Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Front Neurol ; 11: 999, 2020.
Article in English | MEDLINE | ID: mdl-33178093

ABSTRACT

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.

2.
Cytotherapy ; 20(2): 245-261, 2018 02.
Article in English | MEDLINE | ID: mdl-29274773

ABSTRACT

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.


Subject(s)
Brain Injuries, Traumatic/therapy , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Nerve Growth Factor/therapeutic use , Animals , Brain Injuries, Traumatic/pathology , Chemokine CCL3/metabolism , Hematopoietic Stem Cell Transplantation , Humans , Interleukin-10/metabolism , Leukocyte Common Antigens/metabolism , Mice, Inbred C57BL , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Tissue Extracts
3.
J Basic Clin Physiol Pharmacol ; 27(3): 209-16, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26565551

ABSTRACT

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.


Subject(s)
Amino Acids/pharmacology , Amino Acids/therapeutic use , Brain Injuries, Traumatic/drug therapy , Endocannabinoids/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Animals , Arachidonic Acids/pharmacology , Arachidonic Acids/therapeutic use , Brain/drug effects , Endocannabinoids/pharmacology , Endocannabinoids/therapeutic use , Glycerides/pharmacology , Glycerides/therapeutic use , Humans
5.
J Neuroimmune Pharmacol ; 10(2): 356-63, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25721934

ABSTRACT

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.


Subject(s)
Brain Injuries/prevention & control , Endocannabinoids/therapeutic use , Neuroprotective Agents/therapeutic use , Palmitates/therapeutic use , Serine/therapeutic use , Animals , Arachidonic Acids/pharmacology , Arachidonic Acids/therapeutic use , Brain Injuries/pathology , Dose-Response Relationship, Drug , Endocannabinoids/pharmacology , Glycerides/pharmacology , Glycerides/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/pharmacology , Palmitates/pharmacology , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/deficiency , Serine/pharmacology
6.
Neurotherapeutics ; 11(3): 665-78, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24957202

ABSTRACT

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.


Subject(s)
Brain Injuries/drug therapy , Neurogenesis/physiology , Neuroprotective Agents/therapeutic use , Oligopeptides/therapeutic use , Receptor, Angiotensin, Type 2/agonists , Receptor, Angiotensin, Type 2/metabolism , Animals , Brain/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Nerve Growth Factor/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Oligopeptides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Recognition, Psychology/drug effects
7.
J Cereb Blood Flow Metab ; 34(8): 1381-90, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24849663

ABSTRACT

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.


Subject(s)
Acclimatization , Brain Injuries/prevention & control , Head Injuries, Closed/prevention & control , Hot Temperature , Neurogenesis , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Behavior, Animal/physiology , Brain Injuries/metabolism , Brain Injuries/physiopathology , Disease Models, Animal , Head Injuries, Closed/metabolism , Head Injuries, Closed/physiopathology , Imidazoles/pharmacology , Male , Mice , Mice, Inbred Strains , Motor Activity/physiology , Pyridines/pharmacology , Signal Transduction
8.
J Neurotrauma ; 31(16): 1405-16, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24640955

ABSTRACT

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.


Subject(s)
Brain Injuries/therapy , Cord Blood Stem Cell Transplantation/methods , Recovery of Function , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , Humans , Leukocyte Common Antigens/immunology , Male , Mice
9.
Brain Res ; 1555: 78-88, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24502983

ABSTRACT

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.


Subject(s)
Brain Injuries/drug therapy , Inosine/therapeutic use , Animals , Brain Edema/complications , Brain Edema/drug therapy , Brain Injuries/complications , Brain Injuries/pathology , Brain Injuries/psychology , Disease Models, Animal , GAP-43 Protein/metabolism , Hippocampus/metabolism , Male , Maze Learning/drug effects , Mice , Pyramidal Tracts/pathology , Rats , Recognition, Psychology/drug effects , Recovery of Function , Rotarod Performance Test , Synaptophysin/metabolism
10.
PLoS One ; 8(10): e76129, 2013.
Article in English | MEDLINE | ID: mdl-24124534

