Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(5): e0300787, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38753634

RESUMEN

The Presenilin (Psn) gene is closely related to aging, but it is still unclear the role of Psn genes in skeletal muscle. Here, the Psn-UAS/Mhc-GAL4 system in Drosophila was used to regulate muscle Psn overexpression(MPO) and muscle Psn knockdown(MPK). Drosophila were subjected to endurance exercise from 4 weeks to 5 weeks old. The results showed that MPO and exercise significantly increased climbing speed, climbing endurance, lifespan, muscle SOD activity, Psn expression, Sirt1 expression, PGC-1α expression, and armadillo (arm) expression in aged Drosophila, and they significantly decreased muscle malondialdehyde levels. Interestingly, when the Psn gene is knockdown by 0.78 times, the PGC-1α expression and arm expression were also down-regulated, but the exercise capacity and lifespan were increased. Furthermore, exercise combined with MPO further improved the exercise capacity and lifespan. MPK combined with exercise further improves the exercise capacity and lifespan. Thus, current results confirmed that the muscle Psn gene was a vital gene that contributed to the healthy aging of skeletal muscle since whether it was overexpressed or knocked down, the aging progress of skeletal muscle structure and function was slowed down by regulating the activity homeostasis of Sirt1/PGC-1α pathway and Psn/arm pathway. Exercise enhanced the function of the Psn gene to delay skeletal muscle aging by up regulating the activity of the Sirt1/PGC-1α pathway and Psn/arm pathway.


Asunto(s)
Longevidad , Músculo Esquelético , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Condicionamiento Físico Animal , Sirtuina 1 , Animales , Sirtuina 1/metabolismo , Sirtuina 1/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Longevidad/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Transducción de Señal , Envejecimiento Saludable/genética , Envejecimiento Saludable/metabolismo , Envejecimiento Saludable/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Envejecimiento/fisiología , Envejecimiento/genética , Envejecimiento/metabolismo
2.
Biomed Pharmacother ; 165: 115100, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37418977

RESUMEN

The use of oral agents that can modify the gut microbiota (GM) could be a novel preventative or therapeutic option for Parkinson's disease (PD). Maslinic acid (MA), a pentacyclic triterpene acid with GM-dependent biological activities when it is taken orally, has not yet been reported to be effective against PD. The present study found both low and high dose MA treatment significantly prevented dopaminergic neuronal loss in a classical chronic PD mouse model by ameliorating motor functions and improving tyrosine hydroxylase expressions in the substantia nigra pars compacta (SNpc) and increasing dopamine and its metabolite homovanillic acid levels in the striatum. However, the effects of MA in PD mice were not dose-responsive, since similar beneficial effects for low and high doses of MA were observed. Further mechanism studies indicated that low dose MA administration favored probiotic bacterial growth in PD mice, which helped to increase striatal serotonin, 5-hydroxyindole acetic acid, and γ-aminobutyric acid levels. High dose MA treatment did not influence GM composition in PD mice but significantly inhibited neuroinflammation as indicated by reduced levels of tumor necrosis factor alpha and interleukin 1ß in the SNpc; moreover, these effects were mainly mediated by microbially-derived acetic acid in the colon. In conclusion, oral MA at different doses protected against PD via distinct mechanisms related to GM. Nevertheless, our study lacked in-depth investigations of the underlying mechanisms involved; future studies will be designed to further delineate the signaling pathways involved in the interactive actions between different doses of MA and GM.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad de Parkinson , Ratones , Animales , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/prevención & control , Enfermedad de Parkinson/metabolismo , Sustancia Negra , Dopamina/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo
3.
Food Res Int ; 155: 111067, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35400445

RESUMEN

Gut microbiota (GM) dysbiosis plays key roles in aggravating Parkinson's disease (PD) and discovery of agents targeting GM may open new avenues for PD therapy. This study aims to investigate the potentially neuroprotective effects and underlying mechanisms of polymannuronic acid (PM) or Lacticaseibacillus rhamnosus GG (LGG), or their combination in a chronic PD mice model. Our results found oral administration of prebiotic PM or LGG separately or in combination for 5 weeks could prevent dopaminergic neuronal loss via improving reduced walking distance and activity or weakened muscle strength in behavior tests by enhancing tyrosine hydroxylase (TH) gene and/or protein expressions in the midbrain and striatum of PD mice. Strikingly, PM and LGG in combination had a much better neuroprotective effects than separate PM or LGG. PM provided neuroprotection via a short chain fatty acids (SCFAs)-mediated anti-inflammation and anti-apoptosis mechanism. The neuroprotective effects of LGG might be associated with its ability to improve the expression of striatal glial cell-derived neurotrophic factor (GDNF) and to increase bacteria abundance of Clostridiales. When PD mice were administered with PM + LGG, PM as prebiotic favored bacterial growth (from Bacilli class to Lactobacillus genus) in the colon, which helped to improve blood brain barrier (BBB) integrity and increase brain-derived neurotrophic factor (BDNF) and GDNF expressions, thereby inhibiting apoptosis in the striatum. In conclusion, PM and LGG in combination promoted their separate neuroprotection against PD. Our study discovered and testified a novel synbiotic that might be one of the ideal oral agents for PD therapy.


Asunto(s)
Lacticaseibacillus rhamnosus , Fármacos Neuroprotectores , Enfermedad de Parkinson , Probióticos , Simbióticos , Ácido Algínico , Animales , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Ratones , Neuroprotección , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/prevención & control , Prebióticos , Probióticos/farmacología
4.
Mol Nutr Food Res ; 66(2): e2100739, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34811884

RESUMEN

SCOPE: This study aims to investigate and compare the potentially neuroprotective effects and underlying mechanisms for brown seaweed polysaccharides (PS) of Alginate (Alg) and its two components, including polymannuronic acid (PM) and polyguluronic acid (PG), against Parkinson's disease (PD) pathogenesis. METHODS AND RESULTS: Model mice of PD are pretreated with Alg or PM or PG, separately via oral gavage once per day for four weeks. Our results found PM improved motor functions of PD mice, but Alg or PG did not. PM or PG, but not Alg, can prevent dopaminergic neuronal loss by increasing tyrosine hydroxylase (TH) expressions in midbrain of PD mice. The neuroprotective effects of PM rely on its anti-inflammation effects and its ability to improve striatal neurotransmitters (serotonin (5-HT) and 5-hydroxyindole acetic acid (5-HIAA)) levels in PD mice. PM inhibits inflammation, but PG or Alg induces inflammation in systemic circulation of PD mice. The neuroprotection provided by PG might be related to its ability to increase striatal neurotransmitter of 5-hydroxyindole acetic acid levels in PD mice. CONCLUSION: PM plays better than PG to provide neuroprotection, but Alg did not show any neuroprotection against PD. Alg and its two components acted differently in preventing dopaminergic neuronal loss in PD mice.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Alginatos/farmacología , Animales , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
5.
J Steroid Biochem Mol Biol ; 214: 105989, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34478828

RESUMEN

Neuroinflammation plays an important role in the pathogenesis of Parkinson's disease (PD). Genistein is an estrogen-like phytoestrogen that can exert biological effects via the crosstalk of estrogen receptor and insulin-like growth factor 1 receptor (IGF-1R). The present study aimed to evaluate the involvement of G protein-coupled estrogen receptor (GPER) and IGF-1R in the anti-inflammatory effects of genistein against lipopolysaccharide (LPS)-induced nigrostriatal injury in ovariectomized rats. Our results showed that genistein treatment could ameliorate the apomorphine-induced rotational behavior in LPS-induced inflammatory PD rat model. Genistein attenuated LPS-induced decrease of the contents of dopamine (DA) and its metabolites in striatum as well as the loss of tyrosine hydroxylase-immunoreactive (TH-IR) neurons in the substantia nigra (SN) of the lesioned side, which could be blocked by GPER antagonist G15 or IGF-1R antagonist JB1. Meanwhile, G15 or JB1 could attenuate the anti-inflammatory effects of genistein in LPS-induced microglial activation and production of tumor necrosis factor-α (TNF-α), interleukin 1ß (IL-1ß), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Moreover, genistein could inhibit the LPS-induced phosphorylation of p38, JNK, ERK and IκB in the lesioned side of SN and these effects could also be blocked by G15 or JB1. Taken together, our data provide the first evidence that genistein can inhibit the increase of microglia and protect dopaminergic neurons at least in part via GPER and IGF-1R signaling pathways in ovariectomized PD rat model.


Asunto(s)
Genisteína/farmacología , Lipopolisacáridos/metabolismo , Microglía/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sustancia Negra/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Neuronas Dopaminérgicas/metabolismo , Femenino , Sistema de Señalización de MAP Quinasas , FN-kappa B/metabolismo , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Wistar , Transducción de Señal
6.
Eur J Pharmacol ; 906: 174269, 2021 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-34147477

RESUMEN

Icariin and icaritin, the major active components of Epimedii Genus, are considered as promising drugs with anti-inflammatory, anti-aging and neuroprotective effects. Our previous studies have demonstrated that icariin and icaritin can protect against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity on dopaminergic neurons via insulin-like growth factor-1 receptor (IGF-1 receptor) signaling. In the present study, we aimed to evaluate the role of IGF-1 receptor signaling in mediating the anti-inflammatory effects of icariin and icaritin against lipopolysaccharide (LPS)-induced neuroinflammation as well as their biological regulation effects in midbrain primary astrocytes. Our results showed that both icariin and icaritin significantly inhibited LPS-induced mRNA expressions of tumor necrosis factor (TNF-α) and interleukin-1ß (IL-1ß). Pre-treatment with IGF-1 receptor antagonist JB-1 could significantly block the anti-inflammatory effects of icariin and icaritin on LPS-induced up-regulations of TNF-α, IL-1ß, cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS). Under basal conditions of astrocytes, icariin and icaritin treatment alone increased the phosphorylation of ERK1/2 and AKT, which could be blocked by JB-1. Moreover, the mRNA expressions of glutamate transptor-1 (GLT-1) and glutamate-aspartate transporter (GLAST) could be up-regulated by icariin and icaritin in a time-dependent manner via IGF-1 receptor. Taken together, our results suggest for the first time that both icariin and icaritin exert regulatory effects in astrocytes under basal conditions and after an inflammatory challenge via IGF-1 receptor signaling pathway.


Asunto(s)
Astrocitos/patología , Flavonoides/farmacología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Receptor IGF Tipo 1/metabolismo , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Flavonoides/aislamiento & purificación , Flavonoides/uso terapéutico , Humanos , Lipopolisacáridos/inmunología , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/patología , Ratones , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/patología , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
7.
Front Aging Neurosci ; 13: 649627, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33912026

RESUMEN

The role of gut-brain axis in the pathogenesis of Parkinson's disease (PD) have become a research hotspot, appropriate animal model to study gut-brain axis in PD is yet to be confirmed. Our study employed a classical PD mice model achieved by chronic MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) injection to study concurrent changes of dopaminergic neurons in the midbrain and the colon of mice. Our results showed such a PD model exhibited apparent locomotor deficits but not gastrointestinal dysfunction. Tyrosine hydroxylase expressions and dopamine content reduced greatly in the substantia nigra pars compacta (SNpc) or striatum, but increased in the colon of PD mice. Mechanism investigation indicated autophagy activity and apoptosis were stimulated in the SNpc, but inhibited in the colon of PD mice. Interplay of gut microbiota (GM) and autophagy in response to chronic MPTP injection led to GM dysbiosis and defective autophagy in mice colon. Meanwhile, fecal short chain fatty acids (SCFAs), acetate and propionate in particular, declined greatly in PD mice, which could be attributed to the decreased bacteria abundance of phylum Bacteroidetes, but increased abundance of phylum Firmicutes. GM dysbiosis derived fecal SCFAs might be one of the mediators of downregulated autophagy in the colon of PD mice. In conclusion, colonic dopaminergic neurons changed in the opposition direction with those in the midbrain via GM dysbiosis-mediated autophagy inhibition followed by suppressed apoptosis in response to chronic MPTP injection. Such a chronic PD mice model might not be an ideal model to study role of gut-brain axis in PD progression.

8.
Int J Biol Macromol ; 164: 994-1005, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-32710966

RESUMEN

The study aims to investigate the potentially neuroprotective effects and underlying mechanisms for brown seaweed polysaccharide of polymannuronic acid (PM) against Parkinson's disease (PD) pathogenesis. PD model mice were pretreated with PM via oral gavage once per day for 4 weeks and the preventative effects of PM against neuronal loss together with its modulation on brain-gut-microbiota axis were systematically explored. The results showed PM administration improved motor functions by preventing dopaminergic neuronal loss in the substantia nigra pars compacta (SNpc) and enhanced contents of striatal homovanillic acid (HVA), serotonin (5-HT), 5-hydroxyindole acetic acid (5-HIAA) and γ-aminobutyric acid (GABA) in PD mice. PM significantly alleviated inflammation in gut, brain and systemic circulation as shown by reduced levels or expressions of pro-inflammatory cytokines concurrently and inhibited mitogen-activated protein kinases (MAPK) signaling pathway in mice colon. Meanwhile, PM greatly improved integrity of intestinal barrier and blood brain barrier (BBB) as indicated by increased expressions of tight junction associated proteins in both mice colon and SNpc. Further studies indicated PM treatment resulted in changes of gut microbial compositions, together with great alterations of digestion and metabolism of dietary proteins and fats, which led to surge increase of fecal short chain fatty acids (SCFAs) in the colon of PD mice. In conclusion, pre-administration of PM could provide neuroprotective effects against PD pathogenesis by suppressing inflammation in gut, brain and systemic circulation, and by improving integrity of intestinal barrier and BBB. PM might modulate brain-gut-microbiota axis, at least in part, via gut microbiota derived SCFAs as mediators.


Asunto(s)
Ácido Algínico/farmacología , Encéfalo/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Microbioma Gastrointestinal , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Administración Oral , Animales , Barrera Hematoencefálica , Inflamación , Intestinos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , ARN Ribosómico 16S/genética , RNA-Seq , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Front Neurosci ; 13: 1168, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31787867

RESUMEN

Neuroinflammation plays a pivotal role in the pathogenesis of Parkinson's disease. Ginsenoside Rg1, the most active ingredient of ginseng, has been reported to exert neuroprotective effects via estrogen and glucocorticoid receptors. The present study evaluated the involvement of the G protein-coupled estrogen receptor (GPER) in the anti-inflammatory effects of ginsenoside Rg1 against lipopolysaccharide (LPS)-induced microglia activation in the BV2 microglial cell line and ventral mesencephalic primary microglial culture. The pharmacological blockade and lentivirus-mediated small interfering RNA (siRNA) knockdown of GPER were used to study the underlying mechanism. Rg1 attenuated LPS-induced upregulation of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) mRNA and protein levels. The GPER antagonist G15 blocked the inhibitory effects of Rg1 and the GPER-specific agonist G1 on LPS-induced microglia activation. Rg1 mimicked the effects of G1 by inhibiting the LPS-induced activation of nuclear transcription factor-kappa B (NF-κB) and mitogen activated protein kinase signaling pathways, which was also blocked by G15. Moreover, lentivirus-mediated siRNA knockdown of GPER inhibited the anti-inflammatory effects of Rg1. Taken together, our results indicate that GPER is involved in the anti-inflammatory effects of Rg1 against LPS-induced microglia activation. These findings provide a new biological target of Rg1 for the treatment of neuroinflammatory disorders.

10.
Phytomedicine ; 43: 11-20, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29747742

RESUMEN

BACKGROUND: Genistein (GEN), a phytoestrogen that is extracted from leguminous plants, can bind to estrogen receptor and exert biological effects. G protein-coupled estrogen receptor (GPER), a novel membrane estrogen receptor, has been reported to be involved in the anti-inflammatory process. In the present study, using BV2 microglial cell line and primary microglial culture, we evaluated the involvement of GPER in the anti-inflammatory effects of genistein against lipopolysaccharide (LPS)-induced microglia activation. METHODS: The anti-inflammatory effects of genistein were investigated in LPS-induced microglial activation in murine BV2 microglial cell line and primary microglial culture. Anti-inflammatory properties of genistein were determined by MTT, real time PCR, ELISA and western blot analysis. The pharmacological blockade and lentivirus-mediated siRNA knockdown of GPER were used to study the underlying mechanism. RESULTS: The results showed that genistein exerted inhibitory effects on LPS-induced expressions of cyclooxygenase-2 (COX-2), inducible nitric oxide (iNOS), tumor necrosis factor-α (TNF-α), interleukin-1 ß (IL-1ß) and interleukin-6 (IL-6). Pre-treatment with GPER antagonist G15 could significantly block the anti-inflammatory effects of genistein. Moreover, the inhibitory effects of genistein on LPS-induced activation of MAPKs and NF-κB signaling pathways could also be blocked by G15. Lentivirus-mediated siRNA knockdown of GPER significantly inhibited the anti-inflammatory effects of genistein in BV2 cells. Further study revealed that genistein treatment could increase the gene and protein expressions of GPER in BV2 cells. CONCLUSION: Taken together, these data provide the first evidence that genistein exerts anti-inflammatory effects in microglial cells via GPER activation. These beneficial effects of genistein may represent a new strategy for the treatment of neuroinflammatory diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Genisteína/farmacología , Microglía/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Línea Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas Sprague-Dawley , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
11.
Biomed Pharmacother ; 91: 656-663, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28494419

RESUMEN

Flavonoids, the active components of Epimedii Genus, have been demonstrated to protect against osteoporosis, cardiovascular diseases and rheumatoid arthritis. The present study aimed to investigate the neuroprotective effects of total flavonoid (TF) fraction of Epimedium koreanum Nakai on dopaminergic neurons in the cellular and mice models of Parkinson's disease (PD). TF pretreatment could ameliorate the decrease of striatal dopamine (DA) content and the loss of tyrosine hydroxylase (TH)-immunoreactive neurons in the substantia nigra pars compacta (SNpc) induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). TF treatment could reverse the changes of Bcl-2 and Bax protein expressions in the striatum of PD mice. 1-Methyl-4-phenylpyridinium ion (MPP+) significantly decreased the cell viability and mitochondrial membrane potential in MES23.5 cells. These effects could be reversed by TF treatment. In addition, MPP+-induced changes of Bcl-2 and Bax mRNA and protein expressions were also reversed by TF pretreatment. These data demonstrated that TF of E. koreanum Nakai could protect against MPTP-induced dopaminergic neuronal death in mice and MPP+-induced neurotoxicity in dopaminergic MES23.5 cells. Anti-apoptosis might be involved in this process.


Asunto(s)
Neuronas Dopaminérgicas/patología , Epimedium/química , Flavonoides/farmacología , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Flavonoides/uso terapéutico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/uso terapéutico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
12.
Phytomedicine ; 25: 93-99, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-28190476

RESUMEN

BACKGROUND: Epimedium sagittatum is a traditional Chinese herb normally which is used to treat the osteoporosis, cardiovascular dysfunction, and to improve neurological and sexual function in China, Korea and Japan. Icariin is the major active ingredient in Epimedium sagittatum. In the present research, we examined the neuroprotective effects of icariin on dopaminergic neurons and the possible mechanisms in a mouse model of Parkinson's disease (PD). METHODS: Ovariectomized PD mice were treated with vehicle or icariin (3 days before MPTP injections) with or without the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 or mitogen-activated protein kinase kinase (MEK) inhibitor PD98059. The dopamine (DA) content in the striatum was studied by HPLC. Western blot was used to determine the protein expressions of Bcl-2, Bax and Caspase 3 in the striatum. The numbers of tyrosine hydroxylase-immunoreactive (TH-IR) neurons in the substantial nigra pars compacta (SNpc) were assessed by immunohistochemistry. The activation of Akt and ERK by icariin were detected in doparminergic MES23.5 cells. RESULTS: Icariin pretreatment could ameliorate the decreased striatum DA content and the loss of TH-IR neurons in the SNpc induced by MPTP. The MPTP-induced changes of Bcl-2, Bax and caspase 3 protein expressions in the striatum could be reversed by icariin pretreatment. Blockade of PI3K/Akt or MEK/ERK signaling pathway by LY294002 or PD98059 could attenuate the increase of DA content in the striatum and TH-IR in the SNpc induced by icariin in PD mice model. Additionally, icariin treatment alone significantly induced the phosphorylation of Akt and ERK in a time dependent pattern in dopaminergic MES 23.5 cells. These effects were abolished by co-treatment with LY294002 or PD98059. CONCLUSION: These data demonstrated that icariin has neuroprotective effect on dopaminergic neurons in PD mice model and the potential mechanisms might be related to PI3K/Akt and MEK/ERK pathways.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Flavonoides/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Animales , Cromonas/farmacología , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Inhibidores Enzimáticos/farmacología , Epimedium/química , Femenino , Ratones Endogámicos C57BL , Morfolinas/farmacología , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/etiología , Fosforilación/efectos de los fármacos , Fitoterapia , Transducción de Señal/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
14.
Artículo en Chino | MEDLINE | ID: mdl-24175548

RESUMEN

OBJECTIVE: To observe and compare the effects of 17beta-estradiol (EST) on the phasic and tonic contractile activities of the uterine smooth muscles of SD rats in vitro. METHODS: Different concentrations of 17beta-estradiol were added into the perfusion muscular sockets containing uterine smooth muscles of SD rats, and the activities of muscle contraction were recorded at the same time. RESULTS: 17beta-estradiol had obvious depression effects on spontaneous rhythmic contraction of the uterine smooth muscles in a concentration-dependent manner, it could considerably decrease muscular tension, the mean amplitudes and frequencies of contractile waves (P < 0.01); it could also suppress the uterine contraction stimulated by KCl, CaCl2 or prostaglandin F2alpha (PGF2alpha). Based on the contraction of uterine smooth muscle stimulated by KCl, IC50 was 7.278 micromol/L and pD2 was -0.862 when calculated by linear regression method. 17beta-estradiol could also inhibit the maximal CaC12 contraction of uterine smooth muscle in the Ca2+ free Krebs solution, which the ECQ was 1.422 x 10(-3) mol/L, pD2 was 2.847 (control), but the E50 was 3.028 x 10(-3) mol/L, p2 was 2.519 (added with EST) when calculated by linear regression method. CONCLUSION: The depression effects of 17beta-estradiol on the spontaneous rhythmic contraction and activated contraction of the uterine smooth muscles of SD rats could be mediated through the blockage of C2+ influx through potential-dependent Ca2+ channels of plasma membrane.


Asunto(s)
Estradiol/farmacología , Músculo Liso/efectos de los fármacos , Contracción Uterina/efectos de los fármacos , Animales , Femenino , Miometrio/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...