Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Hepatobiliary Pancreat Dis Int ; 14(2): 215-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25865696

ABSTRACT

BACKGROUND: Pancreatic stellate cells (PSCs) play a critical role in the development of pancreatic fibrosis. In this study we used a novel method to isolate and culture rat PSCs and then investigated the inhibitory effects of adipose-derived stem cells (ADSCs) on activation and proliferation of PSCs. METHODS: Pancreatic tissue was obtained from Sprague-Dawley rats for PSCs isolation. Transwell cell cultures were adopted for co-culture of ADSCs and PSCs. PSCs proliferation and apoptosis were determined using CCK-8 and flow cytometry, respectively. alpha-SMA expressions were analyzed using Western blotting. The levels of cytokines [nerve growth factor (NGF), interleukin-10 (IL-10) and transforming growth factor-beta1 (TGF-beta1)] in conditioned medium were detected by ELISA. Gene expression (MMP-2, MMP-9 and TIMP-1) was analyzed using qRT-PCR. RESULTS: This method produced 17.6+/-6.5X10(3) cells per gram of the body weight with a purity of 90%-95% and a viability of 92%-97%. Co-culture of PSCs with ADSCs significantly inhibited PSCs proliferation and induced PSCs apoptosis. Moreover, alpha-SMA expression was significantly reduced in PSCs+ADSCs compared with that in PSC-only cultures, while expression of fibrinolytic proteins (e.g., MMP-2 and MMP-9) was up-regulated and anti-fibrinolytic protein (TIMP-1) was down-regulated. In addition, NGF expression was up-regulated, but IL-10 and TGF-beta1 expressions were down-regulated in the co-culture conditioned medium compared with those in the PSC-only culture medium. CONCLUSIONS: This study provided an easy and reliable technique to isolate PSCs. The data demonstrated the inhibitory effects of ADSCs on the activation and proliferation of PSCs in vitro.


Subject(s)
Adipose Tissue/cytology , Apoptosis , Cell Proliferation , Pancreatic Stellate Cells/physiology , Stem Cells , Actins/metabolism , Animals , Coculture Techniques , Culture Media, Conditioned , Down-Regulation/genetics , Gene Expression , Interleukin-10/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Nerve Growth Factor/metabolism , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-1/genetics , Transforming Growth Factor beta1/metabolism , Up-Regulation/genetics
2.
J Formos Med Assoc ; 114(2): 130-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25678175

ABSTRACT

BACKGROUND/PURPOSE: Previous studies suggested that mesenchymal stem cells may ameliorate fibrogenesis through the inhibition of hepatic stellate cells (HSCs) activation. This study aimed to investigate whether adipose derived mesenchymal stem cells (ADSCs) could modulate the activation of HSCs and contribute to the recovery of liver fibrogenesis. METHODS: ADSCs and HSCs were isolated from Sprague-Dawley rats and co-cultured using a transwells insert. Cell proliferation, apoptosis and smooth muscle α-actin (α-SMA) expression in HSCs were examined. Rats were injected with CCl4 to induce liver fibrogenesis. After injection of ADSCs through portal vein, the rats were examined for pathological changes in the liver. α-SMA expression and hydroxyproline content in the liver and serum levels of collagen III and hyaluronic acid was detected. RESULTS: After co-culturing for 72 h, the proliferation and activation of HSCs was inhibited by ADSCs and the apoptosis of HSCs was promoted by ADSCs. Transplantation of ADSCs inhibited liver fibrogenesis in the rats. CONCLUSION: ADSCs inhibit the proliferation and activation of HSCs in vitro and inhibit liver fibrogenesis in rat model, suggesting the potential application of ADSCs in liver fibrogenesis therapy.


Subject(s)
Hepatic Stellate Cells/cytology , Liver Cirrhosis/therapy , Liver/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Actins/metabolism , Adipose Tissue/cytology , Animals , Apoptosis , Carbon Tetrachloride , Cell Proliferation , Cells, Cultured , Liver Cirrhosis/chemically induced , Male , Rats , Rats, Sprague-Dawley
3.
Am J Med Sci ; 346(5): 353-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23276892

ABSTRACT

BACKGROUND: Extensive controversies exist over the use of preoperative biliary drainage preceding radical resection of hilar cholangiocarcinoma. This study assessed the effectiveness and safety of percutaneous transhepatic cholangiodrainage (PTCD) with bile re-infusion in the preoperative optimization of hilar cholangiocarcinoma patients. METHODS: Eligible hilar cholangiocarcinoma patients received preoperative PTCD with bile re-infusion (treatment group, n = 56) through a nasoduodenal tube for 2 weeks, and the control group (n = 60) received conservative treatment alone. Operable patients were assigned to undergo either a radical or palliative resection. The outcome measures included the overall resection rate, R0 resection rate, surgical morbidity rate and 1-year and 5-year overall survival rates. RESULTS: The treatment group exhibited a significant decrease in serum bilirubin levels after PTCD with bile re-infusion. The overall resection rate was significantly higher in the treatment group than in the control group (85.5% vs. 65.0%, P < 0.05), and the palliative resection rate was also significantly higher in the treatment group (53.5% vs. 35.0%, P < 0.05). However, the R0 resection rate was comparable between the 2 groups (32.1% vs. 30.0%, P > 0.05). The morbidity rate was significantly lower in the treatment group than in the control group (29.1% vs. 51.3%, P < 0.05). One-year and 5-year survival rates were similar between the 2 groups (69.6% vs. 66.7%, P > 0.05; 5.3% vs. 3.6%, P > 0.05). CONCLUSIONS: Preoperative PTCD with bile re-infusion improves the resection rate and shows a good safety profile in patients with hilar cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms/surgery , Bile Ducts, Intrahepatic , Bile , Cholangiocarcinoma/surgery , Drainage/methods , Preoperative Care/methods , Bile Duct Neoplasms/mortality , Cholangiocarcinoma/mortality , Digestive System Surgical Procedures , Drainage/adverse effects , Female , Humans , Male , Middle Aged , Palliative Care , Preoperative Care/adverse effects , Retrospective Studies , Risk Factors , Survival Rate , Time Factors , Treatment Outcome
4.
Mol Cell Biochem ; 369(1-2): 35-43, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22752386

ABSTRACT

In order to explore the effects of fat-specific protein 27 (Fsp27) on regulation of hepatic stellate cell (HSC) activation and liver fibrosis. HSCs were isolated from rat liver tissues and cultivated in vitro for gene expression and lentivirus infection. CCK-8 cell viability assay, immunofluorescence staining, qRT-PCR, and western blot assays were used to assess phenotypic changes and gene expression in HSCs. The rat liver fibrosis model was produced by intraperitoneal injection of carbon tetrachloride for assessing the effects of Fsp27 in the rat liver. Gene expression was then detected by immunohistochemistry and ELISA assays. The results of the study showed that Fsp27 was constitutively expressed in primary quiescent HSCs, but was absent in activated HSCs. Ectopic expression of Fsp27 significantly inhibited HSC proliferation and activation, as well as expression of α-smooth muscle actin. Fsp27 expression also significantly reduced collagen I production and matrix metalloproteinases 2 protein levels, and to a lesser degree, reduced tissue inhibitors of metalloproteinases 1 expression. In vivo data showed that ectopic expression of Fsp27 protein significantly reduced levels of hydroxyproline in liver tissue, and decreased serum levels of collagen III and hyaluronic acid, which in turn, suppressed liver fibrosis in rats. From these findings, it can be concluded that Fsp27 expression suppressed HSC activation in vitro and liver fibrogenesis in vivo. Further studies are needed to explore whether expression of Fsp27 can be selected as a potential novel strategy for anti-fibrotic therapy against liver fibrosis.


Subject(s)
Genetic Therapy , Hepatic Stellate Cells/metabolism , Liver Cirrhosis , Proteins/genetics , Proteins/metabolism , Animals , Carbon Tetrachloride/toxicity , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Collagen Type I/metabolism , Gene Expression Regulation , Hepatic Stellate Cells/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/therapy , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...