Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 63
1.
Pestic Biochem Physiol ; 200: 105831, 2024 Mar.
Article En | MEDLINE | ID: mdl-38582594

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.


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.
Cell Cycle ; 20(8): 808-818, 2021 04.
Article En | MEDLINE | ID: mdl-33794722

The cyclin D-CDK4/6 complex has two distinct functions. Its kinase-dependent role involves its ability to act as serine/threonine kinase, responsible for phosphorylation of substrates required for cell cycle transitions, while its kinase-independent function involves its ability to act as a reservoir for p27Kip1. This association sequesters p27 from cyclin E-CDK2 complexes, allowing them to remain active. The aim of this current study is two-fold: to understand the contribution of the kinase-dependent and kinase-independent functions of CDK4 and CDK6 in epithelial cells and to directly compare CDK4 and CDK6 in a simple model system, TGF-ß treatment, where arrest is initiated by the expression of p15Ink4b. Cells that overexpressed a catalytically inactive, p15-insensitive CDK6 variant (p27 sequestration only mutant) were able to overcome TGF-ß-mediated arrest by maintaining CDK2 activity, while cells expressing the identical mutations in CDK4 were not. This result can be partially explained by the presence of a previously unidentified cyclin D-CDK6 dimer, which serves as a sink for free p27 during TGF-ß treatment, enabling CDK2 to remain inhibitor free. The use of the TGF-ß model system and the characterization of CDK pool dynamics and p27 switching is relevant to the CDK4/6 specific inhibitors, such as palbociclib, whose mechanism of action may resemble that of p15.


Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 6/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Multimerization/physiology , Transforming Growth Factor beta/toxicity , Cell Cycle Checkpoints/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/antagonists & inhibitors , Cyclins/metabolism , Humans , Protein Multimerization/drug effects
3.
Biochem Pharmacol ; 183: 114356, 2021 01.
Article En | MEDLINE | ID: mdl-33285108

Abnormal activation of transforming growth factor (TGF)-ß is a common cause of fibroblast activation and fibrosis. In bleomycin (BLM)-induced lung fibrosis, the marked expression of phospho-Src homology-2 domain-containing phosphatase (SHP) 2, phospho-signal transducer and activator of transcription (STAT) 3, and suppressor of cytokine signaling (SOCS) 3 was highly associated with pulmonary parenchymal lesions and collagen deposition. Human pulmonary fibroblasts differentiated into myofibroblasts exhibited activation of SHP2, SOCS3, protein inhibitor of activated STAT1, STAT3, interleukin (IL)-6, and IL-10. The significant retardation of interferon (IFN)-γ signaling in myofibroblasts was revealed by the decreased expression of phospho-STAT1, IFN-γ-associated genes, and IFN-γ-inducible protein (IP) 10. Microarray analysis showed an induction of fibrotic genes in TGF-ß1-differentiated myofibroblasts, whereas IFN-γ-regulated anti-fibrotic genes were suppressed. Interestingly, BIBF 1120 treatment effectively inhibited both STAT3 and SHP2 phosphorylation in TGF-ß1-differentiated myofibroblasts and BLM fibrotic lung tissues, which was accompanied by suppression of fibroblast-myofibroblast transition. Moreover, the combined treatment of BIBF 1120 plus IFN-γ or SHP2 inhibitor PHPS1 plus IFN-γ markedly reduced TGF-ß1-induced α-smooth muscle actin and further ameliorated BLM lung fibrosis. Accordingly, myofibroblasts were hyporesponsiveness to IFN-γ, while blockade of SHP2 contributed to the anti-fibrotic efficacy of IFN-γ.


Bleomycin/toxicity , Fibroblasts/metabolism , Interferon-gamma/metabolism , Myofibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Interferon-gamma/antagonists & inhibitors , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/toxicity
4.
FASEB J ; 34(7): 8920-8940, 2020 07.
Article En | MEDLINE | ID: mdl-32519817

In the current work we show that the profibrotic actions of TGF-ß are mediated, at least in part, through a metabolic maladaptation in glutamine metabolism and how the inhibition of glutaminase 1 (GLS1) reverses pulmonary fibrosis. GLS1 was found to be highly expressed in fibrotic vs normal lung fibroblasts and the expression of profibrotic targets, cell migration, and soft agar colony formation stimulated by TGF-ß required GLS1 activity. Moreover, knockdown of SMAD2 or SMAD3 as well as inhibition of PI3K, mTORC2, and PDGFR abrogated the induction of GLS1 by TGF-ß. We further demonstrated that the NAD-dependent protein deacetylase, SIRT7, and the FOXO4 transcription factor acted as endogenous brakes for GLS1 expression, which are inhibited by TGF-ß. Lastly, administration of the GLS1 inhibitor CB-839 attenuated bleomycin-induced pulmonary fibrosis. Our study points to an exciting and unexplored connection between epigenetic and transcriptional processes that regulate glutamine metabolism and fibrotic development in a TGF-ß-dependent manner.


Fibroblasts/pathology , Gene Expression Regulation , Glutaminase/metabolism , Pulmonary Fibrosis/pathology , Sirtuins/metabolism , Transforming Growth Factor beta/toxicity , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Cell Movement , Cells, Cultured , Female , Fibroblasts/metabolism , Glutaminase/genetics , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Signal Transduction , Sirtuins/genetics , Smad Proteins/genetics , Smad Proteins/metabolism
5.
Biochem Biophys Res Commun ; 528(1): 154-159, 2020 07 12.
Article En | MEDLINE | ID: mdl-32451085

Renal fibrosis is one of the characteristic features of chronic kidney disease (CKD). Fibrotic change not only impairs the filtration function of the kidney but is also recognized as a marker of end-stage renal disease (ESRD). The epithelial to mesenchymal transition (EMT) is known to play a role in embryonic development and organ formation, but it is getting much attention for its pathological role in the invasion and metastasis of carcinoma. Recently, it has also been reported that EMT plays a role in the formation of fibrosis during chronic inflammation. EMT contribute to the development of the fibrosis in CKD. Moreover, glomerular podocytes and tubular epithelial cells can also undergo mesenchymal transition in CKD. Hesperetin is a flavonoid present in citrus and is well known for its antioxidant and anti-inflammatory properties. In this study, we investigated the effects of hesperetin on the EMT-elicited podocytes. First, we generated an EMT model by treating transforming growth factor (TGF)-ß1, a potent inducer of EMT to the podocytes. TGF-ß1 decreased the expression of epithelial markers such as nephrin, zonula occludens-1 (ZO-1), while it increased the mesenchymal markers, including fibronectin (FN), vimentin, and α-smooth muscle actin (α-SMA) in the podocytes. Hesperetin suppressed EMT-like changes elicited by TGF-ß1. Interestingly, hesperetin did not interfere with the Smad signaling-the classical TGF-ß signaling-pathway, which was confirmed by the experiment with smad 2/3 -/- podocytes. Instead, hesperetin suppressed EMT-like changes by inhibiting the mTOR pathway-one of the alternative TGF-ß signaling pathways. In conclusion, hesperetin has a protective effect on the TGF-ß1 elicited EMT-like changes of podocytes through regulation of mTOR pathway. It could be a good candidate for the suppression of kidney fibrosis in various CKD.


Epithelial-Mesenchymal Transition/drug effects , Hesperidin/pharmacology , Podocytes/metabolism , Podocytes/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/toxicity , Cell Death/drug effects , Hesperidin/chemistry , Humans , Podocytes/drug effects , Signal Transduction/drug effects , Smad Proteins/metabolism
6.
Respir Res ; 21(1): 58, 2020 Feb 19.
Article En | MEDLINE | ID: mdl-32075634

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible disease characterized by excessive fibroblast to myofibroblast differentiation with limited therapeutic options. Curdione, a sesquiterpene compound extracted from the essential oil of Curcuma aromatica Salisb, has anti-inflammatory and anti-tumor effects. However, the role of curdione in IPF is still unclear. METHODS: The effects of curdione were evaluated in a bleomycin (BLM)-induced pulmonary fibrosis mouse model. C57BL/6 mice were treated with BLM on day 0 by intratracheal injection and intraperitoneal administered curdione or vehicle. In vitro study, expression of fibrotic protein was examined and the transforming growth factor (TGF)-ß-related signaling was evaluated in human pulmonary fibroblasts (HPFs) treated with curdione following TGF-ß1 stimulation. RESULTS: Histological and immunofluorescent examination showed that curdione alleviated BLM-induced lung injury and fibrosis. Specifically, curdione significantly attenuated fibroblast to myofibroblast differentiation in the lung in BLM induced mice. Furthermore, curdione also decreased TGF-ß1 induced fibroblast to myofibroblast differentiation in vitro, as evidenced by low expression of α-SMA, collagen 1 and fibronectin in a dose dependent manner. Mechanistically, curdione suppressed the phosphorylation of Smad3 following TGF-ß1 treatment, thereby inhibiting fibroblast differentiation. CONCLUSIONS: Overall, curdione exerted therapeutic effects against pulmonary fibrosis via attenuating fibroblast to myofibroblast differentiation. As curdione had been shown to be safe and well-tolerated in BLM-induced mouse model, curdione might be useful for developing novel therapeutics for IPF.


Bleomycin/toxicity , Cell Differentiation/physiology , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Myofibroblasts/metabolism , Sesquiterpenes, Germacrane/therapeutic use , Transforming Growth Factor beta/toxicity , Animals , Antibiotics, Antineoplastic/toxicity , Cell Differentiation/drug effects , Cells, Cultured , Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Transforming Growth Factor beta/antagonists & inhibitors
7.
J Appl Toxicol ; 40(7): 931-946, 2020 07.
Article En | MEDLINE | ID: mdl-32061184

Small-molecule inhibitors of transforming growth factor beta receptor 1 (TGFßRI) have a history of significant class-based toxicities (eg, cardiac valvulopathy) in preclinical species that have limited their development as new medicines. Nevertheless, some TGFßRI inhibitors have entered into clinical trials using intermittent-dosing schedules and exposure limits in an attempt to avoid these toxicities. This report describes the toxicity profile of the small-molecule TGFßRI inhibitor, BMS-986260, in rats and dogs. Daily oral dosing for 10 days resulted in valvulopathy and/or aortic pathology at systemic exposures that would have been targeted clinically, preventing further development with this dosing schedule. These toxicities were not observed in either species in 1-month studies using the same doses on an intermittent-dosing schedule of 3 days on and 4 days off (QDx3 once weekly). Subsequently, 3-month studies were conducted (QDx3 once weekly), and while there were no cardiovascular findings in dogs, valvulopathy and mortality occurred early in rats. The only difference compared to the 1-month study was that the rats in the 3-month study were 2 weeks younger at the start of dosing. Therefore, a follow-up 1-month study was conducted to evaluate whether the age of rats influences sensitivity to target-mediated toxicity. Using the same dosing schedule and similar doses as in the 3-month study, there was no difference in the toxicity of BMS-986260 in young (8 weeks) or adult (8 months) rats. In summary, an intermittent-dosing schedule mitigated target-based cardiovascular toxicity in dogs but did not prevent valvulopathy in rats, and thus the development of BMS-986260 was terminated.


Aortic Diseases/chemically induced , Aortic Diseases/therapy , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Inhibitors/therapeutic use , Receptor, Transforming Growth Factor-beta Type I/therapeutic use , Transforming Growth Factor beta/toxicity , Animals , Dogs , Female , Humans , Male , Models, Animal , Rats
8.
J Cell Mol Med ; 23(10): 6872-6884, 2019 10.
Article En | MEDLINE | ID: mdl-31397957

Peritoneal fibrosis (PF) is an intractable complication of peritoneal dialysis (PD) that leads to peritoneal membrane failure. This study investigated the role of suppression of tumorigenicity (ST)2 in PF using patient samples along with mouse and cell-based models. Baseline dialysate soluble (s)ST2 level in patients measured 1 month after PD initiation was 2063.4 ± 2457.8 pg/mL; patients who switched to haemodialysis had elevated sST2 levels in peritoneal effluent (1576.2 ± 199.9 pg/mL, P = .03), which was associated with PD failure (P = .04). Baseline sST2 showed good performance in predicting PD failure (area under the receiver operating characteristic curve = 0.780, P = .001). In mice with chlorhexidine gluconate-induced PF, ST2 was expressed in fibroblasts and mesothelial cells within submesothelial zones. In primary cultured human peritoneal mesothelial cells (HPMCs), transforming growth factor-ß treatment increased ST2, fibronectin, ß-galactosidase and Snail protein levels and decreased E-cadherin level. Anti-ST2 antibody administration reversed the up-regulation of ST2 and fibronectin expression; it also reduced fibrosis induced by high glucose (100 mmol/L) in HPMCs. Thus, high ST2 level in dialysate is a marker for fibrosis and inflammation during peritoneal injury, and blocking ST2 may be an effective therapeutic strategy for renal preservation.


Glucose/toxicity , Interleukin-1 Receptor-Like 1 Protein/antagonists & inhibitors , Peritoneal Fibrosis/pathology , Transforming Growth Factor beta/toxicity , Animals , Cells, Cultured , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Epithelium/pathology , Female , Humans , Interleukin-1 Receptor-Like 1 Protein/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Peritoneal Dialysis , Peritoneum/pathology , Proportional Hazards Models , Survival Analysis
9.
BMC Nephrol ; 20(1): 245, 2019 07 05.
Article En | MEDLINE | ID: mdl-31277592

BACKGROUND: It has been proposed that peroxisome proliferator-activated receptor-γ (PPARγ) agonists might reduce renal fibrosis, however, several studies had contradictory results. Moreover, the possible interaction of TGF-ß1, PPARγ, and transcription factors in renal fibrosis have not been investigated. We hypothesized that oral pioglitazone treatment would inhibit TGF-ß-driven renal fibrosis and its progression, by modulating profibrotic transcription factors in TGF-ß1 transgenic mice. METHODS: Male C57Bl/6 J mice (control, CTL, n = 14) and TGF-ß overexpressing transgenic mice (TGFß, n = 14, having elevated plasma TGF-ß1 level) were divided in two sets at 10 weeks of age. Mice in the first set were fed with regular rodent chow (CTL and TGFß, n = 7/group). Mice in the second set were fed with chow containing pioglitazone (at a dose of 20 mg/kg/day, CTL + Pio and TGFß+Pio, n = 7/group). After 5 weeks of treatment, blood pressure was assessed and urine samples were collected, and the kidneys were analyzed for histology, mRNA and protein expression. RESULTS: TGF-ß1 induced glomerulosclerosis and tubulointerstitial damage were significantly reduced by pioglitazone. Pioglitazone inhibited renal mRNA expression of all the profibrotic effectors: type-III collagen, TGF-ß1, CTGF and TIMP-1, and alike transcription factors cFos/cJun and protein expression of EGR-1, and STAT3 protein phosphorylation. CONCLUSIONS: Oral administration of PPARγ agonist pioglitazone significantly reduces TGF-ß1-driven renal fibrosis, via the attenuation of EGR-1, STAT3 and AP-1. This implies that PPARγ agonists might be effective in the treatment of chronic kidney disease patients.


Early Growth Response Protein 1/antagonists & inhibitors , Kidney Diseases/prevention & control , PPAR gamma/agonists , Pioglitazone/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Early Growth Response Protein 1/metabolism , Fibrosis , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pioglitazone/pharmacology , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta/toxicity
10.
Phytomedicine ; 57: 223-235, 2019 Apr.
Article En | MEDLINE | ID: mdl-30785018

BACKGROUND: Renal fibrosis is the pathological feature of chronic kidney disease (CKD) which leads to end-stage renal disease (ESRD) and renal failure. Resveratrol [3,5,4'-trihydroxy-trans-stilbene (RSV)] has shown benefits for metabolic diseases and anti-cancer therapy, but its potential risk on renal health has not been fully evaluated. PURPOSE: To investigate the global effects of RSV on renal fibrosis in human tubular epithelial cell (TEC) line HK-2, and in mice with unilateral ureteral obstruction (UUO). METHODS: A TGF-ß-induced in vitro model of epithelial-mesenchymal transition (EMT) in TEC was established. The effects of RSV on cell viability, pro-fibrotic factors, oxidative stress, mitochondria function, and underlying pathway proteins were analyzed. In vivo, the effects of RSV on renal function and fibrosis were assayed in UUO mice. RESULTS: Our results showed that low concentrations of RSV (5-20 µM) decreased TGF-ß-induced EMT via Sirt1-dependent deacetylation of Smad3/Smad4 mechanism. By contrast, long-term (72 h) exposure to high concentrations of RSV (≥ 40 µM) promoted EMT in HK-2 cells via mitochondrial oxidative stress and ROCK1-mediated disordered cytoskeleton remodeling. In vivo, low-dose treatment of RSV (≤ 25 mg/kg) partly improved renal function, whereas high-dose treatment of RSV (≥ 50 mg/kg) lost its anti-fibrotic role and even aggravated renal fibrosis. However, mice with UUO were more susceptible to high RSV-induced renal injury than normal mice. CONCLUSION: Dependent on dose, RSV activated either anti-fibrotic or pro-fibrotic effects in kidneys. The risk of RSV consumption in individuals with impaired kidney function should be carefully considered.


Epithelial-Mesenchymal Transition/drug effects , Fibrosis/drug therapy , Kidney/drug effects , Resveratrol/administration & dosage , Resveratrol/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , Fibrosis/pathology , Humans , Kidney/pathology , Kidney/physiopathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/pathology , Sirtuin 1/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/toxicity , Ureteral Obstruction/drug therapy , Ureteral Obstruction/pathology
11.
Eur Rev Med Pharmacol Sci ; 22(19): 6551-6559, 2018 10.
Article En | MEDLINE | ID: mdl-30338826

OBJECTIVE: This study aims to explore the protective effect of cyclosporine on inflammation-induced renal tubular epithelial cells and its potential mechanism. MATERIALS AND METHODS: Human kidney-2 (HK-2) cells were induced by transforming growth factor-ß (TGF-ß) for constructing an inflammatory injury model. Cells were then treated with different concentrations of cyclosporine for further investigating the biological functions. Cell viability was detected via cell counting kit-8 assay (CCK-8). The cytotoxicity was detected via lactate dehydrogenase (LDH) release assay. Expression levels of cell damage factors and mammalian target of rapamycin (mTOR) pathway-related genes were detected via polymerase chain reaction (PCR), immunofluorescence and Western blotting, respectively. RESULTS: TGF-ß inhibited the viability of HK-2 cells, increased expressions of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and apoptosis-related genes. Cyclosporine treatment greatly reversed the cell damage on HK-2 cells induced by TGF-ß. Expression levels of mTOR pathway-related genes were downregulated after cyclosporine treatment. CONCLUSIONS: Cyclosporine protects HK-2 cells from inflammatory injury via regulating mTOR pathway.


Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cyclosporine/pharmacology , Epithelial Cells/drug effects , Kidney Tubules/drug effects , Nephritis, Interstitial/prevention & control , Cell Line , Cytoprotection , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Interleukin-1beta/metabolism , Kidney Tubules/metabolism , Kidney Tubules/pathology , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/toxicity , Tumor Necrosis Factor-alpha/metabolism
12.
Nutrients ; 9(9)2017 Sep 06.
Article En | MEDLINE | ID: mdl-28878195

Pulmonary fibrosis is a progressive fibrotic lung disease of persisting lung injury and ineffective wound repair, with poor prognosis. Epithelial-mesenchymal transition (EMT) of alveolar epithelia cells is an early event in the development of pulmonary fibrosis, and transforming growth factor β (TGF-β) is an acknowledged inducer of EMT. Epidemiological studies demonstrated that serum levels of 25-hydroxy-vitamin D were associated with the presence of fibrosis diseases. We investigated whether vitamin D attenuated TGF-β-induced pro-fibrotic effects through inhibiting EMT in human alveolar epithelia A549 cells. A549 cells were cultured with TGF-β alone or in combination with 1α,25-dihydroxyvitamin D3 (1α,25(OH)₂D₃). TGF-β increased the expression of the mesenchymal markers (N-cadherin and Vimentin), and decreased the expression of epithelial markers (E-cadherin). 1α,25(OH)₂D₃ attenuated these TGF-β-induced alterations. Furthermore, the EMT-related transcription factors (Snail and β-catenin) and the extracellular matrix genes (Collagen I and fibronectin) were inhibited by 1α,25(OH)₂D₃, while the expression of vitamin D receptor (VDR) was elevated. In addition, 1α,25(OH)₂D₃ alleviated the cell migration and the invasion abilities in TGF-β-stimulated A549 cells, determined by the scratch wound healing and transwell assays. Our findings suggested that 1α,25(OH)₂D₃ inhibited the pro-fibrotic phenotype of lung epithelial cells under TGF-β stimulation and provided new clues in the clinical management of pulmonary fibrosis.


Calcitriol/pharmacology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lung/drug effects , Pulmonary Fibrosis/prevention & control , Transforming Growth Factor beta/toxicity , A549 Cells , Antigens, CD/metabolism , Cadherins/metabolism , Cell Movement/drug effects , Cell Shape/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibronectins/genetics , Fibronectins/metabolism , Humans , Lung/metabolism , Lung/pathology , Phenotype , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Receptors, Calcitriol/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors/metabolism , Time Factors , Vimentin/metabolism , beta Catenin/metabolism
13.
Transl Res ; 183: 71-86.e1, 2017 05.
Article En | MEDLINE | ID: mdl-28027929

The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) governs antioxidant, innate immune and cytoprotective responses and its deregulation is prominent in chronic inflammatory conditions. To examine the hypothesis that Nrf2 might be implicated in systemic sclerosis (SSc), we investigated its expression, activity, and mechanism of action in SSc patient samples and mouse models of fibrosis and evaluated the effects of a novel pharmacologic Nrf2 agonist. We found that both expression and activity of Nrf2 were significantly reduced in SSc patient skin biopsies and showed negative correlation with inflammatory gene expression. In skin fibroblasts, Nrf2 mitigated fibrotic responses by blocking canonical transforming growth factor-ß (TGF-ß)-Smad signaling, whereas silencing Nrf2 resulted in constitutively elevated collagen synthesis, spontaneous myofibroblast differentiation, and enhanced TGF-ß responses. Bleomycin treatment of Nrf2-null mice resulted in exaggerated fibrosis. In wild-type mice, treatment with a novel pharmacologic Nrf2 agonist 2-trifluoromethyl-2'-methoxychalcone prevented dermal fibrosis induced by TGF-ß. These findings are the first to identify Nrf2 as a cell-intrinsic antifibrotic factor with key roles in maintaining extracellular matrix homeostasis and a pathogenic role in SSc. Pharmacologic reactivation of Nrf2, therefore, represents a novel therapeutic strategy toward effective treatment of fibrosis in SSc.


NF-E2-Related Factor 2/antagonists & inhibitors , Oleanolic Acid/analogs & derivatives , Scleroderma, Systemic/metabolism , Animals , Bleomycin/toxicity , Cells, Cultured , Chalcones/pharmacology , Down-Regulation , Fibroblasts/metabolism , Fibrosis/chemically induced , Fibrosis/genetics , Fibrosis/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Genetic Predisposition to Disease , Humans , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta/toxicity
14.
Sci Rep ; 6: 23836, 2016 Mar 31.
Article En | MEDLINE | ID: mdl-27030015

Most stromal corneal dystrophies are associated with aggregation and deposition of the mutated transforming growth factor-ß induced protein (TGFßIp). The 4(th)_FAS1 domain of TGFßIp harbors ~80% of the mutations that forms amyloidogenic and non-amyloidogenic aggregates. To understand the mechanism of aggregation and the differences between the amyloidogenic and non-amyloidogenic phenotypes, we expressed the 4(th)_FAS1 domains of TGFßIp carrying the mutations R555W (non-amyloidogenic) and H572R (amyloidogenic) along with the wild-type (WT). R555W was more susceptible to acidic pH compared to H572R and displayed varying chemical stabilities with decreasing pH. Thermal denaturation studies at acidic pH showed that while WT did not undergo any conformational transition, the mutants exhibited a clear pH-dependent irreversible conversion from αß conformation to ß-sheet oligomers. The ß-oligomers of both mutants were stable at physiological temperature and pH. Electron microscopy and dynamic light scattering studies showed that ß-oligomers of H572R were larger compared to R555W. The ß-oligomers of both mutants were cytotoxic to primary human corneal stromal fibroblast (pHCSF) cells. The ß-oligomers of both mutants exhibit variations in their morphologies, sizes, thermal and chemical stabilities, aggregation patterns and cytotoxicities.


Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/toxicity , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/toxicity , Fibroblasts/drug effects , Mutation , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/toxicity , Amino Acid Sequence , Amyloidogenic Proteins/genetics , Cell Survival/drug effects , Cloning, Molecular , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Corneal Stroma/cytology , Corneal Stroma/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Extracellular Matrix Proteins/genetics , Fibroblasts/cytology , Gene Expression , Humans , Hydrogen-Ion Concentration , Primary Cell Culture , Protein Denaturation , Protein Domains , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/toxicity , Transforming Growth Factor beta/genetics
15.
Prostate ; 74(8): 901-10, 2014 Jun.
Article En | MEDLINE | ID: mdl-24728978

BACKGROUND: NEDD9 is one of the Crk-associated substrate (Cas) family proteins that mediate downstream signaling processes including cytoskeletal organization, cell-cycle and tumorigenesis. While NEDD9 plays a crucial role in epithelial-mesenchymal transition (EMT), the functional mechanism underlying NEDD9-mediated EMT in prostate cancer (PCa) remains uncertain. METHODS: The expression levels of NEDD9 and its downstream molecules in PC-3, LNCaP, and VCaP cells exposed to transforming growth factor-ß (TGF-ß) were determined by western blotting. The invasion of these cells with ectopic overexpression of NEDD9 or silencing of NEDD9 expression was measured by transwell invasion assay. Human tissue samples comprising 45 PCa specimens and ten specimens of normal prostatic tissue were used for immunohistochemical (IHC) analysis of NEDD9 expression. RESULTS: Both NEDD9 and its downstream signaling molecules associated with EMT were strongly induced by TGF-ß in PCa cells. PC-3 cells with stable overexpression of NEDD9 had a mesenchymal phenotype and significantly enhanced cell invasion, despite their decreased cell proliferation. Knockdown of endogenous NEDD9 expression completely diminished TGF-ß-triggered tumor invasion in several PCa cell lines. The IHC data revealed a significant positive correlation between the NEDD9 staining score and tumor aggressiveness (e.g., Gleason grade, serum PSA level). The NEDD9 staining score in primary PCa with bone metastasis was significantly higher than that in PCa without metastasis. CONCLUSIONS: NEDD9 may be a key mediator involved in TGF-ß-mediated EMT and cell motility in PCa cells and a novel target in the treatment of metastatic PCa and prevention of spread of localized PCa cells to other organs.


Adaptor Proteins, Signal Transducing/physiology , Disease Progression , Epithelial-Mesenchymal Transition/physiology , Neoplasm Invasiveness , Phosphoproteins/physiology , Prostatic Neoplasms/metabolism , Transforming Growth Factor beta/toxicity , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Humans , Male , Neoplasm Invasiveness/pathology , Prostatic Neoplasms/pathology
16.
Drug Chem Toxicol ; 37(1): 40-7, 2014 Jan.
Article En | MEDLINE | ID: mdl-23841455

Escherichia coli-derived recombinant human (rh)BMP-2 (E.BMP-2) can be used as a bone graft substitute because to its high osteoinductivity, but its toxicity is not well understood. Thus, we report on the toxicity of E.BMP-2 in Sprague-Dawley rats under the condition of repetitive injection for 2 weeks. Randomly selected 10 male and female rats were administered with E.BMP-2 at a dose of 0.05, 0.18 or 0.5 mg/kg as an experimental group. A control group with another 10 rats was given E.BMP-2 carrier. Both E.BMP-2 and E.BMP-2 carrier were administered through intravenous injection for 2 weeks. For toxicokinetics study, 3 male and female rats were randomly selected from each group. During the observation period, general symptom, weight and food intakes were monitored, and ophthalmic and urine tests were performed as well. After the observation period, all animals were subjected to blood test, biochemical analysis and organ-weight measurement. During autopsy, visual inspections and histopathological examinations were done. Toxicokinetics study confirmed systemic exposure of the test material. No death or abnormal clinical sign was found during the injection period. Toxicity changes induced by the injection were not detected in autopsy or the tests for weight, food intakes, ophthalmology, hematology and serum biochemistry. The female groups administered with 0.18 and 0.5 mg/kg (the female 0.18-mg/kg group and the female 0.5-mg/kg group) showed absolute and relative weight loss in ovaries and reduced corpora lutea. It was the expected pharmacologic activity, rather than toxicity. The histopathological test revealed cartilage formation and increased fibroblast around the tail vein, but these were thought to be the result of osteoinductivity of the test material. In the male group with 0.5 mg/kg of E.BMP-2 (the male 0.5-mg/kg group), local appearance of multinucleated cells in lung parenchyma was observed, but it was considered as the natural reaction to remove E.BMP-2, which is a recombinant protein. In toxicokinetics study, systemic exposure (area under the serum concentration-time curve and maximum observed serum concentration) increased as the injection dose was increased in both male and female rats, and no clear difference was noticed between the sexes. Blood drug content did not change during the injection period, but the half-life was shortened as the injection dose was increased. Under the condition of this study, the no observed adverse effects level of E.BMP-2 was over 0.5 mg/kg in both male and female rats.


Bone Morphogenetic Protein 2/pharmacokinetics , Bone Morphogenetic Protein 2/toxicity , Escherichia coli/chemistry , Transforming Growth Factor beta/pharmacokinetics , Transforming Growth Factor beta/toxicity , Animals , Blood Chemical Analysis , Body Weight/drug effects , Bone Morphogenetic Protein 2/administration & dosage , Bone Morphogenetic Protein 2/blood , Corpus Luteum/drug effects , Dose-Response Relationship, Drug , Eating/drug effects , Female , Injections, Intravenous , Male , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Ovary/drug effects , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/toxicity , Statistics, Nonparametric , Toxicity Tests , Transforming Growth Factor beta/administration & dosage , Transforming Growth Factor beta/blood , Urinalysis
17.
Arthritis Res Ther ; 14(3): R151, 2012 Jun 21.
Article En | MEDLINE | ID: mdl-22721434

INTRODUCTION: The mechanism by which intra-articular injection of hyaluronan (HA) ameliorates joint pathology is unknown. Animal studies have shown that HA can reduce synovial activation, periarticular fibrosis and cartilage erosion; however, its specific effects on the different cell types involved remain unclear. We have used the TTR (TGFbeta1 injection and Treadmill Running) model of murine osteoarthritis (OA), which exhibits many OA-like changes, including synovial activation, to examine in vivo tissue-specific effects of intra-articular HA. METHODS: The kinetics of clearance of fluorotagged HA from joints was examined with whole-body imaging. Naïve and treated knee joints were examined macroscopically for cartilage erosion, meniscal damage and fibrosis. Quantitative histopathology was done with Safranin O for cartilage and with Hematoxylin & Eosin for synovium. Gene expression in joint tissues for Acan, Col1a1, Col2a1, Col3a1, Col5a1, Col10a1, Adamts5 and Mmp13 was done by quantitative PCR. The abundance and distribution of aggrecan, collagen types I, II, III, V and X, ADAMTS5 and MMP13 were examined by immunohistochemistry. RESULTS: Injected HA showed a half-life of less than 2 h in the murine knee joint. At the tissue level, HA protected against neovascularization and fibrosis of the meniscus/synovium and maintained articular cartilage integrity in wild-type but not in Cd44 knockout mice. HA injection enhanced the expression of chondrogenic genes and proteins and blocked that of fibrogenic/degradative genes and proteins in cartilage/subchondral bone, whereas it blocked activation of both groups in meniscus/synovium. In all locations it reduced the expression/protein for Mmp13 and blocked Adamts5 expression but not its protein abundance in the synovial lining. CONCLUSIONS: The injection of HA, 24 h after TGFbeta1 injection, inhibited the cascade of OA-like joint changes seen after treadmill use in the TTR model of OA. In terms of mechanism, tissue protection by HA injection was abrogated by Cd44 ablation, suggesting that interaction of the injected HA with CD44 is central to its protective effects on joint tissue remodeling and degeneration in OA progression.


Cartilage, Articular/drug effects , Hyaluronan Receptors/metabolism , Hyaluronic Acid/administration & dosage , Osteoarthritis/pathology , Viscosupplements/administration & dosage , ADAM Proteins/biosynthesis , ADAMTS5 Protein , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Disease Models, Animal , Extracellular Matrix Proteins/toxicity , Fibrosis , Immunohistochemistry , Injections, Intra-Arterial , Male , Matrix Metalloproteinase 13/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Osteoarthritis/metabolism , Polymerase Chain Reaction , Synovial Membrane/blood supply , Synovial Membrane/pathology , Transcriptome , Transforming Growth Factor beta/toxicity
18.
Muscle Nerve ; 45(1): 55-9, 2012 Jan.
Article En | MEDLINE | ID: mdl-22190307

INTRODUCTION: Transforming growth factor-beta (TGF-ß) is a well-known regulator of fibrosis and inflammation in many tissues. During embryonic development, TGF-ß signaling induces expression of the transcription factor scleraxis, which promotes fibroblast proliferation and collagen synthesis in tendons. In skeletal muscle, TGF-ß has been shown to induce atrophy and fibrosis, but the effect of TGF-ß on muscle contractility and the expression of scleraxis and atrogin-1, an important regulator of muscle atrophy, were not known. METHODS: We treated muscles from mice with TGF-ß and measured force production, scleraxis, procollagen Iα2, and atrogin-1 protein levels. RESULTS: TGF-ß decreased muscle fiber size and dramatically reduced maximum isometric force production. TGF-ß also induced scleraxis expression in muscle fibroblasts, and increased procollagen Iα2 and atrogin-1 levels in muscles. CONCLUSION: These results provide new insight into the effect of TGF-ß on muscle contractility and the molecular mechanisms behind TGF-ß-mediated muscle atrophy and fibrosis.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Muscle Proteins/metabolism , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Transforming Growth Factor beta/toxicity , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Collagen Type I/metabolism , Fibrosis/chemically induced , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , In Vitro Techniques , Mice , Mice, Transgenic , Muscle Contraction/drug effects , Muscular Atrophy/physiopathology
19.
Toxicology ; 282(1-2): 39-46, 2011 Mar 28.
Article En | MEDLINE | ID: mdl-21251946

Reactive oxygen species (ROS) have been implicated in hepatic stellate cell activation and liver fibrosis. We previously reported that α-lipoic acid (LA) and its reduced form dihydrolipoic acid (DHLA) inhibited toxicant-induced inflammation and ROS generation. In the present study, we further examined the effects of LA/DHLA on thioacetamide (TAA)-induced liver fibrosis in rats and the possible underlying mechanisms in hepatic stellate cells in vitro. We found that co-administration of LA to rats chronically treated with TAA inhibited the development of liver cirrhosis, as indicated by reductions in cirrhosis incidence, hepatic fibrosis, and AST/ALT activities. We also found that DHLA inhibited TGF-ß/PDGF-stimulated HSC-T6 activation and ROS generation. These effects could be mediated by the MAPK and PI3K/Akt pathways. According to our current results, LA may have a beneficial role in the treatment of chronic liver diseases caused by ongoing hepatic damage.


Antioxidants/pharmacology , Cytokines/toxicity , Hepatic Stellate Cells/drug effects , MAP Kinase Signaling System/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Thioctic Acid/pharmacology , Animals , Antioxidants/therapeutic use , Becaplermin , Biomarkers/blood , Biomarkers/metabolism , Cell Line , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/metabolism , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/blood , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Male , Platelet-Derived Growth Factor/toxicity , Proto-Oncogene Proteins c-sis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recombinant Proteins/toxicity , Thioctic Acid/analogs & derivatives , Thioctic Acid/therapeutic use , Transforming Growth Factor beta/toxicity
20.
Spine (Phila Pa 1976) ; 36(3): E149-54, 2011 Feb 01.
Article En | MEDLINE | ID: mdl-21242879

STUDY DESIGN: In vivo and in vitro model. OBJECTIVE: Investigate soft-tissue inflammation caused by rhBMP-2. SUMMARY OF BACKGROUND DATA: Although rhBMP-2 produces excellent rates of fusion in the spine, dysphagia and respiratory compromise have occurred when used in the neck. The mechanism of the swelling and inflammatory response has yet to be fully elucidated. METHODS: ELISA kits (IL-6, IL-10, TNF-α) were used to measure cytokine levels at different concentrations of rhBMP-2. Absorbable collagen sponges were implanted with or without different concentrations of rhBMP-2 into the backs of rats subcutaneously (SC) and intramuscularly (IM). Magnetic resonance imaging was used to measure inflammation at 3 hours and 2, 4, and 7 days. The inflammatory volumes were measured and compared using MIPAV software. Rats were killed after 7 days and studied. RESULTS: IL-6, IL-10, and TNF-α release was dose-dependent. Soft-tissue edema after rhBMP-2 implantation was also dose-dependent, peaking at 3 hours SC, after SC and IM implantations, and on day 2 IM after IM implantation. All formed a granuloma-type mass after SC insertion. The mass was much larger in the 10 and 20 µg/10 µL (high-concentration) groups. The inflammatory response did not diffuse across physiologic barriers (subcutaneous fascia). Both high-dose groups were associated with encapsulated hematomas and a significant increase in the inflammatory zone. CONCLUSION: Swelling and inflammation after rhBMP-2 use are dose-dependent. Swelling may be due to direct contact as well as spread in the plane of access. The causes are a robust inflammatory reaction as well as sterile seroma and encapsulated hematoma formation.


Bone Morphogenetic Protein 2/toxicity , Disease Models, Animal , Hematoma/chemically induced , Hematoma/pathology , Seroma/chemically induced , Seroma/pathology , Transforming Growth Factor beta/toxicity , Animals , Bone Morphogenetic Protein 2/administration & dosage , Dose-Response Relationship, Drug , Inflammation/chemically induced , Inflammation/pathology , Neck/pathology , Rats , Rats, Inbred Lew , Recombinant Proteins/administration & dosage , Recombinant Proteins/toxicity , Rodentia , Transforming Growth Factor beta/administration & dosage
...