Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell ; 161(3): 610-621, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25910210

RESUMEN

Cytotoxic brain edema triggered by neuronal swelling is the chief cause of mortality following brain trauma and cerebral infarct. Using fluorescence lifetime imaging to analyze contributions of intracellular ionic changes in brain slices, we find that intense Na(+) entry triggers a secondary increase in intracellular Cl(-) that is required for neuronal swelling and death. Pharmacological and siRNA-mediated knockdown screening identified the ion exchanger SLC26A11 unexpectedly acting as a voltage-gated Cl(-) channel that is activated upon neuronal depolarization to membrane potentials lower than -20 mV. Blockade of SLC26A11 activity attenuates both neuronal swelling and cell death. Therefore cytotoxic neuronal edema occurs when sufficient Na(+) influx and depolarization is followed by Cl(-) entry via SLC26A11. The resultant NaCl accumulation causes subsequent neuronal swelling leading to neuronal death. These findings shed light on unique elements of volume control in excitable cells and lay the ground for the development of specific treatments for brain edema.


Asunto(s)
Edema Encefálico/patología , Antiportadores de Cloruro-Bicarbonato/metabolismo , Neuronas/metabolismo , Animales , Edema Encefálico/metabolismo , Muerte Celular , Células Cultivadas , Antiportadores de Cloruro-Bicarbonato/química , Humanos , Técnicas In Vitro , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/metabolismo , Ratones , Neuronas/patología , Ratas , Sodio/metabolismo , Transportadores de Sulfato
2.
Glia ; 69(3): 567-578, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32946147

RESUMEN

Immunometabolism refers to the rearrangement of metabolic pathways in response to immune stimulation, and the ability of these metabolic pathways themselves to control immune functions. Many aspects of immunometabolism have been revealed through studies of peripheral immune cells. However, immunometabolic reprogramming of microglia, the resident immune cell of the central nervous system, and the consequential outcome on neuronal activity have remained difficult to unravel. Microglia are highly sensitive to subtle changes in their environment, limiting the techniques available to study their metabolic and inflammatory profiles. Here, using fluorescence lifetime imaging of endogenous NAD(P)H, we measure the metabolic activity of individual microglia within acute hippocampal slices. We observed an LPS-induced increase in aerobic glycolysis, which was blocked by the addition of 5 mM 2-deoxyglucose (2DG). This LPS-induced glycolysis in microglia was necessary for the stabilization of hypoxia inducible factor-1α (HIF-1α) and production of the proinflammatory cytokine, interleukin-1ß (IL-1ß). Upon release, IL-1ß acted via the neuronal interleukin-1 receptor to inhibit the formation of synaptic long-term potentiation (LTP) following high frequency stimulation. Remarkably, the addition of 2DG to blunt the microglial glycolytic increase also inhibited HIF-1α accumulation and IL-1ß production, and therefore rescued LTP in LPS-stimulated slices. Overall, these data reveal the importance of metabolic reprogramming in regulating microglial immune functions, with appreciable outcomes on cytokine release and neuronal activity.


Asunto(s)
Potenciación a Largo Plazo , Microglía , Citocinas/metabolismo , Hipocampo/metabolismo , Lipopolisacáridos/farmacología , Microglía/metabolismo
3.
J Neurosci ; 37(9): 2403-2414, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28137973

RESUMEN

Cerebral blood flow (CBF) is controlled by arterial blood pressure, arterial CO2, arterial O2, and brain activity and is largely constant in the awake state. Although small changes in arterial CO2 are particularly potent to change CBF (1 mmHg variation in arterial CO2 changes CBF by 3%-4%), the coupling mechanism is incompletely understood. We tested the hypothesis that astrocytic prostaglandin E2 (PgE2) plays a key role for cerebrovascular CO2 reactivity, and that preserved synthesis of glutathione is essential for the full development of this response. We combined two-photon imaging microscopy in brain slices with in vivo work in rats and C57BL/6J mice to examine the hemodynamic responses to CO2 and somatosensory stimulation before and after inhibition of astrocytic glutathione and PgE2 synthesis. We demonstrate that hypercapnia (increased CO2) evokes an increase in astrocyte [Ca2+]i and stimulates COX-1 activity. The enzyme downstream of COX-1 that synthesizes PgE2 (microsomal prostaglandin E synthase-1) depends critically for its vasodilator activity on the level of glutathione in the brain. We show that, when glutathione levels are reduced, astrocyte calcium-evoked release of PgE2 is decreased and vasodilation triggered by increased astrocyte [Ca2+]iin vitro and by hypercapnia in vivo is inhibited. Astrocyte synthetic pathways, dependent on glutathione, are involved in cerebrovascular reactivity to CO2 Reductions in glutathione levels in aging, stroke, or schizophrenia could lead to dysfunctional regulation of CBF and subsequent neuronal damage.SIGNIFICANCE STATEMENT Neuronal activity leads to the generation of CO2, which has previously been shown to evoke cerebral blood flow (CBF) increases via the release of the vasodilator PgE2 We demonstrate that hypercapnia (increased CO2) evokes increases in astrocyte calcium signaling, which in turn stimulates COX-1 activity and generates downstream PgE2 production. We demonstrate that astrocyte calcium-evoked production of the vasodilator PgE2 is critically dependent on brain levels of the antioxidant glutathione. These data suggest a novel role for astrocytes in the regulation of CO2-evoked CBF responses. Furthermore, these results suggest that depleted glutathione levels, which occur in aging and stroke, will give rise to dysfunctional CBF regulation and may result in subsequent neuronal damage.


Asunto(s)
Astrocitos/metabolismo , Hipocampo/patología , Hipercapnia/patología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Animales , Animales Recién Nacidos , Dióxido de Carbono/metabolismo , Dióxido de Carbono/farmacología , Circulación Cerebrovascular/efectos de los fármacos , Clonidina/farmacología , Cicloleucina/análogos & derivados , Cicloleucina/farmacología , Ciclooxigenasa 1/metabolismo , Dinoprostona/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutatión/metabolismo , Técnicas In Vitro , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Norepinefrina/farmacología , Ratas , Ratas Wistar , Vibrisas/inervación
4.
J Neurosci ; 34(32): 10511-27, 2014 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-25100586

RESUMEN

Microglia are morphologically dynamic cells that rapidly extend their processes in response to various stimuli including extracellular ATP. In this study, we tested the hypothesis that stimulation of neuronal NMDARs trigger ATP release leading to communication with microglia. We used acute mouse hippocampal brain slices and two-photon laser scanning microscopy to study microglial dynamics and developed a novel protocol for fixation and immunolabeling of microglia processes. Similar to direct topical ATP application in vivo, short multiple applications of NMDA triggered transient microglia process outgrowth that was reversible and repeatable indicating that this was not due to excitotoxic damage. Stimulation of NMDAR was required as NMDAR antagonists, but not blockers of AMPA/kainate receptors or voltage-gated sodium channels, prevented microglial outgrowth. We report that ATP release, secondary to NMDAR activation, was the key mediator of this neuron-microglia communication as both blocking purinergic receptors and inhibiting hydrolysis of ATP to prevent locally generated gradients abolished outgrowth. Pharmacological and genetic analyses showed that the NMDA-triggered microglia process extension was independent of Pannexin 1, the ATP releasing channels, ATP release from astrocytes via connexins, and nitric oxide generation. Finally, using whole-cell patch clamping we demonstrate that activation of dendritic NMDAR on single neurons is sufficient to trigger microglia process outgrowth. Our results suggest that dendritic neuronal NMDAR activation triggers ATP release via a Pannexin 1-independent manner that induces outgrowth of microglia processes. This represents a novel uncharacterized form of neuron-microglial communication mediated by ATP.


Asunto(s)
Adenosina Trifosfato/metabolismo , Encéfalo/citología , Microglía/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Encéfalo/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/genética , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Femenino , Técnicas In Vitro , Magnesio/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/citología , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Receptores Purinérgicos P2Y12/metabolismo , Factores de Tiempo
5.
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
6.
J Neurosci ; 33(6): 2562-70, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23392684

RESUMEN

Cortical spreading depression (CSD) is associated with release of arachidonic acid, impaired neurovascular coupling, and reduced cerebral blood flow (CBF), caused by cortical vasoconstriction. We tested the hypothesis that the released arachidonic acid is metabolized by the cytochrome P450 enzyme to produce the vasoconstrictor 20-hydroxyeicosatetraenoic acid (20-HETE), and that this mechanism explains cortical vasoconstriction and vascular dysfunction after CSD. CSD was induced in the frontal cortex of rats and the cortical electrical activity and local field potentials recorded by glass microelectrodes, CBF by laser Doppler flowmetry, and tissue oxygen tension (tpO(2)) using polarographic microelectrodes. 20-HETE synthesis was measured in parallel experiments in cortical brain slices exposed to CSD. We used the specific inhibitor HET0016 (N-hydroxy-N'-(4-n-butyl-2-methylphenyl)formamidine) to block 20-HETE synthesis. CSD increased 20-HETE synthesis in brain slices for 120 min, and the time course of the increase in 20-HETE paralleled the reduction in CBF after CSD in vivo. HET0016 blocked the CSD-induced increase in 20-HETE synthesis and ameliorated the persistent reduction in CBF, but not the impaired neurovascular coupling after CSD. These findings suggest that CSD-induced increments in 20-HETE cause the reduction in CBF after CSD and that the attenuation of stimulation-induced CBF responses after CSD has a different mechanism. We suggest that blockade of 20-HETE synthesis may be clinically relevant to ameliorate reduced CBF in patients with migraine and acute brain cortex injuries.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/metabolismo , Circulación Cerebrovascular/fisiología , Depresión de Propagación Cortical/fisiología , Ácidos Hidroxieicosatetraenoicos/biosíntesis , Animales , Masculino , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Ratas Wistar
7.
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
8.
Nature ; 456(7223): 745-9, 2008 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-18971930

RESUMEN

Calcium signalling in astrocytes couples changes in neural activity to alterations in cerebral blood flow by eliciting vasoconstriction or vasodilation of arterioles. However, the mechanism for how these opposite astrocyte influences provide appropriate changes in vessel tone within an environment that has dynamic metabolic requirements remains unclear. Here we show that the ability of astrocytes to induce vasodilations over vasoconstrictions relies on the metabolic state of the rat brain tissue. When oxygen availability is lowered and astrocyte calcium concentration is elevated, astrocyte glycolysis and lactate release are maximized. External lactate attenuates transporter-mediated uptake from the extracellular space of prostaglandin E(2), leading to accumulation and subsequent vasodilation. In conditions of low oxygen concentration extracellular adenosine also increases, which blocks astrocyte-mediated constriction, facilitating dilation. These data reveal the role of metabolic substrates in regulating brain blood flow and provide a mechanism for differential astrocyte control over cerebrovascular diameter during different states of brain activation.


Asunto(s)
Arteriolas/metabolismo , Astrocitos/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Vasoconstricción/fisiología , Vasodilatación/fisiología , Adenosina/metabolismo , Adenosina/farmacología , Animales , Arteriolas/efectos de los fármacos , Dinoprostona/metabolismo , Glucólisis , Ácido Láctico/metabolismo , Masculino , Transportadores de Anión Orgánico/metabolismo , Oxígeno/metabolismo , Presión , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas , Ratas Sprague-Dawley , Vasoconstricción/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
9.
Cell Rep ; 42(10): 113128, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37742194

RESUMEN

Neuronal swelling during cytotoxic edema is triggered by Na+ and Cl- entry and is Ca2+ independent. However, the causes of neuronal death during swelling are unknown. Here, we investigate the role of large-conductance Pannexin-1 (Panx1) channels in neuronal death during cytotoxic edema. Panx1 channel inhibitors reduce and delay neuronal death in swelling triggered by voltage-gated Na+ entry with veratridine. Neuronal swelling causes downstream production of reactive oxygen species (ROS) that opens Panx1 channels. We confirm that ROS activates Panx1 currents with whole-cell electrophysiology and find scavenging ROS is neuroprotective. Panx1 opening and subsequent ATP release attract microglial processes to contact swelling neurons. Depleting microglia using the CSF1 receptor antagonist PLX3397 or blocking P2Y12 receptors exacerbates neuronal death, suggesting that the Panx1-ATP-dependent microglia contacts are neuroprotective. We conclude that cytotoxic edema triggers oxidative stress in neurons that opens Panx1 to trigger death but also initiates neuroprotective feedback mediated by microglia contacts.


Asunto(s)
Conexinas , Microglía , Microglía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Conexinas/metabolismo , Muerte Celular , Adenosina Trifosfato/metabolismo
10.
Redox Biol ; 56: 102448, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36037587

RESUMEN

The inter-relationship between microglia dynamics and oxidative stress (Ox-stress) in dystrophic neurites (DNs) at Alzheimer's Disease (AD) plaques may contribute to the pathological changes in neurons. We developed new in vivo imaging strategies to combine EGFP expression in microglia with neuronal expression of genetically encoded ratiometric redox sensors (rogRFP2 or roGFP1), and immunohistochemistry to investigate how microglia influence Ox-stress at amyloid plaques in 5xFAD AD mice. By simultaneously imaging microglia morphology and neuronal Ox-stress over time in vivo and in fixed brains we found that microglia preferentially enwrapped DNs exhibiting the greatest degree of Ox-stress. After microglia were partially depleted with the CSF1 receptor antagonist PLX3397, Ox-stress in DNs increased in a manner that was inversely correlated to the extent of coverage of the adjacent Aß plaques by the remaining microglia. These data suggest that microglia do not create Ox-stress at Aß plaques but instead create protective barriers around Aß plaques possibly reducing the spread of Aß. Intracranial injection of Aß was sufficient to induce neuronal Ox-stress suggesting it to be the initial trigger of Ox-stress generation. Although Ox-stress is increased in DNs, neuronal survival is enhanced following microglia depletion indicating complex and multifactorial roles of microglia with both neurotoxic and neuroprotective components. Increased Ox-stress of DNs was correlated with higher LAMP1 and ubiquitin immunoreactivity supporting proposed mechanistic links between lysosomal accumulation in DNs and their intrinsic generation of Ox-stress. Our results suggest protective as well as neurotoxic roles for microglia at plaques and that the generation of Ox-stress of DNs could intrinsically be generated via lysosomal disruption rather than by microglia. In Brief: Simultaneous imaging of microglia and neuronal Ox-stress revealed a double-edged role for microglia in 5xFAD mice. Plaque associated microglia were attracted to and enwrapped Aß plaques as well as the most highly oxidized DNs. After partial depletion of microglia, DNs were larger with greater levels of Ox-stress. Despite increased Ox-stress after microglia removal neuronal survival improved. Greater Ox-stress was correlated with increased levels of LAMP1 and ubiquitin thereby linking lysosome accumulation and Ox-stress in DNs.


Asunto(s)
Enfermedad de Alzheimer , Placa Amiloide , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Lisosomas/metabolismo , Ratones , Ratones Transgénicos , Neuritas , Oxidación-Reducción , Estrés Oxidativo , Placa Amiloide/metabolismo , Placa Amiloide/patología , Ubiquitinas/metabolismo , Ubiquitinas/farmacología
11.
Hippocampus ; 21(9): 958-67, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20865744

RESUMEN

In cultured hippocampal neurons, transient receptor potential 5 (TRPC5) channels are translocated and inserted into plasma membranes of hippocampal neurons to generate nonselective cation (NSC) currents. We investigated whether TRPC5 channel translocation also contributes to the generation of NSC currents underlying the afterdepolarizations and plateau potentials (PPs) in hippocampal pyramidal cells that are induced by muscarinic receptor activation. Using a biotinylation assay to quantify the change in surface membrane proteins in acute hippocampal slices, we found that muscarinic stimulation significantly enhanced the levels of TRPC5 protein on the membrane surface but not those of TRPC1 or TRPC4 channels. We then investigated the pharmacological sensitivity of the cation current observed during muscarinic stimulation to determine if a component could be due to TRPC5 channels. The TRPC channel antagonists 2-APB and SKF96365 strongly depressed the generation of PPs, the underlying tail currents (I(tail)) and the associated dendritic Ca(2+) influx induced by muscarinic receptor activation in pyramidal neurons. High intracellular concentrations of ATP, which specifically inhibit TRPC5 channels, depressed I(tail). In addition, pretreatment with the calmodulin (CaM) inhibitor W-7, which depresses recombinant TRPC5 currents, inhibited both the cation current (I(tail)) and the surface insertion of TRPC5 channels. Finally, the phosphatidylinositide 3-kinase (PI(3)K) inhibitor wortmannin, which blocks translocation of TRPC5 channels in cell culture, also inhibited both the I(tail) and the surface insertion of TRPC5 channels. Therefore, we conclude that insertion of TRPC5 channels contributes to the generation of the prolonged afterdepolarizations following muscarinic stimulation. This altered plasma membrane expression of TRPC5 channels in pyramidal neurons may play an important role in the generation of prolonged neuronal depolarization and bursting during the epileptiform seizure discharges of epilepsy.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Membrana Celular/metabolismo , Potenciales de la Membrana/fisiología , Células Piramidales/metabolismo , Receptores Muscarínicos/metabolismo , Canales Catiónicos TRPC/metabolismo , Androstadienos/farmacología , Animales , Compuestos de Boro/farmacología , Señalización del Calcio , Calmodulina/antagonistas & inhibidores , Dendritas/metabolismo , Epilepsia/fisiopatología , Epilepsia/terapia , Humanos , Potenciales de la Membrana/efectos de los fármacos , Microscopía Fluorescente , Técnicas de Placa-Clamp , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fotones , Ratas , Ratas Sprague-Dawley , Sulfonamidas/farmacología , Canales Catiónicos TRPC/antagonistas & inhibidores , Wortmanina
12.
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
13.
Nat Commun ; 11(1): 1559, 2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32214088

RESUMEN

Microglia are highly motile cells that continuously monitor the brain environment and respond to damage-associated cues. While glucose is the main energy substrate used by neurons in the brain, the nutrients metabolized by microglia to support surveillance of the parenchyma remain unexplored. Here, we use fluorescence lifetime imaging of intracellular NAD(P)H and time-lapse two-photon imaging of microglial dynamics in vivo and in situ, to show unique aspects of the microglial metabolic signature in the brain. Microglia are metabolically flexible and can rapidly adapt to consume glutamine as an alternative metabolic fuel in the absence of glucose. During insulin-induced hypoglycemia in vivo or in aglycemia in acute brain slices, glutaminolysis supports the maintenance of microglial process motility and damage-sensing functions. This metabolic shift sustains mitochondrial metabolism and requires mTOR-dependent signaling. This remarkable plasticity allows microglia to maintain their critical surveillance and phagocytic roles, even after brain neuroenergetic homeostasis is compromised.


Asunto(s)
Encéfalo/inmunología , Metabolismo Energético/fisiología , Microglía/metabolismo , Animales , Encéfalo/patología , Receptor 1 de Quimiocinas CX3C/genética , Movimiento Celular , Ácidos Grasos/metabolismo , Glucosa/deficiencia , Glucosa/metabolismo , Glutamina/metabolismo , Vigilancia Inmunológica , Ratones , Ratones Transgénicos , Microglía/citología , Microglía/inmunología , NAD/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
14.
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
15.
Cell Rep ; 27(10): 2895-2908.e4, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167136

RESUMEN

Microglia, the brain's immune cells, maintain homeostasis and sense pathological changes by continuously surveying the parenchyma with highly motile large processes. Here, we demonstrate that microglia also use thin actin-dependent filopodia that allow fast nanoscale sensing within discrete regions. Filopodia are distinct from large processes by their size, speed, and regulation mechanism. Increasing cyclic AMP (cAMP) by activating norepinephrine Gs-coupled receptors, applying nitric oxide, or inhibiting phosphodiesterases rapidly increases filopodia but collapses large processes. Alternatively, Gi-coupled P2Y12 receptor activation collapses filopodia but triggers large processes extension with bulbous tips. Similar control of cytoskeletal dynamics and microglial morphology by cAMP is observed in ramified primary microglia, suggesting that filopodia are intrinsically generated sensing structures. Therefore, nanoscale surveillance of brain parenchyma by microglia requires localized cAMP increases that drive filopodia formation. Shifting intracellular cAMP levels controls the polarity of microglial responses to changes in brain homeostasis and alters the scale of immunosurveillance.


Asunto(s)
Encéfalo/diagnóstico por imagen , AMP Cíclico/metabolismo , Microglía/metabolismo , Seudópodos/metabolismo , Actinas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microtúbulos/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Seudópodos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal
16.
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
17.
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
18.
Methods Mol Biol ; 438: 103-21, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18369753

RESUMEN

We document the protocols and methods for the production of immortalized cell lines of human neural stem cells from the human fetal central nervous system (CNS) cells by using a retroviral vector encoding v-myc oncogene. One of the human neural stem cell lines (HB1.F3) was found to express nestin and other specific markers for human neural stem cells, giving rise to three fundamental cell types of the CNS: neurons, astrocytes, and oligodendrocytes. After transplantation into the brain of mouse model of stroke, implanted human neural stem cells were observed to migrate extensively from the site of implantation into other anatomical sites and to differentiate into neurons and glial cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Sistema Nervioso Central/citología , Células Madre Multipotentes/citología , Línea Celular , Línea Celular Transformada , Células Cultivadas , Análisis Citogenético , Feto/citología , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Immunoblotting , Inmunohistoquímica , Células Madre Multipotentes/metabolismo , Especificidad de Órganos , Retroviridae , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , beta-Galactosidasa/metabolismo
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA