Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Neurotrauma ; 37(11): 1342-1357, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31830858

RESUMEN

Traumatic brain injury (TBI) is a chronic pathology, inducing long-term deficits that remain understudied in pre-clinical studies. In this context, exploration, anxiety-like behavior, cognitive flexibility, and motor coordination were assessed until 5 and 10 months after an experimental TBI in the adult mouse, using two cohorts. In order to differentiate age, surgery, and remote gray and white matter lesions, three groups (unoperated, sham-operated, and TBI) were studied. TBI induced delayed motor coordination deficits at the pole test, 4.5 months after injury, that could be explained by gray and white matter damages in ipsilateral nigrostriatal structures (striatum, internal capsule) that were spreading to new structures between cohorts, at 5 versus 10 months after the injury. Further, TBI induced an enhanced exploratory behavior during stressful situations (active phase during actimetry test, object exploration in an open field), risk-taking behaviors in the elevated plus maze 5 months after injury, and a cognitive inflexibility in the Barnes maze that persisted until 9 months after the injury. These behavioral modifications could be related to the white and gray matter lesions observed in ipsi- and contralateral limbic structures (amygdala, hilus/cornu ammonis 4, hypothalamus, external capsule, corpus callosum, and cingular cortex) that were spreading to new structures between cohorts, at 5 months versus 10 months after the injury. The present study corroborates clinical findings on TBI and provides a relevant rodent chronic model which could help in validating pharmacological strategies against the chronic consequences of TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/psicología , Encéfalo/patología , Conducta Exploratoria/fisiología , Aprendizaje por Laberinto/fisiología , Animales , Lesiones Traumáticas del Encéfalo/cirugía , Estudios de Seguimiento , Masculino , Ratones , Factores de Tiempo
2.
PLoS One ; 12(9): e0184811, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28910378

RESUMEN

Traumatic brain injury (TBI) results in white matter injury (WMI) that is associated with neurological deficits. Neuroinflammation originating from microglial activation may participate in WMI and associated disorders. To date, there is little information on the time courses of these events after mild TBI. Therefore we investigated (i) neuroinflammation, (ii) WMI and (iii) behavioral disorders between 6 hours and 3 months after mild TBI. For that purpose, we used experimental mild TBI in mice induced by a controlled cortical impact. (i) For neuroinflammation, IL-1b protein as well as microglial phenotypes, by gene expression for 12 microglial activation markers on isolated CD11b+ cells from brains, were studied after TBI. IL-1b protein was increased at 6 hours and 1 day. TBI induced a mixed population of microglial phenotypes with both pro-inflammatory, anti-inflammatory and immunomodulatory markers from 6 hours to 3 days post-injury. At 7 days, microglial activation was completely resolved. (ii) Three myelin proteins were assessed after TBI on ipsi- and contralateral corpus callosum, as this structure is enriched in white matter. TBI led to an increase in 2',3'-cyclic-nucleotide 3'-phosphodiesterase, a marker of immature and mature oligodendrocyte, at 2 days post-injury; a bilateral demyelination, evaluated by myelin basic protein, from 7 days to 3 months post-injury; and an increase in myelin oligodendrocyte glycoprotein at 6 hours and 3 days post-injury. Transmission electron microscopy study revealed various myelin sheath abnormalities within the corpus callosum at 3 months post-TBI. (iii) TBI led to sensorimotor deficits at 3 days post-TBI, and late cognitive flexibility disorder evidenced by the reversal learning task of the Barnes maze 3 months after injury. These data give an overall invaluable overview of time course of neuroinflammation that could be involved in demyelination and late cognitive disorder over a time-scale of 3 months in a model of mild TBI. This model could help to validate a pharmacological strategy to prevent post-traumatic WMI and behavioral disorders following mild TBI.


Asunto(s)
Conmoción Encefálica/inmunología , Trastornos del Conocimiento/etiología , Microglía/inmunología , Vaina de Mielina/patología , Sustancia Blanca/patología , Animales , Biomarcadores/metabolismo , Conmoción Encefálica/complicaciones , Conmoción Encefálica/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Aprendizaje por Laberinto , Ratones , Microscopía Electrónica de Transmisión , Vaina de Mielina/metabolismo
3.
PLoS One ; 9(2): e87367, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586272

RESUMEN

Poly(ADP-ribose)polymerase and sirtuin 1 are both NAD(+)-dependent enzymes. In vitro oxidative stress activates poly(ADP-ribose)polymerase, decreases NAD(+) level, sirtuin 1 activity and finally leads to cell death. Poly(ADP-ribose)polymerase hyperactivation contributes to cell death. In addition, poly(ADP-ribose)polymerase inhibition restores NAD(+) level and sirtuin 1 activity in vitro. In vitro sirtuin 1 induction protects neurons from cell loss induced by oxidative stress. In this context, the role of sirtuin 1 and its involvement in beneficial effects of poly(ADP-ribose)polymerase inhibition were evaluated in vivo in a model of cerebral oxidative stress induced by intrastriatal infusion of malonate in rat. Malonate promoted a NAD(+) decrease that was not prevented by 3-aminobenzamide, a poly(ADP-ribose)polymerase inhibitor, at 4 and 24 hours. However, 3-aminobenzamide increased nuclear SIRT1 activity/expression ratio after oxidative stress. Malonate induced a neurological deficit associated with a striatal lesion. Both were reduced by 3-aminobenzamide and SRT1720, a sirtuin 1 activator, showing beneficial effects of poly(ADP-ribose)polymerase inhibition and sirtuin 1 activation on oxidative stress consequences. EX527, a sirtuin 1 inhibitor, given alone, modified neither the score nor the lesion, suggesting that endogenous sirtuin 1 was not activated during cerebral oxidative stress. However, its association with 3-aminobenzamide suppressed the neurological improvement and the lesion reduction induced by 3-aminobenzamide. The association of 3-aminobenzamide with SRT1720, the sirtuin 1 activator, did not lead to a better protection than 3-aminobenzamide alone. The present data represent the first demonstration that the sirtuin 1 activator SRT1720 is neuroprotective during in vivo cerebral oxidative stress. Furthermore sirtuin 1 activation is involved in the beneficial effects of poly(ADP-ribose)polymerase inhibition after in vivo cerebral oxidative stress.


Asunto(s)
Encéfalo/fisiopatología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/fisiología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Sirtuina 1/metabolismo , Análisis de Varianza , Animales , Benzamidas/farmacología , Western Blotting , Carbazoles/farmacología , Immunoblotting , Masculino , Malonatos/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Sirtuina 1/antagonistas & inhibidores , Estadísticas no Paramétricas
4.
Exp Neurol ; 248: 416-28, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23876515

RESUMEN

Recombinant tissue plasminogen activator (rt-PA) is the only pharmacological treatment approved for thrombolysis in patients suffering from ischemic stroke, but its administration aggravates the risk of hemorrhagic transformations. Experimental data demonstrated that rt-PA increases the activity of poly(ADP-ribose)polymerase (PARP). The aim of the present study was to investigate whether PJ34, a potent (PARP) inhibitor, protects the blood-brain barrier components from rt-PA toxicity. In our mouse model of cerebral ischemia, administration of rt-PA (10 mg/kg, i.v.) 6h after ischemia aggravated the post-ischemic degradation of ZO-1, claudin-5 and VE-cadherin, increased the hemorrhagic transformations (assessed by brain hemoglobin content and magnetic resonance imaging). Furthermore, rt-PA also aggravated ischemia-induced functional deficits. Combining PJ34 with rt-PA preserved the expression of ZO-1, claudin-5 and VE-cadherin, reduced the hemorrhagic transformations and improved the sensorimotor performances. In vitro studies also demonstrated that PJ34 crosses the blood-brain barrier and may thus exert its protective effect by acting on endothelial and/or parenchymal cells. Thus, co-treatment with a PARP inhibitor seems to be a promising strategy to reduce rt-PA-induced vascular toxicity after stroke.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Fenantrenos/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/efectos adversos , Animales , Barrera Hematoencefálica/patología , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/patología , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Ratones , Fenantrenos/farmacología , Accidente Cerebrovascular/patología , Activador de Tejido Plasminógeno/farmacología , Activador de Tejido Plasminógeno/uso terapéutico
5.
Fundam Clin Pharmacol ; 27(4): 393-401, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22436003

RESUMEN

Recombinant tissue-type plasminogen activator (rt-PA) is presently the only pharmacological treatment approved for thrombolysis in patients suffering from ischemic stroke. Although reperfusion of ischemic tissue is essential, the use of rt-PA is limited due to its narrow therapeutic window and risk of hemorrhagic transformations. Recent studies have shown that rt-PA amplifies the post-ischemic activation of the nuclear enzyme poly(ADP-ribose)polymerase (PARP). This enzyme has been shown to contribute to both the breakdown of the blood brain barrier and spontaneous hemorrhagic transformations after ischemia. We therefore examined the capacity of PJ34 (N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-2-(N,N-dimethylamino) acetamide hydrochloride), a potent inhibitor of PARP, to reduce the hemorrhagic transformations that occur after rt-PA in mice with permanent focal cerebral ischemia. Ischemia was produced by intraluminal occlusion of the left middle cerebral artery and treated with vehicle, rt-PA (10 mg/kg, i.v., 6 h after occlusion) or rt-PA plus PJ34 (3, 6 or 12 mg/kg, i.p., at ischemia onset and 4 h later). Hemorrhagic transformations, neurological examination, and infarct volumes were evaluated 48 h after the onset of ischemia. Delayed administration of rt-PA resulted in increased hemorrhagic transformations and aggravated the neurological deficit. Giving PJ34 (3 mg/kg) markedly reduced the hemorrhagic transformations, an effect not owing to a modification of matrix metalloprotease activity. Furthermore, PJ34 improved the neurological functions of rt-PA-treated ischemic mice. To conclude, the PARP inhibitor PJ34 makes rt-PA safer in experimental ischemic stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Hemorragia/tratamiento farmacológico , Fenantrenos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Activador de Tejido Plasminógeno/metabolismo , Animales , Isquemia Encefálica/metabolismo , Inhibidores Enzimáticos/farmacología , Hemorragia/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Arteria Cerebral Media/efectos de los fármacos , Arteria Cerebral Media/metabolismo , Proteínas Recombinantes/farmacología
6.
Neuropharmacology ; 67: 183-92, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23178198

RESUMEN

Traumatic brain injury (TBI) evokes an intense neuroinflammatory reaction that is essentially mediated by activated microglia and that has been reported to act as a secondary injury mechanism that further promotes neuronal death. It involves the excessive production of inflammatory cytokines and the diminution of neuroprotective and neurotrophic factors, such as the soluble form alpha of the amyloid precursor protein (sAPPα), generated by the activity of α-secretases. Hence, the aim of this study was to examine the effects of etazolate, an α-secretase activator, on acute and belated post-TBI consequences. The mouse model of TBI by mechanical percussion was used and injured mice received either the vehicle or etazolate at the dose of 1, 3 or 10 mg/kg at 2 h post-TBI. Neurological score, cerebral œdema, IL-1ß and sAPPα levels, microglial activation and lesion size were evaluated from 6 to 24 h post-TBI. Spontaneous locomotor activity was evaluated from 48 h to 12 weeks post-TBI, memory function at 5 weeks and olfactory bulb lesions at 13 weeks post-TBI. A single administration of etazolate exerted a dose-dependent anti-inflammatory and anti-œdematous effect accompanied by lasting memory improvement, reduction of locomotor hyperactivity and olfactory bulb tissue protection, with a therapeutic window of at least 2 h. These effects were associated with the restoration of the levels of the sAPPα protein post-TBI. Taken together, these results highlight for the first time the therapeutic interest of an α-secretase activator in TBI.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Edema Encefálico/enzimología , Edema Encefálico/prevención & control , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/prevención & control , Etazolato/uso terapéutico , Animales , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Etazolato/farmacología , Inflamación/enzimología , Inflamación/prevención & control , Masculino , Ratones , Fármacos Neuroprotectores , Distribución Aleatoria
7.
Brain Res ; 1491: 78-87, 2013 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-23122881

RESUMEN

The implication of cyclooxygenase (COX) type 2 in post-traumatic consequences is so far controversial. In experimental models of traumatic brain injury (TBI), genetic disruption or pharmacological inhibition of COX-2 has been shown to be neuroprotective, deleterious or without effect. Therefore, the aim of our study was to investigate the effect of COX-2 inhibition against neurological deficit and brain oedema after TBI that was induced by mechanical percussion in male Swiss mice. Despite the increased level and activity of COX-2, its inhibition either with nimesulide (12 mg/kg) or meloxicam (2mg/kg) modified neither the neurological score nor the brain water content that were evaluated at 6 and 24h after injury. Interestingly, the non-selective COX inhibition with indomethacin (5mg/kg) significantly promoted neurological recovery at 6 and 24h after trauma, without improving brain oedema. In conclusion, the present study yields considerable evidence that COX-2 may not solely constitute an interesting target for the treatment of TBI consequences. Our data point to a potentially deleterious role of COX-1 in the development of neurological impairment in brain-injured mice. However, the neuroprotective mechanism of indomethacin remains to be clarified.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Inhibidores de la Ciclooxigenasa/uso terapéutico , Traumatismos Cerrados de la Cabeza/complicaciones , Traumatismos Cerrados de la Cabeza/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , 6-Cetoprostaglandina F1 alfa/biosíntesis , Animales , Conducta Animal/fisiología , Western Blotting , Encéfalo/patología , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Conducta Exploratoria/fisiología , Traumatismos Cerrados de la Cabeza/enzimología , Técnicas para Inmunoenzimas , Indometacina/uso terapéutico , Masculino , Ratones , Enfermedades del Sistema Nervioso/patología , Desempeño Psicomotor/fisiología , Recuperación de la Función , Especificidad por Sustrato
8.
Exp Neurol ; 238(2): 138-44, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22921458

RESUMEN

Recombinant tissue plasminogen activator (rt-PA) is currently the only approved drug for ischemic stroke treatment, with a dose of 0.9 mg/kg. Since the fibrinolytic activity of rt-PA has been reported in vitro to be 10-fold less potent in rodent than in human, in most in vivo experimental models of cerebral ischemia rt-PA is used at 10 mg/kg. The purpose of this study was to compare the effects of the "human" (0.9 mg/kg) and "rodent" (10 mg/kg) doses of rt-PA given at an early or a delayed time point in a mouse model of cerebral ischemia. Cerebral ischemia was induced by thrombin injection into the left middle cerebral artery of mice. Rt-PA (0.9 or 10 mg/kg) was intravenously administered 30 min or 4 h after the onset of ischemia. The degree of reperfusion after rt-PA was followed for 90 min after its injection. The neurological deficit, infarct volumes, edema and hemorrhagic transformations (HT) were assessed at 24 h. Reperfusion was complete after early administration of rt-PA at 10 mg/kg but partial with rt-PA at 0.9 mg/kg. Both doses given at 4 h induced partial reperfusion. Early administration of both doses of rt-PA reduced the neurological deficit, lesion volume and brain edema, without modifying post-ischemic HT. Injected at 4 h, rt-PA at 0.9 and 10 mg/kg lost its beneficial effects and worsened HT. In conclusion, in the mouse thrombin stroke model, the "human" dose of rt-PA exhibits effects close to those observed in clinic.


Asunto(s)
Modelos Animales de Enfermedad , Fibrinolíticos/uso terapéutico , Infarto de la Arteria Cerebral Media/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/etiología , Activador de Tejido Plasminógeno/uso terapéutico , Análisis de Varianza , Animales , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Esquema de Medicación , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Humanos , Masculino , Ratones , Enfermedades del Sistema Nervioso/etiología , Distribución Aleatoria , Accidente Cerebrovascular/complicaciones , Factores de Tiempo , Resultado del Tratamiento
9.
Endocrinology ; 153(8): 3747-57, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22635678

RESUMEN

Progesterone receptors (PR) are expressed throughout the brain. However, their functional significance remains understudied. Here we report a novel role of PR as crucial mediators of neuroprotection using a model of transient middle cerebral artery occlusion and PR knockout mice. Six hours after ischemia, we observed a rapid increase in progesterone and 5α-dihydroprogesterone, the endogenous PR ligands, a process that may be a part of the natural neuroprotective mechanisms. PR deficiency, and even haploinsufficiency, increases the susceptibility of the brain to stroke damage. Within a time window of 24 h, PR-dependent signaling of endogenous brain progesterone limits the extent of tissue damage and the impairment of motor functions. Longer-term improvement requires additional treatment with exogenous progesterone and is also PR dependent. The potent and selective PR agonist Nestorone is also effective. In contrast to progesterone, levels of the neurosteroid allopregnanolone, which modulates γ-aminobutyric acid type A receptors, did not increase after stroke, but its administration protected both wild-type and PR-deficient mice against ischemic damage. These results show that 1) PR are linked to signaling pathways that influence susceptibility to stroke, and 2) PR are direct key targets for both endogenous neuroprotection and for therapeutic strategies after stroke, and they suggest a novel indication for synthetic progestins already validated for contraception. Although allopregnanolone may not be an endogenous neuroprotective agent, its administration protects the brain against ischemic damage by signaling mechanisms not involving PR. Collectively, our data clarify the relative roles of PR and allopregnanolone in neuroprotection after stroke.


Asunto(s)
Receptores de Progesterona/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Masculino , Ratones , Ratones Noqueados , Norprogesteronas/farmacología , Norprogesteronas/uso terapéutico , Pregnanolona/farmacología , Pregnanolona/uso terapéutico , Progesterona/farmacología , Progesterona/uso terapéutico , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/prevención & control
10.
Neurosci Lett ; 511(2): 110-5, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22314279

RESUMEN

Comorbidity of cognitive and stress disorders is a common clinical sequel of traumatic brain injury (TBI) that is essentially determined by the site and severity of the insult, but also by the extent of the ensuing neuroinflammatory response. The present study sought to examine the late effects of closed-head TBI on memory function and anxiety in mice, in order to further examine the potential efficacy of an acute anti-inflammatory treatment with minocycline. The mouse model of closed-head injury by mechanical percussion was applied on anesthetized Swiss mice. The treatment protocol included three injections of minocycline (i.p.) at 5 min (90 mg/kg), 3 h and 9 h (45 mg/kg) post-TBI. The Novel Object Recognition Test as well as the Elevated Plus Maze (EPM) and Elevated Zero Maze (EZM) tasks were employed to assess post-TBI memory and anxiety respectively. Our results revealed a recognition memory deficit that was significant up to at least 13 weeks post-TBI. However, neither EPM nor EZM revealed any alteration in post-TBI anxiety levels albeit some mild disinhibition. Most importantly, minocycline was able to attenuate the memory impairment in an effective and lasting manner, highlighting its therapeutic potential in TBI.


Asunto(s)
Antiinflamatorios/uso terapéutico , Ansiedad/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Trastornos del Conocimiento/tratamiento farmacológico , Minociclina/uso terapéutico , Animales , Ansiedad/etiología , Lesiones Encefálicas/complicaciones , Trastornos del Conocimiento/etiología , Masculino , Ratones
11.
J Neurotrauma ; 29(2): 354-61, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21910642

RESUMEN

Permanent olfactory dysfunction can often arise after traumatic brain injury (TBI) and while one of the main causes is the immediate loss of neurons in the olfactory bulb (OB), the emergent neuroinflammatory environment following TBI may further promote OB deterioration. Therefore, we examined the effects of acute anti-inflammatory treatment with minocycline on post-TBI olfactory behavior and on OB surface. The mouse model of closed-head injury by mechanical percussion was applied to anesthetized Swiss mice. The treatment protocol included three injections of minocycline (i.p.) at 5 min (90 mg/kg), 3 h, and 9 h (45 mg/kg) post-TBI. An olfactory avoidance test was run up to 12 weeks post-TBI. The mice were then sacrificed and their OB surface was measured. Our results demonstrated a post-TBI olfactory behavior deficit that was significant up to at least 12 weeks post-TBI. Additionally, substantial post-TBI OB atrophy was observed that was strongly correlated with the behavioral impairment. Minocycline was able to attenuate both the olfactory lesions and corresponding functional deficit in the short and long term. These results emphasize the potential role of minocycline as a promising neuroprotective agent for the treatment of TBI-related olfactory bulb lesions and deficits.


Asunto(s)
Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Minociclina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Trastornos del Olfato/tratamiento farmacológico , Bulbo Olfatorio/lesiones , Bulbo Olfatorio/fisiopatología , Olfato/fisiología , Animales , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Masculino , Ratones , Minociclina/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Trastornos del Olfato/etiología , Trastornos del Olfato/fisiopatología , Bulbo Olfatorio/patología , Olfato/efectos de los fármacos
12.
J Neurotrauma ; 28(10): 2135-43, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21770756

RESUMEN

Traumatic brain injury (TBI) induces both focal and diffuse lesions that are concurrently responsible for the ensuing morbidity and mortality and for which no established treatment is available. It has been recently reported that an endogenous neuroprotector, the soluble form α of the amyloid precursor protein (sAPPα), exerts neuroprotective effects following TBI. However, the emergent post-traumatic neuroinflammatory environment compromises sAPPα production and may promote neuronal degeneration and consequent brain atrophy. Hence, the aim of this study was to examine the effects of the anti-inflammatory drug minocycline on sAPPα levels, as well as on long-term histological consequences post-TBI. The weight-drop model was used to induce TBI in mice. Minocycline or its vehicle were administered three times: at 5 min (90 mg/kg, i.p.) and at 3 and 9 h (45 mg/kg, i.p.) post-TBI. The levels of sAPPα, the extent of brain atrophy, and reactive gliosis were evaluated by ELISA, cresyl violet, and immunolabeling of GFAP and CD11b, respectively. Our results revealed a post-TBI sAPPα decrease that was significantly attenuated by minocycline. Additionally, corpus callosum and striatal atrophy, ventriculomegaly, astrogliosis, and microglial activation were observed at 3 months post-TBI. All the above consequences were significantly reduced by minocycline. In conclusion, inhibition of the acute phase of post-TBI neuroinflammation was associated with the sparing of sAPPα and the protection of brain tissue in the long-term, emphasizing the potential role of minocycline as an effective treatment for TBI.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Antibacterianos/farmacología , Química Encefálica/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Minociclina/farmacología , Fármacos Neuroprotectores , Animales , Lesiones Encefálicas/patología , Antígeno CD11b/metabolismo , Ventrículos Cerebrales/patología , Cuerpo Calloso/metabolismo , Cuerpo Calloso/patología , Cuerpo Estriado/patología , Ensayo de Inmunoadsorción Enzimática , Gliosis/patología , Inmunohistoquímica , Masculino , Ratones
13.
Crit Care Med ; 39(10): 2300-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21666443

RESUMEN

OBJECTIVES: Traumatic brain injury causes deleterious brain edema, leading to high mortality and morbidity. Brain edema exacerbates neurologic deficits and may be attributable to the breakdown of endothelial cell junction protein, leukocyte infiltration, and matrix metalloproteinase activation. These all contribute to loss of blood-brain barrier integrity. The pleiotropic effects of statins, hydroxymethylglutaryl-coenzyme A reductase inhibitors, may inhibit posttraumatic brain edema. We therefore investigated the effect of acute simvastatin on neurologic deficits, cerebral edema, and its origins. DESIGN: Randomized laboratory animal study. SETTINGS: University-affiliated research laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: Rats were subjected to lateral fluid percussion traumatic brain injury. Our preliminary dose-effect study indicated that 37.5 mg/kg simvastatin, administered orally 1 hr and 6 hrs after traumatic brain injury, has the greatest anti-edematous effect. This dose was used to study its effects on brain edema and on its mechanisms. MEASUREMENTS AND MAIN RESULTS: We first assessed the effects of simvastatin 24 hrs after traumatic brain injury on brain edema, brain claudin-5 expression, and the vascular endothelial-cadherin (pTyr731)/total vascular endothelial-cadherin ratio, matrix metalloproteinase-9 activity, intercellular adhesion molecule-1 expression, and polymorphonuclear neutrophil infiltration. We also evaluated blood-brain barrier permeability by measuring Evans blue and fluorescein sodium salt extravasation into the cerebral parenchyma. We then investigated whether simvastatin reduces neurologic deficits, edema, and blood-brain barrier permeability earlier than 24 hrs; these effects were evaluated 6 hrs after traumatic brain injury. The anti-edematous effect of simvastatin 24 hrs after traumatic brain injury was associated with increased claudin-5 and decreased intercellular adhesion molecule-1, polymorphonuclear neutrophil infiltration, and blood-brain barrier permeability, with no effect on matrix metalloproteinase-9 activity or vascular endothelial-cadherin phosphorylation. Earlier, 6-hrs after traumatic brain injury, simvastatin reduced neurologic deficits, cerebral edema, and blood-brain barrier permeability. CONCLUSIONS: Simvastatin could be a new therapy for reducing posttraumatic edema by preventing damage to tight junctions and neutrophil infiltration into the parenchyma, thus preserving blood-brain barrier integrity.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/complicaciones , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Simvastatina/farmacología , Animales , Antígenos CD/biosíntesis , Barrera Hematoencefálica/metabolismo , Edema Encefálico/etiología , Edema Encefálico/patología , Cadherinas/biosíntesis , Claudina-5 , Células Endoteliales/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Proteínas de la Membrana/biosíntesis , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Ratas , Ratas Sprague-Dawley
14.
J Neurosci Methods ; 191(2): 180-90, 2010 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-20600312

RESUMEN

Although intracerebral haemorrhage (ICH) entails the highest rates of mortality and disability of all stroke subtypes, efficient neuroprotective therapy is still needed. As functional recovery is a major endpoint in clinical trials, preclinical studies must demonstrate the potential of drugs to improve the sensorimotor and cognitive function of animals. In addition, behavioural studies should be performed on the long-term in order to truly mimic clinical needs. The aim of our study was to characterise a model of intracerebral haemorrhage using both histology and long-term behaviour. ICH was induced in rats by an intrastriatal injection of collagenase. Histology was performed 24h, 7 days and 2 months after ICH. Among a set of sensorimotor tests, we discriminate those able to reveal long-term deficits (up to 2 months) after cerebral haemorrhage. Our five behavioural tests (a neurological score, an adhesive removal test, two beam-walking tests and ipsilateral circling induced by dexamphetamine) proved to be effective in revealing sensorimotor deficits up to 35 days or more after cerebral haemorrhage. In conclusion, these behavioural tests appear of particular interest to screen protective agents that may exhibit benefits in patients who suffer ICH.


Asunto(s)
Encéfalo/patología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/patología , Hemorragia Cerebral/inducido químicamente , Colagenasas/toxicidad , Evaluación de la Discapacidad , Animales , Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Arterias Cerebrales/fisiopatología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/fisiopatología , Dextroanfetamina , Modelos Animales de Enfermedad , Cojera Animal/diagnóstico , Cojera Animal/etiología , Cojera Animal/fisiopatología , Masculino , Examen Neurológico , Ratas , Ratas Sprague-Dawley , Tiempo
15.
J Neurotrauma ; 27(6): 1069-79, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20380552

RESUMEN

Recent evidence supports a crucial role for matrix metalloproteinase-9 (MMP-9) in blood-brain barrier (BBB) disruption and vasogenic edema formation after traumatic brain injury (TBI). Although the exact causes of MMP-9 upregulation after TBI are not fully understood, several arguments suggest a contribution of the enzyme poly(ADP-ribose)polymerase (PARP) in the neuroinflammatory response leading to MMP-9 activation. The objectives of this study were to evaluate the effect of PARP inhibition by 3-aminobenzamide (3-AB) (1) on MMP-9 upregulation and BBB integrity, (2) on edema formation as assessed by magnetic resonance imaging (MRI), (3) on neuron survival as assessed by (1)H magnetic resonance spectroscopy ((1)H-MRS), and (4) on neurological deficits at the acute phase of TBI. Western blots and zymograms showed blunting of MMP-9 upregulation 6 h after TBI. BBB permeability was decreased at the same time point in 3-AB-treated rats compared to vehicle-treated rats. Cerebral MRI showed less "free" water in 3-AB-treated than in vehicle-treated rats 6 h after TBI. MRI findings 24 h after TBI indicated predominant cytotoxic edema, and at this time point no significant differences were found between 3-AB- and vehicle-treated rats with regard to MMP-9 upregulation, BBB permeability, or MRI changes. At both 6 and 24 h, neurological function was better in the 3-AB-treated than in the vehicle-treated rats. These data suggest that PARP inhibition by 3-AB protected the BBB against hyperpermeability induced by MMP-9 upregulation, thereby decreasing vasogenic edema formation 6 h after TBI. Furthermore, our data confirm the neuroprotective effect of 3-AB at the very acute phase of TBI.


Asunto(s)
Benzamidas/farmacología , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/patología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/patología , Inhibidores Enzimáticos/farmacología , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Permeabilidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Ratas , Ratas Sprague-Dawley , Estadísticas no Paramétricas
16.
J Neurotrauma ; 27(5): 911-21, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20166806

RESUMEN

Traumatic brain injury (TBI) causes a wide spectrum of consequences, such as microglial activation, cerebral inflammation, and focal and diffuse brain injury, as well as functional impairment. In this study we aimed to investigate the effects of acute treatment with minocycline as an inhibitor of microglial activation on cerebral focal and diffuse lesions, and on the spontaneous locomotor activity following TBI. The weight-drop model was used to induce TBI in mice. Microglial activation and diffuse axonal injury (DAI) were detected by immunohistochemistry using CD11b and ss-amyloid precursor protein (ss-APP) immunolabeling, respectively. Focal injury was determined by the measurement of the brain lesion volume. Horizontal and vertical locomotor activities were measured for up to 12 weeks post-injury by an automated actimeter. Minocycline or vehicle were administered three times post-insult, at 5 min (90 mg/kg i.p.), 3 h, and 9 h post-TBI (45 mg/kg i.p.). Minocycline treatment attenuated microglial activation by 59% and reduced brain lesion volume by 58%, yet it did not affect DAI at 24 h post-TBI. More interestingly, minocycline significantly decreased TBI-induced locomotor hyperactivity at 48 h post-TBI, and its effect lasted for up to 8 weeks. Taken together, the results indicate that microglial activation appears to play an important role in the development of TBI-induced focal injury and the subsequent locomotor hyperactivity, and its short-term inhibition provides long-lasting functional recovery after TBI. These findings emphasize the fact that minocycline could be a promising new therapeutic strategy for head-injured patients.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Gliosis/tratamiento farmacológico , Traumatismos Cerrados de la Cabeza/tratamiento farmacológico , Microglía/efectos de los fármacos , Microglía/metabolismo , Minociclina/farmacología , Animales , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Estudios de Seguimiento , Gliosis/patología , Traumatismos Cerrados de la Cabeza/patología , Hipercinesia/tratamiento farmacológico , Hipercinesia/patología , Hipercinesia/prevención & control , Masculino , Ratones , Microglía/patología , Fármacos Neuroprotectores/farmacología , Factores de Tiempo , Resultado del Tratamiento
17.
J Neurotrauma ; 27(1): 85-94, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19705964

RESUMEN

Magnetic resonance imaging (MRI) is widely used to evaluate the consequences of traumatic brain injury (TBI) in both experimental and clinical studies. Improved assessment of experimental TBI using the same methods as those used in clinical investigations would help to translate laboratory research into clinical advances. Here our goal was to characterize lateral fluid percussion-induced TBI, with special emphasis on differentiating the contused cortex from the pericontusional subcortical tissue. We used both in vivo MRI and proton magnetic resonance spectroscopy ((1)H-MRS) to evaluate adult male Sprague-Dawley rats 24 h and 48 h and 7 days after TBI. T2 and apparent diffusion coefficient (ADC) maps were derived from T2-weighted and diffusion-weighted images, respectively. Ratios of N-acetylaspartate (NAA), choline compounds (Cho), and lactate (Lac) over creatine (Cr) were estimated by (1)H-MRS. T2 values were high in the contused cortex 24 h after TBI, suggesting edema development; ADC was low, consistent with cytotoxic edema. At the same site, NAA/Cr was decreased and Lac/Cr elevated during the first week after TBI. In the ipsilateral subcortical area, NAA/Cr was markedly decreased and Lac/Cr was elevated during the first week, although MRI showed no evidence of edema, suggesting that (1)H-MRS detected "invisible" damage. (1)H-MRS combined with MRI may improve the detection of brain injury. Extensive assessments of animal models may increase the chances of developing successful neuroprotective strategies.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Animales , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Encéfalo/patología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Edema Encefálico/fisiopatología , Lesiones Encefálicas/patología , Mapeo Encefálico , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Colina/metabolismo , Creatina/metabolismo , Difusión , Modelos Animales de Enfermedad , Ácido Láctico/metabolismo , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
18.
Brain Res ; 1291: 122-32, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19631631

RESUMEN

One of the severe complications following traumatic brain injury (TBI) is cerebral edema and its effective treatment is of great interest to prevent further brain damage. This study investigated the effects of minocycline, known for its anti-inflammatory properties, on cerebral edema and its respective inflammatory markers by comparing different dose regimens, on oxidative stress and on neurological dysfunction following TBI. The weight drop model was used to induce TBI in mice. The brain water content was measured to evaluate cerebral edema. Inflammatory markers were detected by ELISA (IL-1beta), zymography and Western blot (MMP-9). The oxidative stress marker (glutathione levels) and neurological function were measured by Griffith technique and string test, respectively. Minocycline was administered i.p. once (5 min), twice (5 min and 3 h) or triple (5 min, 3 h and 9 h) following TBI. The first dose of minocycline only varied (45 or 90 mg/kg), whereas the following doses were all at 45 mg/kg. The single and double administrations of minocycline reduced the increase of inflammatory markers at 6 h post-TBI. Minocycline also reduced cerebral edema at this time point, only after double administration and at the high dose regimen, although with no effect on the TBI-induced oxidized glutathione increase. The anti-edematous effect of minocycline persisted up to 24 h, upon a triple administration, and accompanied by a neurological recovery. In conclusion, we reported an anti-edematous effect of minocycline after TBI in mice according to a specific treatment regimen. These findings emphasize that the beneficial effects of minocycline depend on the treatment regimen following a brain injury.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Edema Encefálico/etiología , Lesiones Encefálicas/complicaciones , Corteza Cerebral/metabolismo , Interleucina-1beta/metabolismo , Minociclina/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Análisis de Varianza , Animales , Antiinflamatorios/uso terapéutico , Western Blotting , Agua Corporal , Edema Encefálico/metabolismo , Edema Encefálico/fisiopatología , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/fisiopatología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiopatología , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Examen Neurológico , Estrés Oxidativo/fisiología
19.
BMC Mol Biol ; 9: 62, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18611280

RESUMEN

BACKGROUND: Traumatic brain injury models are widely studied, especially through gene expression, either to further understand implied biological mechanisms or to assess the efficiency of potential therapies. A large number of biological pathways are affected in brain trauma models, whose elucidation might greatly benefit from transcriptomic studies. However the suitability of reference genes needed for quantitative RT-PCR experiments is missing for these models. RESULTS: We have compared five potential reference genes as well as total cDNA level monitored using Oligreen reagent in order to determine the best normalizing factors for quantitative RT-PCR expression studies in the early phase (0-48 h post-trauma (PT)) of a murine model of diffuse brain injury. The levels of 18S rRNA, and of transcripts of beta-actin, glyceraldehyde-3P-dehydrogenase (GAPDH), beta-microtubulin and S100beta were determined in the injured brain region of traumatized mice sacrificed at 30 min, 3 h, 6 h, 12 h, 24 h and 48 h post-trauma. The stability of the reference genes candidates and of total cDNA was evaluated by three different methods, leading to the following rankings as normalization factors, from the most suitable to the less: by using geNorm VBA applet, we obtained the following sequence: cDNA(Oligreen); GAPDH > 18S rRNA > S100beta > beta-microtubulin > beta-actin; by using NormFinder Excel Spreadsheet, we obtained the following sequence: GAPDH > cDNA(Oligreen) > S100beta > 18S rRNA > beta-actin > beta-microtubulin; by using a Confidence-Interval calculation, we obtained the following sequence: cDNA(Oligreen) > 18S rRNA; GAPDH > S100beta > beta-microtubulin > beta-actin. CONCLUSION: This work suggests that Oligreen cDNA measurements, 18S rRNA and GAPDH or a combination of them may be used to efficiently normalize qRT-PCR gene expression in mouse brain trauma injury, and that beta-actin and beta-microtubulin should be avoided. The potential of total cDNA as measured by Oligreen as a first-intention normalizing factor with a broad field of applications is highlighted. Pros and cons of the three methods of normalization factors selection are discussed. A generic time- and cost-effective procedure for normalization factor validation is proposed.


Asunto(s)
Lesiones Encefálicas/genética , Perfilación de la Expresión Génica/normas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/normas , Actinas/genética , Animales , ADN Complementario/análisis , ADN Complementario/normas , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Ratones , Modelos Animales , Factores de Crecimiento Nervioso/genética , ARN Ribosómico 18S/genética , Estándares de Referencia , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/genética , Tubulina (Proteína)/genética , Estudios de Validación como Asunto
20.
J Pharmacol Exp Ther ; 326(3): 966-74, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18562561

RESUMEN

We and others have demonstrated that fibrates [peroxisome proliferator-activated receptor (PPAR)alpha agonists] and statins (3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors) exerted neuroprotective and pleiotropic effects in experimental models of traumatic brain injury (TBI). Because the combination of statins and fibrates synergistically enhanced PPARalpha activation, we hypothesized that the combination of both drugs may exert more important and/or prolonged beneficial effects in TBI than each alone. In this study, we examined the combination of fenofibrate with simvastatin, administered 1 and 6 h after injury, on the consequences of TBI. First, our dose-effect study demonstrated that the most efficient dose of simvastatin (37.5 mg/kg) reduced post-traumatic neurological deficits and brain edema. Then, the effects of the combination of fenofibrate (50 mg/kg) and simvastatin (37.5 mg/kg), given p.o. at 1 and 6 h after TBI, were evaluated on the TBI consequences in the early and late phase after injury. The combination exerted more sustained neurological recovery-promoting and antiedematous effects than monotherapies, and it synergistically decreased the post-traumatic brain lesion. Furthermore, a delayed treatment given p.o. at 3 and 8 h after TBI with the combination was still efficient on neurological deficits induced by TBI, but it failed to reduce the brain edema at 48 h. The present data represent the first demonstration that the combination of fenofibrate and simvastatin exerts prolonged and synergistic neuroprotective effects than each drug alone. Thus, these results may have important therapeutic significance for the treatment of TBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Modelos Animales de Enfermedad , Fenofibrato/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , PPAR alfa/agonistas , Simvastatina/administración & dosificación , Animales , Lesiones Encefálicas/metabolismo , Quimioterapia Combinada , Masculino , PPAR alfa/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA