Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Inflammopharmacology ; 32(3): 1903-1928, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38630361

ABSTRACT

Penconazole (PEN) is a systemic triazole fungicide used to control various fungal diseases on grapes, stone fruits, cucurbits, and strawberries. Still, it leaves residues on treated crops after collection with many hazardous effects on population including neurotoxicity. Withania somnifera leaves extract (WSLE) is known for its memory and brain function enhancing ability. To evoke such action efficiently, WSLE bioactive metabolites are needed to cross the blood-brain barrier, that could limit the availability of such compounds to be localized within the brain. Therefore, in the present study, the association between PEN exposure and neurotoxicity was evaluated, and formulated WSLE nanoemulsion was investigated for improving the permeability of the plant extract across the blood-brain barrier. The rats were divided into five groups (n = 6). The control group was administered distilled water, group II was treated with W. somnifera leaves extract nanoemulsion (WSLE NE), group III received PEN, group IV received PEN and WSLE, and group V received PEN and WSLE NE. All rats were gavaged daily for 6 weeks. Characterization of compounds in WSLE using LC-MS/MS analysis was estimated. Neurobehavioral disorders were evaluated in all groups. Oxidative stress biomarkers, antioxidant enzyme activities, and inflammatory cytokines were measured in brain tissue. Furthermore, the gene expression patterns of GFAP, APP, vimentin, TGF-ß1, Smad2 and Bax were measured. Histopathological changes and immunohistochemical expression in the peripheral sciatic nerve and cerebral cortex were evaluated. A total of 91 compounds of different chemo-types were detected and identified in WSLE in both ionization modes. Our data showed behavioral impairment in the PEN-treated group, with significant elevation of oxidative stress biomarkers, proinflammatory cytokines, neuronal damage, and apoptosis. In contrast, the PEN-treated group with WSLE NE showed marked improvement in behavioral performance and histopathological alteration with a significant increase in antioxidant enzyme activity and anti-inflammatory cytokines compared to the group administered WSLE alone. The PEN-treated group with WSLE NE in turn significantly downregulated the expression levels of GFAP, APP, vimentin, TGF-ß1, Smad2 and Bax in brain tissue. In conclusion, WSLE NE markedly enhanced the permeability of plant extract constituents through the blood brain barrier to boost its neuroprotective effect against PEN-induced neurotoxicity.


Subject(s)
Neuroprotective Agents , Oxidative Stress , Plant Extracts , Plant Leaves , Signal Transduction , Smad2 Protein , Transforming Growth Factor beta1 , Withania , Animals , Male , Rats , Antioxidants/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Emulsions , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Plant Leaves/chemistry , Rats, Wistar , Signal Transduction/drug effects , Smad2 Protein/drug effects , Smad2 Protein/metabolism , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/metabolism , Withania/chemistry , Triazoles/pharmacology
2.
Anticancer Drugs ; 32(8): 786-792, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33675608

ABSTRACT

Stachydrine is a bioactive alkaloid that has been found to exert tumor-suppressive potential. However, the effect of stachydrine on hepatocellular carcinoma (HCC) has not been previously investigated. In the present study, we investigated the effect of transforming growth factor-ß1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) in HepG2 cells. Our results showed that stachydrine significantly suppressed TGF-ß1-induced HepG2 cell migration and invasion in a dose-dependent manner. Stachydrine prevented TGF-ß1-induced EMT in HepG2 cells, as proved by the increased expression level of E-cadherin and decreased expression levels of N-cadherin and vimentin. In addition, stachydrine attenuated TGF-ß1-induced upregulation of TGF-ß receptor I (TßRI) in both protein and mRNA levels. Further mechanism investigations proved that stachydrine prevented TGF-ß1-induced activation of Smad2/3 and phosphoinositol-3-kinase (PI3K)/Akt/mTOR signaling pathways in HepG2 cells. In conclusion, these findings demonstrated that stachydrine prevented TGF-ß1-induced EMT in HCC cells through Smad2/3 and PI3K/Akt/mTOR signaling pathways. Thus, stachydrine might be a potential therapeutic agent for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition/drug effects , Liver Neoplasms/pathology , Proline/analogs & derivatives , Transforming Growth Factor beta1/drug effects , Cadherins/drug effects , Cell Movement/drug effects , Cell Survival , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Phosphatidylinositol 3-Kinases , Proline/pharmacology , Signal Transduction/drug effects , Smad2 Protein/drug effects , TOR Serine-Threonine Kinases/drug effects , Vimentin/drug effects
3.
J Cell Physiol ; 234(3): 2606-2617, 2019 03.
Article in English | MEDLINE | ID: mdl-30317545

ABSTRACT

BACKGROUND: Tissue contraction and the extracellular matrix deposition are part of the pathogenesis of hypertrophic scars. The transcriptional factor NFE2L2 inhibits fibroblast differentiation in idiopathic pulmonary fibrosis and promotes myofibroblast dedifferentiation. Our previous study showed that the transcription factor NFE2L2 was strongly induced on treatment with arsenic trioxide (ATO). OBJECTIVE: The present study sought to investigate the effect of ATO on myofibroblast formation to determine its potential role in hypertrophic scar treatment. METHODS: Small interfering RNA against NFE2L2 was used on treatment with ATO in human skin myofibroblasts. The expression levels of fibrosis markers were assessed by reverse transcription polymerase chain reaction, western blot, and immunofluorescence staining. The transforming growth factor-ß1 (TGF-ß1)/Smad2/3 signaling was detected by western blot. A rabbit ear model was used to evaluate the antifibrotic role of ATO. RESULTS: At the cellular level, ATO abolished fibroblast differentiation in response to TGF-ß1. ATO reduced TGF-ß1-induced reactive oxygen species accumulation through increased expression of the antioxidant gene HO-1 in fibroblasts. In addition, ATO promoted the nuclear translocation of NFE2L2 and inhibited the phosphorylation of Smad2/3. In the rabbit ear model, ATO prevented the progression of hypertrophic scar formation. CONCLUSIONS: This study provides the first evidence implying that ATO inhibits the formation of myofibroblasts in vivo and in vitro and provides a possible treatment for hypertrophic scars.


Subject(s)
Arsenic Trioxide/pharmacology , Cell Differentiation/drug effects , Fibroblasts/drug effects , Myofibroblasts/drug effects , NF-E2-Related Factor 2/metabolism , Animals , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Humans , Myofibroblasts/cytology , Myofibroblasts/metabolism , NF-E2-Related Factor 2/drug effects , Rabbits , Signal Transduction/drug effects , Skin/metabolism , Smad2 Protein/drug effects , Smad3 Protein/drug effects
4.
Clin Sci (Lond) ; 132(4): 437-447, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29343616

ABSTRACT

Rapamycin, the macrolide immunosuppressant and active pharmaceutic in drug-eluting stents (DES), has a well-recognized antiproliferative action that involves inhibition of the mTOR pathway after binding to the cytosolic protein FKBP12. TGF receptor-type I (TGFRI) spontaneous activation is inhibited by the association with FKBP12. We hypothesized that rapamycin, in addition to inhibition of mTOR signaling, activates TGFRI independent of TGFß. Human umbilical vein endothelial cells (HUVECs) were treated with rapamycin (10 nmol/l) and/or TGFß RI kinase inhibitor (TGFRIi, 100 nmol/l) for 24 h. Rapamycin induced SMAD phosphorylation (SMAD1, SMAD2, and SMAD5) and PAI-1 up-regulation, which was specifically abrogated by SMAD2 knockdown. TGFRIi efficiently blocked phosphorylation of SMAD2, but not SMAD1/5. Interestingly, the inhibitor did not alter cell proliferation arrest induced by rapamycin. Active TGFß secretion was not affected by the treatment. Neutralizing TGFß experiments did not influence SMAD2 phosphorylation or PAI-1 expression indicating that activation of this pathway is independent of the ligand. In addition, rapamycin induction of endothelial-to-mesenchymal transition (EndMT) was potentiated by IL-1ß and efficiently blocked by TGFRIi. In vivo, the prothrombogenic effects of rapamycin and up-regulation of PAI-1 in murine carotid arteries were reduced by TGFRIi treatment. In conclusion, we provide evidence that rapamycin activates TGF receptor independent of its ligand TGFß, in concert with promotion of PAI-1 expression and changes in endothelial phenotype. These undesirable effects, the prothrombogenic state, and activation of EndMT are SMAD2-dependent and independent of the therapeutic rapamycin-induced cell proliferation arrest.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Receptors, Transforming Growth Factor beta/drug effects , Sirolimus/pharmacology , Smad2 Protein/drug effects , Carrier Proteins/drug effects , Humans , Phosphorylation/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects , Transforming Growth Factor beta/drug effects
5.
Osteoarthritis Cartilage ; 24(2): 315-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26318657

ABSTRACT

OBJECTIVE: The aetiology of OA is not fully understood although several adipokines such as leptin are known mediators of disease progression. Since leptin levels were increased in synovial fluid compared to serum in OA patients, it was suggested that joint cells themselves could produce leptin. However, exact mechanisms underlying leptin production by chondrocytes are poorly understood. Nevertheless, prednisolone, although displaying powerful anti-inflammatory properties has been recently reported to be potent stimulator of leptin and its receptor in OA synovial fibroblasts. Therefore, we investigated, in vitro, spontaneous and prednisolone-induced leptin production in OA chondrocytes, focusing on transforming growth factor-ß (TGFß) and Wnt/ß-catenin pathways. DESIGN: We used an in vitro dedifferentiation model, comparing human freshly isolated hip OA chondrocytes cultivated in monolayer during 1 day (type II, COL2A1 +; type X, COL10A1 + and type I collagen, COL1A1 -) or 14 days (COL2A1 -; COL10A1 - and COL1A1+). RESULTS: Leptin expression was not detected in day1 OA chondrocytes whereas day14 OA chondrocytes produced leptin, significantly increased with prednisolone. Activin receptor-like kinase 1 (ALK1)/ALK5 ratio was shifted during dedifferentiation, from high ALK5 and phospho (p)-Smad2 expression at day1 to high ALK1, endoglin and p-Smad1/5 expression at day14. Moreover, inactive glycogen synthase kinase 3 (GSK3) and active ß-catenin were only found in dedifferentiated OA chondrocytes. Smad1 and ß-catenin but not endoglin stable lentiviral silencing led to a significant decrease in leptin production by dedifferentiated OA chondrocytes. CONCLUSIONS: Only dedifferentiated OA chondrocytes produced leptin. Prednisolone markedly enhanced leptin production, which involved Smad1 and ß-catenin activation.


Subject(s)
Chondrocytes/metabolism , Leptin/metabolism , Osteoarthritis, Hip/metabolism , RNA, Messenger/metabolism , Activin Receptors, Type II/drug effects , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Adult , Aged , Aged, 80 and over , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/genetics , Chondrocytes/drug effects , Collagen Type X/drug effects , Collagen Type X/genetics , Collagen Type X/metabolism , Core Binding Factor Alpha 1 Subunit/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Glucocorticoids/pharmacology , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , In Vitro Techniques , Lymphotoxin-alpha/drug effects , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Male , Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Middle Aged , Osteoarthritis, Hip/genetics , Prednisolone/pharmacology , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/drug effects , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , SOX9 Transcription Factor/drug effects , SOX9 Transcription Factor/metabolism , Smad1 Protein/drug effects , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad2 Protein/drug effects , Smad2 Protein/genetics
6.
Osteoarthritis Cartilage ; 23(3): 478-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25529198

ABSTRACT

OBJECTIVE: Pain is the main problem for patients with osteoarthritis (OA). Pain is linked to inflammation, but in OA a subset of patients suffers from pain without inflammation, indicating an alternative source of pain. Nerve Growth Factor (NGF) inhibition is very efficient in blocking pain during OA, but the source of NGF is unclear. We hypothesize that damaged cartilage in OA releases Transforming Growth Factor-ß (TGF-ß), which in turn stimulates chondrocytes to produce NGF. DESIGN: Murine and human chondrocyte cell lines, primary bovine and human chondrocytes, and cartilage explants from bovine metacarpal joints and human OA joints were stimulated with TGF-ß1 and/or Interleukin-1 (IL-1)ß. We analyzed NGF expression on mRNA level with QPCR and stained human OA cartilage for NGF immunohistochemically. Cultures were additionally pre-incubated with inhibitors for TAK1, Smad2/3 or Smad1/5/8 signaling to identify the TGF-ß pathway inducing NGF. RESULTS: NGF expression was consistently induced in higher levels by TGF-ß than IL-1 in all of our experiments: murine, bovine and human origin, in cell lines, primary chondrocytes and explants cultures. TAK1 inhibition consistently reduced TGF-ß-induced NGF whereas it fully blocked IL-1ß-induced NGF expression. In contrast, ALK5-Smad2/3 inhibition fully blocked TGF-ß-induced NGF expression. Despite the large variation in basal NGF in human OA samples (mRNA and histology), TGF-ß exposure led to a consistent high level of NGF induction. CONCLUSION: We show for the first time that TGF-ß induces NGF expression in chondrocytes, in a ALK5-Smad2/3 dependent manner. This reveals a potential alternative non-inflammatory source of pain in OA.


Subject(s)
Cartilage, Articular/drug effects , Chondrocytes/drug effects , Interleukin-1beta/pharmacology , Nerve Growth Factor/drug effects , Osteoarthritis/metabolism , Pain/metabolism , RNA, Messenger/metabolism , Transforming Growth Factor beta1/pharmacology , Animals , Cartilage, Articular/metabolism , Cattle , Cell Line , Chondrocytes/metabolism , Humans , Mice , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Osteoarthritis/complications , Osteoarthritis/genetics , Pain/etiology , Pain/genetics , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/drug effects , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/drug effects , Smad3 Protein/genetics , Smad3 Protein/metabolism
7.
Australas J Dermatol ; 56(1): e7-14, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24645968

ABSTRACT

BACKGROUND/OBJECTIVES: The objective of this study was to investigate the effects of daidzein on collagen metabolism and its underlying mechanism in cultured skin fibroblast and nude mouse skin. METHODS: Skin fibroblasts were exposed to different concentrations of daidzein (0.5-50 µg/mL) for 24 h or 48 h, respectively. Female nude mice were treated topically with 200 µg/mL daidzein once a day for 6 weeks. Cell viability and cell cycle were determined by MTT and flow cytometer. The transcriptional activity of collagen type I was evaluated and the expression of procollagen, matrix metalloproteinase-1 (MMP1) and MMP2 were measured by real-time polymerase chain reaction. A Western blot analysis was applied to detect the levels of phosphorylated-Smad2 and Smad3. RESULTS: In the daidzein-treated cells the expression of type I procollagen increased markedly while the expressions of MMP1, and MMP2 was significantly inhibited. Additionally, the mouse skin showed more collagen deposition after daidzein treatment. The levels of transforming growth factor (TGF)-ß, phosphorylated-smad2 and smad3 were also higher in the daidzein treated skin fibroblasts than in the controls. CONCLUSIONS: The results showed that daidzein treatment can increase skin collagen synthesis and inhibit collagen degradation in vitro and in vivo. It seems that TGF-ß/smad signalling pathways play an important role in daidzein-induced collagen accumulation.


Subject(s)
Collagen Type I/drug effects , Fibroblasts/drug effects , Isoflavones/pharmacology , Phytoestrogens/pharmacology , Signal Transduction/drug effects , Animals , Cell Line , Cell Survival/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Female , Fibroblasts/metabolism , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 2/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Procollagen/genetics , Smad2 Protein/drug effects , Smad2 Protein/metabolism , Smad5 Protein/drug effects , Smad5 Protein/metabolism , Transcription, Genetic/drug effects , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism
8.
Andrologia ; 47(9): 1020-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25385280

ABSTRACT

Corpus cavernosum smooth muscle cells (CCSMCs) are important functional cells for penile erection. We evaluated the effect of transforming growth factor ß1 (TGFß1) and hepatocyte growth factor (HGF) on the viability and apoptosis of CCSMCs in vitro. CCSMCs from healthy male Sprague Dawley rats were randomly divided into four groups: a negative control group, a TGFß1 group, a HGF group and a HGF+ TGFß1 group. Differences in cell viability and apoptosis among groups were observed by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay and flow cytometry. Western blot was used to detect the change of apoptosis-related proteins. The level of reactive oxygen species (ROS) was detected by colorimetry. In the TGFß1 group, the MTT values were obviously decreased at 12 h, 24 h, 48 h-0.320, 0.383 and 0.432 respectively. However, compared with the normal group, the apoptosis index was markedly increased, reaching 26.86% at the 48-h time point. After TGFß1 treatment, the levels of cleaved caspase-3 and p-Smad2 were increased in the cells, but the levels of Bcl-xL, Bcl-2 and p-Akt were significantly lower. However, HGF co-treatment partially reversed these changes and could decrease the intracellular ROS level while increasing the Akt phosphorylation level. These results indicate that TGFß1 might induce apoptosis of CCSMCs in vitro and that HGF could interfere with the above process through downregulation of apoptosis signalling and oxidative stress reaction.


Subject(s)
Apoptosis/drug effects , Hepatocyte Growth Factor/pharmacology , Myocytes, Smooth Muscle/drug effects , Oxidative Stress/drug effects , Penis/cytology , Transforming Growth Factor beta1/pharmacology , Animals , Blotting, Western , Caspase 3/drug effects , Caspase 3/metabolism , Cell Survival/drug effects , In Vitro Techniques , Male , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , Smad2 Protein/drug effects , Smad2 Protein/metabolism , bcl-X Protein/drug effects , bcl-X Protein/metabolism
9.
Eur J Pharmacol ; 963: 176250, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38092315

ABSTRACT

Diabetic nephropathy (DN) is a serious concern in patients with diabetes mellitus. Prolonged hyperglycemia induces oxidative damage, chronic inflammation, and build-up of extracellular matrix (ECM) components in the renal cells, leading to kidney structural and functional changes. Imperatorin (IMP) is a naturally occurring furanocoumarin derivative with proven antioxidative and anti-inflammatory properties. We investigated whether IMP could improve DN and employed high glucose (HG)-induced HK-2 cells and high-fat diet-fed streptozotocin (HFD/STZ)-generated DN experimental model in C57BL/6 mice. In vitro, IMP effectively reduced the HG-activated reactive oxygen species generation, disturbance in the mitochondrial membrane potential (MMP) and epithelial-to-mesenchymal transition (EMT)-related markers, and the transforming growth factor (TGF)-ß and collagen 1 expression in HK-2 cells. In vivo, we found an elevation of serum creatinine, kidney histology alterations, and collagen build-up in the kidneys of the DN control group. Also, we found an altered expression of EMT-related markers, upregulation of the TGF-ß/Smad2/3 axis, and elevated pro-inflammatory molecules, TNF-α, IL-1ß, IL-18 and phospho-NF-kB (p65) in the DN control group. IMP treatment did not significantly reduce the blood glucose level compared to the DN control group. However, IMP treatment effectively improved renal damage by ameliorating kidney histological changes and serum renal injury markers. IMP treatment restored renal antioxidants and exhibited anti-inflammatory effects in the kidneys. Moreover, the abnormal manifestation of EMT-related attributes and elevated levels of TGF-ß, phospho-Smad2/3, and collagen 1 were also normalized in the IMP treatment group. Our findings highlight that IMP may be a potential candidate for treating DN.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Nephritis , Animals , Humans , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/metabolism , Collagen/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Fibrosis , Inflammation/drug therapy , Inflammation/metabolism , Kidney , Mice, Inbred C57BL , Nephritis/pathology , Transforming Growth Factor beta/metabolism , Smad2 Protein/drug effects , Smad2 Protein/metabolism , Smad3 Protein/drug effects , Smad3 Protein/metabolism , Epithelial-Mesenchymal Transition/drug effects , Furocoumarins/pharmacology , Furocoumarins/therapeutic use
10.
Biochem Biophys Res Commun ; 438(3): 551-6, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23916701

ABSTRACT

Human hair follicle mesenchymal stem cells (hHF-MSCs) are capable of differentiating into smooth muscle cells (SMCs) in response to transforming growth factor-ß (TGF-ß), and thus can be used for cardiovascular tissue engineering and regenerative medicine. However, the precise molecular mechanisms underlying SMC conversion of hHF-MSCs are still undefined. MicroRNAs (miRNAs) are small noncoding RNAs that modulate gene expression post-transcriptionally by binding to the complementary sequences of targeted mRNAs. Accumulating evidence indicates that miRNAs are associated with SMC differentiation in vitro andin vivo. In this study, we revealed that miR-18b was significantly downregulated during TGF-ß1-induced hHF-MSCs differentiation into SMC using miRNA array profiling and quantitative RT- PCR (qRT-PCR). Over-expression of miR-18b in hHF-MSCs led to remarkable downregulation of SMC-specific markers such as SMA and calponin proteins. On the contrary, inhibition of endogenous miR-18b by its antisense oligonucleotide antagomir-18b reversed the changes of SMA and calponin proteins. We also showed that SMAD2, a key transcription regulator in TGF-ß signaling which was involved in SMC differentiation, is regulated by miR-18b. miR-18b could suppress the expression of SMAD2 protein by targeting the 3'UTR of SMAD2 gene without affecting its mRNA level in hHF-MSCs. Moreover, knockdown of SMAD2 by RNA interference could block the effect of inhibition of miR-18b on SMC differentiation, indicating that SMAD2 contributed to miR-18b mediated regulation of TGF-ß-induced SMC differentiation. In conclusion, this study demonstrated that miR-18b regulated the TGF-ß1-induced differentiation of hHF-MSCs into SMCs by targeting SMAD2 gene, and provided novel insights into the regulatory mechanisms of TGF-ß-induced SMC differentiation.


Subject(s)
Hair Follicle/cytology , MicroRNAs/physiology , Myocytes, Smooth Muscle/cytology , Smad2 Protein/drug effects , Stem Cells/cytology , Cell Differentiation/drug effects , Gene Knockdown Techniques , Humans , RNA Interference , RNA, Messenger/metabolism , Stem Cells/metabolism , Transforming Growth Factor beta/pharmacology
11.
Cytokine ; 61(1): 285-96, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23134667

ABSTRACT

TGF-ß is a multifunctional cytokine that regulates cell proliferation, differentiation, apoptosis and extracellular matrix production. Deregulation of TGF-ß production or signaling plays a pivotal role in a variety of pathological processes such as cancer, metastasis, angiogenesis and fibrosis. Therefore, TGF-ß inhibitors should be promising therapeutic agents for the suppression of cancer progression and metastasis as well as fibrotic disorders. In a screening program of natural compounds from fungi inhibiting the TGF-ß dependent expression of a reporter gene in HepG2 cells, we found that the fungal lactones (S)-curvularin, dehydrocurvularin, oxacyclododecindione and galiellalactone inhibited the binding of the activated Smad2/3 transcription factors to the DNA and antagonized the cellular effects of TGF-ß including reporter gene activation and expression of TGF-ß induced genes in HepG2 and MDA-MB-231 cells. The most active compound oxacyclododecindione inhibited TGF-ß dependent reporter activity with IC50-values of 190-217 nM. In an in vitro angiogenesis assay, the fungal lactones strongly decreased the formation of capillary-like tubules of MDA-MB-231 cells on Matrigel.


Subject(s)
Lactones/pharmacology , Macrocyclic Compounds/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Zearalenone/analogs & derivatives , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/drug effects , Electrophoretic Mobility Shift Assay , Genes, Reporter/drug effects , Hep G2 Cells , Humans , Neovascularization, Physiologic/drug effects , Signal Transduction/drug effects , Smad2 Protein/drug effects , Smad2 Protein/metabolism , Smad3 Protein/drug effects , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Zearalenone/pharmacology
12.
J Periodontal Res ; 47(2): 149-58, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21883230

ABSTRACT

BACKGROUND AND OBJECTIVE: Ciclosporin A (CsA)-induced gingival overgrowth is attributed to an exaggerated accumulation of extracellular matrix, which is mainly due to an increased expression of transforming growth factor-ß1 (TGF-ß1). Herein, the in vitro investigation of effects of overexpression of Smad7, a TGF-ß1 signaling inhibitor, in the events associated with CsA-induced extracellular matrix accumulation was performed. MATERIAL AND METHODS: The effects of Smad7 were assessed by stable overexpression of Smad7 in fibroblasts from normal gingiva. Smad7-overexpressing cells and control cells were incubated with CsA, and synthesis of type I collagen, production and activity of MMP-2 and cellular proliferation were evaluated by ELISA, zymography, growth curve, bromodeoxyuridine incorporation assay and cell cycle analysis. The effects of CsA on cell viability and apoptosis of fibroblasts from normal gingiva were also evaluated. Western blot and immunofluorescence for phospho-Smad2 were performed to measure the activation of TGF-ß1 signaling. RESULTS: Although the treatment with CsA stimulated TGF-ß1 production in both control and Smad7-overexpressing fibroblasts, its signaling was markedly inhibited in Smad7-overexpressing cells, as revealed by low levels of phospho-Smad2. In Smad7-overexpressing cells, the effects of CsA on proliferation, synthesis of type I collagen and the production and activity of MMP-2 were significantly blocked. Smad7 overexpression blocked CsA-induced fibroblast proliferation via p27 regulation. Neither CsA nor Smad7 overexpression induced cell death. CONCLUSION: The data presented here confirm that TGF-ß1 expression is related to the molecular events associated with CsA-induced gingival overgrowth and suggest that Smad7 overexpression is effective in blocking these events, including proliferation, type I collagen synthesis and MMP-2 activity.


Subject(s)
Cyclosporine/adverse effects , Fibroblasts/drug effects , Gingiva/drug effects , Gingival Overgrowth/chemically induced , Smad7 Protein/pharmacology , Antimetabolites , Apoptosis/drug effects , Bromodeoxyuridine , Cell Culture Techniques , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Collagen Type I/drug effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclosporine/antagonists & inhibitors , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/genetics , Gingiva/cytology , Gingiva/metabolism , Humans , Male , Matrix Metalloproteinase 2/drug effects , Phosphorylation , Protein Kinase Inhibitors/metabolism , Signal Transduction/drug effects , Smad2 Protein/drug effects , Smad7 Protein/genetics , Transfection , Transforming Growth Factor beta1/antagonists & inhibitors , Young Adult
13.
Cell Biochem Funct ; 29(5): 371-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21491471

ABSTRACT

It is recognized that Wnt3a affects bone metabolism via the canonical Wnt/ß-catenin signalling pathway. We have previously shown that transforming growth factor-ß (TGF-ß) stimulates the synthesis of vascular endothelial growth factor (VEGF) via p44/p42 mitogen-activated protein (MAP) kinase, stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK) and p38 MAP kinase in osteoblast-like MC3T3-E1 cells. In the present study, we investigated the effect of Wnt3a on TGF-ß-stimulated VEGF synthesis in these cells. Wnt3a, which alone had little effect on the VEGF levels, significantly enhanced the TGF-ß-stimulated VEGF release. Lithium chloride and SB216763, inhibitors of glycogen synthase kinase 3ß, markedly amplified the TGF-ß-stimulated VEGF release. Wnt3a failed to affect the TGF-ß-induced phosphorylation of Smad2, p44/p42 MAP kinase, p38 MAP kinase or SAPK/JNK. Wnt3a and lithium chloride strengthened the VEGF mRNA expression induced by TGF-ß. These results strongly suggest that Wnt3a upregulates VEGF synthesis stimulated by TGF-ß via activation of the canonical pathway in osteoblasts.


Subject(s)
Osteoblasts/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Wnt Proteins/metabolism , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Indoles/pharmacology , Lithium Chloride/pharmacology , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase 4/metabolism , Maleimides/pharmacology , Mice , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Osteoblasts/drug effects , Phosphorylation/drug effects , RNA, Messenger , Signal Transduction , Smad2 Protein/drug effects , Smad2 Protein/metabolism , Transforming Growth Factor beta/pharmacology , Up-Regulation , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics , Wnt Proteins/pharmacology , Wnt3 Protein , Wnt3A Protein , beta Catenin/metabolism , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Biomed Pharmacother ; 137: 111318, 2021 May.
Article in English | MEDLINE | ID: mdl-33556875

ABSTRACT

Cardiac fibrosis plays an important role in hypertension-related contractile dysfunction and heart failure. Qingda granule (QDG), derived from the Qingxuan Jiangya decoction, has been used clinically for more than 60 years to treat hypertension. However, the effect of QDG on hypertensive cardiac fibrosis remains largely unknown. The objective of this study was to investigate the effect of QDG on cardiac fibrosis and explore the underlying mechanism in vivo and in vitro. For in vivo experiments, 30 male spontaneously hypertensive rats were randomly divided into groups that received no QDG or one of three doses (0.45, 0.9 or 1.8 g/kg/day). Positive-control animals received valsartan (VAL, 7.2 mg/kg/day). Treatments were administered by gavage for 10 weeks. All three doses of QDG and VAL led to significantly lower blood pressure than in SHR animals. Besides, all three doses of QDG and VAL attenuated pathological changes in SHR animals. However, only intermediate, high concentrations of QDG and VAL led to significantly lower left ventricle ejection fraction and left ventricle fractional shortening than in SHR animals. Therefore, the minimum and effective QDG dose (intermediate concentration of QDG) was selected for subsequent animal experiments in this study. Our results showed that intermediate concentration of QDG also significantly mitigated the increases in levels of α-smooth muscle actin (α-SMA), proliferating cell nuclear antigen (PCNA), collagen III, transforming growth factor-ß1 (TGF-ß1) and in the ratio of phospho-Smad2/3 to total Smad2/3 protein in cardiac tissue, based on immunohistochemistry, Western blotting, and Masson staining. For in vitro experiments, primary cardiac fibroblasts were stimulated with 100 nM angiotensin II in the presence or absence of QDG. And we tested different concentrations of QDG (3.125, 6.25, 12.5, 25, 50 µg/mL) in the cell viability experiment. Our results showed that 3.125, 6.25 and 12.5 µg/mL of QDG treatment for 24 h didn't affect the cell viability of cardiac fibroblasts. Consistently, QDG at 6.25 or 12.5 µg/mL significantly reduced cell viability and down-regulated α-SMA in primary cardiac fibroblasts were stimulated with 100 nM angiotensin II. Therefore, QDG at 12.5 µg/mL was chosen for the following cell experiment. Our results showed that QDG at 12.5 µg/mL alleviated the increase of PCNA, collagen Ⅲ, TGF-ß1 expression, and the ratio of phospho-Smad2/3 to total Smad2/3 protein. Our studies in vitro and in vivo suggest that QDG reduces blood pressure and cardiac fibrosis as well as protecting cardiac function, and that it exerts these effects in part by suppressing TGF-ß1/Smad2/3 signaling.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Myocardium/pathology , Signal Transduction/drug effects , Smad2 Protein/drug effects , Smad3 Protein/drug effects , Transforming Growth Factor beta1/drug effects , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Blood Pressure/drug effects , Dose-Response Relationship, Drug , Echocardiography , Fibrosis , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Valsartan/therapeutic use
15.
Anal Cell Pathol (Amst) ; 2021: 8883427, 2021.
Article in English | MEDLINE | ID: mdl-33628711

ABSTRACT

Keloids are scars characterized by abnormal proliferation of fibroblasts and overproduction of extracellular matrix components including collagen. We previously showed that LY2109761, a transforming growth factor- (TGF-) ß receptor inhibitor, suppressed the secretion of matrix components and slowed the proliferation of fibroblasts derived from human hypertrophic scar tissue. However, the exact mechanism underlying this effect remains unclear. Here, we replicated the above results in keloid-derived fibroblasts and show that LY2109761 promoted apoptosis, decreased the phosphorylation of Smad2 and Smad3, and suppressed TGF-ß1. These results suggest that the development and pathogenesis of keloids are positively regulated by the Smad2/3 signaling pathway and the upregulation of TGF-ß1 receptors. LY2109761 and other inhibitors of these processes may therefore serve as therapeutic targets to limit excessive scarring after injury.


Subject(s)
Fibroblasts/drug effects , Keloid/metabolism , Pyrazoles/pharmacology , Pyrroles/pharmacology , Adult , Apoptosis/drug effects , Cells, Cultured , Female , Fibroblasts/metabolism , Humans , Male , Smad2 Protein/drug effects , Smad2 Protein/metabolism , Smad3 Protein/drug effects , Smad3 Protein/metabolism , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/metabolism
16.
Ulus Travma Acil Cerrahi Derg ; 16(4): 293-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20849043

ABSTRACT

BACKGROUND: This prospective, controlled experimental study was planned to investigate the effects of levosimendan on transforming growth factor (TGF)-beta3 and Smad1, Smad2 and Smad3 expression in the early stages of sepsis. METHODS: Twenty-four rats were randomized into four groups: (1) sham-operated controls, (2) dobutamine group--subjected to abdominal hypertension and peritonitis-induced sepsis using cecal ligation and puncture (CLP), then treated with 10 microg x kg(-1) min(-1) intravenous (IV) dobutamine infusion, (3) levosimendan group--as in 2, then treated with levosimendan IV bolus 200 microg x kg(-1) followed by 200 microg x kq(-1) min(-1) IV infusion, and (4) a control group as in 2, with no treatment. All rats were killed 8 hours after CLP. Aorta tissue samples were analyzed by immunohistochemical staining. RESULTS: CLP caused mild interleukin (IL)-1 immunostaining in both control and dobutamine groups. Immunoreactivity of tumor necrosis factor (TNF)-alpha was mild in both sham and control groups. TGF-beta3 immunostaining was mildly increased in groups sham, control and dobutamine, whereas it was found moderate in group levosimendan. Smad1, Smad2 and Smad3 were found moderately increased only in group levosimendan. CONCLUSION: Beneficial effects of levosimendan on hemodynamics and global oxygen transport were reported in experimental and clinical trials. Besides its potency on C++ ion sensitivity, it should influence inflammatory cytokine production by diminishing TGF-beta3 and Smad1, Smad2 and Smad3 expression.


Subject(s)
Aorta/physiology , Hydrazones/pharmacology , Pyridazines/pharmacology , Sepsis/physiopathology , Transforming Growth Factor beta3/physiology , Animals , Aorta/drug effects , Aorta/physiopathology , Blood Pressure/drug effects , Dopamine/pharmacology , Male , Rats , Rats, Wistar , Sepsis/genetics , Simendan , Smad1 Protein/drug effects , Smad1 Protein/genetics , Smad2 Protein/drug effects , Smad2 Protein/genetics , Smad3 Protein/drug effects , Smad3 Protein/genetics , Transforming Growth Factor beta3/drug effects , Transforming Growth Factor beta3/genetics , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Vasodilator Agents/pharmacology
17.
Acta Histochem ; 122(7): 151600, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33066828

ABSTRACT

OBJECTIVES: Aim of this study is to explore whether quercetin can inhibit the enlarged fibrogenic responses of endometrial stromal cells by increasing the level of microRNA-145 (miR-145) and mediating the TGFß1/Smad2/Smad3 signaling pathway, and to discuss the mechanism of signal transduction, further to provide experimental basis for revealing the pathophysiological mechanism and seeking new strategies for effective prevention and treatment of endometrial fibrosis. METHODS: The expression levels of miR-145 and TGF-ß receptor 2 (TGFBR2) were detected by RT-qPCR analysis. Expressions of α-smooth muscle actin (α-SMA) and vimentin were examined by immunofluorescence staining. Cell viability was measured by MTT assay. The protein expression of collagen type 1 alpha 1 (Col1a1), α-SMA, fibronectin (FN), TGFBR2, transforming growth factor (TGF-ß1), Smad2/3, phospho-Smad2/3 (p-Smad2/3) were detected by western blot analysis. The interaction between miR-145 and TGFBR2 was confirmed by dual-luciferase reporter gene assay. RESULTS: The expression level of miR-145 was decreased, whereas TGFBR2 was increased in intrauterine adhesion tissue. The expression levels of COL1A1, α-SMA, FN, TGFBR2, and p-Smad2/3 were increased, whereas miR-145 and cell proliferation were decreased in human endometrial stromal cells (hESCs) in response to TGF-ß1 stimulation in a time and dose-dependent manner, which could be reversed by quercetin. Furthermore, quercetin regulates cell fibrogenic responses of endometrial stromal cells via miR-145/TGF-ß1/Smad2/Smad3 pathway. CONCLUSIONS: These findings indicated that quercetin have a significant anti-fibrotic effect and could upregulate miR-145 and inhibit activation of TGF-ß1/Smad2/Smad3 pathway to regulate TGF-ß1 induced fibrogenic responses of endometrial stromal cells, which may serve as a potential therapeutic agent for endometrial fibrosis.


Subject(s)
MicroRNAs/drug effects , Quercetin/pharmacology , Receptor, Transforming Growth Factor-beta Type II/drug effects , Smad2 Protein/drug effects , Smad3 Protein/drug effects , Adult , Female , Humans , Male , Stromal Cells/drug effects , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/genetics
18.
Acta Histochem ; 122(7): 151603, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33066831

ABSTRACT

Dermal papilla cells (DPCs), an important component of hair follicles, its proliferation and apoptosis directly regulate and maintain the growth of hair follicles. All-trans-retinoic acid (ATRA) plays a critical role in hair growth. In this study, the effects of ATRA on cultured mink hair follicle growth were studied by administration of different concentrations of ATRA for 12 days in vitro. In addition, the proliferation and apoptosis of DPCs were measured after treating with ATRA. The mRNA and protein levels of hair follicle growth associated factor transforming growth factor-ß2 (TGF-ß2) and the phosphorylation levels of Smad2/3 were determined. Moreover, TGF-ß type I and type II receptor inhibitor LY2109761 and specific inhibitor of Smad3 (SIS3) were administered to investigate the underlying molecular mechanism. The results showed that ATRA inhibited hair follicle growth, promoted TGF-ß2 expression and activated phosphorylation of Smad2/3. In addition, ATRA inhibited cell proliferation by arresting the cell cycle at G1 phase and induced apoptosis of DPCs by enhancing the ratio of Bax/Bcl-2 and promoted the cleavage of caspase-3. Furthermore, LY2109761 or SIS3 partially reversed the decreased cell viability, increased apoptosis that were induced by ATRA. In conclusion, ATRA could inhibit hair follicle growth via inhibiting proliferation and inducing apoptosis of DPCs partially through the TGF-ß2/Smad2/3 pathway.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Hair Follicle/metabolism , Smad2 Protein/drug effects , Tretinoin/pharmacology , Animals , Cell Cycle/drug effects , Male , Mink/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects
19.
Respir Physiol Neurobiol ; 276: 103412, 2020 05.
Article in English | MEDLINE | ID: mdl-32044448

ABSTRACT

Allergic asthma is a chronic inflammatory disease characterized by airflow obstruction, airway hyperresponsiveness (AHR), airway inflammation, and mucus overproduction. Cordyceps polysaccharide (CPS) is one of the main bioactive compounds of Cordyceps militarisis, a traditional Chinese medicine. In this study, we established a mouse model of asthma using ovalbumin (OVA) challenge and evaluated the potential regulatory effect of CPS (25, 50, and 100 mg/kg) on asthmatic mice. These results showed that the asthmatic mice treated with CPS suppressed the secretion of eotaxin, IL-4, IL-5, IL-13, and IFN-γ in the blood and bronchoalveolar lavage fluid (BALF), and decreased serum IgE levels compared to the vehicle-treated mice. CPS also alleviated inflammatory cell infiltration, goblet cell hyperplasia, and the increases of inflammatory cells in the mouse model of asthma. In addition, OVA-induced AHR was inhibited by CPS treatment. Further analyses of protein expression revealed that CPS inhibited the activation of transforming growth factor ß1 (TGF-ß1)/Smad pathway in mice with asthma. These findings indicated that CPS might serve as a potential therapeutic agent for the management of allergic asthma.


Subject(s)
Asthma/metabolism , Cordyceps , Fungal Polysaccharides/pharmacology , Lung/drug effects , Smad2 Protein/drug effects , Smad3 Protein/drug effects , Transforming Growth Factor beta1/drug effects , Animals , Asthma/chemically induced , Asthma/physiopathology , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Interleukin-5/metabolism , Lung/metabolism , Lung/physiopathology , Medicine, Chinese Traditional , Mice , Ovalbumin , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/physiopathology , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
20.
Physiol Rep ; 8(1): e14343, 2020 01.
Article in English | MEDLINE | ID: mdl-31925944

ABSTRACT

One of the key characteristics of idiopathic pulmonary fibrosis (IPF) is accumulation of excess fibrous tissue in the lung, which leads to hypoxic conditions. Transforming growth factor (TGF) ß is a major mediator that promotes the differentiation of fibroblasts to myofibroblasts. However, how hypoxia and TGFß together contribute the pathogenesis of IPF is poorly understood. Long non-coding RNAs (lncRNAs) have regulatory effects on certain genes and are involved in many diseases. In this study, we determined the effects of hypoxia and/or TGFß on mRNA and lncRNA transcriptomes in pulmonary fibroblasts. Hypoxia and TGFß1 synergistically increased myofibroblast marker expression. RNA sequencing revealed that hypoxia and TGFß1 treatment resulted in significant changes in 669 lncRNAs and 2,676 mRNAs compared to 150 lncRNAs and 858 mRNAs in TGFß1 alone group and 222 lncRNAs and 785 mRNAs in hypoxia alone group. TGFß1 induced the protein expression of HIF-1α, but not HIF-2α. On the other hand, hypoxia enhanced the TGFß1-induced phosphorylation of Smad3, suggesting a cross-talk between these two signaling pathways. In all, 10 selected lncRNAs (five-up and five-down) in RNA sequencing data were validated using real-time PCR. Two lncRNAs were primarily located in cytoplasm, three in nuclei and five in both nuclei and cytoplasm. The silencing of HIF-1α and Smad3, but not Smad2 and HIF-2α rescued the downregulation of FENDRR by hypoxia and TGFß1. In conclusion, hypoxia and TGFß1 synergistically regulate mRNAs and lncRNAs involved in several cellular processes, which may contribute to the pathogenesis of IPF.


Subject(s)
Fibroblasts/metabolism , Hypoxia/genetics , Idiopathic Pulmonary Fibrosis/genetics , Myofibroblasts/metabolism , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Transforming Growth Factor beta1/physiology , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Fibroblasts/drug effects , Gene Expression Regulation , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/cytology , Myofibroblasts/drug effects , Phosphorylation/drug effects , RNA, Long Noncoding/drug effects , RNA, Long Noncoding/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Smad2 Protein/drug effects , Smad2 Protein/metabolism , Smad3 Protein/drug effects , Smad3 Protein/metabolism , Transcriptome/drug effects , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL