Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Neurosci Methods ; 164(2): 218-24, 2007 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-17553568

RESUMEN

In vitro culture of rodent microglia is a common system used to model proinflammatory changes in the brain. However, typical postnatal brain isolation protocols are time consuming and cell numbers acquired are often a rate-limiting factor for experimental progress. Large studies that rely on the use of primary microglia can, therefore, require excessive numbers of animals at considerable expense, additional technical support and culture incubator space. Although the addition of mitogens such as macrophage colony-stimulating factor, granulocyte macrophage-colony stimulating factor, and epidermal growth factor to the cultures can facilitate a higher yield, this adds additional expense and likely alters the microglial phenotype. We have defined a simple, inexpensive modification of our standard culture protocol that allows us to repetitively isolate microglia. In order to define a method for improving microglia yield, we utilized our standard mixed glial culture preparation derived from postnatal day 1-3 mouse brains. After isolating microglia from mixed cultures at 14 days in vitro, we added fresh media to the cultures for an additional 7 and 14 days to monitor microglial proliferation. We acquired a constant number of cells at each successive time point although the numbers were reduced from the first isolation. More importantly, in order to determine if our successive microglia isolates differed phenotypically we characterized several parameters of function. We compared their ability to secrete the proinflammatory cytokines interleukin-6 and tumor necrosis factor alpha after LPS stimulation. We also contrasted the phagocytic ability, morphology, and specific immunoreactivity (CD11b, CD68, CD45 and MHC II) of the culture ages. Our data demonstrate that microglia can be obtained from extended-time cultures provided periodic isolation is performed. Moreover, the cells retain a comparable in vitro phenotype. This demonstrates that cells from all ages can be combined for any given study. These findings are a viable and inexpensive way to increase and extend the microglial yield without increasing the number of animals used or adding costly mitogens. This method will be particularly useful for the preparation of microglia cultures from limited transgenic colonies.


Asunto(s)
Separación Celular/métodos , Microglía/fisiología , Fenotipo , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Encéfalo/citología , Recuento de Células/métodos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
2.
J Neurochem ; 97(2): 449-61, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16539666

RESUMEN

Beta amyloid peptide-containing neuritic plaques are a defining feature of Alzheimer's disease pathology. Beta amyloid are 38-43 residue peptides derived by proteolytic cleavage of amyloid precursor protein. Although much attention has focused on the proteolytic events leading to beta amyloid generation, the function of amyloid precursor protein remains poorly described. Previously, we reported that amyloid precursor protein functions as a pro-inflammatory receptor on monocytic lineage cells and defined a role for amyloid precursor protein in adhesion by demonstrating that beta(1) integrin-mediated pro-inflammatory activation of monocytes is amyloid precursor protein dependent. We demonstrated that antibody-induced cross-linking of amyloid precursor protein in human THP-1 monocytes and primary mouse microglia stimulates a tyrosine kinase-based pro-inflammatory signaling response leading to acquisition of a reactive phenotype. Here, we have identified pro-inflammatory mediators released upon amyloid precursor protein-dependent activation of monocytes and microglia. We show that amyloid precursor protein cross-linking stimulated tyrosine kinase-dependent increases in pro-inflammatory cytokine release and a tyrosine kinase-independent increase in beta amyloid 1-42 generation. These data provide much needed insight into the function of amyloid precursor protein and provide potential therapeutic targets to limit inflammatory changes associated with the progression of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/farmacología , Citocinas/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/inmunología , Animales , Anticuerpos/farmacología , Western Blotting/métodos , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Inmunohistoquímica/métodos , L-Lactato Deshidrogenasa/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Octoxinol/farmacología , ARN Interferente Pequeño/farmacología , Tensoactivos/farmacología , Transfección/métodos
3.
Neurochem Int ; 39(5-6): 449-57, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11578780

RESUMEN

Amyloid deposition within the brains of Alzheimer's Disease patients results in the activation of microglial cells and the induction of a local inflammatory response. The interaction of microglia or monocytes with beta-amyloid (A beta) fibrils elicits the activation a complex tyrosine kinase-based signal transduction cascade leading to stimulation of multiple independent signaling pathways and ultimately to changes in proinflammatory gene expression. The A beta-stimulated expression of proinflammatory genes in myeloid lineage cells is antagonized by the action of a family of ligand-activated nuclear hormone receptors, the peroxisome proliferator-activated receptors (PPARs). We report that THP-1 monocytes express predominantly PPAR gamma isoform and lower levels of PPAR alpha and PPAR delta isoforms. PPAR mRNA levels are not affected by differentiation of the cells into a macrophage phenotype, nor are they altered following exposure to the classical immune stimulus, lipopolysaccharide. Previous studies have found that PPAR gamma agonists act broadly to inhibit inflammatory responses. The present study explored the action of the PPAR alpha isoform and found that PPAR alpha agonists inhibited the A beta-stimulated expression of TNFalpha and IL-6 reporter genes in a dose-dependent manner. Moreover, the PPAR alpha agonist WY14643 inhibited macrophage differentiation and COX-2 gene expression. However, the PPAR alpha agonists failed to inhibit A beta-stimulated elaboration of neurotoxic factors by THP-1 cells. These findings demonstrate that PPAR alpha acts to suppress a diverse array of inflammatory responses in monocytes.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Inflamación/etiología , Proteínas de la Membrana , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Lipoproteína , Factores de Transcripción/fisiología , Amiloide/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Ciclooxigenasa 2 , Humanos , Interleucina-6/antagonistas & inhibidores , Isoenzimas/metabolismo , Macrófagos/citología , Microglía/fisiología , Monocitos/citología , Monocitos/fisiología , Neurotoxinas/antagonistas & inhibidores , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Tirosina Quinasas/fisiología , Receptor para Productos Finales de Glicación Avanzada , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Inmunológicos/fisiología , Receptores Depuradores , Receptores Depuradores de Clase B , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Transcripción/agonistas , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
4.
J Neurosci ; 21(4): 1179-88, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11160388

RESUMEN

Reactive microglia associated with the beta-amyloid plaques in Alzheimer's disease (AD) brains initiate a sequence of inflammatory events integral to the disease process. We have observed that fibrillar beta-amyloid peptides activate a tyrosine kinase-based signaling response in primary mouse microglia and the human monocytic cell line, THP-1, resulting in production of neurotoxic secretory products, proinflammatory cytokines, and reactive oxygen species. We report that most of the amyloid-induced tyrosine kinase activity was stimulated after activation of Src family members such as Lyn. However, transduction of the signaling response required for increased production of the cytokines TNFalpha and IL1-beta was mediated by the nonreceptor tyrosine kinase, Syk. Additionally, beta-amyloid stimulated an NFkappaB-dependent pathway in parallel that was required for cytokine production. Importantly, TNFalpha generated by the monocytes and microglia was responsible for the majority of the neuorotoxic activity secreted by these cells after beta-amyloid stimulation but must act in concert with other factors elaborated by microglia to elicit neuronal death. Moreover, we observed that the neuronal loss was apoptotic in nature and involved increased neuronal expression of inducible nitric oxide synthase and subsequent peroxynitrite production. Selective inhibitors of inducible nitric oxide synthase effectively protected cells from toxicity associated with the microglial and monocytic secretory products. This study demonstrates a functional linkage between beta-amyloid-dependent activation of microglia and several characteristic markers of neuronal death occurring in Alzheimer's disease brains.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Microglía/metabolismo , Monocitos/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis , Células Cultivadas , Contraindicaciones , Precursores Enzimáticos/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , FN-kappa B/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/fisiología , Quinasa Syk , Factores de Transcripción/metabolismo , Familia-src Quinasas/metabolismo
5.
J Neurosci ; 20(2): 558-67, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10632585

RESUMEN

Alzheimer's disease (AD) is characterized by the extracellular deposition of beta-amyloid fibrils within the brain and the subsequent association and phenotypic activation of microglial cells associated with the amyloid plaque. The activated microglia mount a complex local proinflammatory response with the secretion of a diverse range of inflammatory products. Nonsteroidal anti-inflammatory drugs (NSAIDs) are efficacious in reducing the incidence and risk of AD and significantly delaying disease progression. A recently appreciated target of NSAIDs is the ligand-activated nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma). PPARgamma is a DNA-binding transcription factor whose transcriptional regulatory actions are activated after agonist binding. We report that NSAIDs, drugs of the thiazolidinedione class, and the natural ligand prostaglandin J2 act as agonists for PPARgamma and inhibit the beta-amyloid-stimulated secretion of proinflammatory products by microglia and monocytes responsible for neurotoxicity and astrocyte activation. The activation of PPARgamma also arrested the differentiation of monocytes into activated macrophages. PPARgamma agonists were shown to inhibit the beta-amyloid-stimulated expression of the cytokine genes interleukin-6 and tumor necrosis factor alpha. Furthermore, PPARgamma agonists inhibited the expression of cyclooxygenase-2. These data provide direct evidence that PPARgamma plays a critical role in regulating the inflammatory responses of microglia and monocytes to beta-amyloid. We argue that the efficacy of NSAIDs in the treatment of AD may be a consequence of their actions on PPARgamma rather than on their canonical targets the cyclooxygenases. Importantly, the efficacy of these agents in inhibiting a broad range of inflammatory responses suggests PPARgamma agonists may provide a novel therapeutic approach to AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/farmacología , Antiinflamatorios no Esteroideos/farmacología , Astrocitos/fisiología , Microglía/fisiología , Fragmentos de Péptidos/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Tiazolidinedionas , Factores de Transcripción/agonistas , Animales , Animales Recién Nacidos , Astrocitos/citología , Encéfalo/citología , Encéfalo/fisiología , Diferenciación Celular , Cromanos/farmacología , Ciclooxigenasa 2 , Dinoprost/farmacología , Genes Reporteros , Humanos , Inflamación , Interleucina-6/genética , Isoenzimas/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Microcuerpos/fisiología , Microglía/citología , Microglía/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/fisiología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Recombinantes/biosíntesis , Acetato de Tetradecanoilforbol/farmacología , Tiazoles/farmacología , Transfección , Troglitazona , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/genética
6.
J Neurosci ; 19(19): 8182-98, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10493720

RESUMEN

Post-traumatic cystic cavitation, in which the size and severity of a CNS injury progress from a small area of direct trauma to a greatly enlarged secondary injury surrounded by glial scar tissue, is a poorly understood complication of damage to the brain and spinal cord. Using minimally invasive techniques to avoid primary physical injury, this study demonstrates in vivo that inflammatory processes alone initiate a cascade of secondary tissue damage, progressive cavitation, and glial scarring in the CNS. An in vitro model allowed us to test the hypothesis that specific molecules that stimulate macrophage inflammatory activation are an important step in initiating secondary neuropathology. Time-lapse video analyses of inflammation-induced cavitation in our in vitro model revealed that this process occurs primarily via a previously undescribed cellular mechanism involving dramatic astrocyte morphological changes and rapid migration. The physical process of cavitation leads to astrocyte abandonment of neuronal processes, neurite stretching, and secondary injury. The macrophage mannose receptor and the complement receptor type 3 beta2-integrin are implicated in the cascade that induces cavity and scar formation. We also demonstrate that anti-inflammatory agents modulating transcription via the nuclear hormone receptor peroxisome proliferator-activated receptor-gamma may be therapeutic in preventing progressive cavitation by limiting inflammation and subsequent secondary damage after CNS injury.


Asunto(s)
Astrocitos/patología , Lesiones Encefálicas/patología , Corteza Cerebral/patología , Ganglios Espinales/patología , Inflamación , Neuroglía/patología , Neuronas/patología , Tiazolidinedionas , Animales , Animales Recién Nacidos , Astrocitos/fisiología , Astrocitos/ultraestructura , Axones/patología , Axones/ultraestructura , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/fisiopatología , Movimiento Celular , Células Cultivadas , Ganglios Espinales/lesiones , Ganglios Espinales/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/análisis , Indometacina/farmacología , Inflamación/inducido químicamente , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Activación de Macrófagos , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/fisiología , Microscopía por Video , Neuritas/patología , Neuritas/fisiología , Neuritas/ultraestructura , Neuroglía/fisiología , Neuroglía/ultraestructura , Neuronas/fisiología , Neuronas/ultraestructura , Proteoglicanos/biosíntesis , Proteoglicanos/genética , Ratas , Ratas Sprague-Dawley , Tiazoles/farmacología , Zimosan/administración & dosificación , Zimosan/toxicidad
7.
J Neurosci ; 19(3): 928-39, 1999 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9920656

RESUMEN

Microglial interaction with amyloid fibrils in the brains of Alzheimer's and prion disease patients results in the inflammatory activation of these cells. We observed that primary microglial cultures and the THP-1 monocytic cell line are stimulated by fibrillar beta-amyloid and prion peptides to activate identical tyrosine kinase-dependent inflammatory signal transduction cascades. The tyrosine kinases Lyn and Syk are activated by the fibrillar peptides and initiate a signaling cascade resulting in a transient release of intracellular calcium that results in the activation of classical PKC and the recently described calcium-sensitive tyrosine kinase PYK2. Activation of the MAP kinases ERK1 and ERK2 follows as a subsequent downstream signaling event. We demonstrate that PYK2 is positioned downstream of Lyn, Syk, and PKC. PKC is a necessary intermediate required for ERK activation. Importantly, the signaling response elicited by beta-amyloid and prion fibrils leads to the production of neurotoxic products. We have demonstrated in a tissue culture model that conditioned media from beta-amyloid- and prion-stimulated microglia or from THP-1 monocytes are neurotoxic to mouse cortical neurons. This toxicity can be ameliorated by treating THP-1 cells with specific enzyme inhibitors that target various components of the signal transduction pathway linked to the inflammatory responses.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Amiloide/biosíntesis , Microglía/fisiología , Neurotoxinas/metabolismo , Fragmentos de Péptidos/farmacología , Priones/farmacología , Transducción de Señal/fisiología , Péptidos beta-Amiloides/química , Animales , Calcio/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células Cultivadas , Activación Enzimática/fisiología , Precursores Enzimáticos/fisiología , Quinasa 2 de Adhesión Focal , Membranas Intracelulares/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Fosforilación , Priones/química , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/fisiología , Quinasa Syk , Tirosina/metabolismo , Familia-src Quinasas/fisiología
8.
J Neurosci ; 18(12): 4451-60, 1998 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-9614222

RESUMEN

The senile plaques of Alzheimer's disease are foci of local inflammatory responses, as evidenced by the presence of acute phase proteins and oxidative damage. Fibrillar forms of beta-amyloid (Abeta), which are the primary constituents of senile plaques, have been shown to activate tyrosine kinase-dependent signal transduction cascades, resulting in inflammatory responses in microglia. However, the downstream signaling pathways mediating Abeta-induced inflammatory events are not well characterized. We report that exposure of primary rat microglia and human THP1 monocytes to fibrillar Abeta results in the tyrosine kinase-dependent activation of two parallel signal transduction cascades involving members of the mitogen-activated protein kinase (MAPK) superfamily. Abeta stimulated the rapid, transient activation of extracellular signal-regulated kinase 1 (ERK1) and ERK2 in microglia and ERK2 in THP1 monocytes. A second superfamily member, p38 MAPK, was also activated with similar kinetics. Scavenger receptor and receptor for advanced glycated end products (RAGE) ligands failed to activate ERK and p38 MAPK in the absence of significant increases in protein tyrosine phosphorylation, demonstrating that scavenger receptors and RAGE are not linked to these pathways. Importantly, the stress-activated protein kinases (SAPKs) were not significantly activated in response to Abeta. Downstream effectors of the MAPK signal transduction cascades include MAPKAP kinases, such as RSK1 and RSK2, as well as transcription factors. Exposure of microglia and THP1 monocytes to Abeta resulted in the activation of RSK1 and RSK2 and phosphorylation of cAMP response element-binding protein at Ser133, providing a mechanism for Abeta-induced changes in gene expression.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Microglía/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Monocitos/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Humanos , Fragmentos de Péptidos/farmacología , Fosforilación , Proteínas Quinasas/metabolismo , Ratas , Estilbenos/farmacología , Tirosina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
9.
Brain Res Dev Brain Res ; 107(1): 143-58, 1998 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-9602100

RESUMEN

Attempts to describe a mechanism of neurofibrillary tangle formation often focus on site specific phosphorylations of tau protein. These have typically been described in both Alzheimer's disease and developing brains. Therefore, study of the developmental regulation of Alzheimer epitope tau phosphorylations may help explain their persistence or recurrence during Alzheimer's disease. Using fetal rat hippocampal cultures, we report a spatial and temporal expression of tau phosphorylation during neuronal differentiation. We have examined phosphorylation at the epitopes recognized by monoclonal antibodies, PHF-1 and Tau 1. Tau was highly phosphorylated at the PHF-1 epitope at all culture ages examined using both immunohistochemical staining and Western blots. Tau was heavily phosphorylated at the Tau 1 epitope only in older cultures. The populations of tau recognized by the two antibodies also exhibited different solubilities, suggesting different microtubule binding behaviors: tau phosphorylated at PHF-1 was retained in axons following solubilization whereas Tau 1 immunoreactive tau was not retained in any cell compartment. Finally, in this culture system, maintenance of phosphorylation at the PHF-1 epitope, but not the Tau 1 epitope, required protein kinase C activity. These results indicate unique regulatory mechanisms and roles for each of these phosphorylated tau epitopes.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Feto/fisiología , Hipocampo/embriología , Neuronas/metabolismo , Proteína Quinasa C/fisiología , Proteínas tau/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Diferenciación Celular/fisiología , Células Cultivadas , Desarrollo Embrionario y Fetal/fisiología , Epítopos/inmunología , Feto/citología , Inmunohistoquímica , Neuronas/citología , Neuronas/inmunología , Fosforilación , Ratas , Ratas Sprague-Dawley
10.
Neurobiol Aging ; 18(5): 475-81, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9390773

RESUMEN

A rapid reversible tau phosphorylation at Ser 396/404 was observed in adult human cortical biopsy tissue and rat primary cortical cell cultures. Tau phosphorylation increased usually during the first 20-30 min in phosphate-buffered saline, followed by a decrease. The time course of tau phosphorylation and dephosphorylation in biopsy tissue could be lengthened by culturing in defined, oxygenated medium, instead of in phosphate-buffered saline. Phosphorylation of total protein in biopsy tissue occurred in two phases, with peaks at 30 and 90 min. The first peak of total protein phosphorylation coincided with the peak of tau phosphorylation, although both the first and second peaks of total protein phosphorylation coincided with the first and second peaks of neurofilament-H phosphorylation.


Asunto(s)
Química Encefálica/fisiología , Proteínas tau/metabolismo , Adolescente , Adulto , Animales , Biopsia , Western Blotting , Muerte Celular , Niño , Medios de Cultivo , Epilepsia/metabolismo , Epilepsia/cirugía , Humanos , Técnicas In Vitro , Persona de Mediana Edad , Neocórtex/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Neurofilamentos/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA