Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Publication year range
1.
Pestic Biochem Physiol ; 200: 105831, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38582594

ABSTRACT

Paraquat (PQ) causes fatal poisoning that leads to systemic multiple organ fibrosis, and transforming growth factor (TGF)-ß1 plays a critical role in this process. In this study, we aimed to investigate the effects of AZ12601011 (a small molecular inhibitor of TGFßRI) on PQ-induced multiple organ fibrosis. We established a mouse model of PQ in vivo and used PQ-treated lung epithelial cell (A549) and renal tubular epithelial cells (TECs) in vitro. Haematoxylin-eosin and Masson staining revealed that AZ12601011 ameliorated pulmonary, hepatic, and renal fibrosis, consistent with the decrease in the levels of fibrotic indicators, alpha-smooth muscle actin (α-SMA) and collagen-1, in the lungs and kidneys of PQ-treated mice. In vitro data showed that AZ12601011 suppressed the induction of α-SMA and collagen-1 in PQ-treated A549 cells and TECs. In addition, AZ12601011 inhibited the release of inflammatory factors, interleukin (IL)-1ß, IL-6, and tumour necrosis factor-α. Mechanistically, TGF-ß and TGFßRI levels were significantly upregulated in the lungs and kidneys of PQ-treated mice. Cellular thermal shift assay and western blotting revealed that AZ12601011 directly bound with TGFßRI and blocked the activation of Smad3 downstream. In conclusion, our findings revealed that AZ12601011 attenuated PQ-induced multiple organ fibrosis by blocking the TGF-ß/Smad3 signalling pathway, suggesting its potential for PQ poisoning treatment.


Subject(s)
Acute Lung Injury , Paraquat , Pulmonary Fibrosis , Mice , Animals , Paraquat/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Receptor, Transforming Growth Factor-beta Type I , Transforming Growth Factor beta/toxicity , Transforming Growth Factor beta1/toxicity , Transforming Growth Factor beta1/metabolism , Collagen/toxicity , Collagen/metabolism , Transforming Growth Factors/toxicity
2.
J Neurosci ; 41(5): 947-959, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33298536

ABSTRACT

Long-term memory (LTM) formation is a critical survival process by which an animal retains information about prior experiences to guide future behavior. In the experimentally advantageous marine mollusk Aplysia, LTM for sensitization can be induced by the presentation of two aversive shocks to the animal's tail. Each of these training trials recruits distinct growth factor signaling systems that promote LTM formation. Specifically, whereas intact TrkB signaling during Trial 1 promotes an initial and transient increase of the immediate early gene apc/ebp mRNA, a prolonged increase in apc/ebp gene expression required for LTM formation requires the addition of TGFß signaling during Trial 2. Here we explored the molecular mechanisms by which Trial 2 achieves the essential prolonged gene expression of apc/ebp We find that this prolonged gene expression is not dependent on de novo transcription, but that apc/ebp mRNA synthesized by Trial 1 is post-transcriptionally stabilized by interacting with the RNA-binding protein ApELAV. This interaction is promoted by p38 MAPK activation initiated by TGFß. We further demonstrate that blocking the interaction of ApELAV with its target mRNA during Trial 2 blocks both the prolonged increase in apc/ebp gene expression and the behavioral induction of LTM. Collectively, our findings elucidate both when and how ELAV proteins are recruited for the stabilization of mRNA in LTM formation. Stabilization of a transiently expressed immediate early gene mRNA by a repeated training trial may therefore serve as a "filter" for learning, permitting only specific events to cause lasting transcriptional changes and behavioral LTM.SIGNIFICANCE STATEMENT: In the present paper, we significantly extend the general field of molecular processing in long-term memory (LTM) by describing a novel form of pretranslational processing required for LTM, which relies on the stabilization of a newly synthesized mRNA by a class of RNA binding proteins (ELAVs). There are now compelling data showing that important processing can occur after transcription of a gene, but before translation of the message into protein. Although the potential importance of ELAV proteins in LTM formation has previously been reported, the specific actions of ELAV proteins during LTM formation remained to be understood. Our new findings thus complement and extend this literature by demonstrating when and how this post-transcriptional gene regulation is mediated in the induction of LTM.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , ELAV Proteins/metabolism , Memory, Long-Term/physiology , RNA, Messenger/metabolism , Animals , Aplysia , CCAAT-Enhancer-Binding Protein-beta/genetics , ELAV Proteins/genetics , Memory, Long-Term/drug effects , Protein Binding/physiology , RNA, Messenger/genetics , Transforming Growth Factor beta1/toxicity
3.
Am J Physiol Renal Physiol ; 321(4): F517-F526, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34486400

ABSTRACT

Increasing evidence shows that long noncoding RNAs (lncRNAs) play an important role in kidney disease. In this study, we investigated the role of the lncRNA growth arrest-specific 5 (GAS5) in the pathogenesis of renal fibrosis. We found that GAS5 was markedly decreased in the fibrotic kidney of a unilateral ureteral obstructive nephropathy mouse model. In addition, GAS5 was expressed in mouse tubular epithelial cells (mTECs) and interstitial fibroblasts in normal renal tissue and was especially highly expressed in the cytoplasm. In vitro experiments showed that GAS5 was downregulated by transforming growth factor-ß1 (TGF-ß1) in a dose- and time-dependent manner. Overexpression of GAS5 blocked TGF-ß1-induced collagen type I and fibronectin expression and vice versa. Mechanistic experiments revealed that Smad3 but not Smad2 drove the regulation of GAS5. More importantly, GAS5 interacted with miR-142-5p and was involved in the renoprotective effect by participating in the competing endogenous RNA network. Finally, we also found that knockdown of GAS5 promoted TGF-ß1-induced mouse tubular epithelial cell apoptosis via the Smad3 pathway. Taken together, our results uncovered a lncRNA/miRNA competing endogenous RNA network-based mechanism that modulates extracellular matrix formation and cell apoptosis via the Smad3 pathway.NEW & NOTEWORTHY In this work, we mainly discuss long noncoding RNA growth arrest-specific 5 (GAS5), acting in a renoprotective role via the Smad3/miRNA-142-5p axis, that modulates extracellular matrix formation and cell apoptosis. Overexpression of GAS5 effectively blocked renal fibrosis in vitro. This study reveals that GAS5 may represent as a novel and precision therapeutic target for alleviating renal fibrosis.


Subject(s)
Epithelial Cells/drug effects , Kidney Diseases/prevention & control , Kidney Tubules/drug effects , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/toxicity , Animals , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Humans , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Mice, Inbred C57BL , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Signal Transduction , Smad3 Protein/genetics , Ureteral Obstruction/complications
4.
Respir Res ; 22(1): 51, 2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33579280

ABSTRACT

BACKGROUND: Little is known on the role of transient receptor potential ankyrin 1 (TRPA1) in fibroblast-myofibroblast transition (FMT) that can lead to airway remodeling which is a major problem for severe asthma and fibrosis. Thus, this study investigated the effect of TRPA1 modulators on transforming growth factor beta 1(TGF-ß1) -treated lung fibroblasts. METHODS: MRC-5 cells were preincubated with TGF-ß1 for 24 h. TRPA1 agonist or antagonist were added and further incubated for 24 h. The changes in TRPA1 and alpha-smooth muscle actin (α-SMA) expressions by stimuli were evaluated using qRT-PCR, western blot and immunohistochemical analyses. Statistical significance was determined by using one- or two-way ANOVA, followed by Bonferroni's post hoc analysis for comparison of multiple groups and paired 2-tailed Student's t-test between 2 groups. RESULTS: MRC-5 cells treated by TGF-ß1 significantly upregulated α-SMA mRNA expressions (P < 0.01), but downregulated TRPA1 gene expression (P < 0.001). Post-treatment of TRPA1 activator, allyl isothiocyanate (AITC), after TGF-ß1 significantly downregulated the α-SMA gene induction (P < 0.01 at 24 h), protein expression (P < 0.05) and immunoreactivity with stress fibers (P < 0.05). On the other hand, TRPA1 antagonist HC-030031 did not prevent this effect, and instead tended to facilitate the suppressive effect of AITC when co-stimulated. AITC significantly increased phosphorylated- extracellular signal-regulated kinase (ERK) 1/2 and heme oxygenase (HO)-1 protein expressions (P < 0.05) in TGF-ß1-treated cells. Combined inhibition with ERK1/2 mitogen-activated protein kinase (MAPK) and nuclear factor erythroid 2-related factor (NRF2) almost completely reversed AITC-induced α-SMA suppression (P < 0.05). Dexamethasone was not able to inhibit the upregulated α-SMA induction by TGF-ß1. However, AITC improved dexamethasone-insensitive myodifferentiation in the presence of the corticosteroid (P < 0.01). CONCLUSION: We found that AITC exerts protective effect on TGF-ß1-induced α-SMA induction by activating ERK1/2 MAPK and NRF2/HO-1 pathways in lung fibroblasts. It also overcomes corticosteroids insensitivity in TGF-ß1-induced α-SMA induction. TRPA1 antagonist modulates the suppressive effect, but not prevent it. AITC and TRPA1 antagonist may be therapeutic agents in treating chronic respiratory diseases.


Subject(s)
Adrenal Cortex Hormones/toxicity , Heme Oxygenase-1/metabolism , Isothiocyanates/pharmacology , Lung/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-E2-Related Factor 2/metabolism , TRPA1 Cation Channel/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung/drug effects , Myofibroblasts/drug effects , Myofibroblasts/metabolism , TRPA1 Cation Channel/antagonists & inhibitors , Transforming Growth Factor beta1/toxicity
5.
Respir Res ; 21(1): 256, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33032603

ABSTRACT

Glucocorticoids (GCs) and ß2-adrenergic receptor (ß2AR) agonists improve asthma outcomes in most patients. GCs also modulate gene expression in human airway smooth muscle (HASM), thereby attenuating airway inflammation and airway hyperresponsiveness that define asthma. Our previous studies showed that the pro-fibrotic cytokine, transforming growth factor- ß1 (TGF-ß1) increases phosphodiesterase 4D (PDE4D) expression that attenuates agonist-induced levels of intracellular cAMP. Decreased cAMP levels then diminishes ß2 agonist-induced airway relaxation. In the current study, we investigated whether glucocorticoids reverse TGF-ß1-effects on ß2-agonist-induced bronchodilation and modulate pde4d gene expression in HASM. Dexamethasone (DEX) reversed TGF-ß1 effects on cAMP levels induced by isoproterenol (ISO). TGF-ß1 also attenuated G protein-dependent responses to cholera toxin (CTX), a Gαs stimulator downstream from the ß2AR receptor. Previously, we demonstrated that TGF-ß1 treatment increased ß2AR phosphorylation to induce hyporesponsiveness to a ß2 agonist. Our current data shows that expression of grk2/3, kinases associated with attenuation of ß2AR function, are not altered with TGF-ß1 stimulation. Interestingly, DEX also attenuated TGF-ß1-induced pde4d gene expression. These data suggest that steroids may be an effective therapy for treatment of asthma patients whose disease is primarily driven by elevated TGF-ß1 levels.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/biosynthesis , Dexamethasone/pharmacology , Myocytes, Smooth Muscle/metabolism , Receptors, Adrenergic, beta-2/metabolism , Respiratory Mucosa/metabolism , Transforming Growth Factor beta1/toxicity , Anti-Inflammatory Agents/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic , Humans , Myocytes, Smooth Muscle/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Respiratory Mucosa/drug effects , Trachea/chemistry , Trachea/drug effects , Trachea/metabolism
6.
Respir Res ; 21(1): 108, 2020 May 07.
Article in English | MEDLINE | ID: mdl-32381012

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a rapidly progressing disease with challenging management. To find novel effective therapies, better preclinical models are needed for the screening of anti-fibrotic compounds. Activated fibroblasts drive fibrogenesis and are the main cells responsible for the accumulation of extracellular matrix (ECM). Here, a prolonged Scar-in-a-Jar assay was combined with clinically validated biochemical markers of ECM synthesis to evaluate ECM synthesis over time. To validate the model as a drug screening tool for novel anti-fibrotic compounds, two approved compounds for IPF, nintedanib and pirfenidone, and a compound in development, omipalisib, were tested. METHODS: Primary human lung fibroblasts from healthy donors were cultured for 12 days in the presence of ficoll and were stimulated with TGF-ß1 with or without treatment with an ALK5/TGF-ß1 receptor kinase inhibitor (ALK5i), nintedanib, pirfenidone or the mTOR/PI3K inhibitor omipalisib (GSK2126458). Biomarkers of ECM synthesis were evaluated over time in cell supernatants using ELISAs to assess type I, III, IV, V and VI collagen formation (PRO-C1, PRO-C3, PRO-C4, PRO-C5, PRO-C6), fibronectin (FBN-C) deposition and α-smooth muscle actin (α-SMA) expression. RESULTS: TGF-ß1 induced synthesis of PRO-C1, PRO-C6 and FBN-C as compared with unstimulated fibroblasts at all timepoints, while PRO-C3 and α-SMA levels were not elevated until day 8. Elevated biomarkers were reduced by suppressing TGF-ß1 signalling with ALK5i. Nintedanib and omipalisib were able to reduce all biomarkers induced by TGF-ß1 in a concentration dependent manner, while pirfenidone had no effect on α-SMA. CONCLUSIONS: TGF-ß1 stimulated synthesis of type I, III and VI collagen, fibronectin and α-SMA but not type IV or V collagen. Synthesis was increased over time, although temporal profiles differed, and was modulated pharmacologically by ALK5i, nintedanib, pirfenidone and omipalisib. This prolonged 12-day Scar-in-a-Jar assay utilising biochemical markers of ECM synthesis provides a useful screening tool for novel anti-fibrotic compounds.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cicatrix/chemically induced , Cicatrix/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Protein Kinase Inhibitors/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/metabolism , Cells, Cultured , Cicatrix/drug therapy , Collagen/antagonists & inhibitors , Collagen/metabolism , Drug Evaluation, Preclinical/methods , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Fibrosis/chemically induced , Fibrosis/drug therapy , Fibrosis/metabolism , Humans , Indoles/antagonists & inhibitors , Indoles/metabolism , Protein Kinase Inhibitors/therapeutic use , Pyridones/antagonists & inhibitors , Pyridones/metabolism , Transforming Growth Factor beta1/toxicity
7.
Clin Exp Pharmacol Physiol ; 47(6): 940-946, 2020 06.
Article in English | MEDLINE | ID: mdl-32012313

ABSTRACT

Renal fibrosis compromises kidney function, and it is a risk factor for chronic kidney disease (CKD). CKD ultimately progresses to end-stage kidney disease that can be cured only by kidney transplantation. Owing to the increasing number of CKD patients, effective treatment strategies are urgently required for renal fibrosis. TGF-ß is a well-established fibrogenic factor that signals through SMAD2/3 signaling pathway. It was shown that there is a cross-talk between TGF-ß/SMAD and WNT/ß-catenin signaling pathways in renal tubular epithelial cells, and that a WNT/ß-catenin inhibitor, ICG-001, ameliorates TGF-ß1induced renal fibrosis. IC-2, a derivative of ICG-001, has been shown to potently induce hepatocyte differentiation of human mesenchymal stem cells by inhibiting WNT/ß-catenin signaling. In the present study, we examined the effect of ICG-001, IC-2, and IC-2 derivatives (IC-2-506-1, IC-2-506-2, IC-2-506-3, IC-2-Ar-Cl, IC-2-OH, IC-2-OTBS, and IC-2-F) on TGF-ß1-induced SMAD activation and fibrogenic response in immortalized human renal tubular epithelial HK-2 cells. All these compounds inhibited LiCl-induced WNT/ß-catenin reporter activation to a similar extent, whereas ICG-001, IC-2-OTBS, and IC-2-F almost completely suppressed TGF-ß1-induced SMAD reporter activation without apparent cytotoxicity. Phosphorylation of SMAD2/3 by TGF-ß1 was more potently inhibited by IC-2-OTBS and IC-2-F than by ICG-001 and IC-2. IC-2-F suppressed TGF-ß1-induced COL1A1 protein expression, whereas IC-2-506-1 and IC-2-OTBS suppressed TGF-ß1-induced epithelial-mesenchymal transition. These results demonstrated that IC-2 derivatives suppress the TGF-ß1-induced fibrogenic response of tubular epithelial cells and thus could be promising therapeutic agents for the treatment of renal fibrosis.


Subject(s)
Epithelial Cells/drug effects , Kidney Diseases/prevention & control , Kidney Tubules/drug effects , Protective Agents/pharmacology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/toxicity , Wnt Signaling Pathway/drug effects , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Fibrosis , Humans , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Phosphorylation
8.
Microvasc Res ; 121: 14-23, 2019 01.
Article in English | MEDLINE | ID: mdl-30189210

ABSTRACT

PURPOSE: Diabetic retinopathy (DR) is characterized by pro-inflammatory, pro-angiogenic and pro-fibrotic environment during the various stages of the disease progression. Basement membrane changes in the retina and formation of fibrovascular membrane are characteristically seen in DR. In the present study the effect of Alcoholic (AlE) extracts of Triphala an ayurvedic herbal formulation and its chief compounds, Chebulagic (CA), Chebulinic (CI) and Gallic acid (GA) were evaluated for TGFß1-induced anti-fibrotic activity in choroid-retinal endothelial cells (RF/6A). METHOD: RF/6A cells were treated with TGFß1 alone or co-treated with AlE, CA, CI or GA. The mRNA and protein expression of fibrotic markers (αSMA, CTGF) were assessed by qPCR and western blot/ELISA. Functional changes were assessed using proliferation assay and migration assay. To deduce the mechanism of action, downstream signaling was assessed by western blot analysis along with in silico docking studies. RESULT: AlE (50 µg/ml) CA and CI at 10 µM reduced the expression of pro-fibrotic genes (αSMA and CTGF) induced by TGFß1, by inhibiting ERK phosphorylation. GA did not inhibit TGFß1 mediated changes in RF/6A cells. In silico experiments shows that CA and CI has favourable binding energy to bind with TGFß receptor and inhibit the downstream signaling, while GA did not. CONCLUSION: Hence this study identifies Triphala and its chief compounds CA and CI as potential adjuvants in the management of DR.


Subject(s)
Benzopyrans/pharmacology , Choroid/blood supply , Diabetic Retinopathy/drug therapy , Endothelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucosides/pharmacology , Hydrolyzable Tannins/pharmacology , Plant Extracts/pharmacology , Retinal Vessels/drug effects , Transforming Growth Factor beta1/toxicity , Animals , Benzopyrans/metabolism , Binding Sites , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Diabetic Retinopathy/enzymology , Diabetic Retinopathy/pathology , Endothelial Cells/enzymology , Endothelial Cells/pathology , Fibrosis , Glucosides/metabolism , Hydrolyzable Tannins/metabolism , Macaca mulatta , Molecular Docking Simulation , Neovascularization, Pathologic , Phosphorylation , Protein Binding , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Retinal Vessels/enzymology , Retinal Vessels/pathology , Signal Transduction/drug effects
9.
Toxicol Appl Pharmacol ; 369: 17-29, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30826375

ABSTRACT

Crystalline silica (SiO2) particles have very strong toxicity to the lungs, and silicosis is an excessive pulmonary interstitial remodeling disease that follows persistent SiO2 injury. We showed here that DNA double strand breaks (DSBs) and apoptosis were aggravated during rat silicosis induced by SiO2 exposure. Ac-SDKP attenuates lung parenchymal distortion and collagen deposition, and decreases the expression of γH2AX, p21, and cleaved caspase-3, as well as improves the reduction of pulmonary function caused by silicosis. In vitro, we found an evolution of smooth muscle actin α (α-SMA), collagen type I (Col I) in both A549 and MRC-5 cells in response to transforming growth factor-beta 1 (TGF-ß1) + SiO2. Only A549 cells showed any reduction in the rate of apoptosis induced by the double stimulation, because of the anti-apoptotic effects of TGF-ß1. N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is an anti-fibrotic tetrapeptide. It also has the ability to promote the apoptosis of leukemia cells. However its role in promoting cell apoptosis in silicosis is still unknown. We here found that Ac-SDKP could induce cell apoptosis and inhibit fibrotic response in A549 and MRC-5 cells treated with TGF-ß1 + SiO2, and these effects depended on regulation of α-tubulin acetyltransferase 1 (α-TAT1). These findings suggest that Ac-SDKP may have therapeutic value in the treatment of silicotic fibrosis.


Subject(s)
Acetyltransferases/metabolism , Apoptosis/drug effects , Epithelial Cells/drug effects , Fibroblasts/drug effects , Lung/drug effects , Microtubule Proteins/metabolism , Oligopeptides/pharmacology , Silicon Dioxide/toxicity , Silicosis/drug therapy , Transforming Growth Factor beta1/toxicity , A549 Cells , Animals , Collagen Type I/metabolism , DNA Breaks, Double-Stranded , Disease Models, Animal , Epithelial Cells/enzymology , Epithelial Cells/pathology , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Lung/enzymology , Lung/pathology , Male , Rats, Sprague-Dawley , Signal Transduction , Silicosis/enzymology , Silicosis/pathology , Up-Regulation
10.
Respir Res ; 20(1): 119, 2019 Jun 11.
Article in English | MEDLINE | ID: mdl-31185973

ABSTRACT

BACKGROUND: Pirfenidone, an antifibrotic agent used for the treatment of idiopathic pulmonary fibrosis (IPF), functions by inhibiting myofibroblast differentiation, which is involved in transforming growth factor (TGF)-ß1-induced IPF pathogenesis. However, unlike normal lung fibroblasts, the relationship between pirfenidone responses of TGF-ß1-induced human fibrotic lung fibroblasts and lung fibrosis has not been elucidated. METHODS: The effects of pirfenidone were evaluated in lung fibroblasts isolated from fibrotic human lung tissues after TGF-ß1 exposure. The ability of two new pharmacological targets of pirfenidone, collagen triple helix repeat containing protein 1(CTHRC1) and four-and-a-half LIM domain protein 2 (FHL2), to mediate contraction of collagen gels and migration toward fibronectin were assessed in vitro. RESULTS: Compared to control lung fibroblasts, pirfenidone significantly restored TGF-ß1-stimulated fibroblast-mediated collagen gel contraction, migration, and CTHRC1 release in lung fibrotic fibroblasts. Furthermore, pirfenidone attenuated TGF-ß1- and CTHRC1-induced fibroblast activity, upregulation of bone morphogenic protein-4(BMP-4)/Gremlin1, and downregulation of α-smooth muscle actin, fibronectin, and FHL2, similar to that observed post-CTHRC1 inhibition. In contrast, FHL2 inhibition suppressed migration and fibronectin expression, but did not downregulate CTHRC1. CONCLUSIONS: Overall, pirfenidone suppressed fibrotic fibroblast-mediated fibrotic processes via inverse regulation of CTHRC1-induced lung fibroblast activity. Thus, CTHRC1 can be used for predicting pirfenidone response and developing new therapeutic targets for lung fibrosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Fibroblasts/drug effects , Lung/drug effects , Pyridones/pharmacology , Transforming Growth Factor beta1/toxicity , Adult , Aged , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Fibroblasts/pathology , Humans , Lung/pathology , Male , Middle Aged , Rats
11.
Respir Res ; 20(1): 249, 2019 Nov 08.
Article in English | MEDLINE | ID: mdl-31703732

ABSTRACT

Pulmonary fibrosis is a chronic, progressive lung disease associated with lung damage and scarring. The pathological mechanism causing pulmonary fibrosis remains unknown. Emerging evidence suggests prominent roles of epithelial-mesenchymal transition (EMT) of alveolar epithelial cells (AECs) in myofibroblast formation and progressive pulmonary fibrosis. Our previous work has demonstrated the regulation of YY1 in idiopathic pulmonary fibrosis and pathogenesis of fibroid lung. However, the specific function of YY1 in AECs during the pathogenesis of pulmonary fibrosis is yet to be determined. Herein, we found the higher level of YY1 in primary fibroblasts than that in primary epithelial cells from the lung of mouse. A549 and BEAS-2B cells, serving as models for type II alveolar pulmonary epithelium in vitro, were used to determine the function of YY1 during EMT of AECs. TGF-ß-induced activation of the pro-fibrotic program was applied to determine the role YY1 may play in pro-fibrogenesis of type II alveolar epithelial cells. Upregulation of YY1 was associated with EMT and pro-fibrotic phenotype induced by TGF-ß treatment. Targeted knockdown of YY1 abrogated the EMT induction by TGF-ß treatment. Enforced expression of YY1 can partly mimic the TGF-ß-induced pro-fibrotic change in either A549 cell line or primary alveolar epithelial cells, indicating the induction of YY1 expression may mediate the TGF-ß-induced EMT and pro-fibrosis. In addition, the translocation of NF-κB p65 from the cytoplasm to the nucleus was demonstrated in A549 cells after TGF-ß treatment and/or YY1 overexpression, suggesting that NF-κB-YY1 signaling pathway regulates pulmonary fibrotic progression in lung epithelial cells. These findings will shed light on the better understanding of mechanisms regulating pro-fibrogenesis in AECs and pathogenesis of lung fibrosis.


Subject(s)
Alveolar Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fibroblasts/drug effects , Pulmonary Fibrosis/chemically induced , Transforming Growth Factor beta1/toxicity , YY1 Transcription Factor/metabolism , A549 Cells , Active Transport, Cell Nucleus , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Cell Communication , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Signal Transduction , Transcription Factor RelA/metabolism , YY1 Transcription Factor/genetics
12.
Clin Exp Nephrol ; 23(12): 1345-1356, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31482255

ABSTRACT

BACKGROUND: Previous studies showed that microRNA-29b (miR-29b) inhibits renal fibrosis. Therefore, miR-29b replacement therapy represents a promising approach for treating renal fibrosis. However, an efficient method of kidney-targeted miRNA delivery has yet to be established. Recombinant adeno-associated virus (rAAV) vectors have great potential for clinical application. For kidney-targeted gene delivery, the most suitable AAV serotype has yet to be established. Here, we identified the most suitable AAV serotype for kidney-targeted gene delivery and determined that AAV-mediated miR-29b delivery can suppress renal fibrosis in vivo. METHOD: To determine which AAV serotype is suitable for kidney cells, GFP-positive cells were identified by flow cytometry after the infection of rAAV serotype 1-9 vectors containing the EGFP gene. Next, we injected rAAV vectors into the renal pelvis to determine transduction efficiency in vivo. GFP expression was measured seven days after injecting rAAV serotype 1-9 vectors carrying the EGFP gene. Finally, we investigated whether rAAV6-mediated miR-29b delivery can suppress renal fibrosis in UUO mouse model. RESULTS: We found that rAAV6 vector is the most suitable for targeting kidney cells regardless of animal species in vitro and rAAV6 is the most suitable vector for kidney-targeted in vivo gene delivery in mice. Intra-renal pelvic injection of rAAV vectors can transduce genes into kidney TECs. Furthermore, rAAV6-mediated miR-29b delivery attenuated renal fibrosis in UUO model by suppressing Snail1 expression. CONCLUSION: Our study has revealed that rAAV6 is the most suitable serotype for kidney-targeted gene delivery and rAAV6-mediated miR-29b delivery into kidney TECs can suppress established renal fibrosis.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Kidney Diseases/prevention & control , Kidney Tubules, Proximal/metabolism , MicroRNAs/genetics , Parvovirinae/genetics , Ureteral Obstruction/therapy , Animals , Cell Line , Dependovirus , Disease Models, Animal , Fibrosis , Humans , Kidney Diseases/diagnosis , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Parvovirinae/metabolism , Rats , Transforming Growth Factor beta1/toxicity , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
13.
Toxicol Appl Pharmacol ; 358: 35-42, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30201452

ABSTRACT

Fibroblast-to-myofibroblast differentiation is a highly important pathological characteristic of pulmonary fibrosis. In this study, we aimed to investigate the effects and mechanisms of baicalein on the differentiation of human lung fibroblasts. Baicalein reduced the levels of α-smooth muscle actin (α-SMA) mRNA and protein expression in TGF-ß1-treated human lung fibroblasts. It also decreased the contents of collagen type I and fibronectin in time- and dose-dependent manners, and retarded TGF-ß1-stimulated α-SMA filament formation. Baicalein diminished the expression of miR-21, and miR-21 mimics partially antagonized the effects of baicalein. Additionally, Baicalein inhibited the miR-21 transcriptor STAT3 activity but not AP-1 activity. Moreover, the expression of Spry 1 protein, a miR-21 known target, was improved by baicalein treatment, but the level of Smurf2 protein, another miR-21 target, was not interfered. Collectively, these results demonstrated that baicalein can attenuate TGF-ß1-induced human lung fibroblast differentiation by inhibiting the miR-21 expression.


Subject(s)
Fibroblasts/metabolism , Flavanones/pharmacology , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Antioxidants/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Humans , Transforming Growth Factor beta1/toxicity
14.
Respir Res ; 19(1): 91, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29747634

ABSTRACT

BACKGROUND: Fibroblast apoptosis is a critical component of normal repair and the acquisition of an apoptosis-resistant phenotype contributes to the pathogenesis of fibrotic repair. Fibroblasts from fibrotic lungs of humans and mice demonstrate resistance to apoptosis induced by Fas-ligand and prior studies have shown that susceptibility to apoptosis is enhanced when Fas (CD95) expression is increased in these cells. Moreover, prior work shows that Fas expression in fibrotic lung fibroblasts is reduced by epigenetic silencing of the Fas promoter. However, the mechanisms by which microenvironmental stimuli such as TGF-ß1 and substrate stiffness affect fibroblast Fas expression are not well understood. METHODS: Primary normal human lung fibroblasts (IMR-90) were cultured on tissue culture plastic or on polyacrylamide hydrogels with Young's moduli to recapitulate the compliance of normal (400 Pa) or fibrotic (6400 Pa) lung tissue and treated with or without TGF-ß1 (10 ng/mL) in the presence or absence of protein kinase inhibitors and/or inflammatory cytokines. Expression of Fas was assessed by quantitative real time RT-PCR, ELISA and Western blotting. Soluble Fas (sFas) was measured in conditioned media by ELISA. Apoptosis was assessed using the Cell Death Detection Kit and by Western blotting for cleaved PARP. RESULTS: Fas expression and susceptibility to apoptosis was diminished in fibroblasts cultured on 6400 Pa substrates compared to 400 Pa substrates. TGF-ß1 reduced Fas mRNA and protein in a time- and dose-dependent manner dependent on focal adhesion kinase (FAK). Surprisingly, TGF-ß1 did not significantly alter cell-surface Fas expression, but did stimulate secretion of sFas. Finally, enhanced Fas expression and increased susceptibility to apoptosis was induced by combined treatment with TNF-α/IFN-γ and was not inhibited by TGF-ß1. CONCLUSIONS: Soluble and matrix-mediated pro-fibrotic stimuli promote fibroblast resistance to apoptosis by decreasing Fas transcription while stimulating soluble Fas secretion. These findings suggest that distinct mechanisms regulating Fas expression in fibroblasts may serve different functions in the complex temporal and spatial evolution of normal and fibrotic wound-repair responses.


Subject(s)
Apoptosis/physiology , Fibroblasts/metabolism , Fibroblasts/pathology , fas Receptor/biosynthesis , fas Receptor/genetics , Apoptosis/drug effects , Cells, Cultured , Fibroblasts/drug effects , Fibrosis , Gene Expression , Humans , Transforming Growth Factor beta1/toxicity
15.
J Cell Physiol ; 232(8): 2104-2111, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27714812

ABSTRACT

Chronic kidney disease (CKD) is a worldwide public health problem that affects millions of men and women of all ages and racial groups. Loss of mesangial cells (MC) represents an early common feature in the pathogenesis of CKD. Transforming growth factor-ß1 (TGF-ß1) is a key inducer of kidney damage and triggers several pathological changes in renal cells, notably MC apoptosis. However, the mechanism of MC apoptosis induced by TGF-ß1 remains elusive. Here, we demonstrate for the first time a novel regulatory pathway in which the disheveled-binding antagonist of ß-catenin 1 (Dact1) gene is upregulated by TGF-ß1, inducing MC apoptosis. We also show that the inhibitory effect of Dact1 and TGF-ß1 on the transcriptional activation of the pro-survival Wnt pathway is the mechanism of death induction. In addition, Dact1 mRNA/protein levels are increased in kidney remnants from 5/6 nephrectomized rats and strongly correlate with TGF-ß1 expression. Together, our results point to Dact1 as a novel element controlling MC survival that is causally related to CKD progression. J. Cell. Physiol. 232: 2104-2111, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Mesangial Cells/drug effects , Nuclear Proteins/metabolism , Renal Insufficiency, Chronic/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/toxicity , Wnt Signaling Pathway/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Male , Mesangial Cells/metabolism , Mesangial Cells/pathology , Nephrectomy , Nuclear Proteins/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Transfection , Transforming Growth Factor beta1/genetics , Up-Regulation
16.
Toxicol Appl Pharmacol ; 318: 1-7, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27989784

ABSTRACT

Pulmonary fibrosis is characterized by the destruction of lung tissue architecture and the formation of fibrous foci, currently has no satisfactory treatment. Emodin is a component of Chinese herb that has been reported to be medicament on pancreatic fibrosis and liver fibrosis. However, its role in pulmonary fibrosis has not been established yet. In the present study, we investigated the hypothesis that Emodin plays an inhibitory role in TGF-ß1 induced epithelial-mesenchymal transition (EMT) of alveolar epithelial cell, and Emodin exerts its effect through the Notch signaling pathway. Emodin inhibits the proliferation of Rat alveolar type II epithelial cells RLE-6TN in a concentration-dependent manner; reduces the expression of Collagen I, α-SMA and Vimentin, promotes the expression of E-cadherin. Moreover, Emodin could regulate the expression patterns of the Notch signaling pathway-related factors and reduce the Notch-1 nucleus translocation. Knockdown of Notch-1 enhances the inhibitory effect of Emodin on TGF-ß1-induced EMT in RLE-6TN cells. In conclusion, the data of the present study suggests that Emodin suppresses TGF-ß1-induced EMT in alveolar epithelial cells through Notch signaling pathway and shows the potential to be effective in the treatment of pulmonary fibrosis.


Subject(s)
Alveolar Epithelial Cells/drug effects , Emodin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Receptor, Notch1/antagonists & inhibitors , Signal Transduction/drug effects , Transforming Growth Factor beta1/antagonists & inhibitors , Alveolar Epithelial Cells/physiology , Animals , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/physiology , Rats , Receptor, Notch1/physiology , Signal Transduction/physiology , Transforming Growth Factor beta1/toxicity
17.
J Gastroenterol Hepatol ; 31(6): 1220-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26574150

ABSTRACT

BACKGROUND AND AIM: Epithelial-mesenchymal transition (EMT) of biliary epithelial cells (BECs) plays an important role in biliary fibrosis. This study investigated the effects of simvastatin on the lipopolysaccharide (LPS)-induced EMT and related signal pathways in BECs. METHODS: Biliary epithelial cells were exposed to LPS (2 µg/mL) or transforming growth factor ß1 (TGF-ß1) (5 ng/mL) for 5 days. The EMT was assessed by a gain of mesenchymal cell markers (vimentin, N-cadherin, slug, and Twist-1) and a loss of epithelial cell markers (E-cadherin). The effects of simvastatin on the EMT induced by LPS or TGF-ß1 were determined by the changes in the levels of EMT markers and TLR4 and in the c-Jun N-terminal kinase (JNK), p38, and nuclear factor-κB (NF-κB) signaling pathways. RESULTS: Compared with the BECs treated with LPS alone, co-treatment with simvastatin and LPS induced an increase in the expression of E-cadherin and decreases in the expression levels of mesenchymal cell markers. The LPS-induced TLR4 expression level was slightly decreased by co-treatment with simvastatin. LPS-induced BEC growth was markedly inhibited by co-treatment with simvastatin. Furthermore, pretreatment with simvastatin inhibited the LPS-induced EMT in BECs by downregulating NF-κB and JNK phosphorylation. The suppressive effects of simvastatin pretreatment on the induction of the EMT by TGF-ß1 were also demonstrated in H69 cells. CONCLUSIONS: Our results demonstrate that LPS or TGF-ß1 promote the EMT in BECs that that pretreatment with simvastatin inhibited the induced EMT by downregulating toll-like receptor 4 and NF-κB phosphorylation. This finding suggests that simvastatin can be considered a new agent for preventing biliary fibrosis associated with the EMT of BECs.


Subject(s)
Bile Ducts/drug effects , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipopolysaccharides/toxicity , Liver Cirrhosis, Biliary/prevention & control , NF-kappa B/metabolism , Simvastatin/pharmacology , Toll-Like Receptor 4/metabolism , Bile Ducts/metabolism , Bile Ducts/pathology , Biomarkers/metabolism , Cell Line, Transformed , Cell Proliferation/drug effects , Cytoprotection , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Cirrhosis, Biliary/chemically induced , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/pathology , Phosphorylation , Signal Transduction/drug effects , Time Factors , Toll-Like Receptor 4/genetics , Transforming Growth Factor beta1/toxicity
18.
Clin Exp Pharmacol Physiol ; 42(6): 680-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25882815

ABSTRACT

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


Subject(s)
Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Kidney Tubules, Proximal/metabolism , Kruppel-Like Transcription Factors/physiology , Transforming Growth Factor beta1/toxicity , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Fibrosis/chemically induced , Fibrosis/metabolism , Fibrosis/pathology , Humans , Inflammation Mediators/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Kruppel-Like Factor 4 , Mice , Mice, Knockout
19.
J Exp Med ; 204(5): 1083-93, 2007 May 14.
Article in English | MEDLINE | ID: mdl-17485510

ABSTRACT

Semaphorin (SEMA) 7A regulates neuronal and immune function. In these studies, we tested the hypothesis that SEMA 7A is also a critical regulator of tissue remodeling. These studies demonstrate that SEMA 7A and its receptors, plexin C1 and beta1 integrins, are stimulated by transforming growth factor (TGF)-beta(1) in the murine lung. They also demonstrate that SEMA 7A plays a critical role in TGF-beta(1)-induced fibrosis, myofibroblast hyperplasia, alveolar remodeling, and apoptosis. TGF-beta(1) stimulated SEMA 7A via a largely Smad 3-independent mechanism and stimulated SEMA 7A receptors, matrix proteins, CCN proteins, fibroblast growth factor 2, interleukin 13 receptor components, proteases, antiprotease, and apoptosis regulators via Smad 2/3-independent and SEMA 7A-dependent mechanisms. SEMA 7A also played an important role in the pathogenesis of bleomycin-induced pulmonary fibrosis. TGF-beta(1) and bleomycin also activated phosphatidylinositol 3-kinase (PI3K) and protein kinase B (PKB)/AKT via SEMA 7A-dependent mechanisms, and PKB/AKT inhibition diminished TGF-beta(1)-induced fibrosis. These observations demonstrate that SEMA 7A and its receptors are induced by TGF-beta(1) and that SEMA 7A plays a central role in a PI3K/PKB/AKT-dependent pathway that contributes to TGF-beta(1)-induced fibrosis and remodeling. They also demonstrate that the effects of SEMA 7A are not specific for transgenic TGF-beta(1), highlighting the importance of these findings for other fibrotic stimuli.


Subject(s)
Antigens, CD/metabolism , Pulmonary Alveoli/physiology , Pulmonary Fibrosis/chemically induced , Semaphorins/metabolism , Transforming Growth Factor beta1/toxicity , Analysis of Variance , Animals , Apoptosis/drug effects , Collagen/analysis , DNA Damage/drug effects , Immunoblotting , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Integrin beta1/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/metabolism , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/metabolism
20.
Arch Toxicol ; 85(6): 663-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21337027

ABSTRACT

Transforming growth factor-ß (TGF-ß) is closely associated with progressive renal fibrosis. A central component of TGF-ß-stimulated mesangial cell fibrogenesis is the TGF-ß family-specific Smad signal transduction pathway. This study investigated the expression of TGF-ß-receptor--activated Smad2, its common partner Smad4, and the phosphorylated Smad2 (p-Smad2) in adriamycin-induced toxicity of cultured rat mesangial cells. This in vitro study showed that amlodipine (10(-9) to 10(-5) mol/l) had no effect on the toxicity of rat mesangial cells induced by adriamycin in the absence of TGF-ß1. However, amlodipine (10(-7) to 10(-5) mol/l) reduced the toxicity of rat mesangial cells induced by TGF-ß1 in the absence of adriamycin; moreover, amlodipine (10(-8) to 10(-5) mol/l) significantly reduced adriamycin-induced cytotoxicity when it was given in combination with TGF-ß1; amlodipine (10(-6), 10(-5) mol/l) had no effect on Smad2 mRNA and protein expression induced by adriamycin + TGF-ß1, but it (10(-6), 10(-5) mol/l) dramatically inhibited the down-regulation of p-Smad2 protein expression as well as Smad4 mRNA and protein expression induced by adriamycin + TGF-ß1 in rat mesangial cells. Present study shows that amlodipine exerts a significant inhibition on adriamycin-induced toxicity in rat mesangial cells by affecting the expression of TGF-ß/Smad signaling intermediates p-Smad2 and Smad4.


Subject(s)
Amlodipine/pharmacology , Doxorubicin/toxicity , Gene Expression Regulation/drug effects , Mesangial Cells/drug effects , Protective Agents/pharmacology , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Antihypertensive Agents/pharmacology , Cell Line , Cell Survival/drug effects , Humans , Mesangial Cells/metabolism , Osmolar Concentration , RNA, Messenger/metabolism , Rats , Recombinant Proteins/toxicity , Signal Transduction/drug effects , Smad2 Protein/genetics , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta1/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL