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.
Curr Protoc ; 1(2): e124, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33555621

ABSTRACT

Glutathione (GSH) plays fundamental roles in cellular redox buffering and is a common detoxification pathway for excretion of xenobiotics. This is especially crucial during vertebrate embryogenesis, when an organism is at one of its most vulnerable life stages. Importantly, GSH content and redox potential can dictate cell fate decisions, which can have profound consequences if altered by early life xenobiotic exposures. Owing to technical limitations, the best available method to detect and quantify changes in GSH has been high-pressure liquid chromatography, a terminal method that prevents suborganism-level resolution of these changes in developing embryos. Here, we describe a protocol that leverages the transparent nature of zebrafish embryos and the compatibility of monochlorobimane with the zebrafish GSH-S-transferase enzymes, to allow for the visualization of changes in GSH via S-glutathionylation in a live, developing embryo. This method can find broad application in developmental biology and toxicology. © 2021 Wiley Periodicals LLC.


Subject(s)
Glutathione , Zebrafish , Animals , Embryo, Nonmammalian , Pyrazoles
2.
Redox Biol ; 38: 101788, 2021 01.
Article in English | MEDLINE | ID: mdl-33321464

ABSTRACT

Emerging evidence suggests that redox-active chemicals perturb pancreatic islet development. To better understand potential mechanisms for this, we used zebrafish (Danio rerio) embryos to investigate roles of glutathione (GSH; predominant cellular redox buffer) and the transcription factor Nrf2a (Nfe2l2a; zebrafish Nrf2 co-ortholog) in islet morphogenesis. We delineated critical windows of susceptibility to redox disruption of ß-cell morphogenesis, interrogating embryos at 24, 48 and 72 h post fertilization (hpf) and visualized Nrf2a expression in the pancreas using whole-mount immunohistochemistry at 96 hpf. Chemical GSH modulation at 48 hpf induced significant islet morphology changes at 96 hpf. Pro-oxidant exposures to tert-butylhydroperoxide (77.6 µM; 10-min at 48 hpf) or tert-butylhydroquinone (1 µM; 48-56 hpf) decreased ß-cell cluster area at 96 hpf. Conversely, exposures to antioxidant N-acetylcysteine (bolsters GSH pools; 100 µM; 48-72 hpf) or sulforaphane (activates Nrf2a; 20 µM; 48-72 hpf) significantly increased islet areas. Nrf2a was also stabilized in ß-cells: 10-min exposures to 77.6 µM tert-butylhydroperoxide significantly increased Nrf2a protein compared to control islet cells that largely lack stabilized Nrf2a; 10-min exposures to higher (776 µM) tert-butylhydroperoxide concentration stabilized Nrf2a throughout the pancreas. Using biotinylated-GSH to visualize in situ protein glutathionylation, islet cells displayed high protein glutathionylation, indicating oxidized GSH pools. The 10-min high (776 µM) tert-butylhydroperoxide exposure (induced Nrf2a globally) decreased global protein glutathionylation at 96 hpf. Mutant fish expressing inactive Nrf2a were protected against tert-butylhydroperoxide-induced abnormal islet morphology. Our data indicate that disrupted redox homeostasis and Nrf2a stabilization during pancreatic ß-cell development impact morphogenesis, with implications for disease states at later life stages. Our work identifies a potential molecular target (Nrf2) that mediates abnormal ß-cell morphology in response to redox disruptions. Moreover, our findings imply that developmental exposure to exogenous stressors at distinct windows of susceptibility could diminish the reserve redox capacity of ß-cells, rendering them vulnerable to later-life stresses and disease.


Subject(s)
Glutathione , Zebrafish , Animals , Embryo, Nonmammalian , Organogenesis , Sulfhydryl Compounds , Zebrafish/genetics , Zebrafish Proteins/genetics
4.
Redox Biol ; 26: 101235, 2019 09.
Article in English | MEDLINE | ID: mdl-31202080

ABSTRACT

Glutathione (GSH), the most abundant vertebrate endogenous redox buffer, plays key roles in organogenesis and embryonic development, however, organ-specific GSH utilization during development remains understudied. Monochlorobimane (MCB), a dye conjugated with GSH by glutathione-s-transferase (GST) to form a fluorescent adduct, was used to visualize organ-specific GSH utilization in live developing zebrafish (Danio rerio) embryos. Embryos were incubated in 20 µM MCB for 1 h and imaged on an epifluorescence microscope. GSH conjugation with MCB was high during early organogenesis, decreasing as embryos aged. The heart had fluorescence 21-fold above autofluorescence at 24 hpf, dropping to 8.5-fold by 48 hpf; this increased again by 72 hpf to 23.5-fold, and stayed high till 96 hpf (18-fold). The brain had lower fluorescence (10-fold) at 24 and 48 hpf, steadily increasing to 30-fold by 96 hpf. The sensitivity and specificity of MCB staining was then tested with known GSH modulators. A 10-min treatment at 48 hpf with 750 µM tert-butylhydroperoxide, caused organ-specific reductions in staining, with the heart losing 30% fluorescence, and, the brain ventricle losing 47% fluorescence. A 24 h treatment from 24-48 hpf with 100 µM of N-Acetylcysteine (NAC) resulted in significantly increased fluorescence, with the brain ventricle and heart showing 312% and 240% increases respectively, these were abolished upon co-treatment with 5 µM BSO, an inhibitor of the enzyme that utilizes NAC to synthesize GSH. A 60 min 100 µM treatment with ethacrynic acid, a specific GST inhibitor, caused 30% reduction in fluorescence across all measured structures. MCB staining was then applied to test for GSH disruptions caused by the toxicants perfluorooctanesulfonic acid and mono-(2-ethyl-hexyl)phthalate; MCB fluorescence responded in a dose, structure and age-dependent manner. MCB staining is a robust, sensitive method to detect spatiotemporal changes in GSH utilization, and, can be applied to identify sensitive target tissues of toxicants.


Subject(s)
Brain/metabolism , Fluorescent Dyes/chemistry , Glutathione/metabolism , Pyrazoles/chemistry , Staining and Labeling/methods , Zebrafish/metabolism , Acetylcysteine/pharmacology , Alkanesulfonic Acids/toxicity , Animals , Brain/drug effects , Brain/growth & development , Diethylhexyl Phthalate/analogs & derivatives , Diethylhexyl Phthalate/toxicity , Embryo, Nonmammalian , Ethacrynic Acid/pharmacology , Fluorocarbons/toxicity , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/metabolism , Heart/drug effects , Heart/growth & development , Organogenesis/drug effects , Organogenesis/physiology , Toxicity Tests, Chronic , Zebrafish/embryology , Zebrafish/growth & development , tert-Butylhydroperoxide/pharmacology
5.
Semin Cell Dev Biol ; 80: 17-28, 2018 08.
Article in English | MEDLINE | ID: mdl-28927759

ABSTRACT

Vertebrate embryonic development requires specific signaling events that regulate cell proliferation and differentiation to occur at the correct place and the correct time in order to build a healthy embryo. Signaling pathways are sensitive to perturbations of the endogenous redox state, and are also susceptible to modulation by reactive species and antioxidant defenses, contributing to a spectrum of passive vs. active effects that can affect redox signaling and redox stress. Here we take a multi-level, integrative approach to discuss the importance of redox status for vertebrate developmental signaling pathways and cell fate decisions, with a focus on glutathione/glutathione disulfide, thioredoxin, and cysteine/cystine redox potentials and the implications for protein function in development. We present a tissue-specific example of the important role that reactive species play in pancreatic development and metabolic regulation. We discuss NFE2L2 (also known as NRF2) and related proteins, their roles in redox signaling, and their regulation of glutathione during development. Finally, we provide examples of xenobiotic compounds that disrupt redox signaling in the context of vertebrate embryonic development. Collectively, this review provides a systems-level perspective on the innate and inducible antioxidant defenses, as well as their roles in maintaining redox balance during chemical exposures that occur in critical windows of development.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Embryonic Development/physiology , Organogenesis/physiology , Oxidation-Reduction , Animals , Humans , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...