Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 38
Filtrer
Plus de filtres










Gamme d'année
1.
Cell Commun Signal ; 22(1): 17, 2024 01 05.
Article de Anglais | MEDLINE | ID: mdl-38183022

RÉSUMÉ

BACKGROUND: The epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells participated in the development of retinal fibrosis. SB431542 is a small molecule inhibitor with inhibitory effects on the ALK4, ALK5 and ALK7. Our study aimed to explore the effect of SB431542 on the EMT of RPE cells and to provide new ideas for the treatment of retinal fibrosis. METHODS: We performed fundus fluorescein angiography, optical coherence tomography and hematoxylin-eosin staining in vivo to observe the effect of SB431542 on choroidal neovascularization (CNV)-induced retinopathy. The proliferation, migration, cytoskeleton, adhesion, reactive oxygen species (ROS), mitochondrial morphology and membrane potential of RPE cells were observed in vitro through fluorescein diacetate staining, Cell Counting Kit-8 experiment, wound healing assay, phalloidin staining, immunofluorescence, MitoSOX, DCFH-DA, MitoTracker and JC-10 staining. Western blot, reverse transcription quantitative and immunofluorescence were used to detect the expression of EMT-related markers, pERK1/2, pGSK3ß and ß-catenin. RESULTS: SB431542 significantly alleviated retinopathy in the CNV model. The proliferation, migration and adhesion in RPE cells decreased to a certain extent in SB431542 treatment. SB431542 partially normalized the structure of RPE cells. The expression levels of E-cadherin increased, while the expression levels of laminin and N-cadherin decreased with SB431542 treatment. SB431542 reduced the production of total ROS, mitochondrial SOX and recovered the mitochondrial membrane potential to a certain degree. In addition, our study showed that SB431542 downregulated the phosphorylation of ERK1/2, GSK3ß and the expression of ß-catenin. CONCLUSION: SB431542 improved EMT in RPE cells by maintaining mitochondrial homeostasis via the ERK1/2 and GSK3ß/ß-catenin pathways. Video Abstract SB431542 inhibits EMT in RPE cells under high glucose conditions.


Sujet(s)
Néovascularisation choroïdienne , Rétinopathies , Humains , bêta-Caténine , Glycogen synthase kinase 3 beta , Espèces réactives de l'oxygène , Homéostasie , Fibrose , Glucose/toxicité
2.
Biomed Pharmacother ; 161: 114537, 2023 May.
Article de Anglais | MEDLINE | ID: mdl-36933378

RÉSUMÉ

Silicosis is a devastating interstitial lung disease characterized by silicon nodules and diffuse pulmonary fibrosis. To date, inefficient therapy is still a challenge of this disease due to its complicated pathogenesis. Hepatocyte growth factor (HGF) which is highly expressed in hepatocyte with anti-fibrotic and anti-apoptotic function was downregulated in silicosis. In addition, the upregulation of transforming growth factor-beta (TGF-ß), another pathological molecular was observed to aggravate the severity and accelerate the progression of silicosis. Here AAV expressed HGF with targeting pulmonary capillaries and SB431542, the inhibitor of TGF-ß signal pathway, were simultaneously adopted to synergistically reduce silicosis fibrosis. In vivo result demonstrated that the cooperation of HGF with SB431542 showed strong anti-fibrosis effects on the silicosis mice via tracheal administration of silica, compared to the separate treatment. The high efficacy was mainly achieved by remarkably by reducing ferroptosis of lung tissue. In our point, the combination of AAV9-HGF with SB431542 provide an alternative to relieve silicosis fibrosis from the perspective of targeting pulmonary capillaries.


Sujet(s)
Ferroptose , Silicose , Souris , Animaux , Facteur de croissance transformant bêta/métabolisme , Facteur de croissance des hépatocytes , Facteur de croissance transformant bêta-1/métabolisme , Fibrose , Silicose/traitement médicamenteux , Silicose/métabolisme
3.
J Autoimmun ; 132: 102894, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-36030617

RÉSUMÉ

Lupus nephritis (LN) is the most common cause of morbidity and mortality in patients with systemic lupus erythematosus (SLE). Currently, immunosuppressive treatments for LN are suboptimal and can induce significant side effects. SB431542 is a selective and potent inhibitor of the TGFß/Activin/NODAL pathway. Here, we study the effects of SB431542 treatment on LN and discuss the potential mechanisms. SB431542 ameliorated clinical outcomes with a consequent histological improvement in NZB/W mice. A comparative transcriptional profiling analysis revealed 586 differentially expressed genes (247 downregulated genes) in the SB431542 group compared to the control group. We found that the downregulated genes were mainly enriched in the biological processes of B cell activation, B cell proliferation, B cell differentiation, and B cell receptor signaling. Kyoto encyclopedia of genes and genomes pathway analysis revealed that the hematopoietic cell linage pathway was significantly downregulated in the SB431542 group. In addition, we observed that SB431542 reduced the splenic or renal levels of CD20 and the serum levels of anti-dsDNA antibody (IgG) in NZB/W mice. Furthermore, qRT-PCR and immunohistochemistry confirmed that SB431542 inhibits the production of TLR9, TGFß1, and PDGFB. Thus, due to its immunomodulatory activities, SB431542 could be considered for clinical therapy development for LN.


Sujet(s)
Glomérulonéphrite lupique , Animaux , Souris , Protéines proto-oncogènes c-sis , Glomérulonéphrite lupique/traitement médicamenteux , Glomérulonéphrite lupique/génétique , Récepteur-9 de type Toll-like/génétique , Bécaplermine , Souris de lignée NZB , Facteur de croissance transformant bêta
4.
Biochim Biophys Acta Gen Subj ; 1866(11): 130215, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-35905921

RÉSUMÉ

BACKGROUND: Breast cancer is a heterogenous disease composed of multiple clonal populations and the mechanism by which the tumor microenvironment induces cancer stem cell plasticity is not fully understood. METHODS: MCF7 breast cancer cells were treated with macrophage conditioned medium (MɸCM). PD98059 and SB431542 were used for ERK and TGF-ßR inhibition respectively. Epithelial-mesenchymal transition (EMT) and cancer stem cell markers (CSC) were studied using qRT-PCR and flowcytometry. SCID mice were used for animal experiments. RESULTS: MɸCM- induced ERK/TGF-ß1 signaling led to enrichment of CSC and EMT in MCF7 cells and mammospheres. These effects were abrogated by both MEK inhibitor PD98059 (TGF-ß1 synthesis) and SB431542 (TGF-ß1 signaling). The increase in CSC was both hybrid (ALDH1+) and mesenchymal (CD44+ CD24- cells). Increase in hybrid E/M state was at a single cell level as confirmed by the increase in both claudin-1 (E) and vimentin (M). This did not have any growth advantage in SCID mice and monitoring of CSC and EMT markers before and after growth in SCID mice indicated reversal of these markers in tumor cells recovered from mice. Removal of MɸCM and neutralization of TNF-α, IL-6 and IL-1ß in MɸCM abrogated ERK phosphorylation, TGF-ß and CSC enrichment indicating the requirement of continuous signaling for maintenance. CONCLUSIONS: ERK signaling plays an important role in MɸCM- induced EMT and CSC plasticity which is completely reversible upon withdrawal of signals. GENERAL SIGNIFICANCE: Our experimental observations support the semi-independent nature of EMT-stemness connection which is very dynamic and reversible depending on the microenvironment.


Sujet(s)
Transition épithélio-mésenchymateuse , Tumeurs , Animaux , Humains , Cellules MCF-7 , Macrophages , Souris , Souris SCID , Cellules souches tumorales , Facteur de croissance transformant bêta-1
5.
Regen Ther ; 20: 165-186, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35620640

RÉSUMÉ

Introduction: Efficient induction of the otic placode, the developmental origin of the inner ear from human pluripotent stem cells (hPSCs), provides a robust platform for otic development and sensorineural hearing loss modelling. Nevertheless, there remains a limited capacity of otic lineage specification from hPSCs by stepwise differentiation methods, since the critical factors for successful otic cell differentiation have not been thoroughly investigated. In this study, we developed a novel differentiation system involving the use of a three-dimensional (3D) floating culture with signalling factors for generating otic cell lineages via stepwise differentiation of hPSCs. Methods: We differentiated hPSCs into preplacodal cells under a two-dimensional (2D) monolayer culture. Then, we transferred the induced preplacodal cells into a 3D floating culture under the control of the fibroblast growth factor (FGF), bone morphogenetic protein (BMP), retinoic acid (RA) and WNT signalling pathways. We evaluated the characteristics of the induced cells using immunocytochemistry, quantitative PCR (qPCR), population averaging, and single-cell RNA-seq (RNA-seq) analysis. We further investigated the methods for differentiating otic progenitors towards hair cells by overexpression of defined transcription factors. Results: We demonstrated that hPSC-derived preplacodal cells acquired the potential to differentiate into posterior placodal cells in 3D floating culture with FGF2 and RA. Subsequent activation of WNT signalling induced otic placodal cell formation. By single-cell RNA-seq (scRNA-seq) analysis, we identified multiple clusters of otic placode- and otocyst marker-positive cells in the induced spheres. Moreover, the induced otic cells showed the potential to generate hair cell-like cells by overexpression of the transcription factors ATOH1, POU4F3 and GFI1. Conclusions: We demonstrated the critical role of FGF2, RA and WNT signalling in a 3D environment for the in vitro differentiation of otic lineage cells from hPSCs. The induced otic cells had the capacity to differentiate into inner ear hair cells with stereociliary bundles and tip link-like structures. The protocol will be useful for in vitro disease modelling of sensorineural hearing loss and human inner ear development and thus contribute to drug screening and stem cell-based regenerative medicine.

6.
Int J Radiat Biol ; : 1-15, 2022 May 13.
Article de Anglais | MEDLINE | ID: mdl-35446183

RÉSUMÉ

PURPOSE: We have earlier characterized increased TGF-ß signaling in radioresistant breast cancer cells. In this study, we wanted to determine the effect of prolonged treatment of TGF-ßR inhibitor SB431542 on radiation-induced signaling, viz., genes regulating apoptosis, EMT, anti and pro-inflammatory cytokines. MATERIALS AND METHODS: Breast cancer cells were pretreated with TGF-ßR inhibitor (SB 431542) followed by exposure to 6 Gy and recovery period of 7 days (D7-6G). We assessed cell survival by MTT assay, cytokines by ELISA and expression analysis by RT-PCR, flow cytometry, and western blot. We carried out migration assays using trans well inserts. We performed bioinformatics analyses of human cancer database through cBioportal. RESULTS: There was an upregulation of TGF-ß1 and 3 and downregulation of TGF-ß2, TGF-ßR1, and TGF-ßR2 in invasive breast carcinoma samples compared to normal tissue. TGF-ß1 and TNF-α was higher in radioresistant D7-6G cells with upregulation of pSMAD3, pNF-kB, and ERK signaling. Pretreatment of D7-6G cells with TGF-ßR inhibitor SB431542 abrogated pSMAD3, increased proliferation, and migration along with an increase in apoptosis and pro-apoptotic genes. This was associated with hybrid E/M phenotype and downregulation of TGF-ß downstream genes, HMGA2 and Snail. There was complete agreement in the expression of mRNA and protein data in genes like vimentin, Snail and HMGA2 in different treatment groups. However, there was disagreement in expression of mRNA and protein in genes like Bax, Bcl-2, E-cadherin, Zeb-1 among the different treatment groups indicating post-transcriptional and post-translational processing of these proteins. Treatment of cells with only SB431542 also increased expression of some E/M genes indicating TGF-ß independent effects. Increased IL-6 and IL-10 secretion by SB431542 along with increase in pSTAT3 and pCREB1 could probably explain these TGF-ß/Smad3 independent effects. CONCLUSION: These results highlight that TGF-ß-pSMAD3 and TNF-α-pNF-kB are the predominant signaling pathways in radioresistant cells and possibility of some TGF-ß/Smad3 independent effects on prolonged treatment with the drug SB431542.

7.
Naunyn Schmiedebergs Arch Pharmacol ; 394(11): 2309-2322, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34499199

RÉSUMÉ

TGF-ß contributes to drug resistance and the invasiveness of tumor cells and weakens the anti-tumor immune responses. The present study aimed at examining the efficacy of the combination of SB431542, as a specific inhibitor of TGF-ßR, and doxorubicin in controlling the melanoma tumor in mice. The impact of the combination of the doxorubicin and SB431542 on the cell growth, apoptosis, migration, and invasiveness of B16-F10 cells was examined. Besides, the B16-F10 tumor was induced in C57BL/6 mice, and the effects of the mentioned treatment on the tumor volume, survival, and the exhaustion state of T cells were evaluated. Although the combination of doxorubicin and SB431542 did not exhibit synergism in the inhibition of cell growth and apoptosis induction, it efficiently prohibited the migration and the epithelial to mesenchymal transition of B16-F10 cells, and the combination of doxorubicin and SB431542 caused an increase in mRNA levels of E-cadherin and, on the other hand, led to a decline in the expression of Vimentin. Tumor volume and the survival of tumor-bearing mice were efficiently controlled by the combination therapy. This treatment also eventuated in a decrease in the percentage of PD-L1+, TCD4+, and TCD8+ cells as indicators of exhausted T cells within the spleens of tumor-bearing mice. Blockade of TGF-ßR also propelled the RAW 264.7 cells towards an anti-tumor M1 macrophage phenotype. The inhibition of TGF-ßR demonstrated a potential to increase the efficacy of doxorubicin chemotherapy by the means of affecting cellular motility and restoring the anti-tumor immune responses.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Mélanome expérimental/traitement médicamenteux , Récepteurs TGF-bêta/antagonistes et inhibiteurs , Animaux , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Benzamides/administration et posologie , Cadhérines/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Dioxoles/administration et posologie , Doxorubicine/administration et posologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Femelle , Mélanome expérimental/anatomopathologie , Souris , Souris de lignée C57BL , Invasion tumorale/prévention et contrôle , Cellules RAW 264.7 , Vimentine/génétique
8.
Arthritis Res Ther ; 23(1): 223, 2021 08 25.
Article de Anglais | MEDLINE | ID: mdl-34433493

RÉSUMÉ

BACKGROUND: Screening abnormal pathways and complement components in the kidneys of patients with lupus nephritis (LN) and NZB/W mice may help to identify complement-related therapeutic targets for LN. METHODS: KEGG and GO enrichment assays were used to analyze kidney microarray data of LN patients and NZB/W mice. Immunohistochemistry and immunofluorescence assays were used to measure renal expression of complement-related proteins and TGFß1. Cytokines were measured using RT-qPCR and ELISA. RESULTS: We screened the renal pathogenic pathways present in LN patients and NZB/W mice and selected the complement activation pathway for further study. The results indicated greater renal expression of C1qa, C1qb, C3, C3aR1, and C5aR1 at the mRNA and protein levels. C3 appeared to be a key factor in LN and the renal signaling downstream of C1 was inhibited. There were significant correlations between the expression of TGFß1 and C3. Analysis of primary cell cultures indicated that TGFß1 promoted the expression of C3 and that a TGFß1 antagonist decreased the levels of C3 and C3aR. TGFß1 inhibition significantly inhibited the deposition of complement-related factors in the kidneys of NZB/W mice. CONCLUSIONS: At the onset of LN, there are significant increases in the renal levels of C3 and other complement pathway-related factors in patients with LN and NZB/W mice. C3 may lead to albuminuria and participate in the pathogenesis of LN. TGFß1 promotes C3 synthesis, and TGFß1 inhibition may block the progression of LN by inhibiting the synthesis of C3 and other complement components.


Sujet(s)
Glomérulonéphrite lupique , Animaux , Activation du complément , Humains , Rein , Glomérulonéphrite lupique/traitement médicamenteux , Glomérulonéphrite lupique/génétique , Souris , Souris de lignée NZB , Transduction du signal
9.
Front Cell Dev Biol ; 9: 602197, 2021.
Article de Anglais | MEDLINE | ID: mdl-33968919

RÉSUMÉ

Mutations in gap junction beta-2 (GJB2), the gene that encodes connexin 26 (CX26), are the most frequent cause of hereditary deafness worldwide. We recently developed an in vitro model of GJB2-related deafness (induced CX26 gap junction-forming cells; iCX26GJCs) from mouse induced pluripotent stem cells (iPSCs) by using Bone morphogenetic protein 4 (BMP4) signaling-based floating cultures (serum-free culture of embryoid body-like aggregates with quick aggregation cultures; hereafter, SFEBq cultures) and adherent cultures. However, to use these cells as a disease model platform for high-throughput drug screening or regenerative therapy, cell yields must be substantially increased. In addition to BMP4, other factors may also induce CX26 gap junction formation. In the SFEBq cultures, the combination of BMP4 and the Activin/Nodal/TGF-ß pathway inhibitor SB431542 (SB) resulted in greater production of isolatable CX26-expressing cell mass (CX26+ vesicles) and higher Gjb2 mRNA levels than BMP4 treatment alone, suggesting that SB may promote BMP4-mediated production of CX26+ vesicles in a dose-dependent manner, thereby increasing the yield of highly purified iCX26GJCs. This is the first study to demonstrate that SB accelerates BMP4-induced iCX26GJC differentiation during stem cell floating culture. By controlling the concentration of SB supplementation in combination with CX26+ vesicle purification, large-scale production of highly purified iCX26GJCs suitable for high-throughput drug screening or regenerative therapy for GJB2-related deafness may be possible.

10.
Anticancer Res ; 41(5): 2431-2440, 2021 May.
Article de Anglais | MEDLINE | ID: mdl-33952468

RÉSUMÉ

BACKGROUND/AIM: Oncolytic reovirus, which is a non-enveloped virus possessing a 10-segmented double-stranded RNA genome, has been anticipated as a novel class of antitumor agent. Hepatocellular carcinoma (HCC) is considered to be a target suitable for reovirus-mediated virotherapy. Transforming growth factor (TGF)-ß plays an important role in the pathogenesis of HCC. TGF-ß-signaling inhibitors have proceeded to clinical trials as potential antitumor agents for HCC. On the other hand, TGF-ß is involved in induction of expression of cathepsins B and L, which are important for reovirus infection. It remains to be examined whether TGF-ß signaling inhibitors affect reovirus-mediated lysis of HCC cells. The aim of this study was to evaluate the effects of TGF-ß-signaling inhibitors on tumor cell lysis efficiency of reovirus in human HCC cells. MATERIALS AND METHODS: Reovirus was added to four types of human HCC cell lines pretreated with one of three TGF-ß type I receptor inhibitors: SB431542, A-83-01, or galunisertib (LY2157299). Cell viability, virus genome copy numbers, and virus protein expression were evaluated following reovirus infection. RESULTS: SB431542 significantly inhibited reovirus-mediated killing of human HCC cell lines, while A-83-01 and galunisertib did not inhibit. CONCLUSION: These data indicate that SB431542 inhibited reovirus-mediated lysis of human HCC cells in a TGF-ß signaling-independent manner.


Sujet(s)
Benzamides/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Dioxoles/pharmacologie , Tumeurs du foie/traitement médicamenteux , Facteur de croissance transformant bêta-1/génétique , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/virologie , Survie cellulaire/effets des médicaments et des substances chimiques , Composés époxy , Humains , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/virologie , Orthoréovirus mammalien/effets des médicaments et des substances chimiques , Orthoréovirus mammalien/génétique , Pyrazoles/pharmacologie , Quinoléines/pharmacologie , ARN double brin/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta-1/antagonistes et inhibiteurs , Tyrosine/analogues et dérivés , Tyrosine/génétique
11.
Dent Mater J ; 40(3): 615-624, 2021 May 29.
Article de Anglais | MEDLINE | ID: mdl-33814531

RÉSUMÉ

Human induced pluripotent stem cells (hiPSCs) are exciting for regenerative medicine due to their multi-potent differentiation. SB431542 bioactive molecule can activate bone morphogenetic protein-signalling in osteoblasts. The objectives were to: (1) develop a novel injectable calcium phosphate cement (CPC)-SB431542 scaffold for dental/craniofacial bone engineering; and (2) investigate cell proliferation and osteo-differentiation of hiPSC-derived mesenchymal stem cells (hiPSC-MSCs) on CPC-SB431542 scaffold. Three groups were tested: CPC control; CPC with SB431542 inside CPC (CPCSM); CPC with SB431542 in osteogenic medium (CPC+SMM). SB431542 in CPC promoted stem cell proliferation and viability. hiPSC-MSCs differentiated into osteogenic lineage and synthesized bone minerals. CPC with SB431542 showed much greater osteo-expressions and more bone minerals than those without SB431542. In conclusion, hiPSC-MSCs on CPC scaffold containing SB431542 showed excellent osteo-differentiation and bone mineral synthesis for the first time. CPC was a suitable scaffold for delivering stem cells and SB431542 to promote bone regeneration in dental/craniofacial applications.


Sujet(s)
Cellules souches pluripotentes induites , Ostéogenèse , Ciments osseux , Phosphates de calcium/pharmacologie , Différenciation cellulaire , Cellules cultivées , Humains , Ingénierie tissulaire , Structures d'échafaudage tissulaires
12.
Cancer Immunol Immunother ; 70(1): 153-168, 2021 Jan.
Article de Anglais | MEDLINE | ID: mdl-32661685

RÉSUMÉ

The contribution of immune cells in soft tissue sarcomas (STS) is not completely known and understanding their role is very essential for employing immunotherapy strategies. Here, we show that murine fibrosarcoma-conditioned medium promoted total spleen cell proliferation but inhibited T cell responses to mitogenic and allo-antigen-mediated stimulation. This increased proliferation was found to be in B cells resulting in generation of Breg further leading to Treg population. This was found to be the same in vitro and in vivo. The phenotype of these B cells was CD19+CD81+CD27+CD25+PD-L1hi and they secreted both IL-10 and TGF-ß. These tumor evoked Bregs (tBreg), when co-cultured with B depleted T cells, suppressed their proliferation in response to anti-CD3/CD28 stimulation. tBreg-induced suppression of T cell responses was not abrogated by the inhibition or neutralization of IL-10 but by the small molecule inhibitor of TGFß Receptor type I, SB431542. While SB531542 per se was not cytotoxic to tumor cells, administration of SB431542 in tumor-bearing mice (TBM) significantly reduced the tumor burden. In addition, the treatment significantly reduced Treg cells and rescued proliferation of T cells in response to mitogen and allo-antigen. Collectively, our results identify that tumor evoked Breg cells mediate T cell immune suppression through TGFß-mediated pathway and that targeting the Breg-Treg axis can be potentially used as an immunotherapy agent.


Sujet(s)
Lymphocytes B régulateurs/effets des médicaments et des substances chimiques , Benzamides/pharmacologie , Dioxoles/pharmacologie , Fibrosarcome/traitement médicamenteux , Récepteurs TGF-bêta/antagonistes et inhibiteurs , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Charge tumorale/effets des médicaments et des substances chimiques , Animaux , Antigènes CD/métabolisme , Lymphocytes B régulateurs/immunologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Fibrosarcome/immunologie , Fibrosarcome/métabolisme , Facteurs de transcription Forkhead/métabolisme , Interleukine-10/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée BALB C , Rate/effets des médicaments et des substances chimiques , Rate/immunologie , Rate/métabolisme , Lymphocytes T régulateurs/immunologie , Facteur de croissance transformant bêta/métabolisme , Charge tumorale/immunologie
13.
Mol Pharm ; 17(11): 4152-4162, 2020 11 02.
Article de Anglais | MEDLINE | ID: mdl-33089693

RÉSUMÉ

Liver fibrosis is a common outcome of most chronic liver diseases, but there is no clinically approved drug for its treatment. Previous studies have reported the potential of SB431542 as an inhibitor of TGF-ß signaling in the treatment of liver fibrosis, but it shows poor water solubility and low bioavailability. Here, we improve these characteristics of SB431542 by loading it into liposomes (SB-Lips) with two FDA-approved excipients: soya phosphatidyl S100 and Solutol HS15. In vitro, SB-Lips had stronger inhibitory effects on the proliferation and activation of hepatic stellate cells LX-2 than free SB. After an intravenous injection in a CCl4-induced liver fibrosis mouse model, SB-Lips accumulated preferentially in the liver, its area under the concentration-time curve was significantly higher than that of free SB431542, and it alleviated hepatic fibrosis significantly more than free drug, which was associated with greater inhibition of TGF-ß signaling. Furthermore, SB-Lips did not cause significant injury to other organs. These results suggest that our liposomal system is safe and effective for delivering SB431542 to fibrotic liver.


Sujet(s)
Benzamides/administration et posologie , Benzamides/pharmacocinétique , Dioxoles/administration et posologie , Dioxoles/pharmacocinétique , Cirrhose expérimentale/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme , Animaux , Tétrachloro-méthane/effets indésirables , Lignée cellulaire , Modèles animaux de maladie humaine , Libération de médicament , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Humains , Liposomes , Cirrhose expérimentale/induit chimiquement , Mâle , Souris , Souris de lignée C57BL , Rats , Rat Sprague-Dawley , Distribution tissulaire
14.
Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi ; 37(11): 801-805, 2019 Nov 20.
Article de Chinois | MEDLINE | ID: mdl-31826541

RÉSUMÉ

Objective: To investigate the intervention effect of SB431542, which inhibits the TGF-ß/Smad3 signaling pathway, on silicotic fibrosis in rats. Methods: A total of 40 specific pathogen-free Sprague-Dawley rats were divided into normal saline control group, model group, SB431542 inhibitor group, and SB431542 inhibitor control group using a random number table, with 10 rats in each group. All rats except those in the normal saline control group were given non-exposed single intratracheal instillation of free silicon dioxide dust suspension 1 mL (50 mg/mL) ; the rats in the SB431542 inhibitor group were given intraperitoneal injection of SB431542 (5 mg/kg) on days 7 and 30 after dust exposure, those in the SB431542 inhibitor control group were given intraperitoneal injection of SB431542 cosolvent (5 mg/kg) on days 7 and 30 after dust exposure, and those in the normal saline control group were given intratracheal instillation of an equal volume of normal saline (5 mg/kg). On day 60 after dust exposure, the paraffin-embedded section of the right upper lobe of lung was collected for HE staining; the left upper lobe of lung was collected to measure the mRNA levels of fibronectin (FN) , collagen type I (COL-I) , and collagen type III (COL-III) by quantitative real-time PCR; the right inferior lobe of lung was collected to measure the protein levels of FN, COL-I, COL-III, phosphorylated Smad3 (p-Smad3) , and Smad3. Results: Compared with the normal saline control group, the model group had nodules with various sizes in lung tissue, with rupture of some alveolar septa, emphysema changes, and pulmonary interstitial fibrosis, as well as significant increases in the mRNA expression of FN, COL-I, and COL-III and the protein expression of FN, COL-I, COL-III, p-Smad3, and Smad3 in lung tissue (P<0.05) . Compared with the SB431542 inhibitor control group, the SB431542 inhibitor group had a relatively complete structure of lung tissue without marked nodules and with a small amount of exudate in alveolar space and the lumen of bronchioles, as well as significant reductions in the mRNA expression of FN, COL-I, and COL-III and the protein expression of FN, COL-I, COL-III, p-Smad3, and Smad3 in lung tissue (P<0.05) . There were no significant differences in the mRNA expression of FN, COL-I, and COL-III and the protein expression of FN, COL-I, COL-III, p-Smad3, and Smad3 between the model group and the SB431542 inhibitor control group (P>0.05) . Conclusion: SB431542 exerts an intervention effect on silicotic fibrosis by blocking the TGF-ß/Smad3 signaling pathway and reducing the expression of the downstream fibrosis factors FN, COL-I, and COL-III.


Sujet(s)
Benzamides/usage thérapeutique , Dioxoles/usage thérapeutique , Fibrose/traitement médicamenteux , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Benzamides/pharmacologie , Dioxoles/pharmacologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Transduction du signal/effets des médicaments et des substances chimiques , Protéine Smad-3/génétique , Facteur de croissance transformant bêta/génétique
15.
Onco Targets Ther ; 12: 7809-7821, 2019.
Article de Anglais | MEDLINE | ID: mdl-31576139

RÉSUMÉ

BACKGROUND: Despite progress achieved in bladder cancer (BC) treatment, the prognosis of patients with advanced BC (ie, metastasized from the bladder to other organs) is poor. Although mortality in cases of low-grade BC is rare, the treatment, such as a radical cystectomy, often has a serious impact on the quality of life. Thus, research is needed to identify more effective treatment strategies and this work is aiming to examine the potential application of combination of radiofrequency ablation (RFA) and SB435142, a inhibitor of transforming growth factor ß (TGFß)/Smad pathway. METHODS: BC cells were transplanted into nude mice (thymusdeficiency Bal B/c) to form subcutaneous tumors. The mice with subcutaneous tumors were then treated with RFA and oral administration of SB431542, an inhibitor of TGFß/Smad signaling pathway. The antitumor effect of RFA was measured by tumor proliferation curves and micro-positron emission computed tomography (micro-PET). The effect of SB431542 on epithelial-mesenchymal transition (EMT) related regulators in subcutaneous tumor tissues formed by BC cells were examined by quantitative real-time polymerase chain reaction (qPCR) experiments. RESULTS: The SB431542 treatment enhanced the antitumor effect of RFA on subcutaneous growth of BCs. SB431542 also decreased EMT-related regulators in subcutaneous tumor tissues formed by BC cells in nude mice. CONCLUSION: SB431542 enhances the effect of RFA on BC.

16.
Stem Cell Res Ther ; 10(1): 172, 2019 06 13.
Article de Anglais | MEDLINE | ID: mdl-31196174

RÉSUMÉ

BACKGROUND: Clinically, for stem cell-based therapy (SCBT), autologous stem cells are considered better than allogenic stem cells because of little immune rejection and no risk of communicable disease infection. However, severe maxillofacial bone defects restoration needs sufficient autologous stem cells, and this remains a challenge worldwide. Human gingival mesenchymal stem cells (hGMSCs) derived from clinically discarded, easily obtainable, and self-healing autologous gingival tissues, have higher proliferation rate compared with autologous bone marrow mesenchymal stem cells (hBMSCs). But for clinical bone regeneration purpose, GMSCs have inferior osteogenic differentiation capability. In this study, a TGF-ß signaling inhibitor SB431542 was used to enhance GMSCs osteogenesis in vitro and to repair minipig severe maxillofacial bone defects. METHODS: hGMSCs were isolated and cultured from clinically discarded gingival tissues. The effects of SB431542 on proliferation, apoptosis, and osteogenic differentiation of hGMSCs were analyzed in vitro, and then, SB431542-treated hGMSCs composited with Bio-Oss® were transplanted into immunocompromised mice subcutaneously to explore osteogenic differentiation in vivo. After that, SB431542-treated autologous pig GMSCs (pGMSCs) composited with Bio-Oss® were transplanted into circular confined defects (5 mm × 12 mm) in minipigs maxillary to investigate severe bone defect regeneration. Minipigs were sacrificed at 2 months and nude mice at 8 weeks to retrieve specimens for histological or micro-CT or CBCT analysis. Effects of SB431542 on TGF-ß and BMP signaling in hGMSCs were investigated by Western Blot or qRT-PCR. RESULTS: One micromolar of SB431542 treatment induced a robust osteogenesis of hGMSCs in vitro, without adverse effect on apoptosis and growth. In vivo, 1 µM SB431542 treatment also enabled striking osteogenesis of hGMSCs subcutaneously in nude mice and advanced new bone formation of pGMSCs in minipig maxillary bone defect model. In addition, SB431542-treated hGMSCs markedly increased bone-related proteins expression, and BMP2 and BMP4 gene expression. Conversely, SMAD3 protein-dependent TGF-ß signal pathway phosphorylation was decreased. CONCLUSIONS: Our study show that osteogenic differentiation of GMSCs treated with TGF-ß signaling inhibitor SB431542 was increased, and SB431542-treated autologous pig GMSCs could successfully repair minipig severe maxillofacial bone defects. This preclinical study brings about a promising large bone regeneration therapeutic potential of autologous GMSCs induced by SB431542 in clinic settings.


Sujet(s)
Gencive/cytologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme , Animaux , Benzamides/pharmacologie , Protéine morphogénétique osseuse de type 2/génétique , Protéine morphogénétique osseuse de type 2/métabolisme , Protéine morphogénétique osseuse de type 4/génétique , Protéine morphogénétique osseuse de type 4/métabolisme , Régénération osseuse/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Dioxoles/pharmacologie , Humains , Mâle , Cellules souches mésenchymateuses/métabolisme , Souris , Souris de lignée BALB C , Souris nude , Ostéogenèse/effets des médicaments et des substances chimiques , Ostéogenèse/génétique , Protéine Smad-3/génétique , Protéine Smad-3/métabolisme , Suidae , Facteur de croissance transformant bêta/antagonistes et inhibiteurs
17.
Biol Reprod ; 101(1): 63-75, 2019 07 01.
Article de Anglais | MEDLINE | ID: mdl-31004472

RÉSUMÉ

Growth differentiation factor 8 (GDF8), also known as myostatin, is a member of the transforming growth factor-ß (TGF-ß) family and has been identified as a strong physiological regulator of muscle differentiation. Recently, the functional role of GDF8 in reproductive organs has received increased interest following its detection in the human placenta and uterus. To investigate the effects of GDF8 during porcine oocyte in vitro maturation (IVM), we assessed the quality of matured oocytes. Furthermore, we investigated the specific gene transcription and protein activation levels in oocytes and cumulus cells after IVM and subsequent embryonic development after in vitro fertilization and parthenogenetic activation. Prior to these experiments, the concentration of GDF8 in porcine follicular fluid was determined. During the entire IVM period, 1.3 ng/mL GDF8 and its signaling inhibitor SB431542 (SB) at 5 µM were added as control, SB, SB + GDF8, and GDF8 groups, respectively. Our results demonstrate that supplementation with GDF8 during porcine oocyte IVM enhanced both meiotic and cytoplasmic maturation, with altered transcriptional patterns, via activation of Sma- and Mad-related protein 2/3 (SMAD2/3). Using the pharmacological inhibitor SB431542, we demonstrated that inhibition of GDF8-induced Smad2/3 signaling reduces matured oocyte quality. In conclusion, for the first time, we demonstrated paracrine factor GDF8 in porcine follicular fluid in vivo. Furthermore, we showed that GDF8 supplementation improved mature oocyte quality by regulating p38 mitogen-activated protein kinase phosphorylation and intracellular glutathione and reactive oxygen species levels during porcine IVM.


Sujet(s)
Techniques de maturation in vitro des ovocytes , Myostatine/pharmacologie , Ovocytes/cytologie , Ovocytes/effets des médicaments et des substances chimiques , Protéine Smad2/métabolisme , Protéine Smad-3/métabolisme , Animaux , Benzamides/pharmacologie , Cellules cultivées , Dioxoles/pharmacologie , Développement embryonnaire/effets des médicaments et des substances chimiques , Développement embryonnaire/physiologie , Femelle , Fécondation in vitro/normes , Fécondation in vitro/médecine vétérinaire , Techniques de maturation in vitro des ovocytes/médecine vétérinaire , Ovocytes/physiologie , Ovogenèse/effets des médicaments et des substances chimiques , Contrôle de qualité , Transduction du signal/effets des médicaments et des substances chimiques , Protéine Smad2/génétique , Protéine Smad-3/génétique , Suidae
18.
Theranostics ; 9(5): 1387-1400, 2019.
Article de Anglais | MEDLINE | ID: mdl-30867839

RÉSUMÉ

Human CLCN7 encodes voltage-gated chloride channel 7 (ClC-7); mutations of CLCN7 lead to osteopetrosis which is characterized by increased bone mass and impaired osteoclast function. In our previous clinical practice, we noticed that osteopetrosis patients with CLCN7 mutations had some special deformities in craniofacial morphology and tooth dysplasia. It is unclear whether these phenotypes are the typical features of CLCN7 involved osteopetrosis and whether ClC-7 could regulate the development of craniofacial bone and tooth in some signaling pathways. Methods: First, we collected 80 osteopetrosis cases from the literature and compared their craniofacial and dental phenotypes. Second, four osteopetrosis pedigrees with CLCN7 mutations were recruited from our clinic for gene testing and clinical analysis of their craniofacial and dental phenotypes. Third, we used a zebrafish model with clcn7 morpholino treatment to detect the effects of ClC-7 deficiency on the development of craniofacial and dental phenotypes. General observation, whole mount alcian blue and alizarin red staining, whole mount in situ hybridization, scanning electron microscope observation, lysoSensor staining, Q-PCR and western blotting were performed to observe the in vivo characteristics of craniofacial bone and tooth changes. Fourth, mouse marrow stromal cells were further primarily cultured to detect ClC-7 related mRNA and protein changes using siRNA, Q-PCR and western blotting. Results: Over 84% of osteopetrosis patients in the literature had some typical craniofacial and tooth phenotypes, including macrocephaly, frontal bossing, and changes in shape and proportions of facial skeleton, and these unique features are more severe and frequent in autosomal recessive osteopetrosis than in autosomal dominant osteopetrosis patients. Our four pedigrees with CLCN7 mutations confirmed the aforementioned clinical features. clcn7 knockdown in zebrafish reproduced the craniofacial cartilage defects and various dental malformations combined the decreased levels of col10a1, sp7, dlx2b, eve1, and cx43. Loss of clcn7 function resulted in lysosomal storage in the brain and jaw as well as downregulated cathepsin K (CTSK). The craniofacial phenotype severity also presented a dose-dependent relationship with the levels of ClC-7 and CTSK. ClC-7/CTSK further altered the balance of TGF-ß/BMP signaling pathway, causing elevated TGF-ß-like Smad2 signals and reduced BMP-like Smad1/5/8 signals in clcn7 morphants. SB431542 inhibitor of TGF-ß pathway partially rescued the aforementioned craniofacial bone and tooth defects of clcn7 morphants. The ClC-7 involved CTSK/BMP and SMAD changes were also confirmed in mouse bone marrow stromal cells. Conclusion: These findings highlighted the vital role of clcn7 in zebrafish craniofacial bone and tooth development and mineralization, revealing novel insights for the causation of osteopetrosis with CLCN7 mutations. The mechanism chain of ClC-7/CTSK/ TGF-ß/BMP/SMAD might explain the typical craniofacial bone and tooth changes in osteopetrosis as well as pycnodysostosis patients.


Sujet(s)
Canaux chlorure/métabolisme , Os de la face/embryologie , Protéines mutantes/métabolisme , Ostéopétrose/physiopathologie , Crâne/embryologie , Dent/embryologie , Animaux , Canaux chlorure/génétique , Modèles animaux de maladie humaine , Humains , Souris , Modèles théoriques , Protéines mutantes/génétique , Ostéopétrose/anatomopathologie , Danio zébré/embryologie
19.
J Reprod Dev ; 65(3): 215-221, 2019 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-30760649

RÉSUMÉ

All-trans retinoic acid (RA) is a metabolite of vitamin A and has pleiotropic actions on many different biological processes, including cell growth and differentiation, and is involved in different aspects of fertility and developmental biology. In the current study, we investigated the effects of RA on camel (Camelus dromedarius) cumulus-oocyte complex in vitro maturation (IVM). IVM medium was supplemented with 0, 10, 20, and 40 µM RA. Application of 20 µM RA significantly reduced the proportion of degenerated oocytes and significantly improved oocyte meiosis and first polar body extrusion compared to the control and other experimental groups. Retinoic acid significantly reduced the mRNA transcript levels of apoptosis-related genes, including BAX and P53, and reduced the BAX/BCL2 ratio. In addition, RA significantly reduced the expression of the Transforming growth factor beta (TGFß) pathway-related transcripts associated with the actin cytoskeleton, ACTA2 and TAGLN; however, RA increased TGFß expression in cumulus cells. The small molecule SB-431542 inhibits the TGFß pathway by inhibiting the activity of activin receptor-like kinases (ALK-4, ALK-5, and ALK-7); however, combined supplementation with RA during IVM compensated for the inhibitory effect of SB-431542 on cumulus expansion, oocyte meiosis I, and first polar body extrusion in activated oocytes. The current study shows the beneficial effects of RA on camel oocyte IVM and provides a model to study the multifunctional mechanisms involved in cumulus expansion and oocyte meiosis, particularly those involved in the TGFß pathway.


Sujet(s)
Cellules du cumulus/cytologie , Techniques de maturation in vitro des ovocytes , Ovocytes/cytologie , Trétinoïne/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Benzamides/pharmacologie , Blastocyste/cytologie , Chameaux , Milieux de culture , Cellules du cumulus/effets des médicaments et des substances chimiques , Dioxoles/pharmacologie , Techniques de culture d'embryons , Femelle , Fécondité , Techniques de transfert nucléaire , Ovocytes/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme
20.
Int J Clin Exp Pathol ; 12(5): 1835-1845, 2019.
Article de Anglais | MEDLINE | ID: mdl-31934007

RÉSUMÉ

We explored whether transforming growth factor (TGF-ß)/Smads signaling pathway influences rheumatoid arthritis (RA)-associated pulmonary fibrosis (PF) and proliferation of RA synovial fibroblast (RA-SF). Expression levels of TGF-ß1 in RA + PF patients, RA patients and healthy controls were determined. Rat models of RA were successfully induced and assigned into groups. The mRNA, phosphorylation and protein expressions of TGF-ß1, Smad2 and Smad3 were detected. The serum expressions of inflammatory factors were measured by ELISA. Tissue sections were observed using hematoxylin-eosin (HE) and Masson staining. The SF cells were separated and grouped. Cell viability and migration were determined. The highest expressions of TGF-ß1 were found in RA + PF patients, followed by RA patients and then healthy controls. RA + PF rats were characterized by less activity, worse appetite, messy and less shining hair, thin sloppy stool and increased joint swelling. Compared with the normal group, the expressions of TGF-ß1, Smad2, Smad3, IL-6 and TNF-α were elevated in the RA + PF group. Meanwhile, the swelling and pulmonary fibrosis of lung tissues was worse, the lung capacity and serum level of IL-10 were decreased. However, SB431542 can reverse the above results. The cell activity and cell migration ability of cells in the RA + PF + SB431542 group were inhibited compared to those in the blank group. Based on above findings, the inhibition of the TGF-ß/Smads signaling pathway alleviates the pulmonary fibrosis in rats with RA and suppresses cell viability and migration of synovial fibroblasts.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE