Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 483: 116818, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38215994

RESUMEN

The recurrence and metastasis in breast cancer within 3 years after the chemotherapies or surgery leads to poor prognosis with approximately 1-year overall survival. Large-scale scanning research studies have shown that taking lipid-lowering drugs may assist to reduce the risk of death from many cancers, since cholesterol in lipid rafts are essential for maintain integral membrane structure and functional signaling regulation. In this study, we examined five lipid-lowering drugs: swertiamarin, gemfibrozil, clofibrate, bezafibrate, and fenofibrate in triple-negative breast cancer, which is the most migration-prone subtype. Using human and murine triple-negative breast cancer cell lines (Hs 578 t and 4 T1), we found that fenofibrate displays the highest potential in inhibiting the colony formation, wound healing, and transwell migration. We further discovered that fenofibrate reduces the activity of pro-metastatic enzymes, matrix metalloproteinases (MMP)-9 and MMP-2. In addition, epithelial markers including E-cadherin and Zonula occludens-1 are increased, whereas mesenchymal markers including Snail, Twist and α-smooth muscle actin are attenuated. Furthermore, we found that fenofibrate downregulates ubiquitin-dependent GDF-15 degradation, which leads to enhanced GDF-15 expression that inhibits cell migration. Besides, nuclear translocation of FOXO1 is also upregulated by fenofibrate, which may responsible for GDF-15 expression. In summary, fenofibrate with anti-cancer ability hinders TNBC from migration and invasion, and may be beneficial to repurposing use of fenofibrate.


Asunto(s)
Fenofibrato , Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , Neoplasias de la Mama Triple Negativas/metabolismo , Fenofibrato/farmacología , Fenofibrato/uso terapéutico , Factor 15 de Diferenciación de Crecimiento/farmacología , Factor 15 de Diferenciación de Crecimiento/uso terapéutico , Línea Celular Tumoral , Movimiento Celular , Hipolipemiantes/farmacología , Hipolipemiantes/uso terapéutico , Transición Epitelial-Mesenquimal , Lípidos , Proliferación Celular
2.
J Pharmacol Sci ; 155(3): 101-112, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38797534

RESUMEN

Pulmonary inflammation may lead to neuroinflammation resulting in neurological dysfunction, and it is associated with a variety of acute and chronic lung diseases. Paeonol is a herbal phenolic compound with anti-inflammatory and anti-oxidative properties. The aim of this study is to understand the beneficial effects of paeonol on cognitive impairment, pulmonary inflammation and its underlying mechanisms. Pulmonary inflammation-associated cognitive deficit was observed in TNFα-stimulated mice, and paeonol mitigated the cognitive impairment by reducing the expressions of interleukin (IL)-1ß, IL-6, and NOD-like receptor family pyrin domain-containing 3 (NLRP3) in hippocampus. Moreover, elevated plasma miR-34c-5p in lung-inflamed mice was also reduced by paeonol. Pulmonary inflammation induced by intratracheal instillation of TNFα in mice resulted in immune cells infiltration in bronchoalveolar lavage fluid, pulmonary edema, and acute fibrosis, and these inflammatory responses were alleviated by paeonol orally. In MH-S alveolar macrophages, tumor necrosis factor (TNF) α- and phorbol myristate acetate (PMA)-induced inflammasome activation was ameliorated by paeonol. In addition, the expressions of antioxidants were elevated by paeonol, and reactive oxygen species production was reduced. In this study, paeonol demonstrates protective effects against cognitive deficits and pulmonary inflammation by exerting anti-inflammatory and anti-oxidative properties, suggesting a powerful benefit as a potential therapeutic agent.


Asunto(s)
Acetofenonas , Disfunción Cognitiva , Enfermedades Pulmonares , Enfermedades Pulmonares/complicaciones , Acetofenonas/farmacología , Acetofenonas/uso terapéutico , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Macrófagos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratones Endogámicos C57BL , Masculino , Animales , Ratones , Factor de Necrosis Tumoral alfa , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , MicroARNs/sangre , MicroARNs/genética , Especies Reactivas de Oxígeno/metabolismo
3.
Neurobiol Dis ; 187: 106311, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37769745

RESUMEN

Hippocampal oxytocin receptor (OXTR) signaling is crucial for discrimination of social stimuli to guide social recognition, but circuit mechanisms and cell types involved remain incompletely understood. Here, we report a role for OXTR-expressing hilar mossy cells (MCs) of the dentate gyrus in social stimulus discrimination by regulating granule cell (GC) activity. Using a Cre-loxP recombination approach, we found that ablation of Oxtr from MCs impairs discrimination of social, but not object, stimuli in adult male mice. Ablation of MC Oxtr increases spontaneous firing rate of GCs, synaptic excitation to inhibition ratio of MC-to-GC circuit, and GC firing when temporally associated with the lateral perforant path inputs. Using mouse hippocampal slices, we found that bath application of OXTR agonist [Thr4,Gly7]-oxytocin causes membrane depolarization and increases MC firing activity. Optogenetic activation of MC-to-GC circuit ameliorates social discrimination deficit in MC OXTR deficient mice. Together, our results uncover a previously unknown role of MC OXTR signaling for discrimination of social stimuli and delineate a MC-to-GC circuit responsible for social information processing.

4.
Nat Chem Biol ; 15(7): 699-709, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31061498

RESUMEN

Chondroitin sulfate (CS) and heparan sulfate (HS) are glycosaminoglycans that both bind the receptor-type protein tyrosine phosphatase PTPRσ, affecting axonal regeneration. CS inhibits axonal growth, while HS promotes it. Here, we have prepared a library of HS octasaccharides and, together with synthetic CS oligomers, we found that PTPRσ preferentially interacts with CS-E-a rare sulfation pattern in natural CS-and most HS oligomers bearing sulfate and sulfamate groups. Consequently, short and long stretches of natural CS and HS, respectively, bind to PTPRσ. CS activates PTPRσ, which dephosphorylates cortactin-herein identified as a new PTPRσ substrate-and disrupts autophagy flux at the autophagosome-lysosome fusion step. Such disruption is required and sufficient for dystrophic endball formation and inhibition of axonal regeneration. Therefore, sulfation patterns determine the length of the glycosaminoglycan segment that bind to PTPRσ and define the fate of axonal regeneration through a mechanism involving PTPRσ, cortactin and autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Sulfatos de Condroitina/farmacología , Cortactina/metabolismo , Heparitina Sulfato/farmacología , Regeneración Nerviosa/efectos de los fármacos , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Sulfatos de Condroitina/química , Heparitina Sulfato/química , Humanos , Ratones
5.
Environ Toxicol ; 36(12): 2551-2561, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34520103

RESUMEN

Cancer and its associated conditions have significant impacts on public health at many levels worldwide, and cancer is the leading cause of death among adults. Peroxisome proliferator-activated receptor α (PPARα)-specific agonists, fibrates, have been approved by the Food and Drug Administration for managing hyperlipidemia. PPARα-specific agonists exert anti-cancer effects in many human cancer types, including glioblastoma (GBM). Recently, we have reported that the hypoxic state in GBM stabilizes hypoxia-inducible factor-1 alpha (HIF-1α), thus contributing to tumor escape from immune surveillance by activating the expression of the pH-regulating protein carbonic anhydrase IX (CA9). In this study, we aimed to study the regulatory effects of the PPARα agonist fibrate on the regulation of HIF-1α expression and its downstream target protein in GBM. Our findings showed that fenofibrate is the high potency compound among the various fibrates that inhibit hypoxia-induced HIF-1α and CA9 expression in GBM. Moreover, fenofibrate-inhibited HIF-1α expression is mediated by HO-1 activation in GBM cells through the AMP-activated protein kinase (AMPK) pathway. In addition, fenofibrate-enhanced HO-1 upregulation activates SIRT1 and leads to subsequent accumulation of SIRT1 in the nucleus, which further promotes HIF-1α deacetylation and inhibits CA9 expression. Using a protein synthesis inhibitor, cycloheximide, we also observed that fenofibrate inhibited HIF-1α protein synthesis. In addition, the administration of the proteasome inhibitor MG132 showed that fenofibrate promoted HIF-1α protein degradation in GBM. Hence, our results indicate that fenofibrate is a useful anti-GBM agent that modulates hypoxia-induced HIF-1α expression through multiple cellular pathways.


Asunto(s)
Anhidrasas Carbónicas , Fenofibrato , Glioblastoma , Proteínas Quinasas Activadas por AMP/genética , Fenofibrato/farmacología , Glioblastoma/genética , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Sirtuina 1
6.
Toxicol Appl Pharmacol ; 380: 114698, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31394157

RESUMEN

BACKGROUND: During acute lung injury, lung fibroblasts produce chemokines that assist the activation and migration of resident macrophages. The interactions between pulmonary fibroblasts and alveolar macrophages demonstrate the early event in the recruitment of immune cells, and the production of chemokines appear to be central mediators of the initiation and progression of inflammatory responses. In this study, the aim was to investigate the signaling pathway leading to CXCL10 secretion and the effects of CXCL10 released by activated fibroblasts on regulating macrophage polarization in a pro-inflammatory microenvironment. METHODS: The expression of chemokines CCL2, CCL5, CXCL10, and CXCL12, and the phosphorylation of signaling molecules STAT3, FAK, GSK3αß and PKCδ were investigated by real time-PCR, ELISA, or Western blot on TNFα- or IL-1ß-activated MRC-5 pulmonary fibroblasts. By collecting conditioned medium from TNFα-activated fibroblasts, the expression of iNOS and arginase I on MH-S alveolar macrophages were examined by real-time PCR. Surface markers CD86 and CD206 expressions on alveolar macrophages were also evaluated by flow cytometry. RESULTS: We found that CXCL10 production was significantly elevated on MRC-5 fibroblasts under TNFα- or IL-1ß treatment. In addition, we revealed that TNFα and IL-1ß initiated phosphorylation of STAT3, FAK, GSK3αß and PKCδ signaling cascade, leading to the elevation of CXCL10 expression. Moreover, conditioned medium collected from TNFα-activated MRC-5 fibroblasts increased iNOS and CD86 expressions and decreased arginase I and CD206 expressions on MH-S alveolar macrophages, and neutralization of CXCL10 abolished these observed phenomena. CONCLUSION: These results suggest that CXCL10 is crucial in activated fibroblasts-promoted M1 phenotype polarization of alveolar macrophages. In this regard, targeting fibroblasts-released CXCL10 may be promising as anti-inflammatory therapy against acute lung injury.


Asunto(s)
Citocinas/fisiología , Fibroblastos/fisiología , Pulmón/citología , Macrófagos Alveolares/fisiología , Línea Celular , Humanos , Transducción de Señal
7.
J Nat Prod ; 82(4): 1002-1008, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30892032

RESUMEN

Imperatorin is one of the furanocoumarin derivatives and exists in many medicinal herbs with anticancer, antiviral, antibacterial, and antihypertensive activities. In this study, we examined the anti-inflammatory effects of imperatorin on inflammation-associated lung diseases. Imperatorin reduced iNOS and COX-2 expression and also IL-6 and TNFα production enhanced by zymosan. Imperatorin also inhibited the signaling pathways of JAK/STAT and NF-κB. Moreover, in vivo study also revealed that zymosan-induced immune cell infiltration, pulmonary fibrosis, and edema were relieved by imperatorin in mice. We found that imperatorin exerts anti-inflammatory effects that are associated with amelioration of lung inflammation, edema, and rapid fibrosis. Studies on alveolar macrophages also reveal that imperatorin reduced the production of pro-inflammatory mediators and cytokines and inhibited pro-inflammatory JAK1/STAT3 and NF-κB signaling pathways. These results indicate that imperatorin may be a potential anti-inflammatory agent for inflammatory-associated lung diseases.


Asunto(s)
Antiinflamatorios/farmacología , Furocumarinas/farmacología , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Animales , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Ratones
8.
Toxicol Appl Pharmacol ; 338: 182-190, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29180066

RESUMEN

Connexins are widely supported as tumor suppressors due to their downregulation in cancers, nevertheless, more recent evidence suggests roles for connexins in facilitating tumor progression in later stages, including metastasis. One of the key factors regulating the expression, modification, stability, and localization of connexins is hormone receptors in hormone-dependent cancers. It is reasonable to consider that hormones/hormone receptors may modulate connexins expression and play critical roles in the cellular control of connexins during breast cancer progression. In estrogen receptor (ER)-positive breast cancers, tamoxifen and fulvestrant are widely used therapeutic agents and are considered to alter ER signaling. In this present study, we investigated the effects of fulvestrant and tamoxifen in Cx43 expression, and we also explored the role of Cx43 in ER-positive breast cancer migration and the relationship between Cx43 and ER. The involvement of estrogen/ER in Cx43 modulation was further verified by administering tyrosine kinase inhibitors and chemotherapeutic agents. We found that inhibition of ER promoted the binding of E3 ligase Nedd4 to Cx43, leading to Cx43 ubiquitination. Furthermore, inhibition of ER by fulvestrant and tamoxifen phosphorylated p38 MAPK, and inhibition of Rac, MKK3/6, and p38 reversed fulvestrant-reduced Cx43 expression. These findings suggest that Cx43 expression which may positively regulate cell migration is ER-dependent in ER-positive breast cancer cells.


Asunto(s)
Neoplasias de la Mama/patología , Conexina 43/fisiología , Antagonistas de Estrógenos/farmacología , Neoplasias de la Mama/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Conexina 43/análisis , Femenino , Humanos , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Receptores de Estrógenos/fisiología , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
9.
Int J Mol Sci ; 20(1)2018 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-30585203

RESUMEN

OBJECTIVE: Intervertebral disc (IVD) degeneration and disc herniation are major causes of lower back pain, which involve the presence of inflammatory mediators and tissue invasion by immune cells. Intercellular adhesion molecule 1 (ICAM1, also termed CD54) is an adhesion molecule that mediates cell-cell interactions, particularly between immune cells and target tissue. The aim of this study was to examine the intracellular signaling pathways involved in inflammatory stimuli-induced ICAM1 expression in human anulus fibrosus (AF) cells. METHODS: Quantitative reverse transcription-polymerase chain reaction (qPCR), western blotting, and flow cytometry were performed to dissect the roles of different signaling pathways in inflammatory stimuli-mediated ICAM1 expression. RESULTS: Using qPCR and western blot analyses, a significant increase in ICAM1 expression was observed in AF cells after stimulation of lipopolysaccharide (LPS) plus interferon-gamma (IFNγ) in a time-dependent manner. Flow cytometry revealed ICAM1 upregulation on the surface of AF cells. Importantly, LPS plus IFNγ treatment also significantly promoted Chemokine ligand (CCL)2 expression, but not CCL3. The enhanced ICAM1 expression was abolished after incubation with antibody against CCL2. In AF cells, treatment with LPS plus IFNγ activated the FAK/ERK/GSK3 signaling pathways, promoted a time-dependent increase in PKCδ phosphorylation, and promoted PKCδ translocation to the nucleus. Treatment with the pharmacological PKCδ inhibitor; rottlerin, effectively blocked the enhanced productions of ICAM1 and CCL2. CONCLUSIONS: Inflammatory stimuli in AF cells are part of a specific pathophysiology in IVD degeneration and disc herniation that modulates CCL2/ICAM1 activation through the FAK/ERK/GSK3 and PKCδ signaling pathways in AF cells.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Proteína Quinasa C-delta/metabolismo , Acetofenonas/farmacología , Anillo Fibroso/citología , Anillo Fibroso/metabolismo , Benzopiranos/farmacología , Quimiocina CCL2/metabolismo , Humanos , Interferón gamma/farmacología , Janus Quinasa 2/metabolismo , Lipopolisacáridos/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C-delta/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
10.
Int J Mol Sci ; 19(1)2018 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-29301329

RESUMEN

Glioblastoma multiforme (GBM) is the most common type of primary and malignant tumor occurring in the adult central nervous system. Temozolomide (TMZ) has been considered to be one of the most effective chemotherapeutic agents to prolong the survival of patients with glioblastoma. Many glioma cells develop drug-resistance against TMZ that is mediated by increasing O-6-methylguanine-DNA methyltransferase (MGMT) levels. The expression of connexin 43 was increased in the resistant U251 subline compared with the parental U251 cells. The expression of epithelial-mesenchymal transition (EMT)-associated regulators, including vimentin, N-cadherin, and ß-catenin, was reduced in the resistant U251 subline. In addition, the resistant U251 subline exhibited decreased cell migratory activity and monocyte adhesion ability compared to the parental U251 cells. Furthermore, the resistant U251 subline also expressed lower levels of vascular cell adhesion molecule (VCAM)-1 after treatment with recombinant tumor necrosis factor (TNF)-α. These findings suggest differential characteristics in the drug-resistant GBM from the parental glioma cells.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos , Glioma/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Conexina 43/metabolismo , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/patología , Humanos , Monocitos/efectos de los fármacos , Monocitos/patología , Temozolomida , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
Biol Reprod ; 94(3): 60, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26819477

RESUMEN

Endometriosis is the hormone-dependent product of endometrial tissue found outside the uterus. Recently, micro-RNAs (miRNAs) were shown to play a role in endometriotic lesion development. However, the mechanism of steroid hormones responsible for miRNA remains obscure. In the present study, we assayed for the effects of synthetic steroid hormones (danazol, progesterone, and medroxyprogesterone acetate [MPA]) on miRNAs in endometriosis. We used a global miRNA expression profile microarray to evaluate miRNA expression in endometrial mesenchymal stem cells (EN-MSCs) of ovarian endometrioma following treatment with 1 µM danazol, progesterone, or MPA. Furthermore, we selected candidate miRNAs whose expression changed more than fivefold and compared the effects of danazol, progesterone, and MPA treatments and also compared those results with controls in EN-MSCs. Among those with a fivefold change, we found 13 ectopically upregulated miRNAs in EN-MSCs. To understand the function of these 13 miRNAs, we subjected their sequences to Ingenuity Pathway Analysis. According to both the etiology and pathogenesis of endometriosis, we found that miR-199a-5p and miR-34a-5p showed specific association with the disease, including molecular and cellular functions. Steroid hormone treatment elevated the levels of miR-199a-5p and miR-34a-5p. An inhibitor of miR-34a-5p also reduced the synthetic steroid hormones effects on cell proliferation. In vivo data revealed that miRNA levels in endometriotic lesions correlated with findings following in vitro synthetic hormone treatment. Our data show the effects of synthetic steroid hormones on miRNA regulation. These findings contribute to our understanding of the molecular impact of the synthetic steroid hormones and suggest a potential mechanism for endometriosis treatment.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Endometriosis/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/metabolismo , Enfermedades del Ovario/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Danazol/farmacología , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Acetato de Medroxiprogesterona/farmacología , Ratones , MicroARNs/genética , Progesterona/farmacología , Receptor Notch1 , Transcriptoma , Regulación hacia Arriba/efectos de los fármacos
12.
Mol Ther ; 23(4): 656-66, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25531695

RESUMEN

Histone deacetylase inhibitors (HDACi) are novel clinical anticancer drugs that inhibit HDAC gene expression and induce cell apoptosis in human cancers. Nevertheless, the detailed mechanism or the downstream HDAC targets by which HDACi mediates apoptosis in human breast cancer cells remains unclear. Here, we show that HDACi reduce tumorigenesis and induce intrinsic apoptosis of human breast cancer cells through the microRNA miR-125a-5p in vivo and in vitro. Intrinsic apoptosis was activated by the caspase 9/3 signaling pathway. In addition, HDACi mediated the expression of miR-125a-5p by activating RUNX3/p300/HDAC5 complex. Subsequently, miR-125a-5p silenced HDAC5 post-transcriptionally in the cells treated with HDACi. Thus, a regulatory loop may exist in human breast cancer cells involving miR-125a-5p and HDAC5 that is controlled by RUNX3 signaling. Silencing of miR-125a-5p and RUNX3 inhibited cancer progression and activated apoptosis, but silencing of HDAC5 had a converse effect. In conclusion, we demonstrate a possible new mechanism by which HDACi influence tumorigenesis and apoptosis via downregulation of miR-125a-5p expression. This study provides clinical implications in cancer chemotherapy using HDACi.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/efectos de los fármacos , MicroARNs/fisiología , Regulación hacia Arriba/efectos de los fármacos , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Línea Celular Tumoral , Xenoinjertos , Humanos , Ratones
13.
J Pathol ; 232(3): 330-43, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24155090

RESUMEN

It is believed that endometrial miRNAs contribute to the aetiology of endometriosis in stem cells; however, the mechanisms remain unclear. Here we collected serum samples from patients with or without endometriosis and characterized the miRNA expression profiles of these two groups. MicroRNA-199a-5p (miR-199a-5p) was dramatically down-regulated in patients with endometriosis compared with control patients. In addition, we found that the tumour suppressor gene, SMAD4, could elevate miR-199a-5p expression in ectopic endometrial mesenchymal stem cells. Up-regulation of miR-199a-5p suppressed cell proliferation, motility and angiogenesis of these ectopic stem cells by targeting the 3' untranslated region of VEGFA. Furthermore, we established an animal model of endometriosis and found that miR-199a-5p could decrease the size of endometriotic lesions in vivo. Taken together, this newly identified miR-199a-5p module provides a new avenue to the understanding of the processes of endometriosis development, especially proliferation, motility and angiogenesis, and may facilitate the development of potential therapeutics against endometriosis.


Asunto(s)
Endometriosis/genética , Endometrio/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Animales , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Endometriosis/metabolismo , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , MicroARNs/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
14.
Int J Mol Sci ; 16(3): 5572-89, 2015 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-25768341

RESUMEN

Microglial activation has been widely demonstrated to mediate inflammatory processes that are crucial in several neurodegenerative disorders. Pharmaceuticals that can deliver direct inhibitory effects on microglia are therefore considered as a potential strategy to counter balance neurodegenerative progression. Caffeic acid phenethyl ester (CAPE), a natural phenol in honeybee propolis, is known to possess antioxidant, anti-inflammatory and anti-microbial properties. Accordingly, the current study intended to probe the effects of CAPE on microglia activation by using in vitro and in vivo models. Western blot and Griess reaction assay revealed CAPE significantly inhibited the expressions of inducible nitric oxide synthase (NOS), cyclooxygenase (COX)-2 and the production of nitric oxide (NO). Administration of CAPE resulted in increased expressions of hemeoxygenase (HO)-1and erythropoietin (EPO) in microglia. The phosphorylated adenosine monophosphate-activated protein kinase (AMPK)-α was further found to regulate the anti-inflammatory effects of caffeic acid. In vivo results from immunohistochemistry along with rotarod test also revealed the anti-neuroinflammatory effects of CAPE in microglia activation. The current study has evidenced several possible molecular determinants, AMPKα, EPO, and HO-1, in mediating anti-neuroinflammatory responses in microglial cells.


Asunto(s)
Ácidos Cafeicos/farmacología , Microglía/efectos de los fármacos , Alcohol Feniletílico/análogos & derivados , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Línea Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Eritropoyetina/genética , Eritropoyetina/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Microglía/metabolismo , Microglía/patología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Alcohol Feniletílico/farmacología
15.
J Am Chem Soc ; 136(41): 14425-31, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25291402

RESUMEN

Living organisms employ glycans as recognition elements because of their large structural information density. Well-defined sugar structures are needed to fully understand and take advantage of glycan functions, but sufficient quantities of these compounds cannot be readily obtained from natural sources and have to be synthesized. Among the bottlenecks in the chemical synthesis of complex glycans is the preparation of suitably protected monosaccharide building blocks. Thus, easy, rapid, and efficient methods for building-block acquisition are desirable. Herein, we describe routes directly starting from the free sugars toward notable monosaccharide derivatives through microwave-assisted one-pot synthesis. The procedure followed the in situ generation of per-O-trimethylsilylated monosaccharide intermediates, which provided 1,6-anhydrosugars or thioglycosides upon treatment with either trimethylsilyl trifluoromethanesulfonate or trimethyl(4-methylphenylthio)silane and ZnI2, respectively, under microwave irradiation. We successfully extended the methodology to regioselective protecting group installation and manipulation toward a number of thioglucosides and the glycosylation of persilylated derivatives, all of which were conducted in a single vessel. These developed approaches open the possibility for generating arrays of suitably protected building blocks for oligosaccharide assembly in a short period with minimal number of purification stages.


Asunto(s)
Microondas , Oligosacáridos/síntesis química , Tioglicósidos/síntesis química , Conformación de Carbohidratos , Oligosacáridos/química , Tioglicósidos/química
16.
BMC Cancer ; 14: 556, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25081364

RESUMEN

BACKGROUND: The widespread use of phthalates as plasticizers has raised public health concerns regarding their adverse effects, including an association with cancer. Although animal investigations have suggested an association between phthalate exposure and hepatocellular carcinoma, the mechanisms are unknown. METHODS: The hepatocellular carcinoma cell line Huh7 was treated with benzyl butyl phthalate (BBP), and then analyzed by total internal reflection fluorescence microscopy, confocal microscopy and double immunogold transmission electron microscopy. Following BBP treatment, mRNA levels were measured by RT-PCR, protein levels were measured using western blot, and vascular endothelial growth factor levels were measured by an enzyme-linked immunosorbent assay. Cell migration and invasion assays were evaluated by transwell, and angiogenesis were performed by a tube formation assay. Nude mice were used to investigate metastasis and angiogenesis in vivo. RESULTS: BBP affected hepatocellular carcinoma progression through the aryl hydrocarbon receptor (AhR) and that benzyl butyl phthalate (BBP) stimulated AhR at the cell surface, which then interacted with G proteins and triggered a downstream signaling cascade. BBP activated AhR through a nongenomic action involving G-protein signaling rather than the classical genomic AhR action. BBP treatment promoted cell migration and invasion in vitro and metastasis in vivo via the AhR/Gß/PI3K/Akt/NF-κB pathway. In addition, BBP induced both in vitro and in vivo angiogenesis through the AhR/ERK/VEGF pathway. CONCLUSIONS: These findings suggest a novel nongenomic AhR mechanism involving G-protein signaling induced by phthalates, which contributes to tumor progression of hepatocellular carcinoma.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Ácidos Ftálicos/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Animales , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Genoma , Células HEK293 , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica/patología , Trasplante de Neoplasias , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
J Neurooncol ; 118(2): 257-269, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24756349

RESUMEN

Glioblastoma multiforme (GBM) is the most common and lethal type of primary brain tumor characterized by its rapid infiltration to surrounding tissues during the early stages. The fast spreading of GBM obscures the initiation of the tumor mass making the treatment outcome undesirable. Endothelin-1 is known as a secretory protein presented in various types of brain cells, which has been indicated as a factor for cancer pathology. The aim of the present study was to investigate the molecular mechanism of cell migration in GBM. We found that various malignant glioma cells expressed higher amounts of endothelin-1, ETA, and ETB receptors than nonmalignant human astrocytes. The application of endothelin-1 enhanced the migratory activity in human U251 glioma cells corresponding to increased expression of matrix metalloproteinase (MMP)-9 and MMP-13. The endothelin-1-induced cell migration was attenuated by MMP-9 and MMP-13 inhibitors and inhibitors of mitogen-activated protein (MAP) kinase and PI3 kinase/Akt. Furthermore, the elevated levels of phosphate c-Jun accumulation in the nucleus and activator protein-1 (AP-1)-DNA binding activity were also found in endothelin-1 treated glioma cells. In migration-prone sublines, cells with greater migration ability showed higher endothelin-1, ETB receptor, and MMP expressions. These results indicate that endothelin-1 activates MAP kinase and AP-1 signaling, resulting in enhanced MMP-9 and MMP-13 expressions and cell migration in GBM.


Asunto(s)
Movimiento Celular/fisiología , Neoplasias del Sistema Nervioso Central/fisiopatología , Endotelina-1/metabolismo , Glioblastoma/fisiopatología , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Astrocitos/fisiología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Factor de Transcripción AP-1/metabolismo
18.
Int J Mol Sci ; 15(3): 3889-903, 2014 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-24599080

RESUMEN

Glioblastoma multiforme (GBM) is the most common type of primary and malignant tumor occurring in the adult central nervous system. GBM often invades surrounding regions of the brain during its early stages, making successful treatment difficult. Osthole, an active constituent isolated from the dried C. monnieri fruit, has been shown to suppress tumor migration and invasion. However, the effects of osthole in human GBM are largely unknown. Focal adhesion kinase (FAK) is important for the metastasis of cancer cells. Results from this study show that osthole can not only induce cell death but also inhibit phosphorylation of FAK in human GBM cells. Results from this study show that incubating GBM cells with osthole reduces matrix metalloproteinase (MMP)-13 expression and cell motility, as assessed by cell transwell and wound healing assays. This study also provides evidence supporting the potential of osthole in reducing FAK activation, MMP-13 expression, and cell motility in human GBM cells.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Cumarinas/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Western Blotting , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cnidium/química , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Frutas/química , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Fosforilación/efectos de los fármacos , Tirosina/metabolismo
19.
Molecules ; 19(7): 8820-39, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24972270

RESUMEN

Increasing evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play a key role in neurodegeneration. Fisetin, a plant flavonol commonly found in fruits and vegetables, is frequently added to nutritional supplements due to its antioxidant properties. In the present study, treatment with fisetin inhibited microglial cell migration and ROS (reactive oxygen species) production. Treatment with fisetin also effectively inhibited LPS plus IFN-γ-induced nitric oxide (NO) production, and inducible nitric oxide synthase (iNOS) expression in microglial cells. Furthermore, fisetin also reduced expressions of iNOS and NO by stimulation of peptidoglycan, the major component of the Gram-positive bacterium cell wall. Fisetin also inhibited the enhancement of LPS/IFN-γ- or peptidoglycan-induced inflammatory mediator IL (interlukin)-1 ß expression. Besides the antioxidative and anti-inflammatory effects of fisetin, our study also elucidates the manner in fisetin-induced an endogenous anti-oxidative enzyme HO (heme oxygenase)-1 expression. Moreover, the regulatory molecular mechanism of fisetin-induced HO-1 expression operates through the PI-3 kinase/AKT and p38 signaling pathways in microglia. Notably, fisetin also significantly attenuated inflammation-related microglial activation and coordination deficit in mice in vivo. These findings suggest that fisetin may be a candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.


Asunto(s)
Antiinflamatorios/farmacología , Flavonoides/farmacología , Microglía/inmunología , Fármacos Neuroprotectores/farmacología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Flavonoles , Hemo-Oxigenasa 1/metabolismo , Lipopolisacáridos/farmacología , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos ICR , Microglía/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
20.
Pharmacol Biochem Behav ; 241: 173794, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38834160

RESUMEN

Psychological stress affects the neuroendocrine regulation, which modulates mental status and behaviors. Melatonin, a hormone synthesized primarily by the pineal gland, regulates many brain functions, including circadian rhythms, pain, sleep, and mood. Selective pharmacological melatonin agonist ramelteon has been clinically used to treat mood and sleep disorders. Posttraumatic stress disorder (PTSD) is a psychiatric condition associated with severe trauma; it is generally triggered by traumatic events, which lead to severe anxiety and uncontrollable trauma recall. We recently reported that repeated social defeat stress (RSDS) may induce robust anxiety-like behaviors and social avoidance in mice. In the present study, we investigated whether melatonin receptor activation by melatonin and ramelteon regulates RSDS-induced behavioral changes. Melatonin treatment improved social avoidance and anxiety-like behaviors in RSDS mice. Moreover, treatment of the non-selective MT1/MT2 receptor agonist, ramelteon, markedly ameliorated RSDS-induced social avoidance and anxiety-like behaviors. Moreover, activating melatonin receptors also balanced the expression of monoamine oxidases, glucocorticoid receptors, and endogenous antioxidants in the hippocampus. Taken together, our findings indicate that the activation of both melatonin and ramelteon regulates RSDS-induced anxiety-like behaviors and PTSD symptoms. The current study also showed that the regulatory effects of neuroendocrine mechanisms and cognitive behaviors on melatonin receptor activation in repeated social defeat stress.


Asunto(s)
Ansiedad , Indenos , Melatonina , Derrota Social , Estrés Psicológico , Animales , Indenos/farmacología , Ratones , Masculino , Estrés Psicológico/metabolismo , Estrés Psicológico/tratamiento farmacológico , Melatonina/farmacología , Ansiedad/tratamiento farmacológico , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/agonistas , Receptor de Melatonina MT1/agonistas , Receptor de Melatonina MT1/metabolismo , Receptor de Melatonina MT2/agonistas , Receptor de Melatonina MT2/metabolismo , Ratones Endogámicos C57BL , Monoaminooxidasa/metabolismo , Receptores de Melatonina/agonistas , Receptores de Melatonina/metabolismo , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/psicología , Trastornos por Estrés Postraumático/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA