Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
J Neuroinflammation ; 12: 144, 2015 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-26255110

RESUMEN

BACKGROUND: The chemokine interleukin-8 (IL-8) and its receptor CXCR2 contribute to chemotactic responses in Alzheimer's disease (AD); however, properties of the ligand and receptor have not been characterized in animal models of disease. The primary aim of our study was to examine effects of pharmacological antagonism of CXCR2 as a strategy to inhibit receptor-mediated inflammatory reactivity and enhance neuronal viability in animals receiving intrahippocampal injection of amyloid-beta (Aß1-42). METHODS: In vivo studies used an animal model of Alzheimer's disease incorporating injection of full-length Aß1-42 into rat hippocampus. Immunohistochemical staining of rat brain was used to measure microgliosis, astrogliosis, neuronal viability, and oxidative stress. Western blot and Reverse Transcription PCR (RT-PCR) were used to determine levels of CXCR2 in animal tissue with the latter also used to determine expression of pro-inflammatory mediators. Immunostaining of human AD and non-demented (ND) tissue was also undertaken. RESULTS: We initially determined that in the human brain, AD relative to ND tissue exhibited marked increases in expression of CXCR2 with cell-specific receptor expression prominent in microglia. In Aß1-42-injected rat brain, CXCR2 and IL-8 showed time-dependent increases in expression, concomitant with enhanced gliosis, relative to controls phosphate-buffered saline (PBS) or reverse peptide Aß42-1 injection. Administration of the competitive CXCR2 antagonist SB332235 to peptide-injected rats significantly reduced expression of CXCR2 and microgliosis, with astrogliosis unchanged. Double staining studies demonstrated localization of CXCR2 and microglial immunoreactivity nearby deposits of Aß1-42 with SB332235 effective in inhibiting receptor expression and microgliosis. The numbers of neurons in granule cell layer (GCL) were reduced in rats receiving Aß1-42, compared with PBS, with administration of SB332235 to peptide-injected animals conferring neuroprotection. Oxidative stress was indicated in the animal model since both 4-hydroxynonenal (4-HNE) and hydroethidine (HEt) were markedly elevated in Aß1-42 vs. PBS-injected rat brain and diminished with SB332235 treatment. CONCLUSION: Overall, the findings suggest critical roles for CXCR2-dependent inflammatory responses in an AD animal model with pharmacological modulation of the receptor effective in inhibiting inflammatory reactivity and conferring neuroprotection against oxidative damage.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Inflamación/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Receptores de Interleucina-8B/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Animales , Encéfalo/patología , Femenino , Gliosis/patología , Hipocampo , Humanos , Mediadores de Inflamación , Masculino , Microinyecciones , Persona de Mediana Edad , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos , Ratas
2.
BMC Neurosci ; 15: 18, 2014 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-24447580

RESUMEN

BACKGROUND: The properties of Ca2+ signaling mediated by purinergic receptors are intrinsically linked with functional activity of astrocytes. At present little is known concerning Ca2+-dependent purinergic responses in adult human astrocytes. This work has examined effects of purinergic stimulation to alter levels of intracellular Ca2+ in adult human astrocytes. Ca2+-sensitive spectrofluorometry was carried out to determine mobilization of intracellular Ca2+ following adenosine triphosphate (ATP) or 3'-O-(4-benzoyl)benzoyl-ATP (Bz-ATP) stimulation of adult human astrocytes. In some experiments pharmacological modulation of Ca2+ pathways was applied to help elucidate mechanisms of Ca2+ signaling. RT-PCR was also performed to confirm human astrocyte expression of specific purinoceptors which were indicated from imaging studies. RESULTS: The endogenous P2 receptor agonist ATP (at 100 µM or 1 mM) applied in physiological saline solution (PSS) evoked a rapid increase of [Ca2+]i to a peak amplitude with the decay phase of response exhibiting two components. The two phases of decay consisted of an initial rapid component which was followed by a secondary slower component. In the presence of Ca2+-free solution, the secondary phase of decay was absent indicating this prolonged component was due to influx of Ca2+. This prolonged phase of decay was also attenuated with the store-operated channel (SOC) inhibitor gadolinium (at 2 µM) added to standard PSS, suggesting this component was mediated by SOC activation. These results are consistent with ATP activation of P2Y receptor (P2YR) in adult human astrocytes leading to respective rapid [Ca2+]i mobilization from intracellular stores followed by Ca2+ entry through SOC. An agonist for P2X7 receptor (P2X7R), BzATP induced a very different response compared with ATP whereby BzATP (at 300 µM) elicited a slowly rising increase in [Ca2+]i to a plateau level which was sustained in duration. The BzATP-induced increase in [Ca2+]i was not enhanced with lipopolysaccharide pre-treatment of cells as previously found for P2X7R mediated response in human microglia. RT-PCR analysis showed that adult human astrocytes in vitro constitutively express mRNA for P2Y1R, P2Y2R and P2X7R. CONCLUSION: These results suggest that activation of metabotropic P2YR (P2Y1R and/or P2Y2R) and ionotropic P2X7R could mediate purinergic responses in adult human astrocytes.


Asunto(s)
Astrocitos/fisiología , Canales de Calcio/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Activación del Canal Iónico/fisiología , Receptores Purinérgicos/metabolismo , Adulto , Células Cultivadas , Humanos
3.
Curr Alzheimer Res ; 20(7): 515-522, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37702232

RESUMEN

INTRODUCTION: The present study has examined microglial and astrocyte activation in association with neuronal degeneration in an animal model using an injection of amyloid-beta peptide Aß1-42 (Aß42) plus fibrinogen into rat hippocampus. METHODS: The combination of stimuli is suggested as a novel and potent perturbation to induce gliosis and the production of glial-derived neurotoxic factors in an animal model exhibiting a leaky BBB (blood-brain barrier). Specifically, Aß42 + fibrinogen stimulation elevated levels of COX-2 (cyclooxygenase-2) and iNOS (inducible nitric oxide synthase) with a considerable extent of neuronal loss associated with microglia and astrocyte activation. RESULTS: Treatment of injected rats with the broad spectrum anti-inflammatory agent, minocycline or the iNOS inhibitor, 1400 W inhibited gliosis, reduced levels of COX-2 and iNOS, and demonstrated efficacy for neuroprotection. CONCLUSION: The findings suggest the utility of combining amyloid beta peptide plus fibrinogen as a potent and understudied neuroinflammatory stimulus for the induction of glial-derived neurotoxic factors in BBB-compromised AD brain.


Asunto(s)
Péptidos beta-Amiloides , Gliosis , Ratas , Animales , Péptidos beta-Amiloides/metabolismo , Ciclooxigenasa 2/metabolismo , Gliosis/tratamiento farmacológico , Enfermedades Neuroinflamatorias , Microglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fibrinógeno , Hipocampo/metabolismo , Fragmentos de Péptidos/metabolismo
4.
Neurobiol Dis ; 45(1): 438-49, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21946335

RESUMEN

Central nervous system (CNS) inflammatory processes including microglial activation have been implicated in the pathogenesis of neurodegenerative diseases such as Huntington Disease (HD). We report age-dependent changes in striatal microglial morphology and vasculature in the YAC128 mouse model of HD. Decreases in microglial ramification along with a decrease in vessel diameter and increased vessel density and length suggest the presence of microgliosis and proangiogenic activity in YAC128 mice. Our hypothesis for this study was that the changes in microglial morphology and perturbations in vasculature may be involved in the pathogenesis of HD and that peripheral challenge with the bacterial endotoxin, lipopolysaccharide (LPS), will exacerbate these microglial and vascular changes as well as the HD phenotype in YAC128 mice at 12 months. Chronic peripheral LPS (1mg/kg) potentiated microglial activation indicated by an increase in microglial cell body size and retraction of processes. This potentiation in microglial activation with chronic peripheral LPS challenge was paralleled with vascular remodeling including dilatation, increased vessel wall thickness, increased BBB permeability and fibrinogen deposition in YAC128 striatum. Although peripheral LPS caused an increase in microglial activation and degenerative changes in cerebrovasculature, the phenotypic hallmarks of HD in YAC128 mice such as motor coordination deficits and decreased striatal volume were not exacerbated by chronic peripheral LPS exposure. This study identifies age-dependent increases in microglial activation and angiogenesis in YAC128 at 12 months. Peripheral inflammation induced by chronic LPS causes similar changes but does not influence the HD phenotype in YAC128 mice.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/patología , Enfermedad de Huntington/patología , Microglía/patología , Factores de Edad , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Forma de la Célula , Cuerpo Estriado/irrigación sanguínea , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Femenino , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Fenotipo
5.
Antioxidants (Basel) ; 11(1)2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-35052606

RESUMEN

The intactness of blood-brain barrier (BBB) is compromised in Alzheimer's disease (AD). Importantly, evidence suggests that the perturbation and abnormalities appearing in BBB can manifest early in the progression of the disease. The disruption of BBB allows extravasation of the plasma protein, fibrinogen, to enter brain parenchyma, eliciting immune reactivity and response. The presence of amyloid-ß (Aß) peptide leads to the formation of abnormal aggregates of fibrin resistant to degradation. Furthermore, Aß deposits act on the contact system of blood coagulation, altering levels of thrombin, fibrin clots and neuroinflammation. The neurovascular unit (NVU) comprises an ensemble of brain cells which interact with infiltrating fibrinogen. In particular, interaction of resident immune cell microglia with fibrinogen, fibrin and Aß results in the production of reactive oxygen species (ROS), a neurotoxic effector in AD brain. Overall, fibrinogen infiltration through a leaky BBB in AD animal models and in human AD tissue is associated with manifold abnormalities including persistent fibrin aggregation and clots, microglial-mediated production of ROS and diminished viability of neurons and synaptic connectivity. An objective of this review is to better understand how processes associated with BBB leakiness to fibrinogen link vascular pathology with neuronal and synaptic damage in AD.

6.
Curr Alzheimer Res ; 18(12): 925-938, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34789126

RESUMEN

Astrocytes contribute to brain development and homeostasis and support diverse functions of neurons. These cells also respond to the pathological processes in Alzheimer's disease (AD). There is still considerable debate concerning the overall contribution of astrocytes to AD pathogenesis since both the protective and harmful effects of these cells on neuronal survival have been documented. This review focuses exclusively on the neurotoxic potential of astrocytes while acknowledging that these cells can contribute to neurodegeneration through other mechanisms, for example, by lowered neurotrophic support. We identify reactive oxygen and nitrogen species, tumor necrosis factor α (TNF-α), glutamate, and matrix metalloproteinase (MMP)-9 as molecules that can be directly toxic to neurons and are released by reactive astrocytes. There is also considerable evidence suggesting their involvement in AD pathogenesis. We further discuss the signaling molecules that trigger the neurotoxic response of astrocytes with a focus on human cells. We also highlight microglia, the immune cells of the brain, as critical regulators of astrocyte neurotoxicity. Nuclear imaging and magnetic resonance spectroscopy (MRS) could be used to confirm the contribution of astrocyte neurotoxicity to AD progression. The molecular mechanisms discussed in this review could be targeted in the development of novel therapies for AD.


Asunto(s)
Enfermedad de Alzheimer , Astrocitos , Enfermedad de Alzheimer/patología , Humanos , Microglía/patología , Neuronas , Factor de Necrosis Tumoral alfa
7.
J Neurosci ; 29(1): 3-13, 2009 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-19129379

RESUMEN

We hypothesize that microglial chemotactic responses to amyloid-beta peptide (Abeta(1-42)) serve as an early and integral component of inflammatory response in Alzheimer's disease (AD) brain. This study reports a receptor for vascular endothelial growth factor (VEGF), termed VEGF-1 (Flt-1), subserves microglial chemotactic responses induced by Abeta(1-42) stimulation, in vivo and in vitro. Expression of Flt-1 was significantly increased in tissue obtained from AD patients [compared with tissue from nondemented (ND) individuals], in Abeta(1-42)-injected rat hippocampus, and in peptide-stimulated human microglia. Single and double immunohistochemical staining demonstrated marked immunoreactivity, for both Flt-1 and its ligand VEGF, in association with microglia and Abeta deposits in AD, but not ND, brain tissue. Functionally, treatment with anti-Flt-1 antibody was highly effective in inhibiting microglial mobility and chemotactic responses measured in vitro using a transwell migration assay. In vivo, transplanted enhanced green fluorescent protein (EGFP)-labeled microglia exhibited Flt-1-dependent chemotaxis induced by peptide injection with anti-Flt-1 effective in blocking migration of cells. Importantly, anti-Flt-1 reduction of microglial mobility was neuroprotective in peptide-injected hippocampus and associated with a significant increase in numbers of viable hippocampal neurons. The results of this study suggest critical functional roles for Flt-1 in mediating microglial chemotactic inflammatory responses which contribute to pathological conditions in AD brain.


Asunto(s)
Enfermedad de Alzheimer/patología , Movimiento Celular/fisiología , Corteza Entorrinal/patología , Microglía/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/administración & dosificación , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Antígeno CD11b/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Corteza Entorrinal/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Antígenos HLA-DR/metabolismo , Hipocampo/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Fragmentos de Péptidos/administración & dosificación , Fosfopiruvato Hidratasa/metabolismo , Cambios Post Mortem , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Curr Alzheimer Res ; 17(12): 1057-1064, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33463465

RESUMEN

The dysregulation of calcium signaling mechanisms in neurons has been considered a contributing factor to the pathogenesis evident in early-onset Alzheimer's Disease (AD). However, considerably less is known concerning the possible impairment of Ca2+ mobilization in resident immune cell microglia. This review considers findings which suggest that a prominent pathway for non-excitable microglial cells, store-operated calcium entry (SOCE), is altered in the sporadic form of AD. The patterns of Ca2+ mobilization are first discussed with platelet-activating factor (PAF) stimulation of SOCE in adult, fetal and immortalized cell-line, human microglia in the healthy brain. In all cases, PAF was found to induce a rapid transient depletion of Ca2+ from endoplasmic reticulum (ER) stores, followed by a sustained entry of Ca2+ (SOCE). A considerably attenuated duration of SOCE is observed with ATP stimulation of human microglia, suggested as due to agonist actions on differential subtype purinergic receptors. Microglia obtained from AD brain tissue, or microglia treated with full-length amyloid-ß peptide (Aß42), show significant reductions in the amplitude of SOCE relative to controls. In addition, AD brain and Aß42-treated microglia exhibit decreased levels of Ca2+ release from ER stores compared to controls. Changes in properties of SOCE in microglia could lead to altered immune cell response and neurovascular unit dysfunction in the inflamed AD brain.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Señalización del Calcio/fisiología , Calcio/metabolismo , Microglía/metabolismo , Péptidos beta-Amiloides/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Neuronas/patología
9.
J Cell Mol Med ; 13(9A): 2911-25, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18657226

RESUMEN

This study has used immunohistochemical examination of tissue obtained from Alzheimer's disease (AD) brains and rat hippocampus injected with Abeta(1-42) peptide to determine effects of induced inflammatory reactivity on integrity of blood-brain barrier (BBB) and viability of neurons. Tissue from AD, but not non-demented, brains exhibited a diffuse pattern of staining for fibrinogen and immunoglobulin (IgG) indicative of BBB leakiness with considerable fibrinogen immunoreactivity (ir) appearing in association with Abeta deposits. Immunostaining for the endothelial cell specific glycoprotein, von Willebrand factor, showed morphological evidence for altered blood vessels in AD tissue. AD brains also demonstrated extensive areas of fibrinogen ir in association with microglial reactivity. In vivo, intra-hippocampal injection of Abeta(1-42) caused time-dependent (1-7 days after injection) increases in double staining of fibrinogen with areas of microgliosis. Two independent pharmacological strategies were employed to examine how Abeta(1-42) stimulation (7 days injection) may be linked to neurodegeneration. The defibrinogenating compound, ancrod, reduced inflammatory reactivity, levels of parenchymal fibrinogen and IgG, and was neuroprotective. These results prompted use of Abeta(1-42) plus fibrinogen as a novel in vivo inflammatory stimulus and this combination significantly enhanced inflammatory reactivity, vascular perturbations and neuronal damage compared to Abeta(1-42) alone. A second approach, using anti-Mac-1 (antibody for antigen CD11b) to block activation of microglia, was highly effective in attenuating effects of Abeta(1-42) plus fibrinogen amplification of inflammatory and vascular responses and conferred significant neuroprotection. The overall findings from study of AD tissue and in vivo in Abeta(1-42) and Abeta(1-42) plus fibrinogen stimulated rat hippocampus suggest microglial responses to promote increased extravasation of blood protein as a critical component in amplifying inflammatory reactivity and causing neuronal damage in inflamed AD brain.


Asunto(s)
Enfermedad de Alzheimer/patología , Barrera Hematoencefálica/patología , Encéfalo/patología , Fibrinógeno/metabolismo , Inflamación/patología , Microglía/patología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/farmacología , Ancrod/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Demencia/complicaciones , Demencia/patología , Fluorescencia , Humanos , Inflamación/complicaciones , Antígeno de Macrófago-1/metabolismo , Masculino , Microglía/efectos de los fármacos , Persona de Mediana Edad , Fragmentos de Péptidos/farmacología , Permeabilidad/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factor de von Willebrand/metabolismo
10.
Glia ; 57(8): 898-907, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19115379

RESUMEN

Lysophosphatidylcholine (LPC), a major phospholipid component of atherogenic oxidized LDL, is implicated in atherosclerosis and, recently, in neurodegenerative diseases. We investigated the immunomodulatory functions of LPC in the central nervous system (CNS) using both an in vivo rat model, and in vitro culture systems of human primary astrocytes and a microglia cell line, HMO6. Compared with PBS injection, 20 nmol LPC-injection into the rat striatum increased astrocyte and microglial accumulation and elevated iNOS expression; concomitantly a time-dependent decrease in number of neurons was exhibited. In vitro studies on astrocytes and HMO6 cells showed that LPC increased the gene expression of proinflammatory factors IL-1beta, COX-2, and GM-CSF. LPC also induced chemotactic responses in HMO6 cells. Inhibition of rho kinase by fasudil, Y27632, or expressing a dominant negative form of rho kinase inhibited the LPC-induced IL-1beta mRNA expression in both astrocytes and HMO6. Moreover, intraperitoneal fasudil injection inhibited the LPC-induced microglial accumulation and iNOS expression and also was effective in protecting against neuronal loss. Silencing G2A, a specific receptor for LPC, inhibited proinflammatory gene expression and HMO6 migration. Overall, our results indicate that LPC induced considerable neuroinflammatory reactivity in glia mediated by rho kinase-dependent pathways with inhibition of these pathways conferring significant extents of neuroprotection.


Asunto(s)
Lisofosfatidilcolinas/farmacología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Quinasas Asociadas a rho/metabolismo , Amidas/farmacología , Animales , Encéfalo/citología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Feto , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lisofosfatidilcolinas/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo II/metabolismo , Piridinas/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
11.
J Neuroinflammation ; 6: 39, 2009 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20030829

RESUMEN

BACKGROUND: Transplantation of neural progenitor cells (NPC) constitutes a putative therapeutic maneuver for use in treatment of neurodegenerative diseases. At present, effects of NPC transplantation in Alzheimer's disease (AD) brain are largely unknown and a primary objective of this work was to demonstrate possible efficacy of NPC administration in an animal model of AD. The benefits of transplantation could involve a spectrum of effects including replacement of endogenous neurons or by conferring neuroprotection with enhancement of neurotrophic factors or diminishing levels of neurotoxic agents. Since chronic inflammation is a characteristic property of AD brain, we considered that transplantation of NPC could have particular utility in inhibiting ongoing inflammatory reactivity. We have tested intrahippocampal transplantation of NPC for efficacy in attenuating inflammatory responses and for neuroprotection in beta-amyloid (Abeta1-42) peptide-injected rat hippocampus. METHODS: Spheres of neural progenitor cells were grown from dissociated telencephalon tissue of rat embryos. NPC were infected with lentiviral vector green fluorescent protein (GFP) with subsequent cell transplantation into rat hippocampus previously injected (3 d prior) with Abeta1-42 peptide or PBS control. Immunohistochemical analysis was carried out (7 d post-NPC transplantation, 10 d post-peptide/PBS injection) for GFP, microgliosis (Iba-1 marker), astrogliosis (GFAP marker), neuron viability (MAP-2 marker) and levels of the proinflammatory cytokine, TNF-alpha. RESULTS: Successful infection of cultured NPC with lentiviral vector green fluorescent protein (GFP) was demonstrated prior to cell transplantation into rat hippocampus. In vivo, immunohistochemical staining showed migration of GFP-positive cells, in a region of dentate gyrus between Abeta1-42/PBS injection site and NPC transplantation site, was increased x2.8-fold with Abeta1-42 compared to PBS injection. Double immunostaining in peptide-injected brain indicated GFP association with nestin and GFAP, but not MAP-2. Cell-specific immunostaining showed marked increases in microgliosis and astrogliosis in Abeta1-42-injected brain (respective increases of x4.3- and x4.6-fold compared with PBS injection). NPC transplantation significantly reduced microgliosis (by 38%) but not astrogliosis in peptide-injected hippocampus. The proinflammatory cytokine TNF-alpha was elevated by 6.7-fold (peptide vs PBS injection) with NPC administration attenuating levels of TNF-alpha (by 40%). Peptide-injected brain demonstrated neuronal loss (MAP-2 staining reduced by 45% vs PBS injection) with NPC transplantation effective in conferring neuroprotection (26% recovery of neurons). CONCLUSIONS: These findings indicate efficacy for NPC transplantation in an animal model of AD with effects consistent with cellular actions to attenuate inflammatory reactivity induced by intrahippocampal peptide injection.


Asunto(s)
Enfermedad de Alzheimer/cirugía , Trasplante de Tejido Encefálico , Encefalitis/cirugía , Neuronas/trasplante , Trasplante de Células Madre , Telencéfalo/trasplante , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides , Animales , Muerte Celular , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Encefalitis/fisiopatología , Hipocampo/fisiopatología , Hipocampo/cirugía , Masculino , Neuroinmunomodulación/fisiología , Neuronas/fisiología , Fragmentos de Péptidos , Ratas , Ratas Sprague-Dawley , Células Madre/fisiología , Telencéfalo/fisiología
12.
BMC Cancer ; 9: 442, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20003523

RESUMEN

BACKGROUND: Glioma present as one of the most challenging cancers to treat, however, understanding of tumor cell biology is not well understood. Extracellular adenosine triphosphate (ATP) could serve as a critical signaling molecule regulating tumor development. This study has examined pharmacological modulation of calcium (Ca2+) entry through store-operated channels (SOC) on cellular expression and production of immune-cell mobilizing chemokines in ATP-stimulated C6 glioma cells. METHODS: Calcium spectrofluorometry was carried out to measure mobilization of intracellular Ca2+ [Ca2+]i following ATP stimulation of rat C6 glioma cells. Pretreatment with two inhibitors of SOC, SKF96365 or gadolinium, was used to examine for effects on [Ca2+]i. RT-PCR was performed to determine effects of purinergic stimulation on C6 cell expression of metabotropic P2Y receptors (P2YR) and the chemokines, monocyte chemoattractant protein-1 (MCP-1) and interleukin-8 (IL-8). ELISA was carried out to measure production of MCP-1 and IL-8 with ATP stimulation of glioma cells. RESULTS: Application of ATP (at 100 microM) to C6 glioma induced an increase in [Ca2+]i with the response exhibiting two components of decay. In the presence of the SOC inhibitors, SKF96365 or gadolinium, or with Ca2+-free solution, ATP responses lacked a slow phase suggesting the secondary component was due to SOC-mediated influx of Ca2+. RT-PCR confirmed expression of purinergic P2Y-subtype receptors in C6 cells which would serve as a precursor to activation of SOC. In addition, ATP-stimulated C6 cells showed enhanced expression of the chemokines, MCP-1 and IL-8, with SKF96365 or gadolinium effective in reducing chemokine expression. Gadolinium treatment of ATP-stimulated C6 cells was also found to inhibit the production of MCP-1 and IL-8. CONCLUSION: These results suggest ATP-induced Ca2+ entry, mediated by activation of SOC in C6 glioma, as a mechanism leading to increased cellular expression and release of chemokines. Elevated levels of MCP-1 and IL-8 are predicted to enhance the mobility of tumor cells and promote recruitment of microglia into developing tumors thereby supporting tumor growth.


Asunto(s)
Adenosina Trifosfato/farmacología , Calcio/metabolismo , Quimiocinas/metabolismo , Glioma/patología , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocinas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/diagnóstico , Glioma/genética , Glioma/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Microglía/patología , Invasividad Neoplásica , Pronóstico , Ratas , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
13.
Curr Alzheimer Res ; 16(11): 1007-1017, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31692444

RESUMEN

A combinatorial cocktail approach is suggested as a rationale intervention to attenuate chronic inflammation and confer neuroprotection in Alzheimer's disease (AD). The requirement for an assemblage of pharmacological compounds follows from the host of pro-inflammatory pathways and mechanisms present in activated microglia in the disease process. This article suggests a starting point using four compounds which present some differential in anti-inflammatory targets and actions but a commonality in showing a finite permeability through Blood-brain Barrier (BBB). A basis for firstchoice compounds demonstrated neuroprotection in animal models (thalidomide and minocycline), clinical trial data showing some slowing in the progression of pathology in AD brain (ibuprofen) and indirect evidence for putative efficacy in blocking oxidative damage and chemotactic response mediated by activated microglia (dapsone). It is emphasized that a number of candidate compounds, other than ones suggested here, could be considered as components of the cocktail approach and would be expected to be examined in subsequent work. In this case, systematic testing in AD animal models is required to rigorously examine the efficacy of first-choice compounds and replace ones showing weaker effects. This protocol represents a practical approach to optimize the reduction of microglial-mediated chronic inflammation in AD pathology. Subsequent work would incorporate the anti-inflammatory cocktail delivery as an adjunctive treatment with ones independent of inflammation as an overall preventive strategy to slow the progression of AD.


Asunto(s)
Enfermedad de Alzheimer , Quimioterapia Combinada/métodos , Inflamación , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/etiología , Animales , Dapsona/farmacología , Humanos , Ibuprofeno/farmacología , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Minociclina/farmacología , Talidomida/farmacología
14.
J Neurosci ; 27(18): 4957-68, 2007 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-17475804

RESUMEN

We investigated the involvement and roles of the ionotropic purinergic receptor P2X(7)R in microglia in mediating lipopolysaccharide (LPS)-induced inflammatory responses and neuronal damage in rat striatum. A detailed in vivo study showed that LPS injection into striatum markedly increased the expression of P2X(7)R in microglia compared with control (saline)-injected animals. Additionally, LPS injection upregulated a broad spectrum of proinflammatory mediators, including inducible nitric oxide synthase (nitric oxide production marker), 3-nitrotyrosine (peroxynitrite-mediated nitration marker), 4-hydroxynonenal (lipid peroxidation marker), and 8-hydroxy-2'-deoxyguanosine (oxidative DNA damage marker), and reduced neuronal viability. The P2X(7)R antagonist oxidized ATP (oxATP) was effective in attenuating expressions of all inflammatory mediators and in addition inhibited LPS-induced activation of the cellular signaling factors p38 mitogen-activated protein kinase and transcriptional factor nuclear factor kappaB. Most importantly, in vivo, oxATP blockade of P2X(7)R also reduced numbers of caspase-3-positive neurons and increased neuronal survival in LPS-injected brain. In vitro, LPS stimulation of cultured human microglia enhanced cellular expressions of a host of proinflammatory factors, including cyclooxygenase-2, interleukin-1beta (IL-1beta), IL-6, IL-12, and tumor necrosis factor-alpha; all factors were inhibited by oxATP. A novel finding was that LPS potentiated intracellular [Ca(2+)](i) mobilization induced by the P2X(7)R ligand 2',3'-O-(4-benzoyl-benzoyl) ATP, which could serve as a mechanistic link for P2X(7)R amplification of inflammatory responses. Our results suggest critical roles for P2X(7)R in mediating inflammation and inhibition of this subtype purinergic receptor as a novel therapeutic approach to reduce microglial activation and confer neuroprotection in inflamed and diseased brain.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Lipopolisacáridos/toxicidad , Microglía/metabolismo , Neuronas/metabolismo , Neuronas/patología , Receptores Purinérgicos P2/fisiología , Animales , Encéfalo/efectos de los fármacos , Células Cultivadas , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Microglía/efectos de los fármacos , Microglía/patología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X7
15.
J Neuroinflammation ; 5: 18, 2008 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-18492281

RESUMEN

BACKGROUND: Excitotoxic brain insult is associated with extensive neuronal damage but could also cause inflammatory reactivity and vascular remodeling. The effects of the vascular endothelial growth factor (VEGF) inhibitor, Cyclo-VEGI on expression of VEGF, microgliosis and astrogliosis, blood-brain barrier (BBB) integrity and neuronal viability have been studied following intra-striatal injection of the excitotoxin, quinolinic acid (QUIN). The purpose of this study was to examine VEGF-dependent inflammatory responses in excitotoxin-injected brain and their dependence on pharmacological antagonism of VEGF receptors. METHODS: Single and double immunofluorescence staining of cellular (microglia, astrocyte, neuron) responses and dye and protein infiltration of blood-brain barrier have been applied in the absence, and presence, of pharmacological modulation using a VEGF receptor antagonist, Cyclo-VEGI. Dunn-Bonferroni statistical analysis was used to measure for significance between animal groups. RESULTS: Detailed analysis, at a single time point of 1 d post-QUIN injection, showed excitotoxin-injected striatum to exhibit marked increases in microgliosis (ED1 marker), astrogliosis (GFAP marker) and VEGF expression, compared with PBS injection. Single and double immunostaining demonstrated significant effects of Cyclo-VEGI treatment of QUIN-injected striatum to inhibit microgliosis (by 38%), ED1/VEGF (by 42%) and VEGF striatal immunoreactivity (by 43%); astrogliosis and GFAP/VEGF were not significantly altered with Cyclo-VEGI treatment. Leakiness of BBB was indicated by infiltration of Evans blue dye and plasma protein fibrinogen into QUIN-injected striatum with barrier permeability restored by 62% (Evans blue permeability) and 49% (fibrinogen permeability) with Cyclo-VEGI application. QUIN-induced toxicity was demonstrated with loss of striatal neurons (NeuN marker) and increased neuronal damage (Fluoro-Jade marker) with significant neuroprotection conferred by Cyclo-VEGI treatment (33% increase in NeuN and 38% decrease in Fluoro-Jade). CONCLUSION: An antagonist for VEGF receptor-mediated signaling, Cyclo-VEGI, has shown efficacy in a broad spectrum of activity against striatal excitotoxic insult including inhibition of microgliosis, reduction in leakiness of BBB and parenchymal infiltration of plasma fibrinogen and in conferring significant protection for striatal neurons. Antagonism of VEGF-mediated activity, possibly targeting VEGF receptors on reactive microglia, is suggested as a neuroprotective mechanism against inflammatory reactivity and a novel strategy to attenuate acute excitotoxic damage.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Factores de Crecimiento Endotelial/uso terapéutico , Gliosis/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/toxicidad , Péptidos Cíclicos/uso terapéutico , Ácido Quinolínico/toxicidad , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Cuerpo Estriado/fisiopatología , Evaluación Preclínica de Medicamentos , Factores de Crecimiento Endotelial/farmacocinética , Factores de Crecimiento Endotelial/farmacología , Gliosis/inducido químicamente , Gliosis/patología , Gliosis/prevención & control , Inyecciones , Inyecciones Intraventriculares , Masculino , Microglía/efectos de los fármacos , Microglía/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Péptidos Cíclicos/farmacocinética , Péptidos Cíclicos/farmacología , Ratas , Ratas Sprague-Dawley
16.
Cancer Lett ; 260(1-2): 79-87, 2008 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-18039556

RESUMEN

Our results demonstrate the first findings of expression and function of the purinergic P2X7 receptor (P2X7R) in rat C6 glioma cells. P2X7R mRNA and protein were present in unstimulated C6 cells and were up-regulated by cell exposure to the P2X7R agonist, 2',3'-(benzoyl-4-benzoyl)-ATP (BzATP). Activation of P2X7R in C6 in response to BzATP led to increased mobilization of intracellular calcium [Ca2+]i and formation of large pores. Chronic exposure of C6 cells to BzATP enhanced the expression of pro-inflammatory factors including MCP-1, IL-8 and VEGF. In a scratch-wound migration assay, the P2X7R was shown to regulate cell mobility. The overall results suggest that P2X7R activation in C6 is linked with increased pro-inflammatory factors and tumor cell migration.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Agonistas del Receptor Purinérgico P2 , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Calcio/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/patología , Interleucina-8/metabolismo , ARN Mensajero/metabolismo , Ratas , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X7 , Factores de Tiempo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
J Neurosci ; 26(45): 11652-64, 2006 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-17093087

RESUMEN

We investigated the modulating actions of the nonselective K(+) channel blocker 4-aminopyridine (4-AP) on amyloid beta (Abeta(1-42))-induced human microglial signaling pathways and functional processes. Whole-cell patch-clamp studies showed acute application of Abeta(1-42) (5 mum) to human microglia led to rapid expression of a 4-AP-sensitive, non-inactivating outwardly rectifying K(+) current (I(K)). Intracellular application of the nonhydrolyzable analog of GTP, GTPgammaS, induced an outward K(+) current with similar properties to the Abeta(1-42)-induced I(K) including sensitivity to 4-AP (IC(50) = 5 mm). Reverse transcriptase-PCR showed a rapid expression of a delayed rectifier Kv3.1 channel in Abeta(1-42)-treated microglia. Abeta(1-42) peptide also caused a slow, progressive increase in levels of [Ca(2+)](i) (intracellular calcium) that was partially blocked by 4-AP. Chronic exposure of human microglia to Abeta(1-42) led to enhanced p38 mitogen-activated protein kinase and nuclear factor kappaB expression with factors inhibited by 4-AP. Abeta(1-42) also induced the expression and production of the pro-inflammatory cytokines interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha, the chemokine IL-8, and the enzyme cyclooxygenase-2; 4-AP was effective in reducing all of these pro-inflammatory mediators. Additionally, toxicity of supernatant from Abeta(1-42)-treated microglia on cultured rat hippocampal neurons was reduced if 4-AP was included with peptide. In vivo, injection of Abeta(1-42) into rat hippocampus induced neuronal damage and increased microglial activation. Daily administration of 1 mg/kg 4-AP was found to suppress microglial activation and exhibited neuroprotection. The overall results suggest that 4-AP modulation of an Abeta(1-42)-induced I(K) (candidate channel Kv3.1) and intracellular signaling pathways in human microglia could serve as a therapeutic strategy for neuroprotection in Alzheimer's disease pathology.


Asunto(s)
4-Aminopiridina/farmacología , Péptidos beta-Amiloides/farmacología , Microglía/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Encéfalo/citología , Calcio/metabolismo , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Interacciones Farmacológicas , Feto , Expresión Génica/efectos de los fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Humanos , Inmunohistoquímica/métodos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/efectos de la radiación , Proteínas de la Membrana/metabolismo , FN-kappa B/metabolismo , Neuronas/química , Neuronas/fisiología , Técnicas de Placa-Clamp/métodos , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Cancer Lett ; 402: 93-99, 2017 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-28536012

RESUMEN

This review considers evidence suggesting that activation of the ionotropic purinergic receptor P2X7 (P2X7R) is a contributing factor in the growth of brain tumors. Importantly, expression of P2X7R may be upregulated in both glioma cells and in immune responding microglial cells with possible differential effects on tumor progression. The recruitment of immune cells into tumor regions may not only be involved in supporting an immunosuppressive environment aiding tumor growth but activated microglia could secrete inflammatory factors promoting neoangiogenesis in expanding tumors. The subtype P2X7R exhibits a number of unique properties including activation of the receptor in pathological conditions associated with developing brain tumors. In particular, the tumor microenvironment includes elevated levels of ATP required for activation of P2X7R and the sustained tumor and immune cell P2X7R-mediated responses which in total contribute to overall tumor growth and viability. Studies on cultured rat and human glioma show marked increases in expression of P2X7R and enhanced cell mobility relative to control. Glioma cell animal models demonstrate enhanced expression of P2X7R in both glioma and microglia with antagonism of receptor showing differential effects on tumor growth. Overall, P2X7R activation is associated with a complexity of modulatory actions on tumor growth in part due to ubiquitous expression of the receptor in glioma and immune responsive cells.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Microglía/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Proliferación Celular , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Microglía/efectos de los fármacos , Microglía/patología , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/efectos de los fármacos , Transducción de Señal , Carga Tumoral , Microambiente Tumoral
19.
J Neuropathol Exp Neurol ; 65(11): 1090-7, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17086106

RESUMEN

The expression of the purinergic receptor subtype P2X(7)R, a nonselective cationic channel activated by high levels of adenosine triphosphate (ATP), has been studied in adult microglia obtained from Alzheimer disease (AD) and nondemented (ND) brains, in fetal human microglia exposed to Abeta(1-42) peptide and in vivo in Abeta(1-42)-injected rat hippocampus. Semiquantitative reverse transcriptase-polymerase chain reaction showed enhanced expression (increase of 70%) of P2X(7)R in AD microglia compared with ND cells (analysis of 6 AD and 8 ND cases). Immunohistochemical analysis showed prominent P2X(7)R expression in association with Abeta plaques and localized to HLA-DR-immunoreactive microglia. In cultured fetal human microglia, cells exposed to Abeta(1-42) (5 microM for 18 hours) had significantly elevated levels of P2X(7)R (by 106%) compared with untreated cells. Amplitudes of Ca(2+) responses in these cells, induced by the selective P2X(7)R agonist BzATP, were increased by 145% with Abeta(1-42) pretreatment relative to control (no peptide pretreatment) and were largely blocked if the P2X(7)R inhibitor-oxidized ATP (oxATP) was added with peptide in pretreatment solution. In vivo, double immunostaining analysis showed considerable P2X(7)R colocalized with microglia after injection of Abeta(1-42) (1 nmol) into rat hippocampus. The overall results suggest roles of P2X(7)R in mediating microglial purinergic inflammatory responses in AD brain.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Microglía/metabolismo , Receptores Purinérgicos P2/biosíntesis , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Marcadores de Afinidad/farmacología , Enfermedad de Alzheimer/patología , Animales , Calcio/metabolismo , Antígenos HLA-DR/metabolismo , Hipocampo/patología , Humanos , Inmunohistoquímica , Microglía/efectos de los fármacos , Microscopía Fluorescente , Péptidos/metabolismo , Ratas , Receptores Purinérgicos P2X7 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
20.
Am J Neurodegener Dis ; 5(1): 69-73, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27073744

RESUMEN

The effects of pyruvate, the end metabolite of glycolysis, on blood-brain barrier (BBB) impairment and immune reactivity were examined in the quinolinic acid (QA)-injected rat striatum. Extensive disruption of BBB was observed at 7 d post QA-injection as demonstrated by increased immunohistochemical staining using antibody against immunoglobulin G (IgG). Animals receiving pyruvate administration (500 mg/kg) with QA-injection exhibited reduced lgG immunoreactivity (by 45%) relative to QA alone. QA intrastriatal injection also resulted in marked increases in the number of infiltrating T-lymphocytes (by 70-fold) and expression of major histocompatibility complex (MHC-class II) (by 45-fold) relative to unlesioned control. Treatment with pyruvate significantly reduced infiltration of T-cells (by 68%) and MHC class II expression (by 48%) induced by QA. These results indicate that QA injection into rat striatum leads to impairment in BBB function with pyruvate administration reducing immune response and BBB leakiness in excitotoxic injury.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA