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.
PLoS One ; 12(7): e0180280, 2017.
Article in English | MEDLINE | ID: mdl-28686657

ABSTRACT

BACKGROUND: There are no therapies shown to improve outcome after severe traumatic brain injury (TBI) in humans, a leading cause of morbidity and mortality. We sought to verify brain exposure of the systemically administered antioxidant N-acetylcysteine (NAC) and the synergistic adjuvant probenecid, and identify adverse effects of this drug combination after severe TBI in children. METHODS: IRB-approved, randomized, double-blind, placebo controlled Phase I study in children 2 to 18 years-of-age admitted to a Pediatric Intensive Care Unit after severe TBI (Glasgow Coma Scale [GCS] score ≤8) requiring an externalized ventricular drain for measurement of intracranial pressure (ICP). Patients were recruited from November 2011-August 2013. Fourteen patients (n = 7/group) were randomly assigned after obtaining informed consent to receive probenecid (25 mg/kg load, then 10 mg/kg/dose q6h×11 doses) and NAC (140 mg/kg load, then 70 mg/kg/dose q4h×17 doses), or placebos via naso/orogastric tube. Serum and CSF samples were drawn pre-bolus and 1-96 h after randomization and drug concentrations were measured via UPLC-MS/MS. Glasgow Outcome Scale (GOS) score was assessed at 3 months. RESULTS: There were no adverse events attributable to drug treatment. One patient in the placebo group was withdrawn due to adverse effects. In the treatment group, NAC concentrations ranged from 16,977.3±2,212.3 to 16,786.1±3,285.3 in serum and from 269.3±113.0 to 467.9±262.7 ng/mL in CSF, at 24 to 72 h post-bolus, respectively; and probenecid concentrations ranged from 75.4.3±10.0 to 52.9±25.8 in serum and 5.4±1.0 to 4.6±2.1 µg/mL in CSF, at 24 to 72 h post-bolus, respectively (mean±SEM). Temperature, mean arterial pressure, ICP, use of ICP-directed therapies, surveillance serum brain injury biomarkers, and GOS at 3 months were not different between groups. CONCLUSIONS: Treatment resulted in detectable concentrations of NAC and probenecid in CSF and was not associated with undesirable effects after TBI in children. TRIAL REGISTRATION: ClinicalTrials.gov NCT01322009.


Subject(s)
Acetylcysteine/pharmacokinetics , Adjuvants, Pharmaceutic/pharmacokinetics , Antioxidants/pharmacokinetics , Brain Injuries, Traumatic/drug therapy , Probenecid/pharmacokinetics , Acetylcysteine/blood , Acetylcysteine/cerebrospinal fluid , Acetylcysteine/pharmacology , Adjuvants, Pharmaceutic/pharmacology , Adolescent , Antioxidants/pharmacology , Biomarkers/blood , Body Temperature , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/cerebrospinal fluid , Brain Injuries, Traumatic/mortality , Child , Child, Preschool , Double-Blind Method , Drug Administration Schedule , Female , Glasgow Coma Scale , Glasgow Outcome Scale , Humans , Intracranial Pressure/drug effects , Intubation, Gastrointestinal , Male , Probenecid/blood , Probenecid/cerebrospinal fluid , Probenecid/pharmacology , Survival Analysis
2.
Neurotherapeutics ; 13(1): 217-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26572666

ABSTRACT

N-acetyl cysteine (NAC) supports the synthesis of glutathione (GSH), an essential substrate for fast, enzymatically catalyzed oxidant scavenging and protein repair processes. NAC is entering clinical trials for adrenoleukodystrophy, Parkinson's disease, schizophrenia, and other disorders in which oxidative stress may contribute to disease progression. However, these trials are hampered by uncertainty about the dose of NAC required to achieve biological effects in human brain. Here we describe an approach to this issue in which mice are used to establish the levels of NAC in cerebrospinal fluid (CSF) required to affect brain neurons. NAC dosing in humans can then be calibrated to achieve these NAC levels in human CSF. The mice were treated with NAC over a range of doses, followed by assessments of neuronal GSH levels and neuronal antioxidant capacity in ex vivo brain slices. Neuronal GSH levels and antioxidant capacity were augmented at NAC doses that produced peak CSF NAC concentrations of ≥50 nM. Oral NAC administration to humans produced CSF concentrations of up to 10 µM, thus demonstrating that oral NAC administration can surpass the levels required for biological activity in brain. Variations of this approach may similarly facilitate and rationalize drug dosing for other agents targeting central nervous system disorders.


Subject(s)
Acetylcysteine/administration & dosage , Glutathione/cerebrospinal fluid , Acetylcysteine/cerebrospinal fluid , Animals , Antioxidants/analysis , Brain Chemistry/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
3.
Parkinsonism Relat Disord ; 21(5): 500-3, 2015 May.
Article in English | MEDLINE | ID: mdl-25765302

ABSTRACT

INTRODUCTION: Depletion of neuronal glutathione may contribute to the pathogenesis of Parkinson's disease (PD). N-acetylcysteine (NAC) can restore neuronal glutathione levels, but it has not been established whether NAC can cross the blood-brain barrier in humans. METHODS: Twelve patients with PD were given oral NAC twice daily for 2 days. Three doses were compared: 7 mg/kg, 35 mg/kg, and 70 mg/kg. NAC, cysteine, and glutathione were measured in the cerebrospinal fluid (CSF) at baseline and 90 min after the last dose. Cognitive and motor functions were assessed pre- and post-NAC administration using the Montreal Cognitive Assessment (MoCA) and the Unified Parkinson's Disease Rating Scale part III motor subscore (UPDRS-III). RESULTS: Oral NAC produced a dose-dependent increase in CSF NAC concentrations (p < 0.001), with the highest dose producing a CSF concentration of 9.26 ± 1.62 µM. There were no significant adverse events. NAC had no acute effect on motor or cognitive function. CONCLUSION: Orally administered NAC produces biologically relevant CSF NAC concentrations at doses that are well tolerated. The findings support the feasibility of NAC as a potential disease-modifying therapy for PD.


Subject(s)
Acetylcysteine/administration & dosage , Acetylcysteine/cerebrospinal fluid , Parkinson Disease/cerebrospinal fluid , Parkinson Disease/drug therapy , Administration, Oral , Aged , Aged, 80 and over , Biomarkers/cerebrospinal fluid , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged
4.
J Neurochem ; 115(1): 178-87, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20649841

ABSTRACT

Recent findings suggest that a defect in the ubiquitin-proteasome system plays an important role in the pathogenesis of Parkinson's disease (PD). A previous report (McNaught et al. 2004) demonstrated that rats systemically injected with proteasome inhibitors exhibited PD-like clinical symptoms and pathology. However, because these findings have not been consistently replicated, this model is not commonly used to study PD. We used medaka fish to test the effect of systemic administration of proteasome inhibitors because of the high level of accessibility of the cerebrospinal fluid in fish. We injected lactacystin or epoxomicin into the CSF of medaka. With proteasome inhibition in the medaka brain, selective dopaminergic and noradrenergic cell loss was observed. Furthermore, treated fish exhibited reduced spontaneous movement. Treatment with proteasome inhibitors also induced the formation of inclusion bodies resembling Lewy bodies, which are characteristic of PD. Treatment with 6-OHDA also induced dopaminergic cell loss but did not produce inclusion bodies. These findings in medaka are consistent with previous results reporting that non-selective proteasome inhibition replicates the cardinal features of PD: locomotor dysfunction, selective dopaminergic cell loss, and inclusion body formation.


Subject(s)
Brain Chemistry/drug effects , Brain Chemistry/physiology , Oryzias/physiology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Proteasome Inhibitors , Acetylcysteine/analogs & derivatives , Acetylcysteine/cerebrospinal fluid , Acetylcysteine/pharmacology , Animals , Behavior, Animal/drug effects , Blotting, Western , Cysteine Proteinase Inhibitors/cerebrospinal fluid , Cysteine Proteinase Inhibitors/pharmacology , Dopamine/physiology , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/physiopathology , Immunohistochemistry , Inclusion Bodies/pathology , Microscopy, Electron, Transmission , Neurons/drug effects , Neurons/metabolism , Norepinephrine/physiology , Oxidopamine/administration & dosage , Oxidopamine/cerebrospinal fluid , Oxidopamine/pharmacology , Parkinson Disease, Secondary/psychology , Swimming/physiology , Sympatholytics/administration & dosage , Sympatholytics/cerebrospinal fluid , Sympatholytics/pharmacology , Ubiquitination/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL