Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
BMC Cardiovasc Disord ; 22(1): 17, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35081907

RESUMEN

BACKGROUND: The aim of this study was to investigate the effects of Resveratrol (RSV) in rats with dilated cardiomyopathy (DCM). METHODS: Porcine cardiac myosin was used to set up rat model with DCM. RSV (10 mg/kg in RSV-L group and 50 mg/kg in RSV-H group) or vehicle was administered to rats with DCM once daily from the 28th day till the 90th day after the first immunization. Cardiac function of rats was evaluated by echocardiographic analysis. The deposition of fibrous tissues in the hearts was evaluated by Masson and picrosirius red staining. The mRNA levels of collagen type I (Col I), collagen type III (Col III) and silence information regulator 1 (Sirt1) were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The interaction of Sirt1 with Smad3 was revealed by coimmunoprecipitation. RESULTS: The heart weight, heart weight/body weight ratio, left ventricular end diastolic diameter (LVEDD) and left ventricular end systolic diameter (LVESD) were significantly increased in rats with DCM, and attenuated by RSV. RSV also positively decreased fibrosis, and the expression of Col I and Col III in the myocardium. The Sirt1 mRNA was significantly decreased in myosin-immunized hearts and was positively increased by RSV. The Sirt1 combined with Smad3 directly. Acetylation of Smad3 (Ac-Smad3) was significantly increased in DCM and was markedly decreased by RSV. CONCLUSION: RSV effectively ameliorated myocardial fibrosis and improved cardiac function by regulating Sirt1/Smad3 deacetylation pathway in rat model with DCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Regulación de la Expresión Génica , Miocardio/patología , ARN/genética , Resveratrol/farmacología , Sirtuina 1/genética , Proteína smad3/genética , Animales , Biopsia , Cardiomiopatía Dilatada/diagnóstico , Cardiomiopatía Dilatada/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Inhibidores Enzimáticos/farmacología , Fibrosis/diagnóstico , Fibrosis/prevención & control , Masculino , Sirtuina 1/biosíntesis , Proteína smad3/biosíntesis , Porcinos
2.
Am J Physiol Cell Physiol ; 322(1): C63-C72, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34852209

RESUMEN

Pulmonary fibrosis is a chronic, progressive, and irreversible interstitial lung disease. Transforming growth factor-ß1 (TGF-ß1) plays a major role in lung fibroblast cell differentiation to myofibroblast cells and production of extracellular matrix, which are hallmarks of pulmonary fibrosis. G protein-coupled receptor kinase-2 (GRK2) has been shown to play controversial roles in TGF-ß1-induced signal transduction in different cell types; however, the role of GRK2 in TGF-ß1-induced activation of lung fibroblast cells and development of pulmonary fibrosis has not been revealed. In this study, we found that GRK2 levels were increased in lungs and isolated fibroblast cells in a murine model of pulmonary fibrosis, as well as TGF-ß1-treated lung fibroblasts. GRK2 levels were not changed in lungs in the injury phase of pulmonary fibrosis. Posttreatment with GRK2 inhibitor reduced extracellular matrix (ECM) accumulation in lungs in bleomycin-challenged mice, suggesting that GRK2 activation contributes to the progressive phase of pulmonary fibrosis. Inhibition or downregulation of GRK2 attenuates fibronectin, collagen, and α-smooth muscle actin expression in TGF-ß1-induced lung fibroblast cells or myofibroblast cells isolated from patients with pulmonary fibrosis. Furthermore, we showed that GRK2 regulates Smad3 expression, indicating that inhibition of GRK2 attenuates ECM accumulation through downregulation of Smad3 expression. This study reveals that GRK2 is a therapeutic target in treating pulmonary fibrosis and inhibition of GRK2 dampens pulmonary fibrosis by suppression of Smad3 expression, eventually attenuating TGF-ß1 signal pathway and ECM accumulation.


Asunto(s)
Fibroblastos/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/biosíntesis , Pulmón/metabolismo , Fibrosis Pulmonar/metabolismo , Proteína smad3/biosíntesis , Animales , Bleomicina/toxicidad , Línea Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Expresión Génica , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Proteína smad3/antagonistas & inhibidores , Proteína smad3/genética
3.
Immunopharmacol Immunotoxicol ; 43(4): 461-470, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34142927

RESUMEN

AIM: High-mobility group box 1 (HMGB1) protein has been noticed particularly for its pivotal role in several pathologies. However, the relevance between HMGB1 and pathological progress in lung toxicity still remains unclear. In the study, we evaluated the effect of glycyrrhizic acid as an HMGB1 inhibitor on the early inflammation and late fibrosis in bleomycin-induced pulmonary toxicity in mice. METHODS: We established a bleomycin-induced pulmonary toxicity model to detect the relevance between HMGB1 and pathological changes in the early inflammatory and late fibrotic stages. RESULTS: We found that bleomycin-induced increase in inflammatory cytokines interleukin (IL)-ß1, tumor necrosis factor (TNF)-α, monocyte chemotactic protein (MCP)-1, and inflammatory lesions in lung tissue in the early stage of the model. However, markers of fibrosis such as transforming growth factor (TGF)-ß1 and α-smooth muscle actin (α-SMA) were significantly elevated on day 7 after bleomycin instillation. Interestingly, HMGB1 also began to rise on day 7, rather than in the early inflammatory phase. However, early (from day 0 to 14 after bleomycin instillation) or late (from day 14 to 28) intervention with HMGB1 neutralizing antibody or glycyrrhizic acid alleviated inflammation and fibrosis through down-regulating the inflammatory signaling mitogen-activated protein kinase (MAPK) and fibrotic signaling Smad3 pathway. CONCLUSION: Our results suggested that HMGB1 mediates both inflammation and fibrosis in this model. The development of high-potency and low-toxicity HMGB1 inhibitors may be a class of potential drugs for the treatment of pulmonary fibrosis.


Asunto(s)
Bleomicina/toxicidad , Ácido Glicirrínico/farmacología , Proteína HMGB1/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fibrosis Pulmonar/prevención & control , Proteína smad3/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Femenino , Ácido Glicirrínico/uso terapéutico , Proteína HMGB1/biosíntesis , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Proteína smad3/biosíntesis
4.
Dig Dis Sci ; 66(6): 1862-1874, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32705438

RESUMEN

BACKGROUND: There is little known about stem cells in human non-neoplastic and neoplastic esophageal epithelia. We have demonstrated expression of linker threonine-phosphorylated Smad2/3 (pSmad2/3L-Thr), suggesting presence of stem-like cells in mouse esophageal epithelium, and identified presence of pSmad2/3L-Thr-positive cells that might function as cancer stem cells in mouse model of colorectal carcinoma. AIMS: We explore whether pSmad2/3L-Thr can be used as a biomarker for stem cells of human esophageal epithelia and/or neoplasms. METHODS: We have used esophageal tissues from inpatients undergoing endoscopic submucosal dissection and performed double immunofluorescent staining of pSmad2/3L-Thr and Ki67, CDK4, p63, Sox2, CK14, p53, ALDH1, CD44 or D2-40 after which the sections were stained with hematoxylin and eosin. RESULTS: pSmad2/3L-Thr-positive cells showed immunohistochemical co-localization with CDK4, p63, CD44 and Sox2 in the basal and parabasal layers of non-neoplastic esophageal epithelia. In esophageal neoplasms, they showed immunohistochemical co-localization with p53, CDK4, ALDH1 and CD44. There was a significant increase in the percentage of pSmad2/3L-Thr-positive cells in the p53-positive neoplastic cell population with development of esophageal neoplasia. pSmad2/3L-Thr-positive cells localized to the lower section of low-grade intraepithelial neoplasia and were observed up to the upper section in carcinoma in situ. In invasive squamous cell carcinoma, they were scattered throughout the tumor with disappearance of polarity and were found in intraepithelial primary lesions and sites of submucosal and vessel invasion. CONCLUSIONS: We determined significant expression of pSmad2/3L-Thr in human esophageal non-neoplastic and neoplastic epithelia, indicating that these are epithelial stem-like cells and cancer stem cells, respectively, that correlate with developing esophageal neoplasms.


Asunto(s)
Mucosa Esofágica/metabolismo , Neoplasias Esofágicas/metabolismo , Células Madre Neoplásicas/metabolismo , Proteína Smad2/biosíntesis , Proteína smad3/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Mucosa Esofágica/patología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Fosforilación/fisiología , Proteína Smad2/genética , Proteína smad3/genética
5.
J Orthop Surg Res ; 15(1): 173, 2020 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-32410637

RESUMEN

BACKGROUND: It is reported that miR-596 has a potential diagnostic value for non-traumatic osteonecrosis of the femoral head (NOFH), but its underlying mechanisms in NOFH is unclear. METHODS: The expression of miR-596 and Smad3 was detected by western blot and quantitative real-time PCR. The relationship between the two molecules was explored using Dual-Luciferase Reporter Assay. Glucocorticoid (GC)-dexamethasone, was used to induce bone marrow mesenchymal stem cell (BMSC) osteogenic differentiation, and the effects of miR-596 on BMSC osteogenic differentiation and proliferation were determined. RESULTS: MiR-596 expression was upregulated, while Smad3 expression was inhibited in the bone marrow samples of patients with steroid-induced osteonecrosis of femoral head (SANFH). Overexpression of miR-596 inhibited the proliferation and osteogenic differentiation of BMSCs induced by GC. Meanwhile, the opposite results were observed in the miR-596 inhibitor group. In addition, Smad3 was a target gene of miR-596, and negatively regulated by miR-596. The promotion effect of the miR-596 inhibitor on BMSC proliferation and osteogenic differentiation was reversed by si-Smad3. CONCLUSION: MiR-596 can suppress GC-BMSC osteoblastic differentiation and proliferation by regulating Smad3 expression.


Asunto(s)
Diferenciación Celular/fisiología , Necrosis de la Cabeza Femoral/metabolismo , MicroARNs/biosíntesis , Osteoblastos/metabolismo , Proteína smad3/biosíntesis , Esteroides/efectos adversos , Adulto , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Necrosis de la Cabeza Femoral/inducido químicamente , Necrosis de la Cabeza Femoral/genética , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Proteína smad3/antagonistas & inhibidores , Proteína smad3/genética
6.
J Hepatol ; 73(4): 882-895, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32330605

RESUMEN

BACKGROUND & AIMS: Cytoglobin (CYGB) is a respiratory protein that acts as a scavenger of reactive oxygen species. The molecular role of CYGB in human hepatic stellate cell (HSC) activation and human liver disease remains uncharacterised. The aim of this study was to reveal the mechanism by which the TGF-ß1/SMAD2 pathway regulates the human CYGB promoter and the pathophysiological function of CYGB in human non-alcoholic steatohepatitis (NASH). METHODS: Immunohistochemical staining was performed using human NASH biopsy specimens. Molecular and biochemical analyses were performed by western blotting, quantitative PCR, and luciferase and immunoprecipitation assays. Hydroxyl radicals (•OH) and oxidative DNA damage were measured using an •OH-detectable probe and 8-hydroxy-2'-deoxyguanosine (8-OHdG) ELISA. RESULTS: In culture, TGF-ß1-pretreated human HSCs exhibited lower CYGB levels - together with increased NADPH oxidase 4 (NOX4) expression - and were primed for H2O2-triggered •OH production and 8-OHdG generation; overexpression of human CYGB in human HSCs reversed these effects. Electron spin resonance demonstrated the direct •OH scavenging activity of recombinant human CYGB. Mechanistically, pSMAD2 reduced CYGB transcription by recruiting the M1 repressor isoform of SP3 to the human CYGB promoter at nucleotide positions +2-+13 from the transcription start site. The same repression did not occur on the mouse Cygb promoter. TGF-ß1/SMAD3 mediated αSMA and collagen expression. Consistent with observations in cultured human HSCs, CYGB expression was negligible, but 8-OHdG was abundant, in activated αSMA+pSMAD2+- and αSMA+NOX4+-positive hepatic stellate cells from patients with NASH and advanced fibrosis. CONCLUSIONS: Downregulation of CYGB by the TGF-ß1/pSMAD2/SP3-M1 pathway brings about •OH-dependent oxidative DNA damage in activated hepatic stellate cells from patients with NASH. LAY SUMMARY: Cytoglobin (CYGB) is a respiratory protein that acts as a scavenger of reactive oxygen species and protects cells from oxidative DNA damage. Herein, we show that the cytokine TGF-ß1 downregulates human CYGB expression. This leads to oxidative DNA damage in activated hepatic stellate cells. Our findings provide new insights into the relationship between CYGB expression and the pathophysiology of fibrosis in patients with non-alcoholic steatohepatitis.


Asunto(s)
Citoglobina/genética , Regulación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , NADPH Oxidasa 4/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/metabolismo , Biopsia , Células Cultivadas , Citoglobina/biosíntesis , Regulación hacia Abajo , Femenino , Células Estrelladas Hepáticas/patología , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Persona de Mediana Edad , NADPH Oxidasa 4/biosíntesis , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo/genética , Proteína smad3/biosíntesis
7.
Spine (Phila Pa 1976) ; 45(17): E1057-E1065, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32205703

RESUMEN

STUDY DESIGN: Experimental analysis of the thoracic ligamentum flavum cell osteogenic differentiation process. OBJECTIVE: This study aimed to explore the role of miR-29a-5p and special AT-rich sequence-binding protein 2 (SATB2) in a pathological osteogenic process. SUMMARY OF BACKGROUND DATA: Thoracic ossification of the ligamentum flavum (TOLF) is an uncommon disease wherein ligaments within the spine undergo progressive ossification, resulting in stenosis of the spinal canal and myelopathy. MiR-29a-5p was found to be downregulated in ligament cells from ossified ligament tissue in a previous study. However, whether miR-29a-5p is involved in the process of TOLF has not been investigated. METHODS: The expression of miR-29a-5p in ligament tissues or in the context of TOLF osteogenic cell differentiation was measured via qRT-PCR. Alkaline phosphatase activity assay and Alizarin red staining were used to analyze cellular osteogenesis. The protein-level expression of SATB2, SIRT1, and Smad3 were measured via immunohistochemistry or western blotting. Dual luciferase reporter assays and western blotting were used to confirm that miR-29a targets SATB2. RESULTS: SATB2 was found to be upregulated and miR-29a-5p was downregulated in TOLF tissue. We additionally observed decreased miR-29a-5p expression during the process of TOLF osteogenic cell differentiation, and there was a marked reduction in the expression of key mediators of osteogenesis when miR-29a-5p was overexpressed. Consistent with this, when miR-29a-5p was inhibited this led to enhanced osteogenic cell differentiation of these cells. We further found miR-29a-5p to directly target and suppress the expression of SATB2. Knock-down of SATB2 was sufficient to reduce the ability of miR-29a-5p to inhibit osteogenesis, and this also led to decreased SIRT1 expression and Smad3 acetylation. CONCLUSION: Together our findings indicate that miR-29a-5p is able to prevent thoracic ligamentum flavum cell osteogenesis at least in part via targeting SATB2 and thereby suppressing the SIRT1/Smad3 deacetylation pathway. LEVEL OF EVIDENCE: N/A.


Asunto(s)
Ligamento Amarillo/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/biosíntesis , MicroARNs/biosíntesis , Osteogénesis/fisiología , Sirtuina 1/biosíntesis , Proteína smad3/biosíntesis , Factores de Transcripción/biosíntesis , Acetilación , Adulto , Anciano , Células Cultivadas , Femenino , Células HEK293 , Humanos , Ligamento Amarillo/patología , Masculino , Proteínas de Unión a la Región de Fijación a la Matriz/antagonistas & inhibidores , Persona de Mediana Edad , Transducción de Señal/fisiología , Sirtuina 1/antagonistas & inhibidores , Proteína smad3/antagonistas & inhibidores , Vértebras Torácicas/metabolismo , Vértebras Torácicas/patología , Factores de Transcripción/antagonistas & inhibidores
8.
Drug Dev Res ; 80(7): 992-999, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31403228

RESUMEN

Biliary epithelial cells (BECs) can secrete bile and the epithelial-to-mesenchymal transition (EMT) of BECs can cause fibrosis or damage interlobular bile ducts, leading to chronic cholangiopathies, such as primary biliary cholangitis (PBC). Transforming growth factor-ß1 (TGF-ß1) is a potent inducer of the EMT while curcumin, a diarylheptanoid, can inhibit the EMT of hepatocytes in many liver diseases. However, the protection and underlying mechanisms of curcumin against the EMT of BECs have not been clarified. Herein, we show that curcumin treatment significantly mitigates the EMT of BECs in vitro and in vivo. Mechanistically, curcumin significantly attenuated the TGF-ß1-induced Smad and Hedgehog signaling, and upregulated CD109 expression in BECs. Collectively, these findings highlighted the therapeutic potential of curcumin to counteract the EMT process in PBC.


Asunto(s)
Antígenos CD/biosíntesis , Curcumina/farmacología , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteínas de Neoplasias/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Proteínas Ligadas a GPI/biosíntesis , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Proteína Smad2/biosíntesis , Proteína smad3/biosíntesis , Proteína smad7/biosíntesis , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/farmacología
9.
Respir Res ; 20(1): 163, 2019 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-31331325

RESUMEN

BACKGROUND: Pulmonary fibrosis is a progressive and irreversible disease for which therapeutic options are currently limited. A recent in vivo study showed that tenofovir, a nucleotide analogue reverse transcriptase inhibitor, had direct antifibrotic effects on skin and liver fibrosis. Another study in vitro revealed that NS5ATP9 inhibited the activation of human hepatic stellate cells. Because of the similarity of fibrotic diseases, we hypothesized that tenofovir alafenamide fumarate (TAF), the prodrug of tenofovir, and NS5ATP9, is related to and plays a role in the suppression of pulmonary fibrosis. METHODS: We investigated the influence of NS5ATP9 on fibrosis in vitro. Human lung fibroblasts (HFL1) were transfected with short interfering RNAs or overexpression plasmids of NS5ATP9 before stimulation by human recombinant transforming growth factor-ß1. The effect of TAF was evaluated in a bleomycin-induced fibrosis murine model. Male C57BL/6 mice were treated with bleomycin on day 0 by intratracheal injection and intragastrically administered TAF or vehicle. Left lung sections were fixed for histological analysis, while homogenates of the right lung sections and HFL1 cells were analyzed by western blotting and quantitative reverse transcription polymerase chain reaction. RESULTS: NS5ATP9 suppressed the activation of lung fibroblasts. Upregulation of collagen type 3 (α 1 chain) and α-smooth muscle actin was observed in HFL1 cells when NS5ATP9 was silenced, and vice-versa. TAF also showed anti-fibrotic effects in mice, as demonstrated by histological analysis of fibrosis and expression of extracellular matrix components in the lung sections. Additionally, TAF inhibited transforming growth factor-ß1 and phosphorylated-Smad3 synthesis in HFL1 cells and the murine model, which was accompanied by upregulation of NS5ATP9. CONCLUSIONS: Our results suggest that NS5ATP9 forms a negative feedback pathway in pulmonary fibrosis and TAF has anti-fibrotic properties as it upregulates the expression level of NS5ATP9. As TAF has been shown to be safe and well-tolerated in humans, TAF and NS5ATP9 may be useful for developing novel therapeutics for pulmonary fibrosis.


Asunto(s)
Adenina/análogos & derivados , Bleomicina/toxicidad , Proteínas de Unión al ADN/biosíntesis , Fibrosis Pulmonar/metabolismo , Proteína smad3/biosíntesis , Factor de Crecimiento Transformador beta1/biosíntesis , Adenina/farmacología , Adenina/uso terapéutico , Alanina , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/prevención & control , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tenofovir/análogos & derivados , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
10.
J Biochem Mol Toxicol ; 33(5): e22301, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30801894

