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










Base de dados
Intervalo de ano de publicação
1.
Am J Neurodegener Dis ; 5(1): 69-73, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27073744

RESUMO

The effects of pyruvate, the end metabolite of glycolysis, on blood-brain barrier (BBB) impairment and immune reactivity were examined in the quinolinic acid (QA)-injected rat striatum. Extensive disruption of BBB was observed at 7 d post QA-injection as demonstrated by increased immunohistochemical staining using antibody against immunoglobulin G (IgG). Animals receiving pyruvate administration (500 mg/kg) with QA-injection exhibited reduced lgG immunoreactivity (by 45%) relative to QA alone. QA intrastriatal injection also resulted in marked increases in the number of infiltrating T-lymphocytes (by 70-fold) and expression of major histocompatibility complex (MHC-class II) (by 45-fold) relative to unlesioned control. Treatment with pyruvate significantly reduced infiltration of T-cells (by 68%) and MHC class II expression (by 48%) induced by QA. These results indicate that QA injection into rat striatum leads to impairment in BBB function with pyruvate administration reducing immune response and BBB leakiness in excitotoxic injury.

2.
J Neuroinflammation ; 12: 144, 2015 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-26255110

RESUMO

BACKGROUND: The chemokine interleukin-8 (IL-8) and its receptor CXCR2 contribute to chemotactic responses in Alzheimer's disease (AD); however, properties of the ligand and receptor have not been characterized in animal models of disease. The primary aim of our study was to examine effects of pharmacological antagonism of CXCR2 as a strategy to inhibit receptor-mediated inflammatory reactivity and enhance neuronal viability in animals receiving intrahippocampal injection of amyloid-beta (Aß1-42). METHODS: In vivo studies used an animal model of Alzheimer's disease incorporating injection of full-length Aß1-42 into rat hippocampus. Immunohistochemical staining of rat brain was used to measure microgliosis, astrogliosis, neuronal viability, and oxidative stress. Western blot and Reverse Transcription PCR (RT-PCR) were used to determine levels of CXCR2 in animal tissue with the latter also used to determine expression of pro-inflammatory mediators. Immunostaining of human AD and non-demented (ND) tissue was also undertaken. RESULTS: We initially determined that in the human brain, AD relative to ND tissue exhibited marked increases in expression of CXCR2 with cell-specific receptor expression prominent in microglia. In Aß1-42-injected rat brain, CXCR2 and IL-8 showed time-dependent increases in expression, concomitant with enhanced gliosis, relative to controls phosphate-buffered saline (PBS) or reverse peptide Aß42-1 injection. Administration of the competitive CXCR2 antagonist SB332235 to peptide-injected rats significantly reduced expression of CXCR2 and microgliosis, with astrogliosis unchanged. Double staining studies demonstrated localization of CXCR2 and microglial immunoreactivity nearby deposits of Aß1-42 with SB332235 effective in inhibiting receptor expression and microgliosis. The numbers of neurons in granule cell layer (GCL) were reduced in rats receiving Aß1-42, compared with PBS, with administration of SB332235 to peptide-injected animals conferring neuroprotection. Oxidative stress was indicated in the animal model since both 4-hydroxynonenal (4-HNE) and hydroethidine (HEt) were markedly elevated in Aß1-42 vs. PBS-injected rat brain and diminished with SB332235 treatment. CONCLUSION: Overall, the findings suggest critical roles for CXCR2-dependent inflammatory responses in an AD animal model with pharmacological modulation of the receptor effective in inhibiting inflammatory reactivity and conferring neuroprotection against oxidative damage.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Anti-Inflamatórios/uso terapêutico , Inflamação/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Receptores de Interleucina-8B/antagonistas & inibidores , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Animais , Encéfalo/patologia , Feminino , Gliose/patologia , Hipocampo , Humanos , Mediadores da Inflamação , Masculino , Microinjeções , Pessoa de Meia-Idade , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos , Ratos
3.
PLoS One ; 8(10): e76860, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24146937

RESUMO

Neural stem cell (NSC) replacement therapy is considered a promising cell replacement therapy for various neurodegenerative diseases. However, the low rate of NSC survival and neurogenesis currently limits its clinical potential. Here, we examined if hippocampal long-term potentiation (LTP), one of the most well characterized forms of synaptic plasticity, promotes neurogenesis by facilitating proliferation/survival and neuronal differentiation of NSCs. We found that the induction of hippocampal LTP significantly facilitates proliferation/survival and neuronal differentiation of both endogenous neural progenitor cells (NPCs) and exogenously transplanted NSCs in the hippocampus in rats. These effects were eliminated by preventing LTP induction by pharmacological blockade of the N-methyl-D-aspartate glutamate receptor (NMDAR) via systemic application of the receptor antagonist, 3-[(R)-2-carboxypiperazin-4-yl]-propyl-1-phosphonic acid (CPP). Moreover, using a NPC-neuron co-culture system, we were able to demonstrate that the LTP-promoted NPC neurogenesis is at least in part mediated by a LTP-increased neuronal release of brain-derived neurotrophic factor (BDNF) and its consequent activation of tropomysosin receptor kinase B (TrkB) receptors on NSCs. Our results indicate that LTP promotes the neurogenesis of both endogenous and exogenously transplanted NSCs in the brain. The study suggests that pre-conditioning of the host brain receiving area with a LTP-inducing deep brain stimulation protocol prior to NSC transplantation may increase the likelihood of success of using NSC transplantation as an effective cell therapy for various neurodegenerative diseases.


Assuntos
Diferenciação Celular , Potenciação de Longa Duração/fisiologia , Células-Tronco Neurais/citologia , Neurônios/citologia , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Giro Denteado/citologia , Células HEK293 , Humanos , Potenciação de Longa Duração/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/transplante , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transplante de Células-Tronco
4.
Curr Alzheimer Res ; 10(3): 252-60, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23534534

RESUMO

We have examined the anti-angiogenic compound, angiostatin as a modulator of inflammatory reactivity and vascular responses and for neuroprotection in an animal model of Alzheimer's disease (AD). Intra-hippocampal amyloidbeta (Aß1₋42) injection, relative to controls phosphate buffer saline (PBS) or reverse peptide Aß42₋1, increased gliosis in the molecular layer (ML) of rat hippocampus. Vascular remodeling was indicated from increased microvessel immunoreactivity (ir) in ML suggesting the possibility of an angiogenic response to peptide injection. Administration of Aß1₋42 also induced a loss of neurons in the granule cell region of hippocampus relative to controls. Treatment of peptide-injected rats with angiostatin was associated with a spectrum of modulatory effects including reduced microgliosis (by 34%), diminished microvessel ir (by 36%) and increased neuronal viability (by 31%) compared with peptide injection alone. Angiostatin treatment was ineffective in reducing astrogliosis induced by Aß1₋42 and applied alone the compound had no significant effect to alter gliosis, microvessel ir or neuronal viability compared with PBS control. In vitro, angiostatin significantly attenuated secretion of the pro-angiogenic agent, vascular endothelial growth factor (VEGF) in lipopolysaccharide (LPS)-stimulated THP-1 cells. Our findings provide novel evidence for a broad spectrum of angiostatin effects in an animal model of AD including actions to reduce inflammatory reactivity, stabilize vascular remodeling and confer neuroprotection. The overall effects of angiostatin are consistent with actions of the compound to inhibit microglial secretion of VEGF.


Assuntos
Doença de Alzheimer/patologia , Inibidores da Angiogênese/farmacologia , Angiostatinas/farmacologia , Encéfalo/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Masculino , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Neuron ; 75(6): 1094-104, 2012 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-22998876

RESUMO

Astrocytes are proposed to participate in brain energy metabolism by supplying substrates to neurons from their glycogen stores and from glycolysis. However, the molecules involved in metabolic sensing and the molecular pathways responsible for metabolic coupling between different cell types in the brain are not fully understood. Here we show that a recently cloned bicarbonate (HCO3⁻) sensor, soluble adenylyl cyclase (sAC), is highly expressed in astrocytes and becomes activated in response to HCO3⁻ entry via the electrogenic NaHCO3 cotransporter (NBC). Activated sAC increases intracellular cAMP levels, causing glycogen breakdown, enhanced glycolysis, and the release of lactate into the extracellular space, which is subsequently taken up by neurons for use as an energy substrate. This process is recruited over a broad physiological range of [K⁺](ext) and also during aglycemic episodes, helping to maintain synaptic function. These data reveal a molecular pathway in astrocytes that is responsible for brain metabolic coupling to neurons.


Assuntos
Adenilil Ciclases/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Bicarbonatos/farmacologia , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , 1-Metil-3-Isobutilxantina/farmacologia , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/ultraestrutura , Ácidos Cumáricos/farmacologia , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Glucose/deficiência , Glicogênio/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Técnicas In Vitro , Ácido Láctico/metabolismo , Microscopia Imunoeletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neurônios/ultraestrutura , Técnicas de Patch-Clamp , Inibidores de Fosfodiesterase/farmacologia , Potássio/metabolismo , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia
6.
Neurobiol Dis ; 45(1): 438-49, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21946335

RESUMO

Central nervous system (CNS) inflammatory processes including microglial activation have been implicated in the pathogenesis of neurodegenerative diseases such as Huntington Disease (HD). We report age-dependent changes in striatal microglial morphology and vasculature in the YAC128 mouse model of HD. Decreases in microglial ramification along with a decrease in vessel diameter and increased vessel density and length suggest the presence of microgliosis and proangiogenic activity in YAC128 mice. Our hypothesis for this study was that the changes in microglial morphology and perturbations in vasculature may be involved in the pathogenesis of HD and that peripheral challenge with the bacterial endotoxin, lipopolysaccharide (LPS), will exacerbate these microglial and vascular changes as well as the HD phenotype in YAC128 mice at 12 months. Chronic peripheral LPS (1mg/kg) potentiated microglial activation indicated by an increase in microglial cell body size and retraction of processes. This potentiation in microglial activation with chronic peripheral LPS challenge was paralleled with vascular remodeling including dilatation, increased vessel wall thickness, increased BBB permeability and fibrinogen deposition in YAC128 striatum. Although peripheral LPS caused an increase in microglial activation and degenerative changes in cerebrovasculature, the phenotypic hallmarks of HD in YAC128 mice such as motor coordination deficits and decreased striatal volume were not exacerbated by chronic peripheral LPS exposure. This study identifies age-dependent increases in microglial activation and angiogenesis in YAC128 at 12 months. Peripheral inflammation induced by chronic LPS causes similar changes but does not influence the HD phenotype in YAC128 mice.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/patologia , Doença de Huntington/patologia , Microglia/patologia , Fatores Etários , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Forma Celular , Corpo Estriado/irrigação sanguínea , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Modelos Animais de Doenças , Feminino , Doença de Huntington/genética , Doença de Huntington/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Fenótipo
7.
Int J Alzheimers Dis ; 2011: 918280, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21969915

RESUMO

The validity of amyloid-ß peptide (Aß(1-42)) intrahippocampal injection, as an animal model of Alzheimer's disease (AD), has previously been considered in terms of inflammatory reactivity and neuronal damage. In this work, we have extended the testing of the animal model to vasculature by comparison of selected properties of microvessels in vivo with those in human AD brain tissue. The injection of Aß(1-42), relative to control PBS (phosphate buffered saline), increased the mean number of microvessels and diminished the mean length of microvessels in the molecular layer of dentate gyrus. The animal model showed Aß(1-42), but not PBS, injection was associated with abnormalities in morphology of microvessels which were characterized as looping, fragmented, knob-like, uneven, and constricted. In particular, numbers of constricted microvessels, defined as vessels with diameters less than 3 µm, were considerably enhanced for Aß(1-42), compared to PBS, injection. In comparison, human AD brain demonstrated an elevated number of microvessels with a diminished mean length relative to nondemented (ND) brain. Additionally, microvessel perturbations in AD brain showed a similar pattern of morphological abnormalities to those observed in Aß(1-42)-injected rat hippocampus. Constricted microvessels were a prominent feature of AD brain but were rarely observed in ND tissue. These results provide the first evidence that a peptide-injection animal model exhibits a commonality in perturbations of microvessels compared with those evident in AD brain.

8.
J Alzheimers Dis ; 27(3): 531-41, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21841251

RESUMO

Pyrazole derivatives are well documented to possess anti-inflammatory activity but their effects on microglial activation are unknown. We determined the efficacy of the novel pyrazole compound 2-MBAPA (R/S-(±)-2-Methylbenzylamino 2-oxo-N-[4-cyano-1-phenyl-1H-pyrazol-5-yl] acetamide) on activated microglia under conditions relevant to inflammation in Alzheimer's disease (AD) brain. The compound at a non-toxic concentration inhibited secretion of tumor necrosis factor (TNF)-α by activated human microglia and attenuated toxicity of conditioned medium from activated human microglia towards human SH-SY5Y neuroblastoma cells in vitro. The 2-MBAPA neuroprotection was further demonstrated in vivo using an animal model of AD. The compound inhibited microgliosis, but not astrogliosis, in amyloid-ß peptide (Aß)(1-42)-injected rat brain. 2-MBAPA also diminished neuronal loss in the dentate gyrus caused by Aß(1-42) injection. These results indicate that this novel pyrazole compound confers neuroprotection by inhibiting microglial activation. Therefore, further studies with 2-MBAPA and novel analogues based on this lead compound are warranted in an effort to develop new pharmacological agents that may be useful for slowing down progression of AD and other neuroinflammatory disorders associated with activated microglia.


Assuntos
Microglia/efeitos dos fármacos , Microglia/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/patologia , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Humanos , Masculino , Microglia/patologia , Síndromes Neurotóxicas/metabolismo , Ratos , Ratos Sprague-Dawley
9.
J Neuropathol Exp Neurol ; 70(1): 13-22, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21157381

RESUMO

We examined the expression and pharmacological modulation of the purinergic receptor P2X7R in a C6 glioma model. Intrastriatal injection of C6 cells induced a time-dependent growth of tumor; at 2 weeks postinjection immunohistochemical analysis demonstrated higher levels of P2X7R in glioma-injected versus control vehicle-injected brains. P2X7R immunoreactivity colocalized with tumor cells and microglia, but not endogenous astrocytes. Intravenous administration of the P2X7R antagonist brilliant blue G (BBG) inhibited tumor growth in a spatially dependent manner from the C6 injection site. Treatment with BBG reduced tumor volume by 52% versus that in controls. Double immunostaining indicated that BBG treatment did not alter microgliosis, astrogliosis, or vasculature vessels in C6-injected animals. In vitro, BBG reduced the expression of P2X7R and glioma chemotaxis induced by the P2X7R ligand, 2',3'-O-(4-benzoyl-benzoyl)adenosine triphosphate (BzATP). Immunohistochemical staining of human glioblastoma tissue samples demonstrated greater expression of P2X7R compared to control nontumor samples. These results suggest that the efficacy of BBG in inhibiting tumor growth is primarily mediated by direct actions of the compound on P2X7R in glioma cells and that pharmacological inhibition of this purinergic receptor might serve as a strategy to slow the progression of brain tumors.


Assuntos
Neoplasias Encefálicas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Glioma/metabolismo , Inibidores do Crescimento/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7 , Idoso , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Sistemas de Liberação de Medicamentos/métodos , Glioma/tratamento farmacológico , Glioma/patologia , Inibidores do Crescimento/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Antagonistas do Receptor Purinérgico P2X/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7/biossíntese , Receptores Purinérgicos P2X7/genética , Corantes de Rosanilina/administração & dosagem , Corantes de Rosanilina/farmacologia , Células Tumorais Cultivadas
10.
J Neurosci ; 30(17): 5843-54, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20427645

RESUMO

Scar formation in the nervous system begins within hours after traumatic injury and is characterized primarily by reactive astrocytes depositing proteoglycans that inhibit regeneration. A fundamental question in CNS repair has been the identity of the initial molecular mediator that triggers glial scar formation. Here we show that the blood protein fibrinogen, which leaks into the CNS immediately after blood-brain barrier (BBB) disruption or vascular damage, serves as an early signal for the induction of glial scar formation via the TGF-beta/Smad signaling pathway. Our studies revealed that fibrinogen is a carrier of latent TGF-beta and induces phosphorylation of Smad2 in astrocytes that leads to inhibition of neurite outgrowth. Consistent with these findings, genetic or pharmacologic depletion of fibrinogen in mice reduces active TGF-beta, Smad2 phosphorylation, glial cell activation, and neurocan deposition after cortical injury. Furthermore, stereotactic injection of fibrinogen into the mouse cortex is sufficient to induce astrogliosis. Inhibition of the TGF-beta receptor pathway abolishes the fibrinogen-induced effects on glial scar formation in vivo and in vitro. These results identify fibrinogen as a primary astrocyte activation signal, provide evidence that deposition of inhibitory proteoglycans is induced by a blood protein that leaks in the CNS after vasculature rupture, and point to TGF-beta as a molecular link between vascular permeability and scar formation.


Assuntos
Astrócitos/fisiologia , Córtex Cerebral/lesões , Córtex Cerebral/fisiopatologia , Cicatriz/fisiopatologia , Fibrinogênio/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/irrigação sanguínea , Fibrinogênio/genética , Gliose/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neuritos/fisiologia , Neurocam , Fosforilação , Proteoglicanas/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo
11.
BMC Neurosci ; 11: 42, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20346117

RESUMO

BACKGROUND: Midkine (MK), a member of the heparin-binding growth factor family, which includes MK and pleiotrophin, is known to possess neurotrophic and neuroprotective properties in the central nervous system. Previous studies have shown that MK is an effective neuroprotective agent in reducing retinal degeneration caused by excessive light and decreasing hippocampal neuronal death in ischemic gerbil brain. The present study was undertaken to investigate whether MK acts as an anticonvulsant in kainic acid (KA)-induced seizure in mouse and blocks KA-mediated neuronal cell death in hippocampus. RESULTS: Increased expression of MK was found in hippocampus of mouse following seizures induced by intracerebroventricular injection of KA, and MK expression was found in glial fibrillary acidic protein (GFAP)-positive astrocytes. Concurrent injection of MK and KA attenuated KA-induced seizure activity and cell death of hippocampal neurons including pyramidal cells and glutamic acid decarboxylase 67 (GAD67)-positive GABAergic interneurons in the CA3 and hilar area. CONCLUSION: The results of the present study indicate that MK functions as an anticonvulsant and neuroprotective agent in hippocampus during KA-induced seizures.


Assuntos
Anticonvulsivantes/farmacologia , Citocinas/farmacologia , Epilepsia/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Anticonvulsivantes/uso terapêutico , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Biomarcadores/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Citocinas/uso terapêutico , Modelos Animais de Doenças , Epilepsia/induzido quimicamente , Epilepsia/fisiopatologia , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/fisiopatologia , Proteína Glial Fibrilar Ácida/metabolismo , Glutamato Descarboxilase/metabolismo , Hipocampo/patologia , Hipocampo/fisiopatologia , Injeções Intraventriculares , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Ácido Caínico/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Midkina , Degeneração Neural/fisiopatologia , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/antagonistas & inibidores , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Ácido gama-Aminobutírico/metabolismo
12.
J Neuroinflammation ; 6: 39, 2009 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20030829

RESUMO

BACKGROUND: Transplantation of neural progenitor cells (NPC) constitutes a putative therapeutic maneuver for use in treatment of neurodegenerative diseases. At present, effects of NPC transplantation in Alzheimer's disease (AD) brain are largely unknown and a primary objective of this work was to demonstrate possible efficacy of NPC administration in an animal model of AD. The benefits of transplantation could involve a spectrum of effects including replacement of endogenous neurons or by conferring neuroprotection with enhancement of neurotrophic factors or diminishing levels of neurotoxic agents. Since chronic inflammation is a characteristic property of AD brain, we considered that transplantation of NPC could have particular utility in inhibiting ongoing inflammatory reactivity. We have tested intrahippocampal transplantation of NPC for efficacy in attenuating inflammatory responses and for neuroprotection in beta-amyloid (Abeta1-42) peptide-injected rat hippocampus. METHODS: Spheres of neural progenitor cells were grown from dissociated telencephalon tissue of rat embryos. NPC were infected with lentiviral vector green fluorescent protein (GFP) with subsequent cell transplantation into rat hippocampus previously injected (3 d prior) with Abeta1-42 peptide or PBS control. Immunohistochemical analysis was carried out (7 d post-NPC transplantation, 10 d post-peptide/PBS injection) for GFP, microgliosis (Iba-1 marker), astrogliosis (GFAP marker), neuron viability (MAP-2 marker) and levels of the proinflammatory cytokine, TNF-alpha. RESULTS: Successful infection of cultured NPC with lentiviral vector green fluorescent protein (GFP) was demonstrated prior to cell transplantation into rat hippocampus. In vivo, immunohistochemical staining showed migration of GFP-positive cells, in a region of dentate gyrus between Abeta1-42/PBS injection site and NPC transplantation site, was increased x2.8-fold with Abeta1-42 compared to PBS injection. Double immunostaining in peptide-injected brain indicated GFP association with nestin and GFAP, but not MAP-2. Cell-specific immunostaining showed marked increases in microgliosis and astrogliosis in Abeta1-42-injected brain (respective increases of x4.3- and x4.6-fold compared with PBS injection). NPC transplantation significantly reduced microgliosis (by 38%) but not astrogliosis in peptide-injected hippocampus. The proinflammatory cytokine TNF-alpha was elevated by 6.7-fold (peptide vs PBS injection) with NPC administration attenuating levels of TNF-alpha (by 40%). Peptide-injected brain demonstrated neuronal loss (MAP-2 staining reduced by 45% vs PBS injection) with NPC transplantation effective in conferring neuroprotection (26% recovery of neurons). CONCLUSIONS: These findings indicate efficacy for NPC transplantation in an animal model of AD with effects consistent with cellular actions to attenuate inflammatory reactivity induced by intrahippocampal peptide injection.


Assuntos
Doença de Alzheimer/cirurgia , Transplante de Tecido Encefálico , Encefalite/cirurgia , Neurônios/transplante , Transplante de Células-Tronco , Telencéfalo/transplante , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides , Animais , Morte Celular , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Encefalite/fisiopatologia , Hipocampo/fisiopatologia , Hipocampo/cirurgia , Masculino , Neuroimunomodulação/fisiologia , Neurônios/fisiologia , Fragmentos de Peptídeos , Ratos , Ratos Sprague-Dawley , Células-Tronco/fisiologia , Telencéfalo/fisiologia
13.
J Neurosci ; 29(1): 3-13, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19129379

RESUMO

We hypothesize that microglial chemotactic responses to amyloid-beta peptide (Abeta(1-42)) serve as an early and integral component of inflammatory response in Alzheimer's disease (AD) brain. This study reports a receptor for vascular endothelial growth factor (VEGF), termed VEGF-1 (Flt-1), subserves microglial chemotactic responses induced by Abeta(1-42) stimulation, in vivo and in vitro. Expression of Flt-1 was significantly increased in tissue obtained from AD patients [compared with tissue from nondemented (ND) individuals], in Abeta(1-42)-injected rat hippocampus, and in peptide-stimulated human microglia. Single and double immunohistochemical staining demonstrated marked immunoreactivity, for both Flt-1 and its ligand VEGF, in association with microglia and Abeta deposits in AD, but not ND, brain tissue. Functionally, treatment with anti-Flt-1 antibody was highly effective in inhibiting microglial mobility and chemotactic responses measured in vitro using a transwell migration assay. In vivo, transplanted enhanced green fluorescent protein (EGFP)-labeled microglia exhibited Flt-1-dependent chemotaxis induced by peptide injection with anti-Flt-1 effective in blocking migration of cells. Importantly, anti-Flt-1 reduction of microglial mobility was neuroprotective in peptide-injected hippocampus and associated with a significant increase in numbers of viable hippocampal neurons. The results of this study suggest critical functional roles for Flt-1 in mediating microglial chemotactic inflammatory responses which contribute to pathological conditions in AD brain.


Assuntos
Doença de Alzheimer/patologia , Movimento Celular/fisiologia , Córtex Entorrinal/patologia , Microglia/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/administração & dosagem , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Antígeno CD11b/metabolismo , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Córtex Entorrinal/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Antígenos HLA-DR/metabolismo , Hipocampo/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fragmentos de Peptídeos/administração & dosagem , Fosfopiruvato Hidratase/metabolismo , Mudanças Depois da Morte , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
14.
Glia ; 57(8): 898-907, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19115379

RESUMO

Lysophosphatidylcholine (LPC), a major phospholipid component of atherogenic oxidized LDL, is implicated in atherosclerosis and, recently, in neurodegenerative diseases. We investigated the immunomodulatory functions of LPC in the central nervous system (CNS) using both an in vivo rat model, and in vitro culture systems of human primary astrocytes and a microglia cell line, HMO6. Compared with PBS injection, 20 nmol LPC-injection into the rat striatum increased astrocyte and microglial accumulation and elevated iNOS expression; concomitantly a time-dependent decrease in number of neurons was exhibited. In vitro studies on astrocytes and HMO6 cells showed that LPC increased the gene expression of proinflammatory factors IL-1beta, COX-2, and GM-CSF. LPC also induced chemotactic responses in HMO6 cells. Inhibition of rho kinase by fasudil, Y27632, or expressing a dominant negative form of rho kinase inhibited the LPC-induced IL-1beta mRNA expression in both astrocytes and HMO6. Moreover, intraperitoneal fasudil injection inhibited the LPC-induced microglial accumulation and iNOS expression and also was effective in protecting against neuronal loss. Silencing G2A, a specific receptor for LPC, inhibited proinflammatory gene expression and HMO6 migration. Overall, our results indicate that LPC induced considerable neuroinflammatory reactivity in glia mediated by rho kinase-dependent pathways with inhibition of these pathways conferring significant extents of neuroprotection.


Assuntos
Lisofosfatidilcolinas/farmacologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Quinases Associadas a rho/metabolismo , Amidas/farmacologia , Animais , Encéfalo/citologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feto , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lisofosfatidilcolinas/metabolismo , Masculino , Óxido Nítrico Sintase Tipo II/metabolismo , Piridinas/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
15.
J Cell Mol Med ; 13(9A): 2911-25, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18657226

RESUMO

This study has used immunohistochemical examination of tissue obtained from Alzheimer's disease (AD) brains and rat hippocampus injected with Abeta(1-42) peptide to determine effects of induced inflammatory reactivity on integrity of blood-brain barrier (BBB) and viability of neurons. Tissue from AD, but not non-demented, brains exhibited a diffuse pattern of staining for fibrinogen and immunoglobulin (IgG) indicative of BBB leakiness with considerable fibrinogen immunoreactivity (ir) appearing in association with Abeta deposits. Immunostaining for the endothelial cell specific glycoprotein, von Willebrand factor, showed morphological evidence for altered blood vessels in AD tissue. AD brains also demonstrated extensive areas of fibrinogen ir in association with microglial reactivity. In vivo, intra-hippocampal injection of Abeta(1-42) caused time-dependent (1-7 days after injection) increases in double staining of fibrinogen with areas of microgliosis. Two independent pharmacological strategies were employed to examine how Abeta(1-42) stimulation (7 days injection) may be linked to neurodegeneration. The defibrinogenating compound, ancrod, reduced inflammatory reactivity, levels of parenchymal fibrinogen and IgG, and was neuroprotective. These results prompted use of Abeta(1-42) plus fibrinogen as a novel in vivo inflammatory stimulus and this combination significantly enhanced inflammatory reactivity, vascular perturbations and neuronal damage compared to Abeta(1-42) alone. A second approach, using anti-Mac-1 (antibody for antigen CD11b) to block activation of microglia, was highly effective in attenuating effects of Abeta(1-42) plus fibrinogen amplification of inflammatory and vascular responses and conferred significant neuroprotection. The overall findings from study of AD tissue and in vivo in Abeta(1-42) and Abeta(1-42) plus fibrinogen stimulated rat hippocampus suggest microglial responses to promote increased extravasation of blood protein as a critical component in amplifying inflammatory reactivity and causing neuronal damage in inflamed AD brain.


Assuntos
Doença de Alzheimer/patologia , Barreira Hematoencefálica/patologia , Encéfalo/patologia , Fibrinogênio/metabolismo , Inflamação/patologia , Microglia/patologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/complicações , Peptídeos beta-Amiloides/farmacologia , Ancrod/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Demência/complicações , Demência/patologia , Fluorescência , Humanos , Inflamação/complicações , Antígeno de Macrófago 1/metabolismo , Masculino , Microglia/efeitos dos fármacos , Pessoa de Meia-Idade , Fragmentos de Peptídeos/farmacologia , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fator de von Willebrand/metabolismo
16.
Neuroreport ; 19(17): 1715-9, 2008 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-18852683

RESUMO

Pharmacological antagonism of the ionotropic purinergic P2X7R has been studied for effects on inflammatory reactivity and neuronal viability in amyloid-beta1-42-injected rat hippocampus. Amyloid-beta1-42-injected brains (7-day postinjection) demonstrated marked increases in P2X7R expression, gliosis, leakiness of blood-brain barrier and loss of hippocampal neurons. The P2X7R antagonist, brilliant blue G reduced levels of purinergic receptor expression, attenuated gliosis, diminished leakiness of blood-brain barrier and was neuroprotective in peptide-injected brain. Brilliant blue G also demonstrated neuroprotection and antagonism against inflammatory responses induced by the P2X7R agonist, 2',3'-(benzoyl-4-benzoyl)-ATP. The findings constitute the first report that pharmacological inhibition of P2X7R, possibly by acting to inhibit inflammatory reactivity, confers neuroprotection in an animal model of Alzheimer's disease brain.


Assuntos
Doença de Alzheimer/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/metabolismo , Receptores Purinérgicos P2/metabolismo , Trifosfato de Adenosina/administração & dosagem , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/toxicidade , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Gliose/metabolismo , Gliose/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Imuno-Histoquímica , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/agonistas , Fármacos Neuroprotetores/antagonistas & inibidores , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/toxicidade , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/farmacologia , Agonistas do Receptor Purinérgico P2 , Antagonistas do Receptor Purinérgico P2 , Ratos , Ratos Sprague-Dawley
17.
J Neuroinflammation ; 5: 18, 2008 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-18492281

RESUMO

BACKGROUND: Excitotoxic brain insult is associated with extensive neuronal damage but could also cause inflammatory reactivity and vascular remodeling. The effects of the vascular endothelial growth factor (VEGF) inhibitor, Cyclo-VEGI on expression of VEGF, microgliosis and astrogliosis, blood-brain barrier (BBB) integrity and neuronal viability have been studied following intra-striatal injection of the excitotoxin, quinolinic acid (QUIN). The purpose of this study was to examine VEGF-dependent inflammatory responses in excitotoxin-injected brain and their dependence on pharmacological antagonism of VEGF receptors. METHODS: Single and double immunofluorescence staining of cellular (microglia, astrocyte, neuron) responses and dye and protein infiltration of blood-brain barrier have been applied in the absence, and presence, of pharmacological modulation using a VEGF receptor antagonist, Cyclo-VEGI. Dunn-Bonferroni statistical analysis was used to measure for significance between animal groups. RESULTS: Detailed analysis, at a single time point of 1 d post-QUIN injection, showed excitotoxin-injected striatum to exhibit marked increases in microgliosis (ED1 marker), astrogliosis (GFAP marker) and VEGF expression, compared with PBS injection. Single and double immunostaining demonstrated significant effects of Cyclo-VEGI treatment of QUIN-injected striatum to inhibit microgliosis (by 38%), ED1/VEGF (by 42%) and VEGF striatal immunoreactivity (by 43%); astrogliosis and GFAP/VEGF were not significantly altered with Cyclo-VEGI treatment. Leakiness of BBB was indicated by infiltration of Evans blue dye and plasma protein fibrinogen into QUIN-injected striatum with barrier permeability restored by 62% (Evans blue permeability) and 49% (fibrinogen permeability) with Cyclo-VEGI application. QUIN-induced toxicity was demonstrated with loss of striatal neurons (NeuN marker) and increased neuronal damage (Fluoro-Jade marker) with significant neuroprotection conferred by Cyclo-VEGI treatment (33% increase in NeuN and 38% decrease in Fluoro-Jade). CONCLUSION: An antagonist for VEGF receptor-mediated signaling, Cyclo-VEGI, has shown efficacy in a broad spectrum of activity against striatal excitotoxic insult including inhibition of microgliosis, reduction in leakiness of BBB and parenchymal infiltration of plasma fibrinogen and in conferring significant protection for striatal neurons. Antagonism of VEGF-mediated activity, possibly targeting VEGF receptors on reactive microglia, is suggested as a neuroprotective mechanism against inflammatory reactivity and a novel strategy to attenuate acute excitotoxic damage.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Fatores de Crescimento Endotelial/uso terapêutico , Gliose/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/toxicidade , Peptídeos Cíclicos/uso terapêutico , Ácido Quinolínico/toxicidade , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Corpo Estriado/fisiopatologia , Avaliação Pré-Clínica de Medicamentos , Fatores de Crescimento Endotelial/farmacocinética , Fatores de Crescimento Endotelial/farmacologia , Gliose/induzido quimicamente , Gliose/patologia , Gliose/prevenção & controle , Injeções , Injeções Intraventriculares , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Peptídeos Cíclicos/farmacocinética , Peptídeos Cíclicos/farmacologia , Ratos , Ratos Sprague-Dawley
18.
Cancer Lett ; 260(1-2): 79-87, 2008 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-18039556

RESUMO

Our results demonstrate the first findings of expression and function of the purinergic P2X7 receptor (P2X7R) in rat C6 glioma cells. P2X7R mRNA and protein were present in unstimulated C6 cells and were up-regulated by cell exposure to the P2X7R agonist, 2',3'-(benzoyl-4-benzoyl)-ATP (BzATP). Activation of P2X7R in C6 in response to BzATP led to increased mobilization of intracellular calcium [Ca2+]i and formation of large pores. Chronic exposure of C6 cells to BzATP enhanced the expression of pro-inflammatory factors including MCP-1, IL-8 and VEGF. In a scratch-wound migration assay, the P2X7R was shown to regulate cell mobility. The overall results suggest that P2X7R activation in C6 is linked with increased pro-inflammatory factors and tumor cell migration.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Agonistas do Receptor Purinérgico P2 , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/genética , Glioma/patologia , Interleucina-8/metabolismo , RNA Mensageiro/metabolismo , Ratos , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X7 , Fatores de Tempo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Neurobiol Dis ; 29(2): 254-66, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17964176

RESUMO

Injection of Abeta(1-42) peptide into rat hippocampus has been found to induce microglial reactivity and perturbed vasculature in an animal model of inflamed Alzheimer's disease (AD) brain. We report the anti-angiogenic and anti-inflammatory compound, thalidomide, to significantly inhibit peptide-induced vascular changes including endothelial cell proliferation (marker rat endothelial cell antigen-1, RECA-1), angiogenic activity (marker laminin) and leakiness of blood-brain barrier (BBB, marker albumin). Thalidomide also blocked microgliosis and astrogliosis with double immunostaining showing considerable regions of association of activated microglia with vascular remodeling and leaky BBB. Thalidomide inhibition of the glial-derived proinflammatory/angiogenic factor TNF-alpha (tumor necrosis factor-alpha) in Abeta(1-42)-injected brain and also in vitro from peptide-activated human microglia could underly the changes in vascular processes. Thalidomide treatment in vivo was also associated with a significant reduction in hippocampal neuronal loss. Our findings suggest altered cerebral vasculature as an integral component of inflammatory responses with thalidomide an effective inhibitor of gliosis, vascular changes and TNF-alpha leading to neuroprotection in an animal model of inflamed AD brain.


Assuntos
Doença de Alzheimer/metabolismo , Inibidores da Angiogênese/farmacologia , Encéfalo/patologia , Células Endoteliais/efeitos dos fármacos , Talidomida/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Análise de Variância , Inibidores da Angiogênese/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Interações Medicamentosas , Humanos , Laminina/metabolismo , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteínas de Neoplasias/metabolismo , Fragmentos de Peptídeos , Proteoglicanas/metabolismo , Ratos , Ratos Sprague-Dawley , Talidomida/uso terapêutico
20.
J Neurosci ; 27(18): 4957-68, 2007 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-17475804

RESUMO

We investigated the involvement and roles of the ionotropic purinergic receptor P2X(7)R in microglia in mediating lipopolysaccharide (LPS)-induced inflammatory responses and neuronal damage in rat striatum. A detailed in vivo study showed that LPS injection into striatum markedly increased the expression of P2X(7)R in microglia compared with control (saline)-injected animals. Additionally, LPS injection upregulated a broad spectrum of proinflammatory mediators, including inducible nitric oxide synthase (nitric oxide production marker), 3-nitrotyrosine (peroxynitrite-mediated nitration marker), 4-hydroxynonenal (lipid peroxidation marker), and 8-hydroxy-2'-deoxyguanosine (oxidative DNA damage marker), and reduced neuronal viability. The P2X(7)R antagonist oxidized ATP (oxATP) was effective in attenuating expressions of all inflammatory mediators and in addition inhibited LPS-induced activation of the cellular signaling factors p38 mitogen-activated protein kinase and transcriptional factor nuclear factor kappaB. Most importantly, in vivo, oxATP blockade of P2X(7)R also reduced numbers of caspase-3-positive neurons and increased neuronal survival in LPS-injected brain. In vitro, LPS stimulation of cultured human microglia enhanced cellular expressions of a host of proinflammatory factors, including cyclooxygenase-2, interleukin-1beta (IL-1beta), IL-6, IL-12, and tumor necrosis factor-alpha; all factors were inhibited by oxATP. A novel finding was that LPS potentiated intracellular [Ca(2+)](i) mobilization induced by the P2X(7)R ligand 2',3'-O-(4-benzoyl-benzoyl) ATP, which could serve as a mechanistic link for P2X(7)R amplification of inflammatory responses. Our results suggest critical roles for P2X(7)R in mediating inflammation and inhibition of this subtype purinergic receptor as a novel therapeutic approach to reduce microglial activation and confer neuroprotection in inflamed and diseased brain.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Lipopolissacarídeos/toxicidade , Microglia/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Receptores Purinérgicos P2/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Células Cultivadas , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...