Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurotrauma ; 36(13): 2139-2146, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30704349

ABSTRACT

Hypotension worsens outcome after all severities of traumatic brain injury (TBI), with loss of cerebral autoregulation being a potential contributor. Previously, we demonstrated that intravenous injection of a high capacity catalytic antioxidant, poly(ethylene)glycol conjugated hydrophilic carbon clusters (PEG-HCCs) rapidly restored cerebral perfusion and acutely restored brain oxidative balance in a TBI model complicated by hemorrhagic hypotension without evidence of toxicity. Here, we tested whether these acute effects translated into behavioral and structural benefit. TBI was generated by a cortical contusion impactor in 38 Long Evans rats, followed by blood withdrawal to a target mean arterial pressure of 40 mm Hg. PEG-HCC (2 mg/kg) or diluent was injected intravenously 80 min later at the onset of blood resuscitation followed by another injection 2 h later (doses determined in prior studies). Performance on beam walking (performed on days 1-5) and Morris water maze (MWM) (performed on days 11-15) was tested, and lesion size was determined at the termination. PEG-HCC treatment nearly completely prevented motor dysfunction (p < 0.001 vs. diluent), improved MWM performance (p < 0.001; treatment vs. time interaction) and reduced lesion size by 61% (p = 0.054). Here we show that treatment with PEG-HCCs at a clinically realistic time point (onset of resuscitation) prevented a major portion of the neurological dysfunction induced in this TBI model, and that PEG-HCCs are candidates for additional study as a potential therapeutic agent.


Subject(s)
Antioxidants/pharmacology , Brain Injuries, Traumatic , Carbon/pharmacology , Nanoparticles , Polyethylene Glycols/pharmacology , Animals , Antioxidants/chemistry , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Hypotension/complications , Nanoparticles/chemistry , Random Allocation , Rats , Rats, Long-Evans , Recovery of Function/drug effects , Resuscitation
2.
J Cereb Blood Flow Metab ; 32(3): 515-24, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22086196

ABSTRACT

Loss of plasma membrane integrity is a feature of acute cellular injury/death in vitro and in vivo. Plasmalemma-resealing agents are protective in acute central nervous system injury models, but their ability to reseal cell membranes in vivo has not been reported. Using a mouse controlled cortical impact (CCI) model, we found that propidium iodide-positive (PI+) cells pulse labeled at 6, 24, or 48 hours maintained a degenerative phenotype and disappeared from the injured brain by 7 days, suggesting that plasmalemma permeability is a biomarker of fatal cellular injury after CCI. Intravenous or intracerebroventricular administration of Kollidon VA64, poloxamer P188, or polyethylene glycol 8000 resealed injured cell membranes in vivo (P<0.05 versus vehicle or poloxamer P407). Kollidon VA64 (1 mmol/L, 500 µL) administered intravenously to mice 1 hour after CCI significantly reduced acute cellular degeneration, chronic brain tissue damage, brain edema, blood-brain barrier damage, and postinjury motor deficits (all P<0.05 versus vehicle). However, VA64 did not rescue pulse-labeled PI+ cells from eventual demise. We conclude that PI permeability within 48 hours of CCI is a biomarker of eventual cell death/loss. Kollidon VA64 reduces secondary damage after CCI by mechanisms other than or in addition to resealing permeable cells.


Subject(s)
Brain Injuries/prevention & control , Cell Membrane Permeability , Cell Membrane/drug effects , Cerebral Cortex/pathology , Pyrrolidines/therapeutic use , Vinyl Compounds/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Death/drug effects , Cell Membrane/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/injuries , Disease Models, Animal , Injections, Intravenous , Injections, Intraventricular , Magnetic Resonance Imaging , Male , Mice , Motor Activity/drug effects , Motor Activity/physiology , Pyrrolidines/administration & dosage , Vinyl Compounds/administration & dosage
3.
J Cereb Blood Flow Metab ; 29(1): 87-97, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18714331

ABSTRACT

Earlier experiments have shown that cyclosporin A (CsA) and its non-calcineurin inhibitory analog NIM811 attenuate mitochondrial dysfunction after experimental traumatic brain injury (TBI). Presently, we compared the neuroprotective effects of previously determined mitochondrial protective doses of CsA (20 mg/kg intraperitoneally) and NIM811 (10 mg/kg intraperitoneally) when administered at 15 mins postinjury in preventing cytoskeletal (alpha-spectrin) degradation, neurodegeneration, and neurological dysfunction after severe (1.0 mm) controlled cortical impact (CCI) TBI in mice. In a first set of experiments, we analyzed calpain-mediated alpha-spectrin proteolysis at 24 h postinjury. Both NIM811 and CsA significantly attenuated the increased alpha-spectrin breakdown products observed in vehicle-treated animals (P<0.005). In a second set of experiments, treatment of animals with either NIM811 or CsA at 15 mins and again at 24 h postinjury attenuated motor function impairment at 48 h and 7 days (P<0.005) and neurodegeneration at 7 days postinjury (P<0.0001). Delayed administration of NIM811 out to 12 h was still able to significantly reduce alpha-spectrin degradation. These results show that the neuroprotective mechanism of CsA involves maintenance of mitochondrial integrity and that calcineurin inhibition plays little or no role because the non-calcineurin inhibitory analog, NIM811, is as effective as CsA.


Subject(s)
Brain Injuries/prevention & control , Cyclosporine/chemistry , Cyclosporine/pharmacology , Neuroprotective Agents/pharmacology , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Calpain/metabolism , Cytoskeleton/drug effects , Cytoskeleton/enzymology , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Male , Mice , Motor Activity/drug effects , Neuroprotective Agents/chemistry , Physical Conditioning, Animal , Spectrin/metabolism
4.
J Cereb Blood Flow Metab ; 26(11): 1407-18, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16538231

ABSTRACT

In the present study, we investigate the hypothesis that mitochondrial oxidative damage and dysfunction precede the onset of neuronal loss after controlled cortical impact traumatic brain injury (TBI) in mice. Accordingly, we evaluated the time course of post-traumatic mitochondrial dysfunction in the injured cortex and hippocampus at 30 mins, 1, 3, 6, 12, 24, 48, and 72 h after severe TBI. A significant decrease in the coupling of the electron transport system with oxidative phosphorylation was observed as early as 30 mins after injury, followed by a recovery to baseline at 1 h after injury. A statistically significant (P<0.0001) decline in the respiratory control ratio was noted at 3 h, which persisted at all subsequent time-points up to 72 h after injury in both cortical and hippocampal mitochondria. Structural damage seen in purified cortical mitochondria included severely swollen mitochondria, a disruption of the cristae and rupture of outer membranes, indicative of mitochondrial permeability transition. Consistent with this finding, cortical mitochondrial calcium-buffering capacity was severely compromised by 3 h after injury, and accompanied by significant increases in mitochondrial protein oxidation and lipid peroxidation. A possible causative role for reactive nitrogen species was suggested by the rapid increase in cortical mitochondrial 3-nitrotyrosine levels shown as early as 30 mins after injury. These findings indicate that post-traumatic oxidative lipid and protein damage, mediated in part by peroxynitrite, occurs in mitochondria with concomitant ultrastructural damage and impairment of mitochondrial bioenergetics. The data also indicate that compounds which specifically scavenge peroxynitrite (ONOO(-)) or ONOO(-)-derived radicals (e.g. ONOO(-)+H(+) --> ONOOH --> (*)NO(2)+(*)OH) may be particularly effective for the treatment of TBI, although the therapeutic window for this neuroprotective approach might only be 3 h.


Subject(s)
Brain Hemorrhage, Traumatic/drug therapy , Brain Hemorrhage, Traumatic/pathology , Mitochondria/pathology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Animals , Blotting, Northern , Brain Hemorrhage, Traumatic/metabolism , Calpain/physiology , Cytoskeleton/pathology , Male , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Mice , Microscopy, Electron , Mitochondria/metabolism , Nerve Degeneration , Oxygen Consumption/physiology , Peroxynitrous Acid/metabolism , Reactive Oxygen Species , Tyrosine/analogs & derivatives , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...