Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 14(9): e0221230, 2019.
Article in English | MEDLINE | ID: mdl-31513589

ABSTRACT

Thyroid hormone (TH) is essential for brain development. While disruption of TH signaling by environmental chemicals has been discussed as a mechanism of developmental neurotoxicity (DNT) for more than a decade, there remains a paucity of information linking specific TH disrupting chemicals to adverse neurodevelopmental outcomes. This data gap reflects, in part, the fact that the molecular machinery of TH signaling is complex and varies according to cell type and developmental time. Thus, establishing a baseline of the ontogenetic profile of expression of TH signaling molecules in relevant cell types is critical for developing in vitro and alternative systems-based models for screening TH disrupting chemicals for DNT. Here, we characterize the transcriptomic profile of molecules critical to TH signaling across three species-human, rat, and zebrafish-in vitro and in vivo across different stages of neurodevelopment. Our data indicate that while cultured human and rat neural progenitor cells, primary cultures of rat cortical cells, and larval zebrafish all express a fairly comprehensive transcriptome of TH signaling molecules, the spatiotemporal expression profiles as well as the responses to TH vary across species and developmental stages. The data presented here provides a roadmap for identifying appropriate in vitro and in simpler systems-based models for mechanistic studies and screening of chemicals that alter neurodevelopment via interference with TH action.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Thyroid Hormones/metabolism , Zebrafish Proteins/genetics , Zebrafish/growth & development , Animals , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Humans , In Vitro Techniques , Larva/cytology , Male , Neurogenesis , Optogenetics , Primary Cell Culture , Rats , Signal Transduction , Zebrafish/genetics
2.
Sci Rep ; 7: 44861, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28317842

ABSTRACT

Polybrominated diphenyl ethers (PBDEs) are bioaccumulating flame retardants causing developmental neurotoxicity (DNT) in humans and rodents. Their DNT effects are suspected to involve thyroid hormone (TH) signaling disruption. Here, we tested the hypothesis whether disturbance of neural progenitor cell (NPC) differentiation into the oligodendrocyte lineage (O4+ cells) by BDE-99 involves disruption of TH action in human and mouse (h,m)NPCs. Therefore, we quantified differentiation of NPCs into O4+ cells and measured their maturation via expression of myelin-associated genes (hMBP, mMog) in presence and absence of TH and/or BDE-99. T3 promoted O4+ cell differentiation in mouse, but not hNPCs, and induced hMBP/mMog gene expression in both species. BDE-99 reduced generation of human and mouse O4+ cells, but there is no indication for BDE-99 interfering with cellular TH signaling during O4+ cell formation. BDE-99 reduced hMBP expression due to oligodendrocyte reduction, but concentrations that did not affect the number of mouse O4+ cells inhibited TH-induced mMog transcription by a yet unknown mechanism. In addition, ascorbic acid antagonized only the BDE-99-dependent loss of human, not mouse, O4+ cells by a mechanism probably independent of reactive oxygen species. These data point to species-specific modes of action of BDE-99 on h/mNPC development into the oligodendrocyte lineage.


Subject(s)
Cell Differentiation/drug effects , Halogenated Diphenyl Ethers/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Oligodendroglia/cytology , Animals , Cell Line , Cell Lineage , Humans , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Species Specificity , Tyrosine 3-Monooxygenase
3.
Arch Toxicol ; 90(6): 1415-27, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26216354

ABSTRACT

The developing brain is highly vulnerable to the adverse effects of chemicals, resulting in neurodevelopmental disorders in humans. Currently, animal experiments in the rat are the gold standard for developmental neurotoxicity (DNT) testing; however, these guideline studies are insufficient in terms of animal use, time and costs and bear the issue of species extrapolation. Therefore, the necessity for alternative methods that predict DNT of chemicals faster, cheaper and with a high predictivity for humans is internationally agreed on. In this respect, we developed an in vitro model for DNT key event screening, which is based on primary human and rat neural progenitor cells grown as neurospheres. They are able to mimic basic processes of early fetal brain development and enable an investigation of species differences between humans and rodents in corresponding cellular models. The goal of this study was to investigate to what extent human and rat neurospheres were able to correctly predict the DNT potential of a well-characterized training set of nine chemicals by investigating effects on progenitor cell proliferation, migration and neuronal differentiation in parallel to cell viability, and to compare these chemical responses between human and rat neurospheres. We demonstrate that (1) by correlating these human and rat in vitro results to existing in vivo data, human and rat neurospheres classified most compounds correctly and thus may serve as a valuable component of a modular DNT testing strategy and (2) human and rat neurospheres differed in their sensitivity to most chemicals, reflecting toxicodynamic species differences of chemicals.


Subject(s)
Cell Proliferation/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurotoxicity Syndromes/embryology , Animals , Cell Culture Techniques , Cell Movement/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Neural Stem Cells/pathology , Rats , Species Specificity , Spheroids, Cellular
4.
Arch Toxicol ; 88(8): 1537-48, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24599297

ABSTRACT

Polybrominated diphenyl ethers (PBDEs) are bioaccumulating flame retardants found in rising concentrations in human tissue. Epidemiological and animal studies have raised concern for their potential to induce developmental neurotoxicity (DNT). Considering the essential role of calcium homeostasis in neurodevelopment, PBDE-induced disturbance of intracellular calcium concentration ([Ca(2+)]i) may underlie PBDE-induced DNT. To test this hypothesis, we investigated acute effects of BDE-47 and 6-OH-BDE-47 on [Ca(2+)]i in human neural progenitor cells (hNPCs) and unraveled involved signaling pathways. Short-time differentiated hNPCs were exposed to BDE-47, 6-OH-BDE-47, and multiple inhibitors/stimulators of presumably involved signaling pathways to determine possible effects on [Ca(2+)]i by single-cell microscopy with the fluorescent dye Fura-2. Initial characterization of calcium signaling pathways confirmed the early developmental stage of hNPCs. In these cells, BDE-47 (2 µM) and 6-OH-BDE-47 (0.2 µM) induce [Ca(2+)]i transients. This increase in [Ca(2+)]i is due to extracellular Ca(2+) influx and intracellular release of Ca(2+), mainly from the endoplasmic reticulum (ER). While extracellular Ca(2+) seems to enter the cytoplasm upon 6-OH-BDE-47 by interfering with the cell membrane and independent of Ca(2+) ion channels, ER-derived Ca(2+) is released following activation of protein lipase C and inositol 1,4,5-trisphosphate receptor, but independently of ryanodine receptors. These findings illustrate that immature developing hNPCs respond to low concentrations of 6-OH-BDE-47 by an increase in [Ca(2+)]i and provide new mechanistic explanations for such BDE-induced calcium disruption. Thus, these data support the possibility of a critical window of PBDE exposure, i.e., early human brain development, which has to be acknowledged in risk assessment.


Subject(s)
Calcium/metabolism , Fetal Stem Cells/drug effects , Halogenated Diphenyl Ethers/toxicity , Homeostasis/drug effects , Neural Stem Cells/drug effects , Polybrominated Biphenyls/toxicity , Ryanodine Receptor Calcium Release Channel/metabolism , Cells, Cultured , Fetal Stem Cells/metabolism , Gestational Age , Homeostasis/physiology , Humans , Membrane Potentials/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Patch-Clamp Techniques , Primary Cell Culture
5.
Biochim Biophys Acta ; 1788(12): 2594-602, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19814996

ABSTRACT

ETT (originally designated as OCTN1; human gene symbol SLC22A4) and CTT (OCTN2; SLC22A5) are highly specific transporters of ergothioneine and carnitine, respectively. Despite a high degree of sequence homology, both carriers discriminate precisely between substrates: ETT does not transport carnitine, and CTT does not transport ergothioneine. Our aim was to turn ETT into a transporter for carnitine and CTT into a transporter for ergothioneine by a limited number of point mutations. From a multiple alignment of several mammalian amino acid sequences, those positions were selected for conversion that were momentously different between ETT and CTT from human but conserved among all orthologues. Mutants were expressed in 293 cells and assayed for transport of ergothioneine and carnitine. Several ETT mutants clearly catalyzed transport of carnitine, up to 35% relative to wild-type CTT. Amazingly, complementary substitutions in CTT did not provoke transport activity for ergothioneine. In similar contrast, carnitine transport by CTT mutants was abolished by very few substitutions, whereas ergothioneine transport by ETT mutants was maintained even with the construct most active in carnitine transport. To explain these results, we propose that ETT and CTT use dissimilar pathways for conformational change, in addition to incongruent substrate binding sites. In other words, carnitine is excluded from ETT by binding, and ergothioneine is excluded from CTT by turnover movement. Our data indicate amino acids critical for substrate discrimination not only in transmembrane segments 5, 7, 8, and 10, but also in segments 9 and 12 which were hitherto considered as unimportant.


Subject(s)
Antiporters/metabolism , Carnitine/metabolism , Ergothioneine/metabolism , Organic Cation Transport Proteins/metabolism , Amino Acid Substitution , Antiporters/genetics , Biological Transport/physiology , Carnitine/genetics , Cell Line , Ergothioneine/genetics , Humans , Mutation, Missense , Organic Cation Transport Proteins/genetics , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Sequence Homology, Amino Acid , Solute Carrier Family 22 Member 5 , Substrate Specificity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...