Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Bases de datos
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
Plant Cell Environ ; 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39248611

RESUMEN

Sorghum (Sorghum bicolor (L.) Moench) is a highly nutritional multipurpose millet crop. However, the genetic and molecular regulatory mechanisms governing sorghum grain development and the associated agronomic traits remain unexplored. In this study, we performed a comprehensive transcriptomic analysis of pistils collected 1-2 days before pollination, and developing seeds collected -2, 10, 20 and 30 days after pollination of S. bicolor variety M35-1. Out of 31 337 genes expressed in these stages, 12 804 were differentially expressed in the consecutive stages of seed development. These exhibited 10 dominant expression patterns correlated with the distinct pathways and gene functions. Functional analysis, based on the pathway mapping, transcription factor enrichment and orthology, delineated the key patterns associated with pollination, fertilization, early seed development, grain filling and seed maturation. Furthermore, colocalization with previously reported quantitative trait loci (QTLs) for grain weight/size revealed 48 differentially expressed genes mapping to these QTL regions. Comprehensive literature mining integrated with QTL mapping and expression data shortlisted 25, 17 and 8 core candidates for engineering grain size, starch and protein content, respectively.

2.
Brain ; 139(Pt 7): 2063-81, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27190010

RESUMEN

Identifying preventive targets for Alzheimer's disease is a central challenge of modern medicine. Non-steroidal anti-inflammatory drugs, which inhibit the cyclooxygenase enzymes COX-1 and COX-2, reduce the risk of developing Alzheimer's disease in normal ageing populations. This preventive effect coincides with an extended preclinical phase that spans years to decades before onset of cognitive decline. In the brain, COX-2 is induced in neurons in response to excitatory synaptic activity and in glial cells in response to inflammation. To identify mechanisms underlying prevention of cognitive decline by anti-inflammatory drugs, we first identified an early object memory deficit in APPSwe-PS1ΔE9 mice that preceded previously identified spatial memory deficits in this model. We modelled prevention of this memory deficit with ibuprofen, and found that ibuprofen prevented memory impairment without producing any measurable changes in amyloid-ß accumulation or glial inflammation. Instead, ibuprofen modulated hippocampal gene expression in pathways involved in neuronal plasticity and increased levels of norepinephrine and dopamine. The gene most highly downregulated by ibuprofen was neuronal tryptophan 2,3-dioxygenase (Tdo2), which encodes an enzyme that metabolizes tryptophan to kynurenine. TDO2 expression was increased by neuronal COX-2 activity, and overexpression of hippocampal TDO2 produced behavioural deficits. Moreover, pharmacological TDO2 inhibition prevented behavioural deficits in APPSwe-PS1ΔE9 mice. Taken together, these data demonstrate broad effects of cyclooxygenase inhibition on multiple neuronal pathways that counteract the neurotoxic effects of early accumulating amyloid-ß oligomers.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Trastornos de la Memoria/prevención & control , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Inhibidores de la Ciclooxigenasa , Modelos Animales de Enfermedad , Regulación hacia Abajo , Electroencefalografía , Ibuprofeno , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Reconocimiento en Psicología/efectos de los fármacos , Triptófano Oxigenasa/efectos de los fármacos
3.
J Neurosci ; 34(17): 5882-94, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24760848

RESUMEN

A persistent and nonresolving inflammatory response to accumulating Aß peptide species is a cardinal feature in the development of Alzheimer's disease (AD). In response to accumulating Aß peptide species, microglia, the innate immune cells of the brain, generate a toxic inflammatory response that accelerates synaptic and neuronal injury. Many proinflammatory signaling pathways are linked to progression of neurodegeneration. However, endogenous anti-inflammatory pathways capable of suppressing Aß-induced inflammation represent a relatively unexplored area. Here we report that signaling through the prostaglandin-E2 (PGE2) EP4 receptor potently suppresses microglial inflammatory responses to Aß42 peptides. In cultured microglial cells, EP4 stimulation attenuated levels of Aß42-induced inflammatory factors and potentiated phagocytosis of Aß42. Microarray analysis demonstrated that EP4 stimulation broadly opposed Aß42-driven gene expression changes in microglia, with enrichment for targets of IRF1, IRF7, and NF-κB transcription factors. In vivo, conditional deletion of microglial EP4 in APPSwe-PS1ΔE9 (APP-PS1) mice conversely increased inflammatory gene expression, oxidative protein modification, and Aß deposition in brain at early stages of pathology, but not at later stages, suggesting an early anti-inflammatory function of microglial EP4 signaling in the APP-PS1 model. Finally, EP4 receptor levels decreased significantly in human cortex with progression from normal to AD states, suggesting that early loss of this beneficial signaling system in preclinical AD development may contribute to subsequent progression of pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacología , Inflamación/metabolismo , Microglía/metabolismo , Fragmentos de Péptidos/farmacología , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal/fisiología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Humanos , Inflamación/patología , Éteres Metílicos/farmacología , Microglía/efectos de los fármacos , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Transducción de Señal/efectos de los fármacos
4.
Ann Neurol ; 72(5): 788-98, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22915243

RESUMEN

OBJECTIVE: There is significant evidence for a central role of inflammation in the development of Alzheimer disease (AD). Epidemiological studies indicate that chronic use of nonsteroidal anti-inflammatory drugs (NSAIDs) reduces the risk of developing AD in healthy aging populations. As NSAIDs inhibit the enzymatic activity of the inflammatory cyclooxygenases COX-1 and COX-2, these findings suggest that downstream prostaglandin signaling pathways function in the preclinical development of AD. Here, we investigate the function of prostaglandin E(2) (PGE(2) ) signaling through its EP3 receptor in the neuroinflammatory response to Aß peptide. METHODS: The function of PGE(2) signaling through its EP3 receptor was examined in vivo in a model of subacute neuroinflammation induced by administration of Aß(42) peptides. Our findings were then confirmed in young adult APPSwe-PS1ΔE9 transgenic mice. RESULTS: Deletion of the PGE(2) EP3 receptor in a model of Aß(42) peptide-induced neuroinflammation reduced proinflammatory gene expression, cytokine production, and oxidative stress. In the APPSwe-PS1ΔE9 model of familial AD, deletion of the EP3 receptor blocked induction of proinflammatory gene and protein expression and lipid peroxidation. In addition, levels of Aß peptides were significantly decreased, as were ß-secretase and ß C-terminal fragment levels, suggesting that generation of Aß peptides may be increased as a result of proinflammatory EP3 signaling. Finally, deletion of EP3 receptor significantly reversed the decline in presynaptic proteins seen in APPSwe-PS1ΔE9 mice. INTERPRETATION: Our findings identify the PGE(2) EP3 receptor as a novel proinflammatory, proamyloidogenic, and synaptotoxic signaling pathway, and suggest a role for COX-PGE(2) -EP3 signaling in the development of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Dinoprostona/metabolismo , Encefalitis/metabolismo , Regulación de la Expresión Génica/genética , Transducción de Señal/fisiología , Factores de Edad , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Animales Recién Nacidos , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Proteínas de Unión al Calcio , Células Cultivadas , Disfunción Cognitiva/patología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Fragmentos de Péptidos/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , ARN Mensajero/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E/deficiencia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA