Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Intervalo de año de publicación
1.
Am J Rhinol Allergy ; 32(4): 228-235, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29676176

RESUMEN

Purpose Methyl-CpG-binding protein 2 (MeCP2), known as a transcriptional regulator, has been suggested to play an important role in myofibroblast differentiation in the lung. The purpose of this study was to investigate the role of MeCP2 in transforming growth factor (TGF)- ß1-induced myofibroblast differentiation and extracellular matrix (ECM) production in nasal polyp-derived fibroblasts (NPDFs). Methods To identify the expression of MeCP2 in nasal polyp tissues, immunohistochemistry staining and Western blot were performed. TGF- ß1-induced NPDFs were treated with 5-azacytidine, a DNA methylation inhibitor, and the expression levels of α-SMA and fibronectin were determined by semiquantitative reverse transcription polymerase chain reaction, immunofluorescent staining, and Western blotting. The total soluble collagen was analyzed by the Sircol collagen assay. MeCP2 silenced by MeCP2-specific small interference ( si) RNA was verified by Western blot. Results The expression levels of MeCP2 increased in nasal polyp tissues compared to normal inferior turbinate tissues. 5-Azacytidine significantly inhibited the expression of α-SMA and fibronectin mRNA in a dose-dependent manner. In addition, 5-azacytidine suppressed collagen production and the expression of MeCP2 in the same manner. The expression levels of a-SMA and collagen production were significantly blocked by MeCP2 silencing in TGF- ß1-induced NPDFs. Conclusions Our data suggest that MeCP2 plays an essential role in TGF- ß1-induced myofibroblast differentiation and ECM production in NPDFs.


Asunto(s)
Matriz Extracelular/metabolismo , Fibroblastos/fisiología , Proteína 2 de Unión a Metil-CpG/metabolismo , Miofibroblastos/fisiología , Pólipos Nasales/inmunología , Actinas/genética , Actinas/metabolismo , Adulto , Azacitidina/farmacología , Diferenciación Celular , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/genética , ARN Interferente Pequeño/genética , Factor de Crecimiento Transformador beta1/metabolismo , Cornetes Nasales/citología
2.
Am J Rhinol Allergy ; 31(6): 357-363, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29122080

RESUMEN

PURPOSE: Activation of adenosine monophosphate-activated protein kinase (AMPK) by metformin, as a master regulator of metabolism, is involved in airway tissue remodeling. Here, we investigated the physical role of AMPK on cell migration, matrix contraction, and the production of matrix metalloproteinases (MMP) in nasal polyp-derived fibroblasts (NPDF). METHODS: Primary NPDFs from six patients with chronic rhinosinusitis and nasal polyps were isolated and cultured. To assess the effect of AMPK on fibroblast migration, we conducted scratch and migration assays in NPDF treated with metformin and/or compound C. A collagen gel contraction assay measured activity of contractile. MMP expression was measured with reverse transcription-polymerase chain reaction, Western blot, and zymography. To evaluate for specific AMPK action, we examined by AMPK small interfering RNA. RESULTS: Metformin, an activator of AMPK, significantly inhibited cell migration in NPDFs in a dose-dependent manner. Compound C, an inhibitor of AMPK, partially reversed the inhibitory effect of metformin. Metformin also significantly decreased contractile activity, with a concomitant reduction in the production of MMP-1 and MMP-2 but not of MMP-9. Specific silencing that targeted AMPK resulted in the enhancement of mobility and contractility and in the production of MMP-1 and MMP-2. CONCLUSION: AMPK played an important role in regulating cell migration, matrix contraction, and MMP production in NPDFs, which provided data that AMPK activator might be a therapeutic target for the prevention of tissue remodeling in nasal polyps.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Movimiento Celular , Metaloproteinasa 1 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/biosíntesis , Pólipos Nasales/etiología , Movimiento Celular/efectos de los fármacos , Femenino , Fibroblastos/fisiología , Humanos , Masculino , Metformina/farmacología , Pólipos Nasales/metabolismo , Pólipos Nasales/patología
3.
Sci Rep ; 7(1): 7299, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28779150

RESUMEN

To investigate the potential role of vitamin D (1,25(OH)2D3) in preventing the development of nasal polyps, we examined the effect of vitamin D on myofibroblast differentiation and extracellular matrix (ECM) production in TGF-ß1-induced nasal polyp-derived fibroblasts (NPDFs) and elucidated the mechanisms underlying its inhibitory effect. 1,25(OH)2D3 significantly reduced expression levels of α-SMA, a myofibroblast marker, and fibronectin, a representative ECM component, in a dose-dependent manner in TGF-ß1-induced NPDFs. 1,25(OH)2D3 suppressed activated Smad2/3 in time-course. Up-regulation of α-SMA, fibronectin and phosphorylation of Smad2/3 by TGF-ß1 was unaffected by 1,25(OH)2D3 in NPDFs after vitamin D receptor-specific siRNA transfection. We confirmed that the Smad2/3-specific inhibitor SIS3 inactivated Smad2/3 and reduced α-SMA and fibronectin expression. Furthermore, acetylation of histone H3 was compromised by 1,25(OH)2D3, leading to inhibition of collagen 1A1, collagen 1A2 and α-SMA gene expression. Treatment with 1,25(OH)2D3 also significantly suppressed TGF-ß1-enhanced contractility and motility in a contraction assay and Transwell migration assay. Finally, 1,25(OH)2D3 had a similar effect in ex vivo organ cultures of nasal polyps. Taken together, our results suggest that 1,25(OH)2D3 might be an effective therapy for nasal polyps by reducing myofibroblast differentiation and ECM production mediated by Smad2/3-dependent TGF-ß1 signaling pathways in NPDFs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Miofibroblastos/citología , Miofibroblastos/efectos de los fármacos , Pólipos Nasales/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Acetilación , Actinas/metabolismo , Movimiento Celular , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Histonas/metabolismo , Humanos , Modelos Biológicos , Pólipos Nasales/patología , Unión Proteica , Factor de Crecimiento Transformador beta1/metabolismo
4.
Am J Rhinol Allergy ; 30(4): 128-33, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27456588

RESUMEN

BACKGROUND: Wogonin has been shown to have antifibrotic and anti-inflammatory effects in the lower airway. The purpose of this study was to evaluate the effects of wogonin on transforming growth factor (TGF) ß1-induced myofibroblast differentiation, extracellular matrix production, migration, and collagen contraction, and to determine the molecular mechanisms of wogonin in nasal polyp-derived fibroblasts (NPDF). METHODS: NPDFs were isolated from nasal polyps from eight patients. TGF-ß1-induced NPDFs were treated with wogonin. Cytotoxicity was evaluated by using a 3-(4,5- dimethylthiazol-2yl)-2,5-diphenyl-tetrazolium bromide assay. Fibroblast migration was evaluated with transwell and scratch migration assays. The expression levels of α-smooth muscle actin, fibronectin, phosphorylated-p38, and c-Fos were determined by Western blot and/or reverse transcription-polymerase chain reaction. The total collagen amount was analyzed with the Sircol collagen assay, and contractile activity was measured by a collagen gel contraction assay. RESULTS: Wogonin (0-60 µM) had no significant cytotoxic effects on TGF-ß1-induced NPDFs. Migration of NPDFs was significantly inhibited by wogonin treatment. The expression levels of α-smooth muscle actin and fibronectin were significantly reduced in wogonin-treated NPDFs. Collagen production and contraction were also significantly decreased by wogonin treatment. Wogonin markedly inhibited activation of the p38/activator protein 1 pathway in TGF-ß1-induced NPDFs. CONCLUSION: These results indicated that wogonin may inhibit TGF-ß1-induced myofibroblast differentiation, extracellular matrix production, migration, and collagen contraction through the p38/activator protein-1 pathway in NPDFs.


Asunto(s)
Matriz Extracelular/metabolismo , Flavanonas/farmacología , Pólipos Nasales/patología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/fisiología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Humanos , Miofibroblastos/citología , Miofibroblastos/efectos de los fármacos , Transducción de Señal/fisiología
5.
Am J Rhinol Allergy ; 30(6): 211-217, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28124643

RESUMEN

PURPOSE: Rapamycin has antiproliferative and antifibrogenic effects in vitro and in vivo. The purpose of this study was to evaluate the effects of rapamycin on transforming growth factor (TGF) beta 1 induced myofibroblast differentiation (alpha smooth-muscle actin [SMA]), extracellular matrix production, and collagen contraction in nasal polyp-derived fibroblasts (NPDF). The underlying molecular mechanisms of rapamycin were also determined in NPDFs. METHODS: NPDFs were grown in culture and transformed into myofibroblasts by using TGF beta 1 (5 ng/mL). For cytotoxicity evaluation, a 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyl-tetrazolium bromide assay was used. Expression levels of alpha SMA, phosphorylated phosphatidylinositol 3-kinase (PI3K), and phosphorylated mammalian target of rapamycin (mTOR) were determined by using Western blot, reverse transcription-polymerase chain reaction, and immunofluorescence staining. The total amount of collagen was analyzed by using the Sircol collagen assay, and contractile activity was measured with a collagen gel contraction assay. Silencing mTOR with mTOR-specific small interference RNA was determined by using reverse transcription-polymerase chain reaction. RESULTS: Whereas rapamycin (range, 0-400 nM) had no significant cytotoxic effects on TGF beta 1 induced NPDFs, it significantly reduced the expression levels of alpha-SMA in TGF beta 1 induced NPDFs in a dose-dependent manner. TGF beta 1 induced collagen production and collagen contraction were significantly inhibited by rapamycin treatment. Rapamycin also attenuated the TGF beta 1 induced activation of PI3K and mTOR, and its inhibitory effects were similar to those of mTOR silencing and a specific PI3K inhibitor. CONCLUSIONS: Rapamycin inhibited TGF beta 1 induced myofibroblast differentiation, extracellular matrix production, and collagen contraction through the PI3K/mTOR signal pathways in NPDFs.


Asunto(s)
Fibroblastos/efectos de los fármacos , Inmunosupresores/farmacología , Miofibroblastos/efectos de los fármacos , Pólipos Nasales/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Actinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/fisiología , Humanos , Miofibroblastos/fisiología , Pólipos Nasales/metabolismo , Pólipos Nasales/patología , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Factor de Crecimiento Transformador beta/metabolismo
6.
Int Forum Allergy Rhinol ; 5(10): 923-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26097205

RESUMEN

BACKGROUND: Fibroblast migration is crucial for normal wound repair after sinonasal surgery. Histamine is known to be involved in wound healing by its effects on cell proliferation and migration. This study aimed to determine whether histamine affects the migration of nasal fibroblasts and to investigate the mechanism of action of histamine on nasal fibroblasts. METHODS: Primary cultures of nasal fibroblasts were established from inferior turbinate samples. Fibroblast migration was evaluated with scratch assays. Cells were treated with histamine and/or histamine receptor-selective antagonists. U-73122 and pertussis toxin, which are selective inhibitors of the lower signaling pathway of H1R and H4R, were used to confirm the modulation of nasal fibroblast migration by histamine. Fibroblast cytoskeletal structures were visualized with immunocytochemistry. RESULTS: Histamine significantly stimulated the migration of nasal fibroblasts. Antagonists selective for HR1 and HR4 significantly reduced nasal fibroblast migration. In immunocytochemical staining, histamine treatment increased membrane ruffling and pyrilamine, diphenhydramine, fexofenadine, and JNJ7777120 decreased histamine-induced membrane ruffling. U-73122 and pertussis toxin also decreased histamine-induced migration of fibroblasts. Histamine maintains its stimulatory effects on fibroblast migration in the presence of mitomycin C, which blocks proliferation of cells. CONCLUSION: We showed that histamine stimulates fibroblast migration in nasal fibroblasts. This effect appeared to be mediated by HR1 and HR4. However, because fibroblast migration also can be involved in scaring and fibrosis, more research is necessary to determine the effects of antihistamine on wound healing after sinus surgery.


Asunto(s)
Fibroblastos/fisiología , Histamina/metabolismo , Cornetes Nasales/citología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Cultivadas , Citoesqueleto/metabolismo , Estrenos/farmacología , Fibroblastos/efectos de los fármacos , Histamina/inmunología , Humanos , Inmunización , Indoles/farmacología , Toxina del Pertussis/farmacología , Piperazinas/farmacología , Pirrolidinonas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Histamínicos , Receptores Histamínicos H4 , Transducción de Señal/efectos de los fármacos
7.
PLoS One ; 9(11): e90683, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25390332

RESUMEN

Nasal polyposis is characterized by persistent inflammation and remodeling in sinonasal mucosa. Toll-like receptors (TLRs) play a role in the innate immune response to microbes in the sinonasal cavity. The aim of this study was to evaluate whether nasal polyp-derived fibroblasts (NPDFs) and organ-cultured nasal polyps can synthesize pro-inflammatory cytokines and matrix metalloproteinases (MMPs) after exposure to lipopolysaccharide (LPS), a TLR4 agonist. NPDFs and organ-cultured nasal polyps were isolated from nasal polyps of 8 patients and exposed to LPS. The mRNA and protein expression levels of TLRs, cytokines, and MMPs were determined using a gene expression microarray, real-time RT-PCR, western blot analysis, enzyme-linked immunosorbent assay, and immunofluorescence staining. The enzymatic activities of MMPs were analyzed using collagen or gelatin zymography. The protein expression level of MMP-1 increased in nasal polyp tissues compared to inferior turbinate tissues. LPS induced mRNA expression of TLR4, IL-6, IL-8, and MMP-1 and activated MAPK signaling in NPDFs. LPS promoted the release of interleukin (IL)-6 through extracellular signal-related kinase (ERK) and IL-8 through ERK and c-Jun N-terminal kinases (JNK). Production of IL-6 and IL-8 was induced by PI3K/Akt signaling in LPS-stimulated NPDFs. LPS increased the transcript and protein expression levels of MMP-1 and induced collagenase activity of MMP-1 via ERK and p38, but did not induce gelatinase activity of MMP-2 and MMP-9. LPS from Rhodobacter sphaeroides (LPS-RS) inhibited the stimulatory effects of LPS in NPDFs as well as in organ culture of nasal polyp. LPS triggers immune response via TLR 4 and activates MAPK and PI3K/Akt signaling pathway, which is involved in remodeling of nasal polyps.


Asunto(s)
Citocinas/metabolismo , Fibroblastos/metabolismo , Lipopolisacáridos/farmacología , Metaloproteinasas de la Matriz/metabolismo , Pólipos Nasales/metabolismo , Receptor Toll-Like 4/metabolismo , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Pólipos Nasales/patología , Técnicas de Cultivo de Órganos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Am J Rhinol Allergy ; 28(3): e120-4, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24980222

RESUMEN

BACKGROUND: Fibroblast migration is crucial for normal wound repair after sinonasal surgery. Prostaglandin E2 (PGE2) is a potent inhibitor of fibroblast functions including chemotaxis, proliferation, and matrix production. The purpose of this study was to determine whether PGE2 affects the migration of nasal fibroblasts and to investigate the mechanism of action of PGE2 on nasal fibroblasts. METHODS: Primary cultures of nasal fibroblasts were established from inferior turbinate samples. Fibroblast migration was evaluated with scratch assays. Reverse-transcription polymerase chain reaction was performed for E prostanoid (EP) 1, EP2, EP3, and EP4 receptors. EP receptor-selective agonists and antagonists were used to evaluate receptor functions. Stimulatory G (Gs) proteins were activated to evaluate mechanisms. Intracellular cyclic adenosine monophosphate (cAMP) levels were measured by ELISA, and fibroblast cytoskeletal structures were visualized with immunocytochemistry. RESULTS: PGE2 significantly reduced the migration of nasal fibroblasts. Agonists selective for the EP2 and EP4 receptors significantly reduced the nasal fibroblast migration. Antagonists of the EP2 and EP4 receptors inhibited the effect of PGE2 on nasal fibroblast migration. Activation of Gs protein and adenyl cyclase reduced nasal fibroblast migration. CONCLUSION: PGE2 inhibited the migration of nasal fibroblasts via the EP2 and EP4 receptors, and this inhibition was mediated by cAMP elevation. Targeting specific EP receptors could offer therapeutic opportunities for conditions such as delayed wound healing after nasal surgery.


Asunto(s)
Alprostadil/análogos & derivados , Compuestos de Bifenilo/farmacología , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Endoscopía , Fibroblastos/efectos de los fármacos , Senos Paranasales/efectos de los fármacos , Pirrolidinonas/farmacología , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Xantonas/farmacología , Alprostadil/farmacología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Dinoprostona/farmacología , Fibroblastos/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Terapia Molecular Dirigida , Senos Paranasales/patología , Senos Paranasales/cirugía , Subtipo EP1 de Receptores de Prostaglandina E/agonistas , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP3 de Receptores de Prostaglandina E/agonistas , Subtipo EP3 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Cicatrización de Heridas
9.
Otolaryngol Head Neck Surg ; 150(1): 148-53, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24357397

RESUMEN

UNLABELLED: BACKGROUND AND OBJECTS: Metformin is widely used to treat type 2 diabetes mellitus, and adenosine monophosphate-activated protein kinase (AMPK) is thought to be the target that mediates its effects. Recently, it has been demonstrated that metformin has antifibrotic effects beyond its antihyperglycemic action. The purposes of this study were to investigate the effect of metformin on TGF-ß1-induced myofibroblast differentiation (α-smooth muscle actin [α-SMA]) and extracellular matrix (ECM) production and to determine the underlying mechanism of the action of metformin in nasal polyp-derived fibroblasts (NPDFs). STUDY DESIGN: Basic research. SETTING: The rhinology laboratory of Korea University Guro Hospital, Seoul, Korea. METHODS: NPDFs from 7 patients were incubated with TGF-ß1 and treated with metformin or compound C, an inhibitor of AMPK. To determine the proliferation rate of nasal fibroblasts, a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay was performed. The expression levels of α-SMA and fibronectin were determined by reverse transcription-polymerase chain reaction (RT-PCR), Western blotting, and immunofluorescent staining. Phosphorylation of AMPK and phosphorylation of Smad2/3 were evaluated by Western blot analysis. RESULTS: In TGF-ß1-induced NPDFs, metformin inhibited the expression of α-SMA and fibronectin, as confirmed by both RT-PCR and Western blot analysis. Metformin increased the phosphorylation of AMPK and the expression levels of α-SMA and fibronectin. However, compound C reversed these effects. Metformin inhibited TGF-ß1-induced phosphorylation of Smad2/3. CONCLUSIONS: This study showed that metformin inhibits TGF-ß1-induced myofibroblast differentiation and ECM production in NPDFs via the Smad2/3 pathway. AMPK can be a therapeutic target for the prevention of ECM remodeling in nasal polyps.


Asunto(s)
Matriz Extracelular/fisiología , Hipoglucemiantes/farmacología , Metformina/farmacología , Miofibroblastos/diagnóstico por imagen , Pólipos Nasales/patología , Factor de Crecimiento Transformador beta1/fisiología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Actinas/análisis , Adulto , Western Blotting , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Fibronectinas/análisis , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ultrasonografía
10.
Am J Rhinol Allergy ; 27(1): 18-23, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23406592

RESUMEN

BACKGROUND: Nasal polyposis is associated with a chronic inflammatory condition of the sinonasal mucosa and involves myofibroblast differentiation and extracellular matrix (ECM) accumulation. Epigenetic modulation by histone deacetylase (HDAC) inhibitors including trichostatin A (TSA) has been reported to have inhibitory effects on myofibroblast differentiation in lung and renal fibroblasts. The purpose of this study was to investigate the inhibitory effect of TSA on myofibroblast differentiation and ECM production in nasal polyp organ cultures. METHODS: Nasal polyp tissues from 18 patients were acquired during endoscopic sinus surgery. After organ culture, nasal polyps were stimulated with TGF-beta1 and then treated with TSA. Alpha-smooth muscle actin (α-SMA), fibronectin, and collagen type I expression levels were examined by reverse transcription-polymerase chain reaction (PCR), real-time PCR, Western blot, and immunofluorescent staining. HDAC2, HDAC4, and acetylated H4 expression levels were assayed by Western blot. Cytotoxicity was analyzed by the terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling assay. RESULTS: The expression levels of α-SMA, fibronectin, and collagen type 1 were increased in nasal polyp after transforming growth factor (TGF) beta1 treatment. TSA-inhibited TGF-beta1 induced these gene and protein expression levels. Furthermore, TSA suppressed protein expression levels of HDAC2 and HDAC4. However, TSA induced hyperacetylation of histones H4. Treatment with TGF-beta1 with or without TSA did not have cytotoxic effect. CONCLUSION: These findings provide novel insights into the epigenetic regulation in myofibroblast differentiation and ECM production of nasal polyp. TSA could be a candidate of a therapeutic agent for reversing the TGF-beta1-induced ECM synthesis that leads to nasal polyp development.


Asunto(s)
Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/enzimología , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Miofibroblastos/efectos de los fármacos , Pólipos Nasales/enzimología , Pólipos Nasales/patología , Acetilación/efectos de los fármacos , Actinas/genética , Actinas/metabolismo , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Miofibroblastos/enzimología , Miofibroblastos/patología , Técnicas de Cultivo de Órganos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Crecimiento Transformador beta1/inmunología
11.
Phytother Res ; 27(1): 16-20, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22422627

RESUMEN

The purposes of this study were to determine whether berberine has any effect on phenotype changes and extracellular matrix (ECM) production in nasal polyp-derived fibroblasts (NPDFs) and to investigate the underlying molecular mechanism. NPDFs were pre-treated with berberine prior to induction by transforming growth factor (TGF)-ß1. The expression of α-smooth muscle actin (SMA) and collagen type I mRNA was determined by a reverse transcription-polymerase chain reaction, and the expression of α-SMA protein and collagen type I was determined by western blotting and/or immunofluorescent staining. The total soluble collagen production was analysed by the SirCol collagen assay. The expression of several signaling molecules of the TGF-ß1 pathway was evaluated by western blot analysis. In TGF-ß1-induced NPDFs, berberine significantly inhibited the expression of α-SMA and collagen type I mRNA and reduced α-SMA and collagen protein levels. Berberine only suppressed the expression of pp38 among the evaluated signaling molecules. SB203580 (a specific inhibitor of p38 kinase) markedly suppressed the increased expression of collagen type I and α-SMA in TGF-ß1-induced NPDFs. Berberine exerts suppressive effects on phenotype changes and ECM production in NPDFs via p38 signaling pathway interference. The findings provide new therapeutic options for ECM production in nasal polyps.


Asunto(s)
Berberina/farmacología , Diferenciación Celular/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Pólipos Nasales/patología , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Actinas/metabolismo , Adulto , Células Cultivadas , Colágeno Tipo I/metabolismo , Femenino , Humanos , Masculino , Miofibroblastos/citología , Factor de Crecimiento Transformador beta1/farmacología
12.
Exp Biol Med (Maywood) ; 237(6): 663-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22728707

RESUMEN

Nasal polyps are associated with chronic inflammation of the sinonasal mucosa and are involved in myofibroblast differentiation and extracellular matrix (ECM) accumulation. Ginsenoside Rg1, a compound derived from Panax ginseng, shows antifibrotic and anticancer effects. However, the molecular effects of Rg1 on myofibroblast differentiation and ECM production remain unknown. The aims of this study were to investigate the effect of Rg1 on transforming growth factor (TGF)-ß1-induced myofibroblast differentiation and ECM production and to determine the molecular mechanism of Rg1 in nasal polyp-derived fibroblasts (NPDFs). NPDFs were isolated from nasal polyps of seven patients who had chronic rhinosinusitis with nasal polyp. NPDFs were exposed to TGF-ß1 with or without Rg1. Expression levels of α-smooth muscle actin (SMA), fibronectin and collagen type Iα1 were determined by reverse transcription polymerase chain reaction, Western blot and immunofluorescent staining. TGF-ß1 signaling molecules, including Smad2/3, extracellular signal-regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 were analyzed by Western blotting. Transcription factors involved with TGF-ß1 signaling, nuclear factor (NF)-κB and activator protein 1 (AP-1) were also assessed by Western blot. The cytotoxic effect of Rg1 was measured by an established viability assay. The mRNA and protein expression levels of α-SMA, fibronectin and collagen type Iα1 were increased in TGF-ß1-induced NPDFs. Rg1 inhibited these effects. The inhibitory molecular mechanism of Rg1 was involved in the ERK pathway. Rg1 inhibited the transcription factor activation of AP-1. Rg1 itself was not cytotoxic. The ginsenoside Rg1 has inhibitory effects on myofibroblast differentiation and ECM production. The inhibitory mechanism of Rg1 is involved with the ERK and AP-1 signaling pathways. Rg1 may be useful as an inhibitor of ECM deposition, and has potential to be used as a novel treatment option for nasal polyps.


Asunto(s)
Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Ginsenósidos/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Pólipos Nasales/patología , Transducción de Señal/fisiología , Factor de Transcripción AP-1/metabolismo , Actinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Fibroblastos/patología , Fibronectinas/metabolismo , Humanos , FN-kappa B/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA