Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Infect Microbiol ; 14: 1397724, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38966251

RESUMEN

Cryptococcus neoformans is at the top of the list of "most wanted" human pathogens. Only three classes of antifungal drugs are available for the treatment of cryptococcosis. Studies on antifungal resistance mechanisms are limited to the investigation of how a particular antifungal drug induces resistance to a particular drug, and the impact of stresses other than antifungals on the development of antifungal resistance and even cross-resistance is largely unexplored. The endoplasmic reticulum (ER) is a ubiquitous subcellular organelle of eukaryotic cells. Brefeldin A (BFA) is a widely used chemical inducer of ER stress. Here, we found that both weak and strong selection by BFA caused aneuploidy formation in C. neoformans, mainly disomy of chromosome 1, chromosome 3, and chromosome 7. Disomy of chromosome 1 conferred cross-resistance to two classes of antifungal drugs: fluconazole and 5-flucytosine, as well as hypersensitivity to amphotericin B. However, drug resistance was unstable, due to the intrinsic instability of aneuploidy. We found overexpression of AFR1 on Chr1 and GEA2 on Chr3 phenocopied BFA resistance conferred by chromosome disomy. Overexpression of AFR1 also caused resistance to fluconazole and hypersensitivity to amphotericin B. Furthermore, a strain with a deletion of AFR1 failed to form chromosome 1 disomy upon BFA treatment. Transcriptome analysis indicated that chromosome 1 disomy simultaneously upregulated AFR1, ERG11, and other efflux and ERG genes. Thus, we posit that BFA has the potential to drive the rapid development of drug resistance and even cross-resistance in C. neoformans, with genome plasticity as the accomplice.


Asunto(s)
Aneuploidia , Antifúngicos , Brefeldino A , Cryptococcus neoformans , Farmacorresistencia Fúngica , Cryptococcus neoformans/efectos de los fármacos , Cryptococcus neoformans/genética , Brefeldino A/farmacología , Antifúngicos/farmacología , Farmacorresistencia Fúngica/genética , Fluconazol/farmacología , Anfotericina B/farmacología , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Pruebas de Sensibilidad Microbiana , Flucitosina/farmacología , Humanos , Estrés del Retículo Endoplásmico/efectos de los fármacos
2.
Cancer Med ; 12(10): 11073-11096, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36645225

RESUMEN

Colorectal cancer (CRC) is a common gastrointestinal malignancy with high morbidity and fatality. Chemotherapy, as traditional therapy for CRC, has exerted well antitumor effect and greatly improved the survival of CRC patients. Nevertheless, chemoresistance is one of the major problems during chemotherapy for CRC and significantly limits the efficacy of the treatment and influences the prognosis of patients. To overcome chemoresistance in CRC, many strategies are being investigated. Here, we review the common and novel measures to combat the resistance, including drug repurposing (nonsteroidal anti-inflammatory drugs, metformin, dichloroacetate, enalapril, ivermectin, bazedoxifene, melatonin, and S-adenosylmethionine), gene therapy (ribozymes, RNAi, CRISPR/Cas9, epigenetic therapy, antisense oligonucleotides, and noncoding RNAs), protein inhibitor (EFGR inhibitor, S1PR2 inhibitor, and DNA methyltransferase inhibitor), natural herbal compounds (polyphenols, terpenoids, quinones, alkaloids, and sterols), new drug delivery system (nanocarriers, liposomes, exosomes, and hydrogels), and combination therapy. These common or novel strategies for the reversal of chemoresistance promise to improve the treatment of CRC.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , Humanos , MicroARNs/genética , Resistencia a Antineoplásicos/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Interferencia de ARN , Pronóstico , Línea Celular Tumoral
3.
Front Microbiol ; 13: 930495, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36204612

RESUMEN

Candida albicans is a prevalent, opportunistic human fungal pathogen. It usually dwells in the human body as a commensal, however, once in its pathogenic state, it causes diseases ranging from debilitating superficial to life-threatening systemic infections. The switch from harmless colonizer to virulent pathogen is, in most cases, due to perturbation of the fungus-host-microbiota interplay. In this review, we focused on the interactions between C. albicans and the host microbiota in the mouth, gut, blood, and vagina. We also highlighted important future research directions. We expect that the evaluation of these interplays will help better our understanding of the etiology of fungal infections and shed new light on the therapeutic approaches.

4.
Zhen Ci Yan Jiu ; 43(2): 75-9, 2018 Feb 25.
Artículo en Chino | MEDLINE | ID: mdl-29516693

RESUMEN

OBJECTIVE: To study the protective effect of moxibustion for tripterygium-induced premature ovarian failure (POF) and its underlying mechanisms in rats. METHODS: Forty-five female SD rats were randomly divided into normal control, POF model and moxibustion groups (n=15/group). The POF model was induced by intragastric administration of Triptolide (40 mg/kg), once daily for 6 weeks. From the 4th week after modeling, moxibustion was given at "Guanyuan" (CV 4) and bilateral "Sanyinjiao" (SP 6) for 10 min, once daily for 3 weeks. Pathological changes of ovary tissues were determined by hematoxylin-eosin (HE) staining. The serum estradiol (E2), luteinizing hormone (LH), follicle-stimulating hormone (FSH), interleukin-6 (IL-6) and interleukin-1 ß (IL-1 ß) contents were measured by enzyme-linked immunosorbent assay (ELISA). The expression levels of phosphatidyl inositol 3- kinase (PI 3 K), protein kinase B (Akt) and mammalian target of rapamycin (mTOR) proteins of the ovarian tissue were detected by Western blot. RESULTS: After modeling, HE staining showed that the numbers of ovarian follicles and follicular granulocytes and corpora luteum layers were decreased, and the number of corpora atretica was increased in the model group. The content of serum E2 was markedly decreased and those of serum LH, FSH, IL-6 and IL-1 ß were markedly increased in the model group (P<0.01), and the expression levels of ovarian p-PI 3 K, p-Akt and p-mTOR were markedly increased after modeling relevant to the control group (P<0.01). Following moxibustion, the pathological damage of ovarian tissue was improved, the contents of serum LH, FSH, IL-6, IL-1 ß, and the levels of p-PI 3 K, p-Akt and p-mTOR proteins in the ovarian tissue were significantly decreased (P<0.05, P<0.01), and the content of serum E2 was markedly increased (P<0.05) in comparison with the model group. CONCLUSION: Moxibustion can improve POF in POF rats, which may be related to its actions in inhibiting PI 3 K/Akt/mTOR signaling, down-regulating serum IL-6, IL-1 ß, and regulating serum hormones.


Asunto(s)
Moxibustión , Insuficiencia Ovárica Primaria , Puntos de Acupuntura , Animales , Femenino , Fosfatidilinositol 3-Quinasas , Insuficiencia Ovárica Primaria/terapia , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Sirolimus , Serina-Treonina Quinasas TOR
5.
Acta Pharmacol Sin ; 38(11): 1554-1565, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28770824

RESUMEN

HIV infection is often associated with liver failure, which alters the pharmacokinetics of many drugs. In this study we investigated whether acute liver failure (ALF) altered the pharmacokinetics of the first-line anti-HIV agent zidovudine (AZT), a P-gp/BCRP substrate, in rats. ALF was induced in rats by injecting thioacetamide (TAA, 300 mg·kg-1·d-1, ip) for 2 days. On the second day after the last injection of TAA, the pharmacokinetics of AZT was investigated following both oral (20 mg/kg) and intravenous (10 mg/kg) administration. ALF significantly increased the plasma concentrations of AZT after both oral and intravenous doses of AZT, but without affecting the urinary excretion of AZT. AZT metabolism was studied in rat hepatic microsomes in vitro, which revealed that hepatic UGT2B7 was the main enzyme responsible for the formation of AZT O-glucuronide (GAZT); ALF markedly impaired AZT metabolism in hepatic microsomes, which was associated with the significantly decreased hepatic UGT2B7 expression. Intestinal absorption of AZT was further studied in rats via in situ single-pass intestinal perfusion. Intestinal P-gp function and intestinal integrity were assessed with rhodamine 123 and FD-70, respectively. We found that ALF significantly downregulated intestinal P-gp expression, and had a smaller effect on intestinal BCRP. Further studies showed that ALF significantly increased the intestinal absorption of both rhodamine 123 and AZT without altering intestinal integrity, thus confirming an impairment of intestinal P-gp function. In conclusion, ALF significantly increases the oral plasma exposure of AZT in rats, a result partly attributed to the impaired function and expression of hepatic UGT2B7 and intestinal P-gp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Glucuronosiltransferasa/metabolismo , Yeyuno/metabolismo , Fallo Hepático Agudo/enzimología , Hígado/enzimología , Inhibidores de la Transcriptasa Inversa/administración & dosificación , Inhibidores de la Transcriptasa Inversa/farmacocinética , Zidovudina/administración & dosificación , Zidovudina/farmacocinética , Administración Intravenosa , Administración Oral , Animales , Disponibilidad Biológica , Modelos Animales de Enfermedad , Absorción Intestinal , Masculino , Microsomas Hepáticos/enzimología , Ratas Sprague-Dawley , Eliminación Renal , Inhibidores de la Transcriptasa Inversa/sangre , Tioacetamida , Zidovudina/sangre
6.
Eur J Pharmacol ; 781: 190-7, 2016 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-27085896

RESUMEN

The generation of thrombus is not considered as an isolated progression without other pathologic processes, which may also enhance procoagulant state. The purpose of this study was to assess whether HY023016, a novel dabigatran prodrug and an oral direct thrombin inhibitor, or dabigatran etexilate, another thrombin inhibitor can improve the state of whole blood hypercoagulability in vitro/vivo. By using whole blood flow cytometry we explored the effects of HY023016 and dabigatran etexilate on thrombin and ADP-induced human platelet-leukocyte aggregation generated in vitro. With the method of continuous infusion of thrombin intravenous, we successfully established a rat hypercoagulable model and evaluated the effect of HY023016 or dabigatran etexilate in vivo. HY023016 was able to inhibit thrombin- or ADP-induced platelet P-selectin or CD40L expression, leukocyte CD11b expression and formation of platelet-leukocyte aggregates in dose-dependent manner. Dabigatran etexilate was unable to affect ADP-induced platelet P-selectin or CD40L expression, leukocyte CD11b expression and formation of platelet-leukocyte aggregates. Based on rat hypercoagulable model, dabigatran etexilate could reverse thrombin-induced circulatory system hypercoagulable state in a concentration-dependent manner. Dabigatran etexilate also inhibited electrical stimulation induced formation of arterial thrombus in rat under hypercoagulable state, and extracorporal circulation-induced formation of thrombus in dose-dependent manner. Compared with dabigatran etexilate, HY023016 showed nearly equal or even better antithrombotic activity, regardless of reversing the cycle of rat hypercoagulable state or inhibiting platelet-leukocyte aggregation. In surrmary, HY023016 could effectively improve hypercoagulable state of circulatory system.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrinolíticos/farmacología , Animales , Antígeno CD11b/metabolismo , Modelos Animales de Enfermedad , Fibrinolíticos/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Nocicepción/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Ratas , Trombina/administración & dosificación , Trombina/farmacología , Trombosis/tratamiento farmacológico , Trombosis/fisiopatología
7.
Acta Pharmacol Sin ; 37(4): 561-70, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26838075

RESUMEN

AIM: Caderofloxacin is a new fluoroquinolone that is under phase III clinical trials in China. Here we examined the effects of caderofloxacin on rat hepatic cytochrome P450 (CYP450) isoforms as well as the potential of caderofloxacin interacting with co-administered drugs. METHODS: Male rats were treated with caderofloxacin (9 mg/kg, ig) once or twice daily for 14 consecutive days. The effects of caderofloxacin on CYP3A, 2D6, 2C19, 1A2, 2E1 and 2C9 were evaluated using a "cocktail" of 6 probes (midazolam, dextromethorphan, omeprazole, theophylline, chlorzoxazone and diclofenac) injected on d 0 (prior to caderofloxacin exposure) and d 15 (after caderofloxacin exposure). Hepatic microsomes from the caderofloxacin-treated rats were used to assess CYP2E1 activity and chlorzoxazone metabolism. The expression of CYP2E1 mRNA and protein in hepatic microsomes was analyzed with RT-PCR and Western blotting, respectively. RESULTS: Fourteen-day administration of caderofloxacin significantly increased the activity of hepatic CYP2E1, leading to enhanced metabolism of chlorzoxazone. In vitro microsomal study confirmed that CYP2E1 was a major metabolic enzyme involved in chlorzoxazone metabolism, and the 14-d administration of caderofloxacin significantly increased the activity of CYP2E1 in hepatic microsomes, resulting in increased formation of 6-hydroxychlorzoxazone. Furthermore, the 14-d administration of caderofloxacin significantly increased the expression of CYP2E1 mRNA and protein in liver microsomes, which was consistent with the pharmacokinetic results. CONCLUSION: Fourteen-day administration of caderofloxacin can induce the expression and activity of hepatic CYP2E1 in rats. When caderofloxacin is administered, a potential drug-drug interaction mediated by CYP2E1 induction should be considered.


Asunto(s)
Antibacterianos/administración & dosificación , Citocromo P-450 CYP2E1/metabolismo , Fluoroquinolonas/administración & dosificación , Hígado/efectos de los fármacos , Animales , Interacciones Farmacológicas , Hígado/metabolismo , Masculino , Ratas Sprague-Dawley
8.
Br J Pharmacol ; 173(2): 372-85, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26507867

RESUMEN

BACKGROUND AND PURPOSE: There is emerging evidence suggesting that abnormal transport of amyloid-ß (Aß) across the blood-brain barrier (BBB) is involved in diabetes-associated cognitive decline. We investigated whether PPARγ agonists restore Aß transport across the BBB and hippocampal plasticity in db/db mice. EXPERIMENTAL APPROACH: Efflux and influx of Aß across the BBB were determined by stereotaxic intra-cerebral or i.a. infusion of [(125) I]-Aß1-40 respectively. Receptor for advanced glycation end products (RAGE) and low-density lipoprotein receptor-related protein 1 (LRP1), which are involved in Aß influx and efflux, PPARγ and NF-κB p65 at the BBB, as well as hippocampal Aß, caspase-3, Bax and Bcl-2 were assayed by Western blot, immunohistochemistry and RT-PCR. In vivo, hippocampal LTP was recorded, and Morris water maze and Y-maze tasks were performed. KEY RESULTS: Treatment with PPARγ agonists, rosiglitazone (0.8 mg·kg(-1) ) and pioglitazone (9.0 mg·kg(-1) ), for 6 weeks significantly increased Aß efflux and decreased Aß influx across the BBB in db/db mice. Concomitantly, they decreased hippocampal Aß1-40 and Aß1-42 , suppressed neuronal apoptosis, as indicated by decreased caspase-3 activity and increased ratio of Bcl-2/Bax, and increased hippocampal plasticity, characterized by an enhanced in vivo LTP and better performance in behavioural tests. Furthermore, the PPARγ agonists induced the expression of LRP1 gene by activation of PPARγ and suppressed RAGE gene expression by inactivation of NF-κB signalling at the BBB of db/db mice. CONCLUSIONS AND IMPLICATIONS: PPARγ agonists modify abnormal Aß transport across the BBB and this is accompanied by amelioration of ß-amyloidosis and an improvement in hippocampal plasticity in diabetic mice.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/metabolismo , Hipocampo/metabolismo , Plasticidad Neuronal/fisiología , PPAR gamma/agonistas , PPAR gamma/metabolismo , Fragmentos de Péptidos/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Pioglitazona , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Rosiglitazona , Tiazolidinedionas/farmacología
9.
Neuropharmacology ; 101: 123-36, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26211973

RESUMEN

Previous studies have shown significant changes in amyloid-ß (Aß) transport across the blood-brain barrier (BBB) under diabetic conditions with hypoinsulinemia, which is involved in diabetes-associated cognitive impairment. Present study employed db/db mice with hyperinsulinemia to investigate changes in Aß transport across the BBB, hippocampal synaptic plasticity, and restorative effects of antidiabetic drugs. Our results showed that db/db mice exhibited similar changes in Aß transport across the BBB to that of insulin-deficient mice. Chronic treatment of db/db mice with antidiabetic drugs such as metformin, glibenclamide and insulin glargine significantly decreased Aß influx across the BBB determined by intra-arterial infusion of (125)I-Aß(1-40), and expression of the receptor for advanced glycation end products (RAGE) participating in Aß influx. Insulin glargine, but not, metformin or glibenclamide increased Aß efflux across the BBB determined by stereotaxic intra-cerebral infusion of (125)I-Aß(1-40), and expression of the low-density lipoprotein receptor related protein 1 (LRP1) participating in Aß efflux. Moreover, treatment with these drugs significantly decreased hippocampal Aß(1-40) or Aß(1-42) and inhibited neuronal apoptosis. The drugs also ameliorated memory impairment confirmed by improved performance on behavioral tasks. However, insulin glargine or glibenclamide, but not metformin, restored hippocampal synaptic plasticity characterized by enhancing in vivo long-term potentiation (LTP). Further study found that these three drugs significantly restrained NF-κB, but only insulin glargine enhanced peroxisome proliferator-activated receptor γ (PPARγ) activity at the BBB in db/db mice. Our data indicate that the antidiabetic drugs can partially restore abnormal Aß transport across the BBB and memory impairment under diabetic context.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Hipoglucemiantes/uso terapéutico , Trastornos de la Memoria/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Barrera Hematoencefálica/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Ayuno/sangre , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Insulina/sangre , Isótopos de Yodo/farmacocinética , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Ratones , Ratones Mutantes , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptores de Leptina/genética
10.
Int J Neuropsychopharmacol ; 19(1)2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26362775

RESUMEN

BACKGROUND: Emerging data have demonstrated that peroxisome proliferator-activated receptor δ (PPARδ) activation confers a potentially neuroprotective role in some neurodegenerative diseases. However, whether PPARδ is involved in depression is unknown. METHODS: In this study, PPARδ was firstly investigated in the chronic mild stress (CMS) and learned helplessness (LH) models of depression. The changes in depressive behaviors and hippocampal neurogenesis were investigated after PPARδ overexpression by microinfusion of the lentiviral vector, containing the coding sequence of mouse PPARδ (LV-PPARδ), into the bilateral dentate gyri of the hippocampus or PPARδ activation by repeated systemic administration of PPARδ agonist GW0742 (5 or 10mg/kg.d, i.p., for 21 d). RESULTS: We found that both CMS and LH resulted in a significant decrease in the PPARδ expression in the hippocampi of mice, and this change was reversed by treatment with the antidepressant fluoxetine. PPARδ overexpression and PPARδ activation each suppressed the CMS- and LH-induced depressive-like behavior and produced an antidepressive effect. In vivo or in vitro studies also showed that both overexpression and activation of PPARδ enhanced proliferation or differentiation of neural stem cells in the hippocampi of mice. CONCLUSIONS: These results suggest that hippocampal PPARδ upregulation represses stress-induced depressive behaviors, accompanied by enhancement of neurogenesis.


Asunto(s)
Depresión/complicaciones , Depresión/patología , Hipocampo/metabolismo , Neurogénesis/fisiología , PPAR delta/metabolismo , Estrés Psicológico/complicaciones , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Suspensión Trasera , Masculino , Ratones , Ratones Endogámicos ICR , Células-Madre Neurales/fisiología , PPAR delta/genética , Fosfopiruvato Hidratasa/metabolismo , Sincalida/metabolismo , Tiazoles/farmacología , Factores de Tiempo , Transducción Genética
11.
Pharmacol Biochem Behav ; 124: 373-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25038445

RESUMEN

Amyloid-ß peptide (Aß) is thought to be associated with the progressive neuronal death observed in Alzheimer's disease (AD). However, effective neuroprotective approaches against Aß neurotoxicity are unavailable. Here, we investigated possible preventive effects of ibudilast, as a pharmacologic phosphodiesterase inhibitor, currently used for treatment of inflammatory diseases such as asthma, on Aß 1-42-induced neuroinflammatory, apoptotic responses and memory impairment. We found that pretreatment with ibudilast (4 or 12 mg/kg, i.p.) significantly ameliorated impaired spatial learning and memory in intracerebroventricularly (ICV) Aß 1-42-injected mice, as evidenced by decrease in escape latency during acquisition trials and increase in exploratory activities in the probe trial in Morris water maze (MWM) task, and by increase in the number of correct choices and decrease in latency to enter the shock-free compartment in Y-maze test. Further study showed that ibudilast prevented generation of pro-inflammatory cytokines such as NF-κB p65 and TNF-α as well as pro-apoptotic molecule caspase-3 activation and anti-apoptotic protein Bcl-2 downregulation in both hippocampus and cortex of ICV Aß 1-42-injected mice. Taken together, our findings suggest that ibudilast has preventive effects on Aß-induced cognitive impairment via inhibiting neuroinflammatory and apoptotic responses.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Antiasmáticos/uso terapéutico , Trastornos de la Memoria/prevención & control , Fragmentos de Péptidos/toxicidad , Piridinas/uso terapéutico , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Antiasmáticos/farmacología , Caspasa 3/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Corteza Cerebral/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Hipocampo/metabolismo , Ratones , Ratones Endogámicos ICR , Fragmentos de Péptidos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Piridinas/farmacología
12.
Neurochem Int ; 75: 26-31, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24879954

RESUMEN

Amyloid-ß peptide (Aß), which can invoke a cascade of inflammatory responses, is considered to play a causal role in the development and progress of Alzheimer's disease (AD). Montelukast, known as a cysteinyl leukotriene receptor 1 (CysLT1R) antagonist, is currently used for treatment of inflammatory diseases such as asthma. We have previously reported that CysLT1R activation is involved in Aß generation. In this study, we investigated rescuing effect of CysLT1R antagonist montelukast on Aß1-42-induced neurotoxicity in primary neurons. Our data showed that Aß1-42 elicited a marked increase of CysLT1R expression in primary mouse neurons. This increment of CysLT1R expression was accompanied by increases of inflammatory factors such as NF-κB p65, tumor necrosis factor-α (TNFα) and interleukin-1ß (IL-1ß) as well as pro-apoptotic protein Caspase-3 activation and anti-apoptosis protein Bcl-2 reduction. Aß1-42-mediated increase of CysLT1R expression was associated with Aß1-42-induced cytotoxicity as measured by MTT reduction assay and lactate dehydrogenase (LDH) release assay. This observation was confirmed with treatment of montelukast, a selective CysLT1R antagonist, which had significant effect on Aß1-42-induced cytotoxicity. Moreover, blockade of CysLT1R with montelukast reversed Aß1-42-mediated increase of CysLT1R expression, and concomitant changes of the pro-inflammatory factors and the apoptosis-related proteins. The results demonstrate that montelukast rescued neurons against Aß1-42-induced neurotoxicity, neuroinflammation and apoptosis by down-regulating CysLT1R-mediated NF-κB signaling, suggesting that CysLT1R may be a potential target for AD, and its antagonist may have beneficial effects for treatment of AD.


Asunto(s)
Acetatos/farmacología , Péptidos beta-Amiloides/toxicidad , Antagonistas de Leucotrieno/farmacología , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Quinolinas/farmacología , Receptores de Leucotrienos/efectos de los fármacos , Receptores de Leucotrienos/fisiología , Transducción de Señal/fisiología , Animales , Caspasa 3/metabolismo , Ciclopropanos , Citocinas/biosíntesis , Activación Enzimática , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos ICR , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfuros
13.
J Pharmacol Sci ; 124(4): 418-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24671053

RESUMEN

Telmisartan, an angiotensin II type 1-receptor blocker (ARBs), has been reported to exert beneficial effects on the central nervous system (CNS). However, the effect of telmisartan on cognitive impairment associated with type 1 diabetes is not well known. Here, we examined the possibility that telmisartan could improve memory function in a type 1 diabetic mouse model, streptozotocin (STZ)-induced diabetic mice. STZ-induced diabetic mice subjected to the Morris Water Maze (MWM) task exhibited a significant decline of spatial learning and memory. Oral administration of telmisartan at two nonhypotensive doses (0.7 or 0.35 mg/kg) significantly improved memory deficits in STZ-induced diabetic mice. Telmisartan treatment markedly reduced Aß42, APP, BACE1, RAGE, and NF-κB p65 of the hippocampus and cortex, but did not beneficially affect hyperglycemia and hypoinsulinemia in the STZ-induced diabetic mice compared with untreated diabetic mice. Taken together, our findings suggest that telmisartan ameliorates memory deficits in type 1 diabetic mice, at least partly because of attenuation of amyloidosis in the brain.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Benzoatos/farmacología , Benzoatos/uso terapéutico , Angiopatía Amiloide Cerebral/tratamiento farmacológico , Angiopatía Amiloide Cerebral/etiología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/complicaciones , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Administración Oral , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Bencimidazoles/administración & dosificación , Benzoatos/administración & dosificación , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Fragmentos de Péptidos/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Estreptozocina , Telmisartán , Factor de Transcripción ReIA/metabolismo , Resultado del Tratamiento
14.
Neuropharmacology ; 79: 707-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24456746

RESUMEN

Montelukast, known as a cysteinyl leukotriene receptor 1 (CysLT1R) antagonist, is currently used for treatment of inflammatory diseases such as asthma. Here, we investigated effects of montelukast on neuroinflammatory, apoptotic responses, and memory performance following intracerebral infusions of amyloid-ß (Aß). The data demonstrated that intracerebroventrical infusions of aggregated Aß1-42 (410 pmol/mouse) produced deficits in learning ability and memory, as evidenced by increase in escape latency during acquisition trials and decreases in exploratory activities in the probe trial in Morris water maze (MWM) task, and by decrease in the number of correct choices and increase in latency to enter the shock-free compartment in Y-maze test, and caused significant increases in pro-inflammatory cytokines such as NF-κB p65, TNF-α and IL-1ß as well as pro-apoptotic molecule caspase-3 activation and anti-apoptotic protein Bcl-2 downregulation in hippocampus and cortex. Interestingly, this treatment resulted in upregulation of protein or mRNA of CysLT1R in both hippocampus and cortex. Blockade of CysLT1R by repeated treatment with montelukast (1 or 2 mg/kg, ig, 4 weeks) reduced Aß1-42-induced CysLT1R expression and also suppressed Aß1-42-induced increments of NF-κB p65, TNF-α, IL-1ß and caspase-3 activation, and Bcl-2 downregulation in the hippocampus and cortex. Correspondingly, montelukast treatment significantly improved Aß1-42-induced memory impairment in mice, but had little effect on normal mice. Our results show that montelukast may ameliorate Aß1-42-induced memory impairment via inhibiting neuroinflammation and apoptosis mediated by CysLT1R signaling, suggesting that CysLT1R antagonism represents a novel treatment strategy for Alzheimer's disease.


Asunto(s)
Acetatos/farmacología , Péptidos beta-Amiloides/metabolismo , Apoptosis/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/metabolismo , Quinolinas/farmacología , Receptores de Leucotrienos/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Apoptosis/fisiología , Caspasa 3/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiopatología , Ciclopropanos , Citocinas/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Antagonistas de Leucotrieno/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Nootrópicos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfuros
15.
Int J Neuropsychopharmacol ; 17(4): 581-92, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24229499

RESUMEN

Deposition of extracellular amyloid-ß (Aß) peptide is one of the pathological hallmarks of Alzheimer's disease (AD). Accumulation of Aß is thought to associate with cognition deficits, neuroinflammation and apoptosis observed in AD. However, effective neuroprotective approaches against Aß neurotoxicity are unavailable. In the present study, we analysed the effects of pranlukast, a selective cysteinyl leukotriene receptor 1 (CysLT1R) antagonist, on the impairment of learning and memory formation induced by Aß and the probable underlying electrophysiological and molecular mechanisms. We found that bilateral intrahippocampal injection of Aß1₋42 resulted in a significant decline of spatial learning and memory of mice in the Morris water maze (MWM) and Y-maze tests, together with a serious depression of in vivo hippocampal long-term potentiation (LTP) in the CA1 region of the mice. Importantly, this treatment caused significant increases in CysLT1R expression and subsequent NF-κB signaling, caspase-3 activation and Bcl-2 downregulation in the hippocampus or prefrontal cortex. Oral administration of pranlukast at 0.4 or 0.8 mg/kg for 4 wk significantly reversed Aß1₋42-induced impairments of cognitive function and hippocampal LTP in mice. Furthermore, pranlukast reversed Aß1₋42-induced CysLT1R upregulation, and markedly suppressed the Aß1₋42-triggered NF-κB pathway, caspase-3 activation and Bcl-2 downregulation in the hippocampus and prefrontal cortex in mice. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay confirmed its presence in the brain after oral administration of pranlukast in mice. These data disclose novel findings about the therapeutic potential of pranlukast, revealing a previously unknown therapeutic possibility to treat memory deficits associated with AD.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Cromonas/farmacología , Trastornos del Conocimiento/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Antagonistas de Leucotrieno/farmacología , Fragmentos de Péptidos/farmacología , Péptidos beta-Amiloides/administración & dosificación , Animales , Conducta Animal/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/fisiopatología , Cromonas/administración & dosificación , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Hipocampo/fisiopatología , Aprendizaje/efectos de los fármacos , Antagonistas de Leucotrieno/administración & dosificación , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Memoria/efectos de los fármacos , Ratones Endogámicos ICR , Fragmentos de Péptidos/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Receptores de Leucotrienos/efectos de los fármacos
16.
Molecules ; 18(8): 8976-93, 2013 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-23899832

RESUMEN

The potent anti-hyperuricemia activities of Fructus Gardenia Extract (FGE) have been well reported. The aim of this study was to evaluate the uricosuric and nephro-protective effects of FGE and explore its possible mechanisms of action in oxonate-induced hyperuricemic mice. FGE was orally administered to hyperuricemic and normal mice for 1 week. Serum and urinary levels of uric acid, creatinine and blood urea nitrogen (BUN), and fractional excretion of uric acid (FEUA) were measured. The mRNA and protein levels of mouse urate transporter 1 (mURAT1), glucose transporter 9 (mGLUT9), ATP-binding cassette, subfamily G, 2 (mABCG2), organic anion transporter 1 (mOAT1), mOAT3, oncoprotein induced transcript 3 (mOIT3), organic cation/carnitine transporters in the kidney were analyzed. Simultaneously, Tamm-Horsfall glycoprotein (THP) levels in urine and kidney were detected. FGE significantly reduced serum urate levels and increased urinary urate levels and FEUA in hyperuricemic mice. It could also effectively reverse oxonate-induced alterations in renal mURAT1, mGLUT9, mOAT1 and mOIT3 expressions, as well as THP levels, resulting in the enhancement of renal uric acid excretion. Moreover, FGE decreased serum creatinine and BUN levels, and up-regulated expression of organic cation/carnitine transporters, improving renal dysfunction in this model. Furthermore, FGE decreased renal mABCG2 expressions in hyperuricemic mice, contributing to its beneficial actions. However, further investigation is needed in clinical trials of FGE and its bioactive components.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hiperuricemia/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Insuficiencia Renal/tratamiento farmacológico , Animales , Nitrógeno de la Urea Sanguínea , Gardenia/química , Humanos , Hiperuricemia/inducido químicamente , Hiperuricemia/patología , Ratones , Ácido Oxónico/toxicidad , Insuficiencia Renal/inducido químicamente , Insuficiencia Renal/patología , Ácido Úrico/metabolismo
17.
Neuropharmacology ; 65: 182-92, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22982445

RESUMEN

Amyloid plaques in the extracellular parenchyma mainly consist of amyloid-ß peptides (Aß), one of the pathological hallmarks in Alzheimer's disease (AD). In the present study, we examined neuroinflammation, amyloidogenesis, and memory performance following intracerebral infusions of leukotriene D4 (LTD4) in mice. The results demonstrated that intracerebral infusions of LTD4 (1 ng/mouse) produced memory impairment as determined by Morris water maze test and Y-maze test in mice, and caused the accumulation of Aß1-40 and Aß1-42 in the hippocampus and cortex through increased activity of ß- and γ-secretases accompanied with increased expression of amyloid precursor protein (APP). LTD4 also induced expression of cysteinyl leukotriene receptor 1 (CysLT(1)R) and NF-κB p65 in the hippocampus and cortex. Pretreatment with pranlukast (1.5 ng/mouse, intracerebroventricularly), a CysLT(1)R antagonist, blocked LTD4-induced amyloidogenesis, memory deficits. Pranlukast (0.6 µM) also prevented LTD4 (20 nM)-induced amyloidogenesis in the cultured neurons in vitro. Moreover, LTD4-induced increases in CysLT(1)R and NF-κB p65 in the brain were also attenuated by pranlukast. These results suggest that LTD4 increases Aß peptide burden via activation of CysLT(1)R, which further affects APP levels and activity of ß- and γ-secretases via the NF-κB pathway. Our findings identify CysLT(1)R signaling as a novel proinflammatory and proamyloidogenic pathway, and suggest a rationale for development of therapeutics targeting the CysLT(1)R in neuroinflammatory diseases such as AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/biosíntesis , Trastornos del Conocimiento/metabolismo , Mediadores de Inflamación/fisiología , Leucotrieno D4/administración & dosificación , Receptores de Leucotrienos/fisiología , Precursor de Proteína beta-Amiloide/fisiología , Animales , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/patología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Infusiones Intraventriculares , Leucotrieno D4/toxicidad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos ICR , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...