, in txnrd-1 deletion mutants, no protective effects of the Se species are observed anymore, which is also reflected by differential gene expression data. ConclusionSe species protect against chemically-induced reactive species formation. The identified immediate and sustained systemic effects of Se species give rise to speculations on possible benefits facing subsequent periods of inadequate Se intake. KW - antioxidant defense systems KW - caenorhabditis elegans KW - selenium KW - oxidative stress KW - selenoproteins Y1 - 2019 U6 - https://doi.org/10.1002/mnfr.201801304 SN - 1613-4125 SN - 1613-4133 VL - 63 IS - 9 PB - Wiley CY - Hoboken ER - TY - JOUR A1 - Henze, Andrea A1 - Homann, Thomas A1 - Rohn, Isabelle A1 - Aschner, Michael A. A1 - Link, Christopher D. A1 - Kleuser, Burkhard A1 - Schweigert, Florian J. A1 - Schwerdtle, Tanja A1 - Bornhorst, Julia T1 - Caenorhabditis elegans as a model system to study post-translational modifications of human transthyretin JF - Scientific reports N2 - The visceral protein transthyretin (TTR) is frequently affected by oxidative post-translational protein modifications (PTPMs) in various diseases. Thus, better insight into structure-function relationships due to oxidative PTPMs of TTR should contribute to the understanding of pathophysiologic mechanisms. While the in vivo analysis of TTR in mammalian models is complex, time- and resource-consuming, transgenic Caenorhabditis elegans expressing hTTR provide an optimal model for the in vivo identification and characterization of drug-mediated oxidative PTPMs of hTTR by means of matrix assisted laser desorption/ionization – time of flight – mass spectrometry (MALDI-TOF-MS). Herein, we demonstrated that hTTR is expressed in all developmental stages of Caenorhabditis elegans, enabling the analysis of hTTR metabolism during the whole life-cycle. The suitability of the applied model was verified by exposing worms to D-penicillamine and menadione. Both drugs induced substantial changes in the oxidative PTPM pattern of hTTR. Additionally, for the first time a covalent binding of both drugs with hTTR was identified and verified by molecular modelling. KW - n-acetyl-cysteine KW - s-glutathionylation KW - force-field KW - c. elegans KW - life-span KW - protein KW - cells KW - menadione KW - disease KW - binding Y1 - 2016 U6 - https://doi.org/10.1038/srep37346 SN - 2045-2322 VL - 6 PB - Nature Publishing Group CY - London ER - TY - JOUR A1 - Kumar, Kevin K. A1 - Goodwin, Cody R. A1 - Uhouse, Michael A. A1 - Bornhorst, Julia A1 - Schwerdtle, Tanja A1 - Aschner, Michael A. A1 - McLean, John A. A1 - Bowman, Aaron B. T1 - Untargeted metabolic profiling identifies interactions between Huntington's disease and neuronal manganese status JF - Metallomics N2 - Manganese (Mn) is an essential micronutrient for development and function of the nervous system. Deficiencies in Mn transport have been implicated in the pathogenesis of Huntington's disease (HD), an autosomal dominant neurodegenerative disorder characterized by loss of medium spiny neurons of the striatum. Brain Mn levels are highest in striatum and other basal ganglia structures, the most sensitive brain regions to Mn neurotoxicity. Mouse models of HD exhibit decreased striatal Mn accumulation and HD striatal neuron models are resistant to Mn cytotoxicity. We hypothesized that the observed modulation of Mn cellular transport is associated with compensatory metabolic responses to HD pathology. Here we use an untargeted metabolomics approach by performing ultraperformance liquid chromatography-ion mobility-mass spectrometry (UPLC-IM-MS) on control and HD immortalized mouse striatal neurons to identify metabolic disruptions under three Mn exposure conditions, low (vehicle), moderate (non-cytotoxic) and high (cytotoxic). Our analysis revealed lower metabolite levels of pantothenic acid, and glutathione (GSH) in HD striatal cells relative to control cells. HD striatal cells also exhibited lower abundance and impaired induction of isobutyryl carnitine in response to increasing Mn exposure. In addition, we observed induction of metabolites in the pentose shunt pathway in HD striatal cells after high Mn exposure. These findings provide metabolic evidence of an interaction between the HD genotype and biologically relevant levels of Mn in a striatal cell model with known HD by Mn exposure interactions. The metabolic phenotypes detected support existing hypotheses that changes in energetic processes underlie the pathobiology of both HD and Mn neurotoxicity. KW - hallervorden-spatz-syndrome KW - mobility-mass spectrometry KW - energy-metabolism KW - coenzyme-a KW - model KW - neurotoxicity KW - glutathione KW - database KW - cells KW - neurodegeneration Y1 - 2015 U6 - https://doi.org/10.1039/C4MT00223G SN - 1756-591X SN - 1756-5901 VL - 7 SP - 363 EP - 370 PB - RSC Publ. CY - Cambridge ER - TY - GEN A1 - Henze, Andrea A1 - Homann, Thomas A1 - Rohn, Isabelle A1 - Aschner, Michael A. A1 - Link, Christopher D. A1 - Kleuser, Burkhard A1 - Schweigert, Florian J. A1 - Schwerdtle, Tanja A1 - Bornhorst, Julia T1 - Caenorhabditis elegans as a model system to study post-translational modifications of human transthyretin N2 - The visceral protein transthyretin (TTR) is frequently affected by oxidative post-translational protein modifications (PTPMs) in various diseases. Thus, better insight into structure-function relationships due to oxidative PTPMs of TTR should contribute to the understanding of pathophysiologic mechanisms. While the in vivo analysis of TTR in mammalian models is complex, time- and resource-consuming, transgenic Caenorhabditis elegans expressing hTTR provide an optimal model for the in vivo identification and characterization of drug-mediated oxidative PTPMs of hTTR by means of matrix assisted laser desorption/ionization – time of flight – mass spectrometry (MALDI-TOF-MS). Herein, we demonstrated that hTTR is expressed in all developmental stages of Caenorhabditis elegans, enabling the analysis of hTTR metabolism during the whole life-cycle. The suitability of the applied model was verified by exposing worms to D-penicillamine and menadione. Both drugs induced substantial changes in the oxidative PTPM pattern of hTTR. Additionally, for the first time a covalent binding of both drugs with hTTR was identified and verified by molecular modelling. T3 - Zweitveröffentlichungen der Universität Potsdam : Mathematisch-Naturwissenschaftliche Reihe - 312 KW - binding KW - c. elegans KW - cells KW - disease KW - force-field KW - life-span KW - menadione KW - n-acetyl-cysteine KW - protein KW - s-glutathionylation Y1 - 2016 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:kobv:517-opus4-103674 ER - TY - GEN A1 - Chakraborty, Sudipta A1 - Chen, Pan A1 - Bornhorst, Julia A1 - Schwerdtle, Tanja A1 - Schumacher, Fabian A1 - Kleuser, Burkhard A1 - Bowman, Aaron B. A1 - Aschner, Michael A. T1 - Loss of pdr-1/parkin influences Mn homeostasis through altered ferroportin expression in C. elegans N2 - Overexposure to the essential metal manganese (Mn) can result in an irreversible condition known as manganism that shares similar pathophysiology with Parkinson's disease (PD), including dopaminergic (DAergic) cell loss that leads to motor and cognitive impairments. However, the mechanisms behind this neurotoxicity and its relationship with PD remain unclear. Many genes confer risk for autosomal recessive, early-onset PD, including the parkin/PARK2 gene that encodes for the E3 ubiquitin ligase Parkin. Using Caenorhabditis elegans (C. elegans) as an invertebrate model that conserves the DAergic system, we previously reported significantly increased Mn accumulation in pdr-1/parkin mutants compared to wildtype (WT) animals. For the current study, we hypothesize that this enhanced accumulation is due to alterations in Mn transport in the pdr-1 mutants. While no change in mRNA expression of the major Mn importer proteins (smf-1-3) was found in pdr-1 mutants, significant downregulation in mRNA levels of the putative Mn exporter ferroportin (fpn-1.1) was observed. Using a strain overexpressing fpn-1.1 in worms lacking pdr-1, we show evidence for attenuation of several endpoints of Mn-induced toxicity, including survival, metal accumulation, mitochondrial copy number and DAergic integrity, compared to pdr-1 mutants alone. These changes suggest a novel role of pdr-1 in modulating Mn export through altered transporter expression, and provides further support of metal dyshomeostasis as a component of Parkinsonism pathophysiology. T3 - Zweitveröffentlichungen der Universität Potsdam : Mathematisch-Naturwissenschaftliche Reihe - 290 Y1 - 2015 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:kobv:517-opus4-99508 ER - TY - GEN A1 - Kumar, Kevin K. A1 - Goodwin, Cody R. A1 - Uhouse, Michael A. A1 - Bornhorst, Julia A1 - Schwerdtle, Tanja A1 - Aschner, Michael A. A1 - McLean, John A. A1 - Bowman, Aaron B. T1 - Untargeted metabolic profiling identifies interactions between Huntington's disease and neuronal manganese status N2 - Manganese (Mn) is an essential micronutrient for development and function of the nervous system. Deficiencies in Mn transport have been implicated in the pathogenesis of Huntington's disease (HD), an autosomal dominant neurodegenerative disorder characterized by loss of medium spiny neurons of the striatum. Brain Mn levels are highest in striatum and other basal ganglia structures, the most sensitive brain regions to Mn neurotoxicity. Mouse models of HD exhibit decreased striatal Mn accumulation and HD striatal neuron models are resistant to Mn cytotoxicity. We hypothesized that the observed modulation of Mn cellular transport is associated with compensatory metabolic responses to HD pathology. Here we use an untargeted metabolomics approach by performing ultraperformance liquid chromatography-ion mobility-mass spectrometry (UPLC-IM-MS) on control and HD immortalized mouse striatal neurons to identify metabolic disruptions under three Mn exposure conditions, low (vehicle), moderate (non-cytotoxic) and high (cytotoxic). Our analysis revealed lower metabolite levels of pantothenic acid, and glutathione (GSH) in HD striatal cells relative to control cells. HD striatal cells also exhibited lower abundance and impaired induction of isobutyryl carnitine in response to increasing Mn exposure. In addition, we observed induction of metabolites in the pentose shunt pathway in HD striatal cells after high Mn exposure. These findings provide metabolic evidence of an interaction between the HD genotype and biologically relevant levels of Mn in a striatal cell model with known HD by Mn exposure interactions. The metabolic phenotypes detected support existing hypotheses that changes in energetic processes underlie the pathobiology of both HD and Mn neurotoxicity. T3 - Zweitveröffentlichungen der Universität Potsdam : Mathematisch-Naturwissenschaftliche Reihe - 232 KW - cells KW - coenzyme-a KW - database KW - energy-metabolism KW - glutathione KW - hallervorden-spatz-syndrome KW - mobility-mass spectrometry KW - model KW - neurodegeneration KW - neurotoxicity Y1 - 2015 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:kobv:517-opus4-94314 SP - 363 EP - 370 ER - TY - GEN A1 - Avila, Daiana Silva A1 - Benedetto, Alexandre A1 - Au, Catherine A1 - Bornhorst, Julia A1 - Aschner, Michael A. T1 - Involvement of heat shock proteins on Mn-induced toxicity in Caenorhabditis elegans T2 - BMC pharmacology and toxicology N2 - Background: All living cells display a rapid molecular response to adverse environmental conditions, and the heat shock protein family reflects one such example. Hence, failing to activate heat shock proteins can impair the cellular response. In the present study, we evaluated whether the loss of different isoforms of heat shock protein ( hsp ) genes in Caenorhabditis elegans would affect their vulnerability to Manganese (Mn) toxicity. Methods: We exposed wild type and selected hsp mutant worms to Mn (30 min) and next evaluated further the most susceptible strains. We analyzed survi val, protein carbonylation (as a marker of oxidative stress) and Parkinson ’ s disease related gene expression immediately after Mn exposure. Lastly, we observed dopaminergic neurons in wild type worms and in hsp-70 mutants following Mn treatment. Analysis of the data was performed by one-way or two way ANOVA, depending on the case, followed by post-hoc Bonferroni test if the overall p value was less than 0.05. Results: We verified that the loss of hsp-70, hsp-3 and chn-1 increased the vulnerability to Mn, as exposed mutant worms showed lower survival rate and increased protein oxidation. The importance of hsp-70 against Mn toxicity was then corroborated in dopaminergic neurons, where Mn neurotoxicity was aggravated. The lack of hsp-70 also blocked the transcriptional upregulation of pink1 , a gene that has been linked to Parkinson ’ sdisease. Conclusions: Taken together, our data suggest that Mn exposu re modulates heat shock protein expression, particularly HSP-70, in C. elegans .Furthermore,lossof hsp-70 increases protein oxidation and dopaminergic neuronal degeneration following manganese exposure, which is associated with the inhibition of pink1 increased expression, thus pot entially exacerbating the v ulnerability to this metal. T3 - Zweitveröffentlichungen der Universität Potsdam : Mathematisch-Naturwissenschaftliche Reihe - 439 KW - Caenorhabitis elegans KW - Manganese KW - heat shock proteins KW - hsp-70 KW - pink1 Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:kobv:517-opus4-407286 ER -