Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 686: 149118, 2023 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-37931361

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), like other viruses, can induce proliferation of myofibroblasts and even lead to fibrosis in the lung. Epithelial-mesenchymal transition (EMT) is thought to play an essential role in the pathogenesis of Coronavirus disease 19 (COVID-19). EMT is originally a critical process that regulates the development of different tissues in the embryo, but in inflammatory situations, EMT tries to be activated again to control inflammation or even heal inflammatory damage. However, in pathological situations, such as chronic viral infections (e.g., COVID-19) or pulmonary fibrosis initiation, this benign healing transforms into sinister nature, pushing the lung into the fibrotic process. Notably, the cytokines released by inflammatory cells and the chronic inflammatory microenvironment shared by fibrotic cells promote each other as critical factors in the induction of pathological EMT. In the induction of SARS-CoV-2 virus, cytokines are an essential mediator of EMT transformation, and a summary of whether COVID-19 patients, during the infection phase, have many persistent inflammatory mediators (cytokines) that are a causative factor of EMT has not yet appeared. The following common signaling drivers, including Transforming growth factor beta (TGF-ß), cytokines, Notch signaling pathway, Wnt and hypoxia signaling pathways, drive the regulation of EMT. In this review, we will focus on 3 key EMT signaling pathways: TGF-ß, Leucine zipper transcription factor like 1 (LZTFL1) and the common interleukin family expressed in the lung. TGF-ß-induced SNAIL and LZTFL1 were identified as regulatory EMT in COVID-19. For cytokines, the interleukin family is a common inducer of EMT and plays an essential role in the formation of the microenvironment of fibrosis. We sought to demonstrate that cytokines act as "communicators" and build the "microenvironment" of fibrosis together with EMT as a "bridge" to induce EMT in fibrosis. The mechanisms utilized by these two pathways could serve as templates for other mesenchymal transformations and provide new potential therapeutic targets.


Asunto(s)
COVID-19 , Fibrosis Pulmonar , Humanos , Fibrosis Pulmonar/patología , Citocinas/metabolismo , Transición Epitelial-Mesenquimal/fisiología , SARS-CoV-2/metabolismo , Fibrosis , Factor de Crecimiento Transformador beta/metabolismo , Interleucinas , Factor de Crecimiento Transformador beta1/metabolismo
2.
Biol Chem ; 403(3): 305-315, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34643076

RESUMEN

Transforming growth factor beta 1 (TGF-ß1) functions as a coupling factor between bone development and resorption. Matrix metalloproteinase 13 (MMP13) is important in bone remodeling, and skeletal dysplasia is caused by a deficiency in MMP13 expre-ssion. Runx2, a transcription factor is essential for bone development, and MMP13 is one of its target genes. TGF-ß1 promoted Runx2 phosphorylation, which was necessary for MMP13 production in osteoblastic cells, as we previously shown. Since the phosphorylation of some proteins causes them to be degraded by the ubiquitin/proteasome pathway, we hypothesized that TGF-ß1 might stabilize the phosphorylated Runx2 protein for its activity by other post-translational modification (PTM). This study demonstrated that TGF-ß1-stimulated Runx2 acetylation in rat osteoblastic cells. p300, a histone acetyltransferase interacted with Runx2, and it promoted Runx2 acetylation upon TGF-ß1-treatment in these cells. Knockdown of p300 decreased the TGF-ß1-stimulated Runx2 acetylation and MMP13 expression in rat osteoblastic cells. TGF-ß1-treatment stimulated the acetylated Runx2 bound at the MMP13 promoter, and knockdown of p300 reduced this effect in these cells. Overall, our studies identified the transcriptional regulation of MMP13 by TGF-ß1 via Runx2 acetylation in rat osteoblastic cells, and these findings contribute to the knowledge of events presiding bone metabolism.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal , Metaloproteinasa 13 de la Matriz , Osteoblastos , Factor de Crecimiento Transformador beta1 , Acetilación , Animales , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Osteoblastos/metabolismo , Procesamiento Proteico-Postraduccional , Ratas , Factor de Crecimiento Transformador beta1/farmacología
3.
Circ Res ; 126(12): 1723-1745, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32216531

RESUMEN

RATIONALE: Right ventricular (RV) fibrosis in pulmonary arterial hypertension contributes to RV failure. While RV fibrosis reflects changes in the function of resident RV fibroblasts (RVfib), these cells are understudied. OBJECTIVE: Examine the role of mitochondrial metabolism of RVfib in RV fibrosis in human and experimental pulmonary arterial hypertension. METHODS AND RESULTS: Male Sprague-Dawley rats received monocrotaline (MCT; 60 mg/kg) or saline. Drinking water containing no supplement or the PDK (pyruvate dehydrogenase kinase) inhibitor dichloroacetate was started 7 days post-MCT. At week 4, treadmill testing, echocardiography, and right heart catheterization were performed. The effects of PDK activation on mitochondrial dynamics and metabolism, RVfib proliferation, and collagen production were studied in RVfib in cell culture. Epigenetic mechanisms for persistence of the profibrotic RVfib phenotype in culture were evaluated. PDK expression was also studied in the RVfib of patients with decompensated RV failure (n=11) versus control (n=7). MCT rats developed pulmonary arterial hypertension, RV fibrosis, and RV failure. MCT-RVfib (but not left ventricular fibroblasts) displayed excess mitochondrial fission and had increased expression of PDK isoforms 1 and 3 that persisted for >5 passages in culture. PDK-mediated decreases in pyruvate dehydrogenase activity and oxygen consumption rate were reversed by dichloroacetate (in RVfib and in vivo) or siRNA targeting PDK 1 and 3 (in RVfib). These interventions restored mitochondrial superoxide and hydrogen peroxide production and inactivated HIF (hypoxia-inducible factor)-1α, which was pathologically activated in normoxic MCT-RVfib. Redox-mediated HIF-1α inactivation also decreased the expression of TGF-ß1 (transforming growth factor-beta-1) and CTGF (connective tissue growth factor), reduced fibroblast proliferation, and decreased collagen production. HIF-1α activation in MCT-RVfib reflected increased DNMT (DNA methyltransferase) 1 expression, which was associated with a decrease in its regulatory microRNA, miR-148b-3p. In MCT rats, dichloroacetate, at therapeutic levels in the RV, reduced phospho-pyruvate dehydrogenase expression, RV fibrosis, and hypertrophy and improved RV function. In patients with pulmonary arterial hypertension and RV failure, RVfib had increased PDK1 expression. CONCLUSIONS: MCT-RVfib manifest a DNMT1-HIF-1α-PDK-mediated, chamber-specific, metabolic memory that promotes collagen production and RV fibrosis. This epigenetic mitochondrial-metabolic pathway is a potential antifibrotic therapeutic target.


Asunto(s)
Epigénesis Genética , Ventrículos Cardíacos/metabolismo , Hipertensión Pulmonar/metabolismo , Mitocondrias Cardíacas/metabolismo , Miofibroblastos/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Animales , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Fibrosis , Ventrículos Cardíacos/patología , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Dinámicas Mitocondriales , Monocrotalina/toxicidad , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
4.
BMC Pulm Med ; 22(1): 183, 2022 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-35525938

RESUMEN

BACKGROUND: Transforming Growth Factor-ß1 (TGF-ß1) is a genetic modifier in patients with cystic fibrosis (CF). Several single nucleotide polymorphisms (SNPs) of TGF-ß1 are associated with neutrophilic inflammation, lung fibrosis and loss of pulmonary function. AIM: The aim of this study was to assess the relationship between genetic TGF-ß1 polymorphisms and pulmonary disease progression in CF patients. Furthermore, the effect of TGF-ß1 polymorphisms on inflammatory cytokines in sputum was investigated. METHODS: 56 CF-patients and 62 controls were genotyped for three relevant SNPs in their TGF-ß1 sequence using the SNaPshot® technique. Individual "slopes" in forced expiratory volume in 1 s (FEV1) for all patients were calculated by using documented lung function values of the previous five years. The status of Pseudomonas aeruginosa (Pa) infection was determined. Sputum concentrations of the protease elastase, the serine protease inhibitor elafin and the cytokines IL-1ß, IL-8, IL-6, TNF-α were measured after a standardized sputum induction and processing. RESULTS: The homozygous TT genotype at codon 10 was associated with a lower rate of chronic Pa infection (p < 0.05). The heterozygous GC genotype at codon 25 was associated with lower lung function decline (p < 0.05). Patients with homozygous TT genotype at the promotor SNP showed higher levels of TNF-α (p < 0,05). Higher levels of TGF-ß1 in plasma were associated with a more rapid FEV1 decline over five years (p < 0.05). CONCLUSIONS: Our results suggest that polymorphisms in the TGF-ß1 gene have an effect on lung function decline, Pa infection as well as levels of inflammatory cytokines. Genotyping these polymorphisms could potentially be used to identify CF patients with higher risk of disease progression. TGF-ß1 inhibition could potentially be developed as a new therapeutic option to modulate CF lung disease.


Asunto(s)
Fibrosis Quística , Factor de Crecimiento Transformador beta1 , Codón , Fibrosis Quística/genética , Citocinas/análisis , Progresión de la Enfermedad , Genotipo , Humanos , Pulmón , Polimorfismo de Nucleótido Simple , Factor de Crecimiento Transformador beta1/genética
5.
Mol Cell Biochem ; 476(9): 3433-3448, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33973131

RESUMEN

Amiodarone (AMD) is a widely used antiarrhythmic drug prescribed to treat cardiac tachyarrhythmias; however, AMD has been reported to provoke pulmonary fibrosis (PF) and hepatotoxicity. This study aimed to investigate the influence of alpha lipoic acid (ALA) on AMD-induced PF and hepatotoxicity in male Wistar rats. AMD administration resulted in elevated lung contents of hydroxyproline (Hyp), malondialdehyde (MDA), and increased serum levels of transforming growth factor beta-1 (TGF-ß1), interferon-γ (IFN-γ), alanine amino transaminase (ALT), aspartate amino transaminase (AST), total cholesterol (TC), and glucose. On the other side, lung content of glutathione reduced (GSH) and serum levels of total anti-oxidant capacity (TAC) were significantly decreased. Histopathologically, AMD caused PF, produced a mild hepatic injury, and increased expression of alpha smooth muscle actin (α-SMA). Treatment with ALA produced a significant reversal of the oxidative stress, fibrosis, and inflammation parameters with reductions in α-SMA expressions, leading to amelioration of histopathological lesions. ALA might provide supportive therapy in AMD-receiving cardiovascular patients.


Asunto(s)
Amiodarona/toxicidad , Antioxidantes/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Sustancias Protectoras/farmacología , Fibrosis Pulmonar/prevención & control , Ácido Tióctico/farmacología , Alanina Transaminasa/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citocinas/metabolismo , Glutatión/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Malondialdehído/metabolismo , Estrés Oxidativo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Ratas , Ratas Wistar , Vasodilatadores/toxicidad
6.
Biol Chem ; 400(12): 1617-1627, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31120854

RESUMEN

Geranylgeranyl diphosphate synthase (GGPPS) is an enzyme that catalyzes the synthesis of geranylgeranyl pyrophosphate (GGPP). GGPPS is implicated in many disorders, but its role in idiopathic pulmonary fibrosis (IPF) remains unclear. This study aimed to investigate the role of GGPPS in IPF. We established bleomycin-induced lung injury in a lung-specific GGPPS-deficient mouse (GGPPS-/-) and detected GGPPS expression in lung tissues by Western blot and immunohistochemistry analysis. We found that GGPPS expression increased during lung injury and fibrosis in mice induced by bleomycin, and GGPPS deficiency augmented lung fibrosis. GGPPS deficiency activated lung fibroblast by facilitating transforming growth factor ß1 while antagonizing bone morphogenetic protein 4 signaling. Notably, the supplementation of exogenous GGPP mitigated lung fibrosis in GGPPS-/- mice induced by bleomycin. In conclusion, our findings suggest that GGPPS provides protection against pulmonary fibrosis and that the restoration of protein geranylgeranylation may benefit statin-induced lung injury.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Farnesiltransferasa/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Complejos Multienzimáticos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Farnesiltransferasa/deficiencia , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/deficiencia
7.
Osteoarthritis Cartilage ; 27(4): 676-686, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30610922

RESUMEN

OBJECTIVE: Transforming growth factor beta 1 (TGF-ß1) is implicated in osteoarthritis (OA). The purpose of this study was to explore the ability of Losartan to inhibit the inflammatory signaling pathway of TGF-ß1 observed during osteoarthritic progression in the temporomandibular joint (TMJ) and knee joint using a genetic mouse model. METHODS: A murine OA model displaying the heterozygous chondrodysplasia gene (cho/+), a col11a1 mutation, was used to test this hypothesis. Following a 7-month treatment period with Losartan, the synovial joints were analyzed for histopathological improvement comparing two experimental groups. Tissues were fixed in paraformaldehyde, processed to paraffin section, and stained with Safranin O and Fast Green to visualize proteoglycans and collagen proteins in cartilage. Using the Modified Mankin scoring system, the degree of staining and OA progression were evaluated. RESULTS: Results show heterozygous animals receiving Losartan having diminished degeneration of TMJ condylar and knee joint articular cartilage. This was confirmed in the TMJ and knee by a statistically significant decrease in the Mankin histopathology score. Decreased expression of HtrA1, a key regulator to the TGF-ß1 signaling pathway, was demonstrated in vitro as well as in vivo, via Losartan inhibition. CONCLUSION: Using a genetic mouse model of OA, this study demonstrated the utility of Losartan to improve treatment of human OA in the TMJ and knee joint through inhibition of the TGF-ß1 signaling cascade. We further demonstrated inhibition of HtrA1, the lowering of Mankin scores to wild type control levels, and the limiting of OA progressive damage with treatment of Losartan.


Asunto(s)
Cartílago Articular/patología , Articulación de la Rodilla/diagnóstico por imagen , Losartán/farmacología , Osteoartritis/tratamiento farmacológico , Membrana Sinovial/metabolismo , Articulación Temporomandibular/diagnóstico por imagen , Factor de Crecimiento Transformador beta1/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Western Blotting , Cartílago Articular/metabolismo , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Osteoartritis/diagnóstico , Osteoartritis/metabolismo , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología
8.
J Pharmacol Sci ; 139(1): 9-14, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30472056

RESUMEN

Protocatechuic acid (3, 4-dihydroxybenzoic acid, PCA) is a major metabolite of anthocyanins and was reported to possess anti-allergic response. However, the effects of PCA on airway smooth muscle cells (ASMCs) proliferation and migration remain unclear. Therefore, this study aims to investigate the effects of PCA on proliferation and migration of ASMCs. ASMCs were pre-incubated with various concentrations of PCA for 30 min before stimulation with transforming growth factor-ß1 (TGF-ß1) for different times. Cell proliferation was determined using the colony formation assay. Cell migration was detected using the Transwell chamber assay. The levels of type I collagen, fibronectin, phosphorylated Smad2, Smad2, phosphorylated Smad3 and Smad3 were detected by western blot analysis. Our results demonstrated that PCA inhibited the proliferation and migration of ASMCs, as well as suppressed the expression levels of type I collagen and fibronectin in ASMCs induced by TGF-ß1. Furthermore, PCA obviously down-regulated the phosphorylation levels of Smad2/3 in ASMCs exposed to TGF-ß1. Taken together, the present results have revealed that PCA inhibits asthma airway remodeling by suppressing proliferation and extracellular matrix (ECM) protein deposition in TGF-ß1-mediated ASMCs via the inactivation of Smad2/3 signaling pathway. Therefore, PCA may be useful for the prevention or treatment of asthma airway remodeling.


Asunto(s)
Bronquios/citología , Hidroxibenzoatos/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias) , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Miocitos del Músculo Liso/fisiología , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1
9.
Cell Mol Biol (Noisy-le-grand) ; 65(7): 1-4, 2019 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-31880511

RESUMEN

All-trans retinoic acid (ATRA) is a critical component in cell processes such as cell growth, differentiation and apoptosis, and it is also crucial in the regulation of extracellular matrix (ECM) deposition. Prohibitin (PHB) can regulate cell proliferation, apoptosis and differentiation. The current study investigated whether ATRA regulated PHB is induced by hypoxia/reoxygenation injury in renal tubular epithelial cells (RTEC), using gene interference treatments (knockdown or overexpression of RARα). Our results indicate that ATRA can augment the expression of RARα and PHB proteins and reduce the expression of TGF-ß1, FN and Col-IV proteins. PHB expression was reduced in an ATRA treated RARα- group, and TGF-ß1, FN and Col-IV were up-regulated compared to the ATRA treated RARα+ group. We postulate that ATRA can induce the PHB expression by RARα in hypoxia/reperfusion related RTEC injury.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas Represoras/metabolismo , Receptor alfa de Ácido Retinoico/metabolismo , Animales , Antioxidantes/metabolismo , Western Blotting , Matriz Extracelular/metabolismo , Humanos , Oxidación-Reducción , Estrés Oxidativo/fisiología , Prohibitinas , ARN Mensajero/metabolismo , Ratas , Factor de Crecimiento Transformador beta1/metabolismo
10.
Exp Cell Res ; 370(1): 160-167, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29913155

RESUMEN

Peroxisome proliferator-activated receptor coactivator-1 alpha (PGC-1α) is a transcriptional coactivator that regulates energy metabolism and mitochondrial biogenesis. Recently, mitochondrial dysfunction has been indicated as an established risk factor for the development of renal fibrosis. However, whether PGC-1α is involved in the pathogenesis of renal fibrosis is unknown. In this study, we treated NRK-49F (normal rat kidney fibroblast) cells with transforming growth factor-beta 1 (TGF-ß1) for 24 h to establish an in vitro fibrosis model. TGF-ß1 induced the upregulation of type I collagen, fibronectin, TGF-ß receptor I (TGFß-RI), TGFß-RII, Smad4, and pSmad2/3, as well as PGC-1α. NRK-49F cells transfected with pcDNA-PGC-1α showed significantly increased expression of fibronectin and type I collagen, as revealed by western blot assay. Interestingly, transfection with PGC-1α-siRNA caused a stark reversal of TGF-ß1-induced cellular fibrosis, with concomitant suppression of fibronectin and type I collagen, as revealed by western blot and immunofluorescence assays. Moreover, SB431542 (TGFß-RI), LY294002 (PI3K/Akt), and SB203580 (p38 MAPK), inhibitors of TGF-ß-associated pathways, markedly suppressed TGF-ß1-induced PGC-1α upregulation. These results implicate a role of PGC-1α in renal interstitial fibrosis mediated via the TGFß-RI, PI3K/Akt, and p38 MAPK pathways. Our findings that PGC-1α-siRNA downregulates fibronectin and type I collagen suggest that it can be used as a novel molecular treatment for renal fibrosis.


Asunto(s)
Antifibrinolíticos/farmacología , Fibrosis/tratamiento farmacológico , Enfermedades Renales/tratamiento farmacológico , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , ARN Interferente Pequeño/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Línea Celular , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Fibrosis/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Enfermedades Renales/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Transfección/métodos , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Cell Tissue Res ; 372(1): 115-133, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29209813

RESUMEN

Extracellular matrix deposition during tubulointerstitial fibrosis (TIF), a central pathological process in patients with diabetic nephropathy (DN), is driven by locally activated, disease-relevant myofibroblasts. Myofibroblasts can arise from various cellular sources, e.g., tubular epithelial cells via a process named epithelial-to-mesenchymal transition (EMT). Transforming growth factor beta 1 (TGF-ß1) and its downstream Smad signaling play a critical role in both TIF and EMT. Whereas Smad3 is one central mediator, the role of the other prominently expressed variant, Smad2, is not completely understood. In this study, we sought to analyze the role of renal Smad2 in the development of TIF and EMT during streptozotocin-induced DN by using a fibroblast-specific protein 1 (FSP1)-promotor-driven SMAD2 knockout mouse model with decreased tubular, endothelial, and interstitial Smad2 expression. In contrast to wild-type diabetic mice, diabetic SMAD2 knockout mice showed the following features: (1) significantly reduced DN and TIF (shown by KIM1 expression; periodic acid Schiff staining; collagen I and III, fibronectin, and connective tissue growth factor deposition); (2) significantly reduced tubular EMT-like changes (e.g., altered Snail1, E-cadherin, matrix metalloproteinase 2, and vimentin deposition); and (3) significantly decreased expression of myofibroblast markers (α-smooth muscle actin, FSP1). As one mechanism for the protection against diabetes-induced TIF and EMT, decreased Smad3 protein levels and, as a possible consequence, reduced TGF-ß1 levels were observed in diabetic SMAD2 knockout mice. Our findings thus support the important role of Smad2 for pro-fibrotic TGF-ß/Smad3 signaling in experimental DN.


Asunto(s)
Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Túbulos Renales/patología , Proteína de Unión al Calcio S100A4/metabolismo , Proteína Smad2/metabolismo , Animales , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Fibrosis , Eliminación de Gen , Túbulos Renales/metabolismo , Ratones Noqueados , Estreptozocina , Factor de Crecimiento Transformador beta/metabolismo
12.
Exp Cell Res ; 355(2): 57-66, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28363829

RESUMEN

Signal transduction through the transforming growth factor-beta 1 (TGF-ß1) pathway affects epithelial to mesenchymal transition (EMT), partly by modulation of E-Cadherin expression. The concurrent impact of extracellular matrix driven regulation of integrin signaling on EMT has not been well characterized. We assessed the cumulative effect and molecular mechanisms of TGF-ß1 and integrin signal transduction on E-Cadherin in a renal cell cancer (RCC) model. Stimulation of RCC cells with TGF-ß1 demonstrated a three-fold increased expression of integrin αv. A ligand of integrin αv-ß3, (cyclopentapeptide containing Arginyl-Glycyl-Aspartic acid motif, RGD), was used to mimic integrin signaling. Treatment of cells with RGD and TGF-ß1 demonstrated significantly greater E-cadherin depletion than either ligand alone. This cooperative action on E-Cadherin expression is regulated by transcription factor Snai1 and is followed on a cellular level by increased cellular mobility as evidenced in a wound healing assay. Subsequent silencing of potential downstream mediators of the cumulative action of RGD and TGF-ß1 was carried out by small interfering RNA transfection and confirmed by Western blotting and/or RT-PCR. SiRNA mediated silencing of FAK and PINCH1 independently abrogated the cumulative effect of RGD and TGF-ß1 on E-Cadherin expression. We have identified a novel mechanism through which extracellular matrix event transduction by integrins further augments TGF-ß1 related effects on EMT. Molecular machinery involved in the integrin αv-TGF-ß1 interplay may represent a therapeutic target in RCC.


Asunto(s)
Cadherinas/metabolismo , Carcinoma de Células Renales/metabolismo , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Integrinas/metabolismo , Neoplasias Renales/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo/efectos de los fármacos , Humanos , Neoplasias Renales/patología , Oligopéptidos/farmacología
13.
Cell Biochem Funct ; 36(2): 46-55, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29336056

RESUMEN

Renal interstitial fibrosis is the most common process by which chronic kidney diseases progress to end-stage renal failure. Epithelial-to-mesenchymal transitions (EMTs) play a crucial role in the progression of renal interstitial fibrosis. A newly identified cytokine, interleukin-like EMT inducer (ILEI), has been linked to EMT in some diseases. However, the effects of ILEI on renal tubular EMT have not yet been well established. Here, we characterize the expression of ILEI in tubular EMT and describe the role and mechanism of ILEI in transforming growth factor beta 1 (TGF-ß1)-induced renal tubular EMT. The results indicate that ILEI is involved in renal tubular EMT induced by TGF-ß1, as overexpression of ILEI not only induces EMT of HK-2 cells independently but also profoundly enhances EMT in response to TGF-ß1. Supporting this finding, ILEI small interfering RNA was found to block the EMT of HK-2 cells induced by TGF-ß1. This work additionally suggests ILEI mediates TGF-ß1-dependent EMT via the extracellular regulated protein kinases (ERKs) and protein kinase B (Akt) signalling pathways. In conclusion, ILEI appears to play a crucial role in mediating TGF-ß1-induced EMT through the Akt and ERK pathways, which may provide a therapeutic target for the treatment of fibrotic kidney diseases. SIGNIFICANCE OF THE STUDY: There is no study reporting the effect of ILEI in renal EMTs. In this research, we examined the role and mechanism of ILEI in EMT using tubular epithelial cell; we found that ILEI participated in renal tubular EMT, and overexpression of ILEI can not only induce EMT of HK-2 cells independently but also enhance EMT in response to TGF-ß1. Meanwhile, we found ILEI small interfering RNA blocked the EMT induced by TGF-ß1, and ILEI participates in the EMT caused by TGF-ß1 via ERK and Akt signalling pathways. We hoped to provide new ideas in further study on the prevention and treatment of fibrotic kidney diseases.


Asunto(s)
Citocinas/metabolismo , Transición Epitelial-Mesenquimal , Túbulos Renales/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Células Cultivadas , Humanos
14.
BMC Cancer ; 17(1): 350, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28526008

RESUMEN

BACKGROUND: Urokinase plasminogen activator (uPA) receptor (uPAR) is up-regulated at the invasive tumour front of human oral squamous cell carcinoma (OSCC), indicating a role for uPAR in tumour progression. We previously observed elevated expression of uPAR at the tumour-stroma interface in a mouse model for OSCC, which was associated with increased proteolytic activity. The tumour microenvironment regulated uPAR expression, as well as its glycosylation and cleavage. Both full-length- and cleaved uPAR (uPAR (II-III)) are involved in highly regulated processes such as cell signalling, proliferation, migration, stem cell mobilization and invasion. The aim of the current study was to analyse tumour associated factors and their effect on uPAR cleavage, and the potential implications for cell proliferation, migration and invasion. METHODS: Mouse uPAR was stably overexpressed in the mouse OSCC cell line AT84. The ratio of full-length versus cleaved uPAR as analysed by Western blotting and its regulation was assessed by addition of different protease inhibitors and transforming growth factor - ß1 (TGF-ß1). The role of uPAR cleavage in cell proliferation and migration was analysed using real-time cell analysis and invasion was assessed using the myoma invasion model. RESULTS: We found that when uPAR was overexpressed a proportion of the receptor was cleaved, thus the cells presented both full-length uPAR and uPAR (II-III). Cleavage was mainly performed by serine proteases and urokinase plasminogen activator (uPA) in particular. When the OSCC cells were stimulated with TGF-ß1, the production of the uPA inhibitor PAI-1 was increased, resulting in a reduction of uPAR cleavage. By inhibiting cleavage of uPAR, cell migration was reduced, and by inhibiting uPA activity, invasion was reduced. We could also show that medium containing soluble uPAR (suPAR), and cleaved soluble uPAR (suPAR (II-III)), induced migration in OSCC cells with low endogenous levels of uPAR. CONCLUSIONS: These results show that soluble factors in the tumour microenvironment, such as TGF-ß1, PAI-1 and uPA, can influence the ratio of full length and uPAR (II-III) and thereby potentially effect cell migration and invasion. Resolving how uPAR cleavage is controlled is therefore vital for understanding how OSCC progresses and potentially provides new targets for therapy.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias de la Boca/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Factor de Crecimiento Transformador beta1/genética , Animales , Carcinoma de Células Escamosas/patología , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias de la Boca/patología , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Transducción de Señal , Microambiente Tumoral/genética
15.
Bioorg Med Chem Lett ; 27(4): 1081-1088, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28110870

RESUMEN

The epithelial-mesenchymal transition (EMT) is an important cellular process during which polarized epithelial cells become motile mesenchymal cells, which promote cancer metastasis. Ginger, the rhizome of Zingiber officinale, is extensively used in cooking worldwide and also as a traditional medicinal herb with antioxidant, anti-inflammatory and anticancer properties. Several pungent compounds have been identified in ginger, including zingerone, which has anticancer potential. However, the role of zingerone in EMT is unclear. We investigated the synergistic effect of zingerone and its derivative on EMT. Transforming growth factor-beta 1 (TGF-ß1) induces the EMT to promote hepatocellular carcinoma metastasis, including migration and invasion. To understand the repressive role of the combination of zingerone and its derivative (ZD 2) in hepatocellular carcinoma metastasis, we investigated the potential use of each compound of ginger, such as zingerone, ZD 2 and 6-shogaol, or the mixture of zingerone and ZD 2 (ZD 2-1) as inhibitors of TGF-ß1 induced EMT development in SNU182 hepatocellular carcinoma cells in vitro. We show that ZD 2-1, but not zingerone, ZD 2 and 6-shogaol significantly increased expression of the epithelial marker E-cadherin and repressed Snail upregulation and expression of the mesenchymal marker N-cadherin during initiation of the TGF-ß1 induced EMT. In addition, ZD 2-1 inhibited the TGF-ß1 induced increase in cell migration and invasion of SNU182 hepatocellular carcinoma cells. Furthermore, ZD 2-1 significantly inhibited TGF-ß1 regulated matrix metalloproteinase-2/9 and activation of Smad2/3. We also found that ZD 2-1 inhibited nuclear translocation of NF-κB, activation of p42/44 MAPK/AP1 signaling pathway in the TGF-ß1 induced EMT. Our findings provide new evidence that combined treatment with ZD 2, novel zingerone derivative, and zingerone synergistically suppresses hepatocellular carcinoma metastasis in vitro by inhibiting the TGF-ß1 induced EMT.


Asunto(s)
Carcinoma Hepatocelular/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Guayacol/análogos & derivados , Neoplasias Hepáticas/patología , Invasividad Neoplásica/prevención & control , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Línea Celular Tumoral , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal/fisiología , Guayacol/química , Guayacol/farmacología , Humanos , Factor de Crecimiento Transformador beta1/fisiología
16.
Bioorg Med Chem Lett ; 27(15): 3342-3348, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28610976

RESUMEN

Epithelial-to-mesenchymal transition (EMT), an important cellular process, occurs during cancer development and progression, has a crucial role in metastasis by enhancing the motility of tumor cells. Dioscin is a polyphenolic component isolated from Phyllanthus amarus, which exhibits a wide range of pharmacological and physiological activities, such as anti-tumor, anti-inflammatory, anti-obesity, anti-fungal, and anti-viral activities. However, the possible role of dioscin in the EMT is unclear. We investigated the suppressive effect of dioscin on the EMT. Transforming growth factor-beta 1 (TGF-ß1) is known to induce EMT in a number of cancer cell types and promote lung adenocarcinoma migration and invasion. To verify the inhibitory role of dioscin in lung cancer migration and invasion, we investigated the use of dioscin as inhibitors of TGF-ß1-induced EMT in A549 lung cancer cells in vitro. Here, we found that dioscin prominently increased expression of the epithelial marker E-cadherin and expression of the mesenchymal marker N-cadherin and Snail during the TGF-ß1-induced EMT. In addition, dioscin inhibited the TGF-ß1-induced increase in cell migration and invasion of A549 lung cancer cells. Also, dioscin remarkably inhibited TGF-ß1-regulated activation of MMP-2/9, Smad2, and p38. Taken together, our findings provide new evidence that dioscin suppresses lung cancer migration, and invasion in vitro by inhibiting the TGF-ß1-induced EMT.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Diosgenina/análogos & derivados , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diosgenina/síntesis química , Diosgenina/química , Diosgenina/farmacología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias Pulmonares/patología , Estructura Molecular , Relación Estructura-Actividad , Factor de Crecimiento Transformador beta1/farmacología
17.
Bioorg Med Chem Lett ; 25(23): 5508-13, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26508552

RESUMEN

In the epithelial-mesenchymal transition (EMT), an important cellular process, epithelial cells become mesenchymal cells. This process is also critically involved in cancer metastasis. Sanguiin H6 is a compound derived from ellagitannin, which is found in berries. Sanguiin H6 shows various pharmacological properties, including anti-angiogenic activity. Because the possible role of sanguiin H6 in the EMT and the underlying molecular mechanisms are unclear, we investigated the effect of sanguiin H6 on the EMT. Transforming growth factor-beta 1 (TGF-ß1) induces the EMT and promotes lung adenocarcinoma migration and invasion through the Smad2/3 signaling pathway. Thus, to understand the inhibitory effects of sanguiin H6 on lung cancer migration and invasion, we investigated the ability of sanguiin H6 to inhibit TGF-ß1-induced EMT in the A549 cell line. We found that sanguiin H6 significantly prevented the activation of Smad2/3 signaling pathway by TGF-ß1. Additionally, sanguiin H6 increased the expression of the epithelial marker E-cadherin and repressed the expression of Snail and the mesenchymal marker N-cadherin during TGF-ß1-induced EMT. Moreover, sanguiin H6 regulated the expression of EMT-dependent genes induced by TGF-ß1. Finally, sanguiin H6 inhibited the migration and invasion of TGF-ß1-stimulated A549 cells. Taken together, our findings provide new evidence that sanguiin H6 suppresses lung cancer migration and invasion in vitro by inhibiting TGF-ß1 induction of the EMT.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Taninos Hidrolizables/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Factor de Crecimiento Transformador beta1/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Humanos , Estructura Molecular , Invasividad Neoplásica
18.
Bioorg Med Chem Lett ; 25(17): 3529-34, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26169124

RESUMEN

The epithelial-mesenchymal transition (EMT) is an important cellular process during which epithelial polarized cells become motile mesenchymal-appeared cells, which, in turn, induces the metastatic of cancer. Geraniin is a polyphenolic component isolated from Phyllanthus amarus, which exhibits a wide range of pharmacological and physiological activities, such as antitumor, anti-hyperglycemic, anti-hypertensive, antimicrobial, and antiviral activities. However, the possible role of geraniin in the EMT is unclear. We investigated the effect of geraniin on the EMT. Transforming growth factor-beta 1 (TGF-ß1) induces the EMT to promote lung adenocarcinoma migration, invasion, and anoikis resistance. To understand the suppressive role of geraniin in lung cancer migration, invasion, and anoikis resistance, we investigated the use of geraniin as inhibitors of TGF-ß1-induced EMT in A549 lung cancer cells in vitro. Here, we show that geraniin remarkably increased expression of the epithelial marker E-cadherin and repressed Snail upregulation and expression of the mesenchymal marker N-cadherin and vimentin during the TGF-ß1-induced EMT. Geraniin also inhibited the TGF-ß1-induced increase in cell migration, invasion, and anoikis resistance of A549 lung cancer cells. Additionally, geraniin markedly inhibited TGF-ß1-regulated activation of Smad2. Taken together, our findings provide new evidence that geraniin suppresses lung cancer migration, invasion, and anoikis resistance in vitro by inhibiting the TGF-ß1-induced EMT.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anoicis/efectos de los fármacos , Antineoplásicos Fitogénicos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucósidos/farmacología , Taninos Hidrolizables/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Pulmón/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Humanos , Pulmón/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Invasividad Neoplásica/patología , Phyllanthus/química
19.
Clin Exp Pharmacol Physiol ; 42(6): 680-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25882815

RESUMEN

Krüppel-like factor 4 (KLF4) is known to mitigate inflammation in several cell types. Using human proximal tubule cells, the present study aimed to investigate the role of KLF4 in regulating transforming growth factor (TGF)-ß1 induced inflammatory and fibrotic responses. Human kidney proximal tubular cells were exposed to high glucose, or TGF-ß1 and KLF4 expressions were determined. Cells were then transfected with empty vector or KLF4 and exposed to 2-ng/mL TGF-ß1 for up to 72 h. Inflammatory proteins (macrophage migration inhibitory factor and monocyte chemoattractant protein-1) and pro-fibrotic proteins (fibronectin and collagen IV) were measured after 72 h by enzyme-linked immunosorbent assay and western blot, respectively. To determine the relevance to in vivo models of chronic kidney disease, KLF4 protein expression in streptozotocin-induced diabetic mice was determined. Krüppel-like factor 4 messenger RNA (mRNA) levels were significantly reduced in high glucose-treated human kidney proximal tubular cells. High glucose increased TGF-ß1 mRNA expression, which significantly increased migration inhibitory factor and monocyte chemoattractant protein-1 protein secretion. Transforming growth factor-ß1 significantly increased fibronectin and collagen IV protein expression. The overexpression of KLF4 significantly reduced TGF-ß-mediated increases in migration inhibitory factor and monocyte chemoattractant protein-1 but had no effect on TGF-ß-mediated fibronectin and collagen IV mRNA and protein expression. The levels of KLF4 mRNA were significantly reduced in the diabetic kidney, and diabetic animals had a significant reduction in renal tubular expression of KLF4 proteins. This data suggest that KLF4 reduces inflammation induced by TGF-ß1, suggesting a therapeutic role for KLF4 in diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Túbulos Renales Proximales/metabolismo , Factores de Transcripción de Tipo Kruppel/fisiología , Factor de Crecimiento Transformador beta1/toxicidad , Animales , Células Cultivadas , Diabetes Mellitus Experimental/patología , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Mediadores de Inflamación/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Factor 4 Similar a Kruppel , Ratones , Ratones Noqueados
20.
J Med Virol ; 86(8): 1366-76, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24610501

RESUMEN

Hepatitis E virus (HEV) infection is associated with high maternal and fetal mortalities. The aim of the study was to find cytokine gene polymorphisms in relation to HEV infection during pregnancy. A total of 262 pregnant and 208 non-pregnant women with hepatitis, 262 healthy pregnant and 208 non-pregnant women as controls. The study group were pregnant and non-pregnant women with HEV infection, not infected with HEV and controls. Genotyping was carried out by PCR-RFLP and ARMS-PCR methods. The frequencies of TNF-α -308 A allele & AA genotype, IFN-γ +874 T allele & TT genotypes were significantly higher in pregnant women with HEV infection compared to other groups. The frequency of TGF-ß1 codon 10 +869 T allele &TT genotype and codon 25 +915 G allele & GG genotype were significantly higher in pregnant women compared to non-pregnant women with HEV infection. The frequency of IL-6-174 GG genotype was significantly higher in pregnant women with HEV infection compared to not infected with HEV and controls. Cytokine gene polymorphisms shows association with preterm delivery (TNF-α -308 AA, IFN-γ +874 AA, TGF-ß1 codon 10 +869 TT & codon 25 GG genotypes), low birth weight (TNF-α -308 GG & IL-6 -174 CC genotypes), fetal loss (IL-6-174 CC genotype), and small for date (IL-6-174 CC & TGF-ß1 codon 10 +869 TC genotypes) of HEV infected pregnant women compared to not infected with HEV and controls. These findings suggest that cytokines gene polymorphisms were found to be associated with pregnant women with HEV infection and adverse pregnancy outcome.


Asunto(s)
Citocinas/genética , Hepatitis E/genética , Hepatitis E/inmunología , Polimorfismo de Nucleótido Simple , Complicaciones Infecciosas del Embarazo/genética , Complicaciones Infecciosas del Embarazo/inmunología , Resultado del Embarazo , Adolescente , Adulto , Codón , Femenino , Frecuencia de los Genes , Técnicas de Genotipaje , Humanos , Recién Nacido , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Embarazo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA