Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Neural Plast ; 2021: 5554824, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093701

RESUMO

Traumatic axonal injury (TAI) is a major cause of death and disability among patients with severe traumatic brain injury (TBI); however, no effective therapies have been developed to treat this disorder. Neuroinflammation accompanying microglial activation after TBI is likely to be an important factor in TAI. In this review, we summarize the current research in this field, and recent studies suggest that microglial activation plays an important role in TAI development. We discuss several drugs and therapies that may aid TAI recovery by modulating the microglial phenotype following TBI. Based on the findings of recent studies, we conclude that the promotion of active microglia to the M2 phenotype is a potential drug target for the treatment of TAI.


Assuntos
Lesão Axonal Difusa/tratamento farmacológico , Microglia/efeitos dos fármacos , Animais , Lesões Encefálicas Traumáticas/tratamento farmacológico , Encefalite/tratamento farmacológico , Encefalite/etiologia , Humanos
2.
Sci Rep ; 10(1): 7815, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32385407

RESUMO

A high-resolution, three-dimensional, optical imaging technique for the murine brain was developed to identify the effects of different therapeutic windows for preclinical brain research. This technique tracks the same cells over several weeks. We conducted a pilot study of a promising drug to treat diffuse axonal injury (DAI) caused by traumatic brain injury, using two different therapeutic windows, as a means to demonstrate the utility of this novel longitudinal imaging technique. DAI causes immediate, sporadic axon damage followed by progressive secondary axon damage. We administered minocycline for three days commencing one hour after injury in one treatment group and beginning 72 hours after injury in another group to demonstrate the method's ability to show how and when the therapeutic drug exerts protective and/or healing effects. Fewer varicosities developed in acutely treated mice while more varicosities resolved in mice with delayed treatment. For both treatments, the drug arrested development of new axonal damage by 30 days. In addition to evaluation of therapeutics for traumatic brain injury, this hybrid microlens imaging method should be useful to study other types of brain injury and neurodegeneration and cellular responses to treatment.


Assuntos
Axônios/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Lesão Axonal Difusa/tratamento farmacológico , Minociclina/farmacologia , Animais , Axônios/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Lesões Encefálicas/diagnóstico por imagem , Lesões Encefálicas/patologia , Lesão Axonal Difusa/diagnóstico por imagem , Lesão Axonal Difusa/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Imagem Óptica
3.
J Cell Biochem ; 121(1): 430-442, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31232487

RESUMO

BACKGROUND: Nuclear factor erythroid 2-related factor 2 (Nrf2) can alleviate diffuse axonal injury (DAI)-induced apoptosis by regulating expression of heme oxygenase-1 (HO-1), while sulforaphane (SFN) was shown to reduce oxidative stress by increasing the expression of Nrf2. Therefore, we aimed to investigate therapeutic effect of SFN in the treatment of DAI and the ability of SFN to reduce oxidative stress. METHODS: The 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was used to observe the effects of H2 O 2 and SFN on cell viability. Fluorometric assay, Western blot analysis, and flow cytometry were conducted to validate the protective role of SFN in an animal model of DAI. In addition, the levels of malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GPx) were measured in DAI rats treated by SFN, while Western blot, immunohistochemistry assay, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay were carried out to verify the effect of SFN in different animal groups. RESULTS: Cell viability was reduced by H2 O 2 in a dose-dependent manner, while the treatment by SFN significantly promoted cell growth. Meanwhile the administration of SFN effectively reduced the levels of caspase-3/poly(ADP-ribose) polymerase (PARP) activity increased by the H 2 O 2 treatment, indicating that the protective effect of SFN could be mediated by its ability to suppress caspase-3 activation and PARP cleavage. In addition, the SFN treatment reduced the intracellular reactive oxygen species (ROS) generation induced by H 2 O 2 . Moreover, the MDA levels of SOD/GPx activity in various rat groups showed the protective effects of SFN in DAI rats. It is suspected that the protective effect of SFN was exerted via the activation of the Nrf2/HO-1 signaling pathway. In this study, DAI and DAI + phosphate-buffered saline (PBS) groups also showed the presence of more TUNEL-positive cells compared with the sham-operated group, while the SFN treatment reduced the extent of neuronal apoptosis. CONCLUSIONS: By activating the Nrf2/HO-1 signaling pathway and reducing the activity of caspase-3, SFN reduces the apoptosis of neurons in brain trauma-induced DAI.


Assuntos
Axônios/metabolismo , Axônios/patologia , Lesão Axonal Difusa/tratamento farmacológico , Heme Oxigenase (Desciclizante)/metabolismo , Isotiocianatos/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Apoptose , Sobrevivência Celular , Lesão Axonal Difusa/metabolismo , Regulação da Expressão Gênica , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/química , Masculino , Malondialdeído/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Sulfóxidos , Superóxido Dismutase/metabolismo , Resultado do Tratamento
4.
Cell Death Dis ; 10(10): 727, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31562294

RESUMO

Mild traumatic brain injury (mTBI) is a frequently overlooked public health concern that is difficult to diagnose and treat. Diffuse axonal injury (DAI) is a common mTBI neuropathology in which axonal shearing and stretching induces breakdown of the cytoskeleton, impaired axonal trafficking, axonal degeneration, and cognitive dysfunction. DAI is becoming recognized as a principal neuropathology of mTBI with supporting evidence from animal model, human pathology, and neuroimaging studies. As mitochondrial dysfunction and calcium overload are critical steps in secondary brain and axonal injury, we investigated changes in protein expression of potential targets following mTBI using an in vivo controlled cortical impact model. We show upregulated expression of sodium calcium exchanger1 (NCX1) in the hippocampus and cortex at distinct time points post-mTBI. Expression of dynamin-related protein1 (Drp1), a GTPase responsible for regulation of mitochondrial fission, also changes differently post-injury in the hippocampus and cortex. Using an in vitro model of DAI previously reported by our group, we tested whether pharmacological inhibition of NCX1 by SN-6 and of dynamin1, dynamin2, and Drp1 by dynasore mitigates secondary damage. Dynasore and SN-6 attenuate stretch injury-induced swelling of axonal varicosities and mitochondrial fragmentation. In addition, we show that dynasore, but not SN-6, protects against H2O2-induced damage in an organotypic oxidative stress model. As there is currently no standard treatment to mitigate cell damage induced by mTBI and DAI, this work highlights two potential therapeutic targets for treatment of DAI in multiple models of mTBI and DAI.


Assuntos
Concussão Encefálica/tratamento farmacológico , Lesão Axonal Difusa/tratamento farmacológico , Dinaminas/uso terapêutico , Trocador de Sódio e Cálcio/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Neuroproteção
5.
Exp Cell Res ; 383(2): 111546, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31398352

RESUMO

Diffuse axonal injury (DAI) is the predominant effect of severe traumatic brain injury and significantly contributes to cognitive deficits. The mechanisms that underlie these cognitive deficits are often associated with complex molecular alterations. α7nAChR, one of the abundant and widespread nicotinic acetylcholine receptors (nAChRs) in the brain, plays important physiological functions in the central nervous system. However, the relationship between temporospatial alterations in the α7nAChR and DAI-related learning and memory dysfunction are not completely understood. Our study detected temporospatial alterations of α7nAChR in vulnerable areas (hippocampus, internal capsule, corpus callosum and brain stem) of DAI rats and evaluated the development and progression of learning and memory dysfunction via the Morris water maze (MWM). We determined that α7nAChR expression in vulnerable areas was mainly reduced at the recovery of DAI in rats. Moreover, the escape latency of the injured group increased significantly and the percentages of the distance travelled and time spent in the target quadrant were significantly decreased after DAI. Furthermore, α7nAChR expression in the vulnerable area was significantly positively correlated with MWM performance after DAI according to regression analysis. In addition, we determined that a selective α7nAChR agonist significantly improved learning and memory dysfunction. Rats in the α7nAChR agonist group showed better learning and memory performance than those in the antagonist group. These results demonstrate that microstructural injury-induced alterations of α7nAChR in the vulnerable area are significantly correlated with learning and memory dysfunctions after DAI and that augmentation of the α7nAChR level by its agonist contributes to the improvement of learning and memory function.


Assuntos
Aconitina/análogos & derivados , Benzamidas/farmacologia , Compostos Bicíclicos com Pontes/farmacologia , Disfunção Cognitiva/psicologia , Lesão Axonal Difusa/psicologia , Aprendizagem/efeitos dos fármacos , Memória/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7/fisiologia , Aconitina/farmacologia , Animais , Benzamidas/uso terapêutico , Compostos Bicíclicos com Pontes/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Lesão Axonal Difusa/complicações , Lesão Axonal Difusa/tratamento farmacológico , Lesão Axonal Difusa/patologia , Modelos Animais de Doenças , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores
6.
Air Med J ; 38(1): 55-57, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30711088

RESUMO

This case considers underdosing of analgesics as a prime contributor to hypertension in diffuse axonal injury (DAI) patients who are being mechanically ventilated. In the air medical environment, obtunded patients' hemodynamic parameters are the primary tools available in diagnosing complex disorders such as an acute rise in intracranial pressure (ICP) when invasive ICP monitoring is not available. Therefore, differential diagnoses must follow a continuum, from most severe to least, in order to deal with sudden-onset hypertension rapidly. Not until all critical differentials have been eliminated is analgesia considered. Mimicking the signs of ICP, a compensatory rise in the mean arterial pressure (MAP) is displayed in an acute pain response for mechanically ventilated patients. Therefore, poor analgesic coverage should be considered early in DAI patients who are being ventilated, especially when an increased metabolic drive may be occurring, forcing the therapeutic dosing intervals to be increased. This patient was transferred from Europe back to North America via a fixed wing aircraft, a 16-hour transport time.


Assuntos
Analgésicos/uso terapêutico , Lesão Axonal Difusa/complicações , Lesão Axonal Difusa/tratamento farmacológico , Serviços Médicos de Emergência/métodos , Hipertensão/tratamento farmacológico , Hipertensão/etiologia , Pressão Intracraniana/efeitos dos fármacos , Adulto , Lesão Axonal Difusa/mortalidade , Europa (Continente) , Evolução Fatal , Feminino , Humanos , Monitorização Fisiológica/métodos
7.
J Med Life ; 12(4): 468-470, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32025270

RESUMO

We report a case of traumatic brain injury treated with Cerebrolysin, a neurorecovery stimulating agent. Our therapeutic approach was based on the pathophysiology of traumatic brain injury and, in particular, of diffuse axonal injury. The patient registered marked improvement in mood and cognitive performance, indicating the effectiveness of multimodal and multidisciplinary interventions after traumatic brain injury.


Assuntos
Acidentes de Trânsito , Aminoácidos/uso terapêutico , Lesão Axonal Difusa/tratamento farmacológico , Velocidade do Fluxo Sanguíneo , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/fisiopatologia , Lesão Axonal Difusa/fisiopatologia , Humanos , Masculino , Adulto Jovem
8.
Inflammation ; 42(3): 841-856, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30488141

RESUMO

Our early experiments confirmed that rosiglitazone (RSG), a peroxisome proliferator-activated receptor γ (PPARγ) agonist, had therapeutic potential for the treatment of diffuse axonal injury (DAI) by inhibiting the expression of amyloid-beta precursor protein and reducing the loss and abnormal phosphorylation of tau, but the underlying mechanisms were not fully defined. In this study, we aimed to investigate a possible role for PPARγ in the protection of blood-brain barrier (BBB) integrity in a rat model of DAI, and the underlying mechanisms. PPAR agonists and antagonists were intraperitoneally injected after DAI. Treatment with RSG ameliorated axonal injury, cell apoptosis, glia activation, and the release of inflammatory factors such as TNF-α, IL-1ß, and IL-6. It also increased the expression of tight junction-associated proteins like ZO-1, claudin-5, and occludin-1, whereas the PPARγ antagonist GW9662 had the opposite effects. These effects were also studied in a BBB in vitro model, consisting of a monolayer of human microvascular endothelial cells (HBMECs) subjected to oxygen and glucose deprivation (OGD). Treatment with RSG ameliorated the loss of BBB integrity and the increased permeability induced by OGD by reducing the release of inflammatory factors and maintaining the expression of tight junction-associated proteins. Interestingly, caveolin-1 was found located mainly in endothelial cells, and RSG increased the expression of caveolin-1, which decreased following OGD. In contrast, caveolin-1 siRNA abrogated the protective effects of RSG in the in vitro BBB model. In conclusion, we provide evidence that PPARγ plays an important role in a series of processes associated with DAI, and that the PPARγ agonist RSG can protect BBB integrity by decreasing the levels of inflammatory mediators through a caveolin-1-dependent pathway.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Caveolina 1/metabolismo , Lesão Axonal Difusa/patologia , Mediadores da Inflamação/metabolismo , PPAR gama/agonistas , Rosiglitazona/farmacologia , Animais , Células Cultivadas , Lesão Axonal Difusa/tratamento farmacológico , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Glucose/deficiência , Humanos , Ratos , Rosiglitazona/uso terapêutico , Proteínas de Junções Íntimas/metabolismo
9.
Neuroreport ; 29(8): 661-677, 2018 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-29570500

RESUMO

Diffuse axonal injury (DAI) accounts for more than 50% of all traumatic brain injury. In response to the mechanical damage associated with DAI, the abnormal proteins produced in the neurons and axons, namely, ß-APP and p-tau, induce endoplasmic reticulum (ER) stress. Curcumin, a major component extracted from the rhizome of Curcuma longa, has shown potent anti-inflammatory, antioxidant, anti-infection, and antitumor activity in previous studies. Moreover, curcumin is an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2) and promotes its nuclear translocation. In this study, we evaluated the therapeutic potential of curcumin for the treatment of DAI and investigated the mechanisms underlying the protective effects of curcumin against neural cell death and axonal injury after DAI. Rats subjected to a model of DAI by head rotational acceleration were treated with vehicle or curcumin to evaluate the effect of curcumin on neuronal and axonal injury. We observed that curcumin (20 mg/kg intraperitoneal) administered 1 h after DAI induction alleviated the aggregation of p-tau and ß-APP in neurons, reduced ER-stress-related cell apoptosis, and ameliorated neurological deficits. Further investigation showed that the protective effect of curcumin in DAI was mediated by the PERK/Nrf2 pathway. Curcumin promoted PERK phosphorylation, and then Nrf2 dissociated from Keap1 and was translocated to the nucleus, which activated ATF4, an important bZIP transcription factor that maintains intracellular homeostasis, but inhibited the CHOP, a hallmark of ER stress and ER-associated programmed cell death. In summary, we demonstrate for the first time that curcumin confers protection against abnormal proteins and neuronal apoptosis after DAI, that the process is mediated by strengthening of the unfolded protein response to overcome ER stress, and that the protective effect of curcumin against DAI is dependent on the activation of Nrf2.


Assuntos
Apoptose/efeitos dos fármacos , Axônios/efeitos dos fármacos , Curcumina/farmacologia , Lesão Axonal Difusa/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Apoptose/fisiologia , Axônios/metabolismo , Axônios/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesão Axonal Difusa/metabolismo , Lesão Axonal Difusa/patologia , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/fisiologia , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fosforilação/efeitos dos fármacos , Distribuição Aleatória , Ratos Sprague-Dawley , eIF-2 Quinase/metabolismo
10.
Int J Mol Sci ; 18(12)2017 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-29207487

RESUMO

Traumatic brain injury (TBI) is one of the world's leading causes of morbidity and mortality among young individuals. TBI applies powerful rotational and translational forces to the brain parenchyma, which results in a traumatic diffuse axonal injury (DAI) responsible for brain swelling and neuronal death. Following TBI, axonal degeneration has been identified as a progressive process that starts with disrupted axonal transport causing axonal swelling, followed by secondary axonal disconnection and Wallerian degeneration. These modifications in the axonal cytoskeleton interrupt the axoplasmic transport mechanisms, causing the gradual gathering of transport products so as to generate axonal swellings and modifications in neuronal homeostasis. Oxidative stress with consequent impairment of endogenous antioxidant defense mechanisms plays a significant role in the secondary events leading to neuronal death. Studies support the role of an altered axonal calcium homeostasis as a mechanism in the secondary damage of axon, and suggest that calcium channel blocker can alleviate the secondary damage, as well as other mechanisms implied in the secondary injury, and could be targeted as a candidate for therapeutic approaches. Reactive oxygen species (ROS)-mediated axonal degeneration is mainly caused by extracellular Ca2+. Increases in the defense mechanisms through the use of exogenous antioxidants may be neuroprotective, particularly if they are given within the neuroprotective time window. A promising potential therapeutic target for DAI is to directly address mitochondria-related injury or to modulate energetic axonal energy failure.


Assuntos
Cálcio/metabolismo , Lesão Axonal Difusa/patologia , Estresse Oxidativo , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Lesão Axonal Difusa/tratamento farmacológico , Lesão Axonal Difusa/metabolismo , Humanos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
11.
PLoS One ; 12(5): e0178049, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28542502

RESUMO

Many mechanisms or pathways are involved in secondary post-traumatic brain injury, such as the ubiquitin-proteasome pathway (UPP), axonal degeneration and neuronal cell apoptosis. UCH-L1 is a protein that is expressed in high levels in neurons and may have important roles in the UPP, autophagy and axonal integrity. The current study aims to evaluate the role of UCH-L1 in post-traumatic brain injury (TBI) and its potential therapeutic effects. A novel protein was constructed that fused the protein transduction domain (PTD) of trans-activating transduction (TAT) protein with UCH-L1 (TAT-UCH-L1) in order to promote neuronal transduction. The TAT-UCH-L1 protein was readily detected in brain by immunoblotting and immunohistochemistry after i.p. administration in mice. TBI was induced in mice using the controlled cortical impact (CCI) model. TAT-UCH-L1 treatment significantly attenuated K48-linkage polyubiquitin (polyUb)-protein accumulation in hippocampus after CCI compared to vehicle controls, but had no effects on K65-linkage polyUb-protein. TAT-UCH-L1 treatment also attenuated expression of Beclin-1 and LC3BII after CCI. TAT-UCH-L1-treated mice had significantly increased spared tissue volumes and increased survival of CA3 neurons 21 d after CCI compared to control vehicle-treated mice. Axonal injury, detected by APP immunohistochemistry, was reduced in thalamus 24 h and 21 d after CCI in TAT-UCH-L1-treated mice. These results suggest that TAT-UCH-L1 treatment improves function of the UPP and decreases activation of autophagy after CCI. Furthermore, TAT-UCH-L1 treatment also attenuates axonal injury and increases hippocampal neuronal survival after CCI. Taken together these results suggest that UCH-L1 may play an important role in the pathogenesis of cell death and axonal injury after TBI.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/patologia , Lesão Axonal Difusa/tratamento farmacológico , Lesão Axonal Difusa/prevenção & controle , Proteínas Recombinantes de Fusão/uso terapêutico , Ubiquitina Tiolesterase/uso terapêutico , Animais , Autofagia/fisiologia , Proteína Beclina-1/biossíntese , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Lesão Axonal Difusa/patologia , Camundongos , Proteínas Associadas aos Microtúbulos/biossíntese , Neurônios/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterase/metabolismo
12.
Mol Med Rep ; 15(5): 3001-3010, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28339015

RESUMO

Diffuse axonal injury (DAI) is the most common and significant pathological features of traumatic brain injury (TBI). However, there are still no effective drugs to combat the formation and progression of DAI in affected individuals. FK506, also known as tacrolimus, is an immunosuppressive drug, which is widely used in transplantation medicine for the reduction of allograft rejection. Previous studies have identified that FK506 may play an important role in the nerve protective effect of the central nervous system. In the present study, apoptosis of neuronal cells was observed following the induction of experimental DAI. The results demonstrated that it was closely related with the upregulation of death­associated protein kinase 1 (DAPK1). It was hypothesized that FK506 may inhibit the activity of DAPK1 by inhibiting calcineurin activity, which may be primarily involved in anti­apoptosis following DAI induction. Through researching the expression of nerve regeneration associated proteins (NF­H and GAP­43) following DAI, the present study provides novel data to suggest that FK506 promotes axon formation and nerve regeneration following experimental DAI. Therefore, FK506 may be a potent therapeutic for inhibiting nerve injury, as well as promoting the nerve regeneration following DAI.


Assuntos
Apoptose/efeitos dos fármacos , Axônios/efeitos dos fármacos , Lesão Axonal Difusa/tratamento farmacológico , Tacrolimo/farmacologia , Animais , Axônios/metabolismo , Axônios/patologia , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/patologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/patologia , Calcineurina/efeitos dos fármacos , Proteínas Quinases Associadas com Morte Celular/antagonistas & inibidores , Proteínas Quinases Associadas com Morte Celular/metabolismo , Lesão Axonal Difusa/metabolismo , Lesão Axonal Difusa/patologia , Proteína GAP-43/metabolismo , Masculino , Regeneração Nervosa/efeitos dos fármacos , Proteínas de Neurofilamentos/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima
13.
Exp Neurol ; 292: 80-91, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28274861

RESUMO

CHIMERA (Closed Head Impact Model of Engineered Rotational Acceleration) is a recently described animal model of traumatic brain injury (TBI) that primarily produces diffuse axonal injury (DAI) characterized by white matter inflammation and axonal damage. CHIMERA was specifically designed to reliably generate a variety of TBI severities using precise and quantifiable biomechanical inputs in a nonsurgical user-friendly platform. The objective of this study was to define the lower limit of single impact mild TBI (mTBI) using CHIMERA by characterizing the dose-response relationship between biomechanical input and neurological, behavioral, neuropathological and biochemical outcomes. Wild-type male mice were subjected to a single CHIMERA TBI using six impact energies ranging from 0.1 to 0.7J, and post-TBI outcomes were assessed over an acute period of 14days. Here we report that single TBI using CHIMERA induces injury dose- and time-dependent changes in behavioral and neurological deficits, axonal damage, white matter tract microgliosis and astrogliosis. Impact energies of 0.4J or below produced no significant phenotype (subthreshold), 0.5J led to significant changes for one or more phenotypes (threshold), and 0.6 and 0.7J resulted in significant changes in all outcomes assessed (mTBI). We further show that linear head kinematics are the most robust predictors of duration of unconsciousness, severity of neurological deficits, white matter injury, and microgliosis following single TBI. Our data extend the validation of CHIMERA as a biofidelic animal model of DAI and establish working parameters to guide future investigations of the mechanisms underlying axonal pathology and inflammation induced by mechanical trauma.


Assuntos
Axônios/efeitos dos fármacos , Concussão Encefálica/fisiopatologia , Encéfalo/efeitos dos fármacos , Lesão Axonal Difusa/tratamento farmacológico , Animais , Axônios/patologia , Fenômenos Biomecânicos/efeitos dos fármacos , Encéfalo/patologia , Concussão Encefálica/patologia , Concussão Encefálica/terapia , Lesão Axonal Difusa/patologia , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL
14.
Brain Inj ; 31(1): 16-23, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27819489

RESUMO

BACKGROUND AND OBJECTIVE: The benefits of progesterone have been demonstrated in the animal models of traumatic brain injury (TBI). However, the results of clinical studies are conflicting. Considering the heterogenic nature of TBI, the effect of progesterone in patients with diffuse axonal injury (DAI) was investigated in a clinical trial. METHODS: In this study, 48 patients with DAI and Glasgow Coma Scale of 3-12, admitted within 4 hours after injury, were randomly assigned to the progesterone or control group. The dose of progesterone administration was 1 mg kg-1 per 12 hours for 5 days. The effect of progesterone was investigated using extended-Glasgow Outcome Scale (GOS-E), functional independence measure (FIM) scores and also mortality within the follow-up period. RESULTS: The progesterone group exhibited higher GOS-E and FIM scores in comparison to the control group at 6 months post-injury (p < 0.01 and p < 0.05, respectively). Mortality was also found in the control group (p < 0.05). The adverse events attributed to the progesterone administration were not found throughout the study. CONCLUSIONS: Findings of this study suggest that progesterone may be neuroprotective in patients with DAI. However, large clinical trials are needed to assess progesterone as a promising drug in DAI.


Assuntos
Lesão Axonal Difusa/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Progesterona/uso terapêutico , Adulto , Lesão Axonal Difusa/mortalidade , Escala de Resultado de Glasgow , Humanos , Masculino , Método Simples-Cego , Taxa de Sobrevida , Resultado do Tratamento
15.
J Neurotrauma ; 33(16): 1501-13, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-26530250

RESUMO

Diffuse axonal injury is recognized as a progressive and long-term consequence of traumatic brain injury. Axonal injury can have sustained negative consequences on neuronal functions such as anterograde and retrograde transport and cellular processes such as autophagy that depend on cytoarchitecture and axon integrity. These changes can lead to somatic atrophy and an inability to repair and promote plasticity. Obstruction of the autophagic process has been noted after brain injury, and rapamycin, a drug used to stimulate autophagy, has demonstrated positive effects in brain injury models. The optimization of drugs to promote beneficial autophagy without negative side effects could be used to attenuate traumatic brain injury and promote improved outcome. Lanthionine ketimine ethyl ester, a bioavailable derivative of a natural sulfur amino acid metabolite, has demonstrated effects on autophagy both in vitro and in vivo. Thirty minutes after a moderate central fluid percussion injury and throughout the survival period, lanthionine ketimine ethyl ester was administered, and mice were subsequently evaluated for learning and memory impairments and biochemical and histological changes over a 5-week period. Lanthionine ketimine ethyl ester, which we have shown previously to modulate autophagy markers and alleviate pathology and slow cognitive decline in the 3 × TgAD mouse model, spared cognition and pathology after central fluid percussion injury through a mechanism involving autophagy modulation.


Assuntos
Aminoácidos Sulfúricos/farmacologia , Autofagia/efeitos dos fármacos , Lesão Axonal Difusa/tratamento farmacológico , Aminoácidos Sulfúricos/administração & dosagem , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL
16.
J Neurotrauma ; 32(16): 1210-6, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25646701

RESUMO

Estrogen plays an important role as a neuroprotector in the central nervous system (CNS), directly interacting with neurons and regulating physiological properties of non-neuronal cells. Here we evaluated estrogen sulfate (E2-SO4) for traumatic brain injury (TBI) using a Sprague-Dawley rat model. TBI was induced via lateral fluid percussion (LFP) at 24 h after craniectomy. E2-SO4 (1 mg/kg BW in 1 mL/kg BW) or saline (served as control) was intravenously administered at 1 h after TBI (n=5/group). Intracranial pressure (ICP), cerebral perfusion pressure (CPP), and partial brain oxygen pressure (pbtO2) were measured for 2 h (from 23 to 25 h after E2-SO4 injection). Brain edema and diffuse axonal injury (DAI) were assessed by diffusion tensor imaging (DTI), and cerebral glycolysis was measured by (18)F-labeled fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging, at 1 and 7 days after E2-SO4 injection. E2-SO4 significantly decreased ICP, while increasing CPP and pbtO2 (p<0.05) as compared with vehicle-treated TBI rats. The edema size in the brains of the E2-SO4 treated group was also significantly smaller than that of vehicle-treated group at 1 day after E2-SO4 injection (p=0.04), and cerebral glycolysis of injured region was also increased significantly during the same time period (p=0.04). However, E2-SO4 treatment did not affect DAI (p>0.05). These findings demonstrated the potential benefits of E2-SO4 in TBI.


Assuntos
Edema Encefálico/tratamento farmacológico , Lesões Encefálicas/tratamento farmacológico , Estrona/análogos & derivados , Glicólise/efeitos dos fármacos , Animais , Edema Encefálico/diagnóstico , Edema Encefálico/etiologia , Lesões Encefálicas/complicações , Lesões Encefálicas/diagnóstico , Lesão Axonal Difusa/diagnóstico , Lesão Axonal Difusa/tratamento farmacológico , Imagem de Tensor de Difusão , Modelos Animais de Doenças , Estrona/administração & dosagem , Estrona/farmacologia , Fluordesoxiglucose F18 , Masculino , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Ratos , Ratos Sprague-Dawley
17.
Brain Inj ; 27(12): 1454-60, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24088189

RESUMO

OBJECTIVES: This pilot trial was conducted to establish whether Boswellia Serrata (BS), a traditional herbal medicine, could improve the outcome of patients who have diffuse axonal injury (DAI). METHODS: In total, 38 patients with pure DAI were enrolled in this 12-week, double-blind, randomized, cross-over study. The patients were randomly assigned to receive either placebo (group A, n = 20) or BS capsules (group B, n = 18) for 6 weeks and then switched to the other intervention for another 6 weeks. The disability rating scale (DRS) was used to assess the outcome at 2-, 6- and 12-weeks post-trauma. RESULTS: A non-significant trend for improvement of DRS total scores was observed after the use of BS. Regarding the DRS sub-scores, however, there was significant improvement in 'cognitive ability to self-care' during the second 6 weeks in group A on BS compared to an insignificant spontaneous recovery in group B during the same period on placebo. Moreover, both groups experienced a close-to-significant increase in the cognitive function-related items of the DRS during the periods they were on BS. The reported adverse events were all of mild quality and had similar frequency between the groups. CONCLUSION: These results suggest that BS resin does not significantly affect general outcome, but may enhance the cognitive outcome of patients with DAI.


Assuntos
Boswellia , Cognição/efeitos dos fármacos , Lesão Axonal Difusa/tratamento farmacológico , Fitoterapia , Preparações de Plantas/uso terapêutico , Autocuidado/estatística & dados numéricos , Acidentes de Trânsito/estatística & dados numéricos , Adulto , Estudos Cross-Over , Lesão Axonal Difusa/epidemiologia , Lesão Axonal Difusa/fisiopatologia , Avaliação da Deficiência , Método Duplo-Cego , Feminino , Escala de Coma de Glasgow , Humanos , Escala de Gravidade do Ferimento , Irã (Geográfico)/epidemiologia , Masculino , Fitoterapia/métodos , Projetos Piloto , Recuperação de Função Fisiológica , Resultado do Tratamento
18.
Clin Neurol Neurosurg ; 115(10): 2019-22, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23871679

RESUMO

OBJECTIVE: Severe traumatic brain injury (TBI) has a major role in mortality rate among the other types of trauma. The aim of this clinical study was to assess the effect of progesterone on the improvement of neurologic outcome in patients with acute severe TBI. METHODS: A total of 76 patients who had arrived within 8h of injury with a Glasgow Coma Score≤8 were enrolled in the study. In a randomized style 38 received progesterone (1mg/kg per 12h for 5 days) and 38 did not. RESULTS: There was a better recovery rate and GOS score for the patients who were given progesterone than for those in the control group in a 3-months follow-up period (50% vs. 21%); subgroup analysis showed a significant difference in the percentage of favorable outcome between the two groups with GCS of 5-8 (p=0.03). CONCLUSION: The use of progesterone may significantly improve neurologic outcome of patients suffering severe TBI up to 3 months after injury, especially those with 5≤GCS≤8, providing a potential benefit to the treatment of acute severe TBI patients. Considering this drug had no significant side effects, so progesterone could be used in patients with severe TBI as a neuro-protective drug.


Assuntos
Anti-Inflamatórios/uso terapêutico , Traumatismos Craniocerebrais/tratamento farmacológico , Lesão Axonal Difusa/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Progesterona/uso terapêutico , Adulto , Fatores Etários , Idoso , Lesões Encefálicas/tratamento farmacológico , Traumatismos Craniocerebrais/mortalidade , Traumatismos Craniocerebrais/patologia , Lesão Axonal Difusa/mortalidade , Lesão Axonal Difusa/patologia , Escala de Coma de Glasgow , Escala de Resultado de Glasgow , Humanos , Irã (Geográfico)/epidemiologia , Masculino , Pessoa de Meia-Idade , Estado Vegetativo Persistente/epidemiologia , Prognóstico , Método Simples-Cego , Resultado do Tratamento , Adulto Jovem
19.
Brain Res ; 1490: 184-92, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22982593

RESUMO

Traumatic axonal injury (TAI), a feature of traumatic brain injury (TBI), progressively evolves over hours through impaired axonal transport and is thought to be a major contributor to cognitive dysfunction. In spite of various studies suggesting that pharmacologic or physiologic interventions might reduce TAI, clinical neuroprotective treatments are still unavailable. Edaravone, a free radical scavenger, has been shown to exert neuroprotective effects in animal models of several brain disorders. In this study, to evaluate whether edaravone suppresses TAI following TBI, mice were subjected to weight drop injury and had either edaravone (3.0mg/kg) or saline administered intravenously immediately after impact. Axonal injury and oxidative stress were assessed using immunohistochemistry with antibodies against amyloid precursor protein, a marker of impaired axonal transport, and with 8-hydroxy-2'-deoxyguanosine, a marker of oxidative DNA damage. Edaravone significantly suppressed axonal injury and oxidative stress in the cortex, corpus callosum, and hippocampus 24h after injury. The neuroprotective effects of edaravone were observed in mice receiving 1.0, 3.0, or 10mg/kg of edaravone immediately after impact, but not after 0.3mg/kg of edaravone. With treatment 1h after impact, axonal injury was also significantly suppressed and this therapeutic effect persisted up to 6h after impact. Furthermore, behavioral tests performed 9 days after injury showed memory deficits in saline-treated traumatized mice, which were not evident in the edaravone-treated group. These results suggest that edaravone protects against memory deficits following TBI and that this protection is mediated by suppression of TAI and oxidative stress.


Assuntos
Antipirina/análogos & derivados , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/psicologia , Transtornos Cognitivos/prevenção & controle , Transtornos Cognitivos/psicologia , Lesão Axonal Difusa/tratamento farmacológico , Sequestradores de Radicais Livres/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , 8-Hidroxi-2'-Desoxiguanosina , Animais , Antipirina/uso terapêutico , Lesões Encefálicas/complicações , Cognição/efeitos dos fármacos , Transtornos Cognitivos/etiologia , Dano ao DNA , Desoxiguanosina/análogos & derivados , Desoxiguanosina/sangue , Lesão Axonal Difusa/patologia , Relação Dose-Resposta a Droga , Edaravone , Comportamento Exploratório/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/psicologia , Camundongos , Camundongos Endogâmicos C57BL , Reconhecimento Psicológico/efeitos dos fármacos
20.
J Neurotrauma ; 29(7): 1469-82, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22321027

RESUMO

We have previously presented evidence that the development of secondary traumatic axonal injury is related to the degree of local cerebral blood flow (LCBF) and flow-metabolism uncoupling. We have now tested the hypothesis that augmenting LCBF in the acute stages after brain injury prevents further axonal injury. Data were acquired from rats with or without acetazolamide (ACZ) that was administered immediately following controlled cortical impact injury to increase cortical LCBF. Local cerebral metabolic rate for glucose (LCMRglc) and LCBF measurements were obtained 3 h post-trauma in the same rat via ¹8F-fluorodeoxyglucose and ¹4C-iodoantipyrine co-registered autoradiographic images, and compared to the density of damaged axonal profiles in adjacent sections, and in additional groups at 24 h used to assess different populations of injured axons stereologically. ACZ treatment significantly and globally elevated LCBF twofold above untreated-injured rats at 3 h (p<0.05), but did not significantly affect LCMRglc. As a result, ipsilateral LCMRglc:LCBF ratios were reduced by twofold to sham-control levels, and the density of ß-APP-stained axons at 24 h was significantly reduced in most brain regions compared to the untreated-injured group (p<0.01). Furthermore, early LCBF augmentation prevented the injury-associated increase in the number of stained axons from 3-24 h. Additional robust stereological analysis of impaired axonal transport and neurofilament compaction in the corpus callosum and cingulum underlying the injury core confirmed the amelioration of ß-APP axon density, and showed a trend, but no significant effect, on RMO14-positive axons. These data underline the importance of maintaining flow-metabolism coupling immediately after injury in order to prevent further axonal injury, in at least one population of injured axons.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Circulação Cerebrovascular/efeitos dos fármacos , Lesão Axonal Difusa/tratamento farmacológico , Metabolismo Energético/efeitos dos fármacos , Acetazolamida/farmacologia , Animais , Lesões Encefálicas/diagnóstico por imagem , Circulação Cerebrovascular/fisiologia , Lesão Axonal Difusa/diagnóstico por imagem , Lesão Axonal Difusa/metabolismo , Metabolismo Energético/fisiologia , Masculino , Cintilografia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...