Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Ginseng Res ; 47(4): 561-571, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37397414

RESUMEN

Background: Escalating evidence shows that ginseng possesses an antiaging potential with cognitive enhancing activity. As mountain cultivated ginseng (MCG) is cultivated without agricultural chemicals, MCG has emerged as a popular herb medicine. However, little is known about the MCG-mediated pharmacological mechanism on brain aging. Methods: As we demonstrated that glutathione peroxidase (GPx) is important for enhancing memory function in the animal model of aging, we investigated the role of MCG as a GPx inducer using GPx-1 (a major type of GPx) knockout (KO) mice. We assessed whether MCG modulates redox and cholinergic parameters, and memory function in aged GPx-1 knockout KOmice. Results: Redox burden of aged GPx-1 KO mice was more evident than that of aged wild-type (WT) mice. Alteration of Nrf2 DNA binding activity appeared to be more evident than that of NFκB DNA binding activity in aged GPx-1 KO mice. Alteration in choline acetyltransferase (ChAT) activity was more evident than that in acetylcholine esterase activity. MCG significantly attenuated reductions in Nrf2 system and ChAT level. MCG significantly enhanced the co-localization of Nrf2-immunoreactivity and ChAT-immunoreactivity in the same cell population. Nrf2 inhibitor brusatol significantly counteracted MCG-mediated up-regulation in ChAT level and ChAT inhibition (by k252a) significantly reduced ERK phosphorylation by MCG, suggesting that MCG might require signal cascade of Nrf2/ChAT/ERK to enhance cognition. Conclusion: GPx-1 depletion might be a prerequisite for cognitive impairment in aged animals. MCG-mediated cognition enhancement might be associated with the activations of Nrf2, ChAT, and ERK signaling cascade.

2.
Food Chem Toxicol ; 173: 113627, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36682417

RESUMEN

Ginsenoside Re (GRe) upregulates anti-aging klotho by mainly upregulating glutathione peroxidase-1 (GPx-1). However, the anti-aging mechanism of GPx-1 remains elusive. Here we investigated whether the GRe-mediated upregulation of GPx-1 modulates oxidative and proinflammatory insults. GPx-1 gene depletion altered redox homeostasis and platelet-activating factor receptor (PAFR) and nuclear factor kappa B (NFκB) expression, whereas the genetic overexpression of GPx-1 or GRe mitigated this phenomenon in aged mice. Importantly, the NFκB inhibitor pyrrolidine dithiocarbamate (PDTC) did not affect PAFR expression, while PAFR inhibition (i.e., PAFR knockout or ginkgolide B) significantly attenuated NFκB nuclear translocation, suggesting that PAFR could be an upstream molecule for NFκB activation. Iba-1-labeled microgliosis was more underlined in aged GPx-1 KO than in aged WT mice. Triple-labeling immunocytochemistry showed that PAFR and NFκB immunoreactivities were co-localized in Iba-1-positive populations in aged mice, indicating that microglia released these proteins. GRe inhibited triple-labeled immunoreactivity. The microglial inhibitor minocycline attenuated aging-related reduction in phospho-ERK. The effect of minocycline was comparable with that of GRe. GRe, ginkgolide B, PDTC, or minocycline also attenuated aging-evoked memory impairments. Therefore, GRe ameliorated aging-associated memory impairments in the absence of GPx-1 by inactivating oxidative insult, PAFR, NFkB, and microgliosis.


Asunto(s)
Glutatión Peroxidasa GPX1 , FN-kappa B , Ratones , Animales , FN-kappa B/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Minociclina/metabolismo , Minociclina/farmacología , Ratones Noqueados , Hipocampo
3.
Drug Chem Toxicol ; 46(2): 281-296, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35707918

RESUMEN

It has been recognized that serotonergic blocker showed serious side effects, and that ginsenoside modulated serotonergic system with the safety. However, the effects of ginsenoside on serotonergic impairments remain to be clarified. Thus, we investigated ginsenoside Re (GRe), a major bioactive component in the mountain-cultivated ginseng on (±)-8-hydroxy-dipropylaminotetralin (8-OH-DPAT), a 5-HT1A receptor agonist. In the present study, we observed that the treatment with GRe resulted in significant inhibition of protein kinase C δ (PKCδ) phosphorylation induced by the 5-HT1A receptor agonist (±)-8-hydroxy-dipropylaminotetralin (8-OH-DPAT) in the hypothalamus of the wild-type (WT) mice. The inhibition of GRe was comparable with that of the PKCδ inhibitor rottlerin or the 5-HT1A receptor antagonist WAY100635 (WAY). 8-OH-DPAT-induced significant reduction in nuclear factor erythroid-2-related factor 2 (Nrf2)-related system (i.e., Nrf2 DNA binding activity, γ-glutamylcysteine ligase modifier (GCLm) and γ-glutamylcysteine ligase catalytic (GCLc) mRNA expression, and glutathione (GSH)/oxidized glutathione (GSSG) ratio) was significantly attenuated by GRe, rottlerin, or WAY in WT mice. However, PKCδ gene knockout significantly protected the Nrf2-dependent system from 8-OH-DPAT insult in mice. Increases in 5-hydroxytryptophan (5-HT) turnover rate, overall serotonergic behavioral score, and hypothermia induced by 8-OH-DPAT were significantly attenuated by GRe, rottlerin, or WAY in WT mice. Consistently, PKCδ gene knockout significantly attenuated these parameters in mice. However, GRe or WAY did not provide any additional positive effects on the serotonergic protective potential mediated by PKCδ gene knockout in mice. Therefore, our results suggest that PKCδ is an important mediator for GRe-mediated protective activity against serotonergic impairments/oxidative burden caused by the 5-HT1A receptor.


Asunto(s)
Ginsenósidos , Ratones , Animales , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Ginsenósidos/farmacología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Receptor de Serotonina 5-HT1A/genética , Glutatión , Disulfuro de Glutatión , Antagonistas de la Serotonina , Ligasas
4.
Free Radic Biol Med ; 189: 2-19, 2022 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-35840016

RESUMEN

Ginseng is known to possess anti-aging potential. Klotho mutant mice exhibit phenotypes that resemble the phenotype of the human aging process. Similar to Klotho deficient mice, patients with chronic kidney disease (CKD) suffer vascular damage and cognitive impairment, which might upregulate the angiotensin II AT1 receptor. Since AT1 receptor expression was more pronounced than endothelin ET-1 expression in the hippocampus of aged Klotho deficient (±) mice, we focused on the AT1 receptor in this study. Ginsenoside Re (GRe), but not ginsenoside Rb1 (GRb1), significantly attenuated the increase in AT1 receptor expression in aged Klotho deficient mice. Both GRe and the AT1 receptor antagonist losartan failed to attenuate the decrease in phosphorylation of JAK2/STAT3 in aged Klotho deficient (±) mice but significantly activated nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated signaling. Both GRe and losartan attenuated the increased NADPH oxidase (NOX) activity and reactive oxygen species (ROS) in aged Klotho deficient mice. Furthermore, of all the antioxidant enzymes, GRe significantly increased glutathione peroxidase (GPx) activity. GRe significantly attenuated the reduced phosphorylation of ERK and CREB in GPx-1 knockout mice; however, genetic overexpression of GPx-1 did not significantly affect them in aged mice. Klotho-, Nrf2-, and GPx-1-immunoreactivities were co-localized in the same cells of the hippocampus in aged Klotho wild-type mice. Both the GPx inhibitor mercaptosuccinate and Nrf2 inhibitor brusatol counteracted the effects of GRe on all neurobehavioral impairments in aged Klotho deficient (±) mice. Our results suggest that GRe attenuates all alterations, such as AT1 receptor expression, NOX-, ROS-, and GPx-levels, and cognitive dysfunction in aged Klotho deficient (±) mice via upregulation of Nrf2/GPx-1/ERK/CREB signaling.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Receptor de Angiotensina Tipo 1 , Animales , Ratones , Angiotensina II , Antioxidantes/farmacología , Ginsenósidos , Glutatión Peroxidasa , Glutatión Peroxidasa GPX1 , Proteínas Klotho , Losartán/farmacología , Trastornos de la Memoria , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno
5.
Food Chem Toxicol ; 158: 112657, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34740715

RESUMEN

Dextromethorphan (DM) abuse produces mania-like symptoms in humans. ERK/Akt signaling activation involved in manic potential can be attenuated by the inhibition of ouabain-like cardiac steroids. In this study, increased phosphorylations of ERK/Akt and hyperlocomotion induced by DM (30 mg/kg, i.p./day × 7) were significantly protected by the ouabain inhibitor rostafuroxin (ROSTA), suggesting that DM induces the manic potential. ROSTA significantly attenuated DM-induced protein kinase C δ (PKCδ) phosphorylation, GluN2B (i.e., MDA receptor subunit) expression, and phospho-PKCδ/GluN2B interaction. DM instantly upregulated the nuclear factor erythroid-2-related factor 2 (Nrf2)-dependent system. However, DM reduced Nrf2 nuclear translocation, Nrf2 DNA binding activity, γ-glutamylcysteine mRNA expression, and subsequent GSH/GSSG level and enhanced oxidative parameters following 1-h of administration. ROSTA, PKCδ inhibitor rottlerin, and GluN2B inhibitor traxoprodil significantly attenuated DM-induced alterations in Nrf2-related redox parameters and locomotor activity induced by DM in wild-type mice. Importantly, in PKCδ knockout mice, DM failed to alter the above parameters. Further, ROSTA and traxoprodil also failed to enhance PKCδ depletion effect, suggesting that PKCδ is a critical target for the anti-manic potential of ROSTA or GluN2B antagonism. Our results suggest that ROSTA inhibits DM-induced manic potential by attenuating ERK/Akt activation, GluN2B/PKCδ signalings, and Nrf2-dependent system.


Asunto(s)
Androstanoles/farmacología , Trastorno Bipolar , Dextrometorfano/efectos adversos , Ouabaína/antagonistas & inhibidores , Animales , Trastorno Bipolar/inducido químicamente , Trastorno Bipolar/metabolismo , Modelos Animales de Enfermedad , Locomoción/efectos de los fármacos , Masculino , Ratones , Transducción de Señal/efectos de los fármacos
6.
Arch Pharm Res ; 44(7): 668-688, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34286473

RESUMEN

Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.


Asunto(s)
Cuerpo Estriado/patología , Neuronas Dopaminérgicas/efectos de los fármacos , Metanfetamina/toxicidad , Enfermedad de Parkinson Secundaria/patología , Animales , Apoptosis/efectos de los fármacos , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/citología , Humanos , Metanfetamina/administración & dosificación , Ratones , Dinámicas Mitocondriales/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas
7.
Int J Mol Sci ; 22(13)2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-34281274

RESUMEN

It has been recognized that serotonin 2A receptor (5-HT2A) agonist 2,5-dimethoxy-4-iodo-amphetamine (DOI) impairs serotonergic homeostasis. However, the mechanism of DOI-induced serotonergic behaviors remains to be explored. Moreover, little is known about therapeutic interventions against serotonin syndrome, although evidence suggests that ginseng might possess modulating effects on the serotonin system. As ginsenoside Re (GRe) is well-known as a novel antioxidant in the nervous system, we investigated whether GRe modulates 5-HT2A receptor agonist DOI-induced serotonin impairments. We proposed that protein kinase Cδ (PKCδ) mediates serotonergic impairments. Treatment with GRe or 5-HT2A receptor antagonist MDL11939 significantly attenuated DOI-induced serotonergic behaviors (i.e., overall serotonergic syndrome behaviors, head twitch response, hyperthermia) by inhibiting mitochondrial translocation of PKCδ, reducing mitochondrial glutathione peroxidase activity, mitochondrial dysfunction, and mitochondrial oxidative stress in wild-type mice. These attenuations were in line with those observed upon PKCδ inhibition (i.e., pharmacologic inhibitor rottlerin or PKCδ knockout mice). Furthermore, GRe was not further implicated in attenuation mediated by PKCδ knockout in mice. Our results suggest that PKCδ is a therapeutic target for GRe against serotonergic behaviors induced by DOI.


Asunto(s)
Ginsenósidos/farmacología , Proteína Quinasa C-delta/metabolismo , Antagonistas de la Serotonina/farmacología , Síndrome de la Serotonina/prevención & control , Acetofenonas/farmacología , Anfetaminas/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Benzopiranos/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Piperidinas/farmacología , Proteína Quinasa C-delta/deficiencia , Proteína Quinasa C-delta/genética , Inhibidores de Proteínas Quinasas/farmacología , Serotonina/fisiología , Agonistas de Receptores de Serotonina/farmacología , Síndrome de la Serotonina/inducido químicamente , Síndrome de la Serotonina/fisiopatología
8.
Food Chem Toxicol ; 148: 111945, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33359022

RESUMEN

Glutathione peroxidase (GPx) acts in co-ordination with other signaling molecules to exert its own antioxidant role. We have demonstrated the protective effects of GPx,/GPx-1, a selenium-dependent enzyme, on various neurodegenerative disorders (i.e., Parkinson's disease, Alzheimer's disease, cerebral ischemia, and convulsive disorders). In addition, we summarized the recent findings indicating that GPx-1 might play a role as a neuromodulator in neuropsychiatric conditions, such as, stress, bipolar disorder, schizophrenia, and drug intoxication. In this review, we attempted to highlight the mechanistic scenarios mediated by the GPx/GPx-1 gene in impacting these neurodegenerative and neuropsychiatric disorders, and hope to provide new insights on the therapeutic interventions against these disorders.


Asunto(s)
Glutatión Peroxidasa/metabolismo , Trastornos Mentales/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Neuroprotección/fisiología , Animales , Azoles/uso terapéutico , Glutatión Peroxidasa/genética , Humanos , Rayos Infrarrojos , Isoindoles , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/terapia , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/terapia , Óxido Nítrico Sintasa de Tipo III/metabolismo , Compuestos de Organoselenio/uso terapéutico , Fototerapia , Proteína Quinasa C-delta/metabolismo , Receptor Muscarínico M1/metabolismo , Regulación hacia Arriba/efectos de la radiación , Glutatión Peroxidasa GPX1
9.
Curr Mol Pharmacol ; 14(2): 200-209, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32329705

RESUMEN

Attention deficit hyperactivity disorder (ADHD) is a psychiatric disorder commonly found in children, which is recognized by hyperactivity and aggressive behavior. It is known that the pathophysiology of ADHD is associated with neurobiological dysfunction. Although psychostimulants are recognized as the therapeutic drugs of choice for ADHD patients, the side effects might be of great concern. Ginkgo biloba is a promising herbal, complementary supplement that may modulate the neuronal system in an ADHD-like condition. The beneficial effect of Ginkgo biloba on ADHD-like symptoms may be related to the modulation of the system by novel molecular mechanisms. Ginkgo biloba is known to modulate dopamine, serotonin, and norepinephrine signaling. Flavonoid glycosides and terpene trilactones are the two major phytochemical components present in the Ginkgo biloba preparations, which can exhibit antioxidant and neuroprotective activities. The pharmacological mechanisms of the phytochemical components may also contribute to the neuroprotective activity of Ginkgo biloba. In this review, we have summarized recent findings on the potential of various Ginkgo biloba preparations to treat ADHD-like symptoms. In addition, we have discussed the pharmacological mechanisms mediated by Ginkgo biloba against an ADHD-like condition.


Asunto(s)
Antioxidantes/química , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Ginkgo biloba/química , Fármacos Neuroprotectores/química , Extractos Vegetales/farmacología , Animales , Antioxidantes/farmacología , Dopamina/metabolismo , Descubrimiento de Drogas , Flavonoides/química , Glicósidos/química , Humanos , Fármacos Neuroprotectores/farmacología , Norepinefrina/metabolismo , Serotonina/metabolismo , Transducción de Señal , Terpenos/química
10.
Free Radic Res ; 55(1): 11-25, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33222572

RESUMEN

In the present study, we examined whether glutathione peroxidase-1 (GPx-1), a major H2O2 scavenger in the brain, affects memory deficits induced by Aß (1-42) in mice. Treatment with 400 pmol/5 µl Aß (1-42) (i.c.v.) resulted in a reduction of GPx-1 expression in wild-type (WT) mice. An Aß (1-42)-induced reduction in acetylcholine (ACh) level was observed in the hippocampus. Treatment with Aß (1-42) consistently resulted in reduced expression and activity of choline acetyltransferase (ChAT) and in an increase in expression and activity of acetylcholinesterase (AChE). Upon examining each of the muscarinic acetylcholine receptors (mAChRs) and nicotinic AChRs, we noted that Aß (1-42) treatment selectively reduced the levels of M1 mAChR. In addition, Aß (1-42) induced a significant reduction in phospho-cAMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) expression. The cholinergic impairments induced by Aß (1-42) were more pronounced in GPx-1 knockout mice than in WT mice. Importantly, an adenoviral vector encoded with the GPx-1 gene (Ad-GPx-1) significantly rescued Aß (1-42)-induced cholinergic impairments in GPx-1 knockout mice. In addition, M1 mAChR antagonist dicyclomine significantly counteracted Ad-GPx-1-mediated increases in p-CREB and BDNF expression, as well as memory-enhancing effects in GPx-1 knockout mice, thus indicating that M1 mAChR might be a critical mediator for the rescue effects of Ad-GPx-1. Combined, our results suggest that GPx-1 gene protected against Aß (1-42)-induced memory impairments via activation of M1 mAChR-dependent CREB/BDNF signalling.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Glutatión Peroxidasa/genética , Trastornos de la Memoria/inducido químicamente , Fragmentos de Péptidos/farmacología , Receptor Muscarínico M1/metabolismo , Acetilcolina/metabolismo , Adenoviridae/genética , Animales , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Glutatión Peroxidasa/administración & dosificación , Glutatión Peroxidasa/biosíntesis , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Trastornos de la Memoria/genética , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Glutatión Peroxidasa GPX1
11.
Neurochem Res ; 45(12): 2991-3002, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33064252

RESUMEN

A growing body evidence suggests that selenium (Se) deficiency is associated with an increased risk of developing Alzheimer's disease (AD). Se-dependent glutathione peroxidase-1 (GPx-1) of a major antioxidant enzyme, and the most abundant isoform of GPx in the brain. In the present study, we investigated whether GPx-1 is protective against memory impairments induced by beta-amyloid (Aß) (1-42) in mice. As the alteration of protein kinase C (PKC)-mediated ERK activation was recognized in the early stage of AD, we examined whether the GPx-1 gene modulates Aß (1-42)-induced changes in PKC and ERK levels. We observed that Aß (1-42) treatment (400 pmol, i.c.v.) significantly decreased PKC ßII expression in the hippocampus of mice. Aß (1-42)-induced neurotoxic changes [i.e., oxidative stress (i.e., reactive oxygen species, 4-hydroxy-2-noneal, and protein carbonyl), reduced PKC ßII and phospho-ERK expressions, and memory impairment under Y-maze and passive avoidance test] were more pronounced in GPx-1 knockout than in wild type mice. Importantly, exposure to a GPx-1 gene-encoded adenovirus vector (Adv-GPx-1) significantly increased GPx-1 mRNA and GPx activity in the hippocampus of GPx-1 knockout mice. Adv-GPx-1 exposure also significantly blocked the neurotoxic changes induced by Aß (1-42) in GPx-1 knockout mice. Treatment with ERK inhibitor U0126 did not significantly change Adv-GPx-1-mediated attenuation in PKC ßII expression. In contrast, treatment with PKC inhibitor chelerythrine (CHE) reversed Adv-GPx-1-mediated attenuation in ERK phosphorylation, suggesting that PKC ßII-mediated ERK signaling is important for Adv-GPx-1-mediated potentials against Aß (1-42) insult. Our results suggest that treatment with the antioxidant gene GPx-1 rescues Aß (1-42)-induced memory impairment via activating PKC ßII-mediated ERK signaling.


Asunto(s)
Glutatión Peroxidasa/deficiencia , Glutatión Peroxidasa/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Trastornos de la Memoria/enzimología , Memoria/efectos de los fármacos , Proteína Quinasa C beta/metabolismo , Adenoviridae/genética , Péptidos beta-Amiloides , Animales , Expresión Génica/efectos de los fármacos , Terapia Genética , Glutatión Peroxidasa/genética , Hipocampo/enzimología , Hipocampo/metabolismo , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/genética , Trastornos de la Memoria/terapia , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos , Glutatión Peroxidasa GPX1
12.
Clin Exp Pharmacol Physiol ; 47(5): 790-797, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31883280

RESUMEN

In the present study, we investigated whether mood stabilizer lithium (Li) protects against d-amphetamine (AMP)-induced mania-like behaviours via modulating the novel proinflammatory potential. Repeated treatment with AMP resulted in significant increases in proinflammatory cyclooxygenase-2 (COX-2) and indolemaine-2,3-dioxygenase-1 (IDO)-1 expression in the prefrontal cortex (PFC) of mice. However, AMP treatment did not significantly change IDO-2 and 5-lipoxygenase (5-LOX) expression, suggesting that proinflammatory parameters such as COX-2 and IDO-1 are specific for AMP-induced behaviours. AMP-induced initial expression of COX-2 (15 minutes post-AMP) was earlier than that of IDO-1 (1 hour post-AMP). Mood stabilizer Li and COX-2 inhibitor meloxicam significantly attenuated COX-2 expression 15 minutes post-AMP, whereas IDO-1 inhibitor 1-methyl-DL-tryptophan (1-MT) did not affect COX-2 expression. However, AMP-induced IDO-1 expression was significantly attenuated by Li, meloxicam or 1-MT, suggesting that COX-2 is an upstream molecule for the induction of IDO-1 caused by AMP. Consistently, co-immunoprecipitation between COX-2 and IDO-1 was observed at 30 minutes, 1, 3, and 6 hours after the final AMP treatment. This interaction was also significantly inhibited by Li, meloxicam or 1-MT. Furthermore, AMP-induced hyperlocomotion was significantly attenuated by Li, meloxicam or 1-MT. We report, for the first time, that mood stabilizer Li attenuates AMP-induced mania-like behaviour via attenuation of interaction between COX-2 and IDO-1, and that the interaction of COX-2 and IDO-1 may be critical for the therapeutic intervention mediated by mood stabilizer.


Asunto(s)
Antimaníacos/farmacología , Conducta Animal/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Cloruro de Litio/farmacología , Locomoción/efectos de los fármacos , Manía/prevención & control , Corteza Prefrontal/efectos de los fármacos , Anfetamina , Animales , Inhibidores de la Ciclooxigenasa 2/farmacología , Modelos Animales de Enfermedad , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Masculino , Manía/inducido químicamente , Manía/enzimología , Manía/psicología , Meloxicam/farmacología , Ratones Endogámicos C57BL , Corteza Prefrontal/enzimología , Corteza Prefrontal/fisiopatología , Transducción de Señal , Triptófano/análogos & derivados , Triptófano/farmacología
13.
Curr Neuropharmacol ; 17(10): 990-1002, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30819085

RESUMEN

Far-infrared ray (FIR) is an electromagnetic wave that produces various health benefits against pathophysiological conditions, such as diabetes mellitus, renocardiovascular disorders, stress, and depression etc. However, the therapeutic application on the FIR-mediated protective potentials remains to be further extended. To achieve better understanding on FIR-mediated therapeutic potentials, we summarized additional findings in the present study that exposure to FIR ameliorates stressful condition, memory impairments, drug dependence, and mitochondrial dysfunction in the central nervous system. In this review, we underlined that FIR requires modulations of janus kinase 2 / signal transducer and activator of transcription 3 (JAK2/STAT3), nuclear factor E2- related factor 2 (Nrf-2), muscarinic M1 acetylcholine receptor (M1 mAChR), dopamine D1 receptor, protein kinase C δ gene, and glutathione peroxidase-1 gene for exerting the protective potentials in response to neuropsychotoxic conditions.


Asunto(s)
Antioxidantes/uso terapéutico , Rayos Infrarrojos/uso terapéutico , Enfermedades del Sistema Nervioso/radioterapia , Animales , Humanos , Janus Quinasa 2/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores de Dopamina D1/metabolismo , Transducción de Señal/fisiología
14.
Food Funct ; 10(1): 325-332, 2019 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-30574980

RESUMEN

Theanine (γ-glutamylethylamide), an amino acid in tea, is a putative neuroprotective and antioxidant compound capable of improving lifespan and cognitive function. Because we previously reported cognitive dysfunction in klotho mutant mice via down-regulation of janus kinase 2 (JAK2) and signal transducer and activator of transcription3 (STAT3), M1 muscarinic cholinergic receptor (M1 mAChR), and ERK signaling, we, therefore, investigated whether self-administration of theanine affects memory dysfunction in response to klotho gene depletion in mice, and whether theanine modulates the JAK2/STAT3, M1 mAChR, and ERK signaling network. Theanine significantly attenuated memory impairments in klotho mutant mice. Moreover, theanine self-administration significantly attenuated inhibitions of JAK2/STAT3 phosphorylation, M1 mAChR expression, and ERK1/2 phosphorylation in the hippocampus of klotho mutant mice. Consistently, AG490, a JAK2/STAT3 inhibitor, dicyclomine, an M1 mAChR antagonist, or U0126, an ERK1/2 inhibitor, significantly counteracted theanine-induced attenuation of memory impairment induced by klotho gene depletion in mice. Our study suggests that theanine attenuates memory impairments in a genetic aging model via up-regulation of JAK2/STAT3, M1 mAChR, and ERK signaling.


Asunto(s)
Glucuronidasa/deficiencia , Glutamatos/administración & dosificación , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/genética , Animales , Femenino , Glucuronidasa/genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Proteínas Klotho , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/metabolismo , Ratones , Ratones Noqueados , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
15.
Neurochem Res ; 43(5): 1118-1135, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29687308

RESUMEN

Evidence indicates that stress conditions might lead to drug dependence. Recently, we have demonstrated that exposure to far infrared ray (FIR) attenuates acute restraint stress via induction of glutathione peroxidase-1 (GPx-1) gene. We investigated whether FIR affects methamphetamine (MA)-induced behavioral sensitization and whether FIR-mediated pharmacological activity requires interaction between dopamine receptor and GPx-1 gene. We observed that MA treatment significantly increased GPx-1 expression in the striatum of wild-type (WT) mice. Interestingly, exposure to FIR potentiated MA-induced increase in GPx-1 expression. This phenomenon was also observed in animals receiving MA with dopamine D1 receptor antagonist SCH23390. However, dopamine D2 receptor antagonist sulpiride did not affect MA-induced GPx-1 expression. FIR exposure or SCH23390, but not sulpiride, significantly attenuated MA-induced behavioral sensitization. Exposure to FIR significantly attenuated MA-induced dopamine D1 receptor expression, c-Fos induction and oxidative burdens. FIR-mediated antioxidant effects were also more pronounced in mitochondrial- than cytosolic-fraction. In addition, FIR significantly attenuated against MA-induced changes in mitochondrial superoxide dismutase and mitochondrial GPx activities, mitochondrial transmembrane potential, intramitochondrial Ca2+ level, mitochondrial complex-I activity, and mitochondrial oxidative burdens. The attenuation by FIR was paralleled that by SCH23390. Effects of FIR or SCH23390 were more sensitive to GPx-1 KO than WT mice, while SCH23390 treatment did not exhibit any additive effects on the protective activity mediated by FIR, indicating that dopamine D1 receptor constitutes a molecular target of FIR. Our result suggests that exposure to FIR ameliorates MA-induced behavioral sensitization via possible interaction between dopamine D1 receptor and GPx-1 gene.


Asunto(s)
Conducta Animal/efectos de los fármacos , Conducta Animal/efectos de la radiación , Estimulantes del Sistema Nervioso Central/efectos de la radiación , Estimulantes del Sistema Nervioso Central/toxicidad , Glutatión Peroxidasa/genética , Rayos Infrarrojos , Metanfetamina/efectos de la radiación , Metanfetamina/toxicidad , Mitocondrias/efectos de los fármacos , Mitocondrias/efectos de la radiación , Receptores de Dopamina D1/efectos de los fármacos , Receptores de Dopamina D1/efectos de la radiación , Animales , Antioxidantes/metabolismo , Benzazepinas/farmacología , Antagonistas de Dopamina/farmacología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Glutatión Peroxidasa GPX1
16.
Neurochem Int ; 116: 63-76, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29572053

RESUMEN

We demonstrated that activation of protein kinase Cδ (PKCδ) and inactivation of the glutathione peroxidase-1 (GPx-1)-dependent systems are critical for methamphetamine (MA)-induced recognition memory impairment. We also demonstrated that exposure to far-infrared rays (FIR) causes induction of the glutathione (GSH)-dependent system, including induction of the GPx-1 gene. Here, we investigated whether exposure to FIR rays affects MA-induced recognition memory impairment and whether it modulates PKC, cholinergic receptors, and the GSH-dependent system. Because the PKC activator bryostatin-1 mainly induces PKCα, PKCε, and PKCδ, we assessed expression of these proteins after MA treatment. MA treatment selectively increased PKCδ expression and its phosphorylation. Exposure to FIR rays significantly attenuated MA-induced increases in PKCδ phosphorylation. Importantly, bryostatin-1 potentiated MA-induced phosphorylation of PKCδ. MA treatment significantly decreased M1, M3, and M4 muscarinic acetylcholine receptors (mAChRs) and ß2 nicotinic acetylcholine receptor expression. Of these, the decrease was most pronounced in M1 mAChR. Exposure to FIR significantly attenuated MA-induced decreases in the M1 mAChR and phospho-ERK1/2, while it facilitated Nrf2-dependent GSH induction. Dicyclomine, an M1 mAChR antagonist, and l-buthionine-(S, R)-sulfoximine (BSO), an inhibitor of GSH synthesis, counteracted against the protective potentials mediated by FIR. More importantly, the memory-enhancing potential of FIR rays was significantly counteracted by bryostatin-1, dicyclomine, and BSO. Our results suggest that exposure to FIR rays attenuates MA-induced impairment in recognition memory via up-regulation of M1 mAChR, Nrf2-dependent GSH induction, and ERK1/2 phosphorylation by inhibiting PKCδ phosphorylation by bryostatin-1.


Asunto(s)
Trastornos de la Memoria/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Proteína Quinasa C-delta/efectos de los fármacos , Receptor Muscarínico M1/efectos de los fármacos , Animales , Glutatión Peroxidasa , Trastornos de la Memoria/inducido químicamente , Metanfetamina/farmacología , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Quinasa C-delta/metabolismo , Regulación hacia Arriba , Glutatión Peroxidasa GPX1
17.
J Neurosci Res ; 96(7): 1294-1310, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29476655

RESUMEN

We have previously demonstrated that repeated treatment with methamphetamine (MA) results in a recognition memory impairment via upregulation of protein kinase C (PKC) δ and downregulation of the glutathione peroxidase-1 (GPx-1)-dependent antioxidant system. We also demonstrated that far-infrared ray (FIR) attenuates acute restraint stress via induction of the GPx-1 gene. Herein, we investigated whether exposure to FIR modulates MA-induced recognition memory impairment in male mice, and whether cognitive potentials mediated by FIR require modulation of the PKCδ gene, extracellular signal-regulated kinase (ERK) 1/2, and glutathione-dependent system. Repeated treatment with MA significantly increased PKCδ expression and its phosphorylation out of PKC isoenzymes (i.e., PKCα, PKCßI, PKCßII, PKCζ, and PKCδ expression) in the prefrontal cortex of mice. Exposure to FIR significantly attenuated MA-induced increase in phospho-PKCδ and decrease in phospho-ERK 1/2. In addition, FIR further facilitated the nuclear factor E2-related factor 2 (Nrf2)-dependent glutathione synthetic system. Moreover, L-buthionine-(S, R)-sulfoximine, an inhibitor of glutathione synthesis, counteracted the FIR-mediated phospho-ERK 1/2 induction and memory-enhancing activity against MA insult. More important, positive effects of FIR are comparable to those of genetic depletion of PKCδ or the antipsychotic clozapine. Our results indicate that FIR protects against MA-induced memory impairment via activations of the Nrf2-dependent glutathione synthetic system, and ERK 1/2 signaling by inhibition of the PKCδ gene.


Asunto(s)
Clozapina/farmacología , Rayos Infrarrojos , Memoria/efectos de los fármacos , Memoria/efectos de la radiación , Metanfetamina/efectos de la radiación , Metanfetamina/toxicidad , Proteína Quinasa C-delta/antagonistas & inhibidores , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/efectos de la radiación , Animales , Femenino , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Masculino , Metanfetamina/química , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa C-delta/efectos de la radiación , Glutatión Peroxidasa GPX1
18.
Neurochem Int ; 94: 9-22, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26850477

RESUMEN

Exposure to far-infrared ray (FIR) has been shown to exert beneficial effects on cardiovascular and emotional disorders. However, the precise underlying mechanism mediated by FIR remains undetermined. Since restraint stress induces cardiovascular and emotional disorders, the present study investigated whether exposure to FIR affects acute restraint stress (ARS) in mice. c-Fos-immunoreactivity (IR) was significantly increased in the paraventricular hypothalamic nucleus (PVN) and dorsomedial hypothalamic nucleus (DMH) in response to ARS. The increase in c-Fos-IR parallels that in oxidative burdens in the hypothalamus against ARS. Exposure to FIR significantly attenuated increases in the c-Fos-IR, oxidative burdens and corticosterone level. ARS elicited decreases in GSH/GSSG ratio, cytosolic Cu/Zn-superoxide dismutase (SOD-1), glutathione peroxidase (GPx), and glutathione reductase (GR) activities. FIR-mediated attenuation was particularly observed in ARS-induced decrease in GPx, but not in SOD-1 or GR activity. Consistently, ARS-induced decreases in GPx-1-immunoreactivity in PVN and DMH, and decreases in GPx-1 expression in the hypothalamus were significantly attenuated by FIR. ARS-induced significant increases in phosphorylation of JAK2/STAT3, and nuclear translocation and DNA-binding activity of NFκB were observed in the hypothalamus. Exposure to FIR selectively attenuated phosphorylation of JAK2/STAT3, but did not diminish nuclear translocation and DNA-binding activity of NFκB, suggesting that JAK2/STAT3 constitutes a critical target for FIR-mediated pharmacological potential. ARS-induced increase in c-Fos-IR in the PVN and DMH of non-transgenic mice was significantly attenuated by FIR exposure or JAK2/STAT3 inhibitor AG490. GPx-1 overexpressing transgenic mice significantly protected increases in the c-Fos-IR and corticosterone level induced by ARS. However, neither FIR exposure nor AG490 significantly affected attenuations by genetic overexpression of GPx-1. Moreover, AG490 did not exhibit any additional positive effects against the attenuation by genetic overexpression of GPx-1 or FIR exposure. Our results indicate that exposure to FIR significantly protects ARS-induced increases in c-Fos-IR and oxidative burdens via inhibition of JAK2/STAT3 signaling by induction of GPx-1.


Asunto(s)
Glutatión Peroxidasa/biosíntesis , Rayos Infrarrojos/uso terapéutico , Janus Quinasa 2/antagonistas & inhibidores , Restricción Física/psicología , Factor de Transcripción STAT3/antagonistas & inhibidores , Estrés Psicológico/metabolismo , Animales , Núcleo Hipotalámico Dorsomedial/metabolismo , Núcleo Hipotalámico Dorsomedial/efectos de la radiación , Inducción Enzimática , Glutatión Peroxidasa/efectos de la radiación , Janus Quinasa 2/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción STAT3/efectos de la radiación , Transducción de Señal/fisiología , Transducción de Señal/efectos de la radiación , Estrés Psicológico/prevención & control , Glutatión Peroxidasa GPX1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...