Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurovirol ; 20(1): 39-53, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24420448

RESUMO

HIV-associated neurocognitive disorder (HAND), characterized by a wide spectrum of behavioral, cognitive, and motor dysfunctions, continues to affect approximately 50 % of HIV(+) patients despite the success of combination antiretroviral drug therapy (cART) in the periphery. Of note, potential toxicity of antiretroviral drugs in the central nervous system (CNS) remains remarkably underexplored and may contribute to the persistence of HAND in the cART era. Previous studies have shown antiretrovirals (ARVs) to be neurotoxic in the peripheral nervous system in vivo and in peripheral neurons in vitro. Alterations in lipid and protein metabolism, mitochondrial damage, and oxidative stress all play a role in peripheral ARV neurotoxicity. We hypothesized that ARVs also induce cellular stresses in the CNS, ultimately leading to neuronal damage and contributing to the changing clinical and pathological picture seen in HIV-positive patients in the cART era. In this report, we show that ARVs are neurotoxic in the CNS in both pigtail macaques and rats in vivo. Furthermore, in vitro, ARVs lead to accumulation of reactive oxygen species (ROS), and ultimately induction of neuronal damage and death. Whereas ARVs alone caused some activation of the endogenous antioxidant response in vitro, augmentation of this response by a fumaric acid ester, monomethyl fumarate (MMF), blocked ARV-induced ROS generation, and neuronal damage/death. These findings implicate oxidative stress as a contributor to the underlying mechanisms of ARV-induced neurotoxicity and will provide an access point for adjunctive therapies to complement ARV therapy and reduce neurotoxicity in this patient population.


Assuntos
Complexo AIDS Demência/patologia , Antirretrovirais/toxicidade , Encéfalo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Western Blotting , Encéfalo/patologia , Encéfalo/virologia , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Macaca , Masculino , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
J Immunol ; 187(10): 5015-25, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21976775

RESUMO

Despite antiretroviral therapy (ART), HIV infection promotes cognitive dysfunction and neurodegeneration through persistent inflammation and neurotoxin release from infected and/or activated macrophages/microglia. Furthermore, inflammation and immune activation within both the CNS and periphery correlate with disease progression and morbidity in ART-treated individuals. Accordingly, drugs targeting these pathological processes in the CNS and systemic compartments are needed for effective, adjunctive therapy. Using our in vitro model of HIV-mediated neurotoxicity, in which HIV-infected monocyte-derived macrophages release excitatory neurotoxins, we show that HIV infection dysregulates the macrophage antioxidant response and reduces levels of heme oxygenase-1 (HO-1). Furthermore, restoration of HO-1 expression in HIV-infected monocyte-derived macrophages reduces neurotoxin release without altering HIV replication. Given these novel observations, we have identified dimethyl fumarate (DMF), used to treat psoriasis and showing promising results in clinical trials for multiple sclerosis, as a potential neuroprotectant and HIV disease-modifying agent. DMF, an immune modulator and inducer of the antioxidant response, suppresses HIV replication and neurotoxin release. Two distinct mechanisms are proposed: inhibition of NF-κB nuclear translocation and signaling, which could contribute to the suppression of HIV replication, and induction of HO-1, which is associated with decreased neurotoxin release. Finally, we found that DMF attenuates CCL2-induced monocyte chemotaxis, suggesting that DMF could decrease recruitment of activated monocytes to the CNS in response to inflammatory mediators. We propose that dysregulation of the antioxidant response during HIV infection drives macrophage-mediated neurotoxicity and that DMF could serve as an adjunctive neuroprotectant and HIV disease modifier in ART-treated individuals.


Assuntos
Fármacos Anti-HIV/farmacologia , Antioxidantes/metabolismo , Fumaratos/farmacologia , Imunossupressores/farmacologia , Macrófagos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Replicação Viral/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/imunologia , Animais , Antioxidantes/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/imunologia , Córtex Cerebral/patologia , Fumarato de Dimetilo , HIV-1/efeitos dos fármacos , HIV-1/imunologia , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/patologia , Neurônios/patologia , Neurônios/virologia , Ratos , Ratos Sprague-Dawley , Replicação Viral/imunologia
3.
J Neurochem ; 118(6): 1113-23, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21749375

RESUMO

Excitotoxic neuronal damage via over-activation of the NMDA receptor has been implicated in many neurodegenerative diseases. In vitro modeling of excitotoxic injury has shown that activation of G-protein coupled receptors (GPCRs) counteracts such injury through modulation of neuronal pro-survival pathways and/or NMDA receptor signaling. We have previously demonstrated that the GPCR APJ and its endogenous neuropeptide ligand apelin can protect neurons against excitotoxicity, but the mechanism(s) of this neuroprotection remain incompletely understood. We hypothesized that apelin can promote neuronal survival by activating pro-survival signaling as well as inhibiting NMDA receptor-mediated excitotoxic signaling cascades. Our results demonstrate that (i) apelin activates pro-survival signaling via inositol trisphosphate (IP(3) ), protein kinase C (PKC), mitogen-activated protein kinase kinase 1/2 (MEK1/2), and extracellular signal-regulated kinase-1/2 (ERK1/2) to protect against excitotoxicity, and (ii) apelin inhibits excitotoxic signaling by attenuating NMDA receptor and calpain activity, and by modulating NMDA receptor subunit NR2B phosphorylation at serine 1480. These studies delineate a novel apelinergic signaling pathway that concurrently promotes survival and limits NMDA receptor-mediated injury to protect neurons against excitotoxicity. Defining apelin-mediated neuroprotection advances our understanding of neuroprotective pathways and will potentially improve our ability to develop therapeutics for excitotoxicity-associated neurodegenerative disorders.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Fármacos Neuroprotetores , Neurotoxinas/toxicidade , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Animais , Apelina , Western Blotting , Encéfalo/citologia , Cálcio/metabolismo , Calpaína/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fenômenos Eletrofisiológicos , Infecções por HIV/patologia , Humanos , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Macrófagos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transfecção
4.
J Neuroinflammation ; 8: 15, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21324129

RESUMO

BACKGROUND: Lipopolysaccharide (LPS), the major component of the outer membrane of gram-negative bacteria, can activate immune cells including macrophages. Activation of macrophages in the central nervous system (CNS) contributes to neuronal injury. Bowman-Birk inhibitor (BBI), a soybean-derived protease inhibitor, has anti-inflammatory properties. In this study, we examined whether BBI has the ability to inhibit LPS-mediated macrophage activation, reducing the release of pro-inflammatory cytokines and subsequent neurotoxicity in primary cortical neural cultures. METHODS: Mixed cortical neural cultures from rat were used as target cells for testing neurotoxicity induced by LPS-treated macrophage supernatant. Neuronal survival was measured using a cell-based ELISA method for expression of the neuronal marker MAP-2. Intracellular reactive oxygen species (ROS) production in macrophages was measured via 2', 7'-dichlorofluorescin diacetate (DCFH2DA) oxidation. Cytokine expression was determined by quantitative real-time PCR. RESULTS: LPS treatment of macrophages induced expression of proinflammatory cytokines (IL-1ß, IL-6 and TNF-α) and of ROS. In contrast, BBI pretreatment (1-100 µg/ml) of macrophages significantly inhibited LPS-mediated induction of these cytokines and ROS. Further, supernatant from BBI-pretreated and LPS-activated macrophage cultures was found to be less cytotoxic to neurons than that from non-BBI-pretreated and LPS-activated macrophage cultures. BBI, when directly added to the neuronal cultures (1-100 µg/ml), had no protective effect on neurons with or without LPS-activated macrophage supernatant treatment. In addition, BBI (100 µg/ml) had no effect on N-methyl-D-aspartic acid (NMDA)-mediated neurotoxicity. CONCLUSIONS: These findings demonstrate that BBI, through its anti-inflammatory properties, protects neurons from neurotoxicity mediated by activated macrophages.


Assuntos
Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Inibidor da Tripsina de Soja de Bowman-Birk/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Sistema Nervoso Central/imunologia , Citocinas/imunologia , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Síndromes Neurotóxicas/imunologia , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
5.
J Neurophysiol ; 104(2): 654-64, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20522781

RESUMO

Neuromodulation is well known to provide plasticity in pattern generating circuits, but few details are available concerning modulation of motor pattern coordination. We are using the crustacean stomatogastric nervous system to examine how co-expressed rhythms are modulated to regulate frequency and maintain coordination. The system produces two related motor patterns, the gastric mill rhythm that regulates protraction and retraction of the teeth and the pyloric rhythm that filters food. These rhythms have different frequencies and are controlled by distinct mechanisms, but each circuit influences the rhythm frequency of the other via identified synaptic pathways. A projection neuron, MCN1, activates distinct versions of the rhythms, and we show that hormonal dopamine concentrations modulate the MCN1 elicited rhythm frequencies. Gastric mill circuit interactions with the pyloric circuit lead to changes in pyloric rhythm frequency that depend on gastric mill rhythm phase. Dopamine increases pyloric frequency during the gastric mill rhythm retraction phase. Higher gastric mill rhythm frequencies are associated with higher pyloric rhythm frequencies during retraction. However, dopamine slows the gastric mill rhythm frequency despite the increase in pyloric frequency. Dopamine reduces pyloric circuit influences on the gastric mill rhythm and upregulates activity in a gastric mill neuron, DG. Strengthened DG activity slows the gastric mill rhythm frequency and effectively reduces pyloric circuit influences, thus changing the frequency relationship between the rhythms. Overall dopamine shifts dependence of frequency regulation from intercircuit interactions to increased reliance on intracircuit mechanisms.


Assuntos
Dopaminérgicos/farmacologia , Dopamina/farmacologia , Atividade Motora/fisiologia , Neurônios/efeitos dos fármacos , Periodicidade , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Braquiúros , Gânglios dos Invertebrados/citologia , Masculino , Atividade Motora/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos , Neurônios/fisiologia
7.
Dev Neurobiol ; 74(2): 147-77, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23723176

RESUMO

Fragile X Syndrome (FXS) is considered the most common form of inherited intellectual disability. It is caused by reductions in the expression level or function of a single protein, the Fragile X Mental Retardation Protein (FMRP), a translational regulator which binds to approximately 4% of brain messenger RNAs. Accumulating evidence suggests that FXS is a complex disorder of cognition, involving interactions between genetic and environmental influences, leading to difficulties in acquiring key life skills including motor skills, language, and proper social behaviors. Since many FXS patients also present with one or more features of autism spectrum disorders (ASDs), insights gained from studying the monogenic basis of FXS could pave the way to a greater understanding of underlying features of multigenic ASDs. Here we present an overview of the FXS and FMRP field with the goal of demonstrating how loss of a single protein involved in translational control affects multiple stages of brain development and leads to debilitating consequences on human cognition. We also focus on studies which have rescued or improved FXS symptoms in mice using genetic or therapeutic approaches to reduce protein expression. We end with a brief description of how deficits in translational control are implicated in FXS and certain cases of ASDs, with many recent studies demonstrating that ASDs are likely caused by increases or decreases in the levels of certain key synaptic proteins. The study of FXS and its underlying single genetic cause offers an invaluable opportunity to study how a single gene influences brain development and behavior.


Assuntos
Cognição/fisiologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/fisiopatologia , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiopatologia , Transtornos Globais do Desenvolvimento Infantil/genética , Transtornos Globais do Desenvolvimento Infantil/fisiopatologia , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Humanos , Neurônios/fisiologia
8.
Methods Mol Biol ; 1183: 205-19, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25023311

RESUMO

The cellular and molecular mechanisms that underlie brain function are challenging to study in the living brain. The development of organotypic slices has provided a welcomed addition to our arsenal of experimental brain preparations by allowing both genetic and prolonged pharmacological manipulations in a system that, much like the acute slice preparation, retains several core features of the cellular and network architecture found in situ. Neurons in organotypic slices can survive in culture for several weeks, can be molecularly manipulated by transfection procedures and their function can be interrogated by traditional cellular electrophysiological or imaging techniques. Here, we describe a cost-effective protocol for the preparation and maintenance of organotypic slices and also describe a protocol for biolistic transfection that can be used to introduce plasmids in a small subset of neurons living in an otherwise molecularly unperturbed network. The implementation of these techniques offers a flexible experimental paradigm that can be used to study a multitude of neuronal mechanisms.


Assuntos
Biolística/métodos , Neurônios/metabolismo , Técnicas de Cultura de Órgãos/métodos , Transfecção/métodos , Animais , Biolística/economia , Biolística/instrumentação , Encéfalo/citologia , Encéfalo/metabolismo , Desenho de Equipamento , Camundongos , Neurônios/citologia , Técnicas de Cultura de Órgãos/economia , Técnicas de Cultura de Órgãos/instrumentação , Plasmídeos/administração & dosagem , Plasmídeos/genética , Ratos , Transfecção/economia , Transfecção/instrumentação
9.
Cell Rep ; 9(4): 1402-1416, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25456134

RESUMO

Translational control of mRNAs allows for rapid and selective changes in synaptic protein expression that are required for long-lasting plasticity and memory formation in the brain. Fragile X Related Protein 1 (FXR1P) is an RNA-binding protein that controls mRNA translation in nonneuronal cells and colocalizes with translational machinery in neurons. However, its neuronal mRNA targets and role in the brain are unknown. Here, we demonstrate that removal of FXR1P from the forebrain of postnatal mice selectively enhances long-term storage of spatial memories, hippocampal late-phase long-term potentiation (L-LTP), and de novo GluA2 synthesis. Furthermore, FXR1P binds specifically to the 5' UTR of GluA2 mRNA to repress translation and limit the amount of GluA2 that is incorporated at potentiated synapses. This study uncovers a mechanism for regulating long-lasting synaptic plasticity and spatial memory formation and reveals an unexpected divergent role of FXR1P among Fragile X proteins in brain plasticity.


Assuntos
Potenciação de Longa Duração , Memória de Longo Prazo , Biossíntese de Proteínas , Proteínas de Ligação a RNA/metabolismo , Receptores de AMPA/biossíntese , Sinapses/metabolismo , Regiões 5' não Traduzidas/genética , Animais , Comportamento Animal , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
10.
Methods Mol Biol ; 814: 341-52, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22144317

RESUMO

Astrocytes secrete factors that promote neuron survival, synapse formation, and plasticity. Understanding how these factors perform these roles requires a robust in vitro system that can effectively assess the impact of individual glial factors on neuronal properties. A classical approach to studying neuron-glial interactions in vitro uses a system where dissociated embryonic rat neurons are suspended over a feeder layer of rat astrocytes. Here, we describe a useful "sandwich" co-culture system where postnatal mouse hippocampal neurons are grown suspended above a feeder layer of mouse hippocampal astrocytes. We demonstrate that neurons in these cultures remain healthy beyond 3 weeks in vitro and develop more synapses compared to neurons grown without astrocytes. An advantage of this method is that astrocytes and neurons can be prepared separately from postnatal transgenic or knock-out mouse lines allowing one to study, for example, how wild-type neurons develop in the presence of astrocytes from a knock-out mouse line that lacks the expression of a specific astrocyte-secreted factor. We find this culture system to be a convenient and powerful approach to study the contribution of astrocyte-secreted molecules to neuron development.


Assuntos
Astrócitos/citologia , Técnicas de Cultura de Células/métodos , Hipocampo/citologia , Neurogênese/fisiologia , Neurônios/citologia , Animais , Espinhas Dendríticas/fisiologia , Dissecação/métodos , Camundongos , Sinapses/fisiologia
11.
PLoS One ; 6(10): e26120, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22022532

RESUMO

The formation and storage of memories in neuronal networks relies on new protein synthesis, which can occur locally at synapses using translational machinery present in dendrites and at spines. These new proteins support long-lasting changes in synapse strength and size in response to high levels of synaptic activity. To ensure that proteins are made at the appropriate time and location to enable these synaptic changes, messenger RNA (mRNA) translation is tightly controlled by dendritic RNA-binding proteins. Fragile X Related Protein 1 (FXR1P) is an RNA-binding protein with high homology to Fragile X Mental Retardation Protein (FMRP) and is known to repress and activate mRNA translation in non-neuronal cells. However, unlike FMRP, very little is known about the role of FXR1P in the central nervous system. To understand if FXR1P is positioned to regulate local mRNA translation in dendrites and at synapses, we investigated the expression and targeting of FXR1P in developing hippocampal neurons in vivo and in vitro. We found that FXR1P was highly expressed during hippocampal development and co-localized with ribosomes and mRNAs in the dendrite and at a subset of spines in mouse hippocampal neurons. Our data indicate that FXR1P is properly positioned to control local protein synthesis in the dendrite and at synapses in the central nervous system.


Assuntos
Espinhas Dendríticas/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Hipocampo/metabolismo , Ribossomos/metabolismo , Animais , Análise por Conglomerados , Proteína 4 Homóloga a Disks-Large , Feminino , Proteínas de Fluorescência Verde/metabolismo , Guanilato Quinases/metabolismo , Células HEK293 , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/metabolismo , Polirribossomos/metabolismo , Transporte Proteico , Transporte de RNA , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Subunidades Ribossômicas/metabolismo , Extratos de Tecidos
12.
Carcinogenesis ; 27(9): 1909-16, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16632476

RESUMO

Human carcinoembryonic antigen (CEA), a widely used clinical tumor marker, and its close relative, CEACAM6, are often overexpressed in many cancers. This correlation suggests a possible instrumental role in tumorigenesis, which is supported by extensive results obtained with several in vitro systems. The implication that these results could also apply in vivo warrants investigation. Since mice do not possess homologs of the glycophosphatidyl inositol (GPI)-anchored CEACAM family genes CEA, CEACAM6 and CEACAM7, we have constructed transgenic mice harboring a 187 kb portion of the human CEACAM family gene locus contained in a bacterial artificial chromosome (CEABAC) that includes genes coding for CEA, CEACAM6 and CEACAM7. In this study, we treated the CEABAC mice and their wild-type littermates with azoxymethane (AOM) in order to induce colon tumor formation. At 20 weeks post-treatment, the CEABAC transgenics showed more than a 2-fold increase in mean tumor load relative to their wild-type littermates. Cell surface expression of CEA and CEACAM6 increased by 2- and 20-fold, respectively, in colonocytes from the tumors relative to colonocytes from non-AOM treated transgenics and a de-regulated spatial pattern of CEA/CEACAM6 expression was found in 'normal' crypts adjacent to the tumors, thus mimicking closely the situation in human colon tumorigenesis. A modestly increased incidence of beta-catenin mutations also observed in the AOM-induced CEABAC tumors. These results show that expression of the human GPI-anchored CEACAM family genes predisposes mice to acquire and/or retain essential mutations necessary for sporadic colon tumor development.


Assuntos
Azoximetano , Carcinógenos , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Animais , Antígenos CD/genética , Antígeno Carcinoembrionário , Moléculas de Adesão Celular/genética , Proteínas Ligadas por GPI , Predisposição Genética para Doença , Glicosilfosfatidilinositóis/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Mutação , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA