Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Neurobiol Dis ; 158: 105470, 2021 10.
Article in English | MEDLINE | ID: mdl-34371143

ABSTRACT

Mitochondrial superoxide (O2-) production is implicated in aging, neurodegenerative disease, and most recently epilepsy. Yet the specific contribution of neuronal O2- to these phenomena is unclear. Here, we selectively deleted superoxide dismutase-2 (SOD2) in neuronal basic helix-loop-helix transcription factor (NEX)-expressing cells restricting deletion to a subset of excitatory principle neurons primarily in the forebrain (cortex and hippocampus). This resulted in nSOD2 KO mice that lived into adulthood (2-3 months) with epilepsy, selective loss of neurons, metabolic rewiring and a marked mitohormetic gene response. Surprisingly, expression of an astrocytic gene, glial fibrillary acidic protein (GFAP) was significantly increased relative to WT. Further studies in rat primary neuron-glial cultures showed that increased mitochondrial O2-, specifically in neurons, was sufficient to upregulate GFAP. These results suggest that neuron-specific mitochondrial O2- is sufficient to drive a complex and catastrophic epileptic phenotype and highlights the ability of SOD2 to act in a cell-nonautonomous manner to influence an astrocytic response.


Subject(s)
Astrocytes/pathology , Epilepsy/pathology , Glucose Metabolism Disorders/pathology , Mitochondria , Neurons , Oxidative Stress , Animals , Behavior, Animal , Electroencephalography , Epilepsy/psychology , Glial Fibrillary Acidic Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Primary Cell Culture , Rats , Superoxide Dismutase/genetics , Superoxides/metabolism
2.
Redox Biol ; 67: 102895, 2023 11.
Article in English | MEDLINE | ID: mdl-37769522

ABSTRACT

Glutathione (GSH) depletion, and impaired redox homeostasis have been observed in experimental animal models and patients with epilepsy. Pleiotropic strategies that elevate GSH levels via transcriptional regulation have been shown to significantly decrease oxidative stress and seizure frequency, increase seizure threshold, and rescue certain cognitive deficits. Whether elevation of GSH per se alters neuronal hyperexcitability remains unanswered. We previously showed that thiols such as dimercaprol (DMP) elevate GSH via post-translational activation of glutamate cysteine ligase (GCL), the rate limiting GSH biosynthetic enzyme. Here, we asked if elevation of cellular GSH by DMP altered neuronal hyperexcitability in-vitro and in-vivo. Treatment of primary neuronal-glial cerebrocortical cultures with DMP elevated GSH and inhibited a voltage-gated potassium channel blocker (4-aminopyridine, 4AP) induced neuronal hyperexcitability. DMP increased GSH in wildtype (WT) zebrafish larvae and significantly attenuated convulsant pentylenetetrazol (PTZ)-induced acute 'seizure-like' swim behavior. DMP treatment increased GSH and inhibited convulsive, spontaneous 'seizure-like' swim behavior in the Dravet Syndrome (DS) zebrafish larvae (scn1Lab). Furthermore, DMP treatment significantly decreased spontaneous electrographic seizures and associated seizure parameters in scn1Lab zebrafish larvae. We investigated the role of the redox-sensitive mammalian target of rapamycin (mTOR) pathway due to the presence of several cysteine-rich proteins and their involvement in regulating neuronal excitability. Treatment of primary neuronal-glial cerebrocortical cultures with 4AP or l-buthionine-(S,R)-sulfoximine (BSO), an irreversible inhibitor of GSH biosynthesis, significantly increased mTOR complex I (mTORC1) activity which was rescued by pre-treatment with DMP. Furthermore, BSO-mediated GSH depletion oxidatively modified the tuberous sclerosis protein complex (TSC) consisting of hamartin (TSC1), tuberin (TSC2), and TBC1 domain family member 7 (TBC1D7) which are critical negative regulators of mTORC1. In summary, our results suggest that DMP-mediated GSH elevation by a novel post-translational mechanism can inhibit neuronal hyperexcitability both in-vitro and in-vivo and a plausible link is the redox sensitive mTORC1 pathway.


Subject(s)
Glutathione , Zebrafish , Animals , Humans , Zebrafish/metabolism , Glutathione/metabolism , Glutamate-Cysteine Ligase/metabolism , TOR Serine-Threonine Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1 , Seizures/chemically induced , Seizures/drug therapy , Buthionine Sulfoximine/pharmacology , Mammals/metabolism
3.
J Neurochem ; 118(1): 79-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21517855

ABSTRACT

Mitochondrial oxidative stress is a contributing factor in the etiology of numerous neuronal disorders. However, the precise mechanism(s) by which mitochondrial reactive oxygen species modify cellular targets to induce neurotoxicity remains unknown. In this study, we determined the role of mitochondrial aconitase (m-aconitase) in neurotoxicity by decreasing its expression. Incubation of the rat dopaminergic cell line, N27, with paraquat (PQ(2+) ) resulted in aconitase inactivation, increased hydrogen peroxide (H(2) O(2) ) and increased ferrous iron (Fe(2+) ) at times preceding cell death. To confirm the role of m-aconitase in dopaminergic cell death, we knocked down m-aconitase expression via RNA interference. Incubation of m-aconitase knockdown N27 cells with PQ(2+) resulted in decreased H(2) O(2) production, Fe(2+) accumulation, and cell death compared with cells expressing basal levels of m-aconitase. To determine the metabolic role of m-aconitase in mediating neuroprotection, we conducted a complete bioenergetic profile. m-Aconitase knockdown N27 cells showed a global decrease in metabolism (glycolysis and oxygen consumption rates) which blocked PQ(2+) -induced H(+) leak and respiratory capacity deficiency. These findings suggest that dopaminergic cells are protected from death by decreasing release of H(2) O(2) and Fe(2+) in addition to decreased cellular metabolism.


Subject(s)
Aconitate Hydratase/metabolism , Dopamine/metabolism , Hydrogen Peroxide/metabolism , Iron/metabolism , Mitochondria/metabolism , Aconitate Hydratase/deficiency , Aconitate Hydratase/genetics , Analysis of Variance , Animals , Antimycin A/pharmacology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Death/drug effects , Cell Line, Transformed , Fumarate Hydratase/metabolism , Gene Expression Regulation/drug effects , Herbicides/toxicity , Ionophores/pharmacology , L-Lactate Dehydrogenase/metabolism , Mitochondria/drug effects , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Paraquat/toxicity , Protons , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Time Factors , Transfection/methods
4.
PLoS One ; 15(10): e0240451, 2020.
Article in English | MEDLINE | ID: mdl-33052948

ABSTRACT

Cell differentiation and cell fate determination in sensory systems are essential for stimulus discrimination and coding of environmental stimuli. Color vision is based on the differential color sensitivity of retinal photoreceptors, however the developmental programs that control photoreceptor cell differentiation and specify color sensitivity are poorly understood. In Drosophila melanogaster, there is evidence that the color sensitivity of different photoreceptors in the compound eye is regulated by inductive signals between cells, but the exact nature of these signals and how they are propagated remains unknown. We conducted a genetic screen to identify additional regulators of this process and identified a novel mutation in the hibris gene, which encodes an irre cell recognition module protein (IRM). These immunoglobulin super family cell adhesion molecules include human KIRREL and nephrin (NPHS1). hibris is expressed dynamically in the developing Drosophila melanogaster eye and loss-of-function mutations give rise to a diverse range of mutant phenotypes including disruption of the specification of R8 photoreceptor cell diversity. We demonstrate that hibris is required within the retina, and that hibris over-expression is sufficient to disrupt normal photoreceptor cell patterning. These findings suggest an additional layer of complexity in the signaling process that produces paired expression of opsin genes in adjacent R7 and R8 photoreceptor cells.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Retina/growth & development , Animals , Cell Differentiation , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Imaginal Discs/metabolism , Mutation , Organ Specificity , Photoreceptor Cells, Invertebrate/cytology , Retina/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL