Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Ethnopharmacol ; 324: 117737, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38228229

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Leech, as a traditional Chinese medicine for the treatment of blood circulation and blood stasis, was also widely used to cure pulmonary fibrosis in China. In clinical practice, some traditional Chinese medicine preparation such as Shui Zhi Xuan Bi Hua Xian Tang and Shui Zhi Tong Luo Capsule composed of leech, could improve the clinical symptoms and pulmonary function in patients with idiopathic pulmonary fibrosis (IPF). However, the material basis of the leech in the treatment of IPF were not yet clear. AIM OF THE STUDY: Screen out the components of leech that have the anti-pulmonary fibrosis effects, and further explore the therapeutic mechanism of the active components. MATERIALS AND METHODS: In this study, the different molecular weight components of leech extract samples were prepared using the semi-permeable membranes with different pore sizes. The therapeutic effects of the leech extract groups with molecular weight greater than 10 KDa (>10 KDa group), between 3 KDa and 10 KDa (3-10 KDa group), and less than 3 KDa (<3 KDa group) on pulmonary fibrosis were firstly investigated by cell proliferation and cytotoxicity assay (MTT), cell wound healing assay, immunofluorescence staining (IF) and Western blot (WB) assay through the TGF-ß1-induced fibroblast cell model. Then bleomycin-induced pulmonary fibrosis (BML-induced PF) mouse model was constructed to investigate the pharmacological activities of the active component group of leech extract in vivo. Pathological changes of the mouse lung were observed by hematoxylin-eosin staining (H&E) and Masson's trichrome staining (Masson). The hydroxyproline (HYP) content of lung tissues was quantified by HYP detection kit. The levels of extracellular matrix-related fibronectin (FN) and collagen type Ⅰ (Collagen Ⅰ), pyruvate kinase M2 (PKM2) monomer and Smad7 protein were determined via WB method. PKM2 and Smad7 protein were further characterized by IF assays. RESULTS: Using TGF-ß1-induced HFL1 cell line as a PF cell model, the in vitro results demonstrated that the >10 KDa group could significantly inhibited the cell proliferation and migration, downregulated the expression level of cytoskeletal protein vimentin and α-smooth muscle actin (α-SMA), and reduced the deposition of FN and Collagen Ⅰ. In the BML-induced PF mouse model, the >10 KDa group significantly reduced the content of HYP, downregulated the expression levels of FN and Collagen Ⅰ in lung tissues, and delayed the pathological changes of lung tissue structure. The results of WB and IF assays further indicated that the >10 KDa group could up-regulate the expression level of PKM2 monomer and Smad7 protein in the cellular level, thereby delaying the progression of pulmonary fibrosis. CONCLUSIONS: Our study revealed that the >10 KDa group was the main material basis of the leech extract that inhibited pulmonary fibrosis through TGF-ß1/Smad3 signaling pathway.


Subject(s)
Idiopathic Pulmonary Fibrosis , Transforming Growth Factor beta1 , Mice , Animals , Humans , Transforming Growth Factor beta1/metabolism , Smad7 Protein/metabolism , Smad7 Protein/pharmacology , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Collagen Type I/metabolism , Bleomycin , Disease Models, Animal , Signal Transduction
2.
Immunobiology ; 228(2): 152315, 2023 03.
Article in English | MEDLINE | ID: mdl-36608595

ABSTRACT

The development of liver fibrosis is associated with inflammatory responses resulting from chronic liver disease. Immature dendritic cells (imDCs) play an important role in modulating the inflammatory environment of the liver. This study investigated the effects of imDCs on the regulation of hepatic stellate cells (HSCs) during liver fibrosis. We isolated and induced imDCs from monocytes of healthy volunteers, activated LX-2 cells with TGF-ß to establish in vivo liver fibrosis HSCs model, and then set up a cell co-culture system with transwell membranes. imDC surface markers and apoptosis rates of LX-2 cells were detected by flow cytometry. The concentration of IL-10 secreted by imDC was measured through ELISA. The expression of α-SMA in LX-2 after co-culture was examined by qRT­PCR. Proliferation of LX-2 cells were detected by CCK-8. The western blot was used to illustrate the LX-2 activation-related proteins such as Smad3/7 and TGF-ß1. The imDCs co-culture group and the interleukin-10 (IL-10) treatment group had similar results, as they were both able to increase apoptosis, inhibit proliferation, downregulate α-SMA mRNA, and reduce TGF-ß1 and Smad3 protein expression in LX-2 cells. Additionally, the Smad7 protein level was increased after treatment with imDC and IL-10. However, the results in the IL-10 antagonist group showed the opposite trend to that of imDCs and IL-10 groups. Thus, these results suggest that imDC secretion of IL-10 negatively regulates activated LX-2 cells, probably via inhibition of the TGF-ß1/Smad3 pathway and increased expression of Smad7 protein. This may be a potential therapeutic target for liver fibrosis.


Subject(s)
Hepatic Stellate Cells , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Interleukin-10/metabolism , Smad7 Protein/metabolism , Smad7 Protein/pharmacology , Smad7 Protein/therapeutic use , Monocytes/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I/pharmacology , Liver Cirrhosis , Dendritic Cells/metabolism
3.
Nucleic Acids Res ; 50(18): 10526-10543, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36134711

ABSTRACT

Transforming growth factor ß (TGF-ß) superfamily proteins are potent regulators of cellular development and differentiation. Nodal/Activin/TGF-ß and BMP ligands are both present in the intra- and extracellular milieu during early development, and cross-talk between these two branches of developmental signaling is currently the subject of intense research focus. Here, we show that the Nodal induced lncRNA-Smad7 regulates cell fate determination via repression of BMP signaling in mouse embryonic stem cells (mESCs). Depletion of lncRNA-Smad7 dramatically impairs cardiomyocyte differentiation in mESCs. Moreover, lncRNA-Smad7 represses Bmp2 expression through binding with the Bmp2 promoter region via (CA)12-repeats that forms an R-loop. Importantly, Bmp2 knockdown rescues defects in cardiomyocyte differentiation induced by lncRNA-Smad7 knockdown. Hence, lncRNA-Smad7 antagonizes BMP signaling in mESCs, and similarly regulates cell fate determination between osteocyte and myocyte formation in C2C12 mouse myoblasts. Moreover, lncRNA-Smad7 associates with hnRNPK in mESCs and hnRNPK binds at the Bmp2 promoter, potentially contributing to Bmp2 expression repression. The antagonistic effects between Nodal/TGF-ß and BMP signaling via lncRNA-Smad7 described in this work provides a framework for understanding cell fate determination in early development.


Subject(s)
RNA, Long Noncoding , Smad7 Protein/metabolism , Activins/metabolism , Activins/pharmacology , Animals , Cell Differentiation , Ligands , Mice , RNA, Long Noncoding/metabolism , Smad7 Protein/genetics , Smad7 Protein/pharmacology , Transforming Growth Factor beta/metabolism
4.
Mol Biol Rep ; 43(4): 241-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26902861

ABSTRACT

SMAD7 promotes and enhances skeletal muscle differentiation by inhibiting transforming growth factor beta (TGF-ß)/activin signaling and bone morphogenetic protein (BMP) pathways. However, its function, the mechanism regulating its translation, and its association with production meat traits remain unclear in pigs. In this study, we explored SMAD7 gene spatio-temporal and tissue distribution, conducted a single nucleotide polymorphism association analysis, and examined regulation of its expression during skeletal muscle development. We found that SMAD7 was positively related to TGF-ß pathway genes and mainly expressed in prenatal developing muscle, and dual luciferase and western blot assays demonstrated that SMAD7 expression was regulated by miRNA-21 at the protein level via inhibition of mRNA translation. Finally, the association analysis showed that a single nucleotide mutation (Exon 4_28816;C/A) was significantly associated with the weaning weight of piglets among Yorkshire pigs. These data indicate that SMAD7 plays a potentially important role in mammalian prenatal skeletal muscle development and is a candidate gene for promoting greater weaning weight in pig breeding.


Subject(s)
Body Weight/physiology , Muscle, Skeletal/embryology , Signal Transduction , Smad7 Protein/metabolism , Sus scrofa/embryology , Transforming Growth Factor beta/antagonists & inhibitors , Amino Acid Sequence , Animals , Gene Expression Regulation, Developmental , Molecular Sequence Data , Muscle Development , Muscle, Skeletal/metabolism , Organ Specificity , Phylogeny , Polymorphism, Single Nucleotide , Sequence Alignment , Smad7 Protein/genetics , Smad7 Protein/pharmacology , Sus scrofa/metabolism , Weaning
5.
Endocr J ; 59(8): 653-62, 2012.
Article in English | MEDLINE | ID: mdl-22673292

ABSTRACT

The transforming growth factor (TGF)-ß family members, bone morphogenetic protein (BMP)-2 and TGF-ß that signal via the receptor-regulated Smads (R-Smads) induce bone formation in vivo. The inhibitory Smads (I-Smads), Smad6 and Smad7, negatively regulate TGF-ß family ligand signaling by competing with R-Smads for binding to activated type I receptors, and preventing R-Smad activation, Hence, the I-Smads potentially act as suppressors of bone formation although their effects on phenotypic changes in mature osteoblasts are unclear. While Smad7 inhibits both BMP and TGF-ß signaling, Smad6 is less effective in inhibiting TGF-ß signaling. The present study was performed to examine the role of Smad7 on the phenotype of mouse osteoblastic MC3T3-E1 cells. We employed stable Smad7-transfected MC3T3-E1 cells to examine the role of Smad7 in osteoblast proliferation, differentiation and mineralization. Stable Smad7 overexpression significantly inhibited the absorbance in the MTT-dye assay and inhibited the levels of PCNA compared with those in empty vector-transfected cells. Smad7 overexpression suppressed the type 1 collagen mRNA and protein levels. Moreover, Smad7 inhibited ALP activity and mineralization of osteoblastic cells. The effects of stable overexpression of Smad6 were similar to those of Smad7 suggesting the changes mediated by either I-Smad occurred by inhibition of BMP rather than TGF-ß signaling. In addition, PTH-(1-34) elevated the levels of Smad7 in parental MC3T3-E1 cells. In conclusion, the present study demonstrated that Smad7, as well as Smad6, inhibits proliferation, differentiation and mineralization of mouse osteoblastic cells. Therefore, I-Smads are important molecular targets for the negative control of bone formation.


Subject(s)
Cell Differentiation/drug effects , Osteoblasts/drug effects , Smad7 Protein/pharmacology , Alkaline Phosphatase/metabolism , Animals , Collagen Type I/biosynthesis , Mice , Parathyroid Hormone/pharmacology , RNA, Small Interfering/pharmacology , Smad6 Protein/metabolism , Smad6 Protein/pharmacology , Smad7 Protein/metabolism
6.
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
7.
Chin Med J (Engl) ; 124(13): 1988-93, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-22088458

ABSTRACT

BACKGROUND: Transforming growth factor ß (TGFß) is one of the most important growth factors in the development of fibrosis and scarring on cornea. Smad7, an inhibitory Smad, can inhibit TGFß signal transduction. In recent years, effects of lentiviral-mediated Smad7 on inhibition of fibrosis on some organs have been studied, while little is known about the effects on cornea. This study aimed to determine the effects of lentiviral-mediated Smad7 gene expression on keratocyte proliferation and fibrosis induced by TGF ß2 in vitro. METHODS: Keratocytes were cultured from corneal tissue isolated from Sprague-Dawley (SD) rats and transfected with Smad7 expressing lentiviral vector (Lv-Smad7) or non-functioning control vector (Lv-blank). Following the exposure to TGFß2, keratocytes were processed for immunoblotting to assess the phosphorylation of Smad2 as down-stream event of TGFß/Smad signaling. Expression of fibrotic markers α-smooth muscle actin (α-SMA), type III collagen (collagen III) were measured by Western blotting and quantitative real time polymerase chain reaction (PCR). Overall cell proliferation was determined by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the expression of cell cycle-related marker Ki67 at both mRNA and protein levels. RESULTS: The Smad7 gene transfer suppressed TGFß/Smad signaling in keratocytes by down-regulating phosphorylation of Smad2. Markers of cell proliferation and fibrosis including Ki67, α-SMA, collagen III were inhibited by introduction of Smad 7 into TGFß exposed keratocytes. Consequently, the rate of cell proliferation was attenuated. CONCLUSION: Smad7 gene transfer inhibited fibrogenic responses of keratocytes to TGFß2.


Subject(s)
Corneal Keratocytes/cytology , Corneal Keratocytes/metabolism , Smad7 Protein/metabolism , Transforming Growth Factor beta2/pharmacology , Actins/genetics , Actins/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Collagen Type III/genetics , Collagen Type III/metabolism , Corneal Keratocytes/drug effects , Genetic Vectors/genetics , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Lentivirus/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Smad7 Protein/genetics , Smad7 Protein/pharmacology
8.
PLoS One ; 5(4): e10149, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20405019

ABSTRACT

BACKGROUND: TGF-beta has been postulated to play an important role in the maintenance of epithelial homeostasis and the development of epithelium-derived cancers. However, most of previous studies are mainly focused on the function of TGF-beta in immune cells to the development of allergic asthma and how TGF-beta signaling in airway epithelium itself in allergic inflammation is largely unknown. Furthermore, the in vivo TGF-beta function specifically in the airway epithelium during lung cancer development has been largely elusive. METHODOLOGY/PRINCIPAL FINDINGS: To evaluate the in vivo contribution of TGF-beta signaling in lung epithelium to the development of allergic disease and lung cancer, we generated a transgenic mouse model with Smad7, an intracellular inhibitor of TGF-beta signaling, constitutively expressed in mouse airway Clara cells using a mouse CC10 promoter. The mice were subjected to the development of OVA-induced allergic asthma and urethane-induced lung cancer. The Smad7 transgenic animals significantly protected from OVA-induced asthma, with reduced airway inflammation, airway mucus production, extracellular matrix deposition, and production of OVA-specific IgE. Further analysis of cytokine profiles in lung homogenates revealed that the Th2 cytokines including IL-4, IL-5 and IL-13, as well as other cytokines including IL-17, IL-1, IL-6, IP10, G-CSF, and GM-CSF were significantly reduced in the transgenic mice upon OVA induction. In contrast, the Smad7 transgenic animals had an increased incidence of lung carcinogenesis when subjected to urethane treatment. CONCLUSION/SIGNIFICANCE: These studies, therefore, demonstrate for the first time the in vivo function of TGF-beta signaling specifically in airway epithelium during the development of allergic asthma and lung cancer.


Subject(s)
Asthma/etiology , Lung Neoplasms/etiology , Respiratory Mucosa/metabolism , Signal Transduction , Smad7 Protein/pharmacology , Transforming Growth Factor beta/physiology , Animals , Asthma/chemically induced , Asthma/therapy , Cytokines/analysis , Disease Models, Animal , Genetic Therapy , Inflammation/prevention & control , Lung Neoplasms/chemically induced , Lung Neoplasms/therapy , Mice , Mice, Transgenic , Ovalbumin , Signal Transduction/drug effects , Smad7 Protein/genetics , Th2 Cells , Transforming Growth Factor beta/antagonists & inhibitors , Urethane
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 29(10): 2122-3, 2127, 2009 Oct.
Article in Chinese | MEDLINE | ID: mdl-19861282

ABSTRACT

OBJECTIVE: To investigate the effect of Smad7 on the expressions of collagen I and alpha-smooth muscle actin (alpha-SMA) in HSC-T6 cell line activated by transforming growth factor-beta1 (TGF-beta1). METHODS: HSC-T6 cells stably expressing M2-flag protein were selected after co-infection of the cells with pTRE-Smad7-M2-flag and pTet-on. The optimal dose of doxycycline for inducing Smad7 was determined, and the effects of Smad7 over-expression on the expressions of collagen I and alpha-SMA in the cells activated by TGF-beta1 and on Smad2/3 phosphorylation were evaluated using Western blotting. RESULTS: The optimal dose of doxycycline for inducing Smad7 expression was 2 mg/L. Smad7 over-expression induced by doxycycline decreased the expressions of collagen I and alpha-SMA in HSC-T6 cells activated by TGF-beta1, and down-regulated the level of Smad2/3 phosphorylation. CONCLUSION: Smad7 over-expression inhibits Smad2/3 phosphorylation, and decreases the expression of collagen I and alpha-SMA in HSC-T6 cells induced by TGF-beta1 to inhibit the progression of liver fibrosis.


Subject(s)
Actins/metabolism , Collagen Type I/metabolism , Hepatocytes/metabolism , Smad7 Protein/pharmacology , Actins/genetics , Cells, Cultured , Collagen Type I/genetics , Genetic Therapy , Hepatocytes/cytology , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/therapy , Transforming Growth Factor beta1/pharmacology
10.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 24(11): 1074-8, 2008 Nov.
Article in Chinese | MEDLINE | ID: mdl-18992195

ABSTRACT

AIM: To investigate whether Smad6 and Smad7 can regulate TGF-beta-induced epithelial-mesenchymal transition in human renal proximal tubule epithelial cells. METHODS: Two recombinant adeno-associated viruses (AAV) expressing Smad6 and Smad7 genes were produced without helper virus and then they were delivered into human renal proximal tubule epithelial cells (HKCs). The cells were randomly divided into normal controls, TGF-beta1-treated group, Smad7-infected control, LacZ-infected control, TGF-beta1+Smad7 group or TGF-beta1+Smad6 group, and TGF-beta1 + LacZ group. 10 microg/L of TGF-beta1 was added into the cell culture at the time of 15 min, 30 min, 60 min, and 120 min and the third day. The levels of phospho-Smad2, alpha-smooth muscle actin (alpha-SMA) and E-cadherin proteins were measured by Western blot. The concentration of hydroxyproline excreted into the culture supernatant was determinated by ELISA. The morphological changes of the cells detected by inverted microscope. RESULTS: Compared with the cells in absence of TGF-beta1, the expression level of P-Smad2 in HKCs increased at 15 min, 30 min, 60 min, and 120 min with the TGF-beta1 stimulation. It reached peak at 30 min. TGF-beta1 treatment for 72 hours resulted in significant up-regulation of alpha-SMA protein expression and hydroxyproline secretion, but a marked decrease in E-cadherin expression in the culture of HKCs. 10 microg/L of TGF-beta1 treatment for 72 hours also resulted in marked alteration in cell morphology. The cells lost their regular cuboidal appearance, and become elongated and spindle shaped. In the Smad7-infected cells, the overexpression of Smad7 resulted in a marked decrease of alpha-SMA protein and hydroxyproline synthesis, but a increase of E-cadherin protein, as well as the retainment of epithelial phenotypic appearance. These effects were associated with the inhibition of TGF-induced Smad2 activation, whereas the overexpression of Smad6 had no effect on the TGF-beta-induced changes in HKCs. CONCLUSION: The overexpression of Smad7 instead of Smad6 can efficiently inhibit TGF-beta-induced epithelial-mesenchymal transition by blocking Smad2 activation in human renal proximal tubule epithelial cells.


Subject(s)
Cell Differentiation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Mesoderm/drug effects , Mesoderm/pathology , Smad7 Protein/pharmacology , Transforming Growth Factor beta/pharmacology , Actins/metabolism , Blotting, Western , Cadherins/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Humans , Hydroxyproline/metabolism , Kidney Tubules, Proximal/cytology , Mesoderm/metabolism , Smad2 Protein/metabolism , Smad6 Protein/pharmacology
11.
Endocr J ; 54(2): 311-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17384470

ABSTRACT

The present study was conducted to elucidate the role of activin A in capillary formation. When bovine aortic endothelial cells (BAEC) were cultured in a collagen gel, basic fibroblast growth factor (FGF-2) induced tube formation. Activin A also induced tube formation and the addition of two factors together was more effective. BAEC produced both FGF-2 and activin A as autocrine factors. Exogenous FGF-2 did not affect the production of activin A but instead upregulated the type II activin receptor. On the other hand, activin A increased the expression of FGF-2 as well as the FGF receptor. Most importantly, when the action of endogenous activin A was blocked by adding follistatin, the tubulogenic action of FGF-2 was nearly completely inhibited. Activin-induced tubulogenesis was markedly inhibited by overexpression of Smad7, an inhibitory Smad. Similarly, an inhibitor of p44/42 mitogen-activated protein (MAP) kinase attenuated the activin-mediated tubulogenesis, whereas an inhibitor of p38 MAP kinase had no effect. These results indicate that FGF-2 and activin A enhance their signals each other in BAEC, and endogenous activin A is critical for FGF-2-induced capillary formation.


Subject(s)
Activins/physiology , Capillaries/physiology , Fibroblast Growth Factor 2/physiology , Neovascularization, Physiologic/physiology , Activin Receptors, Type II/metabolism , Activins/metabolism , Activins/pharmacology , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Cattle , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/metabolism , Flavonoids/pharmacology , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Recombinant Proteins/pharmacology , Smad7 Protein/genetics , Smad7 Protein/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...