Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pineal Res ; 76(1): e12921, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37846173

RESUMEN

Evidence suggests that the neuroprotective effects of melatonin involve both receptor-dependent and -independent actions. However, little is known about the effects of melatonin receptor activation on the kainate (KA) neurotoxicity. This study examined the effects of repeated post-KA treatment with ramelteon, a selective agonist of melatonin receptors, on neuronal loss, cognitive impairment, and depression-like behaviors following KA-induced seizures. The expression of melatonin receptors decreased in neurons, whereas it was induced in astrocytes 3 and 7 days after seizures elicited by KA (0.12 µg/µL) in the hippocampus of mice. Ramelteon (3 or 10 mg/kg, i.p.) and melatonin (10 mg/kg, i.p.) mitigated KA-induced oxidative stress and impairment of glutathione homeostasis and promoted the nuclear translocation and DNA binding activity of Nrf2 in the hippocampus after KA treatment. Ramelteon and melatonin also attenuated microglial activation but did not significantly affect astroglial activation induced by KA, despite the astroglial induction of melatonin receptors after KA treatment. However, ramelteon attenuated KA-induced proinflammatory phenotypic changes in astrocytes. Considering the reciprocal regulation of astroglial and microglial activation, these results suggest ramelteon inhibits microglial activation by regulating astrocyte phenotypic changes. These effects were accompanied by the attenuation of the nuclear translocation and DNA binding activity of nuclear factor κB (NFκB) induced by KA. Consequently, ramelteon attenuated the KA-induced hippocampal neuronal loss, memory impairment, and depression-like behaviors; the effects were comparable to those of melatonin. These results suggest that ramelteon-mediated activation of melatonin receptors provides neuroprotection against KA-induced neurotoxicity in the mouse hippocampus by activating Nrf2 signaling to attenuate oxidative stress and restore glutathione homeostasis and by inhibiting NFκB signaling to attenuate neuroinflammatory changes.


Asunto(s)
Indenos , Melatonina , Ratones , Animales , Melatonina/farmacología , Melatonina/metabolismo , Receptores de Melatonina/metabolismo , Ácido Kaínico/toxicidad , Ácido Kaínico/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Hipocampo , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Glutatión/metabolismo , ADN
2.
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
3.
J Neuroinflammation ; 19(1): 142, 2022 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690821

RESUMEN

BACKGROUND: It has been demonstrated that reactive astrocytes can be polarized into pro-inflammatory A1 phenotype or anti-inflammatory A2 phenotype under neurotoxic and neurodegenerative conditions. Microglia have been suggested to play a critical role in astrocyte phenotype polarization by releasing pro- and anti-inflammatory mediators. In this study, we examined whether trimethyltin (TMT) insult can induce astrocyte polarization in the dentate gyrus of mice, and whether protein kinase Cδ (PKCδ) plays a role in TMT-induced astrocyte phenotype polarization. METHODS: Male C57BL/6 N mice received TMT (2.6 mg/kg, i.p.), and temporal changes in the mRNA expression of A1 and A2 phenotype markers were evaluated in the hippocampus. In addition, temporal and spatial changes in the protein expression of C3, S100A10, Iba-1, and p-PKCδ were examined in the dentate gyrus. Rottlerin (5 mg/kg, i.p. × 5 at 12-h intervals) was administered 3-5 days after TMT treatment, and the expression of A1 and A2 transcripts, p-PKCδ, Iba-1, C3, S100A10, and C1q was evaluated 6 days after TMT treatment. RESULTS: TMT treatment significantly increased the mRNA expression of A1 and A2 phenotype markers, and the increased expression of A1 markers remained longer than that of A2 markers. The immunoreactivity of the representative A1 phenotype marker, C3 and A2 phenotype marker, S100A10 peaked 6 days after TMT insult in the dentate gyrus. While C3 was expressed evenly throughout the dentate gyrus, S100A10 was highly expressed in the hilus and inner molecular layer. In addition, TMT insult induced microglial p-PKCδ expression. Treatment with rottlerin, a PKCδ inhibitor, decreased Iba-1 and C3 expression, but did not affect S100A10 expression, suggesting that PKCδ inhibition attenuates microglial activation and A1 astrocyte phenotype polarization. Consistently, rottlerin significantly reduced the expression of C1q and tumor necrosis factor-α (TNFα), which has been suggested to be released by activated microglia and induce A1 astrocyte polarization. CONCLUSION: We demonstrated the temporal and spatial profiles of astrocyte polarization after TMT insult in the dentate gyrus of mice. Taken together, our results suggest that PKCδ plays a role in inducing A1 astrocyte polarization by promoting microglial activation and consequently increasing the expression of pro-inflammatory mediators after TMT insult.


Asunto(s)
Astrocitos , Complemento C1q , Acetofenonas , Animales , Astrocitos/metabolismo , Benzopiranos , Complemento C1q/metabolismo , Giro Dentado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Compuestos de Trimetilestaño
4.
Pharmacol Res ; 179: 106226, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35460881

RESUMEN

BKM120 is an inhibitor of class I phosphoinositide 3-kinases and its anti-cancer effects have been demonstrated in various solid cancer models. BKM120 is highly brain permeable and has been reported to induce mood disturbances in clinical trials. Therefore, we examined whether BKM120 produces anxiety- and depression-like behaviors in mice, as with patients receiving BKM120 in clinical trials. In this study, repeated BKM120 treatment (2.0 or 5.0 mg/kg, i.p., five times at 12-h interval) significantly induced anxiety- and depression-like behaviors in mice. Although abnormal changes in hippocampal neurogenesis have been suggested to, at least in part, associated with the pathogenesis of depression and anxiety, BKM120 did not affect the incorporation of 5-bromo-2'-deoxyuridine or the expression of doublecortin (DCX); however, it significantly enhanced the radial migration of DCX-positive cells in the dentate gyrus. BKM120-induced changes in migration were not accompanied by obvious neuronal damage in the hippocampus. Importantly, BKM120-induced anxiety- and depression-like behaviors were positively correlated with the extent of DCX-positive cell migration. Concomitantly, p-Akt expression was significantly decreased in the dentate gyrus. Moreover, the expression of p-c-Jun N-terminal kinase (JNK), p-DCX, and Ras homolog family member A (RhoA)-GTP decreased significantly, particularly in aberrantly migrated DCX-positive cells. Together, the results suggest that repeated BKM120 treatment enhances the radial migration of DCX-positive cells and induces anxiety- and depression-like behaviors by regulating the activity of Akt, JNK, DCX, and RhoA in the dentate gyrus. It also suggests that the altered migration of adult-born neurons in the dentate gyrus plays a role in mood disturbances.


Asunto(s)
Giro Dentado , Neuropéptidos , Aminopiridinas , Animales , Giro Dentado/metabolismo , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Hipocampo/metabolismo , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Morfolinas , Neuropéptidos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
5.
Int J Mol Sci ; 23(4)2022 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-35216059

RESUMEN

The purpose of this review article is to outline the extended applications of polyurethane (PU)-based nanocomposites incorporated with conductive polymeric particles as well as to condense an outline on the chemistry and fabrication of polyurethanes (PUs). Additionally, we discuss related research trends of PU-based conducting materials for EMI shielding, sensors, coating, films, and foams, in particular those from the past 10 years. PU is generally an electrical insulator and behaves as a dielectric material. The electrical conductivity of PU is imparted by the addition of metal nanoparticles, and increases with the enhancing aspect ratio and ordering in structure, as happens in the case of conducting polymer fibrils or reduced graphene oxide (rGO). Nanocomposites with good electrical conductivity exhibit noticeable changes based on the remarkable electric properties of nanomaterials such as graphene, RGO, and multi-walled carbon nanotubes (MWCNTs). Recently, conducting polymers, including PANI, PPY, PTh, and their derivatives, have been popularly engaged as incorporated fillers into PU substrates. This review also discusses additional challenges and future-oriented perspectives combined with here-and-now practicableness.


Asunto(s)
Nanocompuestos/química , Poliuretanos/química , Conductividad Eléctrica , Grafito/química , Nanotubos de Carbono/química , Polímeros/química
6.
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
7.
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
8.
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
9.
Arch Toxicol ; 94(7): 2505-2516, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32296860

RESUMEN

The use of new psychoactive substances (NPSs) as a substitute for illegal drugs is increasing rapidly and is a serious threat to public health. 25C-NBF is a newly synthesized phenethylamine-type NPS that acts as a 5-hydroxyindoleacetic acid (5-HT) receptor agonist, but little is known about its pharmacological effects. Considering that NPSs have caused unexpected harmful effects leading to emergency and even death, scientific confirmation of the potential adverse effects of 25C-NBF is essential. In the present study, we investigated whether 25C-NBF has addictive and neurotoxic potential and causes neurochemical changes. In addictive potential assessments, high conditioned place preference (CPP) scores and stable self-administration (SA) were observed in the 25C-NBF groups (CPP [3 mg kg-1]; SA [0.01, 0.03, 0.1 mg kg-1]), suggesting the addictive liability of 25C-NBF. In neurotoxic potential assessments, 25C-NBF treatment (single super-high dose [1 × 15, 30, 40 mg kg-1]; repeated high dose [4 × 8, 15, 30 mg kg-1]) resulted in reduced motor activity (open field test), abnormal motor coordination (rota-rod test) and impaired recognition memory (novel object recognition test), suggesting that 25C-NBF is neurotoxic leading to motor impairment and memory deficits. Subsequently, immunohistochemistry showed that 25C-NBF treatment decreased tyrosine hydroxylase (TH) expression and increased ionized calcium-binding adapter molecule 1 (Iba-1) expression in the striatum. Taken together, our results clearly demonstrate the dangers of recreational use of 25C-NBF, and we suggest that people stop using 25C-NBF and other NPSs whose pharmacological effects are not precisely known.


Asunto(s)
Conducta Adictiva/inducido químicamente , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Fenetilaminas/toxicidad , Psicotrópicos/toxicidad , Trastornos Relacionados con Sustancias/etiología , Animales , Conducta Adictiva/metabolismo , Conducta Adictiva/psicología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Proteínas de Unión al Calcio/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Locomoción/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Proteínas de Microfilamentos/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/fisiopatología , Prueba de Campo Abierto/efectos de los fármacos , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante , Trastornos Relacionados con Sustancias/metabolismo , Trastornos Relacionados con Sustancias/psicología , Tirosina 3-Monooxigenasa/metabolismo
10.
Clin Exp Pharmacol Physiol ; 46(9): 791-797, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31332816

RESUMEN

The aim of this study was to investigate whether the glutathione peroxidase-1 gene (GPx-1) affects cocaine-induced conditioned place preference (CPP) using a mouse model. Cocaine-induced CPP was accompanied by an increase in the level of σ-1 receptor in the nucleus accumbens (NAc). This phenomenon was more pronounced in the GPx-1 gene knockout (GPx-1 KO) than in wild type (WT) mice. In contrast, the CPP and expression of σ-1 receptor were much less pronounced in GPx-1-overexpressing transgenic (GPx-1 TG) mice than non-transgenic (non-TG) mice. Treatment of the mice with BD1047, a σ-1 receptor antagonist, significantly attenuated both cocaine-induced CPP and c-Fos-immunoreactivity (c-Fos-IR) in WT and GPx-1 KO mice, although the effects were more evident in the latter group. Despite the protective effects of BD1047 on cocaine-induced CPP and c-Fos in non-TG mice, there were no additional protective effects in cocaine-treated GPx-1 TG mice, indicating that the σ-1 receptor is a critical target for GPx-1-mediated psychoprotective activity. Overall, our results suggest that GPx-1 attenuates cocaine-induced CPP via inhibition of σ-1 receptor expression.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cocaína/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/genética , Receptores sigma/genética , Animales , Técnicas de Inactivación de Genes , Glutatión Peroxidasa/deficiencia , Ratones , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Glutatión Peroxidasa GPX1 , Receptor Sigma-1
11.
J Neuroinflammation ; 15(1): 52, 2018 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-29467000

RESUMEN

BACKGROUND: We previously reported that ginsenoside Re (GRe) attenuated against methamphetamine (MA)-induced neurotoxicity via anti-inflammatory and antioxidant potentials. We also demonstrated that dynorphin possesses anti-inflammatory and antioxidant potentials against dopaminergic loss, and that balance between dynorphin and substance P is important for dopaminergic neuroprotection. Thus, we examined whether GRe positively affects interactive modulation between dynorphin and substance P against MA neurotoxicity in mice. METHODS: We examined changes in dynorphin peptide level, prodynorphin mRNA, and substance P mRNA, substance P-immunoreactivity, homeostasis in enzymatic antioxidant system, oxidative parameter, microglial activation, and pro-apoptotic parameter after a neurotoxic dose of MA to clarify the effects of GRe, prodynorphin knockout, pharmacological inhibition of κ-opioid receptor (i.e., nor-binaltorphimine), or neurokinin 1 (NK1) receptor (i.e., L-733,060) against MA insult in mice. RESULTS: GRe attenuated MA-induced decreases in dynorphin level, prodynorphin mRNA expression in the striatum of wild-type (WT) mice. Prodynorphin knockout potentiated MA-induced dopaminergic toxicity in mice. The imbalance of enzymatic antioxidant system, oxidative burdens, microgliosis, and pro-apoptotic changes led to the dopaminergic neurotoxicity. Neuroprotective effects of GRe were more pronounced in prodynorphin knockout than in WT mice. Nor-binaltorphimine, a κ-opioid receptor antagonist, counteracted against protective effects of GRe. In addition, we found that GRe significantly attenuated MA-induced increases in substance P-immunoreactivity and substance P mRNA expression in the substantia nigra. These increases were more evident in prodynorphin knockout than in WT mice. Although, we observed that substance P-immunoreactivity was co-localized in NeuN-immunreactive neurons, GFAP-immunoreactive astrocytes, and Iba-1-immunoreactive microglia. NK1 receptor antagonist L-733,060 or GRe selectively inhibited microgliosis induced by MA. Furthermore, L-733,060 did not show any additive effects against GRe-mediated protective activity (i.e., antioxidant, antimicroglial, and antiapoptotic effects), indicating that NK1 receptor is one of the molecular targets of GRe. CONCLUSIONS: Our results suggest that GRe protects MA-induced dopaminergic neurotoxicity via upregulatgion of dynorphin-mediated κ-opioid receptor and downregulation of substance P-mediated NK1 R.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Dinorfinas/metabolismo , Ginsenósidos/farmacología , Metanfetamina/toxicidad , Receptores de Neuroquinina-1/metabolismo , Receptores Opioides kappa/metabolismo , Sustancia P/metabolismo , Animales , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antagonistas del Receptor de Neuroquinina-1/farmacología , Piperidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
12.
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
13.
Neurochem Res ; 43(1): 66-78, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28589520

RESUMEN

Methamphetamine (MA), an amphetamine-type psychostimulant, is associated with dopaminergic toxicity and has a high abuse potential. Numerous in vivo and in vitro studies have suggested that impaired mitochondria are critical in dopaminergic toxicity induced by MA. Mitochondria are important energy-producing organelles with dynamic nature. Evidence indicated that exposure to MA can disturb mitochondrial energetic metabolism by inhibiting the Krebs cycle and electron transport chain. Alterations in mitochondrial dynamic processes, including mitochondrial biogenesis, mitophagy, and fusion/fission, have recently been shown to contribute to dopaminergic toxicity induced by MA. Furthermore, it was demonstrated that MA-induced mitochondrial impairment enhances susceptibility to oxidative stress, pro-apoptosis, and neuroinflammation in a positive feedback loop. Protein kinase Cδ has emerged as a potential mediator between mitochondrial impairment and oxidative stress, pro-apoptosis, or neuroinflammation in MA neurotoxicity. Understanding the role and underlying mechanism of mitochondrial impairment could provide a molecular target to prevent or alleviate dopaminergic toxicity induced by MA.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Metanfetamina/farmacología , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Neuronas Dopaminérgicas/metabolismo , Humanos , Mitocondrias/metabolismo
14.
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
15.
J Appl Toxicol ; 38(12): 1502-1520, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30027653

RESUMEN

Since the cocaine-induced oxidative stress has been established to lead to hepatotoxicity, we examined the role of the glutathione peroxidase (GPx)-1 gene in cocaine-induced hepatotoxicity. Cocaine treatment significantly increased superoxide dismutase activity in as little as 1 hour, with a maximum level at 6 hours in wild-type mice, while significantly decreasing GPx activity and subsequently inducing oxidative damage (i.e., reactive oxygen species, lipid peroxidation and protein carbonylation). These changes were more prominent in the mitochondrial fraction than in the cytosolic fraction. In contrast, genetic overexpression of GPx-1 significantly attenuated cocaine-induced oxidative damage in mice. Cocaine treatment significantly increased alanine aminotransferase and aspartate aminotransferase levels in the serum. Consistently, cocaine significantly enhanced cleaved caspase-3 expression and intramitochondrial Ca2+ , while significantly reducing mitochondrial transmembrane potential. Cocaine treatment potentiated cleavage of protein kinase C δ (PKCδ), mitochondrial translocation of PKCδ, cytosolic release of cytochrome c and activation of caspase-3, followed by hepatopathologic changes. These results were more prominent in GPx-1 knockout than in wild-type mice, and they were less pronounced in overexpressing transgenic than in non-transgenic mice. Combined, our results suggest that the GPx-1 gene possesses protective potential against mitochondrial oxidative burden, mitochondrial dysfunction and hepatic degeneration induced by cocaine and that the protective mechanisms are associated with anti-apoptotic activity via inactivation of PKCδ.


Asunto(s)
Antioxidantes/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Cocaína/toxicidad , Glutatión Peroxidasa/genética , Estrés Oxidativo/genética , Animales , Calcio/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citosol/efectos de los fármacos , Citosol/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Ratones Noqueados , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Carbonilación Proteica/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transgenes , Glutatión Peroxidasa GPX1
16.
Environ Toxicol ; 33(10): 1019-1028, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30076769

RESUMEN

Although it has been well-recognized that microcystin-leucine-arginine (MCLR), the most common form of microcystins, induces neurotoxicity, little is currently known about the underlying mechanism for this neurotoxicity. Here, we found that MCLR (10 ng/µL/mouse, i.c.v.) induces significant neuronal loss in the hippocampus of mice. MCLR-induced neurotoxicity was accompanied by oxidative stress, as shown by a significant increase in the level of 4-hydroxynonenal, protein carbonyl, and reactive oxygen species (ROS). Superoxide dismutase-1 (SOD-1) activity was significantly increased, but glutathione peroxidase (GPx) level was significantly decreased following MCLR insult. In addition, MCLR significantly inhibited GSH/GSSG ratio, and significantly induced NFκB DNA binding activity. Because reduced activity of GPx appeared to be critical for the imbalance between activities of SODs and GPx, we utilized GPx-1 overexpressing transgenic mice to ascertain the role of GPx-1 in this neurotoxicity. Genetic overexpression of GPx-1 or NFκB inhibitor pyrrolidine dithiocarbamate (PDTC) significantly attenuated MCLR-induced hippocampal neuronal loss in mice. However, PDTC did not exert any additive effect on neuroprotection mediated by GPx-1 overexpression, indicating that NFκB is a neurotoxic target of MCLR. Combined, these results suggest that MCLR-induced neurotoxicity requires oxidative stress associated with failure in compensatory induction of GPx, possibly through activation of the transcription factor NFκB.


Asunto(s)
Arginina/toxicidad , Glutatión Peroxidasa/metabolismo , Hipocampo/efectos de los fármacos , Leucina/toxicidad , Microcistinas/toxicidad , Aldehídos/análisis , Animales , Regulación hacia Abajo/efectos de los fármacos , Glutatión/metabolismo , Glutatión Peroxidasa/genética , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Pirrolidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa-1/metabolismo , Tiocarbamatos/farmacología , Regulación hacia Arriba/efectos de los fármacos , Glutatión Peroxidasa GPX1
17.
J Neuroinflammation ; 14(1): 94, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28449688

RESUMEN

BACKGROUND: Ginkgo biloba has been reported to possess free radical-scavenging antioxidant activity and anti-inflammatory properties. In our pilot study, YY-1224, a terpene trilactone-strengthened extract of G. biloba, showed anti-inflammatory, neurotrophic, and antioxidant effects. RESULTS: We investigated the pharmacological potential of YY-1224 in ß-amyloid (Aß) (1-42)-induced memory impairment using cyclooxygenase-2 (COX-2) knockout (-/-) and APPswe/PS1dE9 transgenic (APP/PS1 Tg) mice. Repeated treatment with YY-1224 significantly attenuated Aß (1-42)-induced memory impairment in COX-2 (+/+) mice, but not in COX-2 (-/-) mice. YY-1224 significantly attenuated Aß (1-42)-induced upregulation of platelet-activating factor (PAF) receptor gene expression, reactive oxygen species, and pro-inflammatory factors. In addition, YY-1224 significantly inhibited Aß (1-42)-induced downregulation of PAF-acetylhydrolase-1 (PAF-AH-1) and peroxisome proliferator-activated receptor γ (PPARγ) gene expression. These changes were more pronounced in COX-2 (+/+) mice than in COX-2 (-/-) mice. YY-1224 significantly attenuated learning impairment, Aß deposition, and pro-inflammatory microglial activation in APP/PS1 Tg mice, whereas it significantly enhanced PAF-AH and PPARγ expression. A preferential COX-2 inhibitor, meloxicam, did not affect the pharmacological activity by YY-1224, suggesting that the COX-2 gene is a critical mediator of the neuroprotective effects of YY-1224. The protective activity of YY-1224 appeared to be more efficacious than a standard G. biloba extract (Gb) against Aß insult. CONCLUSIONS: Our results suggest that the protective effects of YY-1224 against Aß toxicity may be associated with its PAF antagonistic- and PPARγ agonistic-potential as well as inhibition of the Aß-mediated pro-inflammatory switch of microglia phenotypes through suppression of COX-2 expression.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Ciclooxigenasa 2/metabolismo , Ginkgo biloba , Enfermedades Neurodegenerativas/metabolismo , Fragmentos de Péptidos/toxicidad , Extractos Vegetales/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Expresión Génica , Lactonas/aislamiento & purificación , Lactonas/uso terapéutico , Ratones , Ratones Noqueados , Ratones Transgénicos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/prevención & control , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Presenilina-1/biosíntesis , Presenilina-1/genética , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Terpenos/aislamiento & purificación , Terpenos/uso terapéutico
18.
Toxicol Appl Pharmacol ; 334: 158-166, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28916251

RESUMEN

Dextromethorphan (DM) is a dextrorotatory isomer of levorphanol, a typical morphine-like opioid. When administered at supra-antitussive doses, DM produces psychotoxic and neurotoxic effects in humans. Although DM abuse has been well-documented, few studies have examined the effects of high-dose DM. The present study aimed to explore the effects of a single high dose of DM on mortality and seizure occurrence. After intraperitoneal administration with a high dose of DM (80mg/kg), Sprague-Dawley rats showed increased seizure occurrence and intensity. Hippocampal expression levels of N-methyl-d-aspartate (NMDA) receptor subunits (GluN1

Asunto(s)
Dextrometorfano/toxicidad , Maleato de Dizocilpina/farmacología , Naloxona/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Animales , Antitusígenos/farmacología , Dextrometorfano/administración & dosificación , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Antagonistas de Narcóticos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética
19.
Neurochem Res ; 42(11): 3125-3139, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28884396

RESUMEN

Ginseng (Panax ginseng), an herbal medicine, has been used to prevent neurodegenerative disorders. Ginsenosides (e.g., Re, Rb1, or Rg1) were obtained from Korean mountain cultivated ginseng. The anticonvulsant activity of ginsenoside Re (20 mg/kg/day × 3) against trimethyltin (TMT) insult was the most pronounced out of ginsenosides (e.g., Re, Rb1, and Rg1). Re itself did not significantly alter tumor necrosis factor-α (TNF-α), interferon-ϒ (IFN-ϒ), and interleukin-1ß (IL-1ß) expression, however, it significantly increases the interleukin-6 (IL-6) expression. In addition, Re attenuated the TMT-induced decreases in IL-6 protein level. Therefore, IL-6 knockout (-/-) mice were employed to investigate whether Re requires IL-6-dependent neuroprotective activity against TMT toxicity. Re significantly attenuated TMT-induced lipid peroxidation, protein peroxidation, and reactive oxygen species in the hippocampus. Re-mediated antioxidant effects were more pronounced in IL-6 (-/-) mice than in WT mice. Consistently, TMT-induced increase in c-Fos-immunoreactivity (c-Fos-IR), TUNEL-positive cells, and nuclear chromatin clumping in the dentate gyrus of the hippocampus were significantly attenuated by Re. Furthermore, Re attenuated TMT-induced proapoptotic changes. Protective potentials by Re were comparable to those by recombinant IL-6 protein (rIL-6) against TMT-insult in IL-6 (-/-) mice. Moreover, treatment with a phosphoinositol 3-kinase (PI3K) inhibitor, LY294002 (1.6 µg, i.c.v) counteracted the protective potential mediated by Re or rIL-6 against TMT insult. The results suggest that ginsenoside Re requires IL-6-dependent PI3K/Akt signaling for its protective potential against TMT-induced neurotoxicity.


Asunto(s)
Ginsenósidos/farmacología , Interleucina-6/deficiencia , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Compuestos de Trimetilestaño/toxicidad , Animales , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Panax , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Compuestos de Trimetilestaño/antagonistas & inhibidores
20.
Planta Med ; 83(17): 1342-1350, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28561205

RESUMEN

Panax ginseng is the most widely used herbal medicine for improving cognitive functions. The pharmacological activity and underlying mechanisms of mountain-cultivated ginseng, however, have yet to be clearly elucidated, in particular, against trimethyltin-induced cognitive dysfunction. We previously reported that interleukin-6 plays a protective role against trimethyltin-induced cognitive dysfunction. Because of this, we have implemented a study system that uses interleukin-6 null (-/-) and wild-type mice. Interestingly, mountain-cultivated ginseng significantly upregulated interleukin-6 expression. With this study, we sought to determine whether the interleukin-6-dependent modulation of the Janus kinase 2/signal transducer activator of transcription 3 and extracellular signal-regulated kinase signaling network is also associated with the pharmacological activity of mountain-cultivated ginseng against trimethyltin-induced cognitive dysfunction. Trimethyltin treatment (2.4 mg/kg, intraperitoneal) causes the downregulation of Janus kinase 2/signal transducer activator of transcription 3, extracellular signal-regulated kinase signaling, and impairment of the cholinergic system. We found that mountain-cultivated ginseng treatment (50 mg/kg, intraperitoneal) significantly attenuated cognitive impairment normally induced by trimethyltin by upregulating p-Janus kinase 2/signal transducer activator of transcription 3, p-extracellular signal-regulated kinase signaling, and the cholinergic system. Trimethyltin-induced cognitive impairments were more pronounced in interleukin-6 (-/-) mice than wild-type mice, and they were markedly reduced by treatment with either mountain-cultivated ginseng or recombinant interleukin-6 protein (6 ng, intracerebroventricular). Additionally, treatment with either AG490 (20 mg/kg, intraperitoneal), a Janus kinase 2/signal transducer activator of transcription 3 inhibitor, or U0126 (2 µg/head, intracerebroventricular), an extracellular signal-regulated kinase inhibitor, reversed the effects of mountain-cultivated ginseng treatment. The effects of mountain-cultivated ginseng treatment were comparable to those of recombinant interleukin-6 protein in interleukin-6 (-/-) mice. Our results, therefore, suggest that mountain-cultivated ginseng acts through interleukin-6-dependent activation of Janus kinase 2/signal transducer activator of transcription 3/extracellular signal-regulated kinase signaling in order to reverse cognitive impairment caused by trimethyltin treatment.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Interleucina-6/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Panax , Fitoterapia , Animales , Disfunción Cognitiva/inducido químicamente , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Interleucina-6/genética , Janus Quinasa 2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Panax/química , Panax/crecimiento & desarrollo , Filogeografía , Factor de Transcripción STAT3/metabolismo , Compuestos de Trimetilestaño , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA