Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cytotherapy ; 20(2): 245-261, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29274773

RESUMEN

BACKGROUND AIMS: Human umbilical cord blood (HUCB) is an important source of stem cells for therapy of hematopoietic disorders and is a potential therapy for various neurological disorders, including traumatic brain injury (TBI). The expression of nerve growth factor (NGF) and its receptors TrkA, p75NTR and α9ß1 integrin on an HUCB CD45+ pan-hematopoietic subpopulation was investigated in the context of its neurotherapeutic potential after TBI. METHODS: NGF and its receptors were detected on CD45+ cells by reverse transcriptase polymerase chain reaction, flow cytometry analysis and confocal microscopy. CD45+ cells were stimulated by TBI brain extracts, and NGF levels were measured by enzyme-linked immunosorbent assay. TBI mice were divided into six groups for xenogeneic intravenous transplantation, 1 day post-trauma, with 1 × 106 CD45+ cells untreated or treated with the anti-NGF neutralizing antibody K252a, a TrkA antagonist; VLO5, an α9ß1 disintegrin; or negative (vehicle) and positive (NGF) controls. RESULTS: The HUCB CD45+ subpopulation constitutively expresses NGF and its receptors, mainly TrkA and p75NTR and minor levels of α9ß1. In vitro experiments provided evidence that trauma-related mediators from brain extracts of TBI mice induced release of NGF from HUCB CD45+ cell cultures. HUCB CD45+ cells induced a neurotherapeutic effect in TBI mice, abrogated by cell treatment with either anti-NGF antibody or K252a, but not VLO5. CONCLUSIONS: These findings strengthen the role of NGF and its TrkA receptor in the HUCB CD45+ subpopulation's neurotherapeutic effect. The presence of neurotrophin receptors in the HUCB CD45+ pan-hematopoietic subpopulation may explain the neuroprotective effect of cord blood in therapy of a variety of neurological disorders.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Factor de Crecimiento Nervioso/uso terapéutico , Animales , Lesiones Traumáticas del Encéfalo/patología , Quimiocina CCL3/metabolismo , Trasplante de Células Madre Hematopoyéticas , Humanos , Interleucina-10/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Ratones Endogámicos C57BL , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Extractos de Tejidos
2.
Synapse ; 65(7): 634-42, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21108236

RESUMEN

BACKGROUND: Neuroinflammation is involved in several acute-onset neuropathologies such as meningitis, encephalitis, stroke, and traumatic brain injury as well as in neurodegenerative diseases. All of these patholologies are associated with cognitive deficits. Using a model of pure neuroinflammation (intracisternal injection of endotoxin in mice), we tested the hypothesis that brain regions involved in cognition are the most vulnerable to inflammatory insults, and this vulnerability is an inherent property of neocortical neurons. METHODS: Mice (n = 10/group) injected with endotoxin (LPS) or saline in the cisterna magna underwent neurobehavioral and cognitive testing followed by quantitative autoradiographic assessment of regional neuroinflammation with [3H]PK11195, an established marker of microgliosis. In parallel, cocultures of cortical and striatal neurons taken from embryonic day 19 rat embryos or postnatal day 1 mice expressing green fluorescent protein were exposed for 24 h to the proinflammatory cytokine TNFalpha, glutamate, or a combination of the two agents. RESULTS: LPS-treated mice exhibited significant deficits in memory and significant increases in specific PK11195 binding in cortical and hippocampal regions, but not in striatum. Cultured neurons of cortical origin showed significantly lower survival rate relative to striatal neurons in response to TNFalpha, glutamate, or a combination of the two agents. Furthermore, TNFalpha exerted neuroprotective rather than neurotoxic effects in the striatal but not in the cortical neurons. CONCLUSIONS: These results suggest that the cortex is inherently more sensitive than the striatum to the deleterious effects of neuroinflammation, and may offer an explanation for the preponderance of cognitive deficits in neuropathologies with a neuroinflammatory component.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/patología , Encefalitis/patología , Inflamación/patología , Animales , Encéfalo/metabolismo , Encefalitis/inducido químicamente , Encefalitis/metabolismo , Técnica del Anticuerpo Fluorescente , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Ratones
3.
FASEB J ; 23(12): 4266-75, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19723705

RESUMEN

Despite efforts aimed at developing novel therapeutics for traumatic brain injury (TBI), no specific pharmacological agent is currently clinically available. Here, we show that the pan-histone deacetylase (HDAC) inhibitor ITF2357, a compound shown to be safe and effective in humans, improves functional recovery and attenuates tissue damage when administered as late as 24 h postinjury. Using a well-characterized, clinically relevant mouse model of closed head injury (CHI), we demonstrate that a single dose of ITF2357 administered 24 h postinjury improves neurobehavioral recovery from d 6 up to 14 d postinjury (improved neurological score vs. vehicle; P< or =0.05), and that this functional benefit is accompanied by decreased neuronal degeneration, reduced lesion volume (22% reduction vs. vehicle; P< or =0.01), and is preceded by increased acetylated histone H3 levels and attenuation of injury-induced decreases in cytoprotective heat-shock protein 70 kDa and phosphorylated Akt. Moreover, reduced glial accumulation and activation were observed 3 d postinjury, and total p53 levels at the area of injury and caspase-3 immunoreactivity within microglia/macrophages at the trauma area were elevated, suggesting enhanced clearance of these cells via apoptosis following treatment. Hence, our findings underscore the relevance of HDAC inhibitors for ameliorating trauma-induced functional deficits and warrant consideration of applying ITF2357 for this indication.


Asunto(s)
Apoptosis/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Neuroglía/efectos de los fármacos , Animales , Encéfalo/patología , Caspasa 3/metabolismo , Ácidos Hidroxámicos/administración & dosificación , Masculino , Ratones , Fármacos Neuroprotectores/farmacología
4.
Front Neurol ; 11: 999, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178093

RESUMEN

Traumatic brain injury (TBI), caused by mechanical impact to the brain, is a leading cause of death and disability among young adults, with slow and often incomplete recovery. Preemptive treatment strategies may increase the injury resilience of high-risk populations such as soldiers and athletes. In this work, the xanthophyll carotenoid Astaxanthin was examined as a potential nutritional preconditioning method in mice (sabra strain) to increase their resilience prior to TBI in a closed head injury (CHI) model. The effect of Astaxanthin pretreatment on heat shock protein (HSP) dynamics and functional outcome after CHI was explored by gavage or free eating (in pellet form) for 2 weeks before CHI. Assessment of neuromotor function by the neurological severity score (NSS) revealed significant improvement in the Astaxanthin gavage-treated group (100 mg/kg, ATX) during recovery compared to the gavage-treated olive oil group (OIL), beginning at 24 h post-CHI and lasting throughout 28 days (p < 0.007). Astaxanthin pretreatment in pellet form produced a smaller improvement in NSS vs. posttreatment at 7 days post-CHI (p < 0.05). Cognitive and behavioral evaluation using the novel object recognition test (ORT) and the Y Maze test revealed an advantage for Astaxanthin administration via free eating vs. standard chow during recovery post-CHI (ORT at 3 days, p < 0.035; improvement in Y Maze score from 2 to 29 days, p < 0.02). HSP profile and anxiety (open field test) were not significantly affected by Astaxanthin. In conclusion, astaxanthin pretreatment may contribute to improved recovery post-TBI in mice and is influenced by the form of administration.

5.
FASEB J ; 22(1): 285-94, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17704191

RESUMEN

We have recently reported that in bone the cannabinoid CB1 receptor is present in sympathetic terminals. Here we show that traumatic brain injury (TBI), which in humans enhances peripheral osteogenesis and fracture healing, acutely stimulates bone formation in a distant skeletal site. At this site we demonstrate i) a high level of the main endocannabinoid, 2-arachidonoylglycerol (2-AG), and expression of diacylglycerol lipases, enzymes essential for 2-AG synthesis; ii) that the TBI-induced increase in bone formation is preceded by elevation of the 2-AG and a decrease in norepinephrine (NE) levels. The TBI stimulation of bone formation was absent in CB1-null mice. In wild-type animals it could be mimicked, including the suppression of NE levels, by 2-AG administration. The TBI- and 2-AG-induced stimulation of osteogenesis was restrained by the beta-adrenergic receptor agonist isoproterenol. NE from sympathetic terminals is known to tonically inhibit bone formation by activating osteoblastic beta2-adrenergic receptors. The present findings further demonstrate that the sympathetic control of bone formation is regulated through 2-AG activation of prejunctional CB1. Elevation of bone 2-AG apparently suppresses NE release from bone sympathetic terminals, thus alleviating the inhibition of bone formation. The involvement of osteoblastic CB2 signaling in this process is minimal, if any.


Asunto(s)
Desarrollo Óseo/fisiología , Receptor Cannabinoide CB1/fisiología , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/fisiología , Animales , Ácidos Araquidónicos/administración & dosificación , Ácidos Araquidónicos/metabolismo , Secuencia de Bases , Huesos/metabolismo , Cartilla de ADN , Endocannabinoides , Femenino , Glicéridos/administración & dosificación , Glicéridos/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Cannabinoide CB1/genética
6.
J Neurotrauma ; 25(4): 324-33, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18373482

RESUMEN

Memory and neurobehavioral dysfunctions are among the sequelae of traumatic brain injury (TBI). The Neurological Severity Score (NSS) includes 10 tasks and was previously designed to assess the functional status of mice after TBI. The object recognition task (ORT) measures specific episodic memory and is expressed by the percent time spent by an animal at a novel, unfamiliar object (Discrimination Index [DI]). It is an ideal tool for evaluating cognitive function after TBI. The present study sought to validate the use of the NSS and ORT in severe and mild focal TBI in mice, and to confirm that the spontaneous recovery and the radiological abnormalities, shown by T2-weighted magnetic resonance imaging (MRI), are dependent upon injury severity. Mice were subjected to severe and mild closed head injury (NSS at 1 h 7.52 +/- 0.34 and 4.62 +/- 0.14, respectively). NSS was evaluated for 25 days and showed a decrease by 3.86 +/- 0.26 and 2.54 +/- 0.35 units in the severely and mildly injured mice, respectively. ORT revealed DI in severely injured group of 51.7 +/- 6.15%, (vs approximately 75-80% in uninjured animal) on day 3 and 66.2 +/- 6.81% on day 21. In contrast, the mildly injured mice did not show cognitive impairment throughout the same period. The damage seen by MRI at 24 h after injury, strongly correlated with NSS(1h) (R = 0.87, p < 0.001). We conclude that NSS is a reliable tool for evaluation of neurological damage in head-injured mice, NSS(1h) predicts the motor dysfunction, cognitive damage, and brain-damage characteristics as depicted by T2-weighted MRI. The combined assessment of neurobehavioral and cognitive function along with MRI is most useful in evaluating recovery from injury, especially when testing effectiveness of novel treatments or genetic manipulations.


Asunto(s)
Lesiones Encefálicas/patología , Lesiones Encefálicas/psicología , Cognición/fisiología , Actividad Motora/fisiología , Reconocimiento en Psicología/fisiología , Recuperación de la Función/fisiología , Animales , Lesiones Encefálicas/fisiopatología , Imagen por Resonancia Magnética , Masculino , Ratones , Equilibrio Postural/fisiología , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Factores de Tiempo , Índices de Gravedad del Trauma
7.
J Neurotrauma ; 25(6): 569-80, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18447626

RESUMEN

Traumatic brain injury (TBI) remains a frequent and major challenge in neurological and neurosurgical practice. Apoptosis may play a role in cerebral tissue damage induced by the traumatic insult, and thus its detection and inhibition may advance patient care. DDC (N,N'-didansyl-L-cystine) is a novel fluorescent probe for detection of apoptotic cells. We now report on the performance of DDC in experimental TBI. Closed head injury was induced in mice by weight-drop. DDC was administered intravenously in vivo. Two hours later, animals were sacrificed, and brain tissue was subjected to fluorescent microcopy, for assessment of DDC uptake, in correlation with histopathological assessment of apoptosis by TUNEL and caspase substrates, and also in correlation with the neurological deficits, as assessed by Neurological Severity Score (NSS). Selective uptake of DDC was observed at the primary site of injury, and also at remote sites. Uptake was at the cellular level, with accumulation of DDC in the cytoplasm. Cells manifesting DDC uptake were confirmed as apoptotic cells by detection of the characteristic apoptotic DNA fragmentation (positive TUNEL staining) and detection of activated caspases. The damaged region stained by DDC fluorescence correlated with the severity of neuronal deficits. Our study confirms the role of apoptosis in TBI, and proposes DDC as a useful tool for its selective targeting and detection in vivo. Such imaging of apoptosis, following future radiolabeling of DDC, may advance care for patients with head injury, by allowing real-time evaluation of the extent of tissue damage, assessment of novel therapeutic strategies, and optimization of treatment for the individual patient.


Asunto(s)
Apoptosis , Lesiones Encefálicas/diagnóstico , Cistina/análogos & derivados , Compuestos de Dansilo , Colorantes Fluorescentes , Degeneración Nerviosa/diagnóstico , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Caspasas/metabolismo , Cistina/química , Cistina/metabolismo , Citoplasma/metabolismo , Citoplasma/patología , Fragmentación del ADN , Compuestos de Dansilo/química , Compuestos de Dansilo/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Valor Predictivo de las Pruebas , Coloración y Etiquetado/métodos
8.
FASEB J ; 21(9): 2033-41, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17351125

RESUMEN

Traumatic brain injury triggers a massive glutamate efflux, activation of NMDA receptor channels, and cell death. Recently, we reported that NMDA receptors in mice are down-regulated from hours to days following closed head injury (CHI), and treatment with NMDA improved recovery of motor and cognitive functions up to 14 d post-injury. Here we show that a single injection of a low dose of D-cycloserine (DCS), a partial NMDA receptor agonist, in CHI mice 24 h post-injury, resulted in a faster and greater recovery of motor and memory functions as assessed by neurological severity score and object recognition tests, respectively. Moreover, DCS treatment of CHI mice led to a significant improvement of hippocampal long-term potentiation (LTP) in the CA1 region that was completely blunted in CHI control mice. However, DCS did not improve CHI-induced impairment in synaptic glutamate release measured by paired pulse facilitation (PPF) ratio in hippocampal CA1 region. Finally, CHI-induced reduction of brain-derived neurotrophic factor (BDNF) was fully restored following DCS treatment. Since DCS is in clinical use for other indications, the present study offers a novel approach to treat human brain injury.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Cicloserina/uso terapéutico , Agonistas de Aminoácidos Excitadores/uso terapéutico , Traumatismos Cerrados de la Cabeza/complicaciones , Potenciación a Largo Plazo/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Receptores de N-Metil-D-Aspartato/agonistas , Animales , Astrocitos/metabolismo , Astrocitos/patología , Lesiones Encefálicas/etiología , Lesiones Encefálicas/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Cicloserina/farmacología , Evaluación Preclínica de Medicamentos , Agonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Hipocampo/ultraestructura , Masculino , Ratones , Microglía/metabolismo , Microglía/patología , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptores de N-Metil-D-Aspartato/fisiología , Reconocimiento en Psicología/efectos de los fármacos , Método Simple Ciego , Sinaptofisina/biosíntesis , Sinaptofisina/genética
9.
J Neurotrauma ; 24(4): 651-6, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17439348

RESUMEN

Low-level laser therapy (LLLT) has been evaluated in this study as a potential therapy for traumatic brain injury (TBI). LLLT has been found to modulate various biological processes. Following TBI in mice, we assessed the hypothesis that LLLT might have a beneficial effect on their neurobehavioral and histological outcome. TBI was induced by a weight-drop device, and motor function was assessed 1 h post-trauma using a neurological severity score (NSS). Mice were then divided into three groups of eight mice each: one control group that received a sham LLLT procedure and was not irradiated; and two groups that received LLLT at two different doses (10 and 20 mW/cm(2) ) transcranially. An 808-nm Ga-As diode laser was employed transcranially 4 h post-trauma to illuminate the entire cortex of the brain. Motor function was assessed up to 4 weeks, and lesion volume was measured. There were no significant changes in NSS at 24 and 48 h between the laser-treated and non-treated mice. Yet, from 5 days and up to 28 days, the NSS of the laser-treated mice were significantly lower (p < 0.05) than the traumatized control mice that were not treated with the laser. The lesion volume of the laser treated mice was significantly lower (1.4%) than the non-treated group (12.1%). Our data suggest that a non-invasive transcranial application of LLLT given 4 h following TBI provides a significant long-term functional neurological benefit. Further confirmatory trials are warranted.


Asunto(s)
Lesiones Encefálicas/radioterapia , Traumatismos Cerrados de la Cabeza/terapia , Terapia por Luz de Baja Intensidad , Enfermedades del Sistema Nervioso/prevención & control , Animales , Encéfalo/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Corteza Cerebral/patología , Traumatismos Cerrados de la Cabeza/complicaciones , Traumatismos Cerrados de la Cabeza/patología , Masculino , Ratones , Movimiento/fisiología , Enfermedades del Sistema Nervioso/patología , Equilibrio Postural/fisiología , Reflejo/fisiología , Caminata/fisiología
10.
Brain Res ; 1185: 313-20, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17963735

RESUMEN

Long-term exposure to moderate ambient heat (heat acclimation, HA, 30 days at 34+/-1 degrees C) provides protection toward a variety of stressors including traumatic brain injury. As previous studies suggested an anti-inflammatory effect of HA and given the ability of augmented pre-injury anti-inflammatory cytokine expression to harbor neuroprotection and to attenuate early post-injury expression of pro-inflammatory mediators, we hypothesized that HA-induced neuroprotection may involve enhanced pre-injury expression of anti-inflammatory mediators or a reduction in post-injury TNF alpha (TNFalpha) expression. Since the attenuation of inflammatory-associated entities has also been linked to mild hypothermia, an established neuroprotective paradigm, the effect of HA on post-injury body temperature was also studied. HA mice and normothermic (NT) counterparts were examined using a closed head injury model. Cytokines were measured within the ipsilateral cortex. Pre-injury protein levels of anti-inflammatory interleukins 10 and 4 (IL-10, IL-4) were quantified by enzyme-linked immunosorbent assays (ELISA). mRNA and protein levels of TNFalpha were quantified during the initial 2 h post-injury using semi-quantitative and real-time polymerase chain reaction (sqRT-PCR and qRT-PCR) or ELISA, respectively. Rectal temperatures were measured. HA induced augmented pre-injury IL-10 expression and a post-injury reduction in TNFalpha mRNA levels, as well as altered expression dynamics of TNFalpha protein. TNFalpha protein levels decreased relative to the sham state in HA mice only. HA mice displayed sustained post-injury hypothermia, namely significantly lower body temperature at 4 h post-injury. Given the evidence on the neuroprotective nature of hypothermia and anti-inflammatory cytokines, we suggest that these changes may contribute to HA-induced neuroprotection.


Asunto(s)
Aclimatación/fisiología , Lesiones Encefálicas/complicaciones , Citocinas/metabolismo , Regulación de la Expresión Génica/fisiología , Calor , Hipotermia/etiología , Análisis de Varianza , Animales , Ensayo de Inmunoadsorción Enzimática/métodos , Masculino , Ratones , Factores de Tiempo
11.
J Basic Clin Physiol Pharmacol ; 27(3): 209-16, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26565551

RESUMEN

In recent years, a library of approx. 70 N-acyl aminoacids (NAAAs) was discovered in the rat brain. A particular member of this family of compounds is arachidonoyl serine (AraS), which has generated special interest as a potential therapy for traumatic brain injury (TBI). This is due to its structural similarity to the endocannabinoid (eCB) 2-arachidonoyl glycerol (2-AG), which was previously shown to be beneficial in the recovery in a closed head injury model of TBI. Indeed, AraS exerted eCB-mediated neuroprotection, which was evident in numerous aspects related to the secondary damage characterizing TBI. These findings promoted broadening of the research to additional compounds of the NAAA family that share a structural similarity to AraS, namely, palmitoyl serine (PalmS) and oleoyl serine. The latter did not exhibit any improvement in recovery, whereas the former displayed some neuroprotection, albeit inferior to 2-AG and AraS, via unknown mechanisms. Interestingly, when a combined treatment of 2-AG, AraS and PalmS was tested, the overall effect on the severity score was inferior to their individual effects, suggesting not only a lack of direct or indirect synergism, but also possibly some spatial hindrance. Taken together, the complexity of the damage caused by TBI and the many open questions concerning the role of the eCB system in health and disease, the findings so far may serve as a small trace to the understanding of the eCB system, as well as of the mechanisms underlying TBI.


Asunto(s)
Aminoácidos/farmacología , Aminoácidos/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Endocannabinoides/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Animales , Ácidos Araquidónicos/farmacología , Ácidos Araquidónicos/uso terapéutico , Encéfalo/efectos de los fármacos , Endocannabinoides/farmacología , Endocannabinoides/uso terapéutico , Glicéridos/farmacología , Glicéridos/uso terapéutico , Humanos
12.
Peptides ; 26(8): 1520-7, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16042992

RESUMEN

A single administration of the neuroprotective peptide NAP was previously shown to protect against death associated with closed head injury (CHI) and enhance recovery of the surviving mice. The protective effect was accompanied by down-regulation of the relative mRNA content of the complement receptor 3 (Mac-1, a marker for inflammation) as measured about a month after the injury. In contrast, the mRNA transcripts for activity-dependent neuroprotective protein (ADNP, the NAP containing protein) were shown to increase 29 days post CHI in the injured hemisphere of Mac-1 expressing mice. The present study was set out to investigate: (1) are Mac-1-deficient mice less susceptible to the adverse outcome of traumatic head injury; (2) does NAP treatment affect Mac-1-deficient mice subjected to head injury; and (3) is Mac-1 expression associated with ADNP expression. Results showed that (1) Mac-1-deficient mice were partially protected against death associated with severe head injury as compared to Mac-1 expressing mice. (2) Significant protection against death was observed in NAP-treated mice and an increase in recovery was observed in the NAP-treated Mac-1 mice 4 weeks after injury. (3) ADNP expression did not change in the Mac-1-deficient mice following head injury. Our working hypothesis is that a month following injury, gene expression in the injured brain is altered and competing proteins are expressed such as Mac-1 that is associated with inflammation and ADNP that is associated with neuroprotection. Obviously, this plasticity in gene expression is intimately interwoven with the genetic background of the animal. NAP treatment tilts the balance toward neuroprotection.


Asunto(s)
Traumatismos Cerrados de la Cabeza/tratamiento farmacológico , Antígeno de Macrófago-1/genética , Fármacos Neuroprotectores/administración & dosificación , Oligopéptidos/administración & dosificación , Animales , Modelos Animales de Enfermedad , Traumatismos Cerrados de la Cabeza/fisiopatología , Proteínas de Homeodominio/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Examen Neurológico/estadística & datos numéricos , Fármacos Neuroprotectores/uso terapéutico , Pruebas Neuropsicológicas/estadística & datos numéricos , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Factores de Tiempo
13.
J Neuroimmune Pharmacol ; 10(2): 356-63, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25721934

RESUMEN

The endocannabinoid (eCB) system helps recovery following traumatic brain injury (TBI). Treatment with 2-arachidonoylglycerol (2-AG), a cerebral eCB ligand, was found to ameliorate the secondary damage. Interestingly, the fatty acid amino acid amide (FAAA) N-arachidonoyl-L-serine (AraS) exerts similar eCB dependent neuroprotective. The present study aimed to investigate the effects of the FAAA palmitoyl-serine (PalmS) following TBI. We utilized the TBI model in mice to examine the therapeutic potential of PalmS, injected 1 h following closed head injury (CHI). We followed the functional recovery of the injured mice for 28 days post-CHI, and evaluated cognitive and motor function, lesion volume, cytokines levels, molecular signaling, and infarct volume at different time points after CHI. PalmS treatment led to a significant improvement of the neurobehavioral outcome of the treated mice, compared with vehicle. This effect was attenuated in the presence of eCBR antagonists and in CB2-/- mice, compared to controls. Unexpectedly, treatment with PalmS did not affect edema and lesion volume, TNFα and IL1ß levels, anti-apoptotic mechanisms, nor did it exert improvement in cognitive and motor function. Finally, co-administration of PalmS, AraS and 2-AG, did not enhance the effect of the individual drugs. We suggest that the neuroprotective action of PalmS is mediated by indirect activation of the eCB receptors following TBI. One such mechanism may involve receptor palmitoylation which has been reported to result in structural stabilization of the receptors and to an increase in their activity. Further research is required in order to establish this assumption.


Asunto(s)
Lesiones Encefálicas/prevención & control , Endocannabinoides/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Palmitatos/uso terapéutico , Serina/uso terapéutico , Animales , Ácidos Araquidónicos/farmacología , Ácidos Araquidónicos/uso terapéutico , Lesiones Encefálicas/patología , Relación Dosis-Respuesta a Droga , Endocannabinoides/farmacología , Glicéridos/farmacología , Glicéridos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/farmacología , Palmitatos/farmacología , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/deficiencia , Serina/farmacología
14.
J Mol Neurosci ; 24(2): 181-7, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15456931

RESUMEN

Activity-dependent neuroprotective protein (ADNP), a crucial brain development factor, contains a unique sequence, termed NAPVSIPQ, which protects mice against closed head injury (CHI). The aim of this study was to determine whether CHI affects ADNP mRNA expression in the injured brain hemisphere. Male C57JBL/6J mice were subjected to CHI. Brains were removed 5 h, 24 h, 7 d, and 29 d post-CHI. A comparison was made between ADNP mRNA in the injured versus the noninjured hemisphere using real-time polymerase chain reaction. A nonsignificant change (p >0.05) was found 5 h, 24 h, and 7 d post-CHI. However, a significant increase (p <0.05) in ADNP mRNA expression was detected in the injured cerebral hemisphere 29 d post-CHI. The data presented may be associated with ADNP's crucial involvement in brain development and response to injury.


Asunto(s)
Lesiones Encefálicas/metabolismo , Traumatismos Cerrados de la Cabeza/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Homeodominio/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética
15.
Eur J Pharmacol ; 434(3): 109-16, 2002 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-11779573

RESUMEN

Selegiline and rasagiline are selective and irreversible monoamine oxidase-B inhibitors that exert neuroprotective effects in various preclinical models. The aim of the present study was to examine the effect of selegiline and its major metabolite, L-methamphetamine in comparison to rasagiline and its major metabolite, 1-R-aminoindan on oxygen-glucose deprivation induced cell death in nerve growth factor (NGF)-differentiated pheochromocytoma (PC12) cells. Our results show that selegiline reduces oxygen-glucose deprivation induced cell death by 30%. When the cultures were treated with rasagiline at similar concentrations, cell death induced by oxygen-glucose deprivation was reduced by 45-55%. L-methamphetamine, a major selegiline metabolite, but not 1-R-aminoindan, the major rasagiline metabolite, enhanced oxygen-glucose deprivation-induced cell death by 70%. Under normoxic conditions, both metabolites lack neurotoxicity. Concomitant exposure of the cultures under oxygen-glucose deprivation, to a combination of either selegiline and L-methamphetamine or rasagiline and 1-R-aminoindan, indicated that L-methamphetamine, but not 1-R-aminoindan, blocked the neuroprotective effect of the parental drug. These results suggest there may be a neuroprotective advantage of rasagiline over selegiline.


Asunto(s)
Isquemia/enzimología , Isquemia/patología , Inhibidores de la Monoaminooxidasa/farmacología , Monoaminooxidasa/metabolismo , Fármacos Neuroprotectores/farmacología , Células PC12/efectos de los fármacos , Células PC12/enzimología , Animales , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores de la Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/toxicidad , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/toxicidad , Células PC12/patología , Ratas
16.
Neurosci Lett ; 353(2): 115-8, 2003 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-14664914

RESUMEN

Tumor necrosis factor-alpha (TNFalpha) is harmful in the early phase and beneficial in the long-term phase after brain injury. Reactive oxygen species (ROS) are among the most toxic mediators activated by injury. We speculate that part of the TNFalpha toxicity is mediated by its synergism with ROS. Thus, toxicity of TNFalpha and ROS, alone or together, were studied in PC12 cells. PC12 cells were exposed for 18 h to TNFalpha (0-100 ng/ml), to H2O2 (1-300 microM) or to both, each at sub-toxic concentrations. Lactic dehydrogenase release, prostaglandin E2 accumulation and morphology indicated cell death and stress response. TNFalpha toxicity was seen at >50 ng/ml, and that of H2O2 at >150 microM, however, when together, sub-lethal levels (25 ng/ml TNFalpha and 30 microM H2O2) induced toxicity. Dexanabinol, an N-methyl-D-aspartate antagonist with antioxidant and anti-TNFalpha properties, completely rescued the cells. These findings corroborate our hypothesis on the cooperative toxicity exerted by TNFalpha and ROS after brain injury.


Asunto(s)
Muerte Celular/efectos de los fármacos , Dronabinol/análogos & derivados , Peróxido de Hidrógeno/toxicidad , Neurotoxinas/toxicidad , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Antioxidantes/farmacología , Muerte Celular/fisiología , Dinoprostona/metabolismo , Dronabinol/farmacología , Sinergismo Farmacológico , L-Lactato Deshidrogenasa/metabolismo , Estrés Oxidativo , Células PC12 , Ratas , Especies Reactivas de Oxígeno
17.
Neurotherapeutics ; 11(3): 665-78, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24957202

RESUMEN

Angiotensin II receptor type 2 (AT(2)) agonists have been shown to limit brain ischemic insult and to improve its outcome. The activation of AT(2) was also linked to induced neuronal proliferation and differentiation in vitro. In this study, we examined the therapeutic potential of AT(2) activation following traumatic brain injury (TBI) in mice, a brain pathology that displays ischemia-like secondary damages. The AT(2) agonist CGP42112A was continuously infused immediately after closed head injury (CHI) for 3 days. We have followed the functional recovery of the injured mice for 35 days post-CHI, and evaluated cognitive function, lesion volume, molecular signaling, and neurogenesis at different time points after the impact. We found dose-dependent improvement in functional recovery and cognitive performance after CGP42112A treatment that was accompanied by reduced lesion volume and induced neurogenesis in the neurogenic niches of the brain and also in the injury region. At the cellular/molecular level, CGP42112A induced early activation of neuroprotective kinases protein kinase B (Akt) and extracellular-regulated kinases ½ (ERK½), and the neurotrophins nerve growth factor and brain-derived neurotrophic factor; all were blocked by treatment with the AT(2) antagonist PD123319. Our results suggest that AT(2) activation after TBI promotes neuroprotection and neurogenesis, and may be a novel approach for the development of new drugs to treat victims of TBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Neurogénesis/fisiología , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Receptor de Angiotensina Tipo 2/agonistas , Receptor de Angiotensina Tipo 2/metabolismo , Animales , Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proliferación Celular , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Oligopéptidos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Reconocimiento en Psicología/efectos de los fármacos
18.
J Cereb Blood Flow Metab ; 34(8): 1381-90, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24849663

RESUMEN

Long-term exposure of mice to mild heat (34°C±1°C) confers neuroprotection against traumatic brain injury (TBI); however, the underling mechanisms are not fully understood. Heat acclimation (HA) increases hypothalamic angiotensin II receptor type 2 (AT2) expression and hypothalamic neurogenesis. Accumulating data suggest that activation of the brain AT2 receptor confers protection against several types of brain pathologies, including ischemia, a hallmark of the secondary injury occurring following TBI. As AT2 activates the same pro-survival pathways involved in HA-mediated neuroprotection (e.g., Akt phosphorylation, hypoxia-inducible factor 1α (HIF-1α), and brain-derived neurotrophic factor (BDNF)), we examined the role of AT2 in HA-mediated neuroprotection after TBI. Using an AT2-specific antagonist PD123319, we found that the improvements in motor and cognitive recovery as well as reduced lesion volume and neurogenesis seen in HA mice were all diminished by AT2 inhibition, whereas no significant alternations were observed in control mice. We also found that nerve growth factor/tropomyosin-related kinase receptor A (TrkA), BDNF/TrkB, and HIF-1α pathways are upregulated by HA and inhibited on PD123319 administration, suggesting that these pathways play a role in AT2 signaling in HA mice. In conclusion, AT2 is involved in HA-mediated neuroprotection, and AT2 activation may be protective and should be considered a novel drug target in the treatment of TBI patients.


Asunto(s)
Aclimatación , Lesiones Encefálicas/prevención & control , Traumatismos Cerrados de la Cabeza/prevención & control , Calor , Neurogénesis , Receptor de Angiotensina Tipo 2/metabolismo , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Conducta Animal/fisiología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Traumatismos Cerrados de la Cabeza/metabolismo , Traumatismos Cerrados de la Cabeza/fisiopatología , Imidazoles/farmacología , Masculino , Ratones , Ratones Endogámicos , Actividad Motora/fisiología , Piridinas/farmacología , Transducción de Señal
19.
Brain Res ; 1555: 78-88, 2014 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-24502983

RESUMEN

Despite years of research, no effective therapy is yet available for the treatment of traumatic brain injury (TBI). The most prevalent and debilitating features in survivors of TBI are cognitive deficits and motor dysfunction. A potential therapeutic method for improving the function of patients following TBI would be to restore, at least in part, plasticity to the CNS in a controlled way that would allow for the formation of compensatory circuits. Inosine, a naturally occurring purine nucleoside, has been shown to promote axon collateral growth in the corticospinal tract (CST) following stroke and focal TBI. In the present study, we investigated the effects of inosine on motor and cognitive deficits, CST sprouting, and expression of synaptic proteins in an experimental model of closed head injury (CHI). Treatment with inosine (100 mg/kg i.p. at 1, 24 and 48 h following CHI) improved outcome after TBI, significantly decreasing the neurological severity score (NSS, p<0.04 vs. saline), an aggregate measure of performance on several tasks. It improved non-spatial cognitive performance (object recognition, p<0.016 vs. saline) but had little effect on sensorimotor coordination (rotarod) and spatial cognitive functions (Y-maze). Inosine did not affect CST sprouting in the lumbar spinal cord but did restore levels of the growth-associated protein GAP-43 in the hippocampus, though not in the cerebral cortex. Our results suggest that inosine may improve functional outcome after TBI.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Inosina/uso terapéutico , Animales , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Lesiones Encefálicas/psicología , Modelos Animales de Enfermedad , Proteína GAP-43/metabolismo , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Tractos Piramidales/patología , Ratas , Reconocimiento en Psicología/efectos de los fármacos , Recuperación de la Función , Prueba de Desempeño de Rotación con Aceleración Constante , Sinaptofisina/metabolismo
20.
J Neurotrauma ; 31(16): 1405-16, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24640955

RESUMEN

Treatment of traumatic brain injury (TBI) is still an unmet need. Cell therapy by human umbilical cord blood (HUCB) has shown promising results in animal models of TBI and is under evaluation in clinical trials. HUCB contains different cell populations but to date, only mesenchymal stem cells have been evaluated for therapy of TBI. Here we present the neurotherapeutic effect, as evaluated by neurological score, using a single dose of HUCB-derived mononuclear cells (MNCs) upon intravenous (IV) administration one day post-trauma in a mouse model of closed head injury (CHI). Delayed (eight days post-trauma) intracerebroventricular administration of MNCs showed improved neurobehavioral deficits thereby extending the therapeutic window for treating TBI. Further, we demonstrated for the first time that HUCB-derived pan-hematopoietic CD45 positive (CD45(+)) cells, isolated by magnetic sorting and characterized by expression of CD45 and CD11b markers (96-99%), improved the neurobehavioral deficits upon IV administration, which persisted for 35 days. The therapeutic effect was in a direct correlation to a reduction in the lesion volume and decreased by pre-treatment of the cells with anti-human-CD45 antibody. At the site of brain injury, 1.5-2 h after transplantation, HUCB-derived cells were identified by near infrared scanning and immunohistochemistry using anti-human-CD45 and anti-human-nuclei antibodies. Nerve growth factor and vascular endothelial growth factor levels were differentially expressed in both ipsilateral and contralateral brain hemispheres, thirty-five days after CHI, measured by enzyme-linked immunosorbent assay. These findings indicate the neurotherapeutic potential of HUCB-derived CD45(+) cell population in a mouse model of TBI and propose their use in the clinical setting of human TBI.


Asunto(s)
Lesiones Encefálicas/terapia , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Recuperación de la Función , Animales , Separación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Antígenos Comunes de Leucocito/inmunología , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA