Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 509(4): 877-885, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30642634

RESUMEN

Obesity is known to be induced by the accumulation of hypertrophy and hyperplasia of newly created fat in adipose tissues through differentiation of adipocyte precursor cells. Some cytokines are excessively produced in adipose tissues that can negatively regulate differentiation of adipocytes. Impaired adipogenesis is known to contribute to obesity-related diseases. Interleukin-10 (IL-10) is involved in the development of type 2 diabetes, insulin sensitivity, and immune response in obesity state. However, effects of IL-10 on adipogenesis remain unclear. The objective of this study was to determine the inhibitory effect of IL-10 on adipocyte differentiation and mechanisms involved in such effect. The effect of IL-10 on adipogenesis was analyzed by Western blot analysis, Oil Red O staining, qRT-PCR, and flow cytometry. We also examined the part of Wnt5a in adipogenesis using gene interfering technique. IL-10 suppressed lipid accumulation and adipocyte differentiation related gene expression. The inhibitory effect of IL-10 on the differentiation of adipocytes occurred at an early phase. IL-10 treatment caused a G0/G1 phase cell cycle arrest and altered expression levels of cell cycle proteins (CDK2, p21, and p27), thereby preventing re-entry into cell cycle. Additionally, IL-10 treatment reduced Wnt5a expression. Inhibition of Wnt5a by siRNA significantly attenuated lipid accumulation and expression of adipocyte differentiation-related genes. Taken together, these results indicate that IL-10 can inhibit the early phase of adipogenesis via suppressing Wnt5a signaling pathway in 3T3-L1 preadipocytes.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Interleucina-10/farmacología , Transducción de Señal/fisiología , Proteína Wnt-5a/metabolismo , Células 3T3-L1 , Adipocitos/citología , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ratones
2.
Planta Med ; 85(4): 302-311, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30452073

RESUMEN

Lobaric acid (LA) is a constituent of the lichen Stereocaulon alpinum. LA has multiple biological activities, including antibacterial and antioxidant ones. The purpose of this study was to investigate the effect of LA and its mechanism on lipopolysaccharide (LPS)-induced inflammatory responses in macrophages. Macrophages were pretreated with different concentrations of LA (0.2 - 20 µM), followed by LPS stimulation. LA treatment of LPS stimulated macrophages decreased their nitric oxide production and the expression of cyclooxygenase-2 and prostaglandin E2. LA also significantly reduced the production of tumor necrosis factor-α and interleukin (IL)-6 by inhibiting the activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-κB). Additionally, LA inhibited the production of IL-1ß and IL-18, as well as caspase-1 maturation, by inhibition of NLRP3 inflammasome activation in LPS/ATP-stimulated cells. These results strongly suggest that LA could inhibit inflammation by downregulating NF-κB/MAPK pathways and NLRP3 inflammasome activation in activated macrophages. These results reveal a new therapeutic approach to modulate inflammatory diseases linked to deregulated inflammasome activities.


Asunto(s)
Antiinflamatorios/farmacología , Inflamasomas/efectos de los fármacos , Lactonas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Salicilatos/farmacología , Animales , Western Blotting , Ciclooxigenasa 2/metabolismo , Depsidos/farmacología , Dinoprostona/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Transducción de Señal/efectos de los fármacos
3.
J Infect Dis ; 217(12): 1977-1986, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29579238

RESUMEN

Background: Previous studies have focused on colonization resistance of the gut microbiota against antibiotic resistant strains. However, less research has been performed on respiratory colonization resistance. Methods: Because respiratory colonization is the first step of respiratory infections, intervention to prevent colonization would represent a new approach for preventive and therapeutic measures. The Th17 response plays an important role in clearance of respiratory pathogens. Thus, harnessing the Th17 immune response in the mucosal site would be an effective method to design a respiratory mucosal vaccine. Results: In this study, we show that intranasal Δpep27 immunization induces noncanonical Wnt and subsequent interleukin (IL)-17 secretion, and it inhibits Streptococcus pneumoniae, Staphylococcus aureus, and Klebsiella pneumoniae colonization. Moreover, IL-17A neutralization or nuclear factor of activated T-cell inhibition augmented bacterial colonization, indicating that noncanonical Wnt signaling is involved in pulmonary colonization resistance. Conclusions: Therefore, Δpep27 immunization can provide nonspecific respiratory colonization resistance via noncanonical Wnt signaling and IL-17A-related pathways.


Asunto(s)
Interleucina-17/inmunología , Pulmón/inmunología , Pulmón/microbiología , Vía de Señalización Wnt/inmunología , Administración Intranasal/métodos , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Inmunización/métodos , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones Neumocócicas/inmunología , Vacunas Neumococicas/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Streptococcus pneumoniae/inmunología , Células Th17/inmunología , Vacunación/métodos
4.
Biochem Biophys Res Commun ; 503(2): 882-887, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-29928885

RESUMEN

Bromodomain-containing protein 4 (Brd4) is known to play a key role in tumorigenesis. It binds acetylated histones to regulate the expression of numerous genes. Because of the importance of brd4 in tumorigenesis, much research has been undertaken to develop brd4 inhibitors with therapeutic potential. As a result, various scaffolds for bromodomain inhibitors have been identified. To discover new scaffolds, we performed mid-throughput screening using two different enzyme assays, alpha-screen and ELISA. We found a novel bromodomain inhibitor with a unique scaffold, aristoyagonine. This natural compound showed inhibitory activity in vitro and tumor growth inhibition in a Ty82-xenograft mouse model. In addition, we tested Brd4 inhibitors in gastric cancer cell lines, and found that aristoyagonine exerted cytotoxicity not only in I-BET-762-sensitive cancer cells, but also in I-BET-762-resistant cancer cells. This is the first paper to describe a natural compound as a Brd4 bromodomain inhibitor.


Asunto(s)
Productos Biológicos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Isoquinolinas/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/patología , Neoplasias/prevención & control , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Biochim Biophys Acta ; 1860(11 Pt A): 2416-2427, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27424923

RESUMEN

BACKGROUND INFORMATION: Adipose tissue regulates energy metabolism by means of adipocyte hypertrophy and/or the differentiation of pre-existing adipocytes. Excessive production of some cytokines in adipose tissue is known to be a negative regulator of adipocyte differentiation, and the resulting impaired adipogenesis contributes to disorders like insulin resistance. IFN-α is a key immunoregulatory cytokine in the development of type 1 diabetes, lipid disorders and insulin resistance; however, its effect on adipogenesis remains unknown. METHOD: We examined the effect of IFN-α on adipocyte differentiation and its mechanisms. The effect of IFN-α on adipogenesis was evaluated by Western blotting, qRT-PCR, flow cytometric analysis and Oil Red O staining. We also investigated the role of STAT1 in adipogenesis using gene silencing analysis. RESULTS: IFN-α inhibited the accumulation of lipid droplets and the expression of adipogenesis related genes. The inhibition of adipocyte differentiation by IFN-α occurred in the early stages of differentiation. IFN-α arrested the cell cycle at the G0/G1 phase and regulated the expression of CDK2 and p21. These results were confirmed in MEF cells. Treatment with IFN-α increased STAT1 phosphorylation, and STAT1 siRNA or inhibitor prevented IFN-α from inhibiting the expression of PPARγ and C/EBPα as well as cell cycle progression in 3T3-L1 cells. CONCLUSION: We suggest that IFN-α inhibits adipocyte differentiation during the early stage of adipogenesis by regulating the expression of PPARγ and C/EBPα as well as the cell cycle through JAK/STAT1 signaling pathways. GENERAL SIGNIFICANCE: Our study provides new insights into possible mechanisms of the anti-adipogenetic effects of IFN-α.


Asunto(s)
Adipogénesis , Interferón-alfa/farmacología , Quinasas Janus/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Células 3T3 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Ciclo Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , PPAR gamma/metabolismo
6.
Biochem Biophys Res Commun ; 492(1): 121-127, 2017 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-28782519

RESUMEN

Bone morphogenetic protein 9 (BMP9), a member of the TGF-ß superfamily, is considered a regulator of glucose homeostasis as well as a neuronal differentiation factor. BMP9 induces phosphorylation of Smad1/5 through activin receptor-like kinase 1 and 2 (ALK1 and ALK2). Recently, many studies have shown that BMP9 contributes to tumorigenesis, and aberrant ALK2 expression is involved in many diseases. To investigate the role of BMP9-ALK2 signaling in cancer cells, we used TF-1 cells that require granulocyte-macrophage colony-stimulating factor (GM-CSF) for cell proliferation. BMP9 promoted the proliferation of TF-1 cells in media lacking GM-CSF. TF-1 cells overexpressing ALK2 resulted in the autophosphorylation of Smad1/5, leading to consequent increase in cell growth. Through high-throughput screening (HTS), we found two ALK2-specific inhibitors, KRC203 and KRC360, with IC50 values of 0.9 nM and 0.3 nM. These compounds were more potent and specific for the inhibition of ALK2 when compared to LDN193189. In cell-based assays, these compounds effectively inhibited the proliferation and migration of cancer cells induced by ALK2 and BMP9. Therefore, we propose that our compounds are promising candidates for the treatment of cancer or diseases with abnormal ALK2 or BMP9 signaling.


Asunto(s)
Receptores de Activinas Tipo I/antagonistas & inhibidores , Neoplasias/enzimología , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Activinas Tipo I/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , Ratones , Estructura Molecular , Células 3T3 NIH , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
7.
Biochim Biophys Acta Gen Subj ; 1861(11 Pt A): 2663-2673, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28713020

RESUMEN

BACKGROUND INFORMATION: The 53kDa protein pneumolysin (PLY) is the main virulence factor of Streptococcus pneumoniae, a leading cause of invasive pneumococcal diseases. PLY forms pores in cholesterol-containing membranes, thereby interfering with the function of cells. Bone destruction is a serious matter in chronic inflammatory diseases such as septic arthritis and osteomyelitis. S. pneumoniae is increasingly being recognized as a common cause of septic arthritis, but its pathogenesis is poorly defined. METHOD: We examined the effect of PLY on osteoblast differentiation and its mechanisms of action. The effect of PLY on osteoblast differentiation was evaluated by qRT-PCR, ALP activity assay, flow cytometric analysis, and Western blotting. We also examined the role of PLY-induced autophagy in osteoblast differentiation using RNA interference analysis. RESULTS: PLY inhibited osteoblast differentiation by decreasing the expression of osteoblast marker genes such as Runx2 and OCN, along with ALP activity. ROS production was increased by PLY during osteoblast differentiation. PLY induced autophagy through ROS-mediated regulation of AMPK and mTOR, which downregulated the expression of Sp1 and subsequent inhibition of differentiation. Treatment with autophagy inhibitors or Atg5 siRNA alleviated the PLY-induced inhibition of differentiation. CONCLUSION: The results suggest that PLY inhibits osteoblast differentiation by downregulation of Sp1 accompanied by induction of autophagy through ROS-mediated regulation of the AMPK/mTOR pathway. GENERAL SIGNIFICANCE: This study proposes a molecular mechanism for inhibition of osteoblast differentiation in response to PLY.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Osteosarcoma/genética , Factor de Transcripción Sp1/genética , Estreptolisinas/metabolismo , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteogénesis/efectos de los fármacos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Streptococcus pneumoniae/química , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/administración & dosificación , Serina-Treonina Quinasas TOR/metabolismo
8.
J Org Chem ; 82(21): 11566-11572, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-29019238

RESUMEN

The first total synthesis and biological evaluation of penchinone A and its structural analogues are described. The key steps for the preparation of penchinone A derivatives involve the oxime-directed palladium(II)-catalyzed oxidative acylation, Claisen rearrangement, and base-mediated olefin migration. This transformation efficiently provides a range of allyl-substituted biaryl ketones with site-selectivity and functional group compatibility. In addition, all synthetic compounds were screened for anti-inflammatory activity against nitric oxide (NO), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6) with lipopolysaccharide (LPS)-induced RAW264.7 cells. Generally, a range of penchinone A derivatives potently inhibited NO, TNF-α, and IL-6 productions, compared to dexamethasone as a positive control. Notably, penchinone A (8g) and its derivatives (8e and 8f) were found to exhibit anti-inflammatory activity stronger than that of dexamethasone.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Interleucina-6/antagonistas & inhibidores , Lignanos/farmacología , Óxido Nítrico/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interleucina-6/biosíntesis , Lignanos/síntesis química , Lignanos/química , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Ratones , Estructura Molecular , Óxido Nítrico/biosíntesis , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/biosíntesis
9.
J Nat Prod ; 80(1): 149-155, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-28093915

RESUMEN

A new anthranilic acid derivative (1) was isolated from a Philippine sponge, Oscarella stillans (Bergquist and Kelly). The structure of compound 1, named oscarellin, was determined as 2-amino-3-(3'-aminopropoxy)benzoic acid from spectroscopic data and confirmed by synthesis. We examined the immunomodulating effect of compound 1 and its mechanism in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Our data indicated that the expression of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 were significantly reduced by the pretreatment of 1 (0.1-10 µM) for 2 h. In addition, compound 1 suppressed activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun NH2-termimal kinase (JNK), but not p38 mitogen-activated protein kinase (MAPK) in LPS-stimulated RAW 264.7 cells. Compound 1 abrogated LPS-induced nuclear factor-κB (NF-κB) and activator protein-1 (AP-1) activities, whereas the induction of activating transcription factor-3 (ATF-3) was increased. Taken together, our results suggest that compound 1 attenuates pro-inflammatory cytokines via the suppression of JNK, ERK, AP-1, and NF-κB and the activation of the ATF-3 signaling pathway.


Asunto(s)
Aminas/farmacología , Benzoatos/farmacología , Citocinas/metabolismo , Inflamación/metabolismo , Interleucina-6/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Aminas/química , Aminas/aislamiento & purificación , Animales , Benzoatos/química , Benzoatos/aislamiento & purificación , Citocinas/química , Interleucina-6/química , Proteínas Quinasas JNK Activadas por Mitógenos/química , Lipopolisacáridos/química , Ratones , Proteína Quinasa 3 Activada por Mitógenos/química , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/química , Estructura Molecular , FN-kappa B/química , Óxido Nítrico Sintasa de Tipo II/química , Filipinas , Poríferos , Factor de Necrosis Tumoral alfa/química , Proteínas Quinasas p38 Activadas por Mitógenos/química
10.
Phytother Res ; 30(3): 426-38, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26676298

RESUMEN

Breast cancer, the most commonly diagnosed cancer in women worldwide, is treated in various ways. Ramalin is a chemical compound derived from the Antarctic lichen Ramalina terebrata and is known to exhibit antioxidant and antiinflammatory activities. However, its effect on breast cancer cells remains unknown. We examined the ability of ramalin to induce apoptosis and its mechanisms in MCF-7 and MDA-MB-231 human breast cancer cell lines. Ramalin inhibited cell growth and induced apoptosis in both cell lines in a concentration-dependent manner. By upregulating Bax and downregulating Bcl-2, ramalin caused cytochrome c and apoptosis-inducing factor to be released from the mitochondria into the cytosol, thus activating the mitochondrial apoptotic pathway. In addition, activated caspase-8 and caspase-9 were detected in both types of cells exposed to ramalin, whereas ramalin activated caspase-3 only in the MDA-MB-231 cells. Ramalin treatment also increased the levels of LC3-II and p62. Moreover, the inhibition of autophagy by 3-methyladenine or Atg5 siRNA significantly enhanced ramalin-induced apoptosis, which was accompanied by a decrease in Bcl-2 levels and an increase in Bax levels. Therefore, autophagy appears to be activated as a protective mechanism against apoptosis in cancer cells exposed to ramalin. These findings suggest that ramalin is a potential anticancer agent for the treatment of patients with non-invasive or invasive breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Productos Biológicos/farmacología , Neoplasias de la Mama/metabolismo , Glutamatos/farmacología , Líquenes/química , Adenina/análogos & derivados , Adenina/metabolismo , Antineoplásicos/uso terapéutico , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Factor Inductor de la Apoptosis/metabolismo , Proteína 5 Relacionada con la Autofagia , Productos Biológicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Femenino , Glutamatos/uso terapéutico , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
11.
Phytother Res ; 30(12): 1978-1987, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27558640

RESUMEN

Atopic dermatitis (AD) is a chronic inflammatory skin disease that involves eczematous skin lesions with pruritic erythematous papules. In this study, we investigated the mitigating effects of ramalin, a component of the Antarctic lichen Ramalina terebrata against AD in vivo and in vitro. Oral administration of ramalin lessened scratching behaviors and significantly reduced both serum immunoglobulin E and IL-4 levels, and mRNA levels of IL-4 and IL-10 in AD-induced Balb/c mice. In vitro, treatment with ramalin produced significantly less inflammatory chemokines and cytokines, including TARC, MCP-1, RANTES, and IL-8 in TNF-α-stimulated HaCaT cells. In addition, ramalin inhibited the activation of nuclear factor-kappa B as well as the phosphorylation of mitogen-activated protein kinases (MAPK). Furthermore, ramalin treatment resulted in decreased production of ß-hexosaminidase and proinflammatory cytokines IL-4, IL-6, and TNF-α in 2,4 dinitrophenyl-human serum albumin-stimulated RBL-2H3 cells through blocking MAPK signaling pathways. The results suggest that ramalin modulates the production of immune mediators by inhibiting the nuclear factor-kappa B and MAPK signaling pathways. Taken together, ramalin effectively attenuated the development of AD and promoted the mitigating effects on Th2 cell-mediated immune responses and the production of inflammatory mediators in mast cells and keratinocytes. Thus, ramalin may be a potential therapeutic agent for AD. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Dermatitis Atópica/tratamiento farmacológico , Glutamatos/química , Queratinocitos/efectos de los fármacos , Mastocitos/efectos de los fármacos , Enfermedades de la Piel/tratamiento farmacológico , Piel/efectos de los fármacos , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Transducción de Señal
12.
Infect Immun ; 83(1): 108-19, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25312953

RESUMEN

Alcohol impairs the host immune system, rendering the host more vulnerable to infection. Therefore, alcoholics are at increased risk of acquiring serious bacterial infections caused by Streptococcus pneumoniae, including pneumonia. Nevertheless, how alcohol affects pneumococcal virulence remains unclear. Here, we showed that the S. pneumoniae type 2 D39 strain is ethanol tolerant and that alcohol upregulates alcohol dehydrogenase E (AdhE) and potentiates pneumolysin (Ply). Hemolytic activity, colonization, and virulence of S. pneumoniae, as well as host cell myeloperoxidase activity, proinflammatory cytokine secretion, and inflammation, were significantly attenuated in adhE mutant bacteria (ΔadhE strain) compared to D39 wild-type bacteria. Therefore, AdhE might act as a pneumococcal virulence factor. Moreover, in the presence of ethanol, S. pneumoniae AdhE produced acetaldehyde and NADH, which subsequently led Rex (redox-sensing transcriptional repressor) to dissociate from the adhE promoter. An increase in AdhE level under the ethanol condition conferred an increase in Ply and H2O2 levels. Consistently, S. pneumoniae D39 caused higher cytotoxicity to RAW 264.7 cells than the ΔadhE strain under the ethanol stress condition, and ethanol-fed mice (alcoholic mice) were more susceptible to infection with the D39 wild-type bacteria than with the ΔadhE strain. Taken together, these data indicate that AdhE increases Ply under the ethanol stress condition, thus potentiating pneumococcal virulence.


Asunto(s)
Alcohol Deshidrogenasa/metabolismo , Etanol/metabolismo , Regulación Enzimológica de la Expresión Génica , Streptococcus pneumoniae/enzimología , Estreptolisinas/toxicidad , Factores de Virulencia/metabolismo , Alcohol Deshidrogenasa/genética , Animales , Proteínas Bacterianas/toxicidad , Supervivencia Celular , Tolerancia a Medicamentos , Eliminación de Gen , Macrófagos/microbiología , Macrófagos/fisiología , Masculino , Ratones Endogámicos ICR , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/patogenicidad , Virulencia , Factores de Virulencia/genética
13.
Biochem Biophys Res Commun ; 467(4): 638-44, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26499075

RESUMEN

Fisetin (3,7,3',4'-tetrahydroxyflavone) is a naturally found flavonol in many fruits and vegetables and is known to have anti-aging, anti-cancer and anti-viral effects. However, the effects of fisetin on early adipocyte differentiation and the epigenetic regulator controlling adipogenic transcription factors remain unclear. Here, we show that fisetin inhibits lipid accumulation and suppresses the expression of PPARγ in 3T3-L1 cells. Fisetin suppressed early stages of preadipocyte differentiation, and induced expression of Sirt1. Depletion of Sirt1 abolished the inhibitory effects of fisetin on intracellular lipid accumulation and on PPARγ expression. Mechanistically, fisetin facilitated Sirt1-mediated deacetylation of PPARγ and FoxO1, and enhanced the association of Sirt1 with the PPARγ promoter, leading to suppression of PPARγ transcriptional activity, thereby repressing adipogenesis. Lowering Sirt1 levels reversed the effects of fisetin on deacetylation of PPARγ and increased PPARγ transactivation. Collectively, our results suggest the effects of fisetin in increasing Sirt1 expression and in epigenetic control of early adipogenesis.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Flavonoides/farmacología , Sirtuina 1/metabolismo , Células 3T3-L1 , Acetilación , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Flavonoles , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Metabolismo de los Lípidos , Ratones , PPAR gamma/metabolismo
14.
Chem Res Toxicol ; 28(1): 71-86, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25531190

RESUMEN

Senescence, which is irreversible cell cycle arrest, is induced by various types of DNA damage, including genotoxic stress. Senescent cells show dysregulation of tumor suppressor genes and other regulators of cellular proliferation. Activating transcription factor 3 (ATF3) plays a pleiotropic role in biological processes through genotoxic stress. In this study, we examined the effects of acrylamide (ACR), a genotoxic carcinogen, on cellular senescence and the molecular mechanisms of ATF3 function in macrophages. Treatment of macrophages with ACR at low concentrations (<1.0 mM) resulted in senescence-like morphology and an increase in senescence-associated ß-galactosidase (SA-ß-gal) activity. Exposure of macrophages to ACR led to stress-induced, telomerase-independent senescence. In addition, ACR treatment for 1, 3, or 5 days showed a concentration-dependent increase in ATF3 expression and G0/G1 phase arrest. To better understand the role of ATF3 in controlling the senescence response to ACR, SA-ß-gal activity was examined using ATF3 knockdown and overexpression. ACR-mediated senescence was significantly decreased by knockdown of ATF3, whereas it was increased with ATF3 overexpression. We found that ATF3 regulated p53 and p21 levels. ATF3 also played an important role in regulating intracellular reactive oxygen species (ROS) production in response to ACR treatment. Moreover, phosphorylation of p38 and JNK kinases, which were activated during ATF3-mediated senescence, was observed in ACR-treated macrophages. Taken together, these results suggest that ATF3 contributes to ACR-induced senescence by enhancing ROS production, activating p38 and JNK kinases, and promoting the ATF3-dependent expression of p53, resulting in regulation of cellular senescence in macrophages.


Asunto(s)
Acrilamida/toxicidad , Carcinógenos/toxicidad , Senescencia Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/fisiología , Fibroblastos , MAP Quinasa Quinasa 4/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Telomerasa , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Biosci Biotechnol Biochem ; 79(4): 539-52, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25494680

RESUMEN

Cell adhesion molecules play a critical role in inflammatory processes and atherosclerosis. In this study, we investigated the effect of ramalin, a chemical compound from the Antarctic lichen Ramalina terebrata, on vascular cell adhesion molecule-1 (VCAM-1) expression induced by TNF-α in vascular smooth muscular cells (VSMCs). Pretreatment of VSMCs with ramalin (0.1-10 µg/mL) concentration-dependently inhibited TNF-α-induced VCAM-1 expression. Additionally, ramalin inhibited THP-1 (human acute monocytic leukemia cell line) cell adhesion to TNF-α-stimulated VSMCs. Ramalin suppressed TNF-α-induced production of reactive oxygen species (ROS), PADI4 expression, and phosphorylation of p38, ERK, and JNK. Moreover, ramalin inhibited TNF-α-induced translocation of NF-κB and AP-1. Inhibition of PADI4 expression by small interfering RNA or the PADI4-specific inhibitor markedly attenuated TNF-α-induced activation of NF-κB and AP-1 and VCAM-1 expression in VSMCs. Our study provides insight into the mechanisms underlying ramalin activity and suggests that ramalin may be a potential therapeutic agent to modulate inflammation within atherosclerosis.


Asunto(s)
Antioxidantes/farmacología , Glutamatos/farmacología , Hidrolasas/genética , FN-kappa B/genética , Factor de Transcripción AP-1/genética , Molécula 1 de Adhesión Celular Vascular/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Adhesión Celular/efectos de los fármacos , Línea Celular , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Hidrolasas/metabolismo , Líquenes , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Infect Dis ; 210(11): 1745-54, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24951825

RESUMEN

BACKGROUND: Activating transcription factor-3 (ATF3) is known as a suppressor of cytokine production after exposure to lipopolysaccharide or during gram-negative bacterial infection. However, the mechanism by which ATF3 regulates innate immunity against gram-positive bacterial infection, particularly Streptococcus pneumoniae, remains unknown. METHODS: The wild-type and ATF3 knock-out (KO) mice were infected intranasally (i.n) or intraperitoneally with S. pneumoniae, and bacterial colonization or survival rate was determined. Pneumococcal pneumonia was induced by i.n infection, and ATF3 level was determined by Western blot. ATF3 KO cells or ATF3 siRNA transfection were used to determine expression of ATF3 downstream genes. Enzyme-linked immunosorbent assay was used to examine cytokines levels. RESULTS: ATF3 was highly expressed in various cell lines in vitro and in many organs in vivo. Pneumolysin was a novel inducer of ATF3. Pneumococcal infection induced ATF3, which subsequently stimulated production of cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-1ß, and interferon [IFN]-γ). ATF3-mediated cytokine induction protected the host from pneumococcal infection. In the pneumonia infection model, the bacterial clearance of wild-type mice was more efficient than those of ATF3 KO mice. CONCLUSIONS: Taken together, we can conclude that ATF3 regulates innate immunity positively upon pneumococcus infection by enhancing TNF-α, IL-1ß, and IFN-γ expression and modulating bacterial clearance.


Asunto(s)
Factor de Transcripción Activador 3/genética , Citocinas/biosíntesis , Resistencia a la Enfermedad/genética , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae , Factor de Transcripción Activador 3/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Línea Celular , Citocinas/genética , Modelos Animales de Enfermedad , Resistencia a la Enfermedad/inmunología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Ratones , Ratones Noqueados , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/mortalidad , Regiones Promotoras Genéticas , Unión Proteica , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/metabolismo , Estreptolisinas/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Infect Immun ; 82(9): 3802-10, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24980975

RESUMEN

Caseinolytic protease L (ClpL) is a member of the HSP100/Clp chaperone family, which is found mainly in Gram-positive bacteria. ClpL is highly expressed during infection for refolding of stress-induced denatured proteins, some of which are important for adherence. However, the role of ClpL in modulating pneumococcal virulence is poorly understood. Here, we show that ClpL impairs pneumococcal adherence to A549 lung cells by inducing and activating Rap1 and Rac1, thus increasing phosphorylation of cofilin (inactive form). Moreover, infection with a clpL mutant (ΔclpL) causes a greater degree of filopodium formation than D39 wild-type (WT) infection. Inhibition of Rap1 and Rac1 impairs filopodium formation and pneumococcal adherence. Therefore, ClpL can reduce pneumococcal adherence to A549 cells, likely via modulation of Rap1- and Rac1-mediated filopodium formation. These results demonstrate a potential role for ClpL in pneumococcal resistance to host cell adherence during infection. This study provides insight into further understanding the interactions between hosts and pathogens.


Asunto(s)
Adhesión Bacteriana/fisiología , Proteínas Bacterianas/metabolismo , Neoplasias Pulmonares/metabolismo , Infecciones Neumocócicas/metabolismo , Serina Endopeptidasas/metabolismo , Streptococcus pneumoniae/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Factores Despolimerizantes de la Actina/genética , Factores Despolimerizantes de la Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Adhesión Bacteriana/genética , Proteínas Bacterianas/genética , Línea Celular Tumoral , Endopeptidasa Clp , Humanos , Neoplasias Pulmonares/genética , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/microbiología , Serina Endopeptidasas/genética , Complejo Shelterina , Streptococcus pneumoniae/genética , Proteínas de Unión a Telómeros/genética , Virulencia/genética , Proteína de Unión al GTP rac1/genética
19.
Mod Pathol ; 26(3): 327-35, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22996377

RESUMEN

Ribosomal protein S6 is a key regulator of 40S ribosome biogenesis, and its phosphorylation is closely related to cell growth capacity. However, as a downstream target of S6 kinases, the clinical significance and the roles of S6 and S6 phosphorylation in cell viability and motility of esophageal squamous cell carcinoma remain unclear. Here, we show that high level of phosphorylated-ribosomal protein S6 (p-S6) (immunohistochemistry score ≥5) and an increased ratio of p-S6/S6 (immunohistochemistry score ≥0.75) were significantly associated with shortened disease-free survival in patients with esophageal squamous cell carcinoma in univariate analysis (P=0.049 and P<0.001, respectively). After adjusting for age, tumor-nodes-metastasis stage, chemotherapy, and radiation therapy in multivariate analysis, both p-S6 (hazard ratio 2.21, P=0.005) and p-S6/S6 (hazard ratio 2.40, P<0.001) remained independent adverse prognostic factors. In addition, S6 and S6 kinase 1 knockdown resulted in attenuation of viability by suppressing cyclin D1 expression in esophageal cancer cells. Furthermore, depletion of S6 and S6 kinase 1 resulted in a reduction in esophageal cancer cell migration and invasion. This was paralleled by a reduction in focal adhesion and by suppression of extracellular signal-regulated kinase and c-jun N-terminal kinase phosphorylation, which control cell motility. Collectively, these findings suggest that p-S6 and the ratio of p-S6/S6 are closely relevant to tumor progression and have prognostic significance in esophageal squamous cell carcinoma.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Neoplasias Esofágicas/enzimología , Proteína S6 Ribosómica/metabolismo , Anciano , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Ciclina D1/metabolismo , Supervivencia sin Enfermedad , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/terapia , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Adhesiones Focales/enzimología , Humanos , Inmunohistoquímica , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis Multivariante , Invasividad Neoplásica , Fosforilación , Pronóstico , Modelos de Riesgos Proporcionales , Interferencia de ARN , Proteína S6 Ribosómica/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Transfección
20.
Hepatology ; 55(6): 1727-37, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22183976

RESUMEN

UNLABELLED: Nutrient homeostasis is tightly regulated by the balance between energy production and utilization. During fasting, production of ketone bodies as an alternative energy source is critical to maintain nutrient homeostasis. An important component in the nutrient-sensitive signaling pathway is S6 kinase 2 (S6K2), a downstream effector of mammalian target of rapamycin. Here, we show that mice lacking S6K2 exhibit elevated levels of ketone bodies and enhanced peroxisome proliferator-activated receptor alpha (PPARα) activity upon nutrient availability. Consistent with this, knockdown of S6K2 increases the transcriptional activity of PPARα. S6K2 suppresses PPARα by associating with its corepressor, nuclear receptor corepressor 1 (NCoR1), and by inducing the recruitment of NCoR1 to the nucleus. Moreover, ob/ob mice, a genetic model of obesity, have markedly elevated S6K2 activity, and S6K2 was strongly associated with NCoR1 in the nucleus of liver cells. CONCLUSION: Our findings suggest that S6K2 regulates hepatic energy homeostasis by repressing PPARα activity and point to its potential relevance for therapeutic strategies designed to modulate S6K2 activity as a treatment for deregulated ketone body production.


Asunto(s)
Cuerpos Cetónicos/biosíntesis , Hígado/metabolismo , PPAR alfa/fisiología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos , Co-Represor 1 de Receptor Nuclear/fisiología , Obesidad/metabolismo , Fosforilación , Proteínas/fisiología , Proteínas Quinasas S6 Ribosómicas/deficiencia , Serina-Treonina Quinasas TOR , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA