Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Neurol ; 257: 95-105, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24805071

ABSTRACT

Mitochondrial dysfunction is becoming a pivotal target for neuroprotective strategies following contusion spinal cord injury (SCI) and the pharmacological compounds that maintain mitochondrial function confer neuroprotection and improve long-term hindlimb function after injury. In the current study we evaluated the efficacy of cell-permeating thiol, N-acetylcysteine amide (NACA), a precursor of endogenous antioxidant glutathione (GSH), on mitochondrial function acutely, and long-term tissue sparing and hindlimb locomotor recovery following upper lumbar contusion SCI. Some designated injured adult female Sprague-Dawley rats (n=120) received either vehicle or NACA (75, 150, 300 or 600mg/kg) at 15min and 6h post-injury. After 24h the total, synaptic, and non-synaptic mitochondrial populations were isolated from a single 1.5cm spinal cord segment (centered at injury site) and assessed for mitochondrial bioenergetics. Results showed compromised total mitochondrial bioenergetics following acute SCI that was significantly improved with NACA treatment in a dose-dependent manner, with maximum effects at 300mg/kg (n=4/group). For synaptic and non-synaptic mitochondria, only 300mg/kg NACA dosage showed efficacy. Similar dosage (300mg/kg) also maintained mitochondrial GSH near normal levels. Other designated injured rats (n=21) received continuous NACA (150 or 300mg/kg/day) treatment starting at 15min post-injury for one week to assess long-term functional recovery over 6weeks post-injury. Locomotor testing and novel gait analyses showed significantly improved hindlimb function with NACA that were associated with increased tissue sparing at the injury site. Overall, NACA treatment significantly maintained acute mitochondrial bioenergetics and normalized GSH levels following SCI, and prolonged delivery resulted in significant tissue sparing and improved recovery of hindlimb function.


Subject(s)
Acetylcysteine/analogs & derivatives , Energy Metabolism/drug effects , Mitochondria/drug effects , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Acetylcysteine/therapeutic use , Animals , Disease Models, Animal , Double-Blind Method , Drug Delivery Systems , Electron Transport Chain Complex Proteins/metabolism , Female , Ganglia, Spinal/drug effects , Ganglia, Spinal/pathology , Lameness, Animal/drug therapy , Lameness, Animal/etiology , Mitochondria/enzymology , Motor Activity/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord/ultrastructure , Spinal Cord Injuries/complications , Synapses/drug effects , Synapses/enzymology , Synapses/pathology , Synapses/ultrastructure , Time Factors
2.
Exp Neurol ; 257: 106-13, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24792639

ABSTRACT

Traumatic brain injury (TBI) has become a growing epidemic but no approved pharmacological treatment has been identified. Our previous work indicates that mitochondrial oxidative stress/damage and loss of bioenergetics play a pivotal role in neuronal cell death and behavioral outcome following experimental TBI. One tactic that has had some experimental success is to target glutathione using its precursor N-acetylcysteine (NAC). However, this approach has been hindered by the low CNS bioavailability of NAC. The current study evaluated a novel, cell permeant amide form of N-acetylcysteine (NACA), which has high permeability through cellular and mitochondrial membranes resulting in increased CNS bioavailability. Cortical tissue sparing, cognitive function and oxidative stress markers were assessed in rats treated with NACA, NAC, or vehicle following a TBI. At 15days post-injury, animals treated with NACA demonstrated significant improvements in cognitive function and cortical tissue sparing compared to NAC or vehicle treated animals. NACA treatment also was shown to reduce oxidative damage (HNE levels) at 7days post-injury. Mechanistically, post-injury NACA administration was demonstrated to maintain levels of mitochondrial glutathione and mitochondrial bioenergetics comparable to sham animals. Collectively these data provide a basic platform to consider NACA as a novel therapeutic agent for treatment of TBI.


Subject(s)
Acetylcysteine/analogs & derivatives , Brain Injuries/complications , Brain Injuries/drug therapy , Energy Metabolism/drug effects , Maze Learning/drug effects , Neuroprotective Agents/therapeutic use , Acetylcysteine/therapeutic use , Aldehydes/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Disease Models, Animal , Double-Blind Method , Glutathione/metabolism , Male , Mitochondria/drug effects , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...