ABSTRACT

Preconditioning via heat acclimation (34°C 30 d) results in neuroprotection from traumatic brain injury due to constitutive as well as dynamic changes triggered by the trauma. Among these changes is Akt phosphorylation, which decreases apoptosis and induces HIF1α. In the present study we investigated the Akt downstream GSK3ß/ß-catenin pathway and focused on post injury alternations of ß catenin and its impact on the cellular response in preconditioned heat acclimated mice. We found that the reduction in motor disability is accompanied with attenuation of depressive like behavior in heat acclimated mice that correlates with the GSK3ß phosphorylation state. Concomitantly, a robust ß catenin phosphorylation is not followed by its degradation, or by reduced nuclear accumulation. Enhanced tyrosine phosphorylation of ß catenin in the injured area weakens the ß catenin-N cadherin complex. Membrane ß catenin is transiently reduced in heat acclimated mice and its recovery 7 days post TBI is accompanied by induction of the synaptic marker synaptophysin. We suggest a set of cellular events following traumatic brain injury in heat acclimated mice that causes ß catenin to participate in cell-cell adhesion alternations rather than in Wnt signaling. These events may contribute to synaptogenesis and the improved motor and cognitive abilities seen heat acclimated mice after traumatic brain injury.


Subject(s)
Brain Injuries/metabolism , beta Catenin/metabolism , Animals , Cadherins/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Mice , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
11.
J Cereb Blood Flow Metab ; 33(8): 1242-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23695434

ABSTRACT

N-arachidonoyl-L-serine (AraS) is a novel neuroprotective endocannabinoid. We aimed to test the effects of exogenous AraS on neurogenesis after traumatic brain injury (TBI). The effects of AraS on neural progenitor cells (NPC) proliferation, survival, and differentiation were examined in vitro. Next, mice underwent TBI and were treated with AraS or vehicle. Lesion volumes and clinical outcome were evaluated and the effects on neurogenesis were tested using immunohistochemistry. Treatment with AraS led to a dose-dependent increase in neurosphere size without affecting cell survival. These effects were partially reversed by CB1, CB2, or TRPV1 antagonists. AraS significantly reduced the differentiation of NPC in vitro to astrocytes or neurons and led to a 2.5-fold increase in expression of the NPC marker nestin. Similar effects were observed in vivo in mice treated with AraS 7 days after TBI. These effects were accompanied by a reduction in lesion volume and an improvement in neurobehavioral function compared with controls. AraS increases proliferation of NPCs in vitro in cannabinoid-receptor-mediated mechanisms and maintains NPC in an undifferentiated state in vitro and in vivo. Moreover, although given at 7 days post injury, these effects are associated with significant neuroprotective effects leading to an improvement in neurobehavioral functions.


Subject(s)
Arachidonic Acids/pharmacology , Brain Injuries/drug therapy , Neurogenesis/drug effects , Serine/analogs & derivatives , Animals , Astrocytes/drug effects , Behavior, Animal , Blotting, Western , Brain/pathology , Brain Injuries/pathology , Brain Injuries/psychology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coloring Agents , Male , Mice , Neural Stem Cells/drug effects , Propidium , Receptor, Cannabinoid, CB1/drug effects , Receptor, Cannabinoid, CB2/drug effects , Serine/pharmacology , TRPV Cation Channels/physiology , Treatment Outcome
12.
J Cereb Blood Flow Metab ; 33(4): 524-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23281425

ABSTRACT

Heat acclimation (HA), a well-established preconditioning model, confers neuroprotection in rodent models of traumatic brain injury (TBI). It increases neuroprotective factors, among them is hypoxia-inducible factor 1α (HIF-1α), which is important in the response to postinjury ischemia. However, little is known about the role of HIF-1α in TBI and its contribution to the establishment of the HA protecting phenotype. Therefore, we aimed to explore HIF-1α role in TBI defense mechanisms as well as in HA-induced neuroprotection. Acriflavine was used to inhibit HIF-1 in injured normothermic (NT) or HA mice. After TBI, we evaluated motor function recovery, lesion volume, edema formation, and body temperature as well as HIF-1 downstream transcription targets, such as glucose transporter 1 (GLUT1), vascular endothelial growth factor, and aquaporin 4. We found that HIF-1 inhibition resulted in deterioration of motor function, increased lesion volume, hypothermia, and reduced edema formation. All these parameters were significantly different in the HA mice. Western blot analysis and enzyme-linked immunosorbent assay showed reduced levels of all HIF-1 downstream targets in HA mice, however, only GLUT1 was downregulated in NT mice. We conclude that HIF-1 is a key mediator in both spontaneous recovery and HA-induced neuroprotection after TBI.


Subject(s)
Acclimatization , Brain Edema/metabolism , Brain Injuries/metabolism , Hot Temperature , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nerve Tissue Proteins/metabolism , Acriflavine/pharmacology , Animals , Anti-Infective Agents, Local/pharmacology , Aquaporin 4/biosynthesis , Brain Edema/pathology , Brain Injuries/pathology , Glucose Transporter Type 1/biosynthesis , Mice , Remission, Spontaneous
13.
J Neurotrauma ; 29(18): 2831-4, 2012 Dec 10.
Article in English | MEDLINE | ID: mdl-22994850

ABSTRACT

Death of Central Nervous System (CNS) neurons following traumatic brain injury (TBI) is a complex process arising from a combination of factors, many of which are still unknown. It has been found that inhibition of transient receptor potential (TRP) channels constitutes an effective strategy for preventing death of CNS neurons following TBI. TRP channels are classified into seven related subfamilies, most of which are Ca(2+) permeable and involved in many cellular functions, including neuronal cell death. We hypothesized that TRP channels of the TRPC subfamily may be involved in post-TBI pathophysiology and that the compound 5-isopropyl-2-methylphenol (carvacrol), by inhibition of TRP channels, may exert neuroprotective effect after TBI. To test these suppositions, carvacrol was given to mice after TBI and its effect on their functional recovery was followed for several weeks. Our results show that neurological recovery after TBI was significantly enhanced by application of carvacrol. To better define the type of the specific channel involved, the effect of carvacrol on the extent and speed of recovery after TBI was compared among mice lacking TRPC1, TRPC3, or TRPC5, relative to wild type controls. We found that neurological recovery after TBI was significantly enhanced by combining carvacrol with TRPC1 elimination, but not by the absence of TRPC3 or TRPC5, showing a synergistic effect between carvacrol application and TRPC1 elimination. We conclude that TRPC1-sensitive mechanisms are involved in TBI pathology, and that inhibition of this channel by carvacrol enhances recovery and should be considered for further studies in animal models and humans.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/genetics , Monoterpenes/pharmacology , TRPC Cation Channels/genetics , TRPC Cation Channels/physiology , Animals , Attention/physiology , Behavior, Animal/physiology , Cymenes , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoterpenes/administration & dosage , Postural Balance/physiology , Psychomotor Performance/physiology , Rats , Recovery of Function , Reflex/physiology , TRPC Cation Channels/antagonists & inhibitors
14.
J Neurotrauma ; 29(2): 375-84, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-21561314

ABSTRACT

Traumatic brain injury (TBI) initiates acute and chronic inflammatory processes involving cyclooxygenase-2 (COX-2), which may have detrimental effects on outcome and especially on brain regeneration. Therefore we aimed to study whether carprofen, a COX-2 inhibitor, would improve outcome and increase neurogenesis after TBI. TBI was induced in Sabra mice that were then treated with vehicle or carprofen for 7 days. Functional outcome was evaluated with the Neurological Severity Score (NSS).Cytokine levels were assessed 4 h post-TBI and water content was measured 24 h post TBI. Mice were given BrdU to label newborn cells for 10 days. The animals were killed 90 days post-TBI and the lesion size as well as newborn cell fate were assessed. Carprofen significantly reduced lesion size (p=0.002), decreased water content in the lesioned cortex (p=0.03), reduced the number of microglia in the lesioned cortex (p<0.0001), and lowered the levels of proinflammatory cytokines (IL-1ß, p=0.03; IL-6, p=0.02). Carprofen led to significantly larger improvements in functional outcome (p≤0.008) which were durable over 90 days. Carprofen also induced a threefold increase in the proliferation of new cells in the peri-lesion area (p≤0.002), but newborn cells differentiated mainly into glia in both groups. Carprofen is neuroprotective and induces cell proliferation and gliogenesis after TBI. Treatment with carprofen is consistently associated with better functional outcome. Our results imply that anti-inflammatory drugs may represent novel therapeutic options for TBI.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Brain Injuries/drug therapy , Brain Injuries/pathology , Carbazoles/administration & dosage , Neurogenesis/drug effects , Neuroglia/drug effects , Neuroglia/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain Injuries/physiopathology , Carbazoles/therapeutic use , Cell Proliferation/drug effects , Disease Models, Animal , Male , Mice , Mice, Inbred Strains , Neurogenesis/physiology , Neuroglia/cytology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Time Factors
15.
J Neurotrauma ; 29(2): 401-7, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22040267

ABSTRACT

Near-infrared transcranial laser therapy (TLT) has been found to modulate various biological processes including traumatic brain injury (TBI). Following TBI in mice, in this study we assessed the possibility of various near-infrared TLT modes (pulsed versus continuous) in producing a beneficial effect on the long-term neurobehavioral outcome and brain lesions of these mice. TBI was induced by a weight-drop device, and neurobehavioral function was assessed from 1 h to 56 days post-trauma using the Neurological Severity Score (NSS). The extent of recovery is expressed as the difference in NSS (dNSS), the difference between the initial score and that at any other later time point. An 808-nm Ga-Al-As diode laser was employed transcranially 4, 6, or 8 h post-trauma to illuminate the entire cortex of the brain. Mice were divided into several groups of 6-8 mice: one control group that received a sham treatment and experimental groups that received either TLT continuous wave (CW) or pulsed wave (PW) mode transcranially. MRI was taken prior to sacrifice at 56 days post-injury. From 5-28 days post-TBI, the NSS of the laser-treated mice were significantly lower (p<0.05) than those of the non-laser-treated control mice. The percentage of surviving mice that demonstrated full recovery at 56 days post-CHI (NSS=0, as in intact mice) was the highest (63%) in the group that had received TLT in the PW mode at 100 Hz. In addition, magnetic resonance imaging (MRI) analysis demonstrated significantly smaller infarct lesion volumes in laser-treated mice compared to controls. Our data suggest that non-invasive TLT of mice post-TBI provides a significant long-term functional neurological benefit, and that the pulsed laser mode at 100 Hz is the preferred mode for such treatment.


Subject(s)
Behavior, Animal/physiology , Brain Injuries/therapy , Hyperthermia, Induced/methods , Infrared Rays/therapeutic use , Low-Level Light Therapy/methods , Recovery of Function/physiology , Animals , Brain Injuries/physiopathology , Male , Mice , Mice, Inbred Strains
16.
J Cereb Blood Flow Metab ; 31(8): 1768-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21505478

ABSTRACT

N-arachidonoyl-L-serine (AraS) is a brain component structurally related to the endocannabinoid family. We investigated the neuroprotective effects of AraS following closed head injury induced by weight drop onto the exposed fronto-parietal skull and the mechanisms involved. A single injection of AraS following injury led to a significant improvement in functional outcome, and to reduced edema and lesion volume compared with vehicle. Specific antagonists to CB2 receptors, transient receptor potential vanilloid 1 (TRPV1) or large conductance calcium-activated potassium (BK) channels reversed these effects. Specific binding assays did not indicate binding of AraS to the GPR55 cannabinoid receptor. N-arachidonoyl-L-serine blocked the attenuation in phosphorylated extracellular-signal-regulated kinase 1/2 (ERK) levels and led to an increase in pAkt in both the ipsilateral and contralateral cortices. Increased levels of the prosurvival factor Bcl-xL were evident 24 hours after injury in AraS-treated mice, followed by a 30% reduction in caspase-3 activity, measured 3 days after injury. Treatment with a CB2 antagonist, but not with a CB1 antagonist, reversed this effect. Our results suggest that administration of AraS leads to neuroprotection via ERK and Akt phosphorylation and induction of their downstream antiapoptotic pathways. These protective effects are related mostly to indirect signaling via the CB2R and TRPV1 channels but not through CB1 or GPR55 receptors.


Subject(s)
Apoptosis/drug effects , Arachidonic Acids/pharmacology , Brain Injuries/drug therapy , Serine/analogs & derivatives , Animals , Arachidonic Acids/therapeutic use , Brain Injuries/pathology , Male , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Neuroprotective Agents , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Cannabinoid, CB2/metabolism , Recovery of Function , Serine/pharmacology , Serine/therapeutic use , TRPV Cation Channels/metabolism
17.
Synapse ; 65(7): 634-42, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21108236

ABSTRACT

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.


Subject(s)
Behavior, Animal/physiology , Brain/pathology , Encephalitis/pathology , Inflammation/pathology , Animals , Brain/metabolism , Encephalitis/chemically induced , Encephalitis/metabolism , Fluorescent Antibody Technique , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Male , Mice
18.
Eur J Pharmacol ; 629(1-3): 25-30, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-19958766

ABSTRACT

It has been long thought that hyperactivation of N-methyl-D-aspartate (NMDA) receptors underlies neurological decline after traumatic brain injury. However, all clinical trials with NMDA receptor antagonists failed. Since NMDA receptors are down-regulated from 4h to 2weeks after brain injury, activation at 24h, rather than inhibition, of these receptors, was previously shown to be beneficial in mice. Here, we tested the therapeutic window, dose regimen and mechanism of action of the NMDA receptor partial agonist D-cycloserine (DCS) in traumatic brain injury. Male mice were subjected to trauma using a weight-drop model, and administered 10mg/kg (i.p.) DCS or vehicle once (8, 16, 24, or 72h) twice (24 and 48h) or three times (24, 48 and 72h). Functional recovery was assessed for up to 60days, using a Neurological Severity Score that measures neurobehavioral parameters. In all groups in which treatment was begun at 24 or 72h neurobehavioral function was significantly better than in the vehicle-treated groups. Additional doses, on days 2 and 3 did not further improve recovery. Mice treated at 8h or 16h post injury did not differ from the vehicle-treated controls. Co-administration of the NMDA receptor antagonist MK-801 completely blocked the protective effect of DCS given at 24h. Infarct volume measured by 2,3,5-triphenyltetrazolium chloride staining at 48h or by cresyl violet at 28days was not affected by DCS treatment. Since DCS is used clinically for other indications, the present study offers a novel approach for treating human traumatic brain injury with a therapeutic window of at least 24h.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/physiopathology , Cycloserine/administration & dosage , Cycloserine/therapeutic use , Recovery of Function/drug effects , Animals , Brain Injuries/metabolism , Cycloserine/metabolism , Cycloserine/pharmacology , Dizocilpine Maleate/pharmacology , Drug Administration Schedule , Male , Mice , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors , Treatment Outcome
19.
J Cereb Blood Flow Metab ; 30(3): 616-27, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19904288

ABSTRACT

Heat acclimation (HA) offers functional neuroprotection in mice after traumatic brain injury (TBI). This study further characterizes endogenous neuroprotection acquired by HA (34+/-1 degrees C, 30 d) after TBI. We establish here the ability of HA to induce sustained functional benefits and to reduce activation of apoptotic pathways. Neurobehavioral recovery, assessed by the Neurological Severity Score, was greater in HA mice up to 8 days after injury as compared with normothermic controls (P<0.05) and lesion volume was also smaller in the HA group (P<0.05). Reduced apoptotic cell death in HA mice was confirmed using caspase-3 activity measurements and immunohistochemistry. To investigate the underlying molecular pathways, expression levels of intrinsic apoptotic pathway-related proteins were examined. HA mice displayed higher mitochondrial levels of antiapoptotic Bcl-xL, accompanied by lower proapoptotic Bad levels and decreased cytochrome c release, suggesting a higher apoptotic threshold. Taken together with our previous reports, indicating increased Akt phosphorylation and antioxidative capacity, alongside with reduced tumor necrosis alpha levels after TBI in HA animals, the current results support the involvement of an antiapoptotic effect in HA-induced neuroprotection. Current results warrant further study as TBI-induced apoptosis may persist over weeks after injury, possibly providing a target for belated therapeutic intervention.


Subject(s)
Acclimatization/physiology , Apoptosis/physiology , Brain Injuries/pathology , Hot Temperature , Recovery of Function/physiology , Animals , Antioxidants/metabolism , Behavior, Animal/physiology , Blotting, Western , Brain Injuries/physiopathology , Caspase 3/metabolism , Cytochromes c/biosynthesis , Heart/physiopathology , In Situ Nick-End Labeling , Male , Mice , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , bcl-Associated Death Protein/biosynthesis , bcl-X Protein/biosynthesis
20.
FASEB J ; 23(12): 4266-75, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19723705

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Brain Injuries/drug therapy , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Neuroglia/drug effects , Animals , Brain/pathology , Caspase 3/metabolism , Hydroxamic Acids/administration & dosage , Male , Mice , Neuroprotective Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL