Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.560
Filtrar
1.
Int J Mol Sci ; 25(15)2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39125585

RESUMEN

Acute respiratory distress syndrome (ARDS) occurs as an acute onset condition, and patients present with diffuse alveolar damage, refractory hypoxemia, and non-cardiac pulmonary edema. ARDS progresses through an initial exudative phase, an inflammatory phase, and a final fibrotic phase. Pirfenidone, a powerful anti-fibrotic agent, is known as an agent that inhibits the progression of fibrosis in idiopathic pulmonary fibrosis. In this study, we studied the treatment efficiency of pirfenidone on lipopolysaccharide (LPS) and bleomycin-induced ARDS using rats. The ARDS rat model was created by the intratracheal administration of 3 mg/kg LPS of and 3 mg/kg of bleomycin dissolved in 0.2 mL of normal saline. The pirfenidone treatment group was administered 100 or 200 mg/kg of pirfenidone dissolved in 0.5 mL distilled water orally 10 times every 2 days for 20 days. The administration of LPS and bleomycin intratracheally increased lung injury scores and significantly produced pro-inflammatory cytokines. ARDS induction increased the expressions of transforming growth factor (TGF)-ß1/Smad-2 signaling factors. Additionally, matrix metalloproteinase (MMP)-9/tissue inhibitor of metalloproteinase (TIMP)-1 imbalance occurred, resulting in enhanced fibrosis-related factors. Treatment with pirfenidone strongly suppressed the expressions of TGF-ß1/Smad-2 signaling factors and improved the imbalance of MMP-9/TIMP-1 compared to the untreated group. These effects led to a decrease in fibrosis factors and pro-inflammatory cytokines, promoting the recovery of damaged lung tissue. These results of this study showed that pirfenidone administration suppressed inflammation and fibrosis in the ARDS animal model. Therefore, pirfenidone can be considered a new early treatment for ARDS.


Asunto(s)
Bleomicina , Lipopolisacáridos , Piridonas , Síndrome de Dificultad Respiratoria , Transducción de Señal , Animales , Piridonas/farmacología , Piridonas/uso terapéutico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/inducido químicamente , Transducción de Señal/efectos de los fármacos , Ratas , Masculino , Bleomicina/efectos adversos , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Proteína Smad2/metabolismo , Ratas Sprague-Dawley , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Modelos Animales de Enfermedad , Metaloproteinasa 9 de la Matriz/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Pulmón/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Proteínas Smad/metabolismo
2.
Int J Immunopathol Pharmacol ; 38: 3946320241274225, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39140804

RESUMEN

OBJECTIVES: Tuberostemonine has several biological activity, the aim of study examined the impact of tuberostemonine on the proliferation of TGF-ß1 induced cell model, and its ability to alleviate pulmonary fibrosis stimulated by bleomycin in mice. METHODS: In vitro, we assessed the effect of tuberostemonine (350, 550 and 750 µM) on the proliferation of cells stimulated by TGF-ß1 (10 µg/L), as well as on parameters such as α-SMA vitality, human fibronectin, collagen, and hydroxyproline levels in cells. In vivo, we analyzed inflammation, hydroxyproline, collagen activity and metabolomics in the lungs of mice. Additionally, a comprehensive investigation into the TGF-ß/smad signaling pathway was undertaken, targeting lung tissue as well as HFL cells. RESULTS: Within the confines of an in vitro setup, the tuberostemonine manifested a discerned IC50 of 1.9 mM. Furthermore, a significant reduction of over fifty percent was ascertained in the secretion levels of hydroxyproline, fibronectin, collagen type I, collagen type III and α-SMA. In vivo, tuberostemonine obviously improved the respiratory function percentage over 50% of animal model and decreased the hydroxyproline, lung inflammation and collagen deposition. A prominent decline in TGF-ß/smad pathway functioning was identified within both the internal and external cellular contexts. CONCLUSIONS: Tuberostemonine is considered as a modulator to alleviate fibrosis and may become a new renovation for pulmonary fibrosis.


Asunto(s)
Bleomicina , Proliferación Celular , Fibroblastos , Pulmón , Fibrosis Pulmonar , Transducción de Señal , Factor de Crecimiento Transformador beta1 , Animales , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Hidroxiprolina/metabolismo , Proteínas Smad/metabolismo , Ratones Endogámicos C57BL , Masculino , Línea Celular , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibronectinas/metabolismo , Actinas/metabolismo
3.
FASEB J ; 38(15): e23877, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39114961

RESUMEN

Transforming growth factor-ß (TGF-ß) is a pleiotropic cytokine that modulates a wide variety of cellular responses by regulating target gene expression. It principally transmits signals via receptor-activated transcription factors Smad2 and Smad3, which form trimeric complexes with Smad4 upon activation and regulate gene expression by binding to genomic DNA. Here, we examined the mechanisms by which TGF-ß regulates the transcription of target genes in a cell context-dependent manner by screening a double-stranded DNA oligonucleotide library for DNA sequences bound to endogenous activated Smad complexes. Screening was performed by cyclic amplification of selected targets (CASTing) using an anti-Smad2/3 antibody and nuclear extracts isolated from three cell lines (A549, HepG2, and HaCaT) stimulated with TGF-ß. The preference of the activated Smad complexes for conventional Smad-binding motifs such as Smad-binding element (SBE) and CAGA motifs was different in HepG2 than in the other two cell lines, which may indicate the distinct composition of the activated Smad complexes. Several transcription factor-binding motifs other than SBE or CAGA, including the Fos/Jun-binding motifs, were detected in the enriched sequences. Reporter assays using sequences containing these transcription factor-binding motifs together with Smad-binding motifs indicated that some of the motifs may be involved in cell type-dependent transcriptional activation by TGF-ß. The results suggest that the CASTing method is useful for elucidating the molecular basis of context-dependent Smad signaling.


Asunto(s)
ADN , Transducción de Señal , Factor de Crecimiento Transformador beta , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Células Hep G2 , ADN/metabolismo , Unión Proteica , Proteína smad3/metabolismo , Proteína Smad2/metabolismo , Células A549 , Células HaCaT , Proteínas Smad/metabolismo
4.
Zhongguo Zhong Yao Za Zhi ; 49(13): 3583-3590, 2024 Jul.
Artículo en Chino | MEDLINE | ID: mdl-39041130

RESUMEN

To investigate the effects of Luhong Yixin Granules on myocardial fibrosis in rats with heart failure and its possible mechanism, a total of 60 male Wistar rats were randomly divided into the control group, model group, and low-, medium-and high-dose Luhong Yixin Granules groups, with 12 rats in each group. Except for those in the control group, rats in the other groups were induced by intraperitoneal injection of doxorubicin(DOX) into a rat model. After the Luhong Yixin Granules were dissolved in the same amount of normal saline, they were given by gavage at low, medium and high doses(2.8, 5.6, 11.2 g·kg~(-1)·d~(-1)), and the control group and the model group were given the same amount of normal saline by gavage for 40 days. After the end of dosing, echocardiography was used to measure left ventricular ejection fraction(LVEF) and left ventricular fractional shortening(LVFS). Rat body weight(BW) and heart weight(HW) were calculated as HW/BW. Enzyme-linked immunosorbent assay was used to measure the levels of interleukin-6(IL-6), interleukin-17(IL-17), tumor necrosis factor-α(TNF-α), transforming growth factor-ß1(TGF-ß1), growth stimulation expressed gene 2 protein(ST2), N-terminal pro-B-type natriuretic peptide(NT-proBNP), galectin-3(Gal-3) and creatine kinase isoenzyme(CK-MB) in serum. Hematoxylin-eosin(HE) staining and Masson staining were used to observe the pathological morphology of myocardial tissue. Western blot and quantitative real-time polymerase chain reaction were used to detect the protein and mRNA expression levels of IL-6, IL-17, TNF-α, TGF-ß1, Smad3, Smad7, α-smooth muscle actin(α-SMA), and collagen Ⅰ(COL-Ⅰ), respectively. RESULTS:: showed that compared with those in the control group, LVEF, LVFS, and HW/BW in the model group were decreased(P<0.05), and the levels of IL-6, IL-17, TNF-α, TGF-ß1, ST2, NT-proBNP, Gal-3, and CK-MB were increased(P<0.05). HE staining showed inflammatory changes in myocardial tissue; Masson staining showed decreases in the cross-sectional area and ventricular cavity area of the heart, and myocardial fibrosis of varying degrees(P<0.05). The protein and mRNA expression of IL-6, IL-17, TNF-α, TGF-ß1, Smad3, α-SMA, and COL-Ⅰ were increased(P<0.05), and the protein and mRNA expression of Smad7 protein was decreased(P<0.01). Compared with those in the model group, LVEF, LVFS and HW/BW of the low-, medium-and high-dose Luhong Yixin Granules groups were increased(P<0.05), and the levels of IL-6, IL-17, TNF-α, TGF-ß1, ST2, NT-proBNP, Gal-3 and CK-MB were decreased(P<0.05). HE staining showed gradually reduced inflammatory changes of myocardial tissue, and Masson staining showed increased cross-sectional area and ventricular cavity area of the heart and decreased area of myocardial fibrosis(P<0.05). The protein and mRNA expression levels of IL-6, IL-17, TNF-α, TGF-ß1, Smad3, α-SMA, and COL-Ⅰ were decreased(P<0.05), while the protein and mRNA expression levels of Smad7 were increased(P<0.05). Luhong Yixin Granules may be of great value in the treatment of heart failure by regulating the TGF-ß1/Smads signaling pathway, inhibiting the expression of inflammation-related proteins, reducing the deposition of extracellular matrix, and alleviating myocardial fibrosis.


Asunto(s)
Medicamentos Herbarios Chinos , Fibrosis , Insuficiencia Cardíaca , Miocardio , Ratas Wistar , Transducción de Señal , Proteínas Smad , Factor de Crecimiento Transformador beta1 , Animales , Masculino , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/farmacología , Ratas , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Transducción de Señal/efectos de los fármacos , Miocardio/patología , Miocardio/metabolismo , Proteínas Smad/metabolismo , Proteínas Smad/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Humanos
5.
Zhongguo Zhong Yao Za Zhi ; 49(11): 3012-3020, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-39041161

RESUMEN

This study investigated the effects of modified Fangji Huangqi Decoction on the expression of proteins related to epithelial-mesenchymal transition(EMT) in a mouse model of unilateral ureteral obstruction( UUO) and in a rat renal tubular epithelial cell(NRK-52E) model of fibrosis induced by transforming growth factor ß1(TGF-ß1). It aims to decipher the molecular mechanism by which modified Fangji Huangqi Decoction alleviates renal interstitial fibrosis. C57/BL mice were subjected to UUO.After the surgery, the mice were treated with 0. 5-fold and 2-fold concentrations of modified Fangji Huangqi Decoction and fosinopril sodium(positive control) for 7 days. The interstitial collagen deposition in the kidney was assessed by Masson staining. Western blot and RT-qPCR were employed to determine the expression levels of TGF-ß1, phosphorylated Smad2/3(p-Smad2/3), Smad2/3, Snail,epithelial cadherin(E-cadherin), alpha smooth muscle actin(α-SMA), and vimentin. The NRK-52E cell model induced by TGF-ß1was treated with the serum samples collected from SD rats treated with different concentrations of modified Fangji Huangqi Decoction.The CCK-8 assay was employed to examine the effects of the serum samples on NRK-52E cell proliferation. The cell morphology in different groups was observed under a microscope. Furthermore, the modeled cells were treated with the serum containing 1-fold decoction. Western blot and RT-qPCR were then employed to measure the expression levels of p-Smad2/3, Smad2/3, Snail,E-cadherin, α-SMA, and vimentin in the cells. Under the same conditions, sh RNA was used to silence the Snail gene, and measurements were repeated before and after treatment with the serum containing 1-fold decoction. The results indicated that modified Fangji Huangqi Decoction alleviated the fibrotic injury in the mouse model of UUO and the fibrosis in the NRK-52E cell model. The treatment with the decoction down-regulated the protein and m RNA levels of EMT-related indicators including p-Smad2/3, α-SMA,Snail, and vimentin, while it up-regulated the expression of E-cadherin. After sh RNA silencing of the Snail gene, the protein and m RNA levels of E-cadherin, α-SMA, and vimentin showed no significant differences before and after treatment with the serum containing the decoction. The results suggest that modified Fangji Huangqi Decoction may alleviate renal interstitial fibrosis by inhibiting the TGF-ß1/Smad/Snail signaling pathway and regulating the EMT process.


Asunto(s)
Medicamentos Herbarios Chinos , Transición Epitelial-Mesenquimal , Fibrosis , Ratones Endogámicos C57BL , Transducción de Señal , Proteínas Smad , Factores de Transcripción de la Familia Snail , Factor de Crecimiento Transformador beta1 , Animales , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Ratones , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/administración & dosificación , Fibrosis/tratamiento farmacológico , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción de la Familia Snail/genética , Ratas , Transducción de Señal/efectos de los fármacos , Masculino , Proteínas Smad/metabolismo , Proteínas Smad/genética , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Enfermedades Renales/genética
6.
Anticancer Res ; 44(8): 3355-3364, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39060083

RESUMEN

BACKGROUND/AIM: As an antagonist of bone morphogenetic protein (BMP), Noggin facilitates osteolytic bone metastases from breast cancer. The present study aimed to further dissect its role in oestrogen receptor (ER) positive breast cancer. MATERIALS AND METHODS: Noggin expression in ER positive breast cancer cell lines (MCF-7 and T-47D) was determined under conditions of oestrogen deprivation and treatment with 17-ß-oestradiol (E2). Activation of Smad1/5/8 in the oestrogen-regulated Noggin was examined using recombinant human BMP7 (rhBMP7) and a BMP receptor inhibitor (LDN-193189). The influence of Noggin on cellular functions was evaluated in MCF-7 and T-47D cell lines. Responses to tamoxifen and chemotherapy drugs were determined in MCF-7 and T-47D cells with Noggin over-expression using MTT assay. RESULTS: Noggin expression was negatively correlated with ERα in breast cancers. Noggin was up-regulated upon oestrogen deprivation, an effect that was eliminated by E2 Furthermore, increased levels of phosphorylated Smad1/5/8 were observed in the oestrogen-deprived MCF-7 and T-47D cells, which was prevented by E2 and LDN-193189, respectively. BMP7-induced Noggin expression and activation of Smad1/5/8 was also prevented by E2 and LDN-193189. Noggin over-expression resulted in an increase in the proliferation of both MCF-7 and T-47D cells. MCF-7 and T-47D cells over-expressing Noggin exhibited a good tolerance to tamoxifen (TAM), DTX, and 5-FU, but the percentage of viable cells was higher compared with the controls. CONCLUSION: Noggin expression can be repressed by oestrogen through inference with the BMP/Smad signalling. Over-expression of Noggin promotes the proliferation of MCF-7 and T-47D cells, contributing to drug resistance.


Asunto(s)
Neoplasias de la Mama , Proteínas Portadoras , Estrógenos , Transducción de Señal , Proteínas Smad , Tamoxifeno , Humanos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Femenino , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Estrógenos/farmacología , Estrógenos/metabolismo , Células MCF-7 , Tamoxifeno/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteína Morfogenética Ósea 7/metabolismo , Proteína Morfogenética Ósea 7/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Receptor alfa de Estrógeno/metabolismo , Receptor alfa de Estrógeno/genética , Estradiol/farmacología
7.
Anal Cell Pathol (Amst) ; 2024: 2751280, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38946862

RESUMEN

Background: Biliary atresia (BA) is a devastating congenital disease characterized by inflammation and progressive liver fibrosis. Activation of hepatic stellate cells (HSCs) plays a central role in the pathogenesis of hepatic fibrosis. Our study aimed to investigate the pharmacological effect and potential mechanism of pirfenidone (PFD) and andrographolide (AGP) separately and together on liver fibrosis of BA. Materials and Methods: The bile ducts of male C57BL/6J mice were ligated or had the sham operation. The in vivo effects of PFD and/or AGP on liver fibrosis of BA were evaluated. Human hepatic stellate cells (LX-2) were also treated with PFD and/or AGP in vitro. Results: PFD and/or AGP ameliorates liver fibrosis and inflammation in the mice model of BA, as evidenced by significant downregulated in the accumulation of collagen fibers, hepatic fibrosis markers (α-SMA, collagen I, and collagen IV), and inflammatory markers (IL-1ß, IL-6, and TNF-α). Moreover, compared with monotherapy, these changes are more obvious in the combined treatment of PFD and AGP. Consistent with animal experiments, hepatic fibrosis markers (α-SMA, collagen I, and CTGF) and inflammatory markers (IL-1ß, IL-6, and TNF-α) were significantly decreased in activated LX-2 cells after PFD and/or AGP treatment. In addition, PFD and/or AGP inhibited the activation of HSCs by blocking the TGF-ß/Smad signaling pathway, and the combined treatment of PFD and AGP synergistically inhibited the phosphorylation of Smad2 and Smad3. Conclusion: The combined application of PFD and AGP exerted superior inhibitive effects on HSC activation and liver fibrosis by mediating the TGF-ß/Smad signaling pathway as compared to monotherapy. Therefore, the combination of PFD and AGP may be a promising treatment strategy for liver fibrosis in BA.


Asunto(s)
Diterpenos , Células Estrelladas Hepáticas , Cirrosis Hepática , Ratones Endogámicos C57BL , Piridonas , Transducción de Señal , Proteínas Smad , Factor de Crecimiento Transformador beta , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Animales , Cirrosis Hepática/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Transducción de Señal/efectos de los fármacos , Diterpenos/farmacología , Diterpenos/uso terapéutico , Masculino , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Smad/metabolismo , Humanos , Piridonas/farmacología , Línea Celular , Ratones , Atresia Biliar/patología , Atresia Biliar/tratamiento farmacológico , Atresia Biliar/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada
8.
J Orthop Surg Res ; 19(1): 386, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951811

RESUMEN

BACKGROUND: Bone defects, resulting from substantial bone loss that exceeds the natural self-healing capacity, pose significant challenges to current therapeutic approaches due to various limitations. In the quest for alternative therapeutic strategies, bone tissue engineering has emerged as a promising avenue. Notably, excretory proteins from Toxoplasma gondii (TgEP), recognized for their immunogenicity and broad spectrum of biological activities secreted or excreted during the parasite's lifecycle, have been identified as potential facilitators of osteogenic differentiation in human bone marrow mesenchymal stem cells (hBMSCs). Building on our previous findings that TgEP can enhance osteogenic differentiation, this study investigated the molecular mechanisms underlying this effect and assessed its therapeutic potential in vivo. METHODS: We determined the optimum concentration of TgEP through cell cytotoxicity and cell proliferation assays. Subsequently, hBMSCs were treated with the appropriate concentration of TgEP. We assessed osteogenic protein markers, including alkaline phosphatase (ALP), Runx2, and Osx, as well as components of the BMP/Smad signaling pathway using quantitative real-time PCR (qRT-PCR), siRNA interference of hBMSCs, Western blot analysis, and other methods. Furthermore, we created a bone defect model in Sprague-Dawley (SD) male rats and filled the defect areas with the GelMa hydrogel, with or without TgEP. Microcomputed tomography (micro-CT) was employed to analyze the bone parameters of defect sites. H&E, Masson and immunohistochemical staining were used to assess the repair conditions of the defect area. RESULTS: Our results indicate that TgEP promotes the expression of key osteogenic markers, including ALP, Runx2, and Osx, as well as the activation of Smad1, BMP2, and phosphorylated Smad1/5-crucial elements of the BMP/Smad signaling pathway. Furthermore, in vivo experiments using a bone defect model in rats demonstrated that TgEP markedly promoted bone defect repair. CONCLUSION: Our results provide compelling evidence that TgEP facilitates hBMSC osteogenic differentiation through the BMP/Smad signaling pathway, highlighting its potential as a therapeutic approach for bone tissue engineering for bone defect healing.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas , Osteogénesis , Ratas Sprague-Dawley , Transducción de Señal , Toxoplasma , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Humanos , Animales , Transducción de Señal/fisiología , Diferenciación Celular/fisiología , Masculino , Toxoplasma/fisiología , Ratas , Proteínas Smad/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas
9.
Matrix Biol ; 132: 87-97, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39019241

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) represents a severe and progressive manifestation of idiopathic interstitial pneumonia marked by an uncertain etiology along with an unfavorable prognosis. Osteoglycin (OGN), belonging to the small leucine-rich proteoglycans family, assumes pivotal functions in both tissue formation and damage response. However, the roles and potential mechanisms of OGN in the context of lung fibrosis remain unexplored. METHODS: The assessment of OGN expression levels in fibrotic lungs was conducted across various experimental lung fibrosis mouse models. To elucidate the effects of OGN on the differentiation of lung myofibroblasts, both OGN knockdown and OGN overexpression were employed in vitro. The expression of integrin αv, along with its colocalization with lysosomes and latency-associated peptide (LAP), was monitored in OGN-knockdown lung myofibroblasts. Furthermore, the role of OGN in lung fibrosis was investigated through OGN knockdown utilizing adeno-related virus serotype 6 (AAV6)-mediated delivery. RESULTS: OGN exhibited upregulation in both lungs and myofibroblasts across diverse lung fibrosis mouse models. And laboratory experiments in vitro demonstrated that OGN knockdown inhibited the TGF-ß/Smad signaling pathway in lung myofibroblasts. Conversely, OGN overexpression promoted TGF-ß/Smad pathway in these cells. Mechanistic insights revealed that OGN knockdown facilitated lysosome-mediated degradation of integrin αv while inhibiting its binding to latency-associated peptide (LAP). Remarkably, AAV6-targeted OGN knockdown ameliorated the extent of lung fibrosis in experimental mouse models. CONCLUSION: Our results indicate that inhibiting OGN signaling could serve as a promising therapeutic way for lung fibrosis.


Asunto(s)
Modelos Animales de Enfermedad , Fibrosis Pulmonar Idiopática , Integrina alfaV , Pulmón , Miofibroblastos , Transducción de Señal , Factor de Crecimiento Transformador beta , Animales , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ratones , Integrina alfaV/metabolismo , Integrina alfaV/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/genética , Pulmón/metabolismo , Pulmón/patología , Proteínas Smad/metabolismo , Proteínas Smad/genética , Humanos , Técnicas de Silenciamiento del Gen , Masculino , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología
10.
PLoS One ; 19(7): e0305927, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39024326

RESUMEN

Fibrotic skin diseases, such as keloids, are pathological results of aberrant tissue healing and are characterized by overgrowth of dermal fibroblasts. Remdesivir (RD), an antiviral drug, has been reported to have pharmacological activities in a wide range of fibrotic diseases. However, whether RD function on skin fibrosis remains unclear. Therefore, in our study, we explored the potential effect and mechanisms of RD on skin fibrosis both in vivo and in vitro. As expected, the results demonstrated that RD alleviated BLM-induced skin fibrosis and attenuates the gross weight of keloid tissues in vivo. Further studies suggested that RD suppressed fibroblast activation and autophagy both in vivo and in vitro. In addition, mechanistic research showed that RD attenuated fibroblasts activation by the TGF-ß1/Smad signaling pathway and inhibited fibroblasts autophagy by the PI3K/Akt/mTOR signaling pathway. In summary, our results demonstrate therapeutic potential of RD for skin fibrosis in the future.


Asunto(s)
Adenosina Monofosfato , Alanina , Fibroblastos , Fibrosis , Transducción de Señal , Piel , Factor de Crecimiento Transformador beta1 , Animales , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Fibrosis/tratamiento farmacológico , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/metabolismo , Ratones , Piel/efectos de los fármacos , Piel/patología , Piel/metabolismo , Humanos , Autofagia/efectos de los fármacos , Queloide/tratamiento farmacológico , Queloide/metabolismo , Queloide/patología , Antivirales/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Bleomicina , Fosfatidilinositol 3-Quinasas/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Smad/metabolismo
11.
Clin Exp Hypertens ; 46(1): 2380291, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39077772

RESUMEN

OBJECTIVE: This study investigated the expression of TGF-ß/Smad pathway-related indices in patients with isolated iliac artery aneurysms (IIAA) complicated with iliac arteriovenous fistula (IAVF) and their relationship with prognosis. METHODS: From January 2016 to June 2022, 83 patients with IIAA complicated with IAVF (Study group) and 54 patients with IIAA not complicated with IAVF (control group) were studied. The related indices of TGF-ß/Smad pathway were evaluated, and the effects of each index on the formation of IAVF were analyzed. The patients were divided into the survival group (64 cases) and death group (19 cases), and the prognostic value of indices in combination was analyzed. RESULTS: TGF-ß, p-Smad2, p-Smad3, p-JNK, and p-ERK in the study group were higher than those in the control group. Abnormal increase of pSmad3 expression was a risk factor for IAVF formation in patients with IIAA. TGF-ß level in the death group was higher than that in the survival group, and p-Smad3 and p-JNK proteins were higher than those in the survival group. The AUC value of indices in the TGF-ß/Smad pathway in combination was greater than that of each index alone. Abnormal increased expression of pSmad3 was a risk factor for prognosis of patients with IIAA complicated with IAVF. CONCLUSION: The abnormal increase of TGF-ß/Smad pathway-related indices is related to poor prognosis of patients with IIAA complicated with IAVF, and the combined detection of all indices has a predictive value for patients' prognosis.


Asunto(s)
Fístula Arteriovenosa , Aneurisma Ilíaco , Arteria Ilíaca , Factor de Crecimiento Transformador beta , Humanos , Masculino , Femenino , Persona de Mediana Edad , Pronóstico , Factor de Crecimiento Transformador beta/metabolismo , Fístula Arteriovenosa/complicaciones , Fístula Arteriovenosa/metabolismo , Anciano , Aneurisma Ilíaco/complicaciones , Transducción de Señal , Proteína smad3/metabolismo , Proteínas Smad/metabolismo , Proteína Smad2/metabolismo , Vena Ilíaca
12.
Front Biosci (Landmark Ed) ; 29(7): 241, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-39082347

RESUMEN

BACKGROUND: The involvement of gut microbiota in carcinogenesis has gradually been highlighted in past decades. Bacteria could play its role by the secretion of extracellular vesicles (EVs); however, interrelationship between bacterial EVs and hepatocellular carcinoma (HCC) development has not been investigated much. METHODS: Diethylnitrosamine (DEN) was utilized to produce HCC model in mice, of which fecal was collected for detecting Bifidobacterium longum (B.longum) with real-time polymerase chain reaction (PCR). EV isolated from B.longum (B.longum-EV) with ultracentrifugation were stained with PKH26 to investigate the cellular uptake of murine hepatocytes (AML12). After treatment with B.longum-EV, TGF-ß1-induced AML12 cells were subjected to morphological observation, fibrosis- and apoptosis-related marker detection with western blot, apoptotic ratio and reactive oxygen species (ROS) level analysis with flow cytometry, and oxidative stress biomarker assessment with enzyme-linked immunosorbent assay (ELISA); meanwhile, animal studies including liver function, tumor formation rate, and histological analysis, were also performed to investigate the role of B.longum-EV in the fibrosis, apoptosis, oxidative stress, and carcinogenesis of the liver in vivo. RESULTS: The levels of B.longum were significantly reduced in HCC model mice. B.longum-EV could enter AML12 cells and effectively attenuate TGF-ß1-induced fibrosis, apoptosis, and oxidative stress in AML12 cells. In vivo studies showed that B.longum-EV administration alleviated DEN-induced liver fibrosis, apoptosis, and oxidative stress at the early stage. Moreover, B.longum-EV administration also effectively reduced the tumor formation rate and liver function injury in DEN-induced mice and down-regulated TGF-ß1 expression and Smad3 phosphorylation of mouse liver. CONCLUSIONS: B.longum-EVs protect hepatocytes against fibrosis, apoptosis, and oxidative damage, which exert a potential of preventing HCC development.


Asunto(s)
Apoptosis , Bifidobacterium longum , Carcinoma Hepatocelular , Vesículas Extracelulares , Neoplasias Hepáticas , Transducción de Señal , Factor de Crecimiento Transformador beta1 , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Vesículas Extracelulares/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/prevención & control , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/inducido químicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/prevención & control , Neoplasias Hepáticas/patología , Ratones , Bifidobacterium longum/metabolismo , Apoptosis/efectos de los fármacos , Estrés Oxidativo , Masculino , Dietilnitrosamina/toxicidad , Proteínas Smad/metabolismo , Hepatocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Cirrosis Hepática/metabolismo , Cirrosis Hepática/prevención & control , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología
13.
PLoS One ; 19(6): e0304185, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38857261

RESUMEN

OBJECTIVE: The present study aims to investigate the specific protective effects and underlying mechanisms of Ganshuang granule (GSG) on dimethylnitrosamine (DMN)-induced hepatic fibrosis in rat models. METHODS: Hepatic fibrosis was experimentally evoked in rats by DMN administration, and varying dosages of GSG were employed as an intervention. Hepatocellular damage was assessed by measuring serum levels of aminotransferase and bilirubin, accompanied by histopathological examinations of hepatic tissue. The hepatic concentrations of platelet-derived growth factor (PDGF) and transforming growth factor-ß1 (TGF-ß1) were quantitated via enzyme-linked immunosorbent assay (ELISA). The expression of α-smooth muscle actin (α-SMA) within hepatic tissue was evaluated using immunohistochemical techniques. The levels of hepatic interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and a spectrum of interleukins (IL-2, IL-4, IL-6, IL-10) were quantified by quantitative real-time PCR (qRT-PCR). Additionally, hepatic stellate cells (HSCs) were cultured in vitro and exposed to TNF-α in the presence of naringin, a principal component of GSG. The gene expression levels of tissue inhibitor of metalloproteinase-1 (TIMP-1) and matrix metallopeptidase-1 (MMP-1) in these cells were also quantified by qRT-PCR. Proliferative activity of HSCs was evaluated by the Cell Counting Kit-8 assay. Finally, alterations in Smad protein expression were analyzed through Western blotting. RESULTS: Administration of GSG in rats with fibrosis resulted in reduced levels of serum aminotransferases and bilirubin, along with alleviation of histopathological liver injury. Furthermore, the fibrosis rats treated with GSG exhibited significant downregulation of hepatic TGF-ß1, PDGF, and TNF-α levels. Additionally, GSG treatment led to increased mRNA levels of IFN-γ, IL-2, and IL-4, as well as decreased expression of α-SMA in the liver. Furthermore, treatment with naringin, a pivotal extract of GSG, resulted in elevated expression of MMP-1 and decreased levels of TIMP-1 in TNF-α-stimulated HSCs when compared to the control group. Additionally, naringin administration led to a reduction in Smad expression within the HSCs. CONCLUSION: GSG has the potential to mitigate fibrosis induced by DMN in rat models through the regulation of inflammatory and fibrosis factors. Notably, naringin, the primary extract of GSG, may exert a pivotal role in modulating the TGF-ß-Smad signaling pathway.


Asunto(s)
Medicamentos Herbarios Chinos , Flavanonas , Células Estrelladas Hepáticas , Cirrosis Hepática , Transducción de Señal , Proteínas Smad , Animales , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/inducido químicamente , Transducción de Señal/efectos de los fármacos , Flavanonas/farmacología , Flavanonas/uso terapéutico , Masculino , Ratas , Proteínas Smad/metabolismo , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Ratas Sprague-Dawley , Dimetilnitrosamina , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Actinas/metabolismo
14.
J Orthop Surg Res ; 19(1): 329, 2024 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-38825706

RESUMEN

BACKGROUND: Fibrosis is a significant pathological feature of chronic skeletal muscle injury, profoundly affecting muscle regeneration. Fibro-adipogenic progenitors (FAPs) have the ability to differentiate into myofibroblasts, acting as a primary source of extracellular matrix (ECM). the process by which FAPs differentiate into myofibroblasts during chronic skeletal muscle injury remains inadequately explored. METHOD: mouse model with sciatic nerve denervated was constructed and miRNA expression profiles between the mouse model and uninjured mouse were analyzed. qRT/PCR and immunofluorescence elucidated the effect of miR-27b-3p on fibrosis in vivo and in vitro. Dual-luciferase reporter identified the target gene of miR-27b-3p, and finally knocked down or overexpressed the target gene and phosphorylation inhibition of Smad verified the influence of downstream molecules on the abundance of miR-27b-3p and fibrogenic differentiation of FAPs. RESULT: FAPs derived from a mouse model with sciatic nerves denervated exhibited a progressively worsening fibrotic phenotype over time. Introducing agomiR-27b-3p effectively suppressed fibrosis both in vitro and in vivo. MiR-27b-3p targeted Transforming Growth Factor Beta Receptor 1 (TGF-ßR1) and the abundance of miR-27b-3p was negatively regulated by TGF-ßR1/Smad. CONCLUSION: miR-27b-3p targeting the TGF-ßR1/Smad pathway is a novel mechanism for regulating fibrogenic differentiation of FAPs. Increasing abundance of miR-27b-3p, suppressing expression of TGF-ßR1 and inhibiting phosphorylation of smad3 presented potential strategies for treating fibrosis in chronic skeletal muscle injury.


Asunto(s)
Fibrosis , MicroARNs , Músculo Esquelético , Transducción de Señal , Animales , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ratones , Enfermedad Crónica , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Ratones Endogámicos C57BL , Proteínas Smad/metabolismo , Proteínas Smad/genética , Masculino , Modelos Animales de Enfermedad , Diferenciación Celular , Nervio Ciático/lesiones
15.
Mol Med Rep ; 30(2)2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38904198

RESUMEN

The TGF­ß/Smad signaling pathway plays a pivotal role in the onset of glomerular and tubulointerstitial fibrosis in chronic kidney disease (CKD). The present review delves into the intricate post­translational modulation of this pathway and its implications in CKD. Specifically, the impact of the TGF­ß/Smad pathway on various biological processes was investigated, encompassing not only renal tubular epithelial cell apoptosis, inflammation, myofibroblast activation and cellular aging, but also its role in autophagy. Various post­translational modifications (PTMs), including phosphorylation and ubiquitination, play a crucial role in modulating the intensity and persistence of the TGF­ß/Smad signaling pathway. They also dictate the functionality, stability and interactions of the TGF­ß/Smad components. The present review sheds light on recent findings regarding the impact of PTMs on TGF­ß receptors and Smads within the CKD landscape. In summary, a deeper insight into the post­translational intricacies of TGF­ß/Smad signaling offers avenues for innovative therapeutic interventions to mitigate CKD progression. Ongoing research in this domain holds the potential to unveil powerful antifibrotic treatments, aiming to preserve renal integrity and function in patients with CKD.


Asunto(s)
Procesamiento Proteico-Postraduccional , Insuficiencia Renal Crónica , Transducción de Señal , Proteínas Smad , Factor de Crecimiento Transformador beta , Humanos , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Smad/metabolismo , Animales , Fosforilación , Fibrosis , Ubiquitinación , Autofagia
16.
Biomed Pharmacother ; 176: 116823, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38834008

RESUMEN

Ancient Egyptians (including Bedouins and Nubians) have long utilized Ziziphus spina-christi (L.), a traditional Arabian medicinal herb, to alleviate swellings and inflammatory disorders. It is also mentioned in Christian and Muslim traditions. Ziziphus spina-christi L. (Family: Rhamnaceae) is a plentiful source of polyphenols, revealing free radical scavenging, antioxidant, metal chelating, cytotoxic, and anti-inflammatory activities. Herein, different classes of the existing bioactive metabolites in Z. spina-christi L. were detected using liquid chromatography-tandem mass spectrometry (LC-MS/MS) for the first time. The study also aimed to assess the anti-inflammatory and antifibrotic properties of Z. spina-christi L. extract against bleomycin-induced lung fibrosis in an experimental mouse model. 32 male Swiss Albino mice were assigned into 4 groups; the first and second were the normal control group and the bleomycin positive control (single 2.5 U/kg bleomycin intratracheal dose). The third and fourth groups received 100 and 200 mg/kg/day Z. spina-christi L. extract orally for 3 weeks, 2 weeks before bleomycin, and 1 week after. The bioactive metabolites in Z. spina-christi L. extract were identified as phenolic acids, catechins, flavonoids, chalcones, stilbenes, triterpenoid acids, saponins, and sterols. The contents of total phenolic compounds and flavonoids were found to be 196.62 mg GAE/gm and 33.29 mg QE/gm, respectively. In the experimental study, histopathological examination revealed that lung fibrosis was attenuated in both Z. spina-christi L.- treated groups. Z. spina-christi L. extract downregulated the expression of nuclear factor kappa B (NF-κB) p65 and decreased levels of the inflammatory markers tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and c-Jun N-terminal kinase (JNK) in lung tissue. Z. spina-christi L. also downregulated the expression of the fibrotic parameters collagen-1, alpha-smooth muscle actin (α-SMA), transforming growth factor-beta 1 (TGF-ß1), matrix metalloproteinase-9 (MMP-9) and SMAD3, with upregulation of the antifibrotic SMAD7 in lung tissue. Overall, the present study suggests a potential protective effect of Z. spina-christi L. extract against bleomycin-induced lung fibrosis through regulation of the TGF-ß1/SMAD pathway.


Asunto(s)
Bleomicina , Extractos Vegetales , Fibrosis Pulmonar , Transducción de Señal , Proteínas Smad , Espectrometría de Masas en Tándem , Factor de Crecimiento Transformador beta1 , Ziziphus , Animales , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Masculino , Ziziphus/química , Ratones , Extractos Vegetales/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Smad/metabolismo , Espectrometría de Masas en Tándem/métodos , Cromatografía Liquida/métodos , Transducción de Señal/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Metabolómica/métodos , Antiinflamatorios/farmacología , Cromatografía Líquida con Espectrometría de Masas
17.
Eur J Pharmacol ; 977: 176708, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38843945

RESUMEN

Excessive transforming growth factor ß1 (TGF-ß1) secreted by activated hepatic stellate cells (aHSCs) aggravates liver fibrosis via over-activation of TGF-ß1-mediated signaling pathways in a TGF-ß type I receptor (TßRI) dependent manner. TßRI with the C-terminal valine truncated (RIPΔ), as a novel TßRI-mimicking peptide, is an appealing anti-fibrotic candidate by competitive binding of TGF-ß1 to block TGF-ß1 signal transduction. Platelet-derived growth factor receptor ß (PDGFßR) is highly expressed on the surface of aHSCs in liver fibrosis. Herein, we designed a novel RIPΔ variant Z-RIPΔ (PDGFßR-specific affibody ZPDGFßR fused to the N-terminus of RIPΔ) for liver fibrosis therapy, and expect to improve the anti-liver fibrosis efficacy by specifically inhibiting the TGF-ß1 activity in aHSCs. Target peptide Z-RIPΔ was prepared in Escherichia coli by SUMO fusion system. Moreover, Z-RIPΔ specifically bound to TGF-ß1-activated aHSCs, inhibited cell proliferation and migration, and reduced the expression of fibrosis markers (α-SMA and FN) and TGF-ß1 pathway-related effectors (p-Smad2/3 and p-p38) in vitro. Furthermore, Z-RIPΔ specifically targeted the fibrotic liver, alleviated the liver histopathology, mitigated the fibrosis responses, and blocked TGF-ß1-mediated Smad and p38 MAPK cascades. More importantly, Z-RIPΔ exhibited a higher fibrotic liver-targeting capacity and stronger anti-fibrotic effects than its parent RIPΔ. Besides, Z-RIPΔ showed no obvious toxicity effects in treating both an in vitro cell model and an in vivo mouse model of liver fibrosis. In conclusion, Z-RIPΔ represents a promising targeted candidate for liver fibrosis therapy.


Asunto(s)
Células Estrelladas Hepáticas , Cirrosis Hepática , Receptor Tipo I de Factor de Crecimiento Transformador beta , Transducción de Señal , Proteínas Smad , Factor de Crecimiento Transformador beta1 , Proteínas Quinasas p38 Activadas por Mitógenos , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Cirrosis Hepática/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Ratones , Proteínas Smad/metabolismo , Masculino , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Péptidos/farmacología , Péptidos/química , Humanos , Ratones Endogámicos C57BL
18.
J Pharm Pharmacol ; 76(8): 1038-1050, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-38848454

RESUMEN

OBJECTIVE: Breast cancer is a malignant tumor with high invasion and metastasis. TGF-ß1-induced epithelial-mesenchymal transition (EMT) is crucially involved in the growth and metastasis of breast cancer. Wedelolactone (Wed) is extracted from herbal medicine Ecliptae Herba, which is reported to have antineoplastic activity. Here, we aimed to elucidate the efficacy and mechanism of Wed against breast cancer. METHODS: The effects of Wed on migration and invasion of 4T1 were detected. The expression of EMT-related markers was detected by Western blot and qPCR. The 4T1 orthotopic murine breast cancer model was established to evaluate the therapeutic effect of Wed on the growth and metastasis of breast cancer through TGF-ß1/Smad pathway. RESULTS: Wed inhibited the proliferation, migration and invasion of 4T1. It exhibited concentration-dependent inhibition of p-Smad2/3. Wed also reversed the expression of EMT-markers induced by TGF-ß1. In addition, Wed suppressed the growth and metastasis of breast cancer in mice. It also affected p-Smad3 expression as well as EMT-related genes, suggesting that its anti-breast cancer effect may be related to the TGF-ß1/Smad pathway. CONCLUSION: Wed reverses EMT by regulating TGF-ß1/Smad pathway, potentially serving as a therapeutic agent for breast cancer. Wed is expected to be a potential drug to inhibit TGF-ß1/Smad pathway-related diseases.


Asunto(s)
Neoplasias de la Mama , Movimiento Celular , Proliferación Celular , Cumarinas , Transición Epitelial-Mesenquimal , Ratones Endogámicos BALB C , Transducción de Señal , Factor de Crecimiento Transformador beta1 , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Femenino , Transducción de Señal/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Ratones , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Cumarinas/farmacología , Cumarinas/uso terapéutico , Antineoplásicos Fitogénicos/farmacología , Proteínas Smad/metabolismo , Metástasis de la Neoplasia , Proteína smad3/metabolismo , Invasividad Neoplásica , Humanos , Proteína Smad2/metabolismo
19.
Int Immunopharmacol ; 137: 112483, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38880023

RESUMEN

Renal fibrosis is a representative pathological feature of various chronic kidney diseases, and efficient treatment is needed. Interstitial myofibroblasts are a key driver of kidney fibrosis, which is dependent on the binding of TGF-ß1 to type I TGF-ß receptor (TßRI) and TGF-ß1-related signaling pathways. Therefore, attenuating TGF-ß1 activity by competing with TGF-ß1 in myofibroblasts is an ideal strategy for treating kidney fibrosis. Recently, a novel TßRI-mimicking peptide RIPΔ demonstrated a high affinity for TGF-ß1. Thus, it could be speculated that RIPΔ may be used for anti-fibrosis therapy. Platelet-derived growth factor ß receptor (PDGFßR) is highly expressed in fibrotic kidney. In this study, we found that target peptide Z-RIPΔ, which is RIPΔ modified with PDGFßR-specific affibody ZPDGFßR, was specifically and highly taken up by TGF-ß1-activated NIH3T3 fibroblasts. Moreover, Z-RIPΔ effectively inhibited the myofibroblast proliferation, migration and fibrosis response in vitro. In vivo and ex vivo experiments showed that Z-RIPΔ specifically targeted fibrotic kidney, improved the damaged renal function, and ameliorated kidney histopathology and renal fibrosis in UUO mice. Mechanistic studies showed that Z-RIPΔ hold the stronger inhibition of the TGF-ß1/Smad and TGF-ß1/p38 pathways than unmodified RIPΔ in vitro and in vivo. Furthermore, systemic administration of Z-RIPΔ to UUO mice led to minimal toxicity to major organs. Taken together, RIPΔ modified with ZPDGFßR increased its therapeutic efficacy and reduced its systemic toxicity, making it a potential candidate for targeted therapy for kidney fibrosis.


Asunto(s)
Fibrosis , Riñón , Ratones Endogámicos C57BL , Proteínas Smad , Factor de Crecimiento Transformador beta1 , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Fibrosis/tratamiento farmacológico , Ratones , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Riñón/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Células 3T3 NIH , Masculino , Proteínas Smad/metabolismo , Transducción de Señal/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Péptidos/uso terapéutico , Péptidos/farmacología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Enfermedades Renales/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Humanos , Modelos Animales de Enfermedad , Proliferación Celular/efectos de los fármacos
20.
Toxicol Appl Pharmacol ; 489: 117012, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38906511

RESUMEN

Keloid formation has been linked to abnormal fibroblast function, such as excessive proliferation and extracellular matrix (ECM) production. Serum deprivation protein response (SDPR) is a crucial regulator of cellular function under diverse pathological conditions, yet its role in keloid formation remains unknown. The current work investigated the function of SDPR in regulating the proliferation, motility, and ECM production of keloid fibroblasts (KFs), as well as to decipher the mechanisms involved. Analysis of RNA sequencing data from the GEO database demonstrated significant down-regulation of SDPR in KF compared to normal fibroblasts (NFs). This down-regulation was also observed in clinical keloid specimens and isolated KFs. Overexpression of SDPR suppressed the proliferation, motility, and ECM production of KFs, while depletion of SDPR exacerbated the enhancing impact of TGF-ß1 on the proliferation, motility, and ECM production of NFs. Mechanistic studies revealed that SDPR overexpression repressed TGF-ß/Smad signal cascade activation in KFs along with decreased levels of phosphorylated Samd2/3, while SDPR depletion exacerbated TGF-ß/Smad activation in TGF-ß1-stimulated NFs. SDPR overexpression also repressed ERK1/2 activation in KFs, while SDPR depletion exacerbated ERK1/2 activation in TGF-ß1-stimulated NFs. Inhibition of ERK1/2 abolished SDPR-depletion-induced TGF-ß1/Smad activation, cell proliferation, motility, and ECM production in NFs. In conclusion, SDPR represses the proliferation, motility, and ECM production in KFs by blocking the TGF-ß1/Smad pathway in an ERK1/2-dependent manner. The findings highlight the role of SDPR in regulating abnormal behaviors of fibroblasts associated with keloid formation and suggest it as a potential target for anti-keloid therapy development.


Asunto(s)
Movimiento Celular , Proliferación Celular , Matriz Extracelular , Fibroblastos , Queloide , Sistema de Señalización de MAP Quinasas , Proteínas Smad , Factor de Crecimiento Transformador beta1 , Humanos , Queloide/patología , Queloide/metabolismo , Queloide/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Proteínas Smad/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Transducción de Señal , Células Cultivadas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Masculino , Femenino , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Adulto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...