1.Saltwater stir-fried Plantaginis Semen alleviates renal fibrosis by regulating epithelial-mesenchymal transition in renal tubular cells.
Xin-Lei SHEN ; Qing-Ru ZHU ; Wen-Kai YU ; Li ZHOU ; Qi-Yuan SHAN ; Yi-Hang ZHANG ; Yi-Ni BAO ; Gang CAO
China Journal of Chinese Materia Medica 2025;50(5):1195-1208
This study aimed to investigate the effect of saltwater stir-fried Plantaginis Semen(SPS) on renal fibrosis in rats and decipher the underlying mechanism. Thirty-six Sprague-Dawley rats were randomly assigned into control, model, losartan potassium, and low-, medium-, and high-dose(15, 30, and 60 g·kg~(-1), respectively) SPS groups. Rats in other groups except the control group were subjected to unilateral ureteral obstruction(UUO) to induce renal fibrosis, and the modeling and gavage lasted for 14 days. After 14 consecutive days of treatment, the levels of serum creatinine(Scr) and blood urea nitrogen(BUN) in rats of each group were determined by an automatic biochemical analyzer. Hematoxylin-eosin(HE) and Masson staining were used to evaluate pathological changes in the renal tissue. Western blot and immunofluorescence assay were conducted to determine the protein levels of fibronectin(FN), collagen Ⅰ, vimentin, and α-smooth muscle actin(α-SMA) in the renal tissue. The mRNA levels of epithelial-mesenchymal transition(EMT)-associated transcription factors including twist family bHLH transcription factor 1(TWIST1), snail family transcriptional repressor 1(SNAI1), and zinc finger E-box binding homeobox 1(ZEB1), as well as inflammatory cytokines such as interleukin-1β(IL-1β), interleukin-6(IL-6), and tumor necrosis factor-α(TNF-α), were determined by RT-qPCR. Human renal proximal tubular epithelial(HK2) cells exposed to transforming growth factor-β(TGF-β) for the modeling of renal fibrosis were used to investigate the inhibitory effect of SPS on EMT. Network pharmacology and Western blot were employed to explore the molecular mechanism of SPS in alleviating renal fibrosis. The results showed that SPS significantly reduced Scr and BUN levels and alleviated renal injury and collagen deposition in UUO rats. Moreover, SPS notably down-regulated the protein levels of FN, collagen Ⅰ, vimentin, and α-SMA as well as the mRNA levels of SNAI1, ZEB1, TWIST1, IL-1β, IL-6, and TNF-α in the kidneys of UUO rats and TGF-β-treated HK-2 cells. In addition, compared with Plantaginis Semen without stir-frying with saltwater, SPS showed increased content of specific compounds, which were mainly enriched in the mitogen-activated protein kinase(MAPK) signaling pathway. SPS significantly inhibited the phosphorylation of extracellular signal-regulated kinase(ERK) and p38 MAPK in the kidneys of UUO rats and TGF-β-treated HK2 cells. In conclusion, SPS can alleviate renal fibrosis by attenuating EMT through inhibition of the MAPK signaling pathway.
Animals
;
Epithelial-Mesenchymal Transition/drug effects*
;
Rats, Sprague-Dawley
;
Male
;
Rats
;
Fibrosis/genetics*
;
Drugs, Chinese Herbal/administration & dosage*
;
Kidney Diseases/pathology*
;
Kidney Tubules/pathology*
;
Humans
2.Scleromitrion diffusum reverses epithelial-mesenchymal transi-tion of gastric mucosa in rats with gastric precancerous lesions.
Luping MA ; Xin ZUO ; Weikai ZHU ; Jiyan LI ; Yanyan ZHAO ; Jingyuan ZHANG ; Hui SHEN
Journal of Zhejiang University. Medical sciences 2025;54(3):342-349
OBJECTIVES:
To investigate the effect of Scleromitrion diffusum on gastric mucosal epithelial-mesenchymal transition (EMT) in rats with gastric precancerous lesion.
METHODS:
Fifty SD rats were randomly divided into blank control group (n=11), model control group (n=13), Scleromitrion diffusum (SD) group (n=13) and vitase group (n=13). Gastric precancerous lesion animal model was prepared by 1-methyl-3-nitro-1-nitrosoguanidine complex polyfactor method, and the drugs were administrated by gavage once a day for 6 weeks. The pathological changes of gastric mucosa were observed with hematoxylin and eosin staining, the expression of EMT marker proteins were detected with immunohistochemical staining and Western blotting.
RESULTS:
Compared with the model control group, the gastric mucosal injury was significantly attenuated in the Scleromitrion diffusum group, the mucosal tissue structure gradually recovered, the saccular expansion area was reduced, and the inflammatory infiltration was ameliorated. The expression of epithelial cadherin was higher, and the expression of neural cadherin and vimentin in the Scleromitrion diffusum group were lower than those of model control group (all P<0.05).
CONCLUSIONS
Scleromitrion diffusum can ameliorate gastric mucosal injury in rats with gastric precancerous lesion by reversing the EMT.
Animals
;
Rats
;
Rats, Sprague-Dawley
;
Epithelial-Mesenchymal Transition/drug effects*
;
Precancerous Conditions/metabolism*
;
Gastric Mucosa/metabolism*
;
Stomach Neoplasms/drug therapy*
;
Male
;
Cadherins/metabolism*
3.Epithelial-mesenchymal Transition: Biological Basis and Clinical Prospects of Lung Cancer Invasion, Metastasis, and Drug Resistance.
Hengxing SUN ; Mengting XIONG ; Shuanshuan XIE ; Jing WEN
Chinese Journal of Lung Cancer 2025;28(2):155-164
Lung cancer is the leading cause of cancer-related deaths worldwide, characterized by high incidence and mortality rates. The primary reasons for treatment failure in lung cancer patients are tumor invasion and drug resistance, particularly resistance to chemotherapeutic agents and epidermal growth factor receptor (EGFR) mutant targeted therapy, which considerably undermine the therapeutic outcomes for those with advanced lung cancer. Epithelial-mesenchymal transition (EMT) serves as a crucial biological process closely associated with physiological or pathological processes such as tissue embryogenesis, organogenesis, wound repair, and tumor invasion. Numerous studies have indicated that EMT, mediated through various signaling pathways, plays a pivotal role in the initiation, progression, and metastasis of lung cancer, while it is also closely associated with drug resistance in lung cancer cells. Therefore, research focusing on the molecular mechanisms and pathophysiology related to EMT can contribute to reversing drug resistance in drug treatment for lung cancer, thereby improving prognosis. This article reviews the progress in research on EMT in the invasion, metastasis, and drug resistance of lung cancer based on relevant domestic and international literature.
Humans
;
Epithelial-Mesenchymal Transition/drug effects*
;
Drug Resistance, Neoplasm
;
Lung Neoplasms/physiopathology*
;
Neoplasm Metastasis
;
Neoplasm Invasiveness
;
Animals
;
Antineoplastic Agents/therapeutic use*
4.Astragaloside IV delayed the epithelial-mesenchymal transition in peritoneal fibrosis by inhibiting the activation of EGFR and PI3K-AKT pathways.
Ying HUANG ; Chen-Ling CHU ; Wen-Hui QIU ; Jia-Yi CHEN ; Lu-Xi CAO ; Shui-Yu JI ; Bin ZHU ; Guo-Kun WANG ; Quan-Quan SHEN
Journal of Integrative Medicine 2025;23(6):694-705
OBJECTIVE:
Peritoneal fibrosis (PF) is an adverse event that occurs during long-term peritoneal dialysis, significantly impairing treatment efficiency and adversely affecting patient outcomes. Astragaloside IV (AS-IV), a principal active component derived from Astragalus membranaceus (Fisch.) Bunge, has exhibited anti-inflammatory and antifibrotic effects in various settings. This study aims to investigate the potential therapeutic efficacy and mechanism of AS-IV in the treatment of PF.
METHODS:
The PF mouse model was established by intraperitoneal injection of 4.25% peritoneal dialysis fluid (100 mL/kg). The epithelial-mesenchymal transition (EMT) of HMrSV5 cells was induced by the addition of 10 ng/mL transforming growth factor β (TGF-β). The differentially expressed genes in HMrSV5 cells treated with AS-IV were screened using transcriptome sequencing analysis. The potential targets of AS-IV were screened using network pharmacology and analyzed using molecular docking and molecular dynamics simulations.
RESULTS:
Administration of AS-IV at doses of 20, 40, or 80 mg/kg effectively mitigated the increase in peritoneal thickness and the development of fibrosis in mice with PF. The expression of the fibrosis marker α-smooth muscle actin in the peritoneum was significantly decreased in AS-IV-treated mice. The treatment of AS-IV (10, 20, and 40 μmol/L) significantly delayed the EMT of HMrSV5 cells induced by TGF-β, as demonstrated by the decreased number of 5-ethynyl-2'-deoxyuridine-positive cells, reduced migrated area, and decreased expression of fibrosis markers. A total of 460 differentially expressed genes were detected in AS-IV-treated HMrSV5 cells through transcriptome sequencing, with notable enrichment in the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)-AKT serine/threonine kinase 1 (AKT) signaling pathway. The reduced levels of phosphorylated PI3K (p-PI3K) and p-AKT were detected in HMrSV5 cells with AS-IV treatment. Epidermal growth factor receptor (EGFR) was predicted as a direct target of AS-IV, exhibiting strong hydrogen bond interactions. The activation of the PI3K-AKT pathway by the compound 740Y-P, and the activation of the EGFR pathway by NSC 228155 each partially counteracted the inhibitory effect of AS-IV on the EMT of HMrSV5 cells.
CONCLUSION
AS-IV delayed the EMT process in peritoneal mesothelial cells and slowed the progression of PF, potentially serving as a therapeutic agent for the early prevention and treatment of PF. Please cite this article as: Huang Y, Chu CL, Qiu WH, Chen JY, Cao LX, Ji SY, Zhu B, Wang GK, Shen QQ. Astragaloside IV delayed the epithelial-mesenchymal transition in peritoneal fibrosis by inhibiting the activation of EGFR and PI3K-AKT pathways. J Integr Med. 2025; 23(6):694-705.
Epithelial-Mesenchymal Transition/drug effects*
;
Animals
;
Saponins/pharmacology*
;
Triterpenes/pharmacology*
;
Mice
;
Peritoneal Fibrosis/pathology*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
ErbB Receptors/metabolism*
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Signal Transduction/drug effects*
;
Male
;
Humans
;
Molecular Docking Simulation
;
Cell Line
;
Mice, Inbred C57BL
5.Wenxia Changfu Formula inhibits NSCLC metastasis by halting TAMs-induced epithelial-mesenchymal transition via antagonisticallymodulating CCL18.
Qianyu BI ; Mengran WANG ; Li LUO ; Beiying ZHANG ; Siyuan LV ; Zengna WANG ; Xuming JI
Chinese Journal of Natural Medicines (English Ed.) 2025;23(7):838-847
Our previous research demonstrated that the Wenxia Changfu Formula (WCF), as a neoadjuvant therapy, inhibits M2 macrophage infiltration in the tumor microenvironment and prevents lung cancer metastasis. Given tumor-associated macrophages (TAMs) in epithelial-mesenchymal transition (EMT), this study investigated whether WCF impedes lung cancer metastasis by attenuating TAM-induced EMT in non-small cell lung cancer (NSCLC) cells. Utilizing a co-culture model treated with or without WCF, we observed that WCF downregulated cluster of differentiation 163 (CD163) expression in macrophages, reduced CCL18 levels in the conditioned medium, and inhibited the growth, invasion, and EMT of NSCLC cells induced by macrophage co-culture. Manipulation of CCL18 levels and Src overexpression in NSCLC cells revealed that WCF's effects are mediated through CCL18 and Src signaling. In vivo, WCF inhibited recombinant CCL18 (rCCL18)-induced tumor metastasis in nude mice by blocking Src signaling. These findings indicate that WCF inhibits NSCLC metastasis by impeding TAM-induced EMT via antagonistic modulation of CCL18, providing evidence for its potential development and clinical application in NSCLC patients.
Epithelial-Mesenchymal Transition/drug effects*
;
Carcinoma, Non-Small-Cell Lung/metabolism*
;
Humans
;
Animals
;
Lung Neoplasms/metabolism*
;
Chemokines, CC/antagonists & inhibitors*
;
Mice
;
Mice, Nude
;
Drugs, Chinese Herbal/administration & dosage*
;
Cell Line, Tumor
;
Neoplasm Metastasis
;
Tumor-Associated Macrophages/drug effects*
;
Mice, Inbred BALB C
;
Signal Transduction/drug effects*
6.Sanhuang Xiexin Decoction Ameliorates TNBC By Modulating JAK2-STAT3 and Lipid Metabolism.
Ying QI ; Xin-Jie WU ; Jing-Bin SHI ; Xiao-Wei SHI ; Na ZHAO ; Yang XIONG ; Li-Pei WANG
Chinese journal of integrative medicine 2024;30(12):1080-1089
OBJECTIVE:
To investigate the therapeutic effect of Sanhuang Xiexin Decoction (SXD) on triple-negative breast cancer (TNBC) in mice and its underlying mechanism.
METHODS:
The high-performance liquid chromatography (HPLC) was used to quantitate and qualify SXD. A total of 15 female BALB/c mice were inoculated subcutaneously on the right hypogastrium with 3×105 of 4T1-Luc cells to establish TNBC mouse model. All mice were divided randomly into 3 groups, including phosphate buffered solution (PBS), SXD and doxorubicin (DOX) groups (positive drug). Additionally, tumor growth, pathological changes, serum lipid profiles, expression of Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signaling pathway and its key targets including inflammatory factors, cell cycle and epithelial-mesenchymal transition (EMT) markers were investigated. Besides, the biosafety of SXD was also evaluated in mice.
RESULTS:
Rhein, coptisine, berberine hydrochloride and baicalin were all found in SXD, and the concentrations of these 4 components were 0.57, 2.61, 2.93, and 46.04 mg/g, respectively. The mouse experiment showed that SXD could notably suppress the development of tumors and reduce the density of tumor cells (P<0.01). The serum lipid analysis and Oil-Red-O staining both showed the differences, SXD group exhibited higher serum adiponectin and HDL-C levels with lower TC and LDL-C levels compared to the PBS and DOX groups (P<0.05 or P<0.01), respectively. SXD also decreased the levels of phospho-JAK2 (p-JAK2), phospho-STAT3 (p-STAT3) expressions and its downstream factors, including mostly inflammatory cytokine, EMT markers, S phase of tumor cells and vascular endothelial growth factor (VEGF) expression (P<0.05 or P<0.01), respectively. The biosafety assessment of SXD revealed low levels of toxicity in mice.
CONCLUSION
SXD could inhibit TNBC by suppressing JAK2-STAT3 phosphorylation which may be associated with modulation of lipid metabolism.
Animals
;
STAT3 Transcription Factor/metabolism*
;
Drugs, Chinese Herbal/pharmacology*
;
Janus Kinase 2/metabolism*
;
Lipid Metabolism/drug effects*
;
Female
;
Mice, Inbred BALB C
;
Triple Negative Breast Neoplasms/metabolism*
;
Cell Line, Tumor
;
Epithelial-Mesenchymal Transition/drug effects*
;
Signal Transduction/drug effects*
;
Mice
7.Danshen Injection inhibits peritoneal dialysis fluid-induced endothelial-mesenchymal transition in HMrSV5 cells by regulating the TGF-β/Smad signaling pathway.
Lihua YU ; Jingya LI ; Xiaoqi WANG ; Li LI ; Ya CHEN ; Feiyu WANG ; Kun ZHANG ; Tongsheng WANG
Journal of Southern Medical University 2024;44(12):2276-2282
OBJECTIVES:
To investigate the inhibitory effect of Danshen Injection on endothelial-mesenchymal transition (EndMT) induced by peritoneal dialysis fluid in HMrSV5 cells and the role of the TGF‑β/Smad signaling pathway in mediating this effect.
METHODS:
HMrSV5 cells cultured in 40% peritoneal dialysis solution for 72 h to induce EndMT were treated with 0.05%, 0.1% and 0.5% Danshen Injection. CCK-8 assay was used to assess the changes in viability of the treated cells, and the levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), and vascular endothelial growth factor (VEGF) in the cell supernatant were detected using ELISA; Western blotting was performed to detect the protein expressions of E-cadherin, α-smooth muscle actin (α-SMA), p-Smad 2/3, and Smad 7 in the cells.
RESULTS:
Culture in 40% peritoneal dialysis fluid for 72 induced significant EndMT in HMrSV5 cells, which exhibited obviously lowered cell viability. Danshen Injection within the concentration range of 0.025%-1.5% did not significantly affect the viability of the cells. Exposure of HMrSV5 cells to peritoneal dialysis fluid for 72 h significantly increased the production of IL-6, TNF‑α, TGF‑β and VEGF, upregulated the protein expressions of α‑SMA and p-Smad 2/3, and lowered the expressions of E-cadherin and Smad7 proteins. Treatment of the exposed cells with Danshen injection significantly increased cell viability and cellular expressions of E-cadherin and Smad 7 proteins and reduced the production of IL-6, TNF-α, TGF-β and VEGF and the protein expressions of α‑SMA and p-Smad 2/3.
CONCLUSIONS
Danshen Injection can suppress peritoneal dialysis fluid-induced EndMT in HMrSV5 cells possibly by regulating the TGF-β/Smad signaling pathway.
Signal Transduction/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
;
Transforming Growth Factor beta/metabolism*
;
Humans
;
Peritoneal Dialysis/adverse effects*
;
Salvia miltiorrhiza
;
Epithelial-Mesenchymal Transition/drug effects*
;
Smad Proteins/metabolism*
;
Vascular Endothelial Growth Factor A/metabolism*
;
Cell Line
;
Tumor Necrosis Factor-alpha/metabolism*
;
Interleukin-6/metabolism*
;
Cadherins/metabolism*
;
Actins/metabolism*
;
Dialysis Solutions
;
Endothelial-Mesenchymal Transition
8.The novel anthraquinone compound Kanglexin prevents endothelial-to-mesenchymal transition in atherosclerosis by activating FGFR1 and suppressing integrin β1/TGFβ signaling.
Yixiu ZHAO ; Zhiqi WANG ; Jing REN ; Huan CHEN ; Jia ZHU ; Yue ZHANG ; Jiangfei ZHENG ; Shifeng CAO ; Yanxi LI ; Xue LIU ; Na AN ; Tao BAN ; Baofeng YANG ; Yan ZHANG
Frontiers of Medicine 2024;18(6):1068-1086
Endothelial-mesenchymal transition (EndMT) disrupts vascular endothelial integrity and induces atherosclerosis. Active integrin β1 plays a pivotal role in promoting EndMT by facilitating TGFβ/Smad signaling in endothelial cells. Here, we report a novel anthraquinone compound, Kanglexin (KLX), which prevented EndMT and atherosclerosis by activating MAP4K4 and suppressing integrin β1/TGFβ signaling. First, KLX effectively counteracted the EndMT phenotype and mitigated the dysregulation of endothelial and mesenchymal markers induced by TGFβ1. Second, KLX suppressed TGFβ/Smad signaling by inactivating integrin β1 and inhibiting the polymerization of TGFβR1/2. The underlying mechanism involved the activation of FGFR1 by KLX, resulting in the phosphorylation of MAP4K4 and Moesin, which led to integrin β1 inactivation by displacing Talin from its β-tail. Oral administration of KLX effectively stimulated endothelial FGFR1 and inhibited integrin β1, thereby preventing vascular EndMT and attenuating plaque formation and progression in the aorta of atherosclerotic Apoe-/- mice. Notably, KLX (20 mg/kg) exhibited superior efficacy compared with atorvastatin, a clinically approved lipid-regulating drug. In conclusion, KLX exhibited potential in ameliorating EndMT and retarding the formation and progression of atherosclerosis through direct activation of FGFR1. Therefore, KLX is a promising candidate for the treatment of atherosclerosis to mitigate vascular endothelial injury.
Animals
;
Atherosclerosis/prevention & control*
;
Mice
;
Receptor, Fibroblast Growth Factor, Type 1/metabolism*
;
Signal Transduction/drug effects*
;
Anthraquinones/pharmacology*
;
Humans
;
Integrin beta1/metabolism*
;
Epithelial-Mesenchymal Transition/drug effects*
;
Male
;
Transforming Growth Factor beta/metabolism*
;
Disease Models, Animal
;
Mice, Inbred C57BL
;
Human Umbilical Vein Endothelial Cells/drug effects*
9.Morin, a matrix metalloproteinase 9 inhibitor, attenuates endothelial-to-mesenchymal transition in atherosclerosis by downregulating Notch-1 signaling.
Yuan HE ; Xiao-Xuan QIN ; Ming-Wei LIU ; Wei SUN
Journal of Integrative Medicine 2024;22(6):683-695
OBJECTIVE:
Atherosclerotic cardiovascular disease poses a significant health challenge globally. Recent findings highlight the pivotal role of the endothelial-to-mesenchymal transition (EndMT) in atherosclerosis. Morin is a bioflavonoid mainly extracted from white mulberry, a traditional Chinese herbal medicine with anti-inflammatory and antioxidant properties. This study examines whether morin can alleviate atherosclerosis by suppressing EndMT and seeks to elucidate the underlying mechanism.
METHODS:
We induced an in vitro EndMT model in human umbilical vein endothelial cells (HUVECs) by stimulating the cells with transforming growth factor-β1 (TGF-β1) (10 ng/mL) for 48 h. The in vivo experiments were performed in an atherosclerosis model using apolipoprotein E (ApoE)-/- mice fed with a high-fat diet (HFD). Mice in the intervention group were given morin (50 mg/kg) orally for 4 weeks. Molecular docking and microscale thermophoresis were assayed to understand the interactions between morin and matrix metalloproteinase-9 (MMP-9).
RESULTS:
Morin inhibited the expression of EndMT markers in a dose-dependent manner in TGF-β1-treated HUVECs. Administering 50 μmol/L morin suppressed the upregulation of MMP-9 and Notch-1 signaling in TGF-β1-induced EndMT. Moreover, the overexpression of MMP-9 activated Notch-1 signaling, thereby reversing morin's inhibitory effect on EndMT. In the HFD-induced atherosclerotic ApoE-/- mice, morin notably reduced aortic intimal hyperplasia and plaque formation by suppressing EndMT. Furthermore, morin demonstrated a strong binding affinity for MMP-9.
CONCLUSION
Morin acts as an MMP-9 inhibitor to disrupt EndMT in atherosclerosis by limiting the activation of Notch-1 signaling. This study underscores morin's potential utility in the development of anti-atherosclerotic medication. Please cite this article as: He Y, Qin XX, Liu MW, Sun W. Morin, a matrix metalloproteinase 9 inhibitor, attenuates endothelial-to-mesenchymal transition in atherosclerosis by downregulating Notch-1 Signaling. J Integr Med. 2024; 22(6): 684-696.
Flavonoids/pharmacology*
;
Animals
;
Atherosclerosis/metabolism*
;
Humans
;
Receptor, Notch1/genetics*
;
Signal Transduction/drug effects*
;
Matrix Metalloproteinase 9/genetics*
;
Human Umbilical Vein Endothelial Cells/drug effects*
;
Mice
;
Epithelial-Mesenchymal Transition/drug effects*
;
Down-Regulation/drug effects*
;
Male
;
Transforming Growth Factor beta1/genetics*
;
Matrix Metalloproteinase Inhibitors/pharmacology*
;
Mice, Inbred C57BL
;
Flavones
10.Research progress of anti-fibrotic drugs that inhibit epithelial-mesenchymal transition in pulmonary fibrosis.
Li Bing ZHANG ; Na ZHAO ; Qi Ying NONG
Chinese Journal of Industrial Hygiene and Occupational Diseases 2023;41(1):72-77
Pulmonary fibrosis is the end-stage pathological change of lung diseases, which seriously affects the respiratory function of human body. A large number of studies at home and abroad have confirmed that epithelial-mesenchymal transition (EMT) is an important intermediate stage in the development of pulmonary fibrosis. Inhibition of multiple pathways upstream and downstream of EMT, such as the classical Smads pathway and non-Smads pathway of TGF-1 can effectively inhibit the process of EMT and alleviate pulmonary fibrosis. This article will review the main conclusions of the mechanism of action of EMT as a target to improve the pathology of pulmonary fibrosis so far, and provide a theoretical basis and research direction for further research and development of anti-pulmonary fibrosis drugs.
Humans
;
Epithelial-Mesenchymal Transition/drug effects*
;
Fibrosis/drug therapy*
;
Pulmonary Fibrosis/pathology*
;
Signal Transduction
;
Transforming Growth Factor beta1/metabolism*
;
Antifibrotic Agents/therapeutic use*

Result Analysis
Print
Save
E-mail