Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurochem Res ; 49(3): 692-705, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38047987

RESUMEN

Narirutin (Nar) is a flavonoid that is abundantly present in citrus fruits and has attracted considerable attention because of its diverse pharmacological activities and low toxicity. Here, we evaluated the preventive effects of Nar in middle cerebral artery occlusion/reperfusion (MCAO/R)-injured mice and oxygen-glucose deprivation/reperfusion (OGD/R)-injured bEnd.3 cells. Pretreatment with Nar (150 mg/kg) for 7 days effectively reduced infarct volume, improved neurological deficits, and significantly inhibited neuronal death in the hippocampus and cortex in MCAO/R-injured mice. Moreover, anti-apoptotic effects of Nar (50 µM) were observed in OGD/R-injured bEnd.3 cells. In addition, Nar pre-administration regulated blood-brain barrier function by increasing tight junction-related protein expression after MCAO/R and OGD/R injury. Nar also inhibited NOD-like receptor protein 3 (NLRP3) inflammasome activation by reducing the expression of thioredoxin-interacting protein (TXNIP) in vivo and in vitro. Taken together, these results provide new evidence for the use of Nar in the prevention and treatment of ischemic stroke.


Asunto(s)
Isquemia Encefálica , Disacáridos , Flavanonas , Daño por Reperfusión , Ratas , Ratones , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas NLR , Ratas Sprague-Dawley , Células Endoteliales/metabolismo , Inflamasomas/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/prevención & control , Daño por Reperfusión/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/prevención & control , Isquemia Encefálica/metabolismo , Proteínas de Ciclo Celular
2.
J Neurochem ; 165(6): 772-790, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37026513

RESUMEN

Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein characterized by its ability to regulate multiple biological processes, such as the inflammatory response and gene transcriptional signaling activation. Abnormal CHI3L1 expression has been associated with multiple neurological disorders and serves as a biomarker for the early detection of several neurodegenerative diseases. Aberrant CHI3L1 expression is also reportedly associated with brain tumor migration and metastasis, as well as contributions to immune escape, playing important roles in brain tumor progression. CHI3L1 is synthesized and secreted mainly by reactive astrocytes in the central nervous system. Thus, targeting astrocytic CHI3L1 could be a promising approach for the treatment of neurological diseases, such as traumatic brain injury, ischemic stroke, Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, and glioma. Based on current knowledge of CHI3L1, we assume that it acts as a molecule mediating several signaling pathways driving the initiation and progression of neurological disorders. This narrative review is the first to introduce the potential roles of astrocytic CHI3L1 in neurological disorders. We also equally explore astrocytic CHI3L1 mRNA expression under physiological and pathological conditions. Inhibiting CHI3L1 and disrupting its interaction with its receptors through multiple mechanisms of action are briefly discussed. These endeavors highlight the pivotal roles of astrocytic CHI3L1 in neurological disorders and could contribute to the development of effective inhibitors based on the strategy of structure-based drug discovery, which could be an attractive therapeutic approach for neurological disease treatment.


Asunto(s)
Neoplasias Encefálicas , Quitinasas , Enfermedades Neurodegenerativas , Humanos , Proteína 1 Similar a Quitinasa-3/metabolismo , Astrocitos/metabolismo , Quitinasas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Sinapsinas/metabolismo , Neoplasias Encefálicas/metabolismo
3.
Mol Pain ; 19: 17448069231177634, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37207346

RESUMEN

Chronic pain, along with comorbid psychiatric disorders, is a common problem worldwide. A growing number of studies have focused on non-opioid-based medicines, and billions of funds have been put into digging new analgesic mechanisms. Peripheral inflammation is one of the critical causes of chronic pain, and drugs with anti-inflammatory effects usually alleviate pain hypersensitivity. Sophoridine (SRI), one of the most abundant alkaloids in Chinese herbs, has been proved to exert antitumor, antivirus and anti-inflammation effects. Here, we evaluated the analgesic effect of SRI in an inflammatory pain mouse model induced by complete Freund's adjuvant (CFA) injection. SRI treatment significantly decreased pro-inflammatory factors release after LPS stimuli in microglia. Three days of SRI treatment relieved CFA-induced mechanical hypersensitivity and anxiety-like behavior, and recovered abnormal neuroplasticity in the anterior cingulate cortex of mice. Therefore, SRI may be a candidate compound for the treatment of chronic inflammatory pain and may serve as a structural basis for the development of new drugs.


Asunto(s)
Dolor Crónico , Hiperalgesia , Ratones , Animales , Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/inducido químicamente , Adyuvante de Freund/toxicidad , Matrinas , Dolor Crónico/tratamiento farmacológico , Analgésicos/farmacología , Analgésicos/uso terapéutico , Ansiedad/complicaciones , Ansiedad/tratamiento farmacológico
4.
Phytother Res ; 37(10): 4838-4850, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37458182

RESUMEN

Diabetic encephalopathy is a common consequence of diabetes mellitus that causes cognitive dysfunction and neuropsychiatric disorders. Praeruptorin C (Pra-C) from the traditional Chinese medicinal herb Peucedanum praeruptorum Dunn. is a potential antioxidant and neuroprotective agent. This study was conducted to investigate the molecular mechanisms underlying the effect of Pra-C on diabetic cognitive impairment. A novel object recognition test and the Morris water maze test were performed to assess the behavioral performance of mice. Electrophysiological recordings were made to monitor synaptic plasticity in the hippocampus. A protein-protein interaction network of putative Pra-C targets was constructed, and molecular docking simulations were performed to predict the potential mechanisms of the action of Pra-C. Protein expression levels were detected by western blotting. Pra-C administration significantly lowered body weight and fasting blood glucose levels and alleviated learning and memory deficits in type 2 diabetic mice. Network pharmacology and molecular docking results suggested that Pra-C affects the PI3K/AKT/GSK3ß signaling pathway. Western blot analysis confirmed significant increases in phosphorylated PI3K, AKT, and GSK3ß levels in vivo and in vitro upon Pra-C administration. Pra-C alleviated cognitive impairment in type 2 diabetic mice by activating PI3K/AKT/GSK3ß pathway.

5.
Clin Immunol ; 245: 109158, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36244672

RESUMEN

Effective immunotherapies for patients with glioblastoma (GBM) are urgently needed. Chitinase-3 like-protein-1 (CHI3L1) play important roles in the development of gliomas. However, its role in glioma-related immune responses remains unclear. We aimed to comprehensively investigate its biological features and clinical value in glioma, especially in GBM. Transcriptome, proteome and single-cell RNA-sequencing databases were enrolled in the study. Immunostaining and immunoblotting were performed for validation. CHI3L1 was highly correlated with clinical and molecular features, suggesting that high CHI3L1 expression is more likely to be predicted as malignant entities. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed CHI3L1 closely associated with immune responses and inflammatory activities in GBM. In addition, CHI3L1 also correlated with immune cell infiltration and immune checkpoints. Our study suggests that inhibition of CHI3L1 may help to reduce immunosuppression and overcome immunotherapy resistance, which would be an therapeutic area for the treatment of GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioblastoma/genética , Glioblastoma/patología , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Ontología de Genes , Microambiente Tumoral , Proteína 1 Similar a Quitinasa-3/genética
6.
Phytother Res ; 36(10): 3932-3948, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35801985

RESUMEN

Posttraumatic stress disorder (PTSD) is one of the most common psychiatric diseases, which is characterized by the typical symptoms such as re-experience, avoidance, and hyperarousal. However, there are few drugs for PTSD treatment. In this study, conditioned fear and single-prolonged stress were employed to establish PTSD mouse model, and we investigated the effects of Tanshinone IIA (TanIIA), a natural product isolated from traditional Chinese herbal Salvia miltiorrhiza, as well as the underlying mechanisms in mice. The results showed that the double stress exposure induced obvious PTSD-like symptoms, and TanIIA administration significantly decreased freezing time in contextual fear test and relieved anxiety-like behavior in open field and elevated plus maze tests. Moreover, TanIIA increased the spine density and upregulated synaptic plasticity-related proteins as well as activated CREB/BDNF/TrkB signaling pathway in the hippocampus. Blockage of CREB remarkably abolished the effects of TanIIA in PTSD model mice and reversed the upregulations of p-CREB, BDNF, TrkB, and synaptic plasticity-related protein induced by TanIIA. The molecular docking simulation indicated that TanIIA could interact with the CREB-binding protein. These findings indicate that TanIIA ameliorates PTSD-like behaviors in mice by activating the CREB/BDNF/TrkB pathway, which provides a basis for PTSD treatment.


Asunto(s)
Productos Biológicos , Factor Neurotrófico Derivado del Encéfalo , Abietanos , Animales , Ansiedad/tratamiento farmacológico , Productos Biológicos/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a CREB/metabolismo , Proteína de Unión a CREB/farmacología , Miedo , Hipocampo/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Transducción de Señal
7.
Mol Pain ; 17: 1744806921990934, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33590786

RESUMEN

Chronic pain is highly prevalent worldwide and severely affects daily lives of patients and family members. Praeruptorin C (Pra-C) is a main active ingredient derived from Peucedanum praeruptorum Dunn, traditionally used as antibechic, anti-bronchitis and anti-hypertension drug. Here, we evaluated the effects of Pra-C in a chronic inflammatory pain mouse model induced by complete Freund's adjuvant (CFA) injection. Pra-C (3 mg/kg) treatment for just 3 days after CFA challenge relieved CFA-induced mechanical allodynia and hindpaw edema in mice. In the anterior cingulate cortex (ACC), Pra-C treatment inhibited microglia activation and reduced levels of proinflammatory cytokines, TNF-α and IL-1ß, and suppressed upregulation of glutamate receptors caused by CFA injection. In addition, Pra-C attenuated neuronal hyperexcitability in ACC of CFA-injected mice. In vitro studies confirmed the analgesic effect of Pra-C was due to its inhibitory ability on microglial activation. In conclusion, Pra-C administration had a certain effect on relieving chronic pain by inhibiting microglial activation, attenuating proinflammatory cytokine releasing and regulating excitatory synaptic proteins in the ACC of the CFA-injected mice.


Asunto(s)
Analgésicos/farmacología , Cumarinas/farmacología , Giro del Cíngulo/patología , Microglía/patología , Analgésicos/uso terapéutico , Animales , Línea Celular , Dolor Crónico/complicaciones , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/fisiopatología , Cumarinas/química , Cumarinas/uso terapéutico , Citocinas/metabolismo , Modelos Animales de Enfermedad , Edema/complicaciones , Edema/patología , Edema/fisiopatología , Adyuvante de Freund , Hiperalgesia/complicaciones , Hiperalgesia/patología , Hiperalgesia/fisiopatología , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Transmisión Sináptica/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
8.
Phytother Res ; 35(7): 3936-3944, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33856723

RESUMEN

Anxiety disorders are a common frequently psychiatric symptom in patients that lead to disruption of daily life. Scutellarin (Scu) is the main component of Erigeron breviscapus, which has been used as a neuroprotective agent against glutamate-induced excitotoxicity. However, the potential effect of Scu on the stress-related neuropsychological disorders has not been clarified. In this study, Anxiety-like behavior was induced by acute restraint stress in mice. Scu were injected intraperitoneally (twice daily, 3 days). Results showed that Scu exhibited good protective activity on mice by decreasing transmitter release levels. Restraint stress caused significant anxiety like behavior in mice. Treatment of Scu could significantly improve the moving time of open arms in Elevated Plus Maze and central time on open field test. Scu treatment suppressed action potential firing frequency, restored excessive presynaptic quantal release, and down-regulated glutamatergic receptor expression levels in the prefrontal cortex (PFC) of stressed mice. GABAA Rα1 and GABAA γ2 expression in the brain PFC tissues of mice were nearly abrogated by Scu treatment. In stress-induced anxiety mice, stress can increase the frequency of mini excitatory postsynaptic currents (mEPSC), which can be reversed by Scu treatment. Therefore, Scu has a potent anxiolytic activity and may be valuable for the treatment of stress-induced anxiety disorders.


Asunto(s)
Ansiedad , Apigenina , Glucuronatos , Neurotransmisores/fisiología , Animales , Ansiedad/tratamiento farmacológico , Apigenina/farmacología , Glucuronatos/farmacología , Ratones
9.
Glia ; 68(1): 27-43, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31429156

RESUMEN

Ischemic stroke leads to neuronal damage induced by excitotoxicity, inflammation, and oxidative stress. Astrocytes play diverse roles in stroke and ischemia-induced inflammation, and autophagy is critical for maintaining astrocytic functions. Our previous studies showed that the activation of G protein-coupled receptor 30 (GPR30), an estrogen membrane receptor, protected neurons from excitotoxicity. However, the role of astrocytic GPR30 in maintaining autophagy and neuroprotection remained unclear. In this study, we found that the neuroprotection induced by G1 (GPR30 agonist) in wild-type mice after a middle cerebral artery occlusion was completely blocked in GPR30 conventional knockout (KO) mice but partially attenuated in astrocytic or neuronal GPR30 KO mice. In cultured primary astrocytes, glutamate exposure induced astrocyte proliferation and decreased astrocyte autophagy by activating mammalian target of rapamycin (mTOR) and c-Jun N-terminal kinase (JNK) and inhibiting p38 mitogen-activated protein kinase (MAPK) pathway. G1 treatment restored autophagy to its basal level by regulating the p38 pathway but not the mTOR and JNK signaling pathways. Our findings revealed a key role of GPR30 in neuroprotection via the regulation of astrocyte autophagy and support astrocytic GPR30 as a potential drug target against ischemic brain damage.


Asunto(s)
Astrocitos/metabolismo , Autofagia/fisiología , Fármacos Neuroprotectores/farmacología , Quinolinas/farmacología , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Astrocitos/efectos de los fármacos , Autofagia/efectos de los fármacos , Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/uso terapéutico , Quinolinas/uso terapéutico , Receptores Acoplados a Proteínas G/agonistas
10.
J Neurochem ; 153(2): 252-263, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31665810

RESUMEN

Cortical areas including the anterior cingulate cortex (ACC) play critical roles in different types of chronic pain. Most of previous studies focus on the sensory inputs from somatic areas, and less information about plastic changes in the cortex for visceral pain. In this study, chronic visceral pain animal model was established by injection with zymosan into the colon of adult male C57/BL6 mice. Whole cell patch-clamp recording, behavioral tests, western blot, and Cannulation and ACC microinjection were employed to explore the role of adenylyl cyclase 1 (AC1) in the ACC of C57/BL6 and AC1 knock out mice. Integrative approaches were used to investigate possible changes of neuronal AC1 in the ACC after the injury. We found that AC1, a key enzyme for pain-related cortical plasticity, was significantly increased in the ACC in an animal model of irritable bowel syndrome. Inhibiting AC1 activity by a selective AC1 inhibitor NB001 significantly reduced the up-regulation of AC1 protein in the ACC. Furthermore, we found that AC1 is required for NMDA GluN2B receptor up-regulation and increases of NMDA receptor-mediated currents. These results suggest that AC1 may form a positive regulation in the cortex during chronic visceral pain. Our findings demonstrate that the up-regulation of AC1 protein in the cortex may underlie the pathology of chronic visceral pain; and inhibiting AC1 activity may be beneficial for the treatment of visceral pain.


Asunto(s)
Adenilil Ciclasas/metabolismo , Corteza Cerebral/metabolismo , AMP Cíclico/metabolismo , Retroalimentación Fisiológica/fisiología , Dolor Visceral/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Transducción de Señal/fisiología
11.
Brain Behav Immun ; 89: 245-255, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32621847

RESUMEN

Microglia are the resident immune cells of the center nervous system and participate in various neurological diseases. Here we determined the function of microglia in epileptogenesis using microglial ablation approaches. Three different microglia-specific genetic tools were used, CX3CR1CreER/+:R26iDTA/+, CX3CR1CreER/+:R26iDTR/+, and CX3CR1CreER/+:Csf1rFlox/Flox mice. We found that microglial depletion led to worse kainic acid (KA)-induced status epilepticus, higher mortality rate, and increased neuronal degeneration in the hippocampus. In KA-induced chronic spontaneous recurrent seizures, microglial depletion increased seizure frequency, interictal spiking, and seizure duration. Therefore, microglial depletion aggravates the severity of KA-induced acute and chronic seizures. Interestingly, microglial repopulation reversed the effects of depletion upon KA-induced status epilepticus. Our results demonstrate a beneficial role of microglia in suppressing both acute and chronic seizures, suggesting that microglia are a potential therapeutic target for the management of epilepsy.


Asunto(s)
Microglía , Estado Epiléptico , Animales , Modelos Animales de Enfermedad , Hipocampo , Ácido Kaínico , Ratones , Ratones Transgénicos , Convulsiones
12.
J Neuroinflammation ; 16(1): 132, 2019 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-31255170

RESUMEN

BACKGROUND: Liver X receptors (LXRs), including LXRα and LXRß, are key regulators of transcriptional programs for both cholesterol homeostasis and inflammation in the brain. Here, the modes of action of LXRs and the epigenetic mechanisms regulating LXRß expression in anterior cingulate cortex (ACC) of chronic inflammatory pain (CIP) are investigated. METHODS: The deficit of LXR isoform and analgesic effect of LXR activation by GW3965 were evaluated using the mouse model of CIP induced by hindpaw injection of complete Freund's adjuvant (CFA). The mechanisms involved in GW-mediated analgesic effects were analyzed with immunohistochemical methods, ELISA, co-immunoprecipitation (Co-IP), Western blot, and electrophysiological recording. The epigenetic regulation of LXRß expression was investigated by chromatin immunoprecipitation, quantitative real-time PCR, and sequencing. RESULTS: We revealed that CFA insult led to LXRß reduction in ACC, which was associated with upregulated expression of histone deacetylase 5 (HDAC5), and knockdown of LXRß by shRNA led to thermal hyperalgesia. Co-IP showed that LXRß interacted with NF-κB p65 physically. LXRß activation by GW3965 exerted analgesic effects by inhibiting the nuclear translocation of NF-κB, reducing the phosphorylation of mitogen-activated protein kinases (MAPKs) in ACC, and decreasing the promoted input-output and enhanced mEPSC frequency in ACC neurons after CFA exposure. In vitro experiments confirmed that HDAC5 triggered histone deacetylation on the promoter region of Lxrß, resulting in downregulation of Lxrß transcription. CONCLUSION: These findings highlight an epigenetic mechanism underlying LXRß deficits linked to CIP, and LXRß activation may represent a potential novel target for the treatment of CIP with an alteration in inflammation responses and synaptic transmission in ACC.


Asunto(s)
Dolor Crónico/metabolismo , Epigénesis Genética/fisiología , Adyuvante de Freund/toxicidad , Giro del Cíngulo/metabolismo , Histona Desacetilasas/biosíntesis , Receptores X del Hígado/metabolismo , Animales , Secuencia de Bases , Dolor Crónico/inducido químicamente , Dolor Crónico/genética , Epigénesis Genética/efectos de los fármacos , Giro del Cíngulo/efectos de los fármacos , Histona Desacetilasas/genética , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Receptores X del Hígado/antagonistas & inhibidores , Receptores X del Hígado/genética , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Toxicol Appl Pharmacol ; 368: 26-36, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30776389

RESUMEN

Cardiac dysfunction is a vital complication during endotoxemia (ETM). Accumulating evidence suggests that enhanced glycolytic metabolism promotes inflammatory and myocardial diseases. In this study, we performed deep mRNA sequencing analysis on the hearts of control and lipopolysaccharide (LPS)-challenged mice (40 mg/kg, i.p.) and identified that the glycolytic enzyme, 6-phosphofructo-2-kinase (PFK-2)/fructose-2,6-bisphosphatase 3 (PFKFB3) might play an indispensable role in ETM-induced cardiac damage. Quantitative real-time PCR validated the transcriptional upregulation of PFKFB3 in the myocardium of LPS-challenged mice and immunoblotting and immunostaining assays confirmed that LPS stimulation markedly increased the expression of PFKFB3 at the protein level both in vivo and in vitro. The potent antagonist 3-(3pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) was used to block PFKFB3 activity in vivo (50 mg/kg, i.p.) and in vitro (10 µM). Echocardiographic analysis and TUNEL staining showed that 3PO significantly alleviated LPS-induced cardiac dysfunction and apoptotic injury in vivo. 3PO also suppressed the LPS-induced secretion of tumor necrosis factor-α, interleukin (IL)-1ß, IL-6 and lactate in the serum, in addition to lactate in the myocardium. PFKFB3 inhibition also diminished the nuclear translocation and phosphorylation of transcription factor nuclear factor-κB (NF-κB) in both adult cardiomyocytes and HL-1 cells. Furthermore, immunoblotting analysis showed that 3PO inhibited LPS-induced apoptotic induction in cardiomyocytes. Taken together, these findings demonstrate that PFKFB3 participates in LPS-induced cardiac dysfunction via mediating inflammatory and apoptotic signaling pathway.


Asunto(s)
Apoptosis , Endotoxemia/enzimología , Cardiopatías/enzimología , Mediadores de Inflamación/metabolismo , Miocitos Cardíacos/enzimología , Fosfofructoquinasa-2/metabolismo , Animales , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Endotoxemia/inducido químicamente , Endotoxemia/patología , Endotoxemia/prevención & control , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica , Cardiopatías/inducido químicamente , Cardiopatías/patología , Cardiopatías/prevención & control , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fosfofructoquinasa-2/antagonistas & inhibidores , Fosfofructoquinasa-2/genética , Piridinas/farmacología , Transducción de Señal
14.
Mol Pain ; 14: 1744806918814367, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30380983

RESUMEN

Tetrahydroxystilbene glucoside (THSG) is one of the active ingredients of Polygonum multiflorum. It has been shown to exert a variety of pharmacological effects, including antioxidant, anti-aging, and anti-atherosclerosis. Because of its prominent anti-inflammatory effect, we explored whether THSG had analgesic effect. In this study, we used a model of chronic inflammatory pain caused by injecting complete Freund's adjuvant into the hind paw of mice. We found THSG relieved swelling and pain in the hind paw of mice on a dose-dependent manner. In the anterior cingulate cortex, THSG suppressed the upregulation of GluN2B-containing N-methyl-D-aspartate receptors and the downregulation of GluN2A-containing N-methyl-D-aspartate receptors caused by chronic inflammation. In addition, THSG increased Bcl-2 and decreased Bax and Caspase-3 expression by protecting neuronal survival. Furthermore, THSG inhibited the phosphorylation of p38 and the increase of nuclear factor κB (NF-κB) and tumor necrosis factor α (TNF-α). Immunohistochemical staining revealed that THSG blocked the activation of microglia and reduced the release of proinflammatory cytokines TNF-α, interleukin 1ß (IL-1ß), and interleukin 6 (IL-6). In conclusion, this study demonstrated that THSG had a certain effect on alleviating complete Freund's adjuvant-induced chronic inflammatory pain.


Asunto(s)
Apoptosis , Dolor Crónico/tratamiento farmacológico , Glucósidos/uso terapéutico , Giro del Cíngulo/metabolismo , Giro del Cíngulo/patología , Inflamación/tratamiento farmacológico , Microglía/patología , Receptores de N-Metil-D-Aspartato/metabolismo , Estilbenos/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dolor Crónico/complicaciones , Dolor Crónico/patología , Citocinas/metabolismo , Edema/tratamiento farmacológico , Adyuvante de Freund/administración & dosificación , Glucósidos/química , Glucósidos/farmacología , Giro del Cíngulo/efectos de los fármacos , Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Inflamación/complicaciones , Inflamación/patología , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Estilbenos/química , Estilbenos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Brain ; 140(12): 3215-3232, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29096020

RESUMEN

Fragile X syndrome is an inheritable form of intellectual disability caused by loss of fragile X mental retardation protein (FMRP, encoded by the FMR1 gene). Absence of FMRP caused overexpression of progranulin (PGRN, encoded by GRN), a putative tumour necrosis factor receptor ligand. In the present study, we found that progranulin mRNA and protein were upregulated in the medial prefrontal cortex of Fmr1 knock-out mice. In Fmr1 knock-out mice, elevated progranulin caused insufficient dendritic spine pruning and late-phase long-term potentiation in the medial prefrontal cortex of Fmr1 knock-out mice. Partial progranulin knock-down restored spine morphology and reversed behavioural deficits, including impaired fear memory, hyperactivity, and motor inflexibility in Fmr1 knock-out mice. Progranulin increased levels of phosphorylated glutamate ionotropic receptor GluA1 and nuclear factor kappa B in cultured wild-type neurons. Tumour necrosis factor receptor 2 antibody perfusion blocked the effects of progranulin on GluA1 phosphorylation; this result indicates that tumour necrosis factor receptor 2 is required for progranulin-mediated GluA1 phosphorylation and late-phase long-term potentiation expression. However, high basal level of progranulin in Fmr1 knock-out mice prevented further facilitation of synaptic plasticity by exogenous progranulin. Partial downregulation of progranulin or tumour necrosis factor receptor 2/nuclear factor kappa B signalling restored synaptic plasticity and memory deficits in Fmr1 knock-out mice. These findings suggest that elevated PGRN is linked to cognitive deficits of fragile X syndrome, and the progranulin/tumour necrosis factor receptor 2 signalling pathway may be a putative therapeutic target for improving cognitive deficits in fragile X syndrome.


Asunto(s)
Conducta Animal , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Aprendizaje , Sinapsis/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Western Blotting , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Granulinas , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Ratones , Ratones Noqueados , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Corteza Prefrontal/metabolismo , Progranulinas , ARN Mensajero/metabolismo , Receptores AMPA/efectos de los fármacos , Receptores AMPA/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Transducción de Señal
16.
Metab Brain Dis ; 33(5): 1413-1420, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29948656

RESUMEN

Targeting neuroinflammatory disturbances has been acknowledged as a potential strategy for treatment of depressive disorder in humans. Over-activation of tryptophan-degrading pathway by pro-inflammatory cytokines resulted in N-methyl-d-aspartate (NMDA)-mediated excitotoxicity, which is implicated in pathophysiology of depression. Gentiopicroside (Gent) has powerful anti-inflammatory property and exhibits promising antidepressant effect in an animal model of pain/depression dyad by down-regulating GluN2B-containing NMDA receptors. Therefore, the present study aimed to investigate the ability of Gent to abolish depressive-like behavior induced by lipopolysaccharide (LPS) in mice. Acute administration of LPS (0.5 mg/kg, i.p.) increased immobility time in both forced swimming test (FST) and tail suspension test (TST) without affecting spontaneous locomotor activity, indicative of depressive-like behavior. Gent (50 mg/kg, i.p.) administered once a day for three consecutive days prevented the development of depressive-like behavior induced by LPS. The antidepressant-like effect was paralleled with restoration of LPS-induced alterations in brain inflammatory mediators (i.e. IL-1ß and TNF-α). In addition, Gent prevented over-activation of indoleamine 2,3-double oxygen enzyme (IDO) and recovered GluN2B subunit expression in the PFC challenged by LPS. In conclusion, our results suggested that Gent pretreatment provided protection against LPS-induced depressive-like behavior and the effect appeared to be demonstrated, at least partially, by blocking various steps of tryptophan-degrading pathway.


Asunto(s)
Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Depresión/tratamiento farmacológico , Glucósidos Iridoides/farmacología , Actividad Motora/efectos de los fármacos , Animales , Antidepresivos/uso terapéutico , Depresión/inducido químicamente , Modelos Animales de Enfermedad , Suspensión Trasera , Mediadores de Inflamación/metabolismo , Glucósidos Iridoides/uso terapéutico , Lipopolisacáridos , Masculino , Ratones , Transducción de Señal/efectos de los fármacos , Natación , Triptófano/metabolismo
17.
Metab Brain Dis ; 32(1): 259-265, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27670769

RESUMEN

Isopsoralen is a type of furocoumarin that exhibits estrogen-like activities. The aim of this study was to determine the estrogen-like neuroprotection of isopsoralen in an animal model of spinal cord injury (SCI). Results indicated that isopsoralen (intraperitoneal injection of 5 and 10 mg/kg per day for two weeks) significantly enhanced the hindlimb locomotor functions of mice with SCI, as revealed in the BMS score and angle of inclined plane tests. Morphological data showed that isopsoralen significantly attenuated the injury of the gray matter of spinal cord and induced the up-regulation of ERα levels. The neuroprotective effects of isopsolaren were blocked by the ERα antagonist MPP (0.3 mg/kg), but not by the ERß receptor antagonist PHTPP (0.3 mg/kg). Isopsolaren treatment increased phosphorylated PI3K and AKT (P-PI3K and P-AKT) in the spinal cord of SCI mice and showed a significant anti-apoptotic activity. These results suggest that isopsoralen performs estrogen-like neuroprotection against SCI-induced apoptosis by activating ERα and regulating the PI3K/AKT pathway.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Furocumarinas/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Médula Espinal/efectos de los fármacos , Animales , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/antagonistas & inhibidores , Furocumarinas/farmacología , Antagonistas de Hormonas/farmacología , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
Metab Brain Dis ; 32(1): 211-219, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27585465

RESUMEN

Sinomenine, an alkaloid originally isolated from the roots of Sinomeniumacutum, is used as a traditional Chinese medicine for rheumatic arthritis. However, little is known about the neuronal mechanisms underlying the analgesic effects of sinomenine in animals with chronic inflammatory pain. In this study, we investigated the persistent inflammatory pain induced by hind paw injection of complete Freund's adjuvant (CFA) in mice, which was reversed by sinomenine administration. In the anterior cingulate cortex (ACC), a region highly associated with chronic pain processing, the upregulation of GluN2B-containing N-methyl-D-aspartate (NMDA) receptors and Ca2+/calmodulin-dependent protein kinase II, total levels of GluA1, and phosphorylation of GluA1 at Ser831 (p-GluA1-Ser831) were reversed by systemically administrating sinomenine. Furthermore, sinomenine treatment downregulated the mammalian target of rapamycin (mTOR) pathway. Increases in p-mTOR, p-p70S6k, p-S6, and p-4EBP, which were induced by chronic inflammation, were all changed. However, sinomenine did not affect the levels of GluN2A-containing NMDA receptors and p-GluA1-Ser845, as well as the total levels of mTOR, p70S6k, S6, and 4EBP. In conclusion, results indicated that sinomenine reduced the chronic inflammatory pain induced by CFA, at least partially by regulating the GluN2B receptors and mTOR signals in the ACC.


Asunto(s)
Analgésicos/uso terapéutico , Dolor Crónico/tratamiento farmacológico , Giro del Cíngulo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Morfinanos/uso terapéutico , Analgésicos/farmacología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Dolor Crónico/inducido químicamente , Adyuvante de Freund , Giro del Cíngulo/metabolismo , Inflamación/inducido químicamente , Ratones , Morfinanos/farmacología , Fosforilación/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
19.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27612915

RESUMEN

BACKGROUND: Cancer pain, especially the one caused by metastasis in bones, is a severe type of pain. Pain becomes chronic unless its causes and consequences are resolved. With improvements in cancer detection and survival among patients, pain has been considered as a great challenge because traditional therapies are partially effective in terms of providing relief. Cancer pain mechanisms are more poorly understood than neuropathic and inflammatory pain states. Chronic inflammatory pain and neuropathic pain are influenced by NB001, an adenylyl cyclase 1 (AC1)-specific inhibitor with analgesic effects. In this study, the analgesic effects of NB001 on cancer pain were evaluated. RESULTS: Pain was induced by injecting osteolytic murine sarcoma cell NCTC 2472 into the intramedullary cavity of the femur of mice. The mice injected with sarcoma cells for four weeks exhibited significant spontaneous pain behavior and mechanical allodynia. The continuous systemic application of NB001 (30 mg/kg, intraperitoneally, twice daily for three days) markedly decreased the number of spontaneous lifting but increased the mechanical paw withdrawal threshold. NB001 decreased the concentrations of cAMP and the levels of GluN2A, GluN2B, p-GluA1 (831), and p-GluA1 (845) in the anterior cingulate cortex, and inhibited the frequency of presynaptic neurotransmitter release in the anterior cingulate cortex of the mouse models. CONCLUSIONS: NB001 may serve as a novel analgesic to treat bone cancer pain. Its analgesic effect is at least partially due to the inhibition of AC1 in anterior cingulate cortex.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Analgésicos/uso terapéutico , Dolor en Cáncer/tratamiento farmacológico , Adenosina Trifosfato/farmacología , Adenosina Trifosfato/uso terapéutico , Animales , Neoplasias Óseas/complicaciones , Neoplasias Óseas/diagnóstico por imagen , Dolor en Cáncer/diagnóstico por imagen , Dolor en Cáncer/etiología , Dolor en Cáncer/patología , Línea Celular Tumoral , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Estudios de Seguimiento , Giro del Cíngulo/patología , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Sarcoma/patología , Sarcoma Experimental/complicaciones , Sarcoma Experimental/diagnóstico por imagen , Transducción de Señal/efectos de los fármacos
20.
Ann Neurol ; 77(4): 618-36, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25611593

RESUMEN

OBJECTIVE: Fragile X syndrome (FXS) is a form of inherited mental retardation in humans that results from expansion of a CGG repeat in the FMR1 gene. Interaction between estrogen receptor (ER) and lipid raft caveolae is critical for the estrogen signaling. Here, we tested the hypothesis that impaired ER-caveolae coupling contributes to the mental retardation of FXS. METHODS: Fmr1 knockout (KO) mouse was used as the model of FXS. Multiple techniques were performed including primary neuronal culture, short hairpin RNA (shRNA) interference, Western blot, electrophysiological recording, RNA-binding protein immunoprecipitation, reverse transcriptase polymerase chain reaction, and behavioral tests. RESULTS: In this study, we report that GluA1 surface expression and phosphorylation induced by 17ß-estradiol (E2) were impaired in the Fmr1 KO neurons. The E2-mediated facilitation of long-term potentiation and fear memory was impaired in the anterior cingulate cortex of Fmr1 KO mice. The increased coupling of caveolin-1 (CAV1) and the membrane estrogen receptor ERα under basal conditions contributed to the impairment of ER signaling in Fmr1 KO neurons. FMRP (fragile X mental retardation protein) interacted with CAV1 mRNA, and knockdown of CAV1 with shRNA rescued the synaptic GluA1 delivery, plasticity, and memory in Fmr1 KO mice. INTERPRETATION: This is the first demonstration that the coupling between ERα and lipid raft CAV1 is critical for membrane ER signaling in synaptic plasticity. Therefore, increased coupling of CAV1 and ERα may elucidate a critical abnormal mechanism of FXS.


Asunto(s)
Caveolina 1/metabolismo , Receptor alfa de Estrógeno/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Animales , Caveolina 1/genética , Receptor alfa de Estrógeno/genética , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Unión Proteica/fisiología , Receptores AMPA/genética , Receptores AMPA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA