Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 321-329, 2018.
Article in English | WPRIM | ID: wpr-812399

ABSTRACT

The present study was designed to elucidate whether the mechanism by which osthole decreases collagenI/III contents and their ratio is regulating the TGF-β/Smad signaling pathway in TGF-β1-overexpressed mouse cardiac fibroblasts (CFs). These CFs were cultured and treated with different concentrations of osthole. Our results showed that the TGF-β1 expression in the CFs transfected with that the recombinant expression plasmids pcDNA3.1(+)-TGF-β1 was significantly enhanced. After the CFs were treated with 1.25-5 μg·mL of osthole for 24 h, the mRNA and protein expression levels of collagensIand III were reduced. The collagen I/III ratio was also reduced. The mRNA and protein expression levels of TGF-β1, TβRI, Smad2/3, P-Smad2/3, Smad4, and α-SMA were decreased, whereas the expression level of Smad7 was increased. These effects suggested that osthole could inhibit collagen I and III expression and reduce their ratio via the TGF-β/Smad signaling pathway in TGF-β1 overexpressed CFs. These effects of osthole may play beneficial roles in the prevention and treatment of myocardial fibrosis.


Subject(s)
Animals , Mice , Actins , Genetics , Cells, Cultured , Collagen , Genetics , Coumarins , Pharmacology , Fibroblasts , Metabolism , Gene Expression Regulation , Myocardium , Cell Biology , Protein Serine-Threonine Kinases , Genetics , RNA, Messenger , Genetics , Real-Time Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta , Genetics , Signal Transduction , Smad Proteins , Genetics , Transforming Growth Factor beta1 , Genetics
2.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 321-329, 2018.
Article in English | WPRIM | ID: wpr-773610

ABSTRACT

The present study was designed to elucidate whether the mechanism by which osthole decreases collagenI/III contents and their ratio is regulating the TGF-β/Smad signaling pathway in TGF-β1-overexpressed mouse cardiac fibroblasts (CFs). These CFs were cultured and treated with different concentrations of osthole. Our results showed that the TGF-β1 expression in the CFs transfected with that the recombinant expression plasmids pcDNA3.1(+)-TGF-β1 was significantly enhanced. After the CFs were treated with 1.25-5 μg·mL of osthole for 24 h, the mRNA and protein expression levels of collagensIand III were reduced. The collagen I/III ratio was also reduced. The mRNA and protein expression levels of TGF-β1, TβRI, Smad2/3, P-Smad2/3, Smad4, and α-SMA were decreased, whereas the expression level of Smad7 was increased. These effects suggested that osthole could inhibit collagen I and III expression and reduce their ratio via the TGF-β/Smad signaling pathway in TGF-β1 overexpressed CFs. These effects of osthole may play beneficial roles in the prevention and treatment of myocardial fibrosis.


Subject(s)
Animals , Mice , Actins , Genetics , Cells, Cultured , Collagen , Genetics , Coumarins , Pharmacology , Fibroblasts , Metabolism , Gene Expression Regulation , Myocardium , Cell Biology , Protein Serine-Threonine Kinases , Genetics , RNA, Messenger , Genetics , Real-Time Polymerase Chain Reaction , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta , Genetics , Signal Transduction , Smad Proteins , Genetics , Transforming Growth Factor beta1 , Genetics
3.
An. bras. dermatol ; 92(2): 184-190, Mar.-Apr. 2017. tab, graf
Article in English | LILACS | ID: biblio-838060

ABSTRACT

Abstract: Background: A single, effective therapeutic regimen for keloids has not been established yet, and the development of novel therapeutic approaches is expected. Butyrate, a short-chain fatty acid, and docosahexaenoic acid (DHA), a ω-3 polyunsaturated fatty acid, play multiple anti-inflammatory and anticancer roles via their respective mechanisms of action. Objective: In this study, we evaluated the antifibrogenic effects of their single and combined use on keloid fibroblasts. Methods: Keloid fibroblasts were treated with butyrate (0-16 mM) and/or DHA (0-100 µM) for 48 or 96 h. Results: Butyrate inhibited cell proliferation, and α-smooth muscle actin (α-SMA) and type III collagen expressions, with inhibition of the transforming growth factor (TGF)-β1 and TGF-β type I receptor expressions and increased prostaglandin E2 with upregulation of cyclooxygenase-1 expression with induction of histone acetylation. DHA inhibited α-SMA, type III collagen, and TGF-β type I receptor expressions. Then, the butyrate/DHA combination augmented the antifibrogenic effects, resulting in additional inhibition of α-SMA, type I and III collagen expressions, with strong disruption of stress fiber and apoptosis induction. Moreover, the butyrate/DHA combination inhibited the cyclooxygenase-2 expression, suggesting stronger anti-inflammatory effect than each monotherapy. Study limitations: Activation in keloid tissue is affected not only by fibroblasts but also by epithelial cells and immune cells. Evaluation of the effects by butyrate and DHA in these cells or in an in vivo study is required. Conclusion: This study demonstrated that butyrate and docosahexaenoic acid have antifibrogenic effects on keloid fibroblasts and that these may exert therapeutic effects for keloid.


Subject(s)
Humans , Butyrates/therapeutic use , Docosahexaenoic Acids/therapeutic use , Fibroblasts , Keloid/drug therapy , Cells, Cultured , Protein Serine-Threonine Kinases , Receptors, Transforming Growth Factor beta , Combined Modality Therapy , Collagen Type I , Collagen Type III , Cell Proliferation
4.
Protein & Cell ; (12): 39-54, 2017.
Article in English | WPRIM | ID: wpr-757379

ABSTRACT

Human telomerase reverse transcriptase (hTERT) plays a central role in telomere lengthening for continuous cell proliferation, but it remains unclear how extracellular cues regulate telomerase lengthening of telomeres. Here we report that the cytokine bone morphogenetic protein-7 (BMP7) induces the hTERT gene repression in a BMPRII receptor- and Smad3-dependent manner in human breast cancer cells. Chonic exposure of human breast cancer cells to BMP7 results in short telomeres, cell senescence and apoptosis. Mutation of the BMPRII receptor, but not TGFbRII, ACTRIIA or ACTRIIB receptor, inhibits BMP7-induced repression of the hTERT gene promoter activity, leading to increased telomerase activity, lengthened telomeres and continued cell proliferation. Expression of hTERT prevents BMP7-induced breast cancer cell senescence and apoptosis. Thus, our data suggest that BMP7 induces breast cancer cell aging by a mechanism involving BMPRII receptor- and Smad3-mediated repression of the hTERT gene.


Subject(s)
Female , Humans , Actin-Related Protein 2 , Genetics , Metabolism , Activin Receptors, Type II , Genetics , Metabolism , Bone Morphogenetic Protein 7 , Genetics , Metabolism , Bone Morphogenetic Protein Receptors, Type II , Genetics , Metabolism , Breast Neoplasms , Genetics , Metabolism , Cellular Senescence , HeLa Cells , MCF-7 Cells , Neoplasm Proteins , Genetics , Metabolism , Protein Serine-Threonine Kinases , Genetics , Metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta , Genetics , Metabolism , Smad3 Protein , Genetics , Metabolism , Telomerase , Genetics , Metabolism , Telomere Homeostasis
5.
Yonsei Medical Journal ; : 33-40, 2016.
Article in English | WPRIM | ID: wpr-199916

ABSTRACT

PURPOSE: This study aimed to investigate whether Mullerian inhibiting substance (MIS) in combination with calcitriol modulates proliferation and apoptosis of human ovarian cancer (OCa) cell lines (SKOV3, OVCAR3, and OVCA433) and identify the signaling pathway by which MIS mediates apoptosis. MATERIALS AND METHODS: OCa cell lines were treated with MIS in the absence or presence of calcitriol. Cell viability and proliferation were evaluated using the Cell Counting Kit-8 assay and apoptosis was evaluated by DNA fragmentation assay. Western blot and enzyme-linked immunosorbent assay were used to determine the signaling pathway. RESULTS: The cells showed specific staining for the MIS type II receptor. Treatment of OCa cells with MIS and calcitriol led to dose- and time-dependent inhibition of cell growth and survival. The combination treatment significantly suppressed cell growth, down-regulated the expression of B-cell lymphoma 2 (Bcl-2), and up-regulated the expressions of Bcl-2 associated X protein, caspase-3, and caspase-9 through the extracellular signal-regulated kinase signaling pathway. CONCLUSION: These results, coupled with a much-needed decrease in the toxic side effects of currently employed therapeutic agents, provide a strong rationale for testing the therapeutic potential of MIS, alone or in combination with calcitriol, in the treatment of OCa.


Subject(s)
Female , Humans , Anti-Mullerian Hormone/pharmacology , Apoptosis/drug effects , Calcitriol/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Fragmentation/drug effects , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/metabolism , Growth Inhibitors/metabolism , Ovarian Neoplasms/drug therapy , Receptors, Peptide , Receptors, Transforming Growth Factor beta , Signal Transduction/drug effects
6.
Journal of Southern Medical University ; (12): 1245-1250, 2015.
Article in Chinese | WPRIM | ID: wpr-333647

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of nuclear factor I-C (NFI-C) on platelet-derived growth factor (PDGF)-induced up-regulation of TGF-β receptor II (TβRII) in dermal fibroblasts.</p><p><b>METHODS</b>A lentiviral vector containing NFI-C sequence (Lenti-GFP-NFI-C) was transfected into a human foreskin fibroblast cell line (HFF-1). Cultured HFF-1 cells, cells transfected with Lenti-GFP-NFI-C, and cells transfected with a negative virus were stimulated with PDGF-BB, and Western blotting and RT-qPCR were used to detect the expression levels of TβRII in the treated cells.</p><p><b>RESULTS</b>PDGF treatment significantly increased the expression level of TβRII in HFF-1 cells (P<0.05). The cells transfected with Lenti-GFP-NFI-C expressed a significantly lower level of TβRII than non-transfected cells in response to PDGF stimulation (P<0.05), but the negative virus showed no such inhibitory effect (P>0.05). No significant difference was found in the expression level of TβRII protein between cells transfected with Lenti-GFP-NFI-C-transfection before PDGF stimulation and the blank control cells.</p><p><b>CONCLUSION</b>NFI-C can inhibit PDGF-induced up-regulation of TβRII and thus reduce the sensitivity of the dermal fibroblasts to TGF-β.</p>


Subject(s)
Humans , Cell Line , Fibroblasts , Genetic Vectors , Lentivirus , NFI Transcription Factors , Genetics , Platelet-Derived Growth Factor , Pharmacology , Protein Serine-Threonine Kinases , Metabolism , Proto-Oncogene Proteins c-sis , Receptors, Transforming Growth Factor beta , Metabolism , Transfection , Transforming Growth Factor beta , Pharmacology , Up-Regulation
7.
Chinese Journal of Burns ; (6): 372-377, 2015.
Article in Chinese | WPRIM | ID: wpr-327394

ABSTRACT

<p><b>OBJECTIVE</b>To explore the effects of blocking two sites of TGF-β/Smads signaling on the formation of scar-related proteins in human skin fibroblasts.</p><p><b>METHODS</b>Two lentivirus vectors encoding soluble TGF-β receptor II (sTβRII) and mutant Smad 4-Smad 4ΔM4 were respectively transfected into human skin fibroblast cell line human foreskin fibroblast 1 (HFF-1) cells with the optimum multiplicity of infection (MOI) of 50. The protein expressions of sTβRII and Smad 4ΔM4 of the two types of transfected cells were determined by Western blotting so as to compare with those of the untransfected cells. The HFF-1 cells were divided into 6 groups as named below according to the random number table, with 6 dishes in each group, 1×10(4) cells per dish. Co-transfection group, transfected with the two previous lentivirus vectors, mixed with the ratio of 1:1 and MOI of 50, and then stimulated with 5 ng/mL TGF-β1 for 72 h; sTβRII group, transfected with lenti-sTβRII with MOI of 50, with the other treatment as above; Smad 4ΔM4 group, transfected with lenti-Smad 4ΔM4 with MOI of 50, with the other treatment as above; negative virus group, transfected with empty lentivirus vector, with the other treatment as above; positive control group, stimulated with 5 ng/mL TGF-β1 for 72 h; and blank control group, conventionally cultured without any other treatment. After stimulation, Western blotting and real-time fluorescent quantitative RT-PCR were respectively used to determine the protein and mRNA expressions of fibronectin in cells of each group. ELISA and Sircol collagen assay were respectively used to determine the protein expressions of connective tissue growth factor (CTGF) and total collagen in the cell culture supernate of each group. Data were processed with one-way analysis of variance and SNK-(q test).</p><p><b>RESULTS</b>(1) HFF-1 cells transfected with lenti-sTβRII and HFF-1 cells transfected with lenti-Smad 4ΔM4 respectively expressed higher levels of sTβRII protein and Smad 4ΔM4 protein compared with those of untransfected cells, confirming that HFF-1 cells transfected with the two lentivirus vectors can efficiently express the target proteins. (2) There were statistically significant differences in the protein and mRNA expressions of fibronectin in cells of the 6 groups (with F values respectively 53.536 and 24.365, P values below 0.001). The protein and mRNA expressions of fibronectin in cells of positive control group (respectively 1.60 ± 0.18 and 1.99 ± 0.40) were similar with those of negative virus group (respectively 1.60 ± 0.15 and 1.94 ± 0.28, with q values respectively 0.091 and 0.419, P values above 0.05), and they were significantly higher than those of the rest 4 groups (with q values from 5.245 to 18.228, P values below 0.05). The protein and mRNA expressions of fibronectin in cells of co-transfection group (respectively 0.60 ± 0.05 and 0.70 ± 0.11) were significantly lower than those of sTβRII group (respectively 0.89 ± 0.13 and 1.24 ± 0.17) and Smad 4ΔM4 group (respectively 0.91 ± 0.14 and 1.28 ± 0.19, with q values from 3.964 to 4.294, P values below 0.05). (3) There were statistically significant differences in the protein expressions of CTGF and total collagen in the cell culture supernate of the 6 groups (with F values respectively 107.680 and 38.347, P values below 0.001). The protein expressions of CTGF and total collagen in the cell culture supernate of positive control group were similar with those of negative virus group (with q values respectively 1.106 and 0.491, P values above 0.05), and they were significantly higher than those of the rest 4 groups (with q values from 6.414 to 26.420, P values below 0.05). The protein expressions of CTGF and total collagen in the cell culture supernate of co-transfection group were significantly lower than those of sTβRII group and Smad 4ΔM4 group (with q values from 3.424 to 7.143, P values below 0.05).</p><p><b>CONCLUSIONS</b>In human skin fibroblasts, blockage of two sites of TGF-β/Smad signaling can reduce the expression of scar related proteins which are up-regulated by TGF-β1 to a greater extent than that of blocking one single site.</p>


Subject(s)
Humans , Cicatrix , Connective Tissue Growth Factor , Fibroblasts , Metabolism , Genetic Vectors , Lentivirus , Genetics , Protein Serine-Threonine Kinases , RNA, Messenger , Genetics , Receptors, Transforming Growth Factor beta , Signal Transduction , Smad Proteins , Genetics , Metabolism , Smad Proteins, Inhibitory , Genetics , Transfection , Transforming Growth Factor beta , Pharmacology , Transforming Growth Factors
8.
Kidney Research and Clinical Practice ; : 93-97, 2015.
Article in English | WPRIM | ID: wpr-50610

ABSTRACT

BACKGROUND: Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine involved in immune disorders, cancer, asthma, lung fibrosis, and chronic kidney disease, and its signal pathways are considered crucial mediators of a variety of cellular processes. In addition, several recent studies have reported that TGF-beta receptor (TGF-betaR) gene polymorphism is associated with chronic kidney disease. However, the association between end-stage renal disease (ESRD) and the TGF-beta gene polymorphism has not been sufficiently investigated. In this study, we hypothesized that polymorphisms of the TGF-beta ligands or their receptors may be related to ESRD. METHODS: We assessed the relationship between four single-nucleotide polymorphisms (SNPs) in the TGF-betaR2 and TGF-beta2 genes and ESRD, in 312 patients with ESRD and 258 controls. RESULTS: Compared with the control participants, the frequencies of the TGF-betaR2 (rs764522*C) and TGF-betaR2 (rs3087465*G) alleles were significantly higher in the patients with ESRD. Genotyping analysis demonstrated that two SNPs in TGF-betaR2 of the four SNPs included in the study were significantly associated with ESRD in the codominant 1 [rs764522, odds ratio (OR)=1.65; rs3087465, OR=1.63], dominant (rs764522, OR=1.63; rs3087465, OR=1.57), and log-additive (rs764522, OR=1.54; rs3087465, OR=1.39) models after adjusting for age and sex. CONCLUSION: We suggest that TGF-betaR2 polymorphisms (rs764522 and rs3087465) increase the risk of development of ESRD.


Subject(s)
Humans , Alleles , Asthma , Fibrosis , Immune System Diseases , Kidney Failure, Chronic , Ligands , Lung , Odds Ratio , Polymorphism, Single Nucleotide , Receptors, Transforming Growth Factor beta , Renal Insufficiency, Chronic , Signal Transduction , Transforming Growth Factor beta , Transforming Growth Factor beta2
9.
Chinese Journal of Industrial Hygiene and Occupational Diseases ; (12): 340-347, 2014.
Article in Chinese | WPRIM | ID: wpr-306304

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the distribution and expression of transforming growth factor beta (TGF-β) receptors I and II, p38 mitogen-activated protein kinase (p38 MAPK), and type I and type III collagen in the lungs of rats with silicosis and cultured pulmonary fibroblasts, and to investigate the relationship of the anti-fibrosis effect of N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) with its inhibition of TGF-β receptor-mediated p38 MAPK pathway activity.</p><p><b>METHODS</b>Rats were randomly divided into control group, silicosis model group, and AcSDKP treatment group (n = 10 for each group). For the model group and AcSDKP treatment group, rats were intratracheally instilled with silica to establish a silicosis model. Cultured pulmonary fibroblasts from neonatal rats were divided into control group, TGF-β1 stimulation group, TGF-β receptor inhibition group, p38 MAPK pathway inhibition group, and AcSDKP treatment group. The protein expression of TGF-β receptors I and II, p38 MAPK, and type I and type III collagen were determined by immunohistochemistry and Western blot. The mRNA expression of TGF-β receptors I and II were determined by real-time PCR. The distribution and nuclear translocation of phospho-p38 MAPK in cultured fibroblasts were determined by laser scanner confocal microscopy.</p><p><b>RESULTS</b>In the AcSDKP treatment group, AcSDKP reduced the expression of TGF-β receptors I and II, phospho-p38 MAPK, and type I and type III collagen to 86.12%, 41.01%, 42.63%, 89.05%, and 52.71%, respectively, of those of the silicosis model group (P < 0.05). In cultured fibroblasts, AcSDKP reduced the mRNA expression of TGF-β receptors I and II to 42.26% and 54.33%, respectively, of those of the TGF-β1 stimulation group; the protein expression of TGF-β receptors I and II, phospho-p38 MAPK, and type 1 and type III collagen was reduced to 58.14%, 51.40%, 45.6%, 58.04%, and 44.74%, respectively, of those of the TGF-β1 stimulation group. The phospho-p38 MAPK translocation from plasma to the nucleus was also inhibited; the nucleus/plasma ratio of p38 MAPK and the protein expression of type I and type III collagen were reduced to 68.60%, 58.04%, and 44.74%, respectively, of those of the TGF-β stimulation group (P < 0.05).</p><p><b>CONCLUSION</b>AcSDKP can inhibit the expression of collagen through inhibition of TGF-β receptor-mediated p38 MAPK pathway activity, and is thus able to exert anti-fibrosis effect in rats with silicosis.</p>


Subject(s)
Animals , Male , Rats , Cells, Cultured , Collagen , Metabolism , Disease Models, Animal , Fibroblasts , Metabolism , MAP Kinase Signaling System , Oligopeptides , Pharmacology , Protein Serine-Threonine Kinases , Metabolism , Rats, Wistar , Receptors, Transforming Growth Factor beta , Metabolism , Silicosis , Metabolism , Transforming Growth Factor beta , Metabolism , p38 Mitogen-Activated Protein Kinases , Metabolism
10.
Protein & Cell ; (12): 503-517, 2014.
Article in English | WPRIM | ID: wpr-757479

ABSTRACT

Transforming growth factor-β (TGF-β) members are key cytokines that control embryogenesis and tissue homeostasis via transmembrane TGF-β type II (TβR II) and type I (TβRI) and serine/threonine kinases receptors. Aberrant activation of TGF-β signaling leads to diseases, including cancer. In advanced cancer, the TGF-β/SMAD pathway can act as an oncogenic factor driving tumor cell invasion and metastasis, and thus is considered to be a therapeutic target. The activity of TGF-β/SMAD pathway is known to be regulated by ubiquitination at multiple levels. As ubiquitination is reversible, emerging studies have uncovered key roles for ubiquitin-removals on TGF-β signaling components by deubiquitinating enzymes (DUBs). In this paper, we summarize the latest findings on the DUBs that control the activity of the TGF-β signaling pathway. The regulatory roles of these DUBs as a driving force for cancer progression as well as their underlying working mechanisms are also discussed.


Subject(s)
Animals , Humans , Molecular Targeted Therapy , Receptors, Transforming Growth Factor beta , Metabolism , Signal Transduction , Smad Proteins , Physiology , Transforming Growth Factor beta , Physiology , Ubiquitin Thiolesterase , Metabolism , Ubiquitin-Specific Proteases , Ubiquitination
11.
Journal of Huazhong University of Science and Technology (Medical Sciences) ; (6): 415-419, 2014.
Article in English | WPRIM | ID: wpr-351061

ABSTRACT

The deubiquitinating enzyme ubiquitin specific peptidase 15 (USP15) is regarded as a regulator of TGFβ signaling pathway. This process depends on Smad7, the inhibitory factor of the TGFβ signal, and type I TGFβ receptor (TβR-I), one of the receptors of TGFβ. The expression level of USP15 seems to play vital roles in the pathogenesis of many neoplasms, but so far there has been no report about USP15 in psoriasis. In this study, immunohistochemical staining of USP15, TβR-I and Smad7 was performed in 30 paraffin-embedded psoriasis specimens and 10 normal specimens to investigate the expression of USP15, TβR-I and Smad7 in psoriasis and to explore the relevance among them. And USP15 small interfering RNA (USP15 siRNA) was used to transfect Hacat cells to detect the mRNA expression of TβR-I and Smad7. Of 30 cases of psoriasis in active stage, 28, 24 and 26 cases were positive for USP15, TβR-I and Smad7 staining, respectively. The positive rates of USP15 and Smad7 were significantly higher in psoriasis specimens than in normal skin specimens (44.1%±26.0% vs. 6.1%±6.6%, 47.2%±27.1% vs. 6.6%±7.1%), and positive rate of TβR-I (20.3%±22.2%) in psoriasis was lower than that in normal skin specimens (46.7%±18.2%). There was a significant positive correlation between USP15 and Smad7 expression, and significant negative correlations between USP15 and TβR-expression, an I d between TβR- and Smad7 expression I in psoriasis. After transfection of USP15 siRNA in Hacat cells, the expression of TβR-mRNA was up I -regulated and that of Smad7 was down-regulated. It is concluded that USP15 may play a role in the pathogenesis of psoriasis through regulating the TβR-I/Smad7 pathway and there may be other cell signaling pathways interacting with USP15 to take part in the development of psoriasis.


Subject(s)
Adult , Female , Humans , Male , Middle Aged , Young Adult , Cell Line , Gene Expression , Immunohistochemistry , Keratinocytes , Cell Biology , Metabolism , Protein Serine-Threonine Kinases , Genetics , Psoriasis , Genetics , Metabolism , RNA Interference , Receptors, Transforming Growth Factor beta , Genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Genetics , Skin , Metabolism , Smad7 Protein , Genetics , Ubiquitin-Specific Proteases , Genetics
12.
Chinese Journal of Stomatology ; (12): 101-105, 2014.
Article in Chinese | WPRIM | ID: wpr-274132

ABSTRACT

<p><b>OBJECTIVE</b>To examine the expression of proteoglycan 4 (PRG-4) induced by hydrostatic pressure in rat temporomandibular synovial fibroblasts and investigate the possible mechanism.</p><p><b>METHODS</b>The cultured rat temporomandibular synovial fibroblasts were subjected to 100 kPa magnitude intermittent hydrostatic pressure (IHP) at frequency of 4 h/day, and the static group served as control. The expressions of Smad pathway proteins and p38MAPK pathway proteins were analyzed by Western blot and immunofluorescence staining. Then the cells were incubated with SB431542, the inhibitor of transforming growth factor (TGF)-β receptor. Western blot and reverse transcription PCR were used to detect the PRG-4 expression after 72 h.</p><p><b>RESULTS</b>The expression of phosphorylated Smad-2 and phosphorylated Smad-3 were increased after 1 h of IHP, reaching a maximum after 2 h and 4 h of IHP, respectively.However, the protein content of phosphorylated p38 did not vary significantly. In addition, IHP induced nuclear translocation of Smad-2/-3, and the immunofluorescence staining signal intensity markedly increased (24.11 ± 4.70)(P < 0.05). The levels of PRG-4 mRNA were significantly increased by IHP (1.48 ± 0.08)(P < 0.05). Treatment of cells with SB431542 could decrease the expression of PRG-4 mRNA significantly after IHP (0.47 ± 0.05)(P < 0.05). In addition, SB431542 inhibited the expression of PRG-4 protein induced by IHP.</p><p><b>CONCLUSIONS</b>Smad signal acts as an essential signal pathway to regulate PRG-4 expression induced by IHP.</p>


Subject(s)
Animals , Rats , Cells, Cultured , Fibroblasts , Hydrostatic Pressure , Phosphorylation , Protein Serine-Threonine Kinases , Proteoglycans , RNA, Messenger , Receptors, Transforming Growth Factor beta , Signal Transduction , Smad Proteins , Physiology , Synovial Fluid , Metabolism , Temporomandibular Joint , Metabolism , p38 Mitogen-Activated Protein Kinases
13.
The Korean Journal of Internal Medicine ; : 281-290, 2014.
Article in English | WPRIM | ID: wpr-62924

ABSTRACT

Pulmonary fibrosis is a fatal progressive disease with no effective therapy. Transforming growth factor (TGF)-beta1 has long been regarded as a central mediator of tissue fibrosis that involves multiple organs including skin, liver, kidney, and lung. Thus, TGF-beta1 and its signaling pathways have been attractive therapeutic targets for the development of antifibrotic drugs. However, the essential biological functions of TGF-beta1 in maintaining normal immune and cellular homeostasis significantly limit the effectiveness of TGF-beta1-directed therapeutic approaches. Thus, targeting downstream mediators or signaling molecules of TGF-beta1 could be an alternative approach that selectively inhibits TGF-beta1-stimulated fibrotic tissue response while preserving major physiological function of TGF-beta1. Recent studies from our laboratory revealed that TGF-beta1 crosstalk with epidermal growth factor receptor (EGFR) signaling by induction of amphiregulin, a ligand of EGFR, plays a critical role in the development or progression of pulmonary fibrosis. In addition, chitotriosidase, a true chitinase in humans, has been identified to have modulating capacity of TGF-beta1 signaling as a new biomarker and therapeutic target of scleroderma-associated pulmonary fibrosis. These newly identified modifiers of TGF-beta1 effector function significantly enhance the effectiveness and flexibility in targeting pulmonary fibrosis in which TGF-beta1 plays a significant role.


Subject(s)
Animals , Humans , Drug Design , Hexosaminidases/antagonists & inhibitors , Lung/drug effects , Molecular Targeted Therapy , Pulmonary Fibrosis/drug therapy , Receptor Cross-Talk , ErbB Receptors/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction , Transforming Growth Factor beta1/antagonists & inhibitors
14.
Chinese Journal of Contemporary Pediatrics ; (12): 767-770, 2013.
Article in Chinese | WPRIM | ID: wpr-241425

ABSTRACT

<p><b>OBJECTIVE</b>To examine the single nucleotide polymorphism (SNP) (rs1495592) in transforming growth factor-beta receptor 2 (TGFBR2) gene in children, and to investigate its association with Kawasaki disease (KD) and coronary artery lesions (CALs).</p><p><b>METHODS</b>Thirty-five KD patients, 14 of whom had CALs (CAL subgroup), were selected as the case group, and 25 healthy age-matched children were selected as the control group. The SNP (rs1495592) in TGFBR2 gene was studied by gene sequencing. The association of SNP (rs1495592) with KD and (CALs) was analyzed based on the sequencing results.</p><p><b>RESULTS</b>There were no significant differences in genotype frequency distribution (χ(2)=0.566, P=0.452) and allele frequency distribution (χ(2)=0.216, P=0.642) between the two groups. Genotypes in the CAL subgroup included CC (21.4%) and CT+TT (78.6%), while genotypes in the non-CAL subgroup included CC (61.9%) and CT+TT (38.1%). There was significant difference in genotype frequency distribution between the two groups (χ(2)=5.546, P=0.019), but without significant difference in allele frequency distribution (χ(2)=3.673, P=0.055).</p><p><b>CONCLUSIONS</b>The SNP (rs1495592) in TGFBR2 gene may not be associated with development of KD in children, but it is associated with CALs in children with KD.</p>


Subject(s)
Female , Humans , Infant , Male , Coronary Artery Disease , Genetics , Genetic Predisposition to Disease , Genotype , Mucocutaneous Lymph Node Syndrome , Genetics , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases , Genetics , Receptors, Transforming Growth Factor beta , Genetics , Signal Transduction , Transforming Growth Factor beta , Physiology
15.
Chinese Journal of Integrated Traditional and Western Medicine ; (12): 819-824, 2013.
Article in Chinese | WPRIM | ID: wpr-287461

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the regulative mechanism of the diterpene phenol extract of Rosmarinus Officinalis (DERO) on the imbalance of collagen metabolism of the lung tissue in pulmonary fibrosis rats.</p><p><b>METHODS</b>Fifty healthy Sprague-Dawley rats were randomly divided into the normal saline group (NS), the bleomycin-induced lung injury group (BLM), the low dose DERO group (at the daily dose of 50 mg/kg), the moderate dose DERO group (at the daily dose of 100 mg/kg), and the high dose DERO group (at the daily dose of 200 mg/kg), 10 in each group (abbreviated as DERO 1, 2, 3, respectively). The pulmonary fibrosis rat model was prepared by disposable intratracheal instillation of bleomycin. DERO was administered by gastrogavage as intervention during the repairing process of lung injury. On the morning of the 29th day, the rats' lung tissue was extracted. The karyocyte number, collagen protein, type I collagen (collagen I) and transforming growth factor-beta type II receptor (TGFbetaR II), Smad4 mRNA expressions were semi-quantitatively determined using tissue microarray, HE staining, collagen fiber dyeing, immunohistochemical assay, and in situ hybridization. Using real-time fluorescent quantification RT-PCR, the mRNA expression of transforming growth factor-beta1 (TGF-beta1) were detected.</p><p><b>RESULTS</b>Compared with the NS group, the collagen deposition of the lung tissue was obvious and the inflammatory infiltration was more severe in the BLM group (P < 0.05, P < 0.01). There was no statistical difference in the aforesaid 4 indices between the DERO1 group and the BLM group (P > 0.05). The collagen deposition and the inflammatory infiltration were obviously alleviated in the DERO2 and DERO3 groups (P < 0.05, P < 0.01). Compared with the NS group, the mRNA expressions of collagen-I, TGF-beta1 R II, Smad4, and TGF-beta1 were obviously up-regulated in the BLM group (P < 0.05, P < 0.01). Compared with the BLM group, the aforesaid four indices were not statistically changed in the DERO1 group (P > 0.05). But the mRNA expressions of collagen-I, TGF-beta1 R II, Smad4, and TGF-beta1 were obviously downregulated in the DERO2 and DERO3 groups (P < 0.05, P < 0.01). But the down-regulation of Smad4 expression was not obvious in the DERO2 and the DERO3 groups (P > 0.05). Compared with the DERO1 group, the mRNA expressions of collagen-I, TGF-beta1, R II, TGFbeta1 were all obviously lower in the DERO2 and the DERO3 groups (P < 0.05). But there was no statistical difference in the aforesaid 4 indices between the DERO2 group and the DERO3 group (P > 0.05).</p><p><b>CONCLUSIONS</b>DERO could regulate imbalanced collagen metabolism of pulmonary fibrosis. It could inhibit excessive deposition of collagen fibers, especially excessive deposition of collagen- I. Its mechanisms might be realized by inhibiting up-regulation of TGF-beta1 and TGFbetaR II mRNA expressions, thus interfering the activation of TGF-beta-Smad signaling pathway on target genes, especially on type I procollagen target gene.</p>


Subject(s)
Animals , Female , Male , Rats , Collagen Type I , Metabolism , Diterpenes , Pharmacology , Lung , Metabolism , Plant Extracts , Pharmacology , Protein Serine-Threonine Kinases , Metabolism , Pulmonary Fibrosis , Metabolism , RNA, Messenger , Genetics , Rats, Sprague-Dawley , Receptors, Transforming Growth Factor beta , Metabolism , Rosmarinus , Chemistry , Signal Transduction , Transforming Growth Factor beta1 , Metabolism
16.
Chinese Medical Journal ; (24): 3340-3343, 2013.
Article in English | WPRIM | ID: wpr-354484

ABSTRACT

<p><b>BACKGROUND</b>It has been reported that there is a significant difference in the local tissue concentration of transforming growth factor (TGF)-β1 between chronic rhinosinusitis without nasal polyps (CRSsNP) and chronic rhinosinusitis without nasal polyps (CRSwNP) patients. TGF-β has been reported to play an important role in regulating epithelial cell repair in lower airway remodeling and may be a critical factor involved in the remodeling process of chronic rhinosinusitis (CRS).</p><p><b>METHODS</b>Ethmoidal mucosal samples collected from CRS and healthy control patients were analyzed for TGF-β1, TGF-β receptor I, TGF-β receptor II, Smad3, phospho-Smad3, Smad7, and Smad anchor for receptor activation by Western blotting analysis. The proliferation of sinonasal epithelial cells at baseline and after TGF-β1 and/or EGF stimulation was evaluated by the MTT assay.</p><p><b>RESULTS</b>In CRSsNP, TGF-β1, TGF-β receptor I, TGF-β receptor II, and Smad3 protein levels were significantly higher than controls. In CRSwNP, TGF-β1, Smad3, and pSmad3 protein levels were significantly lower than controls. Smad7 protein was significantly higher in CRS than controls. In vitro experiments demonstrated that the baseline proliferation levels of sinonasal epithelial cells were lower in CRS than controls.</p><p><b>CONCLUSIONS</b>CRSwNP is characterized by a lower level of TGF-signaling compared with the control. In CRSsNP, although the upstream signaling of TGF-β was enhanced, the high Smad7 protein expression may restrain the downstream signaling components (e.g., pSmad3) and the TGF-β antiproliferative effect on sinonasal epithelium. The difference in the local tissue concentration of TGF-β1 between CRSsNP and CRSwNP patients did not result in significant differences in epithelial proliferation.</p>


Subject(s)
Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Benzamides , Pharmacology , Cells, Cultured , Dioxoles , Pharmacology , Intracellular Signaling Peptides and Proteins , Metabolism , Protein Serine-Threonine Kinases , Metabolism , Receptors, Transforming Growth Factor beta , Metabolism , Serine Endopeptidases , Metabolism , Signal Transduction , Sinusitis , Metabolism , Smad3 Protein , Metabolism , Smad7 Protein , Metabolism , Transforming Growth Factor beta , Metabolism , Transforming Growth Factor beta1 , Metabolism
17.
Gut and Liver ; : 213-220, 2013.
Article in English | WPRIM | ID: wpr-197295

ABSTRACT

BACKGROUND/AIMS: We aimed to investigate the correlation between a disintegrin and metalloprotease with thrombospondin motif 2 (ADAMTS-2) and transforming growth factor-beta1 (TGF-beta1) in clinical human cirrhotic tissues. METHODS: The liver tissues of 24 patients (16 cases with cirrhotic portal hypertension as the cirrhosis group and eight cases with healthy livers as the normal group) were collected. Immunohistochemistry and Western blots were performed to evaluate the protein expression levels of ADAMTS-2 and TGF-beta1. Western blots for other key mediators of cirrhotic progression, including SMAD2, SMAD3, TGF-beta receptor II (TGFbetaRII), matrix metalloproteinases 2 (MMP2), and tissue inhibitor of matrix metalloproteinases 2 (TIMP2), were also performed. RESULTS: Cirrhotic tissues showed higher percentages of collagen. The protein expression levels of ADAMTS-2 and TGF-beta1 were significantly higher in the cirrhotic group as compared to the matched normal group (p<0.05), and there was a positive correlation between these two proteins (r=0.862, p<0.01). The protein expressions of MMP2, TIMP2, and TGFbetaRII, as well as the phosphorylated forms of SMAD2 and SMAD3, were significant higher in the cirrhotic group (p<0.01 or p<0.05). CONCLUSIONS: These findings suggested that ADAMTS-2 and TGF-beta1 may play important roles in the pathogenesis of human cirrhosis; specifically, TGF-beta1 may induce the expression of ADAMTS-2 through the TGFbeta/SMAD pathway.


Subject(s)
Humans , Blotting, Western , Collagen , Fibrosis , Hypertension, Portal , Immunohistochemistry , Liver , Matrix Metalloproteinases , Proteins , Receptors, Transforming Growth Factor beta , Thrombospondins , Transforming Growth Factor beta1
18.
Arq. bras. endocrinol. metab ; 56(8): 473-478, Nov. 2012. ilus, tab
Article in English | LILACS | ID: lil-660252

ABSTRACT

OBJECTIVE: To screen for mutations in AMH and AMHR2 genes in patients with persistent Müllerian duct syndrome (PMDS). PATIENTS AND METHOD: Genomic DNA of eight patients with PMDS was obtained from peripheral blood leukocytes. Directed sequencing of the coding regions and the exon-intron boundaries of AMH and AMHR2 were performed. RESULTS: The AMH mutations p.Arg95*, p.Arg123Trp, c.556-2A>G, and p.Arg502Leu were identified in five patients; and p.Gly323Ser and p.Arg407* in AMHR2 of two individuals. In silico analyses of the novel c.556-2A>G, p.Arg502Leu and p.Arg407* mutations predicted that they were harmful and were possible causes of the disease. CONCLUSION: A likely molecular etiology was found in the eight evaluated patients with PMDS. Four mutations in AMH and two in AMHR2 were identified. Three of them are novel mutations, c.556-2A>G, and p.Arg502Leu in AMH; and p.Gly323Ser in AMHR2. Arq Bras Endocrinol Metab. 2012;56(8):473-8.


OBJETIVO: Analisar os genes AMH e AMHR2 em indivíduos com síndrome de persistência dos ductos de Müller (SPDM). PACIENTES E MÉTODO: Amostras de DNA genômico de oito pacientes com SPDM foram obtidas de leucócitos de sangue periférico. Sequenciamento direto da região codificadora e das áreas intrônicas próximas aos éxons dos genes AMH e AMHR foi realizado. RESULTADOS: As mutações p.Arg95*, p.Arg123Trp, c.556-2A>G e p.Arg502Leu no gene AMH foram identificadas em cinco pacientes e as mutações p.Gly323Ser e p.Arg407* no gene AMHR2, em dois indivíduos. As análises in silico das mutações c.556-2A>G, p.Arg502Leu e p.Arg407*, não descritas anteriormente na literatura, previram que elas são deletérias e possivelmente a causa da doença. CONCLUSÃO: Uma provável etiologia molecular foi encontrada nos oito pacientes portadores de SPDM avaliados. No gene do AMH foram identificadas quatro mutações e no AMHR2, duas mutações. Três das seis mutações encontradas são mutações novas, c.556-2A>G e p.Arg502Leu no gene AMH; e p.Gly323Ser no AMHR2. Arq Bras Endocrinol Metab. 2012;56(8):473-8.


Subject(s)
Adolescent , Adult , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Male , Young Adult , /genetics , Anti-Mullerian Hormone/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , /blood , Anti-Mullerian Hormone/blood , DNA Mutational Analysis , Polymerase Chain Reaction , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood
19.
Medical Journal of the Islamic Republic of Iran. 2012; 26 (1): 31-40
in English | IMEMR | ID: emr-128604

ABSTRACT

Primary and secondary malignant central nervous system [CNS] tumors are devastating invasive tumors able to give rise to many kinds of differentiated tumor cells. Glioblastoma multiform [GBM], is the most malignant brain tumor, in which its growth and persistence depend on cancer stem cells with enhanced DNA damage repair program that also induces recurrence and resists current chemo- and radiotherapies. Unlike non-tumor stem cells, tumor stem cells lack the normal mechanisms that regulate proliferation and differentiation, resulting in uncontrolled production and incomplete differentiation of tumor cells. In current paper recent developments and new researches in the field of brain tumor stem cells have been reviewed


Subject(s)
Stem Cells , Glioblastoma , Glioma , MicroRNAs , Dacarbazine/analogs & derivatives , Adenoviridae , Phosphofructokinase-2 , Microfilament Proteins , Vesicular Transport Proteins , Protein Serine-Threonine Kinases , Receptors, Transforming Growth Factor beta , Protein Kinase C , Tumor Suppressor Protein p53 , Matrix Metalloproteinases , Y-Box-Binding Protein 1 , Nitric Oxide Synthase Type II , Tubulin , Immunotherapy
20.
Chinese Journal of Applied Physiology ; (6): 284-287, 2012.
Article in Chinese | WPRIM | ID: wpr-329885

ABSTRACT

<p><b>OBJECTIVE</b>To generate the skeletal muscle-specific transforming growth factor beta receptor II (TbetaR II) gene knockout mice for the research on the function of the TbetaR II gene in skeletal muscles.</p><p><b>METHODS</b>TbetaR II (flox/flox) mice were generated using embryonic stem cell technology. The MCK-Cre mice were engineered containing Cre recombinase under the control of creatine kinase (MCK) muscle-specific promoter. TbetaR II (flox/flox) mice were crossed with MCK-Cre mice generating TbetaR II (flox/flox)/MCK-Cre double Tg mice. And then, TbetaR II (flox/wt) /MCK-Cre(+) double Tg mice were crossed with TbetaR II (flox/flox) mice to generate TbetaR II (flox/flox)/MCK- Cre(+) mice genetically ablating TbetaR II in cre-expressing skeletal muscle cells.</p><p><b>RESULTS</b>As predicted, mice lacking TbetaR II by gene targeting in skeletal muscles were generated first in the world using Cre/loxP system. TbetaR II null mutant mice were viable, fertile and showed apparently normal development.</p>


Subject(s)
Animals , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal , Metabolism , Protein Serine-Threonine Kinases , Genetics , Metabolism , Receptors, Transforming Growth Factor beta , Genetics , Metabolism , Recombination, Genetic , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL