Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 209(8): 1532-1544, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36165197

RESUMEN

Streptococcus pneumoniae is major cause of otitis media (OM) and life-threatening pneumonia. Overproduction of mucin, the major component of mucus, plays a critical role in the pathogenesis of both OM and pneumonia. However, the molecular mechanisms underlying the tight regulation of mucin upregulation in the mucosal epithelium by S. pneumoniae infection remain largely unknown. In this study, we show that S. pneumoniae pneumolysin (PLY) activates AMP-activated protein kinase α1 (AMPKα1), the master regulator of energy homeostasis, which is required for S. pneumoniae-induced mucin MUC5AC upregulation in vitro and in vivo. Moreover, we found that PLY activates AMPKα1 via cholesterol-dependent membrane binding of PLY and subsequent activation of the Ca2+- Ca2+/calmodulin-dependent kinase kinase ß (CaMKKß) and Cdc42-mixed-lineage protein kinase 3 (MLK3) signaling axis in a TLR2/4-independent manner. AMPKα1 positively regulates PLY-induced MUC5AC expression via negative cross-talk with TLR2/4-dependent activation of MAPK JNK, the negative regulator of MUC5AC expression. Moreover, pharmacological inhibition of AMPKα1 suppressed MUC5AC induction in the S. pneumoniae-induced OM mouse model, thereby demonstrating its therapeutic potential in suppressing mucus overproduction in OM. Taken together, our data unveil a novel mechanism by which negative cross-talk between TLR2/4-independent activation of AMPKα1 and TLR2/4-dependent activation of JNK tightly regulates the S. pneumoniae PLY-induced host mucosal innate immune response.


Asunto(s)
Otitis Media , Streptococcus pneumoniae , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Proteínas Bacterianas , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Calmodulina/metabolismo , Colesterol/metabolismo , Inmunidad Innata , Ratones , Otitis Media/tratamiento farmacológico , Estreptolisinas/metabolismo , Receptor Toll-Like 2/metabolismo
2.
Chem Pharm Bull (Tokyo) ; 72(4): 365-373, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38569867

RESUMEN

Obesity is known to be associated with increased inflammation and dysregulated autophagy, both of which contribute to insulin resistance. Saikosaponin-A (SSA) has been reported to exhibit anti-inflammatory and lipid-lowering properties. In this research, we employed a combination of computational modeling and animal experiments to explore the effects of SSA. Male C57BL/6 mice were categorized into four groups: normal diet, high-fat diet (HFD), HFD + atorvastatin 10 mg/kg, and HFD + SSA 10 mg/kg. We conducted oral glucose and fat tolerance tests to assess metabolic parameters and histological changes. Furthermore, we evaluated the population of Kupffer cells (KCs) and examined gene expressions related to inflammation and autophagy. Computational analysis revealed that SSA displayed high binding affinity to tumor necrosis factor (TNF)-α, nuclear factor (NF)-κB, fibroblast growth factor 21 (FGF21), and autophagy-related 7 (ATG7). Animal study demonstrated that SSA administration improved fasting and postprandial glucose levels, homeostatic model assessment of insulin resistance (HOMA-IR) index, as well as triglyceride, free fatty acid, total cholesterol, low-density lipoprotein cholesterol (LDL-C)-cholesterol, and high-density lipoprotein cholesterol (HDL-C)-cholesterol levels in HFD-fed mice. Moreover, SSA significantly reduced liver weight and fat accumulation, while inhibiting the infiltration and M1 activation of KCs. At the mRNA level, SSA downregulated TNF-α and NF-κB expression, while upregulating FGF21 and ATG7 expression. In conclusion, our study suggests that SSA may serve as a therapeutic agent for addressing the metabolic complications associated with obesity. This potential therapeutic effect is attributed to the suppression of inflammatory cytokines and the upregulation of FGF21 and ATG7.


Asunto(s)
Experimentación Animal , Resistencia a la Insulina , Ácido Oleanólico/análogos & derivados , Saponinas , Ratones , Masculino , Animales , Resistencia a la Insulina/fisiología , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Hígado , Inflamación/metabolismo , Glucosa/metabolismo , Colesterol , Dieta Alta en Grasa/efectos adversos , Factor de Necrosis Tumoral alfa/metabolismo , Insulina/metabolismo
3.
Int J Mol Sci ; 23(23)2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36499655

RESUMEN

Obesity-induced insulin resistance is the fundamental cause of metabolic syndrome. Accordingly, we evaluated the effect of mangiferin (MGF) on obesity and glucose metabolism focusing on inflammatory response and autophagy. First, an in silico study was conducted to analyze the mechanism of MGF in insulin resistance. Second, an in vivo experiment was conducted by administering MGF to C57BL/6 mice with high-fat-diet (HFD)-induced metabolic disorders. The in silico analysis revealed that MGF showed a high binding affinity with macrophage-related inflammatory cytokines and autophagy proteins. In the in vivo study, mice were divided into three groups: normal chow, HFD, and HFD + MGF 150 mg/kg. MGF administration to obese mice significantly improved the body weight, insulin-sensitive organs weights, glucose and lipid metabolism, fat accumulation in the liver, and adipocyte size compared to HFD alone. MGF significantly reduced the macrophages in adipose tissue and Kupffer cells, inhibited the gene expression ratio of tumor necrosis factor-α and F4/80 in adipose tissue, reduced the necrosis factor kappa B gene, and elevated autophagy-related gene 7 and fibroblast growth factor 21 gene expressions in the liver. Thus, MGF exerted a therapeutic effect on metabolic diseases by improving glucose and lipid metabolism through inhibition of the macrophage-mediated inflammatory responses and activation of autophagy.


Asunto(s)
Resistencia a la Insulina , Ratones , Animales , Ratones Endogámicos C57BL , Dieta Alta en Grasa/efectos adversos , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Inflamación/patología , Tejido Adiposo/metabolismo , Ratones Obesos , Autofagia , Glucosa/metabolismo
4.
Int J Mol Sci ; 22(15)2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34360840

RESUMEN

Obesity is characterized as a chronic, low-grade inflammation state accompanied by the infiltration of immune cells into adipose tissue and higher levels of inflammatory cytokines and chemokines. This study aimed to investigate the mechanisms and effects of Coptidis Rhizoma (CR) on obesity and its associated inflammation. First, we applied a network pharmacology strategy to search the target genes and pathways regulated by CR in obesity. Next, we performed in vivo experiments to confirm the antiobesity and anti-inflammatory effects of CR. Mice were assigned to five groups: normal chow (NC), control (high-fat diet (HFD)), HFD + CR 200 mg/kg, HFD + CR 400 mg/kg, and HFD + metformin 200 mg/kg. After 16 weeks of the experimental period, CR administration significantly reduced the weight of the body, epididymal fat, and liver; it also decreased insulin resistance, as well as the area under the curve of glucose in the oral glucose tolerance test and triglyceride in the oral fat tolerance test. We observed a decrease in adipose tissue macrophages (ATMs) and inflammatory M1 ATMs, as well as an increase in anti-inflammatory M2 ATMs. Gene expression levels of inflammatory cytokines and chemokines, including tumor necrosis factor-α, F4/80, and C-C motif chemokine (CCL)-2, CCL4, and CCL5, were suppressed in adipose tissue in the CR groups than levels in the control group. Additionally, histological analyses suggested decreased fat accumulation in the epididymal fat pad and liver in the CR groups than that in the control group. Taken together, these results suggest that CR has a therapeutic effect on obesity-induced inflammation, and it functions through the inhibition of macrophage-mediated inflammation in adipose tissue.


Asunto(s)
Citocinas/genética , Medicamentos Herbarios Chinos/farmacología , Inflamación/tratamiento farmacológico , Macrófagos/metabolismo , Obesidad/complicaciones , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Simulación por Computador , Coptis chinensis , Dieta Alta en Grasa , Regulación de la Expresión Génica , Inflamación/etiología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Molecules ; 26(22)2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34834083

RESUMEN

A sensitive and reproducible liquid chromatography-tandem mass spectrometry (LC-MS/MS) system was developed and fully validated for the simultaneous determination of ephedrine and pseudoephedrine in human plasma after oral administration of the herbal prescription Ojeok-san (OJS); 2-phenylethylamine was used as the internal standard (IS). Both compounds presented a linear calibration curve (r2 ≥ 0.99) over a concentration range of 0.2-50 ng/mL. The developed method was fully validated in terms of selectivity, lower limit of quantitation, precision, accuracy, recovery, matrix effect, and stability, according to the regulatory guidelines from the U.S. Food and Drug Administration and the Korea Ministry of Food and Drug Safety. This validated method was successfully applied for the pharmacokinetic assessment of ephedrine and pseudoephedrine in 20 healthy Korean volunteers administered OJS.


Asunto(s)
Efedrina , Extractos Vegetales/administración & dosificación , Seudoefedrina , Espectrometría de Masas en Tándem , Administración Oral , Cromatografía Liquida , Efedrina/administración & dosificación , Efedrina/farmacocinética , Femenino , Humanos , Masculino , Seudoefedrina/administración & dosificación , Seudoefedrina/farmacocinética , República de Corea
6.
Proc Natl Acad Sci U S A ; 113(2): E165-71, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26719415

RESUMEN

Myeloid differentiation factor 88 (MyD88) acts as a crucial adaptor molecule for Toll-like receptors (TLRs) and interleukin (IL)-1 receptor signaling. In contrast to the well-studied positive regulation of MyD88 signaling, how MyD88 signaling is negatively regulated still remains largely unknown. Here, we demonstrate for the first time to our knowledge that MyD88 protein undergoes lysine 63 (K63)-linked polyubiquitination, which is functionally critical for mediating TLR-MyD88-dependent signaling. Deubiquitinase CYLD negatively regulates MyD88-mediated signaling by directly interacting with MyD88 and deubiquitinating nontypeable Haemophilus influenzae (NTHi)-induced K63-linked polyubiquitination of MyD88 at lysine 231. Importantly, we further confirmed this finding in the lungs of mice in vivo by using MyD88(-/-)CYLD(-/-) mice. Understanding how CYLD deubiquitinates K63-linked polyubiquitination of MyD88 may not only bring insights into the negative regulation of TLR-MyD88-dependent signaling, but may also lead to the development of a previously unidentified therapeutic strategy for uncontrolled inflammation.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Haemophilus influenzae/fisiología , Inflamación/microbiología , Lisina/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación , Animales , Enzima Desubiquitinante CYLD , Células HeLa , Humanos , Inflamación/metabolismo , Ratones , Modelos Biológicos , Poliubiquitina/metabolismo , Unión Proteica
7.
Int J Mol Sci ; 20(3)2019 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-30744038

RESUMEN

Insulin resistance is strongly associated with the metabolic syndrome, and chronic inflammation is known to be a major mechanism of insulin resistance and is a therapeutic target. This study was designed to evaluate the effect of Scutellaria baicalensis (SB) in high-fat diet (HFD)-induced insulin-resistant mice and to investigate its mechanism based on inflammatory responses. Mice were fed a HFD to induce insulin resistance and then administered SB for nine weeks. Body weight, glucose, lipid, insulin, epididymal fat pad and liver weights, and histologic characteristics were evaluated to determine the effect on insulin resistance. In order to evaluate the effects on the inflammatory process, we analyzed the proportions of macrophages in liver and epididymal fat and measured inflammatory gene expression. Fasting and postprandial glucose, fasting insulin, HOMA-IR, triglycerides, and low density lipoprotein cholesterol levels were significantly decreased by SB administration. The epididymal fat and liver showed significant weight decreases and histological improvements. Total adipose tissue macrophages (ATMs) decreased (27.71 ± 3.47% vs. 45.26 ± 7.26%, p < 0.05), M2 ATMs increased (47.02 ± 6.63% vs. 24.28 ± 8.00%, p < 0.05), and CD11b⁺ Kupffer cells decreased. The expression levels of tumor necrosis factor alpha and F4/80 in the liver were significantly decreased (12.03 ± 1.47% vs. 25.88 ± 4.57%, p < 0.05) compared to HFD group. These results suggest that SB improved insulin resistance through inhibition of macrophage-mediated inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Resistencia a la Insulina , Obesidad/metabolismo , Extractos Vegetales/farmacología , Scutellaria baicalensis/química , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Antiinflamatorios/química , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Insulina/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Macrófagos/metabolismo , Masculino , Ratones , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/patología , Extractos Vegetales/química
8.
Int J Mol Sci ; 20(12)2019 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-31212747

RESUMEN

Obesity is a chronic low-grade inflammatory condition in which hypertrophied adipocytes and adipose tissue immune cells, mainly macrophages, contribute to increased circulating levels of proinflammatory cytokines. Obesity-associated chronic low-grade systemic inflammation is considered a focal point and a therapeutic target in insulin resistance and metabolic diseases. We evaluate the effect of Poncirus fructus (PF) on insulin resistance and its mechanism based on inflammatory responses in high-fat diet (HFD)-induced obese mice. Mice were fed an HFD to induce obesity and then administered PF. Body weight, epididymal fat and liver weight, glucose, lipid, insulin, and histologic characteristics were evaluated to determine the effect of PF on insulin resistance by analyzing the proportion of macrophages in epididymal fat and liver and measured inflammatory gene expression. PF administration significantly decreased the fasting and postprandial glucose, fasting insulin, HOMA-IR, total-cholesterol, triglycerides, and low-density lipoprotein cholesterol levels. The epididymal fat tissue and liver showed a significant decrease of fat accumulation in histological analysis. PF significantly reduced the number of adipose tissue macrophages (ATMs), F4/80+ Kupffer cells, and CD68+ Kupffer cells, increased the proportion of M2 phenotype macrophages, and decreased the gene expression of inflammatory cytokines. These results suggest that PF could be used to improve insulin resistance through modulation of macrophage-mediated inflammation and enhance glucose and lipid metabolism.


Asunto(s)
Inflamación/etiología , Inflamación/metabolismo , Resistencia a la Insulina , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Obesidad/complicaciones , Extractos Vegetales/farmacología , Poncirus/química , Animales , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Expresión Génica , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Inflamación/tratamiento farmacológico , Inflamación/patología , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/anatomía & histología , Hígado/efectos de los fármacos , Hígado/metabolismo , Macrófagos/inmunología , Ratones , Ratones Obesos , Extractos Vegetales/química , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/metabolismo
9.
Proc Natl Acad Sci U S A ; 112(14): E1800-9, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25831493

RESUMEN

Phosphodiesterase 4B (PDE4B) plays a key role in regulating inflammation. Roflumilast, a phosphodiesterase (PDE)4-selective inhibitor, has recently been approved for treating severe chronic obstructive pulmonary disease (COPD) patients with exacerbation. However, there is also clinical evidence suggesting the development of tachyphylaxis or tolerance on repeated dosing of roflumilast and the possible contribution of PDE4B up-regulation, which could be counterproductive for suppressing inflammation. Thus, understanding how PDE4B is up-regulated in the context of the complex pathogenesis and medications of COPD may help improve the efficacy and possibly ameliorate the tolerance of roflumilast. Here we show that roflumilast synergizes with nontypeable Haemophilus influenzae (NTHi), a major bacterial cause of COPD exacerbation, to up-regulate PDE4B2 expression in human airway epithelial cells in vitro and in vivo. Up-regulated PDE4B2 contributes to the induction of certain important chemokines in both enzymatic activity-dependent and activity-independent manners. We also found that protein kinase A catalytic subunit ß (PKA-Cß) and nuclear factor-κB (NF-κB) p65 subunit were required for the synergistic induction of PDE4B2. PKA-Cß phosphorylates p65 in a cAMP-dependent manner. Moreover, Ser276 of p65 is critical for mediating the PKA-Cß-induced p65 phosphorylation and the synergistic induction of PDE4B2. Collectively, our data unveil a previously unidentified mechanism underlying synergistic up-regulation of PDE4B2 via a cross-talk between PKA-Cß and p65 and may help develop new therapeutic strategies to improve the efficacy of PDE4 inhibitor.


Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Factor de Transcripción ReIA/metabolismo , Animales , Dominio Catalítico , Línea Celular , AMP Cíclico/metabolismo , Ciclopropanos/farmacología , Haemophilus influenzae , Humanos , Inflamación , Ratones , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica , ARN Interferente Pequeño/metabolismo , Regulación hacia Arriba
10.
Int J Mol Sci ; 19(11)2018 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-30413022

RESUMEN

Phosphodiesterase 4B (PDE4B) plays an important role in inflammation. Recently we have reported that roflumilast as a PDE4-selective inhibitor, synergizes with nontypeable Haemophilus influenzae (NTHi) to up-regulate PDE4B expression in vitro and in vivo. Clinical evidence and our previous results suggest that synergistic induction of PDE4B could be counterproductive for suppressing inflammation or may contribute to tolerance to roflumilast. We thus investigated if dexamethasone inhibits the synergistic induction of PDE4B by roflumilast and NTHi as well as inflammation. Here, dexamethasone markedly suppressed the synergistic induction of PDE4B in human lung epithelial cells and in vivo. We also found that dexamethasone further suppressed NTHi-induced inflammatory response in vitro and in vivo. Moreover, Compound A, as a dissociating non-steroidal glucocorticoid receptor (GR) ligand, inhibited the synergistic induction of PDE4B, thereby suggesting the requirement of dexamethasone-mediated GR activation in the suppression of PDE4B expression. Taken together, our data suggest that dexamethasone may help attenuate inflammation and tolerance through suppressing the PDE4B expression in chronic obstructive pulmonary disease (COPD) patients using roflumilast.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Dexametasona/administración & dosificación , Inflamación/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Aminopiridinas/administración & dosificación , Animales , Benzamidas/administración & dosificación , Línea Celular , Ciclopropanos/administración & dosificación , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica , Haemophilus influenzae/efectos de los fármacos , Haemophilus influenzae/patogenicidad , Humanos , Inflamación/genética , Inflamación/microbiología , Inflamación/patología , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/microbiología
11.
Int J Mol Sci ; 19(11)2018 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-30463229

RESUMEN

Visfatin, a member of the adipokine family, plays an important role in many metabolic and stress responses. The mechanisms underlying the direct therapeutic effects of visfatin on wound healing have not been reported yet. In this study, we examined the effects of visfatin on wound healing in vitro and in vivo. Visfatin enhanced the proliferation and migration of human dermal fibroblasts (HDFs) and keratinocytes the expression of wound healing-related vascular endothelial growth factor (VEGF) in vitro and in vivo. Treatment of HDFs with visfatin induced activation of both extracellular signal-regulated kinases 1 and 2 (ERK1/2) and c-Jun N-terminal kinases 1 and 2 (JNK1/2) in a time-dependent manner. Inhibition of ERK1/2 and JNK1/2 led to a significant decrease in visfatin-induced proliferation and migration of HDFs. Importantly, blocking VEGF with its neutralizing antibodies suppressed the visfatin-induced proliferation and migration of HDFs and human keratinocytes, indicating that visfatin induces the proliferation and migration of HDFs and human keratinocytes via increased VEGF expression. Moreover, visfatin effectively improved wound repair in vivo, which was comparable to the wound healing activity of epidermal growth factor (EGF). Taken together, we demonstrate that visfatin promotes the proliferation and migration of HDFs and human keratinocytes by inducing VEGF expression and can be used as a potential novel therapeutic agent for wound healing.


Asunto(s)
Citocinas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Nicotinamida Fosforribosiltransferasa/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dermis/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Queratinocitos/metabolismo , Ratones Endogámicos BALB C , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Int J Mol Sci ; 19(10)2018 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-30314368

RESUMEN

A flavonoid Astragalin (kaempferol-3-O-ß-d-glucopyranoside, Ast) has several biological activities including anti-oxidant, anti-HIV, and anti-allergic effects. Nonetheless, its insolubility in hydrophilic solvents imposes restrictions on its therapeutic applications. In this study, we investigated the effects of water-soluble astragalin-galactoside (kaempferol-3-O-ß-d-isomaltotrioside, Ast-Gal) on murine bone marrow-derived dendritic cell (DC) maturation and T helper (Th) cell-mediated immune responses. Ast-Gal significantly increased maturation and activation of DCs through the upregulation of surface markers, such as cluster of differentiation (CD)80, CD86, and Major histocompatibility complex (MHC) II in a dose-dependent manner, while Ast had little effects. Additionally, Ast-Gal-treated DCs markedly secreted immune-stimulating cytokines such as interleukin (IL)-1ß, IL-6, and IL-12. Importantly, Ast-Gal strongly increased expression of IL-12, a polarizing cytokine of Th1 cells. In a co-culture system of DCs and CD4⁺ T cells, Ast-Gal-treated DCs preferentially differentiates naïve CD4⁺ T cells into Th1 cells. The addition of neutralizing IL-12 monoclonal antibody (mAb) to cultures of Ast-Gal-treated DCs and CD4⁺ T cells significantly decreased interferon (IFN)-γ production, thereby indicating that Ast-Gal-stimulated DCs enhance the Th1 response through IL-12 production by DCs. Injection with Ast-Gal-treated DCs in mice increased IFN-γ-secreting Th1 cell population. Collectively, these findings indicate that hydrophilically modified astragalin can enhance Th1-mediated immune responses via DCs and point to a possible application of water-soluble astragalin-galactoside as an immune adjuvant.


Asunto(s)
Células Dendríticas/inmunología , Galactósidos/farmacología , Inmunidad/efectos de los fármacos , Quempferoles/farmacología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Animales , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/metabolismo , Expresión Génica , Inmunofenotipificación , Ratones , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células TH1/metabolismo
13.
J Immunol ; 194(12): 5990-8, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25972475

RESUMEN

Mucin overproduction is a hallmark of otitis media (OM). Streptococcus pneumoniae is one of the most common bacterial pathogens causing OM. Mucin MUC5AC plays an important role in mucociliary clearance of bacterial pathogens. However, if uncontrolled, excessive mucus contributes significantly to conductive hearing loss. Currently, there is a lack of effective therapeutic agents that suppress mucus overproduction. In this study, we show that a currently existing antistroke drug, vinpocetine, a derivative of the alkaloid vincamine, inhibited S. pneumoniae-induced mucin MUC5AC upregulation in cultured middle ear epithelial cells and in the middle ear of mice. Moreover, vinpocetine inhibited MUC5AC upregulation by inhibiting the MAPK ERK pathway in an MKP-1-dependent manner. Importantly, ototopical administration of vinpocetine postinfection inhibited MUC5AC expression and middle ear inflammation induced by S. pneumoniae and reduced hearing loss and pneumococcal loads in a well-established mouse model of OM. Thus, these studies identified vinpocetine as a potential therapeutic agent for inhibiting mucus production in the pathogenesis of OM.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/metabolismo , Mucina 5AC/genética , Otitis Media/genética , Otitis Media/metabolismo , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/metabolismo , Alcaloides de la Vinca/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Otitis Media/tratamiento farmacológico , Otitis Media/microbiología , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae , Alcaloides de la Vinca/administración & dosificación
14.
Biochim Biophys Acta ; 1842(3): 446-62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23707515

RESUMEN

There is increasing evidence showing that inflammation is an important pathogenic mediator of the development of obesity-induced insulin resistance. It is now generally accepted that tissue-resident immune cells play a major role in the regulation of this obesity-induced inflammation. The roles that adipose tissue (AT)-resident immune cells play have been particularly extensively studied. AT contains most types of immune cells and obesity increases their numbers and activation levels, particularly in AT macrophages (ATMs). Other pro-inflammatory cells found in AT include neutrophils, Th1 CD4 T cells, CD8 T cells, B cells, DCs, and mast cells. However, AT also contains anti-inflammatory cells that counter the pro-inflammatory immune cells that are responsible for the obesity-induced inflammation in this tissue. These anti-inflammatory cells include regulatory CD4 T cells (Tregs), Th2 CD4 T cells, and eosinophils. Hence, AT inflammation is shaped by the regulation of pro- and anti-inflammatory immune cell homeostasis, and obesity skews this balance towards a more pro-inflammatory status. Recent genetic studies revealed several molecules that participate in the development of obesity-induced inflammation and insulin resistance. In this review, the cellular and molecular players that participate in the regulation of obesity-induced inflammation and insulin resistance are discussed, with particular attention being placed on the roles of the cellular players in these pathogeneses. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.


Asunto(s)
Tejido Adiposo/inmunología , Inflamación/metabolismo , Resistencia a la Insulina/genética , Obesidad/inmunología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Humanos , Inflamación/inmunología , Inflamación/patología , Resistencia a la Insulina/inmunología , Factor 4 Similar a Kruppel , Macrófagos/citología , Macrófagos/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Obesidad/metabolismo , Obesidad/patología , Células TH1/inmunología , Células TH1/metabolismo
15.
BMC Complement Altern Med ; 15: 380, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26490686

RESUMEN

BACKGROUND: Benign prostatic hyperplasia (BPH) is one of the common male diseases, which is provoked by dihydrotestosterone (DHT) and androgen signals. Several studies showed that curcumin has various effects of prevention and treatment to diseases. We investigated whether curcumin may repress the development of BPH in male Wistar rats. METHODS: Seven weeks male Wistar rats were and divided into 4 groups (normal group, BPH group, finasteride group, curcumin group; n = 8 for each group). In order to induce BPH in rats, rats were castrated and testosterone was injected subcutaneously everyday (s.c., 20 mg/kg). Rats in the curcumin group were treated 50 mg/kg, administered orally for 4 weeks. After 4 weeks, all rats were sacrificed and their prostate and serum were analyzed. RESULTS: Compared to the finasteride group as positive group, the curcumin group showed similarly protective effect on BPH in histopathologic morphology, prostate volume. Results of immunohistochemistry and western-blot showed decreased expressions of VEGF, TGF-ß1, and IGF1 were also decreased in the curcumin group. CONCLUSIONS: These results suggested that curcumin inhibited the development of BPH and might a useful herbal treatment or functional food for BPH.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Curcumina/farmacología , Próstata/efectos de los fármacos , Hiperplasia Prostática/tratamiento farmacológico , Análisis de Varianza , Animales , Biomarcadores/metabolismo , Glucemia/efectos de los fármacos , Western Blotting , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Ratas , Ratas Wistar , Testosterona/sangre , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
J Immunol ; 188(12): 6371-80, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22581859

RESUMEN

Inflammation is a hallmark of many important human diseases. Appropriate inflammation is critical for host defense; however, an overactive response is detrimental to the host. Thus, inflammation must be tightly regulated. The molecular mechanisms underlying the tight regulation of inflammation remain largely unknown. Ecotropic viral integration site 1 (EVI1), a proto-oncogene and zinc finger transcription factor, plays important roles in normal development and leukemogenesis. However, its role in regulating NF-κB-dependent inflammation remains unknown. In this article, we show that EVI1 negatively regulates nontypeable Haemophilus influenzae- and TNF-α-induced NF-κB-dependent inflammation in vitro and in vivo. EVI1 directly binds to the NF-κB p65 subunit and inhibits its acetylation at lysine 310, thereby inhibiting its DNA-binding activity. Moreover, expression of EVI1 itself is induced by nontypeable Haemophilus influenzae and TNF-α in an NF-κB-dependent manner, thereby unveiling a novel inducible negative feedback loop to tightly control NF-κB-dependent inflammation. Thus, our study provides important insights into the novel role for EVI1 in negatively regulating NF-κB-dependent inflammation, and it may also shed light on the future development of novel anti-inflammatory strategies.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Retroalimentación Fisiológica/fisiología , Inflamación/metabolismo , FN-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/inmunología , Ensayo de Cambio de Movilidad Electroforética , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/metabolismo , Haemophilus influenzae/inmunología , Inmunoprecipitación , Inflamación/inmunología , Proteína del Locus del Complejo MDS1 y EV11 , Ratones , Ratones Mutantes , FN-kappa B/inmunología , Proto-Oncogenes Mas , Proto-Oncogenes/inmunología , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción ReIA/inmunología , Factores de Transcripción/inmunología , Transfección , Factor de Necrosis Tumoral alfa/inmunología
17.
J Biol Chem ; 287(27): 22799-811, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22610099

RESUMEN

Otitis media (OM) is the most common childhood bacterial infection and the major cause of conductive hearing loss in children. Mucus overproduction is a hallmark of OM. Streptococcus pneumoniae is the most common gram-positive bacterial pathogen causing OM. Among many mucin genes, MUC5AC has been found to be greatly up-regulated in the middle ear mucosa of human patients with OM. We previously reported that S. pneumoniae up-regulates MUC5AC expression in a MAPK ERK-dependent manner. We also found that MAPK phosphatase-1 (MKP-1) negatively regulates S. pneumoniae-induced ERK-dependent MUC5AC up-regulation. Therapeutic strategies for up-regulating the expression of negative regulators such as MKP-1 may have significant therapeutic potential for treating mucus overproduction in OM. However, the underlying molecular mechanism by which MKP-1 expression is negatively regulated during S. pneumoniae infection is unknown. In this study we show that phosphodiesterase 4B (PDE4B) mediates S. pneumoniae-induced MUC5AC up-regulation by inhibiting the expression of a negative regulator MKP-1, which in turn leads to enhanced MAPK ERK activation and subsequent up-regulation of MUC5AC. PDE4B inhibits MKP-1 expression in a cAMP-PKA-dependent manner. PDE4-specific inhibitor rolipram inhibits S. pneumoniae-induced MUC5AC up-regulation both in vitro and in vivo. Moreover, we show that PDE4B plays a critical role in MUC5AC induction. Finally, topical and post-infection administration of rolipram into the middle ear potently inhibited S. pneumoniae-induced MUC5AC up-regulation. Collectively, these data demonstrate that PDE4B mediates ERK-dependent up-regulation of mucin MUC5AC by S. pneumoniae by inhibiting cAMP-PKA-dependent MKP-1 pathway. This study may lead to novel therapeutic strategy for inhibiting mucus overproduction.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Fosfatasa 1 de Especificidad Dual/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Mucina 5AC/metabolismo , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/metabolismo , Animales , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Oído Medio/citología , Oído Medio/inmunología , Células Epiteliales/citología , Células Epiteliales/enzimología , Células Epiteliales/microbiología , Humanos , Ratones , Ratones Endogámicos C57BL , Moco/metabolismo , Otitis Media/inmunología , Otitis Media/metabolismo , Otitis Media/microbiología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , ARN Interferente Pequeño/genética , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Regulación hacia Arriba/fisiología
18.
J Nanosci Nanotechnol ; 13(11): 7643-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24245307

RESUMEN

Ag nanoparticles, used for halogen (especially iodine) adsorption and an evaluation of halogen behavior, were embedded in synthesized inorganic-organic hybrid gels. In particular, an irradiation method using an electron beam plays a part in introducing Ag nanoparticles to the organofunctionalized silica gels from AgNO3 solutions in a simple way at atmospheric pressure and room temperature. For preparation of the Ag nanoparticle-embedded inorganic-organic hybrid gels, ligands of ethylenediamine (NH2CH2CH2NH-, TMSen) and mercapto (HS-) functionalized three-dimensional porous SiO2 sol-gels were first synthesized through hydrolysis and condensation reactions, and Ag nanoparticles were then embedded into the ethylenediamine- and mercapto-anchored silica gels each, through electron-beam irradiation. The addition of ligands yielded larger average pore sizes than the absence of any ligand. Moreover, the ethylenediamine ligand led to looser structures and better access of the Ag nanoparticles to the ethylenediamine-anchored gel. As a result, more Ag nanoparticles were introduced into the ethylenediamine-anchored gel. The preparation and characterization of Ag nanoparticle-embedded blank and ligand-anchored silica gels are discussed in detail.


Asunto(s)
Cristalización/métodos , Geles/química , Nanopartículas del Metal/química , Nanopartículas del Metal/ultraestructura , Dióxido de Silicio/química , Plata/química , Ligandos , Sustancias Macromoleculares/química , Ensayo de Materiales , Conformación Molecular , Tamaño de la Partícula , Porosidad , Propiedades de Superficie
19.
J Nanosci Nanotechnol ; 13(3): 1951-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23755627

RESUMEN

Nanocrystalline titanium dioxide (TiO2) thin films on silicon wafer substrates were prepared by sol-gel spin coating process. The prepared thin films were treated with electron beam (1.1 MeV, 300 kGy) at air atmosphere. The effects of electron-beam (EB) irradiation on the structural and optical properties of the TiO2 thin films were investigated. The structures of all the TiO2 thin films by XRD analysis showed an anatase phase, and the phase remained unchanged within the investigating range of EB treatment. The thickness of the titania thin film decreased slightly with EB treatment whereas the porosity increased. The EB treatment of TiO2 thin film can increase the proportion of Ti3+ in Ti2p at the thin film surface. The optical transmittance of the film in the wavelength ranges of above 380 nm increased after the EB treatment while its refractive index decreased with increasing EB dose. Therefore, improvement of the optical properties could be due to the change in both surface chemistry and morphology of the TiO2 thin films affected by EB irradiation.

20.
Heliyon ; 9(8): e19162, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37654445

RESUMEN

Dyslipidemia, a major risk factor for atherosclerotic cardiovascular disease, can be prevented by lowering low-density lipoprotein cholesterol (LDL-C) levels. The lipid improvement effects of Daeshiho-tang (DSHT), a herbal formula, have been proven through various preclinical studies based on anti-obesity and anti-inflammatory properties, however, clinical trials were few. This preliminary study aimed to assess the lipid-lowering effect of DSHT in patients with dyslipidemia and examine its safety profile. The study was a randomized, double-blind, placebo-controlled trial. The trial included 60 patients with untreated dyslipidemia: total cholesterol (TC) > 200 mg/dL, triglyceride (TG) > 150 mg/dL, LDL-C >130 mg/dL, or high-density lipoprotein cholesterol (HDL-C) <40 mg/dL. Participants (mean age, 44.7 ± 13.7 years) consumed DSHT extract or an equivalent placebo at a dose of 3 g, thrice a day for 8 weeks. Participants underwent blood tests assessing serum lipid and apolipoprotein (apo) levels, including LDL-C, HDL-C, TC, TG, apoA1, and apoB, at baseline and 4 and 8 weeks. Levels of biochemical safety markers, including AST, ALT, GGT, creatinine, and eGFR, were assessed throughout the study. Between the two groups, significant differences were observed in the changes of LDL-C, TC, and apoB concentrations from baseline to post-intervention. Compared with the placebo group, DSHT-administered participants showed significantly reduced LDL-C by 14.0 ± 4.66 mg/dL (p < 0.01), TC by 13.7 ± 4.73 mg/dL (p < 0.01) and apolipoprotein-B by 7.03 ± 3.23 mg/dL (p < 0.05). No significant changes were observed in the safety biochemical parameters in either group. DSHT treatment for 8 weeks improved LDL-C levels and reduced apoB concentrations without severe adverse events.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA