Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
2.
Sci Rep ; 8(1): 3481, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29472564

ABSTRACT

The interaction of light with biological tissue has been successfully utilized for multiple therapeutic purposes. Previous studies have suggested that near infrared light (NIR) enhances the activity of mitochondria by increasing cytochrome c oxidase (COX) activity, which we confirmed for 810 nm NIR. In contrast, scanning the NIR spectrum between 700 nm and 1000 nm revealed two NIR wavelengths (750 nm and 950 nm) that reduced the activity of isolated COX. COX-inhibitory wavelengths reduced mitochondrial respiration, reduced the mitochondrial membrane potential (ΔΨm), attenuated mitochondrial superoxide production, and attenuated neuronal death following oxygen glucose deprivation, whereas NIR that activates COX provided no benefit. We evaluated COX-inhibitory NIR as a potential therapy for cerebral reperfusion injury using a rat model of global brain ischemia. Untreated animals demonstrated an 86% loss of neurons in the CA1 hippocampus post-reperfusion whereas inhibitory NIR groups were robustly protected, with neuronal loss ranging from 11% to 35%. Moreover, neurologic function, assessed by radial arm maze performance, was preserved at control levels in rats treated with a combination of both COX-inhibitory NIR wavelengths. Taken together, our data suggest that COX-inhibitory NIR may be a viable non-pharmacologic and noninvasive therapy for the treatment of cerebral reperfusion injury.


Subject(s)
Brain Injuries/radiotherapy , Electron Transport Complex IV/genetics , Infrared Rays/therapeutic use , Reperfusion Injury/radiotherapy , Animals , Brain/pathology , Brain/radiation effects , Brain Injuries/genetics , Brain Injuries/pathology , Electron Transport Complex IV/radiation effects , Glucose/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/radiation effects , Humans , Membrane Potential, Mitochondrial , Mitochondria/genetics , Mitochondria/radiation effects , Neurons/metabolism , Neurons/radiation effects , Oxidation-Reduction/radiation effects , Rats , Reperfusion Injury/genetics , Reperfusion Injury/pathology
3.
Mol Cell Neurosci ; 76: 68-75, 2016 10.
Article in English | MEDLINE | ID: mdl-27567688

ABSTRACT

Global brain ischemia/reperfusion induces neuronal damage in vulnerable brain regions, leading to mitochondrial dysfunction and subsequent neuronal death. Induction of neuronal death is mediated by release of cytochrome c (cyt c) from the mitochondria though a well-characterized increase in outer mitochondrial membrane permeability. However, for cyt c to be released it is first necessary for cyt c to be liberated from the cristae junctions which are gated by Opa1 oligomers. Opa1 has two known functions: maintenance of the cristae junction and mitochondrial fusion. These roles suggest that Opa1 could play a central role in both controlling cyt c release and mitochondrial fusion/fission processes during ischemia/reperfusion. To investigate this concept, we first utilized in vitro real-time imaging to visualize dynamic changes in mitochondria. Oxygen-glucose deprivation (OGD) of neurons grown in culture induced a dual-phase mitochondrial fragmentation profile: (i) fragmentation during OGD with no apoptosis activation, followed by fusion of mitochondrial networks after reoxygenation and a (ii) subsequent extensive fragmentation and apoptosis activation that preceded cell death. We next evaluated changes in mitochondrial dynamic state during reperfusion in a rat model of global brain ischemia. Evaluation of mitochondrial morphology with confocal and electron microscopy revealed a similar induction of fragmentation following global brain ischemia. Mitochondrial fragmentation aligned temporally with specific apoptotic events, including cyt c release, caspase 3/7 activation, and interestingly, release of the fusion protein Opa1. Moreover, we uncovered evidence of loss of Opa1 complexes during the progression of reperfusion, and electron microscopy micrographs revealed a loss of cristae architecture following global brain ischemia. These data provide novel evidence implicating a temporal connection between Opa1 alterations and dysfunctional mitochondrial dynamics following global brain ischemia.


Subject(s)
Brain Ischemia/metabolism , Mitochondrial Dynamics , Reperfusion Injury/metabolism , Animals , Apoptosis , CA1 Region, Hippocampal/blood supply , CA1 Region, Hippocampal/metabolism , Cell Hypoxia , Cell Line , Cells, Cultured , GTP Phosphohydrolases/metabolism , Male , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...