Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Chem Res Toxicol ; 37(7): 1086-1103, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38912648

RESUMEN

With the rapid expansion in the development and clinical utility of immune checkpoint inhibitors (ICIs) for oncology, the continual evaluation of the safety profile of such agents is imperative. The safety profile of ICIs as monotherapy is dominated by immune-related adverse events, which can be considered as an extension of the mechanism of action of these immunomodulatory drugs. Further to this, an emerging theme is that ICI treatment can significantly impact upon the tolerability of coadministered medications. Numerous reports in literature indicate that ICIs may alter the immunological perception of coadministered drugs, resulting in undesirable reactions to a variety of concomitant medications. These reactions can be severe in manifestation, including hepatotoxicity and Stevens-Johnson Syndrome (SJS)/toxic epidermal necrolysis (TEN), but may also have detrimental impact on malignancy control. To minimize the impact of such drug-drug interactions on patients, it is imperative to identify medications that may cause these reactions, understand the underlying mechanisms, consider the timing and dosing of comedication, and explore alternative medications with comparable efficacies. Improving our understanding of how concomitant medications affect the safety and efficacy of ICIs can allow for potential culprit drugs to be identified/removed/desensitized. This approach will allow the continuation of ICI therapy that may have been discontinued otherwise, thereby improving malignant control and patient and drug development outcomes.


Asunto(s)
Interacciones Farmacológicas , Inhibidores de Puntos de Control Inmunológico , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
3.
Toxicol Sci ; 186(1): 58-69, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-34850240

RESUMEN

An emerging clinical issue associated with immune-oncology agents is the collateral effects on the tolerability of concomitant medications. One report of this phenomenon was the increased incidence of hypersensitivity reactions observed in patients receiving concurrent immune checkpoint inhibitors (ICIs) and sulfasalazine (SLZ). Thus, the aim of this study was to characterize the T cells involved in the pathogenesis of such reactions, and recapitulate the effects of inhibitory checkpoint blockade on de-novo priming responses to compounds within in vitro platforms. A regulatory competent human dendritic cell/T-cell coculture assay was used to model the effects of ICIs on de novo nitroso sulfamethoxazole- and sulfapyridine (SP) (the sulfonamide component of SLZ) hydroxylamine-specific priming responses. The role of T cells in the pathogenesis of the observed reactions was explored in 3 patients through phenotypic characterization of SP/sulfapyridine hydroxylamine (SPHA)-responsive T-cell clones (TCC), and assessment of cross-reactivity and pathways of T-cell activation. Augmentation of the frequency of responding drug-specific T cells and intensity of the T-cell response was observed with PD-1/PD-L1 blockade. Monoclonal populations of SP- and SPHA-responsive T cells were isolated from all 3 patients. A core secretory effector molecule profile (IFN-γ, IL-13, granzyme B, and perforin) was identified for SP and SPHA-responsive TCC, which proceeded through Pi and hapten mechanisms, respectively. Data presented herein provides evidence that drug-responsive T cells are effectors of hypersensitivity reactions observed in oncology patients administered ICIs and SLZ. Perturbation of drug-specific T-cell priming is a plausible explanation for clinical observations of how an increased incidence of these adverse events is occurring.


Asunto(s)
Hipersensibilidad a las Drogas , Sulfasalazina , Humanos , Incidencia , Activación de Linfocitos , Sulfasalazina/efectos adversos , Sulfonamidas
4.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34049931

RESUMEN

Many adverse reactions associated with immune checkpoint inhibitor (ICI) treatments are immunologically driven and may necessitate discontinuation of the ICI. Herein, we present a patient who had been administered the radio contrast media amidotrizoate multiple times without issue but who then developed a Stevens-Johnson syndrome reaction after coadministration of atezolizumab. Causality was confirmed by a positive re-challenge with amidotrizoate and laboratory investigations that implicated T cells. Importantly, the introduction of atezolizumab appears to have altered the immunologic response to amidotrizoate in terms of the tolerance-elicitation continuum. Proof of concept studies demonstrated enhancement of recall responses to a surrogate antigen panel following in-vitro (healthy donors) and in-vivo (ICI patients) administrations of ICIs. Our findings highlight the importance of considering all concomitant medications in patients on ICIs who develop immune-mediated adverse reactions. In the event of some immune-related adverse reactions, it may be critical to identify the culprit antigen-forming entity that the ICIs have altered the perception of rather than simply attribute causality to the ICI itself in order to optimize both patient safety and treatment of malignancies.


Asunto(s)
Anticuerpos Monoclonales Humanizados/efectos adversos , Carcinoma de Células Renales/tratamiento farmacológico , Medios de Contraste/efectos adversos , Diatrizoato/efectos adversos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias Renales/tratamiento farmacológico , Síndrome de Stevens-Johnson/etiología , Linfocitos T/efectos de los fármacos , Corticoesteroides/uso terapéutico , Carcinoma de Células Renales/diagnóstico por imagen , Carcinoma de Células Renales/inmunología , Humanos , Neoplasias Renales/diagnóstico por imagen , Neoplasias Renales/inmunología , Masculino , Valor Predictivo de las Pruebas , Factores de Riesgo , Síndrome de Stevens-Johnson/diagnóstico , Síndrome de Stevens-Johnson/tratamiento farmacológico , Síndrome de Stevens-Johnson/inmunología , Linfocitos T/inmunología
5.
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
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.
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
8.
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
9.
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
10.
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
11.
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
12.
Porto Alegre; Artmed; 3 ed; 2010. 503 p.
Monografía en Portugués | LILACS, Coleciona SUS | ID: biblio-939172

RESUMEN

Prefácio Como Usar este Livro PARTE 1 - As bases da Pesquisa Capítulo 1. Teoria, Método e Delineamento de Pesquisa Capítulo 2. Questões Práticas e Éticas do Projeto de Pesquisa Capítulo 3. Níveis de Mensuração Capítulo 4. O Método Experimental em Psicologia Capítulo 5. Modelos Quasi-Experimentais Capítulo 6. Levantamento e Amostragem PARTE 2 - Coleta de Dados Capítulo 7. Métodos Observacionais Capítulo 8. Métodos Psicofisiológicos Capítulo 9. Métodos Psicofísicos Capítulo 10. Utilizando Testes Psicométricos Capítulo 11. Delineamento de Questionário Capítulo 12. Métodos de Entrevista Capítulo 13. O Uso do Autorregistro: Métodos de Diário e de Narrativa Capítulo 14. Grupos Focais Capítulo 15. Pesquisa Etnográfica e Pesquisa-Ação PARTE 3 - Tratamento dos Dados Capítulo 16. Análise Fenomenológica Interpretativa Capítulo 17. A Teoria Fundamentada Capítulo 18. Análise do Discurso Capítulo 19. Princípios de Estatística Inferencial Capítulo 20. Introdução à Análise Multivariada de Dados Capítulo 21. Introdução à Modelagem de Equação Estrutural Capítulo 22. Metanálise Referências Índice


Asunto(s)
Masculino , Femenino , Humanos , Psicología , Psicología/métodos , Psicología/normas
13.
Porto Alegre; Artmed; 3 ed; 2010. 503 p.
Monografía en Portugués | LILACS | ID: lil-695599

RESUMEN

Prefácio Como Usar este Livro PARTE 1 - As bases da Pesquisa Capítulo 1. Teoria, Método e Delineamento de Pesquisa Capítulo 2. Questões Práticas e Éticas do Projeto de Pesquisa Capítulo 3. Níveis de Mensuração Capítulo 4. O Método Experimental em Psicologia Capítulo 5. Modelos Quasi-Experimentais Capítulo 6. Levantamento e Amostragem PARTE 2 - Coleta de Dados Capítulo 7. Métodos Observacionais Capítulo 8. Métodos Psicofisiológicos Capítulo 9. Métodos Psicofísicos Capítulo 10. Utilizando Testes Psicométricos Capítulo 11. Delineamento de Questionário Capítulo 12. Métodos de Entrevista Capítulo 13. O Uso do Autorregistro: Métodos de Diário e de Narrativa Capítulo 14. Grupos Focais Capítulo 15. Pesquisa Etnográfica e Pesquisa-Ação PARTE 3 - Tratamento dos Dados Capítulo 16. Análise Fenomenológica Interpretativa Capítulo 17. A Teoria Fundamentada Capítulo 18. Análise do Discurso Capítulo 19. Princípios de Estatística Inferencial Capítulo 20. Introdução à Análise Multivariada de Dados Capítulo 21. Introdução à Modelagem de Equação Estrutural Capítulo 22. Metanálise Referências Índice.


Asunto(s)
Humanos , Masculino , Femenino , Psicología/ética , Psicología/métodos , Psicología/normas
14.
Free Radic Biol Med ; 39(2): 164-81, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15964508

RESUMEN

Excessive brain Mn can produce toxicity with symptoms resembling parkinsonism. This syndrome, called "manganism," correlates with loss of dopamine in the striatum and cell death in the striatum and globus pallidus. A common hypothesis is that cell damage in Mn toxicity is caused by oxidation of important cell components by Mn3+. Determination of the amount of Mn3+ present, under a range of conditions, in neuronal cells and brain mitochondria represents an important step in evaluating the "damage through oxidation by Mn3+ hypothesis." In an earlier paper we used X-ray absorption near-edge structure (XANES) spectroscopy to determine the amount of Mn2+ and Mn3+ in brain mitochondria under a range of conditions. Here we extend the study to investigate the evidence for formation of Mn3+ through oxidation of Mn2+ by ROS in PC12 cells and in PC12 cells induced with nerve growth factor (NGF) to display a phenotype more like that of neurons. Although the results suggest that very small amounts of Mn3+ might be present at low Mn levels, probably in Mn superoxide dismutase, Mn3+ is not stabilized by complex formation in these cells and therefore does not accumulate to detectable amounts.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Animales , Encéfalo/metabolismo , Manganeso/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Oxidantes/metabolismo , Oxígeno/metabolismo , Células PC12 , Ratas , Especies Reactivas de Oxígeno , Espectrometría por Rayos X , Espectrofotometría Atómica/métodos , Superóxido Dismutasa/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA