Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Cell Neurosci ; 18: 1325630, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38638304

RESUMEN

Ischemic stroke is the leading cause of serious long-term disability and the 5th leading cause of death in the United States. Revascularization of the occluded cerebral artery, either by thrombolysis or endovascular thrombectomy, is the only effective, clinically-approved stroke therapy. Several potentially neuroprotective agents, including glutamate antagonists, anti-inflammatory compounds and free radical scavenging agents were shown to be effective neuroprotectants in preclinical animal models of brain ischemia. However, these compounds did not demonstrate efficacy in clinical trials with human patients following stroke. Proposed reasons for the translational failure include an insufficient understanding on the cellular and molecular pathophysiology of ischemic stroke, lack of alignment between preclinical and clinical studies and inappropriate design of clinical trials based on the preclinical findings. Therefore, novel neuroprotective treatments must be developed based on a clearer understanding of the complex spatiotemporal mechanisms of ischemic stroke and with proper clinical trial design based on the preclinical findings from specific animal models of stroke. We and others have demonstrated the clinical potential for neuregulin-1 (NRG-1) in preclinical stroke studies. NRG-1 significantly reduced ischemia-induced neuronal death, neuroinflammation and oxidative stress in rodent stroke models with a therapeutic window of >13 h. Clinically, NRG-1 was shown to be safe in human patients and improved cardiac function in multisite phase II studies for heart failure. This review summarizes previous stroke clinical candidates and provides evidence that NRG-1 represents a novel, safe, neuroprotective strategy that has potential therapeutic value in treating individuals after acute ischemic stroke.

2.
Mol Neurobiol ; 59(12): 7236-7252, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36151369

RESUMEN

Stroke is ranked as the fifth leading cause of death and the leading cause of adult disability in the USA. The progression of neuronal damage after stroke is recognized to be a complex integration of glia, neurons, and the surrounding extracellular matrix, therefore potential treatments must target the detrimental effects created by these interactions. In this study, we examined the spatial cellular and neuroinflammatory mechanisms occurring early after ischemic stroke utilizing Nanostring Digital Spatial Profiling (DSP) technology. Male C57bl/6 mice were subjected to photothrombotic middle cerebral artery occlusion (MCAO) and sacrificed at 3 days post-ischemia. Spatial distinction of the ipsilateral hemisphere was studied according to the regions of interest: the ischemic core, peri-infarct tissues, and peri-infarct normal tissue (PiNT) in comparison to the contralateral hemisphere. We demonstrated that the ipsilateral hemisphere initiates distinct spatial regulatory proteomic profiles with DSP technology that can be identified consistently with the immunohistochemical markers, FJB, GFAP, and Iba-1. The core border profile demonstrated an induction of neuronal death, apoptosis, autophagy, immunoreactivity, and early degenerative proteins. Most notably, the core border resulted in a decrease of the neuronal proteins Map2 and NeuN; an increase in the autophagy proteins BAG3 and CTSD; an increase in the microglial and peripheral immune invasion proteins Iba1, CD45, CD11b, and CD39; and an increase in the neurodegenerative proteins BACE1, APP, amyloid ß 1-42, ApoE, and hyperphosphorylated tau protein S-199. The peri-infarct region demonstrated increased astrocytic, immunoreactivity, apoptotic, and neurodegenerative proteomic profiles, with an increase in BAG3, GFAP, and hyperphosphorylated tau protein S-199. The PiNT region displayed minimal changes compared to the contralateral cortex with only an increase in GFAP. In this study, we showed that mechanisms known to be associated with stroke, such as apoptosis and inflammation, occur in distinct spatial domains of the injured brain following ischemia. We also demonstrated the dysregulation of specific autophagic pathways that may lead to neurodegeneration in peri-infarct brain tissues. Taken together, these data suggest that identifying post-ischemic mechanisms occurring in a spatiotemporal manner may lead to more precise targets for successful therapeutic interventions to treat stroke.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Ratones , Masculino , Proteínas tau/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteómica , Ácido Aspártico Endopeptidasas/metabolismo , Neuronas/metabolismo , Accidente Cerebrovascular/metabolismo , Isquemia Encefálica/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Ratones Endogámicos C57BL , Análisis Espacial , Modelos Animales de Enfermedad
3.
Transl Psychiatry ; 12(1): 83, 2022 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-35220393

RESUMEN

Childhood obesity leads to hippocampal atrophy and altered cognition. However, the molecular mechanisms underlying these impairments are poorly understood. The neurotrophic factor neuregulin-1 (NRG1) and its cognate ErbB4 receptor play critical roles in hippocampal maturation and function. This study aimed to determine whether exogenous NRG1 administration reduces hippocampal abnormalities and neuroinflammation in rats exposed to an obesogenic Western-like diet (WD). Lewis rats were randomly divided into four groups (12 rats/group): (1) control diet+vehicle (CDV); (2) CD + NRG1 (CDN) (daily intraperitoneal injections: 5 µg/kg/day; between postnatal day, PND 21-PND 41); (3) WD + VEH (WDV); (4) WD + NRG1 (WDN). Neurobehavioral assessments were performed at PND 43-49. Brains were harvested for MRI and molecular analyses at PND 49. We found that NRG1 administration reduced hippocampal volume (7%) and attenuated hippocampal-dependent cued fear conditioning in CD rats (56%). NRG1 administration reduced PSD-95 protein expression (30%) and selectively reduced hippocampal cytokine levels (IL-33, GM-CSF, CCL-2, IFN-γ) while significantly impacting microglia morphology (increased span ratio and reduced circularity). WD rats exhibited reduced right hippocampal volume (7%), altered microglia morphology (reduced density and increased lacunarity), and increased levels of cytokines implicated in neuroinflammation (IL-1α, TNF-α, IL-6). Notably, NRG1 synergized with the WD to increase hippocampal ErbB4 phosphorylation and the tumor necrosis alpha converting enzyme (TACE/ADAM17) protein levels. Although the results did not provide sufficient evidence to conclude that exogenous NRG1 administration is beneficial to alleviate obesity-related outcomes in adolescent rats, we identified a potential novel interaction between obesogenic diet exposure and TACE/ADAM17-NRG1-ErbB4 signaling during hippocampal maturation. Our results indicate that supraoptimal ErbB4 activities may contribute to the abnormal hippocampal structure and cognitive vulnerabilities observed in obese individuals.


Asunto(s)
Neurregulina-1 , Obesidad Infantil , Animales , Ansiedad , Dieta , Neurregulina-1/metabolismo , Neurregulina-1/farmacología , Enfermedades Neuroinflamatorias , Ratas , Ratas Endogámicas Lew
4.
Neurobiol Dis ; 161: 105545, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34742879

RESUMEN

Temporal lobe epilepsy (TLE) is the most common form of focal epilepsy. Dysregulation of glutamate transporters has been a common finding across animal models of epilepsy and in patients with TLE. In this study, we investigate NRG-1/ErbB4 signaling in epileptogenesis and the neuroprotective effects of NRG-1 treatment in a mouse model of temporal lobe epilepsy. Using immunohistochemistry, we report the first evidence for NRG-1/ErbB4-dependent selective upregulation of glutamate transporter EAAC1 and bihemispheric neuroprotection by exogeneous NRG-1 in the intrahippocampal kainic acid (IHKA) model of TLE. Our findings provide evidence that dysregulation of glutamate transporter EAAC1 contributes to the development of epilepsy and can be therapeutically targeted to reduce neuronal death following IHKA-induced status epilepticus (SE).


Asunto(s)
Epilepsia del Lóbulo Temporal , Epilepsia , Neurregulina-1 , Neuroprotección , Receptor ErbB-4 , Animales , Modelos Animales de Enfermedad , Epilepsia/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/inducido químicamente , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Transportador 3 de Aminoácidos Excitadores/metabolismo , Hipocampo , Humanos , Ratones , Neurregulina-1/metabolismo , Neurregulina-1/farmacología , Receptor ErbB-4/metabolismo
6.
ACS Cent Sci ; 5(8): 1387-1395, 2019 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-31482121

RESUMEN

Proteinaceous aggregation is a well-known observable in Alzheimer's disease (AD), but failure and storage of lysosomal bodies within neurons is equally ubiquitous and actually precedes bulk accumulation of extracellular amyloid plaque. In fact, AD shares many similarities with certain lysosomal storage disorders though establishing a biochemical connection has proven difficult. Herein, we demonstrate that isomerization and epimerization, which are spontaneous chemical modifications that occur in long-lived proteins, prevent digestion by the proteases in the lysosome (namely, the cathepsins). For example, isomerization of aspartic acid into l-isoAsp prevents digestion of the N-terminal portion of Aß by cathepsin L, one of the most aggressive lysosomal proteases. Similar results were obtained after examination of various target peptides with a full series of cathepsins, including endo-, amino-, and carboxy-peptidases. In all cases peptide fragments too long for transporter recognition or release from the lysosome persisted after treatment, providing a mechanism for eventual lysosomal storage and bridging the gap between AD and lysosomal storage disorders. Additional experiments with microglial cells confirmed that isomerization disrupts proteolysis in active lysosomes. These results are easily rationalized in terms of protease active sites, which are engineered to precisely orient the peptide backbone and cannot accommodate the backbone shift caused by isoaspartic acid or side chain dislocation resulting from epimerization. Although Aß is known to be isomerized and epimerized in plaques present in AD brains, we further establish that the rates of modification for aspartic acid in positions 1 and 7 are fast and could accrue prior to plaque formation. Spontaneous chemistry can therefore provide modified substrates capable of inducing gradual lysosomal failure, which may play an important role in the cascade of events leading to the disrupted proteostasis, amyloid formation, and tauopathies associated with AD.

7.
J Mol Neurosci ; 69(2): 333-342, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31290093

RESUMEN

Identifying novel neuroprotectants that can halt or reverse the neurological effects of stroke is of interest to both clinicians and scientists. We and others previously showed the pre-clinical neuroprotective efficacy of neuregulin-1 (NRG-1) in rats following focal brain ischemia. In this study, we examined neuroprotection by exogenous and endogenous NRG-1 using a mouse model of ischemic stroke. C57BL6 mice were subjected to middle cerebral artery occlusion (MCAO) followed by reperfusion. NRG-1 or vehicle was infused intra-arterially (i.a.) or intravenously (i.v.) after MCAO and before the onset of reperfusion. NRG-1 treatment (16 µg/kg; i.a.) reduced cerebral cortical infarct volume by 72% in mice when delivered post-ischemia. NRG-1 also inhibited neuronal injury as measured by Fluoro Jade B labeling and rescued NeuN immunoreactivity in neurons. Neuroprotection by NRG-1 was also observed in mice when administered i.v. (100 µg/kg) in both male and female mice. We investigated whether endogenous NRG-1 was neuroprotective using male and female heterozygous NRG-1 knockout mice (NRG-1+/-) compared with wild-type mice (WT) littermates. NRG-1+/- and WT mice were subjected to MCAO for 45 min, and infarct size was measured 24 h following MCAO. NRG-1+/- mice displayed a sixfold increase in cortical infarct size compared with WT mice. These results demonstrate that NRG-1 treatment mitigates neuronal damage following cerebral ischemia. We further showed that reduced endogenous NRG-1 results in exacerbated neuronal injury in vivo. These findings suggest that NRG-1 represents a promising therapy to treat stroke in human patients.


Asunto(s)
Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Neurregulina-1/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Femenino , Heterocigoto , Infarto de la Arteria Cerebral Media/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neurregulina-1/genética
8.
Comp Med ; 68(6): 496-502, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30486918

RESUMEN

An adult rhesus macaque developed seizures after the induction of ischemic stroke. Initially, on the day of surgery, a focal ischemic lesion was present exclusively in the right caudate nucleus. By 48 h after stroke induction, the lesion had extended into the putamen, when a seizure was observed. Our report highlights the temporal changes in infarction of unilateral basal ganglia after acute stroke and the accompanying clinical symptoms. This unusual case may provide additional information regarding the involvement of the basal ganglia in seizures, given that prior case reports and studies usually have not described the temporal and spatial evolution of the lesion before clinical symptoms emerge.


Asunto(s)
Ganglios Basales/diagnóstico por imagen , Macaca mulatta , Enfermedades de los Monos/diagnóstico por imagen , Convulsiones/veterinaria , Accidente Cerebrovascular/veterinaria , Animales , Imagen por Resonancia Magnética , Convulsiones/diagnóstico por imagen , Accidente Cerebrovascular/diagnóstico por imagen
9.
Brain Res ; 1698: 161-169, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30099039

RESUMEN

The use of blood biomarkers for stroke has been long considered an excellent method to determine the occurrence, timing, subtype, and severity of stroke. In this study, venous blood was obtained from ischemic stroke patients after stroke onset and compared with age and sex-matched controls. We used a multiplex panel of 37 inflammatory molecules, analyzed using Luminex MagPix technology, to identify the changes in plasma proteins after ischemic stroke. We identified eight key molecules that were altered within the blood of stroke patients as compared to controls. Plasma levels of interleukin 6 signal transducer (sIL-6Rß/gp130), matrix metalloproteinase-2 (MMP-2), osteopontin, sTNF-R1 and sTNF-R2 were significantly higher in stroke patients compared to controls. Interferon-ß, interleukin-28, and thymic stromal lymphopoietin (TSLP) were decreased in plasma from stroke patients. No other immunological markers were significantly different between patient groups. When stroke patients were treated with tissue plasminogen activator (t-PA), plasma levels of interferon-α2 significantly increased while interleukin-2 and pentraxin-3 decreased. The discriminatory power of the molecules was evaluated by receiver operating characteristic (ROC) analysis. According to ROC analysis, the best markers for distinguishing stroke occurrence were MMP-2 (AUC = 0.76, sensitivity 62.5%, specificity 88.5%), sTNF-R2 (AUC = 0.75, sensitivity 83.3%, specificity 65.3%) and TSLP (AUC = 0.81, sensitivity 66.7%, specificity 96.2%). Multivariate logistic regression, used to evaluate the combination of proteins, identified a biomarker panel with high specificity and sensitivity (AUC = 0.96, sensitivity 87.5%, specificity 96.2%). These results indicate a novel set of blood biomarkers that could be used in a panel to identify stroke patients and their responsiveness to therapeutic intervention.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Accidente Cerebrovascular/sangre , Anciano , Biomarcadores/sangre , Biomarcadores Farmacológicos/sangre , Isquemia Encefálica/sangre , Receptor gp130 de Citocinas/sangre , Femenino , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/sangre , Persona de Mediana Edad , Osteopontina/sangre , Curva ROC , Factores de Riesgo , Accidente Cerebrovascular/tratamiento farmacológico , Factor 1 Asociado a Receptor de TNF/sangre , Factor 2 Asociado a Receptor de TNF/sangre , Activador de Tejido Plasminógeno/uso terapéutico
10.
Magn Reson Imaging ; 53: 63-70, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30021123

RESUMEN

Stroke is a devastating neurovascular disorder that results in damage to neurons and white matter tracts. It has been previously demonstrated that neuregulin-1 (NRG-1) protects neurons from ischemic injury following stroke. Here, diffusion tensor imaging (DTI) was utilized to characterize the effects of NRG-1 treatment on cererbral infarction and integrity of white matter after ischemic insult using a permanent middle celebral artery occlusion (pMCAo) rat model. In the present study, sixteen Sprague-Dawley rats underwent pMCAo surgery and received either a single intra-arterial bolus (20 µg/kg) dose of NRG-1 or saline immediately prior to pMCAo. MRI including T2-weighted imaging and DTI was performed in the first 3 h post stroke, and repeated 48 h later. It is found that the stroke infarction was significantly reduced in the NRG-1 treated group. Also, NRG-1 prevented the reduction of fractional anisotropy (FA) in white matter tracts of fornix and corpus callosum (CC), indicating its protection of CC and fornix white matter bundles from ischemia insult. As a conclusion, the present DTI results demonstrate that NRG-1 has significantly neuroprotective effects in both cerebral cortex and white matter including corpus callosum and fornix during acute stroke. In particular, NRG-1 is more effective on stroke lesion with mild ischemia. As CC and fornix white matter bundles play critical roles in transcallosal connectivity and hippocampal projections respectively in the central nervous system, the findings could provide complementary information for better understanding the biological mechanism of NRG-1's neuroprotection in ischemic tissues and neurobehavioral effects.


Asunto(s)
Isquemia Encefálica/diagnóstico por imagen , Imagen de Difusión Tensora , Neurregulina-1/fisiología , Neuroprotección , Animales , Anisotropía , Corteza Cerebral/diagnóstico por imagen , Cuerpo Calloso/diagnóstico por imagen , Fórnix/diagnóstico por imagen , Isquemia , Imagen por Resonancia Magnética , Masculino , Neuronas/metabolismo , Fármacos Neuroprotectores , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular , Sustancia Blanca
11.
PLoS One ; 13(6): e0197092, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29856744

RESUMEN

Ischemic stroke is a major cause of mortality in the United States. We previously showed that neuregulin-1 (NRG1) was neuroprotective in rat models of ischemic stroke. We used gene expression profiling to understand the early cellular and molecular mechanisms of NRG1's effects after the induction of ischemia. Ischemic stroke was induced by middle cerebral artery occlusion (MCAO). Rats were allocated to 3 groups: (1) control, (2) MCAO and (3) MCAO + NRG1. Cortical brain tissues were collected three hours following MCAO and NRG1 treatment and subjected to microarray analysis. Data and statistical analyses were performed using R/Bioconductor platform alongside Genesis, Ingenuity Pathway Analysis and Enrichr software packages. There were 2693 genes differentially regulated following ischemia and NRG1 treatment. These genes were organized by expression patterns into clusters using a K-means clustering algorithm. We further analyzed genes in clusters where ischemia altered gene expression, which was reversed by NRG1 (clusters 4 and 10). NRG1, IRS1, OPA3, and POU6F1 were central linking (node) genes in cluster 4. Conserved Transcription Factor Binding Site Finder (CONFAC) identified ETS-1 as a potential transcriptional regulator of NRG1 suppressed genes following ischemia. A transcription factor activity array showed that ETS-1 activity was increased 2-fold, 3 hours following ischemia and this activity was attenuated by NRG1. These findings reveal key early transcriptional mechanisms associated with neuroprotection by NRG1 in the ischemic penumbra.


Asunto(s)
Isquemia Encefálica/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Neurregulina-1/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Elementos de Respuesta , Accidente Cerebrovascular/metabolismo , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Masculino , Neurregulina-1/genética , Proteína Proto-Oncogénica c-ets-1/genética , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/patología
12.
Endocrinology ; 158(10): 3647-3660, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28938399

RESUMEN

Mammalian ovarian follicular development and maturation of an oocyte competent to be fertilized and develop into an embryo depends on tightly regulated, spatiotemporally orchestrated crosstalk among cell death, survival, and differentiation signals through extra- and intraovarian signals, as well as on a permissive ovarian follicular microenvironment. Neuregulin-1 (NRG1) is a member of the epidermal growth factor-like factor family that mediates its effects by binding to a member of the erythroblastoma (ErbB) family. Our experimental results suggest gonadotropins promote differential expression of NRG1 and erbB receptors in granulosa cells (GCs), and NRG1 in theca cells during follicular development, and promote NRG1 secretions in the follicular fluid (FF) of rat ovaries. During the estrous cycle of rat, NRG1 and erbB receptors are differentially expressed in GCs and correlate positively with serum gonadotropins and steroid hormones. Moreover, in vitro experimental studies suggest that the protein kinase C inhibitor staurosporine (STS) causes the physical destruction of GCs by the activation of caspase-3. Exogenous NRG1 treatment of GCs delayed onset of STS-induced apoptosis and inhibited cleaved caspase-3 expressions. Moreover, exogenous NRG1 treatment of GCs alters STS-induced death by maintaining the expression of ErbB2, ErbB3, pAkt, Bcl2, and BclxL proteins. Taken together, these studies demonstrate that NRG1 is gonadotropin dependent, differentially regulated in GCs and theca cells, and secreted in ovarian FF as an intracellular survival factor that may govern follicular maturation.


Asunto(s)
Apoptosis/efectos de los fármacos , Supervivencia Celular , Receptores ErbB/efectos de los fármacos , Gonadotropinas/farmacología , Células de la Granulosa/efectos de los fármacos , Neurregulina-1/efectos de los fármacos , Animales , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Receptores ErbB/metabolismo , Femenino , Líquido Folicular , Células de la Granulosa/metabolismo , Técnicas In Vitro , Neurregulina-1/metabolismo , Neurregulina-1/farmacología , Folículo Ovárico/crecimiento & desarrollo , Ovario/citología , Ovario/efectos de los fármacos , Ovario/metabolismo , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor ErbB-2/efectos de los fármacos , Receptor ErbB-2/metabolismo , Receptor ErbB-3/efectos de los fármacos , Receptor ErbB-3/metabolismo , Estaurosporina/farmacología , Células Tecales , Proteína bcl-X/efectos de los fármacos , Proteína bcl-X/metabolismo
14.
J Neuroinflammation ; 13(1): 267, 2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27733171

RESUMEN

BACKGROUND: Acute intoxication with organophosphorus (OP) cholinesterase inhibitors can trigger convulsions that progress to life-threatening status epilepticus. Survivors face long-term morbidity including mild-to-severe decline in memory. It is posited that neuroinflammation plays a key role in the pathogenesis of OP-induced neuropsychiatric deficits. Rigorous testing of this hypothesis requires preclinical models that recapitulate relevant phenotypic outcomes. Here, we describe a rat model of acute intoxication with the OP diisopropylfluorophosphate (DFP) that exhibits persistent neuroinflammation and cognitive impairment. METHODS: Neuroinflammation, neurodegeneration, and cognitive function were compared in adult male Sprague Dawley rats injected with an acutely toxic dose of DFP vs. vehicle controls at multiple time points up to 36 days post-exposure. Neuroinflammation was quantified using immunohistochemical biomarkers of microglia (ionized calcium-binding adapter molecule 1, IBA1) and activated astrocytes (glial fibrillary acidic protein, GFAP) and positron emission tomography (PET) imaging of [11C]-(R)-PK11195, a ligand for the 18-kDa mitochondrial membrane translocator protein (TSPO). FluoroJade-B staining was used to assess neurodegeneration; Pavlovian conditioning, to assess cognitive function. RESULTS: Animals exhibited moderate-to-severe seizures within minutes of DFP injection that continued for up to 6 h post-injection. As indicated by IBA1 and GFAP immunoreactivity and by PET imaging of TSPO, acute DFP intoxication triggered neuroinflammation in the hippocampus and cortex during the first 3 days that peaked at 7 days and persisted to 21 days post-exposure in most animals. Neurodegeneration was detected in multiple brain regions from 1 to 14 days post-exposure. All DFP-intoxicated animals exhibited significant deficits in contextual fear conditioning at 9 and 20 days post-exposure compared to vehicle controls. Whole-brain TSPO labeling positively correlated with seizure severity score, but did not correlate with performance in the contextual fear-conditioning task. CONCLUSIONS: We describe a preclinical model in which acute DFP intoxication causes seizures, persistent neuroinflammation, neurodegeneration, and memory impairment. The extent of the neuroinflammatory response is influenced by seizure severity. However, the observation that a subset of animals with moderate seizures and minimal TSPO labeling exhibited cognitive deficits comparable to those of animals with severe seizures and significant TSPO labeling suggests that DFP may impair learning and memory circuitry via mechanisms independent of seizures or neuroinflammation.


Asunto(s)
Inhibidores de la Colinesterasa/toxicidad , Disfunción Cognitiva/inducido químicamente , Encefalitis/inducido químicamente , Isoflurofato/toxicidad , Animales , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Disfunción Cognitiva/diagnóstico por imagen , Condicionamiento Clásico/efectos de los fármacos , Encefalitis/diagnóstico por imagen , Conducta Exploratoria/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Imagen por Resonancia Magnética , Masculino , Proteínas de Microfilamentos/metabolismo , Tomografía de Emisión de Positrones , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Análisis de Regresión , Factores de Tiempo
15.
J Neuroinflammation ; 13(1): 237, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27596278

RESUMEN

BACKGROUND: We previously demonstrated that neuregulin-1 (NRG-1) was neuroprotective in rats following ischemic stroke. Neuroprotection by NRG-1 was associated with the suppression of pro-inflammatory gene expression in brain tissues. Over-activation of brain microglia can induce pro-inflammatory gene expression by activation of transcriptional regulators following stroke. Here, we examined how NRG-1 transcriptionally regulates inflammatory gene expression by computational bioinformatics and in vitro using microglial cells. METHODS: To identify transcriptional regulators involved in ischemia-induced inflammatory gene expression, rats were sacrificed 24 h after middle cerebral artery occlusion (MCAO) and NRG-1 treatment. Gene expression profiles of brain tissues following ischemia and NRG-1 treatment were examined by microarray technology. The Conserved Transcription Factor-Binding Site Finder (CONFAC) bioinformatics software package was used to predict transcription factors associated with inflammatory genes induced following stroke and suppressed by NRG-1 treatment. NF-kappa B (NF-kB) was identified as a potential transcriptional regulator of NRG-1-suppressed genes following ischemia. The involvement of specific NF-kB subunits in NRG-1-mediated inflammatory responses was examined using N9 microglial cells pre-treated with NRG-1 (100 ng/ml) followed by lipopolysaccharide (LPS; 10 µg/ml) stimulation. The effects of NRG-1 on cytokine production were investigated using Luminex technology. The levels of the p65, p52, and RelB subunits of NF-kB and IkB-α were determined by western blot analysis and ELISA. Phosphorylation of IkB-α was investigated by ELISA. RESULTS: CONFAC identified 12 statistically over-represented transcription factor-binding sites (TFBS) in our dataset, including NF-kBP65. Using N9 microglial cells, we observed that NRG-1 significantly inhibited LPS-induced TNFα and IL-6 release. LPS increased the phosphorylation and degradation of IkB-α which was blocked by NRG-1. NRG-1 also prevented the nuclear translocation of the NF-kB p65 subunit following LPS administration. However, NRG-1 increased production of the neuroprotective cytokine granulocyte colony-stimulating factor (G-CSF) and the nuclear translocation of the NF-kB p52 subunit, which is associated with the induction of anti-apoptotic and suppression of pro-inflammatory gene expression. CONCLUSIONS: Neuroprotective and anti-inflammatory effects of NRG-1 are associated with the differential regulation of NF-kB signaling pathways in microglia. Taken together, these findings suggest that NRG-1 may be a potential therapeutic treatment for treating stroke and other neuroinflammatory disorders.


Asunto(s)
Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Microglía/efectos de los fármacos , Neurregulina-1/uso terapéutico , Animales , Línea Celular Transformada , Biología Computacional , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Factor Estimulante de Colonias de Granulocitos/metabolismo , Proteínas I-kappa B/metabolismo , Lipopolisacáridos/farmacología , Masculino , Análisis por Micromatrices , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
16.
BMC Genomics ; 17: 130, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26912237

RESUMEN

BACKGROUND: Delayed or secondary cell death that is caused by a cascade of cellular and molecular processes initiated by traumatic brain injury (TBI) may be reduced or prevented if an effective neuroprotective strategy is employed. Microarray and subsequent bioinformatic analyses were used to determine which genes, pathways and networks were significantly altered 24 h after unilateral TBI in the rat. Ipsilateral hemi-brain, the corresponding contralateral hemi-brain, and naïve (control) brain tissue were used for microarray analysis. RESULTS: Ingenuity Pathway Analysis showed cell death and survival (CD) to be a top molecular and cellular function associated with TBI on both sides of the brain. One major finding was that the overall gene expression pattern suggested an increase in CD genes in ipsilateral brain tissue and suppression of CD genes contralateral to the injury which may indicate an endogenous protective mechanism. We created networks of genes of interest (GOI) and ranked the genes by the number of direct connections each had in the GOI networks, creating gene interaction hierarchies (GIHs). Cell cycle was determined from the resultant GIHs to be a significant molecular and cellular function in post-TBI CD gene response. CONCLUSIONS: Cell cycle and apoptosis signalling genes that were highly ranked in the GIHs and exhibited either the inverse ipsilateral/contralateral expression pattern or contralateral suppression were identified and included STAT3, CCND1, CCND2, and BAX. Additional exploration into the remote suppression of CD genes may provide insight into neuroprotective mechanisms that could be used to develop therapies to prevent cell death following TBI.


Asunto(s)
Lesiones Encefálicas/genética , Ciclo Celular/genética , Muerte Celular/genética , Epistasis Genética , Redes Reguladoras de Genes , Animales , Apoptosis , Encéfalo/fisiopatología , Ciclina D1/genética , Ciclina D2/genética , Masculino , Análisis por Micromatrices , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/genética , Proteína X Asociada a bcl-2/genética
17.
J Neurol Sci ; 357(1-2): 28-34, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26183085

RESUMEN

The neuroprotective effects of neuregulin-1 (NRG-1) on stroke lesions were assessed longitudinally in rats with middle cerebral artery occlusion (MCAo) using MRI. Sprague-Dawley rats (n=16, 250±20g) underwent permanent MCAo surgery with cerebral blood flow (CBF) monitored by laser doppler flowmetry at ipsilateral side of bregma for 20min post-occlusion. A single 50µl bolus dose of NRG-1 or vehicle was administered into the left internal carotid artery immediately prior to MCAo. The expansion of the ischemic lesion into the cortex was attenuated by NRG-1 over a 48-hour (h) time span as measured by diffusion weighted imaging (DWI). The final infarct volumes of NRG-1 treated rats were significantly smaller than those of the vehicle treated rats at 48h (264.8±192.1 vs. 533.4±175.5mm(3), p<0.05). The NRG-1 treated rats were further subdivided into 2 subgroups according to their CBF reduction during stroke surgery: mild ischemia (<70% CBF reduction) or severe ischemia (>70% CBF reduction). In particular, ischemic infarction was not usually observed in the cortex of NRG-1 treated rats with mild ischemia at 3 and 48h post-occlusion. Histological results validated the imaging findings and demonstrated that NRG-1 treated rats had fewer injured neurons in peri-infarct areas 48h post-ischemia. In summary, the neuroprotective effect of NRG-1 in the pMCAo stroke model was demonstrated by prevention of ischemic lesion expansion, reduced infarct volume and protection of neurons from ischemic damage.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Imagen por Resonancia Magnética , Neurregulina-1/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Análisis Espacio-Temporal , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Flujometría por Láser-Doppler , Neurregulina-1/farmacología , Neuroimagen , Fármacos Neuroprotectores/farmacología , Ratas , Accidente Cerebrovascular/complicaciones
18.
J Neuroinflammation ; 12: 64, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25880399

RESUMEN

BACKGROUND: Neuregulin-1 (NRG-1) has been shown to act as a neuroprotectant in animal models of nerve agent intoxication and other acute brain injuries. We recently demonstrated that NRG-1 blocked delayed neuronal death in rats intoxicated with the organophosphate (OP) neurotoxin diisopropylflurophosphate (DFP). It has been proposed that inflammatory mediators are involved in the pathogenesis of OP neurotoxin-mediated brain damage. METHODS: We examined the influence of NRG-1 on inflammatory responses in the rat brain following DFP intoxication. Microglial activation was determined by immunohistchemistry using anti-CD11b and anti-ED1 antibodies. Gene expression profiling was performed with brain tissues using Affymetrix gene arrays and analyzed using the Ingenuity Pathway Analysis software. Cytokine mRNA levels following DFP and NRG-1 treatment was validated by real-time reverse transcription polymerase chain reaction (RT-PCR). RESULTS: DFP administration resulted in microglial activation in multiple brain regions, and this response was suppressed by treatment with NRG-1. Using microarray gene expression profiling, we observed that DFP increased mRNA levels of approximately 1,300 genes in the hippocampus 24 h after administration. NRG-1 treatment suppressed by 50% or more a small fraction of DFP-induced genes, which were primarily associated with inflammatory responses. Real-time RT-PCR confirmed that the mRNAs for pro-inflammatory cytokines interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) were significantly increased following DFP exposure and that NRG-1 significantly attenuated this transcriptional response. In contrast, tumor necrosis factor α (TNFα) transcript levels were unchanged in both DFP and DFP + NRG-1 treated brains relative to controls. CONCLUSION: Neuroprotection by NRG-1 against OP neurotoxicity is associated with the suppression of pro-inflammatory responses in brain microglia. These findings provide new insight regarding the molecular mechanisms involved in the neuroprotective role of NRG-1 in acute brain injuries.


Asunto(s)
Inhibidores de la Colinesterasa/toxicidad , Inhibidores de la Colinesterasa/uso terapéutico , Encefalitis/inducido químicamente , Isoflurofato/toxicidad , Neurregulina-1/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Encéfalo/patología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Intraarteriales , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero , Ratas , Ratas Sprague-Dawley
19.
Neuroscience ; 291: 250-259, 2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-25677097

RESUMEN

(1S,2E,4R,6R,-7E,11E)-2,7,11-cembratriene-4,6-diol (4R) is a precursor to key flavor ingredients in leaves of Nicotiana species. The present study shows 4R decreased brain damage in rodent ischemic stroke models. The 4R-pretreated mice had lower infarct volumes (26.2±9.7 mm3) than those in control groups (untreated: 63.4±4.2 mm3, DMSO: 60.2±14.2 mm3). The 4R-posttreated rats also had less infarct volumes (120±65 mm3) than those in the rats of the DMSO group (291±95 mm3). The results from in vitro experiments indicate that 4R decreased neuro2a cell (neuroblastoma cells) apoptosis induced by oxygen-glucose deprivation (OGD), and improved the population spikes' (PSs) recovery in rat acute hippocampal slices under OGD; a phosphatidylinositol 3-kinase (PI3K) inhibitor, wortmannin, abolished the effect of 4R on PSs recovery. Furthermore, 4R also inhibited monocyte adhesion to murine brain-derived endothelial (bEND5) cells and upregulation of intercellular adhesion molecule-1(ICAM-1) induced by OGD/reoxygenation (OGD/R), and restored the p-Akt level to pre-OGD/R values in bEND5 cells. In conclusion, the present study indicates that 4R has a protective effect in rodent ischemic stroke models. Inhibition of ICAM-1 expression and restoration of Akt phosphorylation are the possible mechanisms involved in cellular protection by 4R.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Diterpenos/farmacología , Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Línea Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Glucosa/deficiencia , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología
20.
J Neuroinflammation ; 11: 9, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24433482

RESUMEN

BACKGROUND: Cerebral Malaria (CM) is a diffuse encephalopathy caused by Plasmodium falciparum infection. Despite availability of antimalarial drugs, CM-associated mortality remains high at approximately 30% and a subset of survivors develop neurological and cognitive disabilities. While antimalarials are effective at clearing Plasmodium parasites they do little to protect against CM pathophysiology and parasite-induced brain inflammation that leads to seizures, coma and long-term neurological sequelae in CM patients. Thus, there is urgent need to explore therapeutics that can reduce or prevent CM pathogenesis and associated brain inflammation to improve survival. Neuregulin-1 (NRG-1) is a neurotrophic growth factor shown to protect against brain injury associated with acute ischemic stroke (AIS) and neurotoxin exposure. However, this drug has not been tested against CM-associated brain injury. Since CM-associated brain injuries and AIS share similar pathophysiological features, we hypothesized that NRG-1 will reduce or prevent neuroinflammation and brain damage as well as improve survival in mice with late-stage experimental cerebral malaria (ECM). METHODS: We tested the effects of NRG-1 on ECM-associated brain inflammation and mortality in P. berghei ANKA (PbA)-infected mice and compared to artemether (ARM) treatment; an antimalarial currently used in various combination therapies against malaria. RESULTS: Treatment with ARM (25 mg/kg/day) effectively cleared parasites and reduced mortality in PbA-infected mice by 82%. Remarkably, NRG-1 therapy (1.25 ng/kg/day) significantly improved survival against ECM by 73% despite increase in parasite burden within NRG-1-treated mice. Additionally, NRG-1 therapy reduced systemic and brain pro-inflammatory factors TNFalpha, IL-6, IL-1alpha and CXCL10 and enhanced anti-inflammatory factors, IL-5 and IL-13 while decreasing leukocyte accumulation in brain microvessels. CONCLUSIONS: This study suggests that NRG-1 attenuates ECM-associated brain inflammation and injuries and may represent a novel supportive therapy for the management of CM.


Asunto(s)
Antimaláricos/uso terapéutico , Encefalitis/tratamiento farmacológico , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/mortalidad , Neurregulina-1/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Arteméter , Artemisininas/uso terapéutico , Conducta Animal/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Encéfalo/parasitología , Encéfalo/patología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/etiología , Encefalitis/patología , Endotelio/efectos de los fármacos , Endotelio/patología , Leucocitos/efectos de los fármacos , Leucocitos/patología , Malaria Cerebral/complicaciones , Ratones , Ratones Endogámicos C57BL , Neurregulina-1/metabolismo , Plasmodium berghei/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...