RESUMEN

Schizandrin is a major bioactive constituent of Schisandra chinensis (Turcz.) Baill with antioxidant and anti-inflammatory properties. The objective of this study was to explore the potential effects of schizandrin on a cell model of myocarditis. The H9c2 cells were treated with schizandrin alone or in combination with lipopolysaccharide (LPS), after which, cell survival, migration, and the release of inflammatory cytokines were assessed. Moreover, downstream effectors and signaling pathways were studied to reveal the possible underlying mechanism. As a result, LPS stimulation induced significant cell damage as cell viability was repressed and the apoptosis was induced. In the meantime, LPS promoted the release of proinflammatory cytokines including interleukin 1ß (IL-1ß), IL-8, IL-6, and tumor necrosis factor (TNF-α) while repressing the release of the anti-inflammatory cytokine IL-10. Schizandrin could promote H9c2 cell migration and long-term treatment (7 days) enhanced cell viability. More interestingly, pretreatment with schizandrin attenuated LPS-induced cell loss and inflammatory response. Besides this, Smad3 was a downstream effector of schizandrin. The beneficial effects of schizandrin on the H9c2 cells were attenuated when Smad3 was overexpressed. Moreover, the silencing of Smad3 deactivated c-Jun N-terminal kinase (JNK) and nuclear factor κB (NF-κB) pathways. This study preliminarily demonstrated that schizandrin prevented LPS-induced injury in the H9c2 cells and promoted the recovery of myocardial tissues by enhancing cell viability and migration. Schizandrin conferred its beneficial effects possibly by downregulating Smad3 and inhibiting the activation of JNK and NF-κB pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclooctanos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Lignanos/farmacología , Lipopolisacáridos/toxicidad , Mioblastos Cardíacos/metabolismo , Compuestos Policíclicos/farmacología , Proteína smad3/biosíntesis , Supervivencia Celular/efectos de los fármacos , Ciclooctanos/química , Citocinas/biosíntesis , Humanos , Lignanos/química , MAP Quinasa Quinasa 4/metabolismo , Mioblastos Cardíacos/patología , FN-kappa B/metabolismo , Compuestos Policíclicos/química , Schisandra/química , Transducción de Señal/efectos de los fármacos
11.
FASEB J ; 33(5): 6667-6681, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30779601

RESUMEN

Cell differentiation is directed by extracellular cues and intrinsic epigenetic modifications, which control chromatin organization and transcriptional activation. Central to this process is PRC2, which modulates the di- and trimethylation of lysine 27 on histone 3; however, little is known concerning the direction of PRC2 to specific loci. Here, we have investigated the physical interactome of EZH2, the enzymatic core of PRC2, during retinoic acid-mediated differentiation of neuroepithelial, pluripotent NT2 cells and the dedifferentiation of neuroretinal epithelial ARPE19 cells in response to TGF-ß. We identified Smad3 as an EZH2 interactor in both contexts. Co-occupation of the CDH1 promoter by Smad3 and EZH2 and the cooperative, functional nature of the interaction were established. We propose that the interaction between Smad3 and EZH2 targets the core polycomb assembly to defined regions of the genome to regulate transcriptional repression and forms a molecular switch that controls promoter access through epigenetic mechanisms leading to gene silencing.-Andrews, D., Oliviero, G., De Chiara, L., Watson, A., Rochford, E., Wynne, K., Kennedy, C., Clerkin, S., Doyle, B., Godson, C., Connell, P., O'Brien, C., Cagney, G., Crean, J. Unravelling the transcriptional responses of TGF-ß: Smad3 and EZH2 constitute a regulatory switch that controls neuroretinal epithelial cell fate specification.


Asunto(s)
Diferenciación Celular , Proteína Potenciadora del Homólogo Zeste 2/biosíntesis , Células Epiteliales/metabolismo , Silenciador del Gen , Epitelio Pigmentado de la Retina/metabolismo , Proteína smad3/biosíntesis , Transcripción Genética , Factor de Crecimiento Transformador beta/biosíntesis , Línea Celular , Proteína Potenciadora del Homólogo Zeste 2/genética , Humanos , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética , Tretinoina/farmacología
12.
BMB Rep ; 52(3): 202-207, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30638178

RESUMEN

Keloids are the most common pathological form of trauma healing, with features that seriously affect appearance and body function, are difficult to treat and have a high recurrence rate. Emerging evidence suggests that miRNAs are involved in a variety of pathological processes and play an important role in the process of fibrosis. In this study, we investigated the function and regulatory network of miR-152-5p in keloids. The miRNA miR-152-5p is frequently downregulated in keloid tissue and primary cells compared to normal skin tissue and fibroblasts. In addition, the downregulation of miR-152-5p is significantly associated with the proliferation, migration and apoptosis of keloid cells. Overexpression of miR-152-5p significantly inhibits the progression of fibrosis in keloids. Smad3 is a direct target of miR-152-5p, and knockdown of Smad3 also inhibits fibrosis progression, consistent with the overexpression of miR-152-5p. The interaction between miR-152-5p and Smad3 occurs through the Erk1/2 and Akt pathways and regulates collagen3 production. In summary, our study demonstrates that miR-152-5p/Smad3 regulatory pathways involved in fibrotic progression may be a potential therapeutic target of keloids. [BMB Reports 2019; 52(3): 202-207].


Asunto(s)
Queloide/genética , MicroARNs/genética , MicroARNs/metabolismo , Proteína smad3/biosíntesis , Adolescente , Adulto , Apoptosis/genética , Proliferación Celular/genética , Regulación hacia Abajo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/fisiología , Humanos , Queloide/metabolismo , Queloide/patología , Masculino , Transducción de Señal/genética , Piel/metabolismo , Piel/patología , Proteína smad3/genética , Proteína smad3/metabolismo
13.
Cancer Gene Ther ; 26(5-6): 119-127, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30177817

RESUMEN

The SMAD family (SMAD1-9) was critically important for regulating cellular process through transforming growth factor-ß signaling pathway, and contributed to carcinogenesis; however, their prognostic roles in acute myeloid leukemia (AML) remained unclear. This study collected 84 de novo AML patients treated with chemotherapy and 71 patients who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT). Kaplan-Meier survival estimate indicated that among SMAD1-9, high SMAD3 and SMAD7 expression were both associated with poor event-free survival (EFS) and overall survival (OS; all P < 0.05) in AML patients undergoing chemotherapy; and high SMAD6 expression was associated with shorter EFS and OS (all P < 0.01) in patients underwent allo-HSCT. Multivariate analysis showed that only high SMAD7 expression had adverse effect on EFS and OS (P = 0.021, 0.026) independently. Furthermore, High SMAD3 and SMAD7 expressers had significantly shorter EFS and OS than low expressers (P = 0.006, 0.001). In AML patients who went through allo-HSCT, there were no significant differences for EFS and OS between patients with high and low-expression SMAD3 or SMAD7. Our study suggested that high expression of SMAD3 and SMAD7 predicted adverse prognosis in AML patients undergoing chemotherapy and SMAD7 was a better prognostic marker than SMAD3. Their prognosis impact may be overcome by allo-HSCT.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Proteína smad3/biosíntesis , Proteína smad7/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Pronóstico , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína smad7/genética , Proteína smad7/metabolismo , Adulto Joven
14.
Respir Res ; 19(1): 262, 2018 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-30594196

RESUMEN

BACKGROUND: This study investigated the function of SMAD3 (SMAD family member 3) in regulating PAX6 (paired box 6) in non-small cell lung cancer. METHODS: First, qRT-PCR was employed to detect SMAD3 expression in cancer tissues along with normal tissues and four cell lines, including BEAS-2B, H125, HCC827 and A549 cells. SMAD3 was knocked down by small interference RNA (siRNA), and then its expression was determined via qRT-PCR and Western blot analysis. The correlation between SMAD3 and PAX6 was determined by double luciferase reporter experiments and chromatin immunoprecipitation (ChIP) assay. Cell viability was evaluated by CCK-8 and colony forming assays, while cell migration and invasion were detected by Transwell analysis. RESULTS: SMAD3 and PAX6 were upregulated in lung cancer tissues and cancer cells. Knocking down SMAD3 and PAX6 by transfection with siRNAs specifically suppressed the expression of SMAD3 and PAX6 mRNA and protein levels. SMAD3 could promote PAX6 transcriptional activity by binding to its promoter. Reduced expression of SMAD3 led to the downregulation of PAX6 mRNA and protein levels along with decreased cell migration, invasion, proliferation and viability in A549 and HCC827 cells. PAX6 overexpression altered the si-SMAD3-induced inhibition of cell migration, invasion, proliferation and viability in A549 and HCC827 cells. Additionally, PAX6 knockdown alone also repressed the cell migration, invasion, proliferation and viability of the cell lines. CONCLUSIONS: SMAD3 promotes the progression of non-small cell lung cancer by upregulating PAX6 expression.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Factor de Transcripción PAX6/biosíntesis , Proteína smad3/biosíntesis , Transcripción Genética/fisiología , Células A549 , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Factor de Transcripción PAX6/genética , Proteína smad3/genética
15.
Eur Rev Med Pharmacol Sci ; 22(22): 7621-7627, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30536302

RESUMEN

OBJECTIVE: To study the role of HGF (stem cell growth factor) in renal interstitial fibrosis and to explore its underlying mechanism. MATERIALS AND METHODS: A unilateral ureteral obstruction (UUO) mouse model was first constructed, and kidney samples of mice were then collected. Fibrosis-related indicators in UUO mice kidney were detected by Western blot. The mRNA and protein levels of HGF in UUO mice were detected by quantitative Real-time-polymerase chain reaction (qRT-PCR) and Western blot, respectively. The HGF overexpression mouse model was established by using UUO mice. For in vitro experiments, fibrosis-related indicators and the expression of HGF were detected in transforming growth factor-ß1 (TGF-ß1)-induced NRK-52E cells. Finally, a p-SMAD3 knockdown mouse model was established to confirm whether p-SMAD3 was involved in HGF-regulated renal interstitial fibrosis. RESULTS: The expression levels of HGF and α-SMA (α-smooth muscle actin) were both significantly increased in UUO mice, while E-cadherin expression was significantly decreased, which were consistent with results of in vitro experiments. Overexpression of HGF remarkably decreased the protein and mRNA levels of α-SMA in fibrotic NRK-52E cells. After overexpression of HGF in UUO mice, α-SMA was remarkably downregulated, whereas E-cadherin was significantly upregulated. Further, results also demonstrated that HGF was upregulated and α-SMA was downregulated after p-SMAD3 knockdown in UUO mice. CONCLUSIONS: HGF is highly expressed during renal interstitial fibrosis, which may suppress renal interstitial fibrosis by inhibiting the TGF-ß1/SMAD signaling pathway.


Asunto(s)
Factor de Crecimiento de Hepatocito/uso terapéutico , Nefritis Intersticial/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Proteína smad3/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Cadherinas/biosíntesis , Línea Celular , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Técnicas de Silenciamiento del Gen , Masculino , Ratones , Nefritis Intersticial/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteína smad3/biosíntesis , Proteína smad3/genética , Obstrucción Ureteral/complicaciones
16.
Neurourol Urodyn ; 37(8): 2502-2509, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30070388

RESUMEN

AIMS: Sacral spinal cord injury (SCI) could induce underactive bladder (UAB). Malfunction of connexin 43 (CX43) regulated by TGF-ß1 might involve in urinary bladder dysfunction. We studied the changes of CX43 and TGF-ß1/Smad3 signaling in detrusor of neurogenic bladder (NB) in sacral SCI rats. METHODS: Sacral SCI was produced by hemisection (SSCH) or transection (SSCT) of spinal cord between L4 and L5 in female Wistar rats. BBB scores, residual urine volume and bladder weight as well as characteristic cystometric parameters at 6th week were used to confirm the successful establishment of NB. Western blotting and qRT-PCR were used to exam the protein and mRNA expression levels of CX43, CX45, TGF-ß1, and Smad3 in detrusor. RESULTS: BBB scores were significantly decreased, with the lowest in SSCT rats (P < 0.01). The residual urine volume, mean bladder weight, and cystometric parameters were increased, with the highest in SSCT rats. CX43 and phospho-CX43 protein levels were significantly decreased, but those of TGF-ß1, Smad3, and phospho-Smad3 were significantly increased. It was the protein and mRNA levels of CX43 but not those of CX45 which were decreased in negative accordance with those of TGF-ß1 and Smad3. Those changes were more significant in SSCT than in SSCH rats. CONCLUSIONS: This study indicates that voiding dysfunction is related to the decreased CX43 function in detrusor from NB. TGF-ß1/Smad3 signaling might be involved in the down-regulation of CX43 in SCI rats. Early regulation of CX43 might be beneficial to patients with voiding dysfunction.


Asunto(s)
Conexina 43/biosíntesis , Traumatismos de la Médula Espinal/fisiopatología , Factor de Crecimiento Transformador beta1/biosíntesis , Vejiga Urinaria Neurogénica/fisiopatología , Animales , Conexina 43/genética , Estado de Descerebración/fisiopatología , Femenino , Tamaño de los Órganos , Ratas , Ratas Wistar , Proteína smad3/biosíntesis , Proteína smad3/genética , Traumatismos de la Médula Espinal/complicaciones , Factor de Crecimiento Transformador beta1/genética , Vejiga Urinaria/patología , Vejiga Urinaria Neurogénica/etiología , Urodinámica
17.
Histol Histopathol ; 33(12): 1287-1298, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29916554

RESUMEN

Drug-induced gingival enlargement (DIGE) is a fibrotic condition associated with systemic administration of the anti-epileptic drug, phenytoin. We have previously demonstrated that periostin, which is transforming growth factor-beta (TGF-ß) inducible gene, is upregulated in various fibrotic conditions including gingival enlargement associated with nifedipine. The objective of this study was to assess periostin expression in phenytoin-induced gingival enlargement (PIGE) tissues and to investigate the mechanisms underlying periostin expression. Human PIGE tissues were assessed using Masson's trichrome, with cell infiltration and changes in extracellular matrix composition characterized through labeling with antibodies to periostin, phospho-SMAD 3, TGF-ß, as well as the macrophage markers CD68 and RM3/1. Using human gingival fibroblasts (HGFs) in vitro we examined the pathways through which phenytoin acts on fibroblasts. In PIGE tissues, which demonstrate altered collagen organization and increased inflammatory cell infiltration, periostin protein was increased compared with healthy tissues. p-SMAD2/3, the transcription factor associated with canonical TGF-ß signaling, is localized to the nuclei in both gingival fibroblasts and oral epithelial cells in PIGE tissues, but not in healthy tissue. In vitro culture of HGFs with 15 and 30 µg/ml of phenytoin increased periostin protein levels, which correlated with p-SMAD3 phosphorylation. Inhibition of canonical TGF-ß signaling with SB431542 significantly reduced phenytoin induction of SMAD3 phosphorylation and periostin expression in HGFs. Analysis of PIGE tissues showed a subset of CD68 stained macrophages were TGF-ß positive and that RM1/3 regenerative macrophages were present in the tissues. Our results demonstrate that phenytoin up-regulates periostin in HGFs in a TGF-ß-dependent manner.


Asunto(s)
Anticonvulsivantes/efectos adversos , Moléculas de Adhesión Celular/biosíntesis , Sobrecrecimiento Gingival/inducido químicamente , Fenitoína/efectos adversos , Proteína smad3/biosíntesis , Adulto , Anciano , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Sobrecrecimiento Gingival/metabolismo , Sobrecrecimiento Gingival/patología , Humanos , Masculino , Persona de Mediana Edad , Fosforilación , Adulto Joven
18.
Int J Biol Macromol ; 116: 537-544, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29704604

RESUMEN

We investigated the transforming growth factor-b1 (TGF-ß1)/Smad3 signaling pathway in rats with cerebral ischemia and oxygen-glucose-deprived (OGD) microglia. Cerebral ischemia is a clinical condition that occurs when insufficient blood flows to the brain to maintain metabolic activity. TGF-ß1 is a well-known functional peptide that regulates cell differentiation, migration, proliferation, and apoptosis. In the current study, we determined the infarct size and TGF-ß1/Smad3 protein expression in stroke-induced rats. Apoptosis and TGF-ß1/Smad3 mRNA and protein expression were determined in transfected OGD human microglial cells. TGF-ß1 treatment resulted in smaller infarct regions than in control cells, whereas TGF-ß1 inhibitor treatment resulted in larger infarcts. The TGF-ß1-treated groups showed substantial TGF-ß1 and Smad3 expression by immunofluorescence compared to the controls. Apoptosis was significantly reduced in TGF-ß1- and Smad3-transfected cells, and an increased rate of apoptosis was observed in Smad3 or TGF-ß1 siRNA-transfected cells. TGF-ß1 and Smad3 mRNA and protein expression increased following TGF-ß1 and Smad3 transfection. Taken together, our experimental results show that Smad3 and TGF-ß1 play a protective role against ischemic stroke, as demonstrated by the reduced infarct size. Smad3 and TGF-ß1 expression was increased in cells transfected with TGF-ß1, whereas Smad3 and TGF-ß1 expression was increased in TGF-ß1 inhibitor-transfected cells.


Asunto(s)
Isquemia Encefálica/metabolismo , Glucosa/metabolismo , Microglía/metabolismo , Transducción de Señal , Proteína smad3/biosíntesis , Factor de Crecimiento Transformador beta1/biosíntesis , Animales , Apoptosis , Isquemia Encefálica/patología , Regulación de la Expresión Génica , Masculino , Microglía/patología , Ratas , Ratas Wistar
19.
Life Sci ; 202: 11-20, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29577879

RESUMEN

BACKGROUND: MicroRNAs (miRs) play an important role in osteoclastogenesis. However, no study has investigated the underlying molecular mechanisms of miR-145 in this process. The purpose of the present study was to investigate the role of miR-145 and its post-transcriptional mechanism in the progression of osteoclast differentiation. METHODS: Macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor-kB ligand (RANKL) were used to induce osteoclastogenesis originated from bone marrow-derived macrophages (BMMs). Female C57BL/6J mice were divided into sham, OVX, OVX + NC-agomir and OVX + miR-145-agomir groups. Tartrate-resistant acid phosphatase (TRAP) staining was performed to identify osteoclasts in-vitro and in-vivo. The mRNA and protein levels in osteoclast and tibia were assayed by qRT-PCR and western blotting, respectively. RESULTS: miR-145 expression was inhibited in RANKL-induced osteoclastogenesis, whereas overexpression of miR-145 attenuated it. We further found that Smad3 is a direct target gene of miR-145 by binding with its 3'-UTR. Overexpression of miR-145 significantly suppressed Smad3 mRNA and protein expression. In-vivo, miR-145 agomir treatment inhibited osteoclast activity in OVX mice by inhibiting Smad3 expression. CONCLUSION: We provide the evidence that over-expression of miR-145 could inhibit osteoclast differentiation, at least partially, by decreasing Smad3 expression.


Asunto(s)
Células de la Médula Ósea/metabolismo , Macrófagos/metabolismo , MicroARNs/biosíntesis , MicroARNs/genética , Osteoclastos/fisiología , Osteogénesis/genética , Ovariectomía , Ligando RANK/genética , Proteína smad3/biosíntesis , Proteína smad3/genética , Regiones no Traducidas 3'/genética , Animales , Diferenciación Celular/genética , Femenino , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ligando RANK/biosíntesis , Fosfatasa Ácida Tartratorresistente/metabolismo , Tibia/citología , Tibia/metabolismo
20.
J Virol ; 92(9)2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29444941

RESUMEN

High plasma lactate is associated with poor prognosis of many malignancies, but its role in virally mediated cancer progression and underlying molecular mechanisms are unclear. Epstein-Barr virus (EBV), the first human oncogenic virus, causes several cancers, including B-cell lymphoma. Here, we report that lactate dehydrogenase A (LDH-A) expression and lactate production are elevated in EBV-immortalized B lymphoblastic cells, and lactic acid (LA; acidic lactate) at low concentration triggers EBV-infected B-cell adhesion, morphological changes, and proliferation in vitro and in vivo Moreover, LA-induced responses of EBV-infected B cells uniquely occurs in viral latency type III, and it is dramatically associated with the inhibition of global viral microRNAs, particularly the miR-BHRF1 cluster, and the high expression of SMAD3, JUN, and COL1A genes. The introduction of miR-BHRF1-1 blocks the LA-induced effects of EBV-infected B cells. Thus, this may be a novel mechanism to explain EBV-immortalized B lymphoblastic cell malignancy in an LA microenvironment.IMPORTANCE The tumor microenvironment is complicated, and lactate, which is created by cell metabolism, contributes to an acidic microenvironment that facilitates cancer progression. However, how LA operates in virus-associated cancers is unclear. Thus, we studied how EBV (the first tumor virus identified in humans; it is associated with many cancers) upregulates the expression of LDH-A and lactate production in B lymphoma cells. Elevated LA induces adhesion and the growth of EBV-infected B cells by inhibiting viral microRNA transcription. Thus, we offer a novel understanding of how EBV utilizes an acidic microenvironment to promote cancer development.


Asunto(s)
Adhesión Celular/genética , Proliferación Celular/genética , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/genética , L-Lactato Deshidrogenasa/biosíntesis , Ácido Láctico/biosíntesis , MicroARNs/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Linfocitos B/fisiología , Linfocitos B/virología , Línea Celular Transformada , Supervivencia Celular/genética , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/metabolismo , Humanos , Isoenzimas/biosíntesis , Lactato Deshidrogenasa 5 , Ácido Láctico/sangre , MAP Quinasa Quinasa 4/biosíntesis , MAP Quinasa Quinasa 4/genética , MicroARNs/biosíntesis , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteína smad3/biosíntesis , Proteína smad3/genética , Microambiente Tumoral/genética , Latencia del Virus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA