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










Base de datos
Intervalo de año de publicación
1.
J Healthc Eng ; 2021: 9937904, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804462

RESUMEN

With the advancement in imaging technology, many commercial systems have been developed for performing motion analysis in mice. However, available commercial systems are expensive and use proprietary software. In this paper, we describe a low-cost, camera-based design of an autonomous gait acquisition and analysis system for inspecting gait deficits in C57BL/6 mice. Our system includes video acquisition, autonomous gait-event detection, gait-parameter extraction, and result visualization. We provide a simple, user-friendly, step-by-step detailed methodology to apply well-known image processing techniques for detecting mice footfalls and calculating various gait parameters for analyzing gait abnormalities in healthy and neurotraumatic mice. The system was used in a live animal study for assessing recovery in a mouse model of Parkinson's disease. Using the videos acquired in the study, we validate the performance of our system with receiver operating characteristic (ROC) and Hit : Miss : False (H : M : F) detection analyses. Our system correctly detected the mice footfalls with an average H : M : F score of 92.1 : 2.3 : 5.6. The values for the area under an ROC curve for all the ROC plots are above 0.95, which indicates an almost perfect detection model. The ROC and H : M : F analyses show that our system produces accurate gait detection. The results observed from the gait assessment study are in agreement with the known literature. This demonstrates the practical viability of our system as a gait analysis tool.


Asunto(s)
Análisis de la Marcha , Marcha , Animales , Análisis de la Marcha/métodos , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Endogámicos C57BL , Programas Informáticos
2.
Toxicol Sci ; 177(2): 506-520, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32692843

RESUMEN

Chronic exposure to manganese (Mn) is associated with neuroinflammation and extrapyramidal motor deficits resembling features of Parkinson's disease. Activation of astrocytes and microglia is implicated in neuronal injury from Mn but it is not known whether early life exposure to Mn may predispose glia to more severe inflammatory responses during aging. We therefore examined astrocyte nuclear factor kappa B (NF-κB) signaling in mediating innate immune inflammatory responses during multiple neurotoxic exposures spanning juvenile development into adulthood. MnCl2 was given in drinking water for 30-day postweaning to both wildtype mice and astrocyte-specific knockout (KO) mice lacking I kappa B kinase 2, the central upstream activator of NF-κB. Following juvenile exposure to Mn, mice were subsequently administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) at 4 months of age. Animals were evaluated for behavioral alterations and brain tissue was analyzed for catecholamine neurotransmitters. Stereological analysis of neuronal and glial cell counts from multiple brain regions indicated that juvenile exposure to Mn amplified glial activation and neuronal loss from MPTP exposure in the caudate-putamen and globus pallidus, as well as increased the severity of neurobehavioral deficits in open field activity assays. These alterations were prevented in astrocyte-specific I kappa B kinase 2 KO mice. Juvenile exposure to Mn increased the number of neurotoxic A1 astrocytes expressing C3 as well as the number of activated microglia in adult mice following MPTP challenge, both of which were inhibited in KO mice. These results demonstrate that exposure to Mn during juvenile development heightens the innate immune inflammatory response in glia during a subsequent neurotoxic challenge through NF-κB signaling in astrocytes.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Astrocitos , Encefalitis , Animales , Astrocitos/efectos de los fármacos , Manganeso/toxicidad , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo
3.
Neurobiol Dis ; 127: 193-209, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30818064

RESUMEN

Neuroinflammatory activation of glia is considered a pathological hallmark of Parkinson's disease (PD) and is seen in both human PD patients and in animal models of PD; however, the relative contributions of these cell types, especially astrocytes, to the progression of disease is not fully understood. The transcription factor, nuclear factor kappa B (NFκB), is an important regulator of inflammatory gene expression in glia and is activated by multiple cellular stress signals through the kinase complex, IKK2. We sought to determine the role of NFκB in modulating inflammatory activation of astrocytes in a model of PD by generating a conditional knockout mouse (hGfapcre/Ikbk2F/F) in which IKK2 is specifically deleted in astrocytes. Measurements of IKK2 revealed a 70% deletion rate of IKK2 within astrocytes, as compared to littermate controls (Ikbk2F/F). Use of this mouse in a subacute, progressive model of PD through exposure to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and probenecid (MPTPp) revealed significant protection in exposed mice to direct and progressive loss of dopaminergic neurons in the substantia nigra (SN). hGfapcre/Ikbk2F/F mice were also protected against MPTPp-induced loss in motor activity, loss of striatal proteins, and genomic alterations in nigral NFκB gene expression, but were not protected from loss of striatal catecholamines. Neuroprotection in hGfapcre/Ikbk2F/F mice was associated with inhibition of MPTPp-induced astrocytic expression of inflammatory genes and protection against nitrosative stress and apoptosis in neurons. These data indicate that deletion of IKK2 within astrocytes is neuroprotective in the MPTPp model of PD and suggests that reactive astrocytes directly contribute the potentiation of dopaminergic pathology.


Asunto(s)
Astrocitos/metabolismo , Neuronas Dopaminérgicas/metabolismo , Quinasa I-kappa B/metabolismo , Intoxicación por MPTP/metabolismo , FN-kappa B/metabolismo , Animales , Muerte Celular/fisiología , Neuronas Dopaminérgicas/patología , Quinasa I-kappa B/genética , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Noqueados , FN-kappa B/genética , Probenecid , Sustancia Negra/metabolismo , Sustancia Negra/patología
4.
Mol Pharmacol ; 94(4): 1174-1186, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30111648

RESUMEN

Inflammatory activation of glial cells promotes loss of dopaminergic neurons in Parkinson disease. The transcription factor nuclear factor κB (NF-κB) regulates the expression of multiple neuroinflammatory cytokines and chemokines in activated glial cells that are damaging to neurons. Thus, inhibition of NF-κB signaling in glial cells could be a promising therapeutic strategy for the prevention of neuroinflammatory injury. Nuclear orphan receptors in the NR4A family, including NR4A1 (Nur77) and NR4A2 (Nurr1), can inhibit the inflammatory effects of NF-κB, but no approved drugs target these receptors. Therefore, we postulated that a recently developed NR4A receptor ligand, 1,1bis (3'indolyl) 1(pmethoxyphenyl) methane (C-DIM5), would suppress NF-κB-dependent inflammatory gene expression in astrocytes after treatment with 1-methyl-4-phenyl 1, 2, 3, 6-tetrahydropyridine (MPTP) and the inflammatory cytokines interferon γ and tumor necrosis factor α C-DIM5 increased expression of Nur77 mRNA and suppressed expression of multiple neuroinflammatory genes. C-DIM5 also inhibited the expression of NFκB-regulated inflammatory and apoptotic genes in quantitative polymerase chain reaction array studies and effected p65 binding to unique genes in chromatin immunoprecipitation next-generation sequencing experiments but did not prevent p65 translocation to the nucleus, suggesting a nuclear-specific mechanism. C-DIM5 prevented nuclear export of Nur77 in astrocytes induced by MPTP treatment and simultaneously recruited Nurr1 to the nucleus, consistent with known transrepressive properties of this receptor. Combined RNAi knockdown of Nur77 and Nurr1 inhibited the anti-inflammatory activity of C-DIM5, demonstrating that C-DIM5 requires these receptors to inhibit NF-κB. Collectively, these data demonstrate that NR4A1/Nur77 and NR4A2/Nurr1 dynamically regulated inflammatory gene expression in glia by modulating the transcriptional activity of NF-κB.


Asunto(s)
Astrocitos/fisiología , Inflamación/genética , FN-kappa B/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Transducción de Señal/genética , Animales , Apoptosis/genética , Núcleo Celular/genética , Citocinas/genética , Neuronas Dopaminérgicas/fisiología , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/genética , Neuroglía/fisiología , Transcripción Genética/genética , Factor de Necrosis Tumoral alfa/genética
5.
J Pharmacol Exp Ther ; 365(3): 636-651, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29626009

RESUMEN

The orphan nuclear receptor Nurr1 (also called nuclear receptor-4A2) regulates inflammatory gene expression in glial cells, as well as genes associated with homeostatic and trophic function in dopaminergic neurons. Despite these known functions of Nurr1, an endogenous ligand has not been discovered. We postulated that the activation of Nurr1 would suppress the activation of glia and thereby protect against loss of dopamine (DA) neurons after subacute lesioning with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our previous studies have shown that a synthetic Nurr1 ligand, 1,1-bis(3'-indolyl)-1-(p-chlorophenyl)methane (C-DIM12), suppresses inflammatory gene expression in primary astrocytes and induces a dopaminergic phenotype in neurons. Pharmacokinetic analysis of C-DIM12 in mice by liquid chromatography-mass spectrometry demonstrated that approximately three times more compound concentrated in the brain than in plasma. Mice treated with four doses of MPTP + probenecid over 14 days were monitored for neurobehavioral function, loss of dopaminergic neurons, and glial activation. C-DIM12 protected against the loss of DA neurons in the substantia nigra pars compacta and DA terminals in the striatum, maintained a ramified phenotype in microglia, and suppressed activation of astrocytes. In vitro reporter assays demonstrated that C-DIM12 was an effective activator of Nurr1 transcription in neuronal cell lines. Computational modeling of C-DIM12 binding to the three-dimensional structure of human Nurr1 identified a high-affinity binding interaction with Nurr1 at the coactivator domain. Taken together, these data suggest that C-DIM12 is an activator of Nurr1 that suppresses glial activation and neuronal loss in vivo after treatment with MPTP, and that this receptor could be an efficacious target for disease modification in individuals with Parkinson's disease and related disorders.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Indoles/metabolismo , Indoles/farmacología , Neuroglía/efectos de los fármacos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Recuento de Células , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Regulación de la Expresión Génica/efectos de los fármacos , Indoles/farmacocinética , Indoles/uso terapéutico , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Neuroglía/patología , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Fenotipo , Transducción de Señal/efectos de los fármacos , Distribución Tisular
6.
PLoS One ; 12(12): e0188830, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29244806

RESUMEN

The non-pathogenic parvovirus, adeno-associated virus (AAV), is an efficient vector for transgene expression in vivo and shows promise for treatment of brain disorders in clinical trials. Currently, there are more than 100 AAV serotypes identified that differ in the binding capacity of capsid proteins to specific cell surface receptors that can transduce different cell types and brain regions in the CNS. In the current study, multiple AAV serotypes expressing a GFP reporter (AAV1, AAV2/1, AAVDJ, AAV8, AAVDJ8, AAV9, AAVDJ9) were screened for their infectivity in both primary murine astrocyte and neuronal cell cultures. AAV2/1, AAVDJ8 and AAV9 were selected for further investigation of their tropism throughout different brain regions and cell types. Each AAV was administered to P0-neonatal mice via intracerebroventricular injections (ICV). Brains were then systematically analyzed for GFP expression at 3 or 6 weeks post-infection in various regions, including the olfactory bulb, striatum, cortex, hippocampus, substantia nigra (SN) and cerebellum. Cell counting data revealed that AAV2/1 infections were more prevalent in the cortical layers but penetrated to the midbrain less than AAVDJ8 and AAV9. Additionally, there were differences in the persistence of viral transgene expression amongst the three serotypes examined in vivo at 3 and 6 weeks post-infection. Because AAV-mediated transgene expression is of interest in neurodegenerative diseases such as Parkinson's Disease, we examined the SN with microscopy techniques, such as CLARITY tissue transmutation, to identify AAV serotypes that resulted in optimal transgene expression in either astrocytes or dopaminergic neurons. AAVDJ8 displayed more tropism in astrocytes compared to AAV9 in the SN region. We conclude that ICV injection results in lasting expression of virally encoded transgene when using AAV vectors and that specific AAV serotypes are required to selectively deliver transgenes of interest to different brain regions in both astrocytes and neurons.


Asunto(s)
Astrocitos/metabolismo , Dependovirus/genética , Terapia Genética/métodos , Neuronas/metabolismo , Sustancia Negra/metabolismo , Transgenes , Animales , Animales Recién Nacidos , Astrocitos/citología , Cerebelo/citología , Cerebelo/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Dependovirus/clasificación , Dependovirus/metabolismo , Expresión Génica , Genes Reporteros , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Hipocampo/metabolismo , Inyecciones Intraventriculares , Ratones , Neuronas/citología , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Imagen Óptica , Especificidad de Órganos , Cultivo Primario de Células , Serogrupo , Sustancia Negra/citología , Transducción Genética
7.
J Virol ; 90(12): 5785-96, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27053560

RESUMEN

UNLABELLED: Venezuelan and western equine encephalitis viruses (VEEV and WEEV; Alphavirus; Togaviridae) are mosquito-borne pathogens causing central nervous system (CNS) disease in humans and equids. Adult CD-1 mice also develop CNS disease after infection with VEEV and WEEV. Adult CD-1 mice infected by the intranasal (i.n.) route, showed that VEEV and WEEV enter the brain through olfactory sensory neurons (OSNs). In this study, we injected the mouse footpad with recombinant WEEV (McMillan) or VEEV (subtype IC strain 3908) expressing firefly luciferase (fLUC) to simulate mosquito infection and examined alphavirus entry in the CNS. Luciferase expression served as a marker of infection detected as bioluminescence (BLM) by in vivo and ex vivo imaging. BLM imaging detected WEEV and VEEV at 12 h postinoculation (hpi) at the injection site (footpad) and as early as 72 hpi in the brain. BLM from WEEV.McM-fLUC and VEEV.3908-fLUC injections was initially detected in the brain's circumventricular organs (CVOs). No BLM activity was detected in the olfactory neuroepithelium or OSNs. Mice were also injected in the footpad with WEEV.McM expressing DsRed (Discosoma sp.) and imaged by confocal fluorescence microscopy. DsRed imaging supported our BLM findings by detecting WEEV in the CVOs prior to spreading along the neuronal axis to other brain regions. Taken together, these findings support our hypothesis that peripherally injected alphaviruses enter the CNS by hematogenous seeding of the CVOs followed by centripetal spread along the neuronal axis. IMPORTANCE: VEEV and WEEV are mosquito-borne viruses causing sporadic epidemics in the Americas. Both viruses are associated with CNS disease in horses, humans, and mouse infection models. In this study, we injected VEEV or WEEV, engineered to express bioluminescent or fluorescent reporters (fLUC and DsRed, respectively), into the footpads of outbred CD-1 mice to simulate transmission by a mosquito. Reporter expression serves as detectable bioluminescent and fluorescent markers of VEEV and WEEV replication and infection. Bioluminescence imaging, histological examination, and confocal fluorescence microscopy were used to identify early entry sites of these alphaviruses in the CNS. We observed that specific areas of the brain (circumventricular organs [CVOs]) consistently showed the earliest signs of infection with VEEV and WEEV. Histological examination supported VEEV and WEEV entering the brain of mice at specific sites where the blood-brain barrier is naturally absent.


Asunto(s)
Barrera Hematoencefálica/virología , Encéfalo/virología , Virus de la Encefalitis Equina Venezolana/fisiología , Virus de la Encefalitis Equina del Oeste/fisiología , Encefalomielitis Equina Venezolana/virología , Internalización del Virus , Adulto , Animales , Barrera Hematoencefálica/fisiopatología , Encéfalo/patología , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/crecimiento & desarrollo , Virus de la Encefalitis Equina del Oeste/genética , Virus de la Encefalitis Equina del Oeste/crecimiento & desarrollo , Humanos , Luciferasas , Mediciones Luminiscentes , Ratones , Neuronas Receptoras Olfatorias/virología , Imagen Óptica/métodos , Carga Viral
8.
Neurosci Lett ; 607: 83-89, 2015 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-26383113

RESUMEN

Degeneration of dopaminergic neurons in Parkinson's disease (PD) is associated with decreased expression of the orphan nuclear receptor Nurr1 (NR4A2), which is critical for both homeostasis and development of dopamine (DA) neurons. The synthetic, phytochemical-based compound, 1,1-bis (3'-indolyl)-1-(p-chlorophenyl) methane (C-DIM12) activates Nurr1 in cancer cells and prevents loss of dopaminergic neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD in mice. In the present study, we examined the capacity of C-DIM12 to induce expression of Nurr1-regulated genes in two dopaminergic neuronal cell lines (N2A, N27) and to protect against 6-hydroxydopamine (6-OHDA) neurotoxicity. C-DIM12 induced expression of Nurr1-regulated genes that was abolished by Nurr1 knockdown. C-DIM12 increased expression of transfected human Nurr1, induced Nurr1 protein expression in primary dopaminergic neurons and enhanced neuronal survival from exposure to 6-OHDA. These data indicate that C-DIM12 stimulates neuroprotective expression Nurr1-regulated genes in DA neurons.


Asunto(s)
Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Indoles/farmacología , Fármacos Neuroprotectores/farmacología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/agonistas , Oxidopamina/toxicidad , Animales , Diferenciación Celular , Línea Celular , Supervivencia Celular/efectos de los fármacos , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Expresión Génica , Humanos , Ratones , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Cultivo Primario de Células
9.
Am J Physiol Regul Integr Comp Physiol ; 309(2): R138-47, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25994957

RESUMEN

We reported that brain (pro)renin receptor (PRR) expression levels are elevated in DOCA-salt-induced hypertension; however, the underlying mechanism remained unknown. To address whether ANG II type 1 receptor (AT1R) signaling is involved in this regulation, we implanted a DOCA pellet and supplied 0.9% saline as the drinking solution to C57BL/6J mice. Sham pellet-implanted mice that were provided regular drinking water served as controls. Concurrently, mice were intracerebroventricularly infused with the AT1R blocker losartan, angiotensin-converting-enzyme inhibitor captopril, or artificial cerebrospinal fluid for 3 wk. Intracerebroventricular infusion of losartan or captopril attenuated DOCA-salt-induced PRR mRNA elevation in the paraventricular nucleus of the hypothalamus, suggesting a role for ANG II/AT1R signaling in regulating PRR expression during DOCA-salt hypertension. To test which ANG II/AT1R downstream transcription factors were involved in PRR regulation, we treated Neuro-2A cells with ANG II with or without CREB (cAMP response element-binding protein) or AP-1 (activator protein-1) inhibitors, or CREB siRNA. CREB and AP-1 inhibitors, as well as CREB knockdown abolished ANG II-induced increases in PRR levels. ANG II also induced PRR upregulation in primary cultured neurons. Chromatin immunoprecipitation assays revealed that ANG II treatment increased CREB binding to the endogenous PRR promoter in both cultured neurons and hypothalamic tissues of DOCA-salt hypertensive mice. This increase in CREB activity was reversed by AT1R blockade. Collectively, these findings indicate that ANG II acts via AT1R to upregulate PRR expression both in cultured cells and in DOCA-salt hypertensive mice by increasing CREB binding to the PRR promoter.


Asunto(s)
Angiotensina II/metabolismo , Proteína de Unión a CREB/metabolismo , Hipertensión/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , ATPasas de Translocación de Protón/metabolismo , Receptores de Superficie Celular/metabolismo , Sistema Renina-Angiotensina , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Animales , Sitios de Unión , Proteína de Unión a CREB/genética , Línea Celular , Acetato de Desoxicorticosterona , Modelos Animales de Enfermedad , Hipertensión/inducido químicamente , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Infusiones Intraventriculares , Ratones Endogámicos C57BL , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Fosforilación , Regiones Promotoras Genéticas , ATPasas de Translocación de Protón/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Superficie Celular/genética , Sistema Renina-Angiotensina/efectos de los fármacos , Transducción de Señal , Cloruro de Sodio , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo , Transfección , Regulación hacia Arriba , Receptor de Prorenina
10.
Mol Pharmacol ; 87(6): 1021-34, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25858541

RESUMEN

NR4A family orphan nuclear receptors are an important class of transcription factors for development and homeostasis of dopaminergic neurons that also inhibit expression of inflammatory genes in glial cells. The identification of NR4A2 (Nurr1) as a suppressor of nuclear factor κB (NF-κB)-related neuroinflammatory genes in microglia and astrocytes suggests that this receptor could be a target for pharmacologic intervention in neurologic disease, but compounds that promote this activity are lacking. Selected diindolylmethane compounds (C-DIMs) have been shown to activate or inactivate nuclear receptors, including Nurr1, in cancer cells and also suppress astrocyte inflammatory signaling in vitro. Based upon these data, we postulated that C-DIM12 [1,1-bis(3'-indolyl)-1-(p-chlorophenyl) methane] would suppress inflammatory signaling in microglia by a Nurr1-dependent mechanism. C-DIM12 inhibited lipopolysaccharide (LPS)-induced expression of NF-κB-regulated genes in BV-2 microglia including nitric oxide synthase (NOS2), interleukin-6 (IL-6), and chemokine (C-C motif) ligand 2 (CCL2), and the effects were attenuated by Nurr1-RNA interference. Additionally, C-DIM12 decreased NF-κB activation in NF-κB-GFP (green fluorescent protein) reporter cells and enhanced nuclear translocation of Nurr1 primary microglia. Chromatin immunoprecipitation assays indicated that C-DIM12 decreased lipopolysaccharide-induced p65 binding to the NOS2 promoter and concurrently enhanced binding of Nurr1 to the p65-binding site. Consistent with these findings, C-DIM12 also stabilized binding of the Corepressor for Repressor Element 1 Silencing Transcription Factor (CoREST) and the Nuclear Receptor Corepressor 2 (NCOR2). Collectively, these data identify C-DIM12 as a modulator of Nurr1 activity that results in inhibition of NF-κB-dependent gene expression in glial cells by stabilizing nuclear corepressor proteins, which reduces binding of p65 to inflammatory gene promoters.


Asunto(s)
Indoles/farmacología , Microglía/efectos de los fármacos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Línea Celular , Proteínas Co-Represoras , Citocinas/genética , Citocinas/metabolismo , Expresión Génica , Humanos , Lipopolisacáridos/farmacología , Ratones , Microglía/inmunología , Microglía/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Regiones Promotoras Genéticas , Transporte de Proteínas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción ReIA/metabolismo , Transcripción Genética
11.
Toxicol Sci ; 143(2): 360-73, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25406165

RESUMEN

The orphan nuclear receptor NR4A2 (Nurr1) constitutively regulates inflammatory gene expression in glial cells by suppressing DNA binding activity of NF-κB. We recently reported that novel 1,1-bis(3'-indolyl)-1-(p-substitutedphenyl)methane (C-DIM) compounds that activate NR4A family nuclear receptors in cancer lines also suppress inflammatory gene expression in primary astrocytes and prevent loss of dopaminergic neurons in mice exposed to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and probenecid (MPTPp). In this study, we postulated that the basis for this neuroprotection involves blockade of glial activation and subsequent expression of NF-κB-regulated inflammatory genes. To examine this mechanism, we treated transgenic NF-κB/EGFP reporter mice with MPTPp for 7 days (MPTPp7d) followed by daily oral gavage with either vehicle (corn oil; MPTPp14d) or C-DIMs containing p-methoxyphenyl (C-DIM5), p-hydroxyphenyl (C-DIM8), or p-chlorophenyl (C-DIM12) groups. Each compound conferred significant protection against progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc), even when given after 7 days of dosing with MPTPp. C-DIM12 had the greatest neuroprotective activity in MPTPp-treated mice, and was also the most potent compound in suppressing activation of microglia and astrocytes, expression of cytokines and chemokines in quantitative polymerase chain reaction (qPCR) array studies, and in reducing expression of NF-κB/EGFP in the SN. C-DIM12 prevented nuclear export of Nurr1 in dopaminergic neurons and enhanced expression of the Nurr1-regulated proteins tyrosine hydroxylase and the dopamine transporter. These data indicate that NR4A-active C-DIM compounds protect against loss of dopamine neurons in the MPTPp model of PD by preventing glial-mediated neuronal injury and by supporting a dopaminergic phenotype in TH-positive neurons in the SNpc.


Asunto(s)
Anisoles/farmacología , Antiparkinsonianos/farmacología , Indoles/farmacología , Intoxicación por MPTP/prevención & control , Neuroglía/efectos de los fármacos , Enfermedad de Parkinson/prevención & control , Fenoles/farmacología , Animales , Anisoles/química , Anisoles/uso terapéutico , Antiparkinsonianos/química , Antiparkinsonianos/uso terapéutico , Conducta Animal/efectos de los fármacos , Recuento de Células , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Proteínas Fluorescentes Verdes/genética , Indoles/química , Indoles/uso terapéutico , Intoxicación por MPTP/metabolismo , Intoxicación por MPTP/patología , Ratones Transgénicos , FN-kappa B/genética , Neuroglía/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Fenoles/química , Fenoles/uso terapéutico
12.
J Cell Sci ; 127(Pt 6): 1179-89, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24434582

RESUMEN

It is unknown how cues from the tumor microenvironment can regulate post-transcriptional mechanisms, such as alternative splicing, that control genes that drive malignant growth. The induction of cyclooxygenase 2 (Cox-2) by integrin α3ß1 in breast cancer cells can promote tumor progression. We have used RNAi to suppress α3ß1 in human MDA-MB-231 breast cancer cells and then investigated changes in global gene expression. Numerous mRNAs, including Cox-2, show altered expression and/or alternative exon usage (AEU) in α3ß1-deficient cells. AEU included patterns predicted to render an mRNA susceptible to degradation, such as 3'-UTR variations or retention of elements that target an mRNA for nonsense-mediated decay (NMD). PCR-based analysis of α3ß1-deficient cells confirmed changes in Cox-2 mRNA that might target it for NMD, including retention of an intron that harbors premature termination codons and changes within the 3'-UTR. Moreover, Cox-2 mRNA has reduced stability in α3ß1-deficient cells, which is partially reversed by knockdown of the essential NMD factor UPF1. Our study identifies α3ß1-mediated AEU as a novel paradigm of integrin-dependent gene regulation that has potential for exploitation as a therapeutic target.


Asunto(s)
Ciclooxigenasa 2/genética , Integrina alfa3beta1/fisiología , Degradación de ARNm Mediada por Codón sin Sentido , ARN Mensajero/genética , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Neoplasias de la Mama , Línea Celular Tumoral , Ciclooxigenasa 2/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Intrones , Laminina/metabolismo , Datos de Secuencia Molecular , Unión Proteica , ARN Helicasas , ARN Mensajero/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA