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 - Chen, Pan A1 - Bornhorst, Julia A1 - Aschner, Michael T1 - Manganese metabolism in humans JF - Frontiers in Bioscience-Landmark N2 - Manganese (Mn) is an essential nutrient for intracellular activities; it functions as a cofactor for a variety of enzymes, including arginase, glutamine synthetase (GS), pyruvate carboxylase and Mn superoxide dismutase (Mn-SOD). Through these metalloproteins, Mn plays critically important roles in development, digestion, reproduction, antioxidant defense, energy production, immune response and regulation of neuronal activities. Mn deficiency is rare. In contrast Mn poisoning may be encountered upon overexposure to this metal. Excessive Mn tends to accumulate in the liver, pancreas, bone, kidney and brain, with the latter being the major target of Mn intoxication. Hepatic cirrhosis, polycythemia, hypermanganesemia, dystonia and Parkinsonism-like symptoms have been reported in patients with Mn poisoning. In recent years, Mn has come to the forefront of environmental concerns due to its neurotoxicity. Molecular mechanisms of Mn toxicity include oxidative stress, mitochondrial dysfunction, protein misfolding, endoplasmic reticulum (ER) stress, autophagy dysregulation, apoptosis, and disruption of other metal homeostasis. The mechanisms of Mn homeostasis are not fully understood. Here, we will address recent progress in Mn absorption, distribution and elimination across different tissues, as well as the intracellular regulation of Mn homeostasis in cells. We will conclude with recommendations for future research areas on Mn metabolism. KW - Manganese KW - Metal Metabolism KW - Homeostasis KW - Blood-Brain Barrier KW - Neurotoxicity KW - Transporters KW - Review Y1 - 2018 U6 - https://doi.org/10.2741/4665 SN - 1093-9946 SN - 1093-4715 VL - 23 IS - 9 SP - 1655 EP - 1679 PB - Frontiers in Bioscience INC CY - Irvine ER - TY - JOUR A1 - Crone, Barbara A1 - Aschner, Michael A. A1 - Schwerdtle, Tanja A1 - Karst, Uwe A1 - Bornhorst, Julia T1 - Elemental bioimaging of Cisplatin in Caenorhabditis elegans by LA-ICP-MS JF - Metallomics N2 - cis-Diamminedichloroplatinum(II) (Cisplatin) is one of the most important and frequently used cytostatic drugs for the treatment of various solid tumors. Herein, a laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) method incorporating a fast and simple sample preparation protocol was developed for the elemental mapping of Cisplatin in the model organism Caenorhabditis elegans (C. elegans). The method allows imaging of the spatially-resolved elemental distribution of platinum in the whole organism with respect to the anatomic structure in L4 stage worms at a lateral resolution of 5 μm. In addition, a dose- and time-dependent Cisplatin uptake was corroborated quantitatively by a total reflection X-ray fluorescence spectroscopy (TXRF) method, and the elemental mapping indicated that Cisplatin is located in the intestine and in the head of the worms. Better understanding of the distribution of Cisplatin in this well-established model organism will be instrumental in deciphering Cisplatin toxicity and pharmacokinetics. Since the cytostatic effect of Cisplatin is based on binding the DNA by forming intra- and interstrand crosslinks, the response of poly(ADP-ribose)metabolism enzyme 1 (pme-1) deletion mutants to Cisplatin was also examined. Loss of pme-1, which is the C. elegans ortholog of human poly(ADP-ribose) polymerase 1 (PARP-1) led to disturbed DNA damage response. With respect to survival and brood size, pme-1 deletion mutants were more sensitive to Cisplatin as compared to wildtype worms, while Cisplatin uptake was indistinguishable. Y1 - 2015 U6 - https://doi.org/10.1039/c5mt00096c SN - 1756-591X SN - 1756-5901 VL - 2015 IS - 7 SP - 1189 EP - 1195 PB - Royal Society of Chemistry CY - Cambridge 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 - Nicolai, Merle Marie A1 - Weishaupt, Ann-Kathrin A1 - Baesler, Jessica A1 - Brinkmann, Vanessa A1 - Wellenberg, Anna A1 - Winkelbeiner, Nicola Lisa A1 - Gremme, Anna A1 - Aschner, Michael A1 - Fritz, Gerhard A1 - Schwerdtle, Tanja A1 - Bornhorst, Julia T1 - Effects of manganese on genomic integrity in the multicellular model organism Caenorhabditis elegans JF - International Journal of Molecular Sciences N2 - Although manganese (Mn) is an essential trace element, overexposure is associated with Mn-induced toxicity and neurological dysfunction. Even though Mn-induced oxidative stress is discussed extensively, neither the underlying mechanisms of the potential consequences of Mn-induced oxidative stress on DNA damage and DNA repair, nor the possibly resulting toxicity are characterized yet. In this study, we use the model organism Caenorhabditis elegans to investigate the mode of action of Mn toxicity, focusing on genomic integrity by means of DNA damage and DNA damage response. Experiments were conducted to analyze Mn bioavailability, lethality, and induction of DNA damage. Different deletion mutant strains were then used to investigate the role of base excision repair (BER) and dePARylation (DNA damage response) proteins in Mn-induced toxicity. The results indicate a dose- and time-dependent uptake of Mn, resulting in increased lethality. Excessive exposure to Mn decreases genomic integrity and activates BER. Altogether, this study characterizes the consequences of Mn exposure on genomic integrity and therefore broadens the molecular understanding of pathways underlying Mn-induced toxicity. Additionally, studying the basal poly(ADP-ribosylation) (PARylation) of worms lacking poly(ADP-ribose) glycohydrolase (PARG) parg-1 or parg-2 (two orthologue of PARG), indicates that parg-1 accounts for most of the glycohydrolase activity in worms. KW - manganese KW - oxidative stress KW - DNA repair KW - DNA damage response KW - Caenorhabditis elegans Y1 - 2021 U6 - https://doi.org/10.3390/ijms222010905 SN - 1422-0067 VL - 22 IS - 20 PB - MDPI CY - Basel ER - TY - JOUR A1 - Ferrer, Beatriz A1 - Peres, Tanara Vieira A1 - dos Santos, Alessandra Antunes A1 - Bornhorst, Julia A1 - Morcillo, Patricia A1 - Goncalves, Cinara Ludvig A1 - Aschner, Michael T1 - Methylmercury affects the expression of hypothalamic neuropeptides that control body weight in C57BL/6J mice JF - Toxicological sciences N2 - Methylmercury (MeHg) is an environmental pollutant that affects primarily the central nervous system (CNS), causing neurological alterations. An early symptom of MeHg poisoning is the loss of body weight and appetite. Moreover, the CNS has an important role in controlling energy homeostasis. It is known that in the hypothalamus nutrient and hormonal signals converge to orchestrate control of body weight and food intake. In this study, we investigated if MeHg is able to induce changes in the expression of key hypothalamic neuropeptides that regulate energy homeostasis. Thus, hypothalamic neuronal mouse cell line GT 1-7 was treated with MeHg at different concentrations (0, 0.5, 1, and 5 mu M). MeHg induced the expression of the anorexigenic neuropeptide pro-omiomelanocortin (Pomc) and the orexigenic peptide Agouti-related peptide (Agrp) in a concentration-dependent manner, suggesting deregulation of mechanisms that control body weight. To confirm these in vitro observations, 8-week-old C57BL/6J mice (males and females) were exposed to MeHg in drinking water, modeling the most prevalent exposure route to this metal. After 30-day exposure, no changes in body weight were detected. However, MeHg treated males showed a significant decrease in fat depots. Moreover, MeHg affected the expression of hypothalamic neuropeptides that control food intake and body weight in a gender-and dose-dependent manner. Thus, MeHg increases Pomc mRNA only in males in a dose-dependent way, and it does not have effects on the expression of Agrp mRNA. The present study shows, for first time, that MeHg is able to induce changes in hypothalamic neuropeptides that regulate energy homeostasis, favoring an anorexigenic/catabolic profile. KW - methylmercury KW - hypothalamus KW - neuropeptides KW - control body weight KW - glucose homeostasis Y1 - 2018 U6 - https://doi.org/10.1093/toxsci/kfy052 SN - 1096-6080 SN - 1096-0929 VL - 163 IS - 2 SP - 557 EP - 568 PB - Oxford Univ. Press CY - Oxford ER - TY - JOUR A1 - Ruszkiewicz, Joanna A. A1 - de Macedo, Gabriel Teixeira A1 - Miranda-Vizuete, Antonio A1 - Bowman, Aaron B. A1 - Bornhorst, Julia A1 - Schwerdtle, Tanja A1 - Antunes Soares, Felix A. A1 - Aschner, Michael T1 - Sex-Specific response of caenorhabditis elegans to Methylmercury Toxicity JF - Neurotoxicity Research N2 - Methylmercury (MeHg), an abundant environmental pollutant, has long been known to adversely affect neurodevelopment in both animals and humans. Several reports from epidemiological studies, as well as experimental data indicate sex-specific susceptibility to this neurotoxicant; however, the molecular bases of this process are still not clear. In the present study, we used Caenorhabditis elegans (C. elegans), to investigate sex differences in response to MeHg toxicity during development. Worms at different developmental stage (L1, L4, and adult) were treated with MeHg for 1h. Lethality assays revealed that male worms exhibited significantly higher resistance to MeHg than hermaphrodites, when at L4 stage or adults. However, the number of worms with degenerated neurons was unaffected by MeHg, both in males and hermaphrodites. Lower susceptibility of males was not related to changes in mercury (Hg) accumulation, which was analogous for both wild-type (wt) and male-rich him-8 strain. Total glutathione (GSH) levels decreased upon MeHg in him-8, but not in wt. Moreover, the sex-dependent response of the cytoplasmic thioredoxin system was observedmales exhibited significantly higher expression of thioredoxin TRX-1, and thioredoxin reductase TRXR-1 expression was downregulated upon MeHg treatment only in hermaphrodites. These outcomes indicate that the redox status is an important contributor to sex-specific sensitivity to MeHg in C. elegans. KW - Methylmercury KW - Sex KW - Male KW - C KW - elegans KW - Antioxidant KW - Thioredoxin Y1 - 2019 U6 - https://doi.org/10.1007/s12640-018-9949-4 SN - 1029-8428 SN - 1476-3524 VL - 35 IS - 1 SP - 208 EP - 216 PB - Springer CY - New York 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 -