Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.008
Filter
1.
Mol Biol Rep ; 51(1): 1031, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39352573

ABSTRACT

BACKGROUND: Fatty liver disease is a metabolic disorder that recently has been classified into two categories: metabolic dysfunction-associated fatty liver disease (MAFLD) and non-MAFLD. TGF-ß signaling pathway is likely a significant factor in the pathogenesis of this condition, exerting its effects through its downstream signaling proteins, Smad2/3. Accordingly, this study aimed to investigate the TGF-ß signaling pathway in the white blood cells (WBCs) of patients with MAFLD compared to those with non-MAFLD and control groups. METHODS AND RESULTS: In this study, 41 patients with fatty liver were evaluated, comprising 22 patients with MAFLD and 19 patients with non-MAFLD, and compared to 22 healthy controls. Gene expression of TGF-ß1, TGF-ß3, and CTGF were quantified using qRT-PCR, and the protein expressions of Smad2/3 and P-Smad2/3 were analyzed using western blotting. Gene expression analysis revealed a significant decrease in the gene expressions of the TGF-ß1 and TGF-ß3 and an increase in CTGF gene expression in patients with MAFLD and non-MAFLD compared to the control group. Notably, the Smad2/3 protein expression was significantly higher in the non-MAFLD group compared to the control group (P < 0.05). On the other hand, the P-smad2/3 protein expression was significantly elevated in the MAFLD group compared to the control group (P < 0.001). CONCLUSIONS: TGF-ß signaling pathway in WBCs of patients with fatty liver are affected by a complex signaling pathway. However, metabolic factors most probably affect TGF-ß1 gene expression and its downstream signaling proteins more than TGF-ß3.


Subject(s)
Connective Tissue Growth Factor , Signal Transduction , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta1 , Humans , Male , Case-Control Studies , Female , Middle Aged , Adult , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Fatty Liver/metabolism , Fatty Liver/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Smad Proteins/metabolism , Smad Proteins/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/genetics , Gene Expression Regulation
2.
F S Sci ; 5(1): 16-23, 2024 Feb.
Article in English | MEDLINE | ID: mdl-39382269

ABSTRACT

OBJECTIVE: To investigate the direct effect of growth differentiation factor 9 (GDF9) on androgen production in human theca cells. DESIGN: Experimental study. SETTING: Tertiary hospital-based research laboratory. PATIENT(S): Women who underwent in vitro fertilization and intracytoplasmic sperm injections at our clinic were included in this study. INTERVENTION(S): Primary cultured human theca cells from women undergoing in vitro fertilization and intracytoplasmic sperm injection treatment were treated with GDF9, an activin receptor-like kinase 5 (ALK5) inhibitor, and a SMAD4 agonist. MAIN OUTCOME MEASURE(S): The expression of androgen synthesis-related genes StAR, CYP17A1, and LHCGR, levels of androstenedione and testosterone, phosphorylation of SMAD2/3, and the interaction between bone morphogenic protein-activated type II receptor and ALK5 were evaluated using reverse transcription-quantitative polymerase chain reaction, Western blot, enzyme-linked immunosorbent assays, and coimmunoprecipitation assays, respectively. RESULT(S): Growth differentiation factor 9 decreased StAR, CYP17A1, and LHCGR expression levels in human theca cells, which was prevented by treatment with the ALK5 inhibitor, and suppressed production of androgen in human theca cells. Growth differentiation factor 9 increased SMAD2/3 phosphorylation, and the ALK5 inhibitor also suppressed this effect. Bone morphogenic protein-activated type II receptor and ALK5 bound to each other after GDF9 stimulation. The SMAD4 agonist kartogenin also decreased messenger RNA levels of StAR and CYP17A1 and protein levels of StAR in human theca cells. CONCLUSION(S): Growth differentiation factor 9 can activate the bone morphogenic protein-activated type II receptor-ALK5-SMAD2/3 signaling pathway, suppress CYP17A1 expression, and decrease androgen production in human theca cells.


Subject(s)
Growth Differentiation Factor 9 , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta , Smad2 Protein , Smad3 Protein , Steroid 17-alpha-Hydroxylase , Theca Cells , Humans , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Theca Cells/metabolism , Theca Cells/drug effects , Female , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/genetics , Growth Differentiation Factor 9/metabolism , Growth Differentiation Factor 9/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Androgens/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , Smad4 Protein/metabolism , Smad4 Protein/genetics , Phosphorylation/drug effects , Cells, Cultured , Oocytes/metabolism , Oocytes/drug effects , Androstenedione/metabolism , Testosterone/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction/drug effects
3.
Anticancer Res ; 44(10): 4435-4448, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39348958

ABSTRACT

BACKGROUND/AIM: Predicting lymph node metastasis (LNM) in early gastric cancer (EGC) is crucial for making treatment decisions. This study aimed to confirm risk factors for LNM and identify novel auxiliary biomarkers for predicting LNM in EGC. PATIENTS AND METHODS: We established a training set, comprising 63 patients with LNM-EGC and 274 patients with non-LNM EGC, and a test set, comprising 19 patients with LNM-EGC and 146 non-LNM EGC. Immunohistochemistry for lymphangiogenic and related pathway components (VEGF-C, TGF-ß1, SMAD2/3, VEGF-D, pSTAT3, E-cadherin, CD44, c-MET, YAP, and HER2), in situ hybridization for Epstein-Barr virus-encoded small RNAs, and multiplex PCR for microsatellite instability were conducted. RESULTS: In the training set, Lauren's diffuse/mixed classification, stromal desmoplasia, submucosal invasion ≥500 µm, lymphatic invasion, and high VEGF-C and SMAD2/3 expression were independent risk factors for LNM (p<0.05). A large tumor size, mixed histology, submucosal invasion, perineural invasion, and ulceration were determined as risk factors using univariate analysis (p<0.05). The tumor cutoff size for predicting LNM was 2.65 cm, based on a ROC analysis. The test set study verified that stromal desmoplasia, submucosal invasion, and high VEGF-C expression were independent risk factors for LNM (p<0.05). Moreover, mixed histology, lymphatic invasion, ulceration, and high SMAD 2/3 expression were identified as additional risk factors using univariate analysis (p<0.05). CONCLUSION: Stromal desmoplasia, submucosal invasion, and high VEGF-C expression are potential biomarkers for LNM in EGC. VEGF-C expression might serve as an adjunct biomarker for predicting LNM on forceps-biopsy tissue at initial diagnosis.


Subject(s)
Biomarkers, Tumor , Herpesvirus 4, Human , Lymphatic Metastasis , Microsatellite Instability , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/virology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Male , Female , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Middle Aged , Herpesvirus 4, Human/genetics , Aged , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/pathology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Adult , Smad2 Protein/metabolism , Smad2 Protein/genetics , Risk Factors
4.
FASEB J ; 38(17): e70045, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39259551

ABSTRACT

Epithelial ovarian cancer is the deadliest gynecologic malignancy, characterized by high metastasis. Transforming growth factor-ß1 (TGF-ß1) drives epithelial- mesenchymal transformation (EMT), a key process in tumor metastasis. Tumor necrosis factor-α-induced protein 8 (TNFAIP8)-like 2 (TIPE2) acts as a negative regulator of innate and adaptive immunity and involves in various cancers. However, its relationship with TGF-ß1 in ovarian cancer and its role in reversing TGF-ß1-induced EMT remain unclear. This study examined TIPE2 mRNA and protein expression using quantitative RT-PCR (qRT-PCR), western blot and immunohistochemistry. The effects of TIPE2 overexpression and knockdown on the proliferation, migration and invasion of epithelial ovarian cancer cells were assessed through 5-ethynyl-2-deoxyuridine, colony-forming, transwell migration and invasion assays. The relationship between TIPE2 and TGF-ß1 was investigated using qRT-PCR and enzyme-linked immunosorbent assay, while the interaction between TIPE2 and Smad2 was identified via co-immunoprecipitation. The results revealed that TIPE2 protein was significantly down-regulated in epithelial ovarian cancer tissues and correlated with the pathological type of tumor, patients' age, tumor differentiation degree and FIGO stage. TIPE2 and TGF-ß1 appeared to play an opposite role to each other during the progression of human ovarian cancer cells. Furthermore, TIPE2 inhibited the metastasis and EMT of ovarian cancer cells by combining with Smad2 in vitro or in an intraperitoneal metastasis model. Consequently, these findings suggest that TIPE2 plays a crucial inhibitory role in ovarian cancer metastasis by modulating the TGF-ß1/Smad2/EMT signaling pathway and may serve as a potential target for ovarian cancer, providing important direction for future diagnostic and therapeutic strategies.


Subject(s)
Carcinoma, Ovarian Epithelial , Cell Movement , Epithelial-Mesenchymal Transition , Intracellular Signaling Peptides and Proteins , Ovarian Neoplasms , Smad2 Protein , Transforming Growth Factor beta1 , Smad2 Protein/metabolism , Smad2 Protein/genetics , Humans , Female , Transforming Growth Factor beta1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Cell Line, Tumor , Animals , Mice , Neoplasm Invasiveness , Cell Proliferation , Gene Expression Regulation, Neoplastic , Mice, Nude , Mice, Inbred BALB C , Signal Transduction
5.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39337713

ABSTRACT

Glial-cell-line-derived neurotrophic factor (GDNF) is highly expressed and is involved in the malignant phenotype in glioblastomas (GBMs). However, uncovering its underlying mechanism for promoting GBM progression is still a challenging work. In this study, we found that serine protease inhibitor family E member 1 (SERPINE1) was a potential downstream gene of GDNF. Further experiments confirmed that SERPINE1 was highly expressed in GBM tissues and cells, and its levels of expression and secretion were enhanced by exogenous GDNF. SERPINE1 knockdown inhibited the migration and invasion of GBM cells promoted by GDNF. Mechanistically, GDNF increased SERPINE1 by promoting the phosphorylation of SMAD2/3. In vivo experiments demonstrated that GDNF facilitated GBM growth and the expressions of proteins related to migration and invasion via SERPINE1. Collectively, our findings revealed that GDNF upregulated SERPINE1 via the SMAD2/3-signaling pathway, thereby accelerating GBM cell migration and invasion. The present work presents a new mechanism of GDNF, supporting GBM development.


Subject(s)
Cell Movement , Glial Cell Line-Derived Neurotrophic Factor , Glioblastoma , Neoplasm Invasiveness , Plasminogen Activator Inhibitor 1 , Signal Transduction , Animals , Humans , Male , Mice , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Mice, Nude , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/genetics , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics
6.
Nat Cardiovasc Res ; 3(7): 869-882, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39196175

ABSTRACT

Differentiation of cardiac fibroblasts to myofibroblasts is necessary for matrix remodeling and fibrosis in heart failure. We previously reported that mitochondrial calcium signaling drives α-ketoglutarate-dependent histone demethylation, promoting myofibroblast formation. Here we investigate the role of ATP-citrate lyase (ACLY), a key enzyme for acetyl-CoA biosynthesis, in histone acetylation regulating myofibroblast fate and persistence in cardiac fibrosis. We show that inactivation of ACLY prevents myofibroblast differentiation and reverses myofibroblasts towards quiescence. Genetic deletion of Acly in post-activated myofibroblasts prevents fibrosis and preserves cardiac function in pressure-overload heart failure. TGFß stimulation enhances ACLY nuclear localization and ACLY-SMAD2/3 interaction, and increases H3K27ac at fibrotic gene loci. Pharmacological inhibition of ACLY or forced nuclear expression of a dominant-negative ACLY mutant prevents myofibroblast formation and H3K27ac. Our data indicate that nuclear ACLY activity is necessary for myofibroblast differentiation and persistence by maintaining histone acetylation at TGFß-induced myofibroblast genes. These findings provide targets to prevent and reverse pathological fibrosis.


Subject(s)
ATP Citrate (pro-S)-Lyase , Cell Differentiation , Fibrosis , Histones , Myofibroblasts , Smad2 Protein , Myofibroblasts/metabolism , Myofibroblasts/drug effects , ATP Citrate (pro-S)-Lyase/metabolism , ATP Citrate (pro-S)-Lyase/genetics , Animals , Fibrosis/metabolism , Cell Differentiation/drug effects , Histones/metabolism , Smad2 Protein/metabolism , Smad2 Protein/genetics , Acetylation/drug effects , Cell Nucleus/metabolism , Cell Nucleus/drug effects , Smad3 Protein/metabolism , Smad3 Protein/genetics , Cells, Cultured , Chromatin/metabolism , Mice, Knockout , Transforming Growth Factor beta/metabolism , Disease Models, Animal , Signal Transduction , Mice, Inbred C57BL , Male , Mice , Gene Expression Regulation/drug effects
7.
Exp Cell Res ; 442(1): 114193, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39103072

ABSTRACT

The liver is innervated by primary sensory nerve fibres releasing the neuropeptide calcitonin gene-related peptide (CGRP). Elevated plasma levels of CGRP have been found in patients with liver fibrosis or cirrhosis. We hypothesised that signalling of CGRP and its receptors might regulate liver fibrosis and propose a novel potential target for the treatment. In this study, hepatic expression of CGRP and its receptor component, the receptor activity-modifying protein 1 (RAMP1), was dramatically increased in diseased livers of patients. In a murine liver fibrosis model, deficiency of RAMP1 resulted in attenuated fibrogenesis characterized by less collagen deposition and decreased activity of hepatic stellate cells (HSC). Mechanistically, activity of the TGFß1 signalling core component Smad2 was severely impaired in the absence of RAMP1, and Yes-associated protein (YAP) activity was found to be diminished in RAMP1-deficient liver parenchyma. In vitro, stimulation of the HSC line LX-2 cells with CGRP induces TGFß1 production and downstream signalling as well as HSC activation documented by increased α-SMA expression and collagen synthesis. We further demonstrate in LX-2 cells that CGRP promotes YAP activation and its nuclear translocation subsequent to TGFß1/Smad2 signals. These data support a promotive effect of CGRP signalling in liver fibrosis via stimulation of TGFß1/Smad2 and YAP activity.


Subject(s)
Calcitonin Gene-Related Peptide , Hepatic Stellate Cells , Liver Cirrhosis , Receptor Activity-Modifying Protein 1 , Signal Transduction , Smad2 Protein , Transforming Growth Factor beta1 , YAP-Signaling Proteins , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Animals , Transforming Growth Factor beta1/metabolism , Receptor Activity-Modifying Protein 1/metabolism , Receptor Activity-Modifying Protein 1/genetics , Humans , Smad2 Protein/metabolism , Smad2 Protein/genetics , Mice , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Male , Mice, Inbred C57BL , Transcription Factors/metabolism , Transcription Factors/genetics , Mice, Knockout
8.
FASEB J ; 38(15): e23495, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39126242

ABSTRACT

Hepatic stellate cell (HSC) activation is the essential pathological process of liver fibrosis (LF). The molecular mechanisms regulating HSC activation and LF are incompletely understood. Here, we explored the effect of transcription factor SRY-related high mobility group box 7 (SOX7) on HSC activation and LF, and the underlying molecular mechanism. We found the expression levels of SOX7 were decreased in human and mouse fibrotic livers, particularly at the fibrotic foci. SOX7 was also downregulated in primary activated HSCs and TGF-ß1 stimulated LX-2 cells. SOX7 knockdown promoted activation and proliferation of LX-2 cells while inhibiting their apoptosis. On the other hand, overexpression of SOX7 suppressed the activation and proliferation of HSCs. Mechanistically, SOX7 attenuates HSC activation and LF by decreasing the expression of ß-catenin and phosphorylation of Smad2 and Smad3 induced by TGF-ß1. Furthermore, overexpression of SOX7 using AAV8-SOX7 mouse models ameliorated the extent of LF in response to CCl4 treatment in vivo. Collectively, SOX7 suppressed HSC activation and LF. Targeting SOX7, therefore, could be a potential novel strategy to protect against LF.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , SOXF Transcription Factors , Hepatic Stellate Cells/metabolism , Animals , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Humans , Male , SOXF Transcription Factors/metabolism , SOXF Transcription Factors/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cell Proliferation , Mice, Inbred C57BL , beta Catenin/metabolism , beta Catenin/genetics , Apoptosis , Smad2 Protein/metabolism , Smad2 Protein/genetics , Cell Line , Smad3 Protein/metabolism , Smad3 Protein/genetics
9.
Adv Sci (Weinh) ; 11(33): e2402804, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38953462

ABSTRACT

Understanding the regulation of normal erythroid development will help to develop new potential therapeutic strategies for disorders of the erythroid lineage. Cellular repressor of E1A-stimulated genes 1 (CREG1) is a glycoprotein that has been implicated in the regulation of tissue homeostasis. However, its role in erythropoiesis remains largely undefined. In this study, it is found that CREG1 expression increases progressively during erythroid differentiation. In zebrafish, creg1 mRNA is preferentially expressed within the intermediate cell mass (ICM)/peripheral blood island (PBI) region where primitive erythropoiesis occurs. Loss of creg1 leads to anemia caused by defective erythroid differentiation and excessive apoptosis of erythroid progenitors. Mechanistically, creg1 deficiency results in reduced activation of TGF-ß/Smad2 signaling pathway. Treatment with an agonist of the Smad2 pathway (IDE2) could significantly restore the defective erythroid development in creg1-/- mutants. Further, Klf1, identified as a key target gene downstream of the TGF-ß/Smad2 signaling pathway, is involved in creg1 deficiency-induced aberrant erythropoiesis. Thus, this study reveals a previously unrecognized role for Creg1 as a critical regulator of erythropoiesis, mediated at least in part by the TGF-ß/Smad2-Klf1 axis. This finding may contribute to the understanding of normal erythropoiesis and the pathogenesis of erythroid disorders.


Subject(s)
Erythropoiesis , Kruppel-Like Transcription Factors , Signal Transduction , Smad2 Protein , Transforming Growth Factor beta , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Smad2 Protein/metabolism , Smad2 Protein/genetics , Erythropoiesis/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Signal Transduction/genetics , Cell Differentiation/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
10.
Mol Cell Endocrinol ; 592: 112326, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38972346

ABSTRACT

Activin E activates brown and beige adipocytes and has been controversially implicated as a factor that induces obesity and fatty liver. Here, we sought to address this controversial issue by producing recombinant human activin E to evaluate its effects on HB2 brown adipocytes in vitro. Activin E increased uncoupling protein 1 (Ucp1) and fibroblast growth factor 21 (Fgf21) mRNA expression in the adipocytes. This upregulation was suppressed by SB431542, an inhibitor of activin receptor-like kinase (Alk) TGF-ß type I receptors. SB431542 also inhibited the activin E-induced phosphorylation of Smad2/3. A promoter assay using a CAGA-Luc reporter and Alk expression vectors revealed that activin E activated the TGF-ß/activin pathway via Alk7. The upregulation of Ucp1 and Fgf21 mRNA might be mediated through Alk7 and Smad2/3 phosphorylation. Activin E is a potential stimulator of energy expenditure by activating brown adipocytes and highlights its potential as a therapeutic target for treating obesity.


Subject(s)
Activin Receptors, Type I , Activins , Adipocytes, Brown , Dioxoles , Fibroblast Growth Factors , Uncoupling Protein 1 , Up-Regulation , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , Humans , Up-Regulation/drug effects , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/genetics , Activins/metabolism , Phosphorylation/drug effects , Dioxoles/pharmacology , Signal Transduction/drug effects , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Cell Line , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Benzamides
11.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38960622

ABSTRACT

A pleiotropic immunoregulatory cytokine, TGF-ß, signals via the receptor-regulated SMADs: SMAD2 and SMAD3, which are constitutively expressed in normal cells. Here, we show that selective repression of SMAD3 induces cDC differentiation from the CD115+ common DC progenitor (CDP). SMAD3 was expressed in haematopoietic cells including the macrophage DC progenitor. However, SMAD3 was specifically down-regulated in CD115+ CDPs, SiglecH- pre-DCs, and cDCs, whereas SMAD2 remained constitutive. SMAD3-deficient mice showed a significant increase in cDCs, SiglecH- pre-DCs, and CD115+ CDPs compared with the littermate control. SMAD3 repressed the mRNA expression of FLT3 and the cDC-related genes: IRF4 and ID2. We found that one of the SMAD transcriptional corepressors, c-SKI, cooperated with phosphorylated STAT3 at Y705 and S727 to repress the transcription of SMAD3 to induce cDC differentiation. These data indicate that STAT3 and c-Ski induce cDC differentiation by repressing SMAD3: the repressor of the cDC-related genes during the developmental stage between the macrophage DC progenitor and CD115+ CDP.


Subject(s)
Cell Differentiation , DNA-Binding Proteins , Dendritic Cells , Proto-Oncogene Proteins , STAT3 Transcription Factor , Smad3 Protein , Animals , Mice , Dendritic Cells/metabolism , Dendritic Cells/cytology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Signal Transduction , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , DNA-Binding Proteins/metabolism
12.
Mol Biol Rep ; 51(1): 831, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39037563

ABSTRACT

BACKGROUND: While many genes linked to colorectal cancer (CRC) contribute to cancer development, a thorough investigation is needed to explore crucial hub genes yet to be fully studied. A pivotal pathway in CRC is transforming growth factor-beta (TGF-ß). This study aimed to assess SMAD2 and SMAD4 gene expression from this pathway. METHODS AND RESULTS: Counted data from the Cancer Genome Atlas (TCGA) were examined, comparing 483 tumor and 41 normal samples. Using clinical data, genes impacting overall survival (OS) were evaluated. GSE39582 was employed to confirmed the levels of genes in CRC compared to the normal samples. Additionally, employing unhealthy samples and the RT-qPCR means our outcomes was validated. Finally, PharmacoGx information were utilized to connect the levels of potential genes to drug tolerance and susceptibility. Our findings showed SMAD2 and SMAD4 levels in TGF-ß signaling were more significant than other pathway genes. Our findings indicated that the protein levels of these genes were lower in malignant tissues than in healthy tissues. Results revealed a significant correlation between low levels of SMAD2 and unfavorable OS in CRC individuals. RT-qPCR results demonstrated decreased expressions of both SMAD2 and SMAD4 in cancer tissues compared to elevated levels in adjacent normal samples. Our results showed significant association between selected genes and immune cell infiltration markers such as CD8+, and B-cells. Our results indicated a potential association among the levels of SMAD2 and SMAD4 genes and tolerance and susceptibility to Nilotinib and Panobinostat drugs. CONCLUSION: Reduced expression of SMAD2 and SMAD4 may be pivotal in CRC progression, impacting downstream genes unrelated to patient OS. These findings suggest a potential role for SMAD2 and SMAD4 as predictive markers for drug response in CRC patients.


Subject(s)
Biomarkers, Tumor , Colorectal Neoplasms , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Smad2 Protein , Smad4 Protein , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Smad4 Protein/genetics , Smad4 Protein/metabolism , Smad2 Protein/genetics , Smad2 Protein/metabolism , Prognosis , Gene Expression Regulation, Neoplastic/drug effects , Drug Resistance, Neoplasm/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Signal Transduction/genetics , Signal Transduction/drug effects , Male , Down-Regulation/genetics , Down-Regulation/drug effects , Female
13.
Genes (Basel) ; 15(7)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39062724

ABSTRACT

TGF-ß1/Smads is a classic signaling pathway, which plays important roles in the development process of organisms. Black porgy Acanthopagrus schlegelii and red porgy Pagrus major are valuable economic fishes, and their hybrid offspring show excellent heterosis traits. Yet the molecular regulation mechanism of the heterosis traits is less clear. Here, we explored the TGF-ß1/Smads pathway's molecular genetic information for heterosis in A. schlegelii ♂ × P. major ♀ (AP) and A. schlegelii ♀ × P. major ♂ (PA) in terms of growth and development. The mRNA expression levels of TGF-ß1, TßR-I, TßR-II, and Smad2 genes in different developmental stages of A. schlegelii were detected. Furthermore, the expression levels of TGF-ß1, TßR-I, TßR-II, and Smad2 genes in different tissues of adult (mRNA level) and larva (mRNA and protein level) of A. schlegelii, P. major, and their hybrids were determined by both real-time quantitative PCR and Western blot techniques. The results indicated the ubiquitous expression of these genes in all developmental stages of A. schlegelii and in all tested tissues of A. schlegelii, P. major, and its hybrids. Among them, the mRNA of TGF-ß1, TßR-I, and TßR-II genes is highly expressed in the liver, gill, kidney, and muscle of black porgy, red porgy, and their hybrid offspring. There are significant changes in gene and protein expression levels in hybrid offspring, which indirectly reflect hybrid advantage. In addition, there was no correlation between protein and mRNA expression levels of Smad2 protein. The results provide novel data for the differential expression of growth and development genes between the reciprocal hybridization generation of black porgy and red porgy and its parents, which is conducive to further explaining the molecular regulation mechanism of heterosis in the growth and development of hybrid porgy.


Subject(s)
Hybrid Vigor , Smad2 Protein , Transforming Growth Factor beta1 , Animals , Smad2 Protein/genetics , Smad2 Protein/metabolism , Hybrid Vigor/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Fish Proteins/genetics , Fish Proteins/metabolism , Hybridization, Genetic , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Perciformes/genetics , Perciformes/growth & development , Perciformes/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Female , Male , Gene Expression Regulation, Developmental
14.
Biomolecules ; 14(7)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39062514

ABSTRACT

Liver fibrosis, a consequence of chronic liver damage or inflammation, is characterized by the excessive buildup of extracellular matrix components. This progressive condition significantly raises the risk of severe liver diseases like cirrhosis and hepatocellular carcinoma. The lack of approved therapeutics underscores the urgent need for novel anti-fibrotic drugs. Hepatic stellate cells (HSCs), key players in fibrogenesis, are promising targets for drug discovery. This study investigated the anti-fibrotic potential of Citrus hystrix DC. (KL) and its bioactive compound, ß-citronellol (ß-CIT), in a human HSC cell line (LX-2). Cells exposed to TGF-ß1 to induce fibrogenesis were co-treated with crude KL extract and ß-CIT. Gene expression was analyzed by real-time qRT-PCR to assess fibrosis-associated genes (ACTA2, COL1A1, TIMP1, SMAD2). The release of matrix metalloproteinase 9 (MMP-9) was measured by ELISA. Proteomic analysis and molecular docking identified potential signaling proteins and modeled protein-ligand interactions. The results showed that both crude KL extract and ß-CIT suppressed HSC activation genes and MMP-9 levels. The MAPK signaling pathway emerged as a potential target of ß-CIT. This study demonstrates the ability of KL extract and ß-CIT to inhibit HSC activation during TGF-ß1-induced fibrogenesis, suggesting a promising role of ß-CIT in anti-hepatic fibrosis therapies.


Subject(s)
Acyclic Monoterpenes , Hepatic Stellate Cells , Liver Cirrhosis , Transforming Growth Factor beta1 , Humans , Actins , Antifibrotic Agents/pharmacology , Cell Line , Collagen Type I/metabolism , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Molecular Docking Simulation , Smad2 Protein/metabolism , Smad2 Protein/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Transforming Growth Factor beta1/pharmacology , Acyclic Monoterpenes/pharmacology
15.
J Cell Mol Med ; 28(13): e18527, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38984969

ABSTRACT

Infected bone defects (IBDs) are the common condition in the clinical practice of orthopaedics. Although surgery and anti-infective medicine are the firstly chosen treatments, in many cases, patients experience a prolonged bone union process after anti-infective treatment. Epimedium-Curculigo herb pair (ECP) has been proved to be effective for bone repair. However, the mechanisms of ECP in IBDs are insufficiency. In this study, Effect of ECP in IBDs was verified by micro-CT and histological examination. Qualitative and quantitative analysis of the main components in ECP containing medicated serum (ECP-CS) were performed. The network pharmacological approaches were then applied to predict potential pathways for ECP associated with bone repair. In addition, the mechanism of ECP regulating LncRNA MALAT1/miRNA-34a-5p/SMAD2 signalling axis was evaluated by molecular biology experiments. In vivo experiments indicated that ECP could significantly promote bone repair. The results of the chemical components analysis and the pathway identification revealed that TGF-ß signalling pathway was related to ECP. The results of in vitro experiments indicated that ECP-CS could reverse the damage caused by LPS through inhibiting the expressions of LncRNA MALAT1 and SMAD2, and improving the expressions of miR-34a-5p, ALP, RUNX2 and Collagen type І in osteoblasts significantly. This research showed that ECP could regulate the TGF-ß/SMADs signalling pathway to promote bone repair. Meanwhile, ECP could alleviate LPS-induced bone loss by modulating the signalling axis of LncRNA MALAT1/miRNA-34a-5p/ SMAD2 in IBDs.


Subject(s)
Epimedium , MicroRNAs , Osteoblasts , RNA, Long Noncoding , Signal Transduction , Smad2 Protein , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/metabolism , Osteoblasts/drug effects , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Smad2 Protein/metabolism , Smad2 Protein/genetics , Mice , Epimedium/chemistry , Signal Transduction/drug effects , Male , Bone Regeneration/drug effects , Humans , Gene Expression Regulation/drug effects , Osteogenesis/drug effects , Osteogenesis/genetics
16.
Mol Biol Rep ; 51(1): 728, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861185

ABSTRACT

INTRODUCTION: Colorectal cancer (CRC) is the second common cancer and the fourth major reason of cancer death worldwide. Dysregulation of intracellular pathways, such as TGF-ß/SMAD signaling, contributes to CRC development. MicroRNAs (miRNAs) are post-transcriptional regulators that are involved in CRC pathogenesis. Here, we aimed to investigate the effect of miR-3613-3p on the TGF-ß /SMAD signaling pathway in CRC. METHODS & RESULTS: Bioinformatics analysis suggested that miR-3613-3p is a regulator of TGF-Β signaling downstream genes. Then, miR-3613-3p overexpression was followed by downregulation of TGF-ßR1, TGF-ßR2, and SMAD2 expression levels, detected by RT-qPCR. Additionally, dual luciferase assay supported the direct interaction of miR-3613-3p with 3'UTR sequences of TGF-ßR1 and TGF-ßR2 genes. Furthermore, reduced SMAD3 protein level following the miR-3613-3p overexpression verified its suppressive effect against TGF-ß signaling in HCT-116 cells, detected by western blot analysis. Finally, miR-3613-3p overexpression induced sub-G1 arrest in HCT116 cells, detected by flow cytometry, and promoted downregulation of cyclin D1 protein expression, which was detected by western blotting analysis. CONCLUSION: Our findings indicated that miR-3613-3p plays an important role in CRC by targeting the TGF-ß/SMAD signaling pathway and could be considered as a new candidate for further therapy investigations.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , MicroRNAs , Signal Transduction , Transforming Growth Factor beta , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Smad2 Protein/metabolism , Smad2 Protein/genetics , Cell Proliferation/genetics , 3' Untranslated Regions/genetics , Cell Line, Tumor , Smad3 Protein/genetics , Smad3 Protein/metabolism
17.
Future Oncol ; 20(21): 1479-1493, 2024.
Article in English | MEDLINE | ID: mdl-38861304

ABSTRACT

Aim: The present study aimed to figure out the potential role of exosomal microRNAs, and their targeted genes in HNC detection/diagnosis.Methods: In the present study, exosomes were extracted from the serum samples of 400 HNC patients and 400 healthy controls. Exosomes were characterized using TEM, NTA, TEM-immunogold labeling and ELISA. Quantitative PCR was used to measure the expression level of exosomal miRNA-19a, miRNA-19b and targeted genes SMAD2 and SMAD4 in HNC patients and controls.Results: The deregulation of miR-19a (p < 0.01), miR-19b (p < 0.03), SMAD2 (p < 0.04) and SMAD4 (p < 0.04) was observed in HNC patients vs controls.Conclusion: ROC curve and Kaplan-Meier analysis showed the good diagnostic/prognostic value of selected exosomal microRNAs and related genes in HNC patients.


[Box: see text].


Subject(s)
Biomarkers, Tumor , Exosomes , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms , MicroRNAs , Humans , Exosomes/genetics , Exosomes/metabolism , MicroRNAs/genetics , MicroRNAs/blood , Female , Male , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/diagnosis , Middle Aged , Biomarkers, Tumor/genetics , Prognosis , Smad4 Protein/genetics , Adult , Case-Control Studies , Smad2 Protein/genetics , Aged , Kaplan-Meier Estimate , ROC Curve
18.
Beijing Da Xue Xue Bao Yi Xue Ban ; 56(3): 384-389, 2024 Jun 18.
Article in Chinese | MEDLINE | ID: mdl-38864121

ABSTRACT

OBJECTIVE: To explore the association between polymorphisms of transforming growth factor-ß (TGF-ß) signaling pathway and non-syndromic cleft lip with or without cleft palate (NSCL/P) among Asian populations, while considering gene-gene interaction and gene-environment interaction. METHODS: A total of 1 038 Asian NSCL/P case-parent trios were ascertained from an international consortium, which conducted a genome-wide association study using a case-parent trio design to investigate the genes affec-ting risk to NSCL/P. After stringent quality control measures, 343 single nucleotide polymorphism (SNP) spanning across 10 pivotal genes in the TGF-ß signaling pathway were selected from the original genome-wide association study(GWAS) dataset for further analysis. The transmission disequilibrium test (TDT) was used to test for SNP effects. The conditional Logistic regression models were used to test for gene-gene interaction and gene-environment interaction. Environmental factors collected for the study included smoking during pregnancy, passive smoking during pregnancy, alcohol intake during pregnancy, and vitamin use during pregnancy. Due to the low rates of exposure to smoking during pregnancy and alcohol consumption during pregnancy (<3%), only the interaction between maternal smoking during pregnancy and multivitamin supplementation during pregnancy was analyzed. The threshold for statistical significance was rigorously set at P =1.46×10-4, applying Bonferroni correction to account for multiple testing. RESULTS: A total of 23 SNPs in 4 genes yielded nominal association with NSCL/P (P<0.05), but none of these associations was statistically significant after Bonferroni' s multiple test correction. However, there were 6 pairs of SNPs rs4939874 (SMAD2) and rs1864615 (TGFBR2), rs2796813 (TGFB2) and rs2132298 (TGFBR2), rs4147358 (SMAD3) and rs1346907 (TGFBR2), rs4939874 (SMAD2) and rs1019855 (TGFBR2), rs4939874 (SMAD2) and rs12490466 (TGFBR2), rs2009112 (TGFB2) and rs4075748 (TGFBR2) showed statistically significant SNP-SNP interaction (P<1.46×10-4). In contrast, the analysis of gene-environment interactions did not yield any significant results after being corrected by multiple testing. CONCLUSION: The comprehensive evaluation of SNP associations and interactions within the TGF-ß signaling pathway did not yield any direct associations with NSCL/P risk in Asian populations. However, the significant gene-gene interactions identified suggest that the genetic architecture influencing NSCL/P risk may involve interactions between genes within the TGF-ß signaling pathway. These findings underscore the necessity for further investigations to unravel these results and further explore the underlying biological mechanisms.


Subject(s)
Cleft Lip , Cleft Palate , Gene-Environment Interaction , Genome-Wide Association Study , Transforming Growth Factor beta , Female , Humans , Male , Pregnancy , Alcohol Drinking/genetics , Asian People/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Epistasis, Genetic , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Risk Factors , Signal Transduction/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Tobacco Smoke Pollution/adverse effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
Open Biol ; 14(6): 230427, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862020

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a monogenic cardiac disorder commonly induced by sarcomere gene mutations. However, the mechanism for HCM is not well defined. Here, we generated transgenic MYH7 R453C and MYH6 R453C piglets and found both developed typical cardiac hypertrophy. Unexpectedly, we found serious fibrosis and cardiomyocyte loss in the ventricular of MYH7 R453C, not MYH6 R453C piglets, similar to HCM patients. Then, RNA-seq analysis and western blotting identified the activation of ERK1/2 and PI3K-Akt pathways in MYH7 R453C. Moreover, we observed an increased expression of fetal genes and an excess of reactive oxygen species (ROS) in MYH7 R453C piglet models, which was produced by Nox4 and subsequently induced inflammatory response. Additionally, the phosphorylation levels of Smad2/3, ERK1/2 and NF-kB p65 proteins were elevated in cardiomyocytes with the MYH7 R453C mutation. Furthermore, epigallocatechin gallate, a natural bioactive compound, could be used as a drug to reduce cell death by adjusting significant downregulation of the protein expression of Bax and upregulated Bcl-2 levels in the H9C2 models with MYH7 R453C mutation. In conclusion, our study illustrated that TGF-ß/Smad2/3, ERK1/2 and Nox4/ROS pathways have synergistic effects on cardiac remodelling and inflammation in MYH7 R453C mutation.


Subject(s)
Myosin Heavy Chains , NADPH Oxidase 4 , NF-kappa B , Reactive Oxygen Species , Signal Transduction , Transforming Growth Factor beta , Animals , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Transforming Growth Factor beta/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Swine , Myocytes, Cardiac/metabolism , Humans , Cardiac Myosins/metabolism , Cardiac Myosins/genetics , Disease Models, Animal , MAP Kinase Signaling System , Animals, Genetically Modified , Smad2 Protein/metabolism , Smad2 Protein/genetics , Mutation , Smad3 Protein/metabolism , Smad3 Protein/genetics , Ventricular Remodeling , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Rats
20.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892032

ABSTRACT

Keloids, marked by abnormal cellular proliferation and excessive extracellular matrix (ECM) accumulation, pose significant therapeutic challenges. Ethyl pyruvate (EP), an inhibitor of the high-mobility group box 1 (HMGB1) and TGF-ß1 pathways, has emerged as a potential anti-fibrotic agent. Our research evaluated EP's effects on keloid fibroblast (KF) proliferation and ECM production, employing both in vitro cell cultures and ex vivo patient-derived keloid spheroids. We also analyzed the expression levels of ECM components in keloid tissue spheroids treated with EP through immunohistochemistry. Findings revealed that EP treatment impedes the nuclear translocation of HMGB1 and diminishes KF proliferation. Additionally, EP significantly lowered mRNA and protein levels of collagen I and III by attenuating TGF-ß1 and pSmad2/3 complex expression in both human dermal fibroblasts and KFs. Moreover, metalloproteinase I (MMP-1) and MMP-3 mRNA levels saw a notable increase following EP administration. In keloid spheroids, EP induced a dose-dependent reduction in ECM component expression. Immunohistochemical and western blot analyses confirmed significant declines in collagen I, collagen III, fibronectin, elastin, TGF-ß, AKT, and ERK 1/2 expression levels. These outcomes underscore EP's antifibrotic potential, suggesting its viability as a therapeutic approach for keloids.


Subject(s)
Fibroblasts , Keloid , Pyruvates , Spheroids, Cellular , Humans , Keloid/metabolism , Keloid/pathology , Fibroblasts/metabolism , Fibroblasts/drug effects , Pyruvates/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 1/genetics , Transforming Growth Factor beta1/metabolism , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , Collagen/metabolism , Collagen/biosynthesis , Cell Proliferation/drug effects , Cells, Cultured , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 3/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Collagen Type I/metabolism , Collagen Type I/genetics , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Up-Regulation/drug effects , Male
SELECTION OF CITATIONS
SEARCH DETAIL