Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
iScience ; 27(4): 109480, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38715940

RESUMO

Ischemic stroke is the second leading cause of death and disability worldwide, and efforts to prevent stroke, mitigate secondary neurological damage, and promote neurological recovery remain paramount. Recent findings highlight the critical importance of microbiome-related metabolites, including vitamin B12 (VB12), in alleviating toxic stroke-associated neuroinflammation. Here, we showed that VB12 tonically programmed genes supporting microglial cell division and activation and critically controlled cellular fatty acid metabolism in homeostasis. Intriguingly, VB12 promoted mitochondrial transcriptional and metabolic activities and significantly restricted stroke-associated gene alterations in microglia. Furthermore, VB12 differentially altered the functions of microglial subsets during the acute phase of ischemic stroke, resulting in reduced brain damage and improved neurological function. Pharmacological depletion of microglia before ischemic stroke abolished VB12-mediated neurological improvement. Thus, our preclinical studies highlight the relevance of VB12 in the functional programming of microglia to alleviate neuroinflammation, minimize ischemic injury, and improve host neurological recovery after ischemic stroke.

2.
Stem Cells ; 41(6): 570-577, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37186298

RESUMO

After ischemia, cells in the brain parenchyma upregulate stromal derived factor 1 (SDF1), driving chemokine receptor CXCR4-mediated migration of adult neural stem cells to the ischemic injury. We discovered a novel regulator of CXCR4 in neural stem cells, low-density lipoprotein receptor related protein 1 (LRP1). We used Nestin-driven knockout of LRP1 and induction of td-tomato in neural stem cells of adult mice. We observed reduced localization of td-tomato positive cells to the lesion, and find disrupted CXCR4-mediated neural stem cell migration in vitro, which is likely driven by LRP1-mediated loss of CXCR4 expression in vivo. Our results suggest that LRP1 is a novel regulator of CXCR4 in neural stem cells. This heretofore unknown interaction between LRP1 and CXCR4 could have significant consequences for multiple aspects of neural stem cell physiology.


Assuntos
Quimiocina CXCL12 , Células-Tronco Neurais , Camundongos , Animais , Quimiocina CXCL12/metabolismo , Células-Tronco Neurais/metabolismo , Movimento Celular/fisiologia , Encéfalo/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Isquemia/metabolismo
3.
Neurotherapeutics ; 18(4): 2707-2721, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34608616

RESUMO

Traumatic brain injury (TBI) remains one of the greatest public health concerns with increasing morbidity and mortality rates worldwide. Our group reported that stimulation of astrocyte mitochondrial metabolism by P2Y1 receptor agonists significantly reduced cerebral edema and reactive gliosis in a TBI model. Subsequent data on the pharmacokinetics (PK) and rapid metabolism of these compounds suggested that neuroprotection was likely mediated by a metabolite, AST-004, which binding data indicated was an adenosine A3 receptor (A3R) agonist. The neuroprotective efficacy of AST-004 was tested in a control closed cortical injury (CCCI) model of TBI in mice. Twenty-four (24) hours post-injury, mice subjected to CCCI and treated with AST-004 (0.22 mg/kg, injected 30 min post-trauma) exhibited significantly less secondary brain injury. These effects were quantified with less cell death (PSVue794 fluorescence) and loss of blood brain barrier breakdown (Evans blue extravasation assay), compared to vehicle-treated TBI mice. TBI-treated mice also exhibited significantly reduced neuroinflammatory markers, glial-fibrillary acidic protein (GFAP, astrogliosis) and ionized Ca2+-binding adaptor molecule 1 (Iba1, microgliosis), both at the mRNA (qRT-PCR) and protein (Western blot and immunofluorescence) levels, respectively. Four (4) weeks post-injury, both male and female TBI mice presented a significant reduction in freezing behavior during contextual fear conditioning (after foot shock). AST-004 treatment prevented this TBI-induced impairment in male mice, but did not significantly affect impairment in female mice. Impairment of spatial memory, assessed 24 and 48 h after the initial fear conditioning, was also reduced in AST-004-treated TBI-male mice. Female TBI mice did not exhibit memory impairment 24 and 48 h after contextual fear conditioning and similarly, AST-004-treated female TBI mice were comparable to sham mice. Finally, AST-004 treatments were found to increase in vivo ATP production in astrocytes (GFAP-targeted luciferase activity), consistent with the proposed mechanism of action. These data reveal AST-004 as a novel A3R agonist that increases astrocyte energy production and enhances their neuroprotective efficacy after brain injury.


Assuntos
Lesões Encefálicas Traumáticas , Fármacos Neuroprotetores , Adenosina/metabolismo , Adenosina/farmacologia , Animais , Astrócitos/metabolismo , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Feminino , Gliose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico
4.
J Cereb Blood Flow Metab ; 40(6): 1256-1273, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31272312

RESUMO

Nearly three million people in the USA suffer traumatic brain injury (TBI) yearly; however, there are no pre- or post-TBI treatment options available. KCNQ2-5 voltage-gated K+ channels underlie the neuronal "M current", which plays a dominant role in the regulation of neuronal excitability. Our strategy towards prevention of TBI-induced brain damage is predicated on the suggested hyper-excitability of neurons induced by TBIs, and the decrease in neuronal excitation upon pharmacological augmentation of M/KCNQ K+ currents. Seizures are very common after a TBI, making further seizures and development of epilepsy disease more likely. Our hypothesis is that TBI-induced hyperexcitability and ischemia/hypoxia lead to metabolic stress, cell death and a maladaptive inflammatory response that causes further downstream morbidity. Using the mouse controlled closed-cortical impact blunt TBI model, we found that systemic administration of the prototype M-channel "opener", retigabine (RTG), 30 min after TBI, reduces the post-TBI cascade of events, including spontaneous seizures, enhanced susceptibility to chemo-convulsants, metabolic stress, inflammatory responses, blood-brain barrier breakdown, and cell death. This work suggests that acutely reducing neuronal excitability and energy demand via M-current enhancement may be a novel model of therapeutic intervention against post-TBI brain damage and dysfunction.


Assuntos
Anticonvulsivantes/farmacologia , Lesões Encefálicas Traumáticas/metabolismo , Carbamatos/farmacologia , Canais de Potássio KCNQ/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenilenodiaminas/farmacologia , Animais , Camundongos , Camundongos Endogâmicos C57BL
5.
Neurosci J ; 2019: 2831501, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31187032

RESUMO

After traumatic brain injury (TBI), multiple ongoing processes contribute to worsening and spreading of the primary injury to create a secondary injury. One major process involves disrupted fluid regulation to create vascular and cytotoxic edema in the affected area. Although understanding of factors that influence edema is incomplete, the astrocyte water channel Aquaporin 4 (AQP4) has been identified as an important mediator and therefore attractive drug target for edema prevention. The FDA-approved drug acetazolamide has been administered safely to patients for years in the United States. To test whether acetazolamide altered AQP4 function after TBI, we utilized in vitro and in vivo models of TBI. Our results suggest that AQP4 localization is altered after TBI, similar to previously published reports. Treatment with acetazolamide prevented AQP4 reorganization, both in human astrocyte in vitro and in mice in vivo. Moreover, acetazolamide eliminated cytotoxic edema in our in vivo mouse TBI model. Our results suggest a possible clinical role for acetazolamide in the treatment of TBI.

6.
Neural Regen Res ; 13(3): 434-439, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29623927

RESUMO

The mechanistic target of rapamycin (mTOR) is an intracellular protein kinase that functions as an energy and nutrient sensor in the cellular microenvironment of neurons. Modulation of mTOR is vital when nutrient and energy sources become limited. Hypoxia, traumatic brain injury, cellular energy states, and growth factors all regulate the phosphorylation and total levels of mTOR in cells. Alterations in the microenvironment induce transduction of signals to downstream proteins by mTOR allowing for cells to make the necessary adjustments to counteract stressors and survive. Progesterone, a hydrophobic steroid hormone, has been shown in studies of non-neural tissue to be a suppressor of mTOR and modulator of mTOR phosphorylation. Our study tested the effects of progesterone on mTOR expression following traumatic brain injury. C57BL/6 mice were treated with progesterone (8 mg/kg) at 1 (intraperitoneal), 6 (subcutaneous), 24 (subcutaneous), and 48 (subcutaneous) hours post closed skull traumatic brain injury. The hippocampus was then harvested 72 hours post injury and prepared for western blot analysis. We found that progesterone significantly decreased total mTOR levels in all groups compared to sham treated with vehicle. This was further confirmed by immunostaining showing decreased cytoplasmic mTOR levels compared to sham. Our study shows progesterone is a significant modulator of mTOR levels in the hippocampus of mice following traumatic brain injury.

7.
Neural Regen Res ; 9(21): 1891-6, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25558238

RESUMO

There are currently no federally approved neuroprotective agents to treat traumatic brain injury. Progesterone, a hydrophobic steroid hormone, has been shown in recent studies to exhibit neuroprotective effects in controlled cortical impact rat models. Akt is a protein kinase known to play a role in cell signaling pathways that reduce edema, inflammation, apoptosis, and promote cell growth in the brain. This study aims to determine if progesterone modulates the phosphorylation of Akt via its threonine 308 phosphorylation site. Phosphorylation at the threonine 308 site is one of several sites responsible for activating Akt and enabling the protein kinase to carry out its neuroprotective effects. To assess the effects of progesterone on Akt phosphorylation, C57BL/6 mice were treated with progesterone (8 mg/kg) at 1 (intraperitonally), 6, 24, and 48 hours (subcutaneously) post closed-skull traumatic brain injury. The hippocampus was harvested at 72 hours post injury and prepared for western blot analysis. Traumatic brain injury caused a significant decrease in Akt phosphorylation compared to sham operation. However, mice treated with progesterone following traumatic brain injury had an increase in phosphorylation of Akt compared to traumatic brain injury vehicle. Our findings suggest that progesterone is a viable treatment option for activating neuroprotective pathways after traumatic brain injury.

8.
BMC Neurosci ; 14: 158, 2013 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-24359611

RESUMO

BACKGROUND: Insulin-like growth factor binding protein-2 (IGFBP-2) regulates the bioavailability, transportation, and localization of insulin-like growth factor-I (IGF-I), an effective neuroprotectant in animal stroke models especially when administered intranasally. Therefore, determining IGFBP-2's endogenous distribution in the normal and ischemic brain is essential in maximizing the neuroprotective potential of the intranasal IGF-I treatment approach. However, current data on IGFBP-2 is limited to mRNA and in situ hybridization studies. The purpose of this study was to determine if there are any changes in IGFBP-2 protein levels and distribution in ischemic brain and also to determine if IGFBPs play a role in the transportation of intranasally administered IGF-I into the brain. RESULTS: Using an in vitro approach, we show that ischemia causes changes in the distribution of IGFBP-2 in primary cortical neurons and astrocytes. In addition, we show using the transient middle cerebral artery occlusion (MCAO) model in mice that there is a significant increase in IGFBP-2 levels in the stroke penumbra and core after 72 h. This correlated with an overall increase in IGF-I after stroke, with the highest levels of IGF-I in the stroke core after 72 h. Brain sections from stroke mice indicate that neurons and astrocytes located in the penumbra both have increased expression of IGFBP-2, however, IGFBP-2 was not detected in microglia. We used binding competition studies to show that intranasally administered exogenous IGF-I uptake into the brain is not receptor mediated and is likely facilitated by IGFBPs. CONCLUSIONS: The change in protein levels indicates that IGFBP-2 plays an IGF-I-dependent and -independent role in the brain's acute (neuroprotection) and chronic (tissue remodeling) response to hypoxic-ischemic injury. Competition studies indicate that IGFBPs may have a role in rapid transportation of exogenous IGF-I from the nasal tissue to the site of injury.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Administração Intranasal , Animais , Astrócitos/metabolismo , Transporte Biológico , Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Fator de Crescimento Insulin-Like I/administração & dosagem , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Microglia/metabolismo , Neurônios/metabolismo , Bulbo Olfatório/metabolismo , Cultura Primária de Células , Ratos
9.
J Neurotrauma ; 30(1): 55-66, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23046422

RESUMO

Traumatic brain injury (TBI) is the leading cause of death and disability in children and young adults. Neuroprotective agents that may promote repair or counteract damage after injury do not currently exist. We recently reported that stimulation of the purinergic receptor subtype P2Y(1)R using 2-methylthioladenosine 5' diphosphate (2MeSADP) significantly reduced cytotoxic edema induced by photothrombosis. Here, we tested whether P2Y(1)R stimulation was neuroprotective after TBI. A controlled closed head injury model was established for mice using a pneumatic impact device. Brains were harvested at 1, 3, or 7 days post-injury and assayed for morphological changes by immunocytochemistry, Western blot analysis, and wet/dry weight. Cerebral edema and expression of both aquaporin type 4 and glial fibrillary acidic protein were increased at all time points examined. Immunocytochemical measurements in both cortical and hippocampal slices also revealed significant neuronal swelling and reactive gliosis. Treatment of mice with 2MeSADP (100 µM) or MRS2365 (100 µM) 30 min after trauma significantly reduced all post-injury symptoms of TBI including edema, neuronal swelling, reactive gliosis, and AQ4 expression. The neuroprotective effect was lost in IP(3)R2-/- mice treated with 2MeSADP. Immunocytochemical labeling of brain slices confirmed that P2Y(1)R expression was defined to cortical and hippocampal astrocytes, but not neurons. Taken together, the data show that stimulation of astrocytic P2Y(1)Rs significantly reduces brain injury after acute trauma and is mediated by the IP(3)-signaling pathway. We suggest that enhancing astrocyte mitochondrial metabolism offers a promising neuroprotective strategy for a broad range of brain injuries.


Assuntos
Astrócitos/metabolismo , Edema Encefálico/metabolismo , Edema Encefálico/prevenção & controle , Lesões Encefálicas/metabolismo , Gliose/metabolismo , Gliose/prevenção & controle , Receptores Purinérgicos P2Y1/metabolismo , Animais , Astrócitos/patologia , Edema Encefálico/etiologia , Lesões Encefálicas/complicações , Córtex Cerebral/lesões , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Gliose/etiologia , Hipocampo/lesões , Hipocampo/metabolismo , Hipocampo/patologia , Receptores de Inositol 1,4,5-Trifosfato/deficiência , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fármacos Neuroprotetores/metabolismo , Receptores Purinérgicos P2Y1/fisiologia , Transdução de Sinais/fisiologia
10.
PLoS One ; 6(4): e18711, 2011 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-21533252

RESUMO

BACKGROUND: The blood brain barrier (BBB) is impermeable to most drugs, impeding the establishment of novel neuroprotective therapies and strategies for many neurological diseases. Intranasal administration offers an alternative path for efficient drug delivery into the CNS. So far, the anatomical structures discussed to be involved in the transport of intranasally administered drugs into the CNS include the trigeminal nerve, olfactory nerve and the rostral migratory stream (RMS), but the relative contributions are debated. METHODS AND FINDINGS: In the present study we demonstrate that surgical transection, and the resulting structural disruption of the RMS, in mice effectively obstructs the uptake of intranasally administered radioligands into the CNS. Furthermore, using a fluorescent cell tracer, we demonstrate that intranasal administration in mice allows agents to be distributed throughout the entire brain, including olfactory bulb, hippocampus, cortex and cerebellum. CONCLUSIONS: This study provides evidence of the vital role the RMS has in the CNS delivery of intranasally administered agents. The identification of the RMS as the major access path for intranasally administered drugs into the CNS may contribute to the development of treatments that are tailored for efficient transport within this structure. Research into the RMS needs to continue to elucidate its limitations, capabilities, mechanisms of transport and potential hazards before we are able to advance this technique into human research.


Assuntos
Sistema Nervoso Central/metabolismo , Administração Intranasal , Animais , Barreira Hematoencefálica , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Neurol Res ; 32(5): 523-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20092679

RESUMO

INTRODUCTION: The neuroprotective nature of exercise has been well established and the mechanisms of this protection are still a subject of much research. This study aims to determine if cerebral blood flow is constituently higher during the ischemia or reperfusion events in stroke. MATERIALS AND METHODS: Adult male Sprague-Dawley rats were randomly assigned into exercise or non-exercise (control) groups. Exercised rats underwent 30 minutes of running on a treadmill for 3 weeks. A 2 hour unilateral middle cerebral artery occlusion using an intraluminal filament was performed to induce ischemic stroke, followed by a 24 hour reperfusion. A sham control without exercise and middle cerebral artery occlusion was used. Laser Doppler flowmetry (LDF) and (15)O-H(2)O positron emission tomography (PET) were used to determine cerebral blood flow, respectively. (18)F-fluorodeoxy-D-glucose was used to determine cerebral metabolism in some animals. Histological analysis determined infarct volume in the same animal after blood flow examination. RESULTS: LDF and PET both indicated that middle cerebral artery occlusion significantly (p<0.05) reduced cerebral blood flow during ischemia and reperfusion in association with reduced cerebral metabolism after stroke. However, pre-ischemic exercise significantly (p<0.05) improved cerebral blood flow during reperfusion, although cerebral blood flow remained at a similar level to that of the non-exercise stroke group during the middle cerebral artery occlusion. This improved cerebral blood flow during reperfusion was associated with decreased brain infarct volume. CONCLUSIONS: This study revealed that pre-ischemic exercise in rats improved cerebral blood flow during reperfusion, suggesting that exercise provides neuroprotection by partially ameliorating the 'no reflow' phenomenon in stroke.


Assuntos
Circulação Cerebrovascular/fisiologia , Infarto da Artéria Cerebral Média/fisiopatologia , Condicionamento Físico Animal/fisiologia , Acidente Vascular Cerebral/fisiopatologia , Envelhecimento , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Glucose/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/fisiologia , Acidente Vascular Cerebral/patologia , Fatores de Tempo
12.
Neurol Res ; 32(7): 756-62, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19682410

RESUMO

OBJECTIVE: We sought to determine whether cerebral inflammation in ischemic rats was reduced by a neuroprotective action of pre-ischemic tumor necrosis factor-alpha up-regulation, which down-regulated matrix metalloproteinase-9 activity via extracellular signal-regulated kinase 1/2 phosphorylation. MATERIAL AND METHODS: Adult male Sprague-Dawley rats were subjected to 30 minutes of exercise on a treadmill for 3 weeks. Stroke was induced by a 2 hour middle cerebral artery occlusion using an intraluminal filament. The exercised animals were treated with tumor necrosis factor-alpha antibody, UO126 (extracellular signal-regulated kinase 1/2 inhibitor), or both UO126 and doxycycline (matrix metalloproteinase-9 inhibitor). Brain infarct volume was assessed using Nissl staining. Leukocyte infiltration was evaluated using myeloperoxidase immunostaining. Intercellular adhesion molecule-1 and matrix metalloproteinase protein levels were determined by Western blot, and enzyme activity was evaluated using zymography. RESULTS: There was a significant decrease in neurological deficits, brain infarct volume and leukocyte infiltration, in association with reduction in matrix metalloproteinase-9 and intercellular adhesion molecule-1 expression in exercised animals. Exercised animals treated with either tumor necrosis factor-alpha antibody or with UO126 showed a reversal of neurological outcome, infarct volume and leukocyte infiltration. Matrix metalloproteinase-9 activity was reversed, at least partially, but the intercellular adhesion molecule-1 expression was not. Neuroprotection remained when the exercised ischemic rats were treated with both UO126 and doxycycline. CONCLUSION: These results suggest that exercise-induced up-regulation of tumor necrosis factor-alpha before stroke and extracellular signal-regulated kinase 1/2 phosphorylation play a role in decreasing brain inflammation by regulating matrix metalloproteinase-9 activity.


Assuntos
Encéfalo/irrigação sanguínea , Encefalite/terapia , Precondicionamento Isquêmico/métodos , Metaloproteinase 9 da Matriz/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Condicionamento Físico Animal/fisiologia , Acidente Vascular Cerebral/terapia , Fator de Necrose Tumoral alfa/metabolismo , Análise de Variância , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Encefalite/metabolismo , Encefalite/patologia , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Regulação para Cima
13.
J Neurosurg ; 111(1): 164-70, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19284235

RESUMO

OBJECT: Individually, the cytokines erythropoietin (EPO) and insulin-like growth factor-I (IGF-I) have both been shown to reduce neuronal damage significantly in rodent models of cerebral ischemia. The authors have previously shown that EPO and IGF-I, when administered together, provide acute and prolonged neuroprotection in cerebrocortical cultures against N-methyl-D-aspartate-induced apoptosis. The aim of this study was to determine whether intranasally applied EPO plus IGF-I can provide acute neuroprotection in an animal stroke model and to show that intranasal administration is more efficient at delivering EPO plus IGF-I to the brain when compared with intravenous, subcutaneous, or intraperitoneal administration. METHODS: The EPO and IGF-I were administered intranasally to mice that underwent transient middle cerebral artery occlusion (MCAO). Stroke volumes were measured after 1 hour of MCAO and 24 hours of reperfusion. To evaluate the long-term effects of this treatment, behavioral outcomes were assessed at 3, 30, 60, and 90 days following MCAO. Radiography and liquid scintillation were used to visualize and quantify the uptake of radiolabeled 125I-EPO and 125I-IGF-I into the mouse brain after intranasal, intravenous, subcutaneous, or intraperitoneal administration. RESULTS: Intranasal administration of EPO plus IGF-I reduced stroke volumes within 24 hours and improved neurological function in mice up to 90 days after MCAO. The 125I-EPO and 125I-IGF-I were found in the brain within 20 minutes after intranasal administration and accumulated within the injured areas of the brain. In addition, intranasal administration delivered significantly higher levels of the applied 125I-EPO and 125I-IGF-I to the brain compared with intravenous, subcutaneous, or intraperitoneal administration. CONCLUSIONS: The data demonstrate that intranasal EPO plus IGF-I penetrates into the brain more efficiently than other drug delivery methods and could potentially provide a fast and efficient treatment to prevent chronic effects of stroke.


Assuntos
Sistemas de Liberação de Medicamentos , Eritropoetina/farmacocinética , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fator de Crescimento Insulin-Like I/farmacocinética , Fármacos Neuroprotetores/farmacocinética , Doença Aguda , Administração Intranasal , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Infarto da Artéria Cerebral Média/patologia , Radioisótopos do Iodo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
J Neurosurg ; 110(6): 1218-26, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19199470

RESUMO

OBJECT: A relationship has been found between peripheral thermal injury and cerebral complications leading to injury and death. In the present study, the authors examined whether tumor necrosis factor-alpha (TNF-alpha) and matrix metalloproteinase-9 (MMP-9) play a causative role in blood-brain barrier (BBB) disruption after peripheral thermal injury. METHODS: Thirty-two male Sprague-Dawley rats were subjected to thermal injury. One hour later, 8 rats were injected with TNF-alpha neutralizing antibody, and 8 were injected with doxycycline, an inhibitor of the MMP family proteins; 16 rats did not receive any treatment. Brain tissue samples obtained 7 hours after injury in the treated animals were examined for BBB function by using fluorescein isothiocyanate-dextran and by assessing parenchymal water content. Protein expression of basement membrane components (collagen IV, laminin, and fibronectin) was quantified on Western blot analysis, and MMP-9 protein expression and enzyme activity were determined using Western blot and gelatin zymography. Thermally injured rats that did not receive treatment were killed at 3, 7, or 24 hours after injury and tested for BBB functioning at each time point. Histological analysis for basement membrane proteins was also conducted in untreated rats killed at 7 hours after injury. Results of testing in injured rats were compared with those obtained in a control group of rats that did not undergo thermal injury. RESULTS: At 7 hours after thermal injury, a significant increase in the fluorescein isothiocyanate-dextran and water content of the brain was found (p < 0.05), but BBB dysfunction was significantly decreased in the rats that received TNF-alpha antibody or doxycycline (p < 0.05). In addition, the components of the basal lamina were significantly decreased at 7 hours after thermal injury (p < 0.01), and there were significant increases in MMP-9 protein expression and enzyme activity (p < 0.05). The basal lamina damage was reversed by inhibition of TNF-alpha and MMP-9, and the increase in MMP-9 protein was reduced in the presence of doxycycline (p < 0.05). The authors found that MMP-9 enzyme activity was significantly increased after thermal injury (p < 0.01) but decreased in the presence of either TNF-alpha antibody or doxycycline (p < 0.01). CONCLUSIONS: The dual, inhibitory activity of both TNF-alpha and MMP-9 in brain injury suggests that a TNF-alpha and MMP-9 cascade may play a key role in BBB disruption. These results offer a better understanding of the pathophysiology of burn injuries, which may open new avenues for burn treatment beyond the level of current therapies.


Assuntos
Barreira Hematoencefálica/fisiologia , Edema Encefálico/etiologia , Queimaduras/complicações , Queimaduras/metabolismo , Metaloproteinase 9 da Matriz/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Edema Encefálico/diagnóstico , Edema Encefálico/metabolismo , Queimaduras/patologia , Proteínas da Matriz Extracelular/metabolismo , Masculino , Inibidores de Metaloproteinases de Matriz , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Fator de Necrose Tumoral alfa/antagonistas & inibidores
15.
Neurosci Lett ; 444(3): 222-6, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18718505

RESUMO

Emerging data suggests the serine proteases, tissue plasminogen activator (tPA), and urokinase plasminogen activator (uPA), may play a detrimental role in traumatic states leading to compromise of the blood brain barrier (BBB). The purpose of our study was to define the role of endogenous tPA and uPA on the BBB following peripheral burn injuries. Adult male Sprague-Dawley rats (n=46) were studied in control and thermal injury groups. Rats were anesthetized and submerged in 100 degrees C water for 6s producing a third degree burn affecting 60-70% of the total body surface area. BBB dysfunction was then evaluated by measuring the amount of Evans blue and by calculating the water content in the brain. Levels of tPA and uPA mRNA in the brain were determined with real-time polymerase chain reaction (PCR) at 3 and 7h post-injury. Results showed an increase in the brain water content and the presence of Evans blue in the brain tissue of thermally injured rats, temporally associated with an increased expression of endogenous tPA and uPA. Our study demonstrates that peripheral thermal injury does induce an increase in the permeability of the BBB. A possible mechanism may be an increased expression of tPA and uPA.


Assuntos
Barreira Hematoencefálica/fisiologia , Queimaduras/metabolismo , Ativador de Plasminogênio Tecidual/biossíntese , Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Animais , Edema Encefálico/metabolismo , Permeabilidade Capilar , Masculino , Ratos , Ratos Sprague-Dawley
16.
J Cereb Blood Flow Metab ; 28(8): 1422-30, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18414498

RESUMO

This study explores the neuroprotective action of tumor necrosis factor-alpha (TNF-alpha) induced during physical exercise, which, consequently, reduces matrix metalloproteinase-9 (MMP-9) activity and ameliorates blood-brain barrier (BBB) dysfunction in association with extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation. Adult male Sprague-Dawley rats were subjected to exercise on a treadmill for 3 weeks. A 2-h middle cerebral artery occlusion and reperfusion was administered to exercised and nonexercised animals to induce stroke. Exercised ischemic rats were subjected to TNF-alpha inhibition and ERK1/2 by TNF-alpha antibody or UO126. Nissl staining of coronal sections revealed the infarct volume. Evans blue extravasation and water content evaluated BBB function. Western blot was performed to analyze protein expression of TNF-alpha, ERK1/2, phosphorylated ERK1/2, the basal laminar protein collagen IV, and MMP-9. The activity of MMP-9 was determined by gelatin zymography. Tumor necrosis factor-alpha expression and ERK1/2 phosphorylation were upregulated during exercise. Infarct volume, brain edema, and Evans blue extravasation all significantly decreased in exercised ischemic rats. Collagen IV production increased in exercised rats and remained high after stroke, whereas MMP-9 protein level and activity decreased. These results were negated and returned toward nonexercised values once TNF-alpha or ERK1/2 was blocked. We concluded that preischemic, exercise-induced TNF-alpha markedly decreases BBB dysfunction by using the ERK1/2 pathway.


Assuntos
Barreira Hematoencefálica/metabolismo , Corpo Caloso/metabolismo , Condicionamento Físico Animal , Acidente Vascular Cerebral/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Animais , Anticorpos/farmacologia , Barreira Hematoencefálica/patologia , Butadienos/farmacologia , Colágeno Tipo IV/biossíntese , Corpo Caloso/patologia , Inibidores Enzimáticos/farmacologia , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos
17.
Acta Neuropathol ; 115(3): 289-96, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18210137

RESUMO

Previous treadmill exercise studies showing neuroprotective effects have raised questions as to whether exercise or the stress related to it may be key etiologic factors. In this study, we examined different exercise regimens (forced and voluntary exercise) and compared them with the effect of stress-only on stroke protection. Adult male Sprague-Dawley rats (n = 65) were randomly assigned to treatment groups for 3 weeks. These groups included control, treadmill exercise, voluntary running wheel exercise, restraint, and electric shock. Levels of the stress hormone, corticosterone, were measured in the different groups using ELISA. Animals from each group were then subjected to stroke induced by a 2-h middle cerebral artery (MCA) occlusion followed by 48-h reperfusion. Infarct volume was determined in each group, while changes in gene expression of stress-induced heat shock proteins (Hsp) 27 and 70 were compared using real-time PCR between voluntary and treadmill exercise groups. The level of corticosterone was significantly higher in both stress (P < 0.05) and treadmill exercise (P < 0.05) groups, but not in the voluntary exercise group. Infarct volume was significantly reduced (P < 0.01) following stroke in rats exercised on a treadmill. However, the amelioration of damage was not duplicated in voluntary exercise, even though running distance in the voluntary exercise group was significantly (P < 0.01) longer than that of the forced exercise group (4,828 vs. 900 m). Furthermore, rats in the electric shock group displayed a significantly increased (P < 0.01) infarct volume. Expression of both Hsp 27 and Hsp 70 mRNA was significantly increased (P < 0.01) in the treadmill exercise group as compared with that in the voluntary exercise group. These results suggest that exercise with a stressful component, rather than either voluntary exercise or stress alone, is better able to reduce infarct volume. This exercise-induced neuroprotection may be attributable to up-regulation of stress-induced heat shock proteins 27 and 70.


Assuntos
Expressão Gênica/fisiologia , Condicionamento Físico Animal/fisiologia , Esforço Físico/fisiologia , Estresse Psicológico/fisiopatologia , Acidente Vascular Cerebral/patologia , Animais , Corticosterona/sangue , Estimulação Elétrica , Ensaio de Imunoadsorção Enzimática , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico/biossíntese , Masculino , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Shock ; 27(6): 652-6, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17505305

RESUMO

Outcomes after mild or moderate head trauma are worsened with associated hypotension, and secondary brain injury can be reduced with timely resuscitation. This study was performed to investigate HBOC-201 as a resuscitation therapy in a combined hemorrhagic shock and brain injury model. Anesthetized rats sustained moderate brain injury using a controlled cortical impact device, followed by rapid hemorrhage to a mean arterial pressure of 30 mmHg. After 30 min of hypotension, animals were resuscitated with HBOC-201, autologous shed blood (SB), or lactated Ringer solution (LR). Brain injury was assessed by measurements of cerebral blood flow (CBF) and cerebral vasoreactivity to hypercapnia (CVH) using a laser Doppler flowmeter. Contusion volume was evaluated histologically, and cerebral edema was determined by total water content. The HBOC rats required significantly less resuscitation volume versus LR and SB. The CBF was significantly diminished at 60 min after resuscitation with HBOC (70.1% +/- 3.8% baseline) compared with LR (105.8% +/- 10.1% baseline; P < 0.01) and SB (96.8% +/- 5% baseline; P < 0.05). The CVH was preserved in the HBOC and SB groups. The CVH was significantly diminished compared with baseline in the LR group at 30 min after resuscitation and showed a significant loss compared with HBOC at 60 min after resuscitation. The contusion volume for HBOC (45.1 mm3) and SB (35.1 mm3) was less than LR (63.5 mm3, P < 0.01). Although CBF was diminished after resuscitation in the HBOC group, HBOC-treated animals maintained CVH and experienced significantly smaller contusion volume than those treated with LR. These results suggest that resuscitation with HBOC-201 protects autoregulatory mechanisms and may reduce secondary brain injury in traumatic brain injury.


Assuntos
Substitutos Sanguíneos/uso terapêutico , Lesões Encefálicas/tratamento farmacológico , Hemoglobinas/uso terapêutico , Ressuscitação/métodos , Choque Hemorrágico/terapia , Animais , Circulação Cerebrovascular , Hemoglobinas/metabolismo , Hemorragia/prevenção & controle , Masculino , Oxigênio/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Resultado do Tratamento
19.
Neurol Res ; 29(6): 610-4, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17535571

RESUMO

High mortality incidence after serious systemic thermal injury is believed to be linked to significant increases in cerebral permeability, ultimately leading to irreversible blood-brain barrier (BBB) breakdown. The aim of this study was to investigate whether disruption of microvascular integrity in a rat thermal injury model is associated with early matrix metalloproteinase (MMP) expression. A total of 35 Sprague-Dawley rats were studied in thermal injury and control groups, each group containing two subgroups, one for brain edema and Evans blue analysis and another for MMP mRNA analysis. Thermally injured animals were anesthetized and submerged vertically in 85 degrees C water to the neck for 6 seconds producing a third degree burn affecting 70% of the total body surface area. BBB integrity was determined by measuring amount of Evans blue after 7 hours of injury with a spectrophotometer. Brain edema was detected by calculating water content. Brain mRNA levels were determined with real-time PCR 3 and 7 hours post-injury. Brain water content was significantly increased after peripheral injury at hour 7. Evans blue leakage was also significantly increased at the same time, suggesting an impaired BBB function after injury. Expressions of MMP-2 and MMP-9 mRNA in brain were increased as early as 3 hours after injury and remained at hour 7. Our study demonstrated a significant increase in cerebral permeability that occurs after serious systemic thermal injury. The underlying mechanisms could be related to early expression of MMPs.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Queimaduras/patologia , Queimaduras/fisiopatologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Metaloproteinases da Matriz/metabolismo , Análise de Variância , Animais , Edema Encefálico/etiologia , Modelos Animais de Doenças , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz/classificação , Metaloproteinases da Matriz/genética , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos
20.
Brain Res ; 1129(1): 26-33, 2007 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-17156757

RESUMO

Mortality after serious systemic thermal injury may be linked to significant increases in cerebral vascular permeability and edema due to blood-brain barrier (BBB) breakdown. This BBB disruption is thought to be mediated by a family of proteolytic enzymes known as matrix metalloproteinases (MMPs). The gelatinases, MMP-2 and MMP-9, digest the endothelial basal lamina of the BBB, which is essential for maintaining BBB integrity. The current study investigated whether disruption of microvascular integrity in a rat thermal injury model is associated with gelatinase expression and activity. Seventy-two adult Sprague-Dawley rats were anesthetized and submerged horizontally, in the supine position, in 100 degrees C (37 degrees C for controls) water for 6 s producing a third-degree burn affecting 60-70% of the total body surface area. Brain edema was detected by calculating water content. Real time PCR, Western blot, and zymography were used to quantify MMP mRNA, protein, and enzyme activity levels. Each group was quantified at 3, 7, 24, and 72 h post thermal injury. Brain water content was significantly increased 7 through 72 h after burn. Expression of brain MMP-9 mRNA was significantly increased as early as 3 h after thermal injury compared to controls, remained at 7 h (p<0.01), and returned to control levels by 24 h. MMP-9 protein levels and enzyme activity began to increase at 7 h and reached significant levels between 7 and 24 h after thermal injury. While MMP-9 protein levels continued to increase significantly through 72 h, enzyme activity returned to control level. The increase in MMP-9 expression and activity, associated with increased BBB permeability following thermal injury, indicates that MMP-9 may contribute to observed cerebral edema in peripheral thermal injury.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Edema Encefálico/etiologia , Edema Encefálico/fisiopatologia , Queimaduras/complicações , Queimaduras/fisiopatologia , Metaloproteinases da Matriz/metabolismo , Animais , Membrana Basal/enzimologia , Membrana Basal/ultraestrutura , Barreira Hematoencefálica/enzimologia , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Edema Encefálico/enzimologia , Modelos Animais de Doenças , Células Endoteliais/enzimologia , Células Endoteliais/ultraestrutura , Ativação Enzimática/fisiologia , Matriz Extracelular/metabolismo , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...