Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Biol Rep ; 51(1): 525, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38632128

RESUMO

BACKGROUND: A series of previous investigations have revealed that p-Smad3 plays a facilitative role in the differentiation and maturation of osteoblasts, while also regulating the expression of certain intercellular communication factors. However, the effects of p-Smad3 in osteoblasts before and after maturation on the proliferation, migration, differentiation, apoptosis and other cellular behaviors of osteoclasts have not been reported. METHODS: MC3T3-E1 cells were cultured in osteogenic induction medium for varying durations, After that, the corresponding conditioned medium was collected and the osteoclast lineage cells were treated. To elucidate the regulatory role of p-Smad3 within osteoblasts, we applied the activator TGF-ß1 and inhibitor SIS3 to immature and mature osteoblasts and collected corresponding conditioned media for osteoclast intervention. RESULTS: We observed an elevation of p-Smad3 and Smad3 during the early stage of osteoblast differentiation, followed by a decline in the later stage. we discovered that as osteoblasts mature, their conditioned media inhibit osteoclasts differentiation and the osteoclast-coupled osteogenic effect. However, it promotes apoptosis in osteoclasts and the angiogenesis coupled with osteoclasts. p-Smad3 in immature osteoblasts, through paracrine effects, promotes the migration, differentiation, and osteoclast-coupled osteogenic effects of osteoclast lineage cells. For mature osteoblasts, p-Smad3 facilitates osteoclast apoptosis and the angiogenesis coupled with osteoclasts. CONCLUSIONS: As pre-osteoblasts undergo maturation, p-Smad3 mediated a paracrine effect that transitions osteoclast cellular behaviors from inducing differentiation and stimulating bone formation to promoting apoptosis and coupling angiogenesis.


Assuntos
Osteoclastos , Osteogênese , Proteína Smad3 , Diferenciação Celular , Meios de Cultivo Condicionados/farmacologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Osteogênese/fisiologia , Animais , Camundongos , Proteína Smad3/genética , Proteína Smad3/metabolismo
2.
Biochem Biophys Res Commun ; 636(Pt 1): 64-74, 2022 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-36332484

RESUMO

Gap junctional intercellular communication (GJIC) is indispensable for the maintenance of physiological balance in articular cartilage. Transforming growth factor-ß3 (TGF-ß3), an important growth factor of TGF-ß superfamily, is well recognized to play a unique regulatory role in cartilage development and diseases. However, the role of TGF-ß3 in GJIC in adult chondrocytes remains elusive. This work aims to investigate the effect of TGF-ß3 on gap-junction mediated intercellular communication in chondrocytes. We first showed that TGF-ß3 could enhance the synaptic connections between chondrocytes by scanning electron microscopy (SEM) and promote the cell-to-cell communication in living chondrocytes by scrape loading/dye transfer assay. We then confirmed that TGF-ß3 enhanced cell-to-cell communication via up-regulation of connexin 43 (Cx43). We next found that TGF-ß3-enhanced GJIC required the participation of TGF-beta type I receptor ALK5 and depended on the activation of p-Smad3 signalling. Finally, through inhibitor experiments of SB525334 and SIS3, we demonstrated that TGF-ß3-induced functional GJIC in chondrocytes via the axis of ALK5/p-Smad3 signalling. Taking together, these results demonstrate a strong correlation between TGF-ß3 and GJIC in chondrocytes, which provides a new perspective on the importance of TGF-ß3 on cartilage physiology and pathobiology.


Assuntos
Cartilagem Articular , Condrócitos , Condrócitos/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fator de Crescimento Transformador beta3/farmacologia , Fator de Crescimento Transformador beta3/metabolismo , Comunicação Celular , Cartilagem Articular/metabolismo , Junções Comunicantes/metabolismo
3.
Ecotoxicol Environ Saf ; 205: 111327, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32961493

RESUMO

Exposure to PM2.5 can cause serious harm to the respiratory system. Until now, although many toxicological studies have shown that pulmonary fibrosis can be caused by long-term PM2.5 exposure, there is no evidence that Endothelial-Mesenchymal Transition (EndMT) can trigger the process of pulmonary fibrosis after exposure. LncRNAs are a class of non-coding RNAs detected in mammalian cells. Nevertheless, researchers have not found whether lncRNAs participate in PM2.5 induced EndMT during pathophysiological duration. The Balb/c mouse model was exposed to PM2.5 for 4 months by dynamic intoxication. The levels of specific endothelial and mesenchymal markers were evaluated by molecular biology experiments to elucidate the mechanisms of EndMT induced by PM2.5 in lung tissues. LncRNA microarray analysis of the established mouse model of PM2.5 exposure was performed. Based on a bioinformatics analysis and RT-qPCR analysis, lncRNA Gm16410 attracted our attention. The change of lncRNA Gm16410 in mouse pulmonary vascular endothelial cells (MHCs) exposed to PM2.5 was verified, and the mechanism of lncRNA Gm16410 in EndMT was discussed. The changes of cell function were evaluated by cell migration and proliferation experiments. The molecular biology experiments proved that PM2.5 induced EndMT by activating the TGF-ß1/Smad3/p-Smad3 pathway in vitro. The relationship of EndMT and lncRNA Gm16410 was verified in mouse lung tissues and MHC cells by PM2.5 exposure. The involvement of lncRNA Gm16410 in PM2.5-induced EndMT highlights the potential of lncRNA to promote pulmonary fibrosis under environmental pollution.


Assuntos
Material Particulado/toxicidade , RNA Longo não Codificante/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Pulmão/metabolismo , Camundongos , Material Particulado/metabolismo , Fibrose Pulmonar/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3 , Fator de Crescimento Transformador beta1/metabolismo
4.
Tumour Biol ; 37(6): 7277-86, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26666823

RESUMO

Hepatocellular carcinoma (HCC) is a common cancer with poor prognosis. The multikinase inhibitor sorafenib is the only clinically proved systematic treatment for HCC. However, few patients respond to sorafenib. Hypoxic microenvironments contribute to sorafenib resistance. LB-100, a serine/threonine protein phosphatase 2A (PP2A) inhibitor was previously found to be a chemosensitizer in HCC. Here, we tested whether LB-100 could sensitize HCC to the effects of sorafenib. Intriguingly, LB-100 enhanced the effects of sorafenib in HCC cells only during hypoxic environments. LB-100 dramatically increased intracellular p-Smad3 level, which was responsible for the effect of LB-100 as a sensitizer. LB-100 downregulated Bcl-2 expression and enhanced sorafenib-induced apoptosis in HCC cells. We further proved that PP2A mediated LB-100-induced p-Smad3 overexpression. In addition, p38 mitogen-activated protein kinase pathway was activated in hypoxic conditions, and enhanced p-Smad3-dependent Bcl-2 inhibition and consequent apoptosis. In conclusion, LB-100 sensitized HCC cells to sorafenib in hypoxic environments. This effect was mediated by inactivation of PP2A, resulting in enhanced level of p-Smad3. Increased p-Smad3 downregulated Bcl-2, causing increased apoptosis of HCC cells.


Assuntos
Antineoplásicos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Carcinoma Hepatocelular/patologia , Hipóxia Celular/efeitos dos fármacos , Neoplasias Hepáticas/patologia , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteína Smad3/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes bcl-2 , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/antagonistas & inibidores , Niacinamida/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 2/antagonistas & inibidores , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Distribuição Aleatória , Proteína Smad3/genética , Sorafenibe , Ensaios Antitumorais Modelo de Xenoenxerto
5.
FASEB J ; 28(6): 2696-704, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24558198

RESUMO

TGFßR1 plays an important role in TGF-ß signaling transduction and serves as a tumor suppressor. Our previous studies show that reduced expression of TGFßR1 is common in non-small cell lung cancer (NSCLC) and TGFßR1 variants confer risk of NSCLC. However, the epigenetic mechanisms underlying the role of TGFßR1 in NSCLC carcinogenesis are still elusive. We investigated the function and regulation of TGF-ß signaling-based miRNAs in NSCLC. Computational algorithms predicted that the 3'-untranslated region (3'-UTR) of TGFßR1 is a target of miR-142-3p. Here a luciferase reporter assay confirmed that miR-142-3p can directly bind to 3'-UTR of TGFßR1. Overexpression of miR-142-3p in NSCLC A549 cells suppressed expression of TGFßR1 mRNA and protein, while knockdown of endogenous miR-142-3p led to increased expression of TGFßR1. On TGF-ß1 stimulation, stable overexpression of miR-142-3p attenuated phosphorylation of SMAD3, an indispensable downstream effector in canonical TGF-ß/Smad signaling, via repression of TGFßR1 in A549 cells. Furthermore, miR-142-3p-mediated down-regulation of TGFßR1 weakened TGF-ß-induced growth inhibition effect, and this effect was reversed by stable knockdown of endogenous miR-142-3p in A549 cells. In NSCLC tissues, miR-142-3p expression was increased and inversely correlated with TGFßR1 expression. These data demonstrate that miR-142-3p influences the proliferation of NSCLC cells through repression of TGFßR1.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Fator de Iniciação 3 em Eucariotos/genética , Neoplasias Pulmonares/metabolismo , MicroRNAs/fisiologia , Regiões 3' não Traduzidas/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo , Fator de Iniciação 3 em Eucariotos/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/biossíntese , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima
6.
Cell Biol Int ; 39(7): 852-7, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25339576

RESUMO

The mesenchymal to epithelial transition (MET) occurs in organ development and anti-tumorigenesis. We have investigated the effects of calcium (Ca(2+)) and epidermal growth factor (EGF) on human mesenchymal stem cell (hMSCs) differentiation into epithelial-like cells. hMSCs lost their biological characteristics after EGF transfection, and MET was achieved by adding 0.4 mmol Ca(2+). Western blotting and immunofluorescence showed expression of EGF, keratin, keratin 19 (K19), ß1-integrin, E-cadherin and phosphorylated focal adhesion kinase (p-FAK, Ser-910) increased in hMSCs infected with EGF and exposed to Ca(2+), although Smad3 activation was downregulated. hMSCs co-stimulated with EGF transfection and Ca(2+) can therefore differentiate into epithelial-like cells in vitro.


Assuntos
Cálcio/metabolismo , Diferenciação Celular , Fator de Crescimento Epidérmico/metabolismo , Células Epiteliais/citologia , Células-Tronco Mesenquimais/citologia , Cátions Bivalentes/metabolismo , Proliferação de Células , Células Cultivadas , Embrião de Mamíferos/citologia , Fator de Crescimento Epidérmico/genética , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Humanos , Células-Tronco Mesenquimais/metabolismo , Proteína Smad3/metabolismo , Transfecção
7.
Cell Biol Int ; 39(6): 690-5, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25612247

RESUMO

Smad ubiquitination regulatory factor 2 (Smurf2) consists of multiple WW domains which can interact with Smad7 molecule and inhibit signaling of transforming growth factor-beta (TGF-ß) cytokine. Arginase I (ArgI) is one of the main products of TGF-ß signaling that plays important roles in tumor escape and airway tissue fibrosis and remodeling in asthma. In this study, the effects of TAT fused to WW2/WW3 (TAT-WW2/WW3) recombinant protein on TGF-ß signaling and ArgI gene expression were evaluated on J774A.1 cell culture. For this purpose, interaction of TAT-WW2/WW3 with Smad7, mRNA expression of ArgI, and phosphorylated Smad3 (P-Smad3) were analyzed in TAT-WW2/WW3-treated J774A.1 cell. The results showed interaction of TAT-WW2/WW3 with Smad7, downregulation of ArgI gene expression (P < 0.05), and higher amount of P-Smad3 in the TAT-WW2/WW3-treated cells. In conclusion, we suggest that TAT-WW2/WW3 could interfere with TGF-ß signaling and reduce ArgI gene expression. Since, ArgI has important effects on tissue remodeling in asthma and cancer progression, so these findings could be used to develop a new approach in the treatment of asthma and cancers.


Assuntos
Arginase/genética , Regulação Enzimológica da Expressão Gênica , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Animais , Arginase/metabolismo , Western Blotting , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Imunoprecipitação , Camundongos , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Proteína Smad3/metabolismo , Proteína Smad7/metabolismo , Relação Estrutura-Atividade
8.
Pulm Pharmacol Ther ; 27(1): 17-28, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23518214

RESUMO

BACKGROUND AND PURPOSE: Pulmonary fibrosis (PF) is a progressing lung injury initiated by pulmonary inflammation (PI). Bleomycin (BLM) is the most common pathogenesis of PF through early PI and extensive extracellular matrix deposition. This study is aimed to determine whether NO-releasing KMUP-1 inhibits PI and PF, and if so, the benefits of KMUP-1S resulted from simvastatin (SIM)-bonding to KMUP-1. EXPERIMENT APPROACH: C57BL/6 male mice were intra-tracheally administered BLM (4 U/kg) at day 0. KMUP-1 (1-5 mg/kg), KMUP-1S (2.5 mg/kg), SIM (5 mg/kg), Plus (KMUP-1 2.5 mg/kg + SIM 2.5 mg/kg), and clarithromycin (CAM, 10 mg/kg) were orally and daily administered for 7 and 28 days, respectively, to mice, sacrificed at day-7 and day-28 to isolate the lung tissues, for examining the inflammatory and fibrotic signaling and measuring the cell population and MMP-2/MMP-9 activity in broncholaveolar lavage fluid (BAL). KEY RESULTS: KMUP-1 and KUP-1S significantly decreased neutrophil counts in BAL fluid. Fibroblastic foci were histologically assessed by H&E and Masson's trichrome stain and treated with KMUP-1 and references. Lung tissues were determined the contents of collagen and the expressions of TGF-ß, α-SMA, HMGB1, CTGF, eNOS, p-eNOS, RhoA, Smad3, p-Smad3, MMP-2 and MMP-9 by Western blotting analyses, respectively. These changes areregulated by NO/cGMP and inhibited by various treatments. KMUP-1 and KMUP-1S predominantly prevented HMGB1/MMP-2 expression at day-7 and reduced TGF-ß/phosphorylated Smad3 and CTGF at day-28. CONCLUSIONS AND IMPLICATIONS: KMUP-1 and KMUP-S restore eNOS, inhibit iNOS/ROCKII/MMP-2/MMP-9, attenuate histologic collagen disposition and reduce BALF inflammatory cells, potentially useful for the treatment of BLM-lung PF.


Assuntos
Piperidinas/farmacologia , Pneumonia/tratamento farmacológico , Fibrose Pulmonar/tratamento farmacológico , Sinvastatina/farmacologia , Xantinas/farmacologia , Animais , Bleomicina/toxicidade , Western Blotting , Líquido da Lavagem Broncoalveolar , Claritromicina/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piperidinas/administração & dosagem , Piperidinas/química , Pneumonia/patologia , Fibrose Pulmonar/patologia , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/administração & dosagem , Sinvastatina/química , Fatores de Tempo , Xantinas/administração & dosagem , Xantinas/química
9.
Biol Reprod ; 89(5): 126, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23986572

RESUMO

The initiation of primordial follicle development is essential for female fertility, but the signals that trigger this process are poorly understood. Given the potentially important roles of microRNAs (miRNAs) in the ovary, we aimed to study the expression patterns and regulatory functions of miRNAs during the initiation of primordial follicle development. Expression patterns of miRNA in the neonatal mouse ovary were profiled by microarray, and 24 miRNAs whose abundances differed significantly between ovaries from 3- and 5-day-old mice were identified. Pathway enrichment analysis revealed that 48 signal transduction pathways are modulated by the up-regulated miRNAs and 29 pathways are modulated by the down-regulated miRNAs (P-value and false discovery rate < 0.001). A miRNA-mRNA regulatory network was established for TGF-beta signaling pathway-related genes. Among the miRNAs involved in this pathway, miR-145 was chosen for further analysis. Down-regulation of miR-145 using an antagomir (AT) decreased the proportion and number of the primordial follicles and increased that of the growing follicles in the cultured ovaries (P < 0.05). The mean oocyte diameter in the primordial follicles was significantly greater in the AT group relative to the AT-negative control group (P < 0.05), whereas the mean oocyte diameter in growing follicles was smaller in the AT group than in the AT-negative control group. In addition, we confirmed that miR-145 targets Tgfbr2. The miR-145 AT caused an increase in TGFBR2 expression and activation of Smad signaling but did not affect the p38 MAPK or JNK pathway. These data suggest that miRNAs and the signaling pathways they modulate are involved in the initiation of primordial follicle development, and miR-145 targets Tgfbr2 to regulate the initiation of primordial follicle development and maintain primordial follicle quiescence.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , MicroRNAs/genética , Folículo Ovariano/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Feminino , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Análise em Microsséries , Ovário/citologia , Ovário/crescimento & desenvolvimento
10.
Front Cell Dev Biol ; 10: 876054, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35478960

RESUMO

The therapeutic effects and mechanism of umbilical cord mesenchymal stem cells (UC-MSC) on kidney injury in MRL/Ipr mice were studied. UC-MSC, methylprednisolone (MP), and their combination were used to treat MRL/Ipr mice. The therapeutic effects were evaluated by renal function assessment, and HE, PAS, and Masson staining were carried out on renal tissues and visualized by electron microscopy. Subsequently, podocyte injury was detected by the presence of podocin in renal tissues by immunofluorescence. To further explore the mechanism, serum TGF-ß1 was measured, and TGF-ß1, p-Smad3, and TRAF6 in the renal tissue were detected by Western blotting. In vitro, TGF-ß1 was used to stimulate podocytes, and the podocyte activity and changes in synaptopodin were observed after UC-MSC treatment. Significant improvements in renal function and pathological injury were observed in the UC-MSC group compared to the lupus nephritis (LN) model group. UC-MSC and MP treatment improved podocyte injury in MRL/Ipr mice. Western blot examination showed a significant increase in TGF-ß1, p-Smad3, and TRAF6 expression in renal tissues of the LN model group, while significant downregulation of those proteins was observed in the UC-MSC group. After TGF-ß1 stimulation in vitro, podocyte activity decreased, and UC-MSC treatment improved podocyte activity and restored synaptopodin expression. UC-MSC therapy could improve the deterioration of renal function and the pathological changes of the renal tissues in MRL/Ipr mice. Our study suggested that UC-MSC may improve kidney injury and podocyte injury in LN mice by inhibiting the TGF-ß1 pathway.

11.
Eur J Med Res ; 27(1): 126, 2022 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-35858881

RESUMO

BACKGROUND: Cleft lip repair surgery always results in visible scarring. It has been proved that scar formation can be reduced by inhibiting the p38 mitogen-activated protein kinases (p38MAPKs) signaling pathway. However, the interaction between p38MAPK and Smads in scar formation is still controversial. METHODS: This study was designed to investigate whether inhibition of p38MAPK reduces postoperative scar formation of cleft lips on rabbits via the Smads signaling pathway. Scar models in rabbits after cleft lip surgery were created and their fibroblasts were extracted. Then the expression of p38MAPK was disturbed by adenovirus in vitro and Vivo. The scar thickness was measured and scar tissues were excised for Sirius red staining and immunohistochemistry to detect the expression of type I collagen (col I), type III collagen (col III), and α-smooth muscle actin (α-SMA). The underlying mechanisms of p38MAPK knockdown on the extracellular matrix and Smad signaling pathway were invested in vitro using the EdU assay, Western blot, RT PCR, and immunofluorescence. RESULTS: p38MAPK knockdown suppresses the expression of p-smad3 and p-smad2 in fibroblasts, modulating the expression of its target genes, such as α-SMA, col I, and col III. When Ad-P38MAPK-1 was injected into lip scar, it reduced the expression of scar-related genes and scar thickness when compared to the negative control groups. CONCLUSIONS: In rabbits, inhibiting p38MAPK expression prevents scar proliferation through inhibiting the Smad signaling pathway after cleft lip surgery.


Assuntos
Fenda Labial , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Proliferação de Células , Cicatriz/metabolismo , Cicatriz/prevenção & controle , Fenda Labial/metabolismo , Fenda Labial/cirurgia , Fibroblastos/metabolismo , Coelhos , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta1/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Biochim Biophys Acta Mol Cell Res ; 1869(2): 119175, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34863793

RESUMO

Connexin 43 (Cx43)-mediated gap junction intercellular communication (GJIC) plays a crucial role in the pathology and physiology of joint tissues. Transforming growth factor-ß2 (TGF-ß2), one of the potent regulatory factors in chondrocytes, plays a key role in the regulation of cell cycle and development of joint diseases. However, it is still unknown how TGF-ß2 mediates GJIC in chondrocytes. The aim of this study was to explore the potential mechanism by which TGF-ß2 regulates GJIC in chondrocytes. CCK-8 assays and scratch assays were performed to define the role of TGF-ß2 on cell proliferation and migration. The scrape loading/dye transfer assay and scanning electron microscopy (SEM) were used to verify the effect of TGF-ß2 on GJIC between chondrocytes. qPCR was performed to analyse the expression of genes in the gap junction protein family in chondrocytes. The expression of the Cx43 protein and phosphorylated Smad3 (p-Smad3) was evaluated by western blot assay. Immunofluorescence staining was used to explore p-Smad3 signalling pathway activation and Cx43 distribution. From these experiments, we found that the Cx43 protein was the most highly expressed member of the gap junction protein family in chondrocytes. We also found that TGF-ß2 facilitated cell-to-cell communication in chondrocytes by upregulating Cx43 expression in chondrocytes. Finally, we found that TGF-ß2 activated Smad3 signalling and promoted the nuclear aggregation of p-Smad3. Inhibition experiments by SIS3 also confirmed that TGF-ß2-mediated GJIC through p-Smad3 signalling. For the first time, this study confirmed that TGF-ß2 could regulate the formation of Cx43-mediated GJIC in chondrocytes via the canonical p-Smad3 signalling pathway.


Assuntos
Comunicação Celular , Condrócitos/metabolismo , Conexina 43/metabolismo , Junções Comunicantes/fisiologia , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta2/farmacologia , Animais , Proliferação de Células , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Conexina 43/genética , Junções Comunicantes/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Transdução de Sinais , Proteína Smad3/genética
13.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 38(5): 559-563, 2022 Sep.
Artigo em Zh | MEDLINE | ID: mdl-37088770

RESUMO

OBJECTIVE: To investigate the effects of chronic intermittent hypoxia (CIH) on the expression of transforming growth factor-ß (TGF-ß), P-samd3, serum laminin (LN) and hyaluronidase (HA) in mouse lung tissues and the protective effects of Bu Zhong Yi Qi decoction on lung interstitial deposition damage in CIH mice. METHODS: Fifty SPF-grade C57BL mice were randomly divided into five groups (n=10): blank control group, CIH model group, and CIH+ low, medium and high doses of Bu Zhong Yi Qi decoction group. Mice were placed under normoxia or CIH conditions, respectively. The Chinese medicine group was given the corresponding doses of drugs. HE staining was performed to assess pathological changes and Masson staining was performed to assess collagen deposition. Western blot was performed to detect the expressions of channel proteins such as TGF-ß1, P-smad3 and down stream α-SMA and Collagen I. ELISA was performed to detect the serum levels of TGF-ß1, LN and HA. RESULTS: HE staining showed alveolar collapse, septal thickening and epithelial cell necrosis in CIH mice, Masson showed massive collagen fiber proliferation and deposition in lung interstitium, while the above changes in lung tissues were significantly improved in the CIH + Bu Zhong Yi Qi decoction groups compared with the CIH group. TGF-ß1, P-smad3 and Collagen I, Collagen Ⅲ, and α-SMA expression levels were increased compared with the blank control group (P<0.05), and the expressions of TGF-ß1 and LN in serum were upregulated (P<0.05). The expressions of TGF-ß1, P-smad3, Collagen I protein and SMA-α in the lung tissues of the CIH+ Bu Zhong Yi Qi decoction groups were downregulated significantly compared with those of the CIH group (P<0.05), and the improvement of multiple indexes in the CIH+high-dose CIH intervention group was better than those of the low-dose group (P<0.05). CONCLUSION: Bu Zhong Yi Qi decoction can inhibit alveolar structural changes and excessive collagen deposition in the interstitium of CIH mice, and then improve lung function in CIH mice. The mechanism may be related to the down-regulation of protein expression related to TGF-ß/smads signaling pathway by Bu Zhong Yi Qi decoction.


Assuntos
Medicamentos de Ervas Chinesas , Fibrose Pulmonar , Camundongos , Animais , Fator de Crescimento Transformador beta1/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta/metabolismo , Fibrose
14.
Int J Biochem Cell Biol ; 129: 105863, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33049375

RESUMO

INTRODUCTION: Tanshinone IIA (TSIIA), an active component of Salvia miltiorrhiza (Danshen), is reported to inhibit cell proliferation in hypertrophic scars (HS). In our previous study, we observed that lncRNA human-specific regulatory loci (HSRL) was up-regulated in HS tissues. However, whether TSIIA serves as an effective treatment for HS through affecting HSRL is still unexplored. METHODS: TGF-ß1-stimulated fibroblast were used as the in vitro HS model. The effects of TSIIA on cell proliferation were evaluated using CCK-8, Edu staining and colony formation assays. By performing loss-of-function and rescue experiments, we explored the role of HSRL and Sorting nexin 9 (SNX9) in TGF-ß1-stimulated fibroblast. Employing RNA-protein pull-down assay and Co-immunoprecipitation, we further investigated the mechanisms through which TSIIA attenuated TGF-ß1-stimulated fibroblast. RESULTS: Our data demonstrated that TSIIA could effectively attenuate TGF-ß1-mediated fibroblast proliferation in a dose-dependent manner. Meanwhile, TSIIA could down-regulate the expression of α-SMA, VEGFA, Collagen 1, HSRL, SNX9 and p-Smad2/3 in TGF-ß1-stimulated HSF. In addition, we found that SNX9 overexpression reversed the effects of HSRL knockdown on TGF-ß1-stimulated HSF. Furthermore, we confirmed that TSIIA treatment weakens the interaction between p-Smad3 and SNX9 in HS models. CONCLUSIONS: Tanshinone IIA down-regulated p-Smad3 signaling to inhibit TGF-ß1-mediated fibroblast proliferation via lncRNA-HSRL/SNX9.


Assuntos
Abietanos/farmacologia , Fibroblastos/efeitos dos fármacos , Fosfoproteínas/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos
15.
Oncotarget ; 11(29): 2863-2872, 2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32754303

RESUMO

TGF-ß1 is an epithelial-mesenchymal transition (EMT)-inducing factor that is critical in tumor progression. However, whether the effect of TGF-ß1 on breast cancer is through the EMT pathway remains to be determined, and drug development based on this mechanism needs to be improved. Results of this study showed that TGF-ß1 dysregulation significantly correlated with the expression levels of EMT-associated markers and transcriptional factors. Exogenous expression of TGF-ß1 promoted breast cancer cell metastasis and EMT progression. In addition, direct binding of baicalin to TGF-ß1 caused its inactivation, which subsequently blocked signal transduction and inhibited breast cancer cell metastasis. In vivo experiment results further invalidated the inhibitory effect of baicalin on TGF-ß1-induced tumor metastasis. These results suggest that baicalin, an active ingredient used in traditional Chinese medicine, exhibits a potential therapeutic effect on breast cancer metastasis by regulating TGF-ß1-dependent EMT progression.

16.
Front Pharmacol ; 9: 209, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29593538

RESUMO

Aim: Salidroside is an active compound extracted from Rhodiola rosea which is used to alleviate fatigue and enhance endurance in high altitude regions. Some studies have demonstrated that salidroside can affect precursor cell differentiation in hematopoietic stem cells, erythrocytes, and osteoblasts. The aim of this study was to investigate the effect of salidroside on myoblast differentiation and to explore the underlying molecular mechanisms of this effect. Methods: C2C12 myoblast cells were treated with different concentrations of salidroside in differentiation media. Real-time PCR, Western blotting, and immunofluorescence assay were employed to evaluate the effects of salidroside on C2C12 differentiation. RNA interference was used to reveal the important role of Myf5 in myogenesis inhibited by salidroside. Chromatin Immunoprecipitation and dual-luciferase reporter assay were utilized to explore the underlying mechanisms of salidroside-induced upregulation of Myf5. Results: We found that salidroside inhibits myogenesis by downregulating MyoD and myogenin, preserves undifferentiated reserve cell pools by upregulating Myf5. Knocking down Myf5 expression significantly rescued the myogenesis inhibited by salidroside. The effect of salidroside on myogenesis was associated with increased phosphorylated Smad3 (p-Smad3). Both SIS3 (Specific inhibitor of p-Smad3) and dominant negative Smad3 plasmid (DN-Smad3) attenuated the inhibitory effect of salidroside on C2C12 differentiation. Moreover, the induction of Myf5 transcription by salidroside was dependent on a Smad-binding site in the promoter region of Myf5 gene. Conclusion and Implications: Our findings identify a novel role and mechanism for salidroside in regulating myogenesis through p-Smad3-induced Myf5 transcription, which may have implications for its further application in combating degenerative muscular diseases caused by depletion of muscle stem cells, such as Duchenne muscular dystrophy or sarcopenia.

17.
Eur J Med Chem ; 157: 229-247, 2018 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-30096654

RESUMO

In this study, anti-IPF lead compounds 42 and 44, derived from natural sesquiterpene lactones Isoalantolactone and alantolactone, were discovered by screening from a high-throughput TGF-ß1 reporter luciferase assay. Notably, they could reduce the myofibroblast activation and extracellular matrix deposition both in vitro and in vivo. Additionally, compounds 42 and 44 could significantly attenuate bleomycin-induced pulmonary fibrosis in mice. Further validation of pharmacokinetics study and toxicity evaluation indicated that compound 44 might be a promising anti-IPF drug candidate.


Assuntos
Descoberta de Drogas , Fibrose Pulmonar Idiopática/tratamento farmacológico , Lactonas/farmacologia , Sesquiterpenos de Eudesmano/farmacologia , Sesquiterpenos/farmacologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Bleomicina , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Lactonas/síntese química , Lactonas/química , Camundongos , Estrutura Molecular , Células NIH 3T3 , Sesquiterpenos/síntese química , Sesquiterpenos/química , Sesquiterpenos de Eudesmano/síntese química , Sesquiterpenos de Eudesmano/química , Relação Estrutura-Atividade , Fator de Crescimento Transformador beta1/metabolismo
18.
Mol Immunol ; 70: 84-93, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26748386

RESUMO

BACKGROUND: Abnormal proliferation of ASM (airway smooth muscle) directly contributes to the airway remodeling during development of lung diseases such as asthma. Here we report that a specific microRNA (miR-23b) controls ASMCs proliferation through directly inhibiting TGFßR2/p-Smad3 pathway. METHODS: The expression of miR-23b in ASMCs was detected by quantitative real-time polymerase chain reaction (RT-PCR). The effects of miR-23b on cell proliferation and apoptosis of ASMCs were assessed by transient transfection of miR-23b mimics and inhibitor. The target gene of miR-23b and the downstream pathway were further investigated. RESULTS: Overexpression of miR-23b significantly inhibited TGF-ß1-induced ASMCs proliferation and promoted apoptosis. RT-PCR and Western blotting analysis showed miR-23b negatively regulates the expression of TGFßR2 and p-Smad3 in ASMCs. Subsequent analyses demonstrated that TGFßR2 was a direct and functional target of miR-23b, which was validated by the dual luciferase reporter assay. CONCLUSIONS: MiR-23b may function as an inhibitor of airway smooth muscle cells proliferation through inactivation of TGFßR2/p-Smad3 pathway.


Assuntos
Remodelação das Vias Aéreas/fisiologia , Regulação da Expressão Gênica/fisiologia , MicroRNAs/genética , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Animais , Apoptose/fisiologia , Asma/metabolismo , Western Blotting , Proliferação de Células/fisiologia , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad3/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA