Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 386(3): 331-343, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37391223

RESUMEN

The effects of a general anesthetic xenon (Xe) on spontaneous, miniature, electrically evoked synaptic transmissions were examined using the "synapse bouton preparation," with which we can clearly evaluate pure synaptic responses and accurately quantify pre- and postsynaptic transmissions. Glycinergic and glutamatergic transmissions were investigated in rat spinal sacral dorsal commissural nucleus and hippocampal CA3 neurons, respectively. Xe presynaptically inhibited spontaneous glycinergic transmission, the effect of which was resistant to tetrodotoxin, Cd2+, extracellular Ca2+, thapsigargin (a selective sarcoplasmic/endoplasmic reticulum Ca2+-ATPase inhibitor), SQ22536 (an adenylate cyclase inhibitor), 8-Br-cAMP (membrane-permeable cAMP analog), ZD7288 (an hyperpolarization-activated cyclic nucleotide-gated channel blocker), chelerythrine (a PKC inhibitor), and KN-93 (a CaMKII inhibitor) while being sensitive to PKA inhibitors (H-89, KT5720, and Rp-cAMPS). Moreover, Xe inhibited evoked glycinergic transmission, which was canceled by KT5720. Like glycinergic transmission, spontaneous and evoked glutamatergic transmissions were also inhibited by Xe in a KT5720-sensitive manner. Our results suggest that Xe decreases glycinergic and glutamatergic spontaneous and evoked transmissions at the presynaptic level in a PKA-dependent manner. These presynaptic responses are independent of Ca2+ dynamics. We conclude that PKA can be the main molecular target of Xe in the inhibitory effects on both inhibitory and excitatory neurotransmitter release. SIGNIFICANCE STATEMENT: Spontaneous and evoked glycinergic and glutamatergic transmissions were investigated using the whole-cell patch clamp technique in rat spinal sacral dorsal commissural nucleus and hippocampal CA3 neurons, respectively. Xenon (Xe) significantly inhibited glycinergic and glutamatergic transmission presynaptically. As a signaling mechanism, protein kinase A was responsible for the inhibitory effects of Xe on both glycine and glutamate release. These results may help understand how Xe modulates neurotransmitter release and exerts its excellent anesthetic properties.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico , Xenón , Ratas , Animales , Ratas Wistar , Xenón/farmacología , Xenón/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neuronas , Transmisión Sináptica , Terminales Presinápticos/metabolismo , Hipocampo/metabolismo , Médula Espinal , Neurotransmisores/metabolismo
2.
Pharmacol Res ; 180: 106227, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35452800

RESUMEN

Alzheimer's disease (AD) has become a major public health problem that affects the elderly population. Therapeutic compounds with curative effects are not available due to the complex pathogenesis of AD. Daphnetin, a natural coumarin derivative and inhibitor of various kinases, has anti-inflammatory and antioxidant activities. In this study, we found that daphnetin improved spatial learning and memory in an amyloid precursor protein (APP)/presenilin 1 (PS1) double-transgenic mouse model of AD. Daphnetin markedly decreased the levels of amyloid-ß peptide 1-40 (Aß40) and 1-42 (Aß42) in the cerebral cortex, downregulated the expressions of enzymes involved in APP processing, e.g., beta-site APP-cleaving enzyme (BACE), nicastrin and presenilin enhancer protein 2 (PEN2). We further found the reduced serum levels of inflammatory factors, including interleukin-1ß (IL-1ß), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and chemokine (C-C motif) ligand 3 (CCL3), while daphnetin increased total antioxidant capacity (T-AOC) and superoxide dismutase (SOD) levels in the serum. Interestingly, daphnetin markedly decreased the expression of glial fibrillary acidic protein (GFAP) and the upstream regulatory molecule- phosphorylated signal transducer and activator of transcription 3 (p-STAT3) in APP/PS1 mice, and mainly inhibited the phosphorylation of STAT3 at Ser727 to decrease GFAP expression evidenced in a LPS-activated glial cell model. These results suggest that daphnetin ameliorates cognitive deficits and that Aß deposition in APP/PS1 mice is mainly correlated with astrocyte activation and APP processing.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Anciano , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Antioxidantes/uso terapéutico , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/genética , Presenilina-1/metabolismo , Presenilina-1/uso terapéutico , Factor de Transcripción STAT3/metabolismo , Umbeliferonas
3.
J Neurophysiol ; 123(6): 2426-2436, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32401126

RESUMEN

In this study, the effect of extracellular pH on glutamatergic synaptic transmission was examined in mechanically dissociated rat hippocampal CA3 pyramidal neurons using a whole-cell patch-clamp technique under voltage-clamp conditions. Native synaptic boutons were isolated without using any enzymes, using a so-called "synapse bouton preparation," and preserved for the electrical stimulation of single boutons. Both the frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) were found to decrease and increase in response to modest acidic (~pH 6.5) and basic (~pH 8.5) solutions, respectively. These changes in sEPSC frequency were not affected by the addition of TTX but completely disappeared by successive addition of Cd2+. However, changes in sEPSC amplitude induced by acidic and basic extracellular solutions were not affected by the addition of neither TTX nor Cd2+. The glutamate-induced whole-cell currents were decreased and increased by acidic and basic solutions, respectively. Acidic pH also decreased the amplitude and increased the failure rate (Rf) and paired-pulse rate (PPR) of glutamatergic electrically evoked excitatory postsynaptic currents (eEPSCs), while a basic pH increased the amplitude and decreased both the Rf and PPR of eEPSCs. The kinetics of the currents were not affected by changes in pH. Acidic and basic solutions decreased and increased voltage-gated Ca2+ but not Na+ channel currents in the dentate gyrus granule cell bodies. Our results indicate that extracellular pH modulates excitatory transmission via both pre- and postsynaptic sites, with the presynaptic modulation correlated to changes in voltage-gated Ca2+ channel currents.NEW & NOTEWORTHY The effects of external pH changes on spontaneous, miniature, and evoked excitatory synaptic transmission in CA3 hippocampal synapses were examined using the isolated nerve bouton preparation, which allowed for the accurate regulation of extracellular pH at the synapses. Acidification generally reduced transmission, partly via effects on presynaptic Ca2+ channel currents, while alkalization generally enhanced transmission. Both pre- and postsynaptic sites contributed to these effects.


Asunto(s)
Región CA3 Hipocampal/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Concentración de Iones de Hidrógeno , Terminales Presinápticos/fisiología , Células Piramidales/fisiología , Animales , Región CA3 Hipocampal/química , Femenino , Ácido Glutámico/metabolismo , Masculino , Técnicas de Placa-Clamp , Terminales Presinápticos/química , Células Piramidales/química , Ratas , Ratas Wistar
4.
Can J Physiol Pharmacol ; 97(10): 909-915, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31100203

RESUMEN

Molecular hydrogen (H2) showed protection against various kinds of oxidative-stress-related diseases. First, it was reported that the mechanism of therapeutic effects of H2 was antioxidative effect due to inhibition of the most cytotoxic reactive oxygen species, hydroxy radical (•OH). However, after chronic administration of H2 in drinking water, oxidative-stress-induced nerve injury is significantly attenuated even in the absence of H2. It suggests indirect signaling of H2 and gastrointestinal tract is involved. Indirect effects of H2 could be tested by giving H2 water only before nerve injury, as preconditioning. For example, preconditioning of H2 for certain a period (∼7 days) in Parkinson's disease model mice shows significant neuroprotection. As the mechanism of indirect effect, H2 in drinking water induces ghrelin production and release from the stomach via ß1-adrenergic receptor stimulation. Released ghrelin circulates in the body, being transported across the blood-brain barrier, activates its receptor, growth-hormone secretagogue receptor. H2-induced upregulation of ghrelin mRNA is also shown in ghrelin-producing cell line, SG-1. These observations help with understanding the chronic effects of H2 and raise intriguing preventive and therapeutic options using H2.


Asunto(s)
Ghrelina/metabolismo , Hidrógeno/administración & dosificación , Enfermedades Neurodegenerativas/terapia , Neuroprotección/efectos de los fármacos , Traumatismos de los Nervios Periféricos/terapia , Animales , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Ingestión de Líquidos , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Ghrelina/sangre , Humanos , Enfermedades Neurodegenerativas/sangre , Estrés Oxidativo/efectos de los fármacos , Traumatismos de los Nervios Periféricos/sangre , Receptores de Ghrelina/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Agua/química
5.
Int J Mol Sci ; 20(4)2019 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-30795555

RESUMEN

As part of the blood-brain-barrier, astrocytes are ideally positioned between cerebral vasculature and neuronal synapses to mediate nutrient uptake from the systemic circulation. In addition, astrocytes have a robust enzymatic capacity of glycolysis, glycogenesis and lipid metabolism, managing nutrient support in the brain parenchyma for neuronal consumption. Here, we review the plasticity of astrocyte energy metabolism under physiologic and pathologic conditions, highlighting age-dependent brain dysfunctions. In astrocytes, glycolysis and glycogenesis are regulated by noradrenaline and insulin, respectively, while mitochondrial ATP production and fatty acid oxidation are influenced by the thyroid hormone. These regulations are essential for maintaining normal brain activities, and impairments of these processes may lead to neurodegeneration and cognitive decline. Metabolic plasticity is also associated with (re)activation of astrocytes, a process associated with pathologic events. It is likely that the recently described neurodegenerative and neuroprotective subpopulations of reactive astrocytes metabolize distinct energy substrates, and that this preference is supposed to explain some of their impacts on pathologic processes. Importantly, physiologic and pathologic properties of astrocytic metabolic plasticity bear translational potential in defining new potential diagnostic biomarkers and novel therapeutic targets to mitigate neurodegeneration and age-related brain dysfunctions.


Asunto(s)
Adaptación Fisiológica , Envejecimiento/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Animales , Encéfalo/crecimiento & desarrollo , Humanos
6.
Physiol Rev ; 91(2): 461-553, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21527731

RESUMEN

Microglial cells are the resident macrophages in the central nervous system. These cells of mesodermal/mesenchymal origin migrate into all regions of the central nervous system, disseminate through the brain parenchyma, and acquire a specific ramified morphological phenotype termed "resting microglia." Recent studies indicate that even in the normal brain, microglia have highly motile processes by which they scan their territorial domains. By a large number of signaling pathways they can communicate with macroglial cells and neurons and with cells of the immune system. Likewise, microglial cells express receptors classically described for brain-specific communication such as neurotransmitter receptors and those first discovered as immune cell-specific such as for cytokines. Microglial cells are considered the most susceptible sensors of brain pathology. Upon any detection of signs for brain lesions or nervous system dysfunction, microglial cells undergo a complex, multistage activation process that converts them into the "activated microglial cell." This cell form has the capacity to release a large number of substances that can act detrimental or beneficial for the surrounding cells. Activated microglial cells can migrate to the site of injury, proliferate, and phagocytose cells and cellular compartments.


Asunto(s)
Microglía/fisiología , Animales , Evolución Biológica , Encéfalo/citología , Encéfalo/fisiología , Señalización del Calcio/fisiología , Movimiento Celular , Sistema Nervioso Central/citología , Sistema Nervioso Central/fisiología , Quimiocinas/fisiología , Citocinas/fisiología , Humanos , Canales Iónicos/fisiología , Microglía/ultraestructura , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Fagocitosis/fisiología , Receptores de Neurotransmisores/fisiología , Receptores de Reconocimiento de Patrones/fisiología , Transducción de Señal/fisiología
7.
J Pharmacol Sci ; 138(2): 123-130, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30360946

RESUMEN

Sodium salt of deoxyribonucleic acid (DNA), Derinat, isolated from the soft roes of Russian sturgeon, has been utilized as an immunomodulator for the treatment of reactive oxygen species (ROS)-associated diseases in clinics. Here we show that treatment with Derinat has an anti-inflammatory and anti-oxidative effects on cutaneous ischemia-reperfusion (IR) injury in pressure ulcer (PU) model mice. Dorsal skin damage and dermal edema in mild PU model mice were attenuated by treatment with Derinat. Immunohistochemical and biochemical analyses showed that Derinat suppressed IR-induced oxidative damage, i.e. accumulation of 8-hydroxy-2'-deoxyguanosine (8-OHdG), and related inflammatory factors such as cyclooxygenase 2 (COX-2) and IL-6 receptor (IL-6R) in dorsal skin from PU model mice. We also verified that phospholyated/non-phosphorylated ratio of extracellular signal-regulated kinase (Erk) and p38 mitogen-activated protein kinase (MAPK) increased after IR, which were attenuated by Derinat. We then compared the effect of Derinat with that of salmon DNA and other PU therapeutic agents, prostaglandin E1 (PGE1) and basic fibroblast growth factor (bFGF), by using severe PU model mice. The effects of Derinat and salmon-DNA were compatible with those of PGE1 and bFGF. These results suggest that Derinat other fish-derived DNA formulation could be effective enough and become intriguing new therapeutic options.


Asunto(s)
ADN/farmacología , ADN/uso terapéutico , Presión/efectos adversos , Daño por Reperfusión/complicaciones , Úlcera Cutánea/tratamiento farmacológico , Úlcera Cutánea/etiología , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Ciclooxigenasa 2/metabolismo , ADN/aislamiento & purificación , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Peces/metabolismo , Mediadores de Inflamación/metabolismo , Ratones Endogámicos BALB C , Receptores de Interleucina-6/metabolismo , Piel/metabolismo , Úlcera Cutánea/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Neurochem Res ; 42(9): 2658-2665, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28462451

RESUMEN

Molecular hydrogen (H2), as a new medical gas, has protective effects in neurological disorders including Parkinson's disease (PD). In our previous report, the neuroprotective effect of drinking water with saturated H2 (H2 water) in PD mice might be due to stomach-brain interaction via release of gastric hormone, ghrelin. In the present study, we assessed the effect of H2-induced ghrelin more precisely. To confirm the contribution of ghrelin in H2 water-drinking PD model mice, ghrelin-knock out (KO) mice were used. Despite the speculation, the effect of H2 water was still observed in ghrelin-KO PD model mice. To further check the involvement of ghrelin, possible contribution of ghrelin-induced vagal afferent effect was tested by performing subdiaphragmatic vagotomy before treating with H2 water and administration of MPTP (1-methyl- 4-phenyl-1,2,3,6-tetrahydropyridine). The protective effect of H2 water was still observed in the vagotomized mice in substantia nigra, suggesting that stimulation of vagal afferent nerves is not involved in H2-induced neuroprotection. Other neuroprotective substitutes in ghrelin-KO mice were speculated because H2-induced neuroprotection was not cancelled by ghrelin receptor antagonist, D-Lys3 GHRP-6, in ghrelin-KO PD model mice, unlike in wild-type PD model mice. Our results indicate that ghrelin may not be the only factor for H2-induced neuroprotection and other factors can substitute the role of ghrelin when ghrelin is absent, raising intriguing options of research for H2-responsive factors.


Asunto(s)
Encéfalo/metabolismo , Deuterio/administración & dosificación , Mucosa Gástrica/metabolismo , Ghrelina/deficiencia , Trastornos Parkinsonianos/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Ghrelina/antagonistas & inhibidores , Ghrelina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/patología , Estómago/efectos de los fármacos , Vagotomía/métodos , Nervio Vago/metabolismo , Nervio Vago/cirugía
9.
Glia ; 63(5): 906-20, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25643925

RESUMEN

L-tri-iodothyronine (3, 3', 5-triiodothyronine; T3) is an active form of the thyroid hormone (TH) essential for the development and function of the CNS. Though nongenomic effect of TH, its plasma membrane-bound receptor, and its signaling has been identified, precise function in each cell type of the CNS remained to be investigated. Clearance of cell debris and apoptotic cells by microglia phagocytosis is a critical step for the restoration of damaged neuron-glia networks. Here we report nongenomic effects of T3 on microglial functions. Exposure to T3 increased migration, membrane ruffling and phagocytosis of primary cultured mouse microglia. Injection of T3 together with stab wound attracted more microglia to the lesion site in vivo. Blocking TH transporters and receptors (TRs) or TRα-knock-out (KO) suppressed T3-induced microglial migration and morphological change. The T3-induced microglial migration or membrane ruffling was attenuated by inhibiting Gi /o -protein as well as NO synthase, and subsequent signaling such as phosphoinositide 3-kinase (PI3K), mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK). Inhibitors for Na(+) /K(+) -ATPase, reverse mode of Na(+) /Ca(2+) exchanger (NCX), and small-conductance Ca(2+) -dependent K(+) (SK) channel also attenuated microglial migration or phagocytosis. Interestingly, T3-induced microglial migration, but not phagocytosis, was dependent on GABAA and GABAB receptors, though GABA itself did not affect migratory aptitude. Our results demonstrate that T3 modulates multiple functional responses of microglia via multiple complex mechanisms, which may contribute to physiological and/or pathophysiological functions of the CNS.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Microglía/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Triyodotironina/farmacología , Adenosina Trifosfato/farmacología , Adyuvantes Farmacéuticos/farmacología , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/fisiología , Probenecid/farmacología , Receptores de Hormona Tiroidea/deficiencia , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Transducción de Señal/efectos de los fármacos , Tiroxina/farmacología
10.
Molecules ; 20(11): 20297-311, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26569211

RESUMEN

Ultraviolet-B (UVB) is one of the most cytotoxic and mutagenic stresses that contribute to skin damage and aging through increasing intracellular Ca(2+) and reactive oxygen species (ROS). Derinat (sodium deoxyribonucleate) has been utilized as an immunomodulator for the treatment of ROS-associated diseases in clinics. However, the molecular mechanism by which Derinat protects skin cells from UVB-induced damage is poorly understood. Here, we show that Derinat significantly attenuated UVB-induced intracellular ROS production and decreased DNA damage in primary skin cells. Furthermore, Derinat reduced intracellular ROS, cyclooxygenase-2 (COX-2) expression and DNA damage in the skin of the BALB/c-nu mice exposed to UVB for seven days in vivo. Importantly, Derinat blocked the transient receptor potential canonical (TRPC) channels (TRPCs), as demonstrated by calcium imaging. Together, our results indicate that Derinat acts as a TRPCs blocker to reduce intracellular ROS production and DNA damage upon UVB irradiation. This mechanism provides a potential new application of Derinat for the protection against UVB-induced skin damage and aging.


Asunto(s)
ADN/farmacología , Sustancias Protectoras/farmacología , Piel/efectos de los fármacos , Piel/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Daño del ADN/efectos de los fármacos , Expresión Génica , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Piel/patología , Piel/efectos de la radiación , Rayos Ultravioleta/efectos adversos
11.
Eur J Neurosci ; 40(8): 3253-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25040499

RESUMEN

We previously reported that an intraperitoneal (i.p.) injection of synthetic double-stranded RNA, polyriboinosinic:polyribocytidylic acid (poly-I:C), produced prolonged fatigue in rats, which might serve as a model for chronic fatigue syndrome. The poly-I:C-induced fatigue was associated with serotonin transporter (5-HTT) overexpression in the prefrontal cortex (PFC), a brain region that has been suggested to be critical for fatigue sensation. In the present study, we demonstrated that microglial activation in the PFC was important for poly-I:C-induced fatigue in rats, as pretreatment with minocycline, an inhibitor of microglial activation, prevented the decrease in running wheel activity. Poly-I:C injection increased the microglial interleukin (IL)-1ß expression in the PFC. An intracerebroventricular (i.c.v.) injection of IL-1ß neutralising antibody limited the poly-I:C-induced decrease in activity, whereas IL-1ß (i.c.v.) reduced the activity in a dose-dependent manner. 5-HTT expression was enhanced by IL-1ß in primary cultured astrocytes but not in microglia. Poly-I:C injection (i.p.) caused an increase in 5-HTT expression in astrocytes in the PFC of the rat, which was inhibited by pretreatment with minocycline (i.p.) and rat recombinant IL-1 receptor antagonist (i.c.v.). Poly-I:C injection (i.p.) led to a breakdown of the blood-brain barrier and enhanced Toll-like receptor 3 signaling in the brain. Furthermore, direct application of poly-I:C enhanced IL-1ß expression in primary microglia. We therefore propose that poly-I:C-induced microglial activation, which may be at least partly caused by a direct action of poly-I:C, enhances IL-1ß expression. Then, IL-1ß induces 5-HTT expression in astrocytes, resulting in the immunologically induced fatigue.


Asunto(s)
Fatiga/inmunología , Interleucina-1beta/metabolismo , Microglía/metabolismo , Corteza Prefrontal/inmunología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Fatiga/inducido químicamente , Masculino , Microglía/efectos de los fármacos , Minociclina/farmacología , Actividad Motora/efectos de los fármacos , Poli I-C/toxicidad , Corteza Prefrontal/metabolismo , Ratas , Ratas Wistar , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
12.
Neurosci Lett ; : 137885, 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38914276

RESUMEN

To investigate the precise mechanism of xenon (Xe), pharmacologically isolated AMPA/KA and NMDA receptor-mediated spontaneous (s) and evoked (e) excitatory postsynaptic currents (s/eEPSCAMPA/KA and s/eEPSCNMDA) were recorded from mechanically isolated single spinal sacral dorsal commissural nucleus (SDCN) neurons attached with glutamatergic nerve endings (boutons) using conventional whole-cell patch-clamp technique. We analysed kinetic properties of both s/eEPSCAMPA/KA and s/eEPSCNMDA by focal single- and/or paired-pulse electrical stimulation to compare them. The s/eEPSCNMDA showed smaller amplitude, slower rise time, and slower 1/e decay time constant (τDecay) than those of s/eEPSCAMPA/KA. We previously examined how Xe modulates s/eEPSCAMPA/KA, therefore, examined the effects on s/eEPSCNMDA in the present study. Xe decreased the frequency and amplitude of sEPSCNMDA, and decreased the amplitude but increased the failure rate and paired-pulse ratio of eEPSCNMDA without affecting their τDecay. It was concluded that Xe might suppress NMDA receptor-mediated synaptic transmission via both presynaptic and postsynaptic mechanisms.

13.
Glia ; 61(6): 881-91, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23468421

RESUMEN

Microglia express AMPA (α-amino-hydroxy-5-methyl-isoxazole-4-propionate)-type of glutamate (Glu) receptors (AMPAR), which are highly Ca(2+) impermeable due to the expression of GluA2. However, the functional importance of AMPAR in microglia remains to be investigated, especially under pathological conditions. As low expression of GluA2 was reported in some neurodegenerative diseases, GluA2(-/-) mice were used to show the functional change of microglial AMPARs in response to Glu or kainate (KA). Here we found that Glu-induced currents in the presence of 100 µM cyclothiazide, an inhibitor of AMPAR desensitization, showed time-dependent decrease after activation of microglia with lipopolysaccharide (LPS) in GluA2(+/+) microglia, but not in GluA2(-/-) microglia. Upon activation of microglia, expression level of GluA2 subunits significantly increased, while expression of GluA1, A3 and A4 subunits on membrane surface significantly decreased. These results suggest that nearly homomeric GluA2 subunits were the main reason for low conductance of AMPAR in activated microglia. Increased expression of GluA2 in microglia was also detected partially in brain slices from LPS-injected mice. Cultured microglia from GluA2(-/-) mice showed higher Ca(2+) -permeability, consequently inducing significant increase in the release of proinflammatory cytokine, such as TNF-α. The conditioning medium from KA-treated GluA2(-/-) microglia had more neurotoxic effect on wild type cultured neurons than that from KA-treated GluA2(+/+) microglia. These results suggest that membrane translocation of GluA2-containing AMPARs in activated microglia has functional importance and thus, dysfunction or decreased expression of GluA2 may accelerate Glu neurotoxicity via excess release of proinflammatory cytokines from microglia.


Asunto(s)
Microglía/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Animales , Calcio/metabolismo , Genotipo , Ácido Glutámico/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Transporte de Proteínas , Receptores AMPA/genética , Factor de Necrosis Tumoral alfa/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
14.
Biochem Biophys Res Commun ; 436(3): 455-61, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23747724

RESUMEN

Microglia, which constitute the resident macrophages of the central nervous system (CNS), are generally considered as the primary immune cells in the brain and spinal cord. Microglial cells respond to various factors which are produced following nerve injury of multiple aetiologies and contribute to the development of neuronal disease. Chemokine (C-C motif) ligand 1 (CCL-1), a well-characterized chemokine secreted by activated T cells, has been shown to play an important role in neuropathic pain induced by nerve injury and is also produced in various cell types in the CNS, especially in dorsal root ganglia (DRG). However, the role of CCL-1 in the CNS and the effects on microglia remains unclear. Here we showed the multiple effects of CCL-1 on microglia. We first showed that CCR-8, a specific receptor for CCL-1, was expressed on primary cultured microglia, as well as on astrocytes and neurons, and was upregulated in the presence of CCL-1. CCL-1 at concentration of 1 ng/ml induced chemotaxis, increased motility at a higher concentration (100 ng/ml), and increased proliferation and phagocytosis of cultured microglia. CCL-1 also activated microglia morphologically, promoted mRNA levels for brain-derived neurotrophic factor (BDNF) and IL-6, and increased the release of nitrite from microglia. These indicate that CCL-1 has a role as a mediator in neuron-glia interaction, which may contribute to the development of neurological diseases, especially in neuropathic pain.


Asunto(s)
Quimiocina CCL1/metabolismo , Regulación de la Expresión Génica , Microglía/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Recuento de Células , Proliferación Celular , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Quimiocina CCL1/farmacología , Quimiotaxis , Interleucina-6/genética , Interleucina-6/metabolismo , Antígeno Ki-67/metabolismo , Ratones , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nitritos/metabolismo , Fagocitosis , Cultivo Primario de Células , ARN Mensajero/genética , ARN Mensajero/metabolismo
15.
Pharmacol Res ; 71: 34-43, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23419834

RESUMEN

Neuroinflammation and oxidative stress are involved in the pathogenesis of neurodegenerative diseases such as Alzheimer's diseases and Parkinson's disease. Naturally derived kavalactones isolated from Piper methysticum (Piperaceae) have been shown to exhibit neuroprotective effects. We have previously reported that a chemically synthesized kavalactone derivative, 2',6'-dichloro-5-methoxymethyl-5,6-dehydrokawain (compound 1) protects against oxidative stress-induced neuronal cell death through activation of Nrf2 signaling. In the present study, we examined the effect of compound 1 on neuroinflammation. In BV2 microglial cells, compound 1 strongly inhibited LPS-stimulated iNOS induction and NO production, but did not affect LPS-stimulated induction of COX2. At 6h after LPS challenge, when iNOS induction was not clearly seen, treatment with LPS or compound 1 alone increased expression of heme oxygenase 1 (HO-1) whose transcription is regulated by Nrf2. When treated with both, compound 1 enhanced LPS-stimulated HO-1 induction, which was more evident at 24h after LPS treatment. Furthermore, LPS-stimulated activation of Nrf2 signaling and nuclear translocation of Nrf2 were potentiated by compound 1. The mechanism by which compound 1 activated Nrf2 signaling was supposed to be a covalent modification of the sulfhydryl groups of Keap1 by an α,ß-unsaturated carbonyl group present in the compound 1. Treatment with hemin, a HO-1 inducer, and with [Ru(CO)3Cl2]2, a CO donor, decreased LPS-stimulated iNOS induction and NO production. In contrast, siRNA-mediated knockdown of HO-1 expression reduced the inhibitory effect of compound 1 on LPS-stimulated iNOS induction and NO production. The compound 1 inhibited LPS-stimulated ERK phosphorylation after LPS treatment. Finally, compound 1 suppressed LPS/IFN-γ-stimulated NO production in primary microglial cells. These results suggest that compound 1 is capable of inhibiting LPS-stimulated iNOS induction and NO production via activation of Nrf2 signaling and HO-1 induction in microglial cells. Taken together, compound 1 has a potential to reduce neuroinflammation as well as oxidative stress in neurodegenerative diseases through activation of Nrf2 signaling.


Asunto(s)
Lactonas/farmacología , Lipopolisacáridos/inmunología , Microglía/citología , Factor 2 Relacionado con NF-E2/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Óxido Nítrico/inmunología , Piper/química , Animales , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Interferón gamma/inmunología , Lactonas/química , Lactonas/aislamiento & purificación , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Factor 2 Relacionado con NF-E2/análisis , Factor 2 Relacionado con NF-E2/metabolismo , Penicilamina/farmacología , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
16.
Nature ; 446(7131): 41-5, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17287729

RESUMEN

CD38, a transmembrane glycoprotein with ADP-ribosyl cyclase activity, catalyses the formation of Ca2+ signalling molecules, but its role in the neuroendocrine system is unknown. Here we show that adult CD38 knockout (CD38-/-) female and male mice show marked defects in maternal nurturing and social behaviour, respectively, with higher locomotor activity. Consistently, the plasma level of oxytocin (OT), but not vasopressin, was strongly decreased in CD38-/- mice. Replacement of OT by subcutaneous injection or lentiviral-vector-mediated delivery of human CD38 in the hypothalamus rescued social memory and maternal care in CD38-/- mice. Depolarization-induced OT secretion and Ca2+ elevation in oxytocinergic neurohypophysial axon terminals were disrupted in CD38-/- mice; this was mimicked by CD38 metabolite antagonists in CD38+/+ mice. These results reveal that CD38 has a key role in neuropeptide release, thereby critically regulating maternal and social behaviours, and may be an element in neurodevelopmental disorders.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Conducta Materna/fisiología , Oxitocina/metabolismo , Conducta Social , ADP-Ribosil Ciclasa 1/deficiencia , ADP-Ribosil Ciclasa 1/genética , Amnesia/genética , Amnesia/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Femenino , Regulación de la Expresión Génica , Humanos , Inyecciones , Masculino , Memoria/fisiología , Ratones , Actividad Motora/fisiología , Oxitocina/administración & dosificación , Oxitocina/sangre , Oxitocina/farmacología , Vasopresinas/sangre
17.
Adv Exp Med Biol ; 961: 289-94, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23224888

RESUMEN

Microglia, the immune cells of the central nervous system (CNS), are busy and vigilant guards of the adult brain, which scan brain parenchyma for damage and activate in response to lesions. Release of danger signals/chemoattractants at the site of damage initiates microglial activation and stimulates migration. The main candidate for a chemoattractant sensed by microglia is adenosine triphosphate (ATP); however, many other substances can have similar effects. Some neuropeptides such as angiotensin II, bradykinin, endothelin, galanin and neurotensin are also chemoattractants for microglia. Among them, bradykinin increases microglial migration using mechanism distinct from that of ATP. Bradykinin-induced migration is controlled by a G(i/o)-protein-independent pathway, while ATP-induced migration involves G(i/o) proteins as well as mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK)-dependent pathway. Galanin was reported to share certain signalling cascades with bradykinin; however, this overlap is only partial. Bradykinin, for example, stimulates Ca(2+) influx through the reversed Na(+)/Ca(2+) exchange (NCX), whereas galanin induces intracellular Ca(2+) mobilization by inositol-3,4,5-trisphosphate (InsP(3))-dependent Ca(2+) release from the intracellular store. These differences in signal cascades indicate that different chemoattractants such as ATP, bradykinin and galanin control distinct microglial functions in pathological conditions such as lesion and inflammation and NCX contributes to a special case of microglial migration.


Asunto(s)
Calcio/metabolismo , Movimiento Celular/fisiología , Microglía/metabolismo , Neuropéptidos/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Fosfatos de Fosfatidilinositol/metabolismo
18.
Adv Exp Med Biol ; 961: 295-305, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23224889

RESUMEN

Astrocytes exhibit their excitability based on variations in cytosolic Ca(2+) levels, which leads to variety of signalling events. Only recently, however, intracellular fluctuations of more abundant cation Na(+) are brought in the limelight of glial signalling. Indeed, astrocytes possess several plasmalemmal molecular entities that allow rapid transport of Na(+) across the plasma membrane: (1) ionotropic receptors, (2) canonical transient receptor potential cation channels, (3) neurotransmitter transporters and (4) sodium-calcium exchanger. Concerted action of these molecules in controlling cytosolic Na(+) may complement Ca(2+) signalling to provide basis for complex bidirectional astrocyte-neurone communication at the tripartite synapse.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio/fisiología , Comunicación Celular/fisiología , Membrana Celular/metabolismo , Neuronas/metabolismo , Sodio/metabolismo , Animales , Astrocitos/citología , Humanos , Transporte Iónico/fisiología , Neuronas/citología , Intercambiador de Sodio-Calcio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
19.
Pharmaceuticals (Basel) ; 16(6)2023 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-37375765

RESUMEN

Knowledge of the biological effects of molecular hydrogen (H2), hydrogen gas, is constantly advancing, giving a reason for the optimism in several healthcare practitioners regarding the management of multiple diseases, including socially significant ones (malignant neoplasms, diabetes mellitus, viral hepatitis, mental and behavioral disorders). However, mechanisms underlying the biological effects of H2 are still being actively debated. In this review, we focus on mast cells as a potential target for H2 at the specific tissue microenvironment level. H2 regulates the processing of pro-inflammatory components of the mast cell secretome and their entry into the extracellular matrix; this can significantly affect the capacity of the integrated-buffer metabolism and the structure of the immune landscape of the local tissue microenvironment. The analysis performed highlights several potential mechanisms for developing the biological effects of H2 and offers great opportunities for translating the obtained findings into clinical practice.

20.
Antioxidants (Basel) ; 12(6)2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37371975

RESUMEN

OBJECTIVE: Molecular hydrogen (H2) exhibits antioxidant, anti-inflammatory and anti-apoptotic effects, and has shown benefits in glucose and lipid metabolism in certain animal metabolic disorder models. However, the potential benefits of H2 treatment in individuals with impaired fasting glucose (IFG) has seldom been studied. This randomized controlled study (RCT) aims to investigate the effects of hydrogen-rich water (HRW) on IFG subjects and explore the underlying mechanism involved. METHODS: Seventy-three patients with IFG were enrolled in a randomized, double-blind, placebo-controlled clinical study. These patients were assigned to receive either 1000 mL per day of HRW or placebo pure water (no H2 infusion) for a duration of eight weeks. Metabolic parameters and fecal gut microbiota were assessed at baseline (week 0) and at week 8. A combined analysis of metabolomics and intestinal microbiota was conducted to investigate the correlation between the effect of H2 on the metabolisms and the diversity of intestinal flora in the IGF patients. RESULTS: Both pure water and HRW demonstrated a significant reduction in fasting blood glucose in IFG patients, with a significant difference between pure water and HRW after eight weeks. Among IFG patients with abnormal pre-experimental fatty liver, 62.5% (10/16) in the HRW group and 31.6% (6/19) in the pure water group achieved remission. Furthermore, 16S RNA analysis revealed HRW-modified gut microbiota dysbiosis in the fecal samples of IGF patients. Through Pearson correlation analysis, the differential gut microbiota obtained by 16S analysis was found to be highly correlated with nine metabolites. CONCLUSION: H2 slightly improved metabolic abnormalities and gut microbiota dysbiosis, providing a novel target and theoretical basis for the prevention and treatment of blood glucose regulation in patients with IFG.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA