Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
BMC Neurosci ; 21(1): 22, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32404052

RESUMEN

BACKGROUND: Polynitroxylated PEGylated hemoglobin (PNPH, aka SanFlow) possesses superoxide dismutase/catalase mimetic activities that may directly protect the brain from oxidative stress. Stabilization of PNPH with bound carbon monoxide prevents methemoglobin formation during storage and permits it to serve as a carbon monoxide donor. We determined whether small volume transfusion of hyperoncotic PNPH is neuroprotective in a polytrauma model of traumatic brain injury (TBI) plus hemorrhagic shock. Guinea pigs were used because, like humans, they do not synthesize their own ascorbic acid, which is important in reducing methemoglobin. RESULTS: TBI was produced by controlled cortical impact and was followed by 20 mL/kg hemorrhage to a mean arterial pressure (MAP) of 40 mmHg. At 90 min, animals were resuscitated with 20 mL/kg lactated Ringer's solution or 10 mL/kg PNPH. Resuscitation with PNPH significantly augmented the early recovery of MAP after hemorrhagic shock by 10-18 mmHg; whole blood methemoglobin was only 1% higher and carboxyhemoglobin was 2% higher. At 9 days of recovery, unbiased stereology analysis revealed that, compared to animals resuscitated with lactated Ringer's solution, those treated with PNPH had significantly more viable neurons in the hippocampus CA1 + 2 region (59 ± 10% versus 87 ± 18% of sham and naïve mean value) and in the dentate gyrus (70 ± 21% versus 96 ± 24%; n = 12 per group). CONCLUSION: PNPH may serve as a small-volume resuscitation fluid for polytrauma involving TBI and hemorrhagic shock. The neuroprotection afforded by PNPH seen in other species was sustained in a species without endogenous ascorbic acid synthesis, thereby supporting potential translatability for human use.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Catalasa/farmacología , Hemorragia/tratamiento farmacológico , Resucitación , Choque Hemorrágico/tratamiento farmacológico , Animales , Cobayas , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología
2.
J Bioenerg Biomembr ; 47(1-2): 43-51, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25348484

RESUMEN

Progesterone has been studied extensively in preclinical models of adult traumatic brain injury (TBI), and has advanced to clinical trials in adults with TBI. However, there are very few preclinical studies in pediatric TBI models investigating progesterone for neuroprotection. Immature male and female rats (postnatal day, PND 17-21) underwent controlled cortical impact (CCI) to the left parietal cortex. Rats received either progesterone (10 mg/kg) at 1 h (i.p.) and 6 h (s.c.) after TBI or vehicle (22.5 % cyclohexdrin), and were compared to naïve, age-matched littermates. At 24 h after CCI, brain mitochondria were isolated from the ipsilateral hemisphere. Active (State 3) and resting (State 4) mitochondrial respiration were measured, and mitochondrial respiratory control ratio (RCR, State 3/State 4) was determined. Total mitochonidral glutathione content was measured. A separate group of rats were studied for histology, and received progesterone or vehicle every 24 h (s.c.) for 7 days. In male rats, TBI reduced mitochondrial RCR, and progesterone preserved mitochondrial RCR. This improvement of RCR was predominantly through significant decreases in State 4 respiratory rates. In female rats, post-injury treatment with progesterone did not significantly improve mitochondrial RCR. Normal (uninjured) male rats had lower mitochondrial glutathione content than normal female rats. After TBI, progesterone prevented loss of mitochondrial glutathione in male rats only. Tissue loss was reduced in progesterone treated female rats at 7d after CCI. Future studies will be directed at correlation with neurologic outcome testing. These preclinical studies could provide information for planning future clinical trials of progesterone treatment in children with TBI.


Asunto(s)
Lesiones Encefálicas/metabolismo , Mitocondrias/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Progesterona/farmacología , Progestinas/farmacología , Caracteres Sexuales , Animales , Lesiones Encefálicas/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Glutatión/metabolismo , Humanos , Masculino , Mitocondrias/patología , Ratas , Ratas Sprague-Dawley
3.
J Neurotrauma ; 40(11-12): 1197-1215, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36416234

RESUMEN

Therapies are limited for pediatric traumatic brain injury (TBI), especially for the very young who can experience long-term consequences to learning, memory, and social behavior. Animal models of pediatric TBI have yielded mechanistic insights, but demonstration of clinically relevant long-term behavioral and/or cognitive deficits has been challenging. We characterized short- and long-term outcomes in a controlled cortical impact (CCI) model of pediatric TBI using a panel of tests between 2 weeks and ∼4 months after injury. Male rats with CCI at postnatal Day (PND) 10 were compared with three control groups: Naïve, Anesthesia, and Craniotomy. Motor testing (PND 25-33), novel object recognition (NOR; PND 40-50), and multiple tasks in water maze (WM; PND 65-100) were followed by social interaction tests (PND 120-140). Anesthesia rats performed the same as Naïve rats in all tasks. TBI rats, when compared with Naïve controls, had functional impairments across most tests studied. The most sensitive cognitive processes affected by TBI included those that required fast one-trial learning (NOR, WM), flexibility of acquired memory traces (reversals in WM), response strategies (WM), or recognition memory in the setting of reciprocal social interactions. Both TBI and Craniotomy groups demonstrated increased rates of decision making across several WM tasks, suggesting disinhibition of motor responses. When the TBI group was compared with the Craniotomy group, however, deficits were detected in a limited number of outcomes. The latter included learning speed (WM), cognitive flexibility (WM), and social recognition memory. Notably, effects of craniotomy, when compared with Naïve controls, spanned across multiple tasks, and in some tasks, could reach the effect sizes observed in TBI. These results highlight the importance of appropriate control groups in pediatric CCI models. In addition, the study demonstrates the high sensitivity of comprehensive cognitive testing to detect long-term effects of early-age craniotomy and TBI and provides a template for future testing of experimental therapies.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Ratas , Animales , Masculino , Ratas Sprague-Dawley , Grupos Control , Aprendizaje por Laberinto/fisiología , Lesiones Traumáticas del Encéfalo/complicaciones , Cognición , Modelos Animales de Enfermedad
4.
J Neurotrauma ; 37(14): 1656-1667, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32079496

RESUMEN

Young children who have sustained severe traumatic brain injury (TBI) can suffer from debilitating neurocognitive deficits. Impairment of adult hippocampal neurogenesis is associated with cognitive deficits and depression. Very few studies have investigated the adult hippocampal neurogenesis after pediatric TBI. Here, we evaluated long-term cognition, adult hippocampal neurogenesis, and microglial activation in a rabbit pediatric TBI model. On Post-natal Day 5-7 (P5-7), New Zealand white rabbits from the same litter were randomized into naïve, sham (craniotomy alone), and TBI (controlled cortical impact). Bromodeoxyuridine (BrdU, 50 mg/kg, intraperitoneally) was administered at 1-month post-injury, once/daily for 5 consecutive days. Novel object recognition and spontaneous alternation in T-maze tests were performed at 2 months post-injury to measure the cognitive functions. The animals were euthanized after behavioral tests at 3 months of age to evaluate adult hippocampal neurogenesis and microglial activation. We found that: 1) pediatric TBI caused significant deficits in hippocampal dependent cognitive functions; 2) the survival rates of adult-born neurons at both ipsilateral and contralateral hippocampus significantly decreased in the TBI group; 3) TBI induced ectopic migration of adult-born neurons at the dorsal dentate gyrus in both ipsilateral and contralateral hippocampus; 4) TBI increased astrogenesis in the hilus of the dentate gyrus; and 5) TBI results in abnormal microglial activation. In conclusion, pediatric TBI causes prolonged neuroinflammation and dysregulation of the adult hippocampal neurogenesis through young adulthood, which might be responsible for the cognitive deficits. Protection of adult hippocampal neurogenesis may potentially improve outcomes.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Trastornos del Conocimiento/patología , Hipocampo/patología , Neurogénesis/fisiología , Factores de Edad , Animales , Lesiones Traumáticas del Encéfalo/psicología , Trastornos del Conocimiento/psicología , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Conejos
5.
Neuroscience ; 406: 202-211, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-30849447

RESUMEN

Neonatal hypoxic ischemic (HI) brain injury causes lifelong neurologic disability. Therapeutic hypothermia (TH) is the only approved therapy that partially mitigates mortality and morbidity. Therapies specifically targeting HI-induced brain cell death are currently lacking. Intracellular calcium dysregulation, oxidative stress, and mitochondrial dysfunction through the formation of the mitochondrial permeability transition pore (mPTP) are drivers of HI cellular injury. GNX-4728, a small molecule direct inhibitor of the mPTP that increases mitochondrial calcium retention capacity, is highly effective in adult neurodegenerative disease models and could have potential as a therapy in neonatal HI. A dose of GNX-4728, equivalent to that used in animal models, 300 mg/kg, IP was highly toxic in p10 mice. We then tested the hypothesis that acute administration of 30 mg/kg, IP of GNX-4728 immediately after HI in a neonatal mouse model would provide neuroprotection. This non-lethal lower dose of GNX-4728 (30 mg/kg, IP) improved the respiratory control ratio of neonatal female HI brain tissue but not in males. Brain injury, assessed histologically with a novel metric approach at 1 and 30 days after HI, was not mitigated by GNX-4728. Our work demonstrates that a small molecule inhibitor of the mPTP has i) an age related toxicity, ii) a sex-related brain mitoprotective profile after HI but iii) this is not sufficient to attenuate forebrain HI neuropathology.


Asunto(s)
Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Animales , Animales Recién Nacidos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Poro de Transición de la Permeabilidad Mitocondrial , Fármacos Neuroprotectores/farmacología , Resultado del Tratamiento
6.
J Neurotrauma ; 25(6): 603-14, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18454682

RESUMEN

Although studies have shown alterations in cerebral metabolism after traumatic brain injury (TBI), clinical data in the developing brain is limited. We hypothesized that post-traumatic metabolic changes occur early (<24 h) and persist for up to 1 week. Immature rats underwent TBI to the left parietal cortex. Brains were removed at 4 h, 24 h, and 7 days after injury, and separated into ipsilateral (injured) and contralateral (control) hemispheres. Proton nuclear magnetic resonance (NMR) spectra were obtained, and spectra were analyzed for N-acetyl-aspartate (NAA), lactate (Lac), creatine (Cr), choline, and alanine, with metabolite ratios determined (NAA/Cr, Lac/Cr). There were no metabolic differences at any time in sham controls between cerebral hemispheres. At 4 and 24 h, there was an increase in Lac/Cr, reflecting increased glycolysis and/or decreased oxidative metabolism. At 24 h and 7 days, there was a decrease in NAA/Cr, indicating loss of neuronal integrity. The NAA/Lac ratio was decreased ( approximately 15-20%) at all times (4 h, 24 h, 7 days) in the injured hemisphere of TBI rats. In conclusion, metabolic derangements begin early (<24 h) after TBI in the immature rat and are sustained for up to 7 days. Evaluation of early metabolic alterations after TBI could identify novel targets for neuroprotection in the developing brain.


Asunto(s)
Envejecimiento/metabolismo , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Alanina/análisis , Alanina/metabolismo , Animales , Animales Recién Nacidos , Ácido Aspártico/análogos & derivados , Ácido Aspártico/análisis , Ácido Aspártico/metabolismo , Encéfalo/crecimiento & desarrollo , Lesiones Encefálicas/patología , Respiración de la Célula , Colina/análisis , Colina/metabolismo , Creatina/análisis , Creatina/metabolismo , Progresión de la Enfermedad , Glucólisis , Ácido Láctico/análisis , Ácido Láctico/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Mitocondrias/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Fosforilación Oxidativa , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
7.
J Neurotrauma ; 2018 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-30019623

RESUMEN

Neuroinflammation after traumatic brain injury (TBI) contributes to widespread cell death and tissue loss. Here, we evaluated sequential inflammatory response in the brain, as well as inflammation-induced changes in brain tryptophan metabolism over time, in a rabbit pediatric TBI model. On post-natal days 5-7 (P5-P7), New Zealand white rabbit littermates were randomized into three groups: naïve (no injury), sham (craniotomy alone), and TBI (controlled cortical impact). Animals were sacrificed at 6 h and 1, 3, 7, and 21 days post-injury for evaluating levels of pro- and anti-inflammatory cytokines, as well as the major components in the tryptophan-kynurenine pathway. We found that 1) pro- and anti-inflammatory cytokine levels in the brain injury area were differentially regulated in a time-dependent manner post-injury; 2) indoleamine 2,3 dioxygeenase 1 (IDO1) was upregulated around the injury area in TBI kits that persisted at 21 days post-injury; 3) mean length of serotonin-staining fibers was significantly reduced in the injured brain region in TBI kits for at least 21 days post-injury; and 4) kynurenine level significantly increased at 7 days post-injury. A significant decrease in serotonin/tryptophan ratio and melatonin/tryptophan ratio at 21 days post-injury was noted, suggesting that tryptophan metabolism is altered after TBI. A better understanding of the temporal evolution of immune responses and tryptophan metabolism during injury and repair after TBI is crucial for the development of novel therapeutic strategies targeting these pathways.

8.
J Comp Neurol ; 487(1): 1-14, 2005 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15861462

RESUMEN

Bex proteins are expressed from a family of "brain expressed X-linked genes" that are closely linked on the X-chromosome. Bex1 and 2 have been characterized as interacting partners of the olfactory marker protein (OMP). Here we report the distribution of Bex1 and Bex2 mRNAs in several brain regions and the development and characterization of an antibody to mouse Bex1 protein that cross-reacts with Bex2 (but not Bex3), and its use to determine the cellular distribution of Bex proteins in the murine brain. The specificity of the antiserum was characterized by immunoprecipitation and Western blots of tissue and transfected cell extracts and by immunocytochemical analyses of cells transfected with either Bex1 or Bex2. Antibodies preabsorbed with Bex2 still recognize Bex1, while blocking with Bex1 eliminates all immunoreactivity to both Bex1 and Bex2. Bex immunoreactivity (ir) was primarily localized to neuronal cells within several regions of the brain, including the olfactory epithelium, bulb, peri/paraventricular nuclei, suprachiasmatic nucleus, arcuate nucleus, median eminence, lateral hypothalamic area, thalamus, hippocampus, and cerebellum. RT-PCR and in situ hybridization demonstrated the presence of Bex mRNA in several of these regions. Double-label immunocytochemistry indicates that Bex-ir is colocalized with OMP in mature olfactory receptor neurons (ORNs) and in the OMP-positive subpopulation of neurons in hypothalamus. This is the first anatomical mapping of Bex proteins in the mouse brain and their colocalization with OMP in ORNs and hypothalamus.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Vías Olfatorias/metabolismo , Factores de Edad , Secuencia de Aminoácidos , Animales , Encéfalo/citología , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ratones , Proteínas del Tejido Nervioso/genética , Proteína Marcadora Olfativa , Mucosa Olfatoria/citología , Mucosa Olfatoria/metabolismo , Vías Olfatorias/citología , Neuronas Receptoras Olfatorias/metabolismo , Especificidad de Órganos , ARN Mensajero/análisis , Alineación de Secuencia
9.
J Neurotrauma ; 32(17): 1369-79, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25758339

RESUMEN

Traumatic brain injury (TBI) is a common cause of disability in childhood, resulting in numerous physical, behavioral, and cognitive sequelae, which can influence development through the lifespan. The mechanisms by which TBI influences normal development and maturation remain largely unknown. Pediatric rodent models of TBI often do not demonstrate the spectrum of motor and cognitive deficits seen in patients. To address this problem, we developed a New Zealand white rabbit model of pediatric TBI that better mimics the neurological injury seen after TBI in children. On postnatal Day 5-7 (P5-7), rabbits were injured by a controlled cortical impact (6-mm impactor tip; 5.5 m/sec, 2-mm depth, 50-msec duration). Rabbits from the same litter served as naïve (no injury) and sham (craniotomy alone) controls. Functional abilities and activity levels were measured 1 and 5 d after injury. Maturation level was monitored daily. We performed cognitive tests during P14-24 and sacrificed the animals at 1, 3, 7, and 21 d after injury to evaluate lesion volume and microglia. TBI kits exhibited delayed achievement of normal developmental milestones. They also demonstrated significant cognitive deficits, with lower percentage of correct alternation rate in the T-maze (n=9-15/group; p<0.001) and less discrimination between novel and old objects (p<0.001). Lesion volume increased from 16% at Day 3 to 30% at Day 7 after injury, indicating ongoing secondary injury. Activated microglia were noted at the injury site and also in white matter regions of the ipsilateral and contralateral hemispheres. The neurologic and histologic changes in this model are comparable to those reported clinically. Thus, this rabbit model provides a novel platform for evaluating neuroprotective therapies in pediatric TBI.


Asunto(s)
Conducta Animal/fisiología , Lesiones Encefálicas , Trastornos del Conocimiento , Modelos Animales de Enfermedad , Animales , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Femenino , Masculino , Microglía/metabolismo , Microglía/patología , Conejos
10.
J Neurotrauma ; 31(4): 395-403, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24050267

RESUMEN

The robustness of plasticity mechanisms during brain development is essential for synaptic formation and has a beneficial outcome after sensory deprivation. However, the role of plasticity in recovery after acute brain injury in children has not been well defined. Traumatic brain injury (TBI) is the leading cause of death and disability among children, and long-term disability from pediatric TBI can be particularly devastating. We investigated the altered cortical plasticity 2-3 weeks after injury in a pediatric rat model of TBI. Significant decreases in neurophysiological responses across the depth of the noninjured, primary somatosensory cortex (S1) in TBI rats, compared to age-matched controls, were detected with electrophysiological measurements of multi-unit activity (86.4% decrease), local field potential (75.3% decrease), and functional magnetic resonance imaging (77.6% decrease). Because the corpus callosum is a clinically important white matter tract that was shown to be consistently involved in post-traumatic axonal injury, we investigated its anatomical and functional characteristics after TBI. Indeed, corpus callosum abnormalities in TBI rats were detected with diffusion tensor imaging (9.3% decrease in fractional anisotropy) and histopathological analysis (14% myelination volume decreases). Whole-cell patch clamp recordings further revealed that TBI results in significant decreases in spontaneous firing rate (57% decrease) and the potential to induce long-term potentiation in neurons located in layer V of the noninjured S1 by stimulation of the corpus callosum (82% decrease). The results suggest that post-TBI plasticity can translate into inappropriate neuronal connections and dramatic changes in the function of neuronal networks.


Asunto(s)
Lesiones Encefálicas/patología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Plasticidad Neuronal/fisiología , Animales , Anisotropía , Encéfalo/fisiopatología , Lesiones Encefálicas/fisiopatología , Cuerpo Calloso/patología , Imagen de Difusión Tensora , Espacio Extracelular/fisiología , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Vías Nerviosas/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Corteza Somatosensorial/fisiología , Transmisión Sináptica/fisiología
11.
J Neurotrauma ; 30(24): 2066-72, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24032394

RESUMEN

Altered cerebral metabolism and mitochondrial function have been identified in experimental and clinical studies of pediatric traumatic brain injury (TBI). Metabolic changes detected using (1)H (proton) magnetic resonance spectroscopy correlate with long-term outcomes in children after severe TBI. We previously identified early (4-h) and sustained (24-h and 7-day) abnormalities in brain metabolites after controlled cortical impact (CCI) in immature rats. The current study aimed to identify specific alterations of cerebral glucose metabolism at 24 h after TBI in immature rats. Rats (postnatal days 16-18) underwent CCI to the left parietal cortex. Sham rats underwent craniotomy only. Twenty-four hours after CCI, rats were injected (intraperitoneally) with [1,6-(13)C]glucose. Brains were removed, separated into hemispheres, and frozen. Metabolites were extracted with perchloric acid and analyzed using (1)H and (13)C-nuclear magnetic resonance spectroscopy. TBI resulted in decreases in N-acetylaspartate in both hemispheres, compared to sham contralateral. At 24 h after TBI, there was significant decrease in the incorporation of (13)C label into [3-(13)C]glutamate and [2-(13)C]glutamate in the injured brain. There were no differences in percent enrichment of [3-(13)C]glutamate, [4-(13)C]glutamate, [3-(13)C]glutamine, or [4-(13)C]glutamine. There was significantly lower percent enrichment of [2-(13)C]glutamate in both TBI sides and the sham craniotomy side, compared to sham contralateral. No differences were detected in enrichment of (13)C glucose label in [2-(13)C]glutamine, [2-(13)C]GABA (gamma-aminobutyric acid), [3-(13)C]GABA, or [4-(13)C]GABA, [3-(13)C]lactate, or [3-(13)C]alanine between groups. Results suggest that overall oxidative glucose metabolism in the immature brain recovers at 24 h after TBI. Specific reductions in [2-(13)C]glutamate could be the result of impairments in either neuronal or astrocytic metabolism. Future studies should aim to identify pathways leading to decreased metabolism and develop cell-selective "metabolic rescue."


Asunto(s)
Lesiones Encefálicas/metabolismo , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Glucosa/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Lesiones Encefálicas/patología , Masculino , Ratas , Ratas Sprague-Dawley
12.
J Neurotrauma ; 28(5): 763-74, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21250918

RESUMEN

Cyclosporin A (CsA) has been shown to be neuroprotective in mature animal models of traumatic brain injury (TBI), but its effects on immature animal models of TBI are unknown. In mature animal models, CsA inhibits the opening of the mitochondrial permeability transition pore (MPTP), thereby maintaining mitochondrial homeostasis following injury by inhibiting calcium influx and preserving mitochondrial membrane potential. The aim of the present study was to evaluate CsA's ability to preserve mitochondrial bioenergetic function following TBI (as measured by mitochondrial respiration and cerebral microdialysis), in two immature models (focal and diffuse), and in two different species (rat and piglet). Three groups were studied: injured+CsA, injured+saline vehicle, and uninjured shams. In addition, we evaluated CsA's effects on cerebral hemodynamics as measured by a novel thermal diffusion probe. The results demonstrate that post-injury administration of CsA ameliorates mitochondrial dysfunction, preserves cerebral blood flow (CBF), and limits neuropathology in immature animals 24 h post-TBI. Mitochondria were isolated 24 h after controlled cortical impact (CCI) in rats and rapid non-impact rotational injury (RNR) in piglets, and CsA ameliorated cerebral bioenergetic crisis with preservation of the respiratory control ratio (RCR) to sham levels. Results were more dramatic in RNR piglets than in CCI rats. In piglets, CsA also preserved lactate pyruvate ratios (LPR), as measured by cerebral microdialysis and CBF at sham levels 24 h after injury, in contrast to the significant alterations seen in injured piglets compared to shams (p<0.01). The administration of CsA to piglets following RNR promoted a 42% decrease in injured brain volume (p<0.01). We conclude that CsA exhibits significant neuroprotective activity in immature models of focal and diffuse TBI, and has exciting translational potential as a therapeutic agent for neuroprotection in children.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Ciclosporina/uso terapéutico , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Animales , Lesiones Encefálicas/patología , Respiración de la Célula/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Femenino , Inmunosupresores/uso terapéutico , Microdiálisis , Ratas , Ratas Sprague-Dawley , Porcinos
13.
J Neurochem ; 101(5): 1248-57, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17403141

RESUMEN

Mitochondria play central roles in acute brain injury; however, little is known about mitochondrial function following traumatic brain injury (TBI) to the immature brain. We hypothesized that TBI would cause mitochondrial dysfunction early (<4 h) after injury. Immature rats underwent controlled cortical impact (CCI) or sham injury to the left cortex, and mitochondria were isolated from both hemispheres at 1 and 4 h after TBI. Rates of phosphorylating (State 3) and resting (State 4) respiration were measured with and without bovine serum albumin. The respiratory control ratio was calculated (State 3/State 4). Rates of mitochondrial H(2)O(2) production, pyruvate dehydrogenase complex enzyme activity, and cytochrome c content were measured. Mitochondrial State 4 rates (ipsilateral/contralateral ratios) were higher after TBI at 1 h, which was reversed with bovine serum albumin. Four hours after TBI, pyruvate dehydrogenase complex activity and cytochrome c content (ipsilateral/contralateral ratios) were lower in TBI mitochondria. These data demonstrate abnormal mitochondrial function early (

Asunto(s)
Lesiones Encefálicas/patología , Mitocondrias/fisiología , Análisis de Varianza , Animales , Animales Recién Nacidos , Células Cultivadas , Corteza Cerebral/patología , Corteza Cerebral/ultraestructura , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Hipocampo/patología , Hipocampo/ultraestructura , Cetona Oxidorreductasas/metabolismo , Masculino , Mitocondrias/patología , Consumo de Oxígeno , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
14.
Exp Neurol ; 197(1): 235-43, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16259981

RESUMEN

Growing literature suggests important sex-based differences in outcome following traumatic brain injury (TBI) in animals and humans. Progesterone has emerged as a key hormone involved in many potential neuroprotective pathways after acute brain injury and may be responsible for some of these differences. Many studies have utilized supraphysiologic levels of post-traumatic progesterone to reverse pathologic processes after TBI, but few studies have focused on the role of endogenous physiologic levels of progesterone in neuroprotection. We hypothesized that progesterone at physiologic serum levels would be neuroprotective in female rats after TBI and that progesterone would reverse early mitochondrial dysfunction seen in this model. Female, Sprague-Dawley rats were ovariectomized and implanted with silastic capsules containing either low or high physiologic range progesterone at 7 days prior to TBI. Control rats received ovariectomy with implants containing no hormone. Rats underwent controlled cortical impact to the left parietotemporal cortex and were evaluated for evidence of early mitochondrial dysfunction (1 h) and delayed hippocampal neuronal injury and cortical tissue loss (7 days) after injury. Progesterone in the low physiologic range reversed the early postinjury alterations seen in mitochondrial respiration and reduced hippocampal neuronal loss in both the CA1 and CA3 subfields. Progesterone in the high physiologic range had a more limited pattern of hippocampal neuronal preservation in the CA3 region only. Neither progesterone dose significantly reduced cortical tissue loss. These findings have implications in understanding the sex-based differences in outcome following acute brain injury.


Asunto(s)
Lesiones Encefálicas/patología , Hipocampo/patología , Mitocondrias/fisiología , Progesterona/fisiología , Animales , Lesiones Encefálicas/metabolismo , Recuento de Células , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Implantes de Medicamentos , Femenino , Hipocampo/metabolismo , Ovariectomía , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Progesterona/administración & dosificación , Progesterona/farmacología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA