Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(6): 609-615, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-38991960

RESUMEN

OBJECTIVE: To explore whether sivelestat sodium could reduce the expression of mucin 5AC (MUC5AC) in intrahepatic bile duct epithelial cells by inhibiting neutrophil elastase (NE) and thus provide new potential therapeutic ideas for the treatment of intrahepatic bile duct stone (IBDS). METHODS: (1) Bioinformatics analysis: differential gene analysis was performed on gallbladder stone cholecystitis sequencing data based on the gene expression omnibus (GEO) to screen for significantly different genes related to neutrophils and mucins. The search tool for the retrieval of interacting genes database (STRING) was used for protein interaction analysis to predict whether there was an interaction between NE and MUC5AC genes. (2) Animal experiment: a total of 18 male SD rats were divided into the sham-operated group, cholangitis model group and sivelestat sodium treatment group according to the random number table method, with 6 rats in each group. The cholangitis rat model was established by a one-time injection of 1.25 mg/kg lipopolysaccharide (LPS) into the right anterior lobe of the liver of rats in combination with the pre-experiment; the liver of the sham-operated group was injected with an equal volume of saline. After the modelling, 100 mg/kg of sivelestat sodium was injected into the tail vein of the cevalexin treatment group once a day for 5 days, and an equal volume of saline was injected into the tail vein of the sham-operated group and the cholangitis model group. Two weeks later, the rats were euthanized and their liver and bile duct tissues were taken. The pathological changes in the liver and bile duct tissues were observed under the light microscope. Immunohistochemical staining was used to detect the expressions of NE and MUC5AC in liver and bile duct tissues. The protein expressions of NE, MUC5AC and Toll-like receptor 4 (TLR4) were detected by Western blotting. (3) Cell experiment: primary human intrahepatic biliary epithelial cell line (HiBEpiC) was divided into blank control group, NE group (10 nmol/L NE), NE+sivelestat sodium low dose group (10 nmol/L NE+1×10-8 g/L sivelestat sodium 1 mL), NE+sivelestat sodium medium dose group (10 nmol/L NE+1×10-7 g/L sivelestat sodium 1 mL), NE+sivelestat sodium high dose group (10 nmol/L NE+1×10-6 g/L sivelestat sodium 1 mL). Cells were collected after 48 hours of culture, and EdU was performed to detect the proliferative activity of cells; enzyme linked immunosorbent assay (ELISA) and Western blotting were performed to detect the expression of MUC5AC in cells. RESULTS: (1) Bioinformatics analysis: the NE gene (ELANE) had a reciprocal relationship with MUC5AC. (2) Animal experiment: light microscopy showed that hepatocyte edema, hepatocyte diffuse point and focal necrosis, confluent area fibrous tissue and intrahepatic bile ducts hyperplasia and inflammatory cell infiltration in the cholangitis model group; hepatic lobule structure of sivelestat sodium treatment group was clear, and the degree of peripheral inflammatory cell infiltration was reduced compared with the cholangitis model group. Immunohistochemical staining showed that the expressions of NE and MUC5AC were increased in the cholangitis model group compared with the sham-operated group, and the expressions of NE and MUC5AC were decreased in the sivelestat sodium group compared with the cholangitis model group [NE (A value): 5.23±2.02 vs. 116.67±23.06, MUC5AC (A value): 5.40±3.09 vs. 23.81±7.09, both P < 0.05]. Western blotting showed that the protein expressions of NE, MUC5AC, and TLR4 in the hepatic biliary tissues of the cholangitis model group were significantly higher than those of the sham-operated group; and the protein expressions of NE, MUC5AC, and TLR4 in the liver biliary tissues of the sivelestat sodium treatment group were significantly higher than those of the sham-operated group (NE/ß-actin: 0.38±0.04 vs. 0.70±0.10, MUC5AC/ß-actin: 0.37±0.03 vs. 0.61±0.05, TLR4/ß-actin: 0.39±0.10 vs. 0.93±0.15, all P < 0.05). (3) Cell experiment: fluorescence microscopy showed that the proliferation of HiBEpiC cells in each group was good, and there was no significant difference in the proportion of positive cells. ELISA and Western blotting showed that the expressions of MUC5AC in cells of the NE group were significantly higher than those of the blank control group. The expressions of MUC5AC in the NE+different dose of sivelestat sodium group were significantly lower than those in the NE group, and showed a decreasing trend with the increase of sevastatin sodium concentration, especially in the highest dose group [MUC5AC (µg/L): 3.46±0.20 vs. 6.33±0.52, MUC5AC/ß-actin: 0.45±0.07 vs. 1.75±0.10, both P < 0.05]. CONCLUSIONS: LPS can upregulate the expression of NE and MUC5AC in rats with cholangitis, while sodium sivelestat can reduce the expression of MUC5AC in in intrahepatic biliary epithelial cells by inhibiting NE, providing a new direction for the treatment of IBDS.


Asunto(s)
Conductos Biliares Intrahepáticos , Glicina , Elastasa de Leucocito , Mucina 5AC , Ratas Sprague-Dawley , Sulfonamidas , Animales , Mucina 5AC/metabolismo , Masculino , Ratas , Elastasa de Leucocito/metabolismo , Glicina/análogos & derivados , Glicina/farmacología , Sulfonamidas/farmacología , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos
2.
Mol Biol Rep ; 51(1): 176, 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38252208

RESUMEN

BACKGROUND: Pancreatic cancer (PC) is a fatal human malignancy with a poor prognosis. Corosolic acid (CRA) is a triterpenoid, has been reported to have inhibitory effects on tumor growth. However, the role of CRA on PC has not been explored. Here, we aimed to uncover the molecular mechanisms of CRA in PC progression. METHODS: Cell viability, lactate dehydrogenase (LDH) release, cell apoptosis and senescence were detected by cell counting kit-8 (CCK-8), LDH, flow cytometry and senescence associated-ß-galactosidase (SA-ß-gal) assay. Levels of relevant proteins and oxidative stress (OS) markers were evaluated by Western blot and enzyme-linked immunosorbent assay (ELISA). A xenograft tumor model was established to explore the in vivo effects of CRA on PC. RESULTS: We found that CRA inhibited PC cell viability and promoted LDH release in a dose-dependent manner, but had no significant effect on human normal pancreatic ductal epithelial cells HPDE6C7. CRA increased OS-induced cell apoptosis and senescence in HAPC and SW1990 cells. And CRA decreased the levels of anti-apoptotic protein Bcl-2, and elevated the expression of pro-apoptotic protein Bax and senescence-associated proteins P21 and P53. Besides, CRA decreased tumor growth in xenograft models. Furthermore, CRA inactivated the Janus kinase-2 (JAK2)/Signal Transducer and Activator of Transcription 3 (STAT3) signaling pathway in HAPC and SW1990 cells. Functional experiments demonstrated that activation of the JAK2/STAT3 pathway by the JAK2 activator coumermycin A1 (C-A1) or the STAT3 activator colivelin (col) reduced the contribution effect of OS, apoptosis and senescence by CRA. CONCLUSION: Taken together, our findings indicated that CRA exerted anti-cancer effects in PC by inhibiting the JAK2/STAT3 pathway.


Asunto(s)
Neoplasias Pancreáticas , Triterpenos , Humanos , Factor de Transcripción STAT3 , Neoplasias Pancreáticas/tratamiento farmacológico , Estrés Oxidativo , Triterpenos/farmacología , Apoptosis , Janus Quinasa 2
3.
In Vitro Cell Dev Biol Anim ; 58(10): 855-866, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36481977

RESUMEN

Cholesterol overloading stress damages normal cellular functions in hepatocytes and induces metabolic disorders to facilitate the development of multiple diseases, including cardiovascular diseases, which seriously degrades the life quality of human beings. Recent data suggest that the Berberis vulgaris L. extract berberine is capable of regulating cholesterol homeostasis, which is deemed as potential therapeutic drug for the treatment of cholesterol overloading-associated diseases, but its detailed functions and molecular mechanisms are still largely unknown. In the present study, we evidenced that berberine suppressed cell apoptosis in high-cholesterol-diet mice liver and cholesterol-overloaded mice hepatocytes. Also, cholesterol overloading promoted reactive oxygen species (ROS) generation to trigger oxidative damages in hepatocytes, which were reversed by co-treating cells with both berberine and the ROS scavenger N-acetylcysteine (NAC). Moreover, the underlying mechanisms were uncovered, and we validated that berberine downregulated Keap1, and upregulated Nrf2 to activate the anti-oxidant Nrf2/HO-1 signaling pathway in cholesterol overloading-treated hepatocytes, and both Keap1 upregulation and Nrf2 downregulation abrogated the suppressing effects of berberine on cell apoptosis in the hepatocytes with cholesterol exposure. Taken together, we concluded that berberine activated the anti-oxidant Keap1/Nrf2/HO-1 pathway to eliminate cholesterol overloading-induced oxidative stress and apoptotic cell death in mice hepatocytes, and those evidences hinted that berberine might be used as putative therapeutic drug for the treatment of cholesterol overloading-associated cardiovascular diseases.


Asunto(s)
Antioxidantes , Apoptosis , Berberina , Berberis , Enfermedades de los Roedores , Animales , Ratones , Antioxidantes/farmacología , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Berberina/farmacología , Berberina/uso terapéutico , Berberis/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Colesterol/metabolismo , Colesterol/farmacología , Hepatocitos/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Enfermedades de los Roedores/tratamiento farmacológico
4.
J BUON ; 25(5): 2449-2455, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33277869

RESUMEN

PURPOSE: To explore the effects of aspirin on the proliferation and apoptosis of human pancreatic cancer cells and its potential molecular mechanism. METHODS: This study included patients with pancreatic cancer who were divided into experimental group and control group. The cell proliferation ability was detected via cell counting kit-8 (CCK-8) assay and colony forming ability via colony formation assay. In addition, changes in proteins in the phosphatidyl inositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway were assessed using Western blotting, and rescue experiment was conducted to investigate whether aspirin can affect cell proliferation by inhibiting the PI3K/Akt/mTOR signaling pathway. RESULTS: The results of CCK-8 assay showed that the proliferation rate of PANC-1 cells was decreased in a time- and dose-dependent manner after they were treated with aspirin at different concentrations. Colony formation assay confirmed that cell colony forming ability was significantly reduced with the increase in aspirin treatment concentration (p<0.05). Besides, the apoptosis rate and the number of cells in the experimental group were higher and larger than those in the control group (p<0.05). According to Western blotting results, the protein expressions of PI3K, phosphorylated (p)-Akt and p-mTOR were decreased after aspirin treatment. Rescue experimental results manifested that insulin-like growth factor 1 (IGF-1) supplementation remarkably elevated the expressions of PI3K, p-Akt and p-mTOR compared with phosphate-buffered saline (PBS) supplementation. It was found in CCK-8 assay that IGF-1 supplementation markedly reversed the inhibition of aspirin on the proliferation of PANC-1 cells in comparison with PBS supplementation. CONCLUSIONS: Aspirin inhibits the proliferation and promotes the apoptosis of pancreatic cancer cells by inactivating the PI3K/Akt/mTOR signaling pathway.


Asunto(s)
Aspirina/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Apoptosis , Aspirina/farmacología , Proliferación Celular , Humanos
5.
Sci Rep ; 7(1): 581, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28373667

RESUMEN

Acute pancreatitis (AP) is a common acute abdominal disease, 10-20% of which can evolve into severe AP (SAP) causing significant morbidity and mortality. Bone marrow-derived mesenchymal stem cells (BMSCs) have the potential of repairing SAP, but the detailed mechanism remains unknown. We demonstrate here that microRNA-9 (miR-9) modified BMSCs (pri-miR-9-BMSCs) can significantly reduce the pancreatic edema, infiltration, hemorrhage, necrosis, the release of amylase and lipase. Meanwhile, decreased local/systemic inflammatory response (TNF-α↓, IL-1ß↓, IL-6↓, HMGB1↓, MPO↓, CD68↓, IL-4↑, IL-10↑, and TGF-ß↑) and enhanced regeneration of damaged pancreas (Reg4↑, PTF1↑, and PDX1↑) are also promoted. But these effects diminish or disappear after antagonizing miR-9 (TuD). Besides, we find that miR-9 is negatively correlated with AP and miR-9 agomir which can mimic the effects of pri-miR-9-BMSCs and protect injured pancreas. Furthermore, we investigate that BMSCs deliver miR-9 to the injured pancreas or peripheral blood mononuclear cell (PBMC), which can target the NF-κB1/p50 gene and inhibit the NF-κB signaling pathway (p-P65↓, NF-κB1/p50↓, IκBα↑, IκBß↑). Taken together, these results show that miR-9 is a key paracrine factor of BMSCs attenuating SAP targeting the NF-κB1/p50 gene and suppressing the NF-κB signaling pathway.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Subunidad p50 de NF-kappa B/genética , Pancreatitis Aguda Necrotizante/etiología , Animales , Apoptosis/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Subunidad p50 de NF-kappa B/metabolismo , Pancreatitis Aguda Necrotizante/metabolismo , Pancreatitis Aguda Necrotizante/patología , Pancreatitis Aguda Necrotizante/terapia , Interferencia de ARN , Ratas , Transducción de Señal , Factores de Tiempo , Transducción Genética
6.
Sci Rep ; 6: 38438, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27917949

RESUMEN

The administration of mesenchymal stem cells/multipotent mesenchymal stromal cells (MSCs) to enhance tissue repair is currently undergoing clinical trials. Some studies, including our previous work, have also revealed the beneficial effect of MSCs in severe acute pancreatitis (SAP); however, their mechanisms or mode of action remain controversial. In this study, we demonstrated that intravenously (i.v.)-administered human MSCs (hMSCs) remarkably promoted recovery from experimental SAP without significant engraftment of hMSCs in the damaged pancreas. Interestingly, we found that i.v.-administered hMSCs with knockdown of TSG-6 expression lost most of their anti-inflammatory effects and thus could not significantly ameliorate SAP. As expected, the effects of hMSCs were also duplicated by i.v. infusion of recombinant TSG-6. Furthermore, our results showed that the increase of oxidative stress, activation of the NLRP3 inflammasome and NF-κB signaling in SAP was substantially inhibited following administration of hMSCs or TSG-6, which was dependent on the presence of CD-44 receptors in acinar cells. In conclusion, our study, for the first time, revealed that novel mechanisms are responsible for the immunomodulatory effect of i.v. hMSCs.


Asunto(s)
Moléculas de Adhesión Celular/genética , Inmunomodulación/genética , Trasplante de Células Madre Mesenquimatosas , Pancreatitis/terapia , Administración Intravenosa , Animales , Moléculas de Adhesión Celular/inmunología , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/inmunología , Inflamasomas/administración & dosificación , Inflamasomas/genética , Ratones , FN-kappa B/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Estrés Oxidativo/efectos de los fármacos , Pancreatitis/inmunología , Pancreatitis/patología , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/genética
7.
Int J Clin Exp Med ; 8(8): 13824-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26550332

RESUMEN

BACKGROUND: Pain is the most common complaint of patients on the first day after laparoscopic cholecystectomy (LC). The aim of this study was to compare the efficacy of local anesthesia with bupivacaine and intravenous parecoxib on postoperative abdominal pain relief up to 24 h after surgery. METHODS: One hundred and eighty patients who underwent LC were randomized to one of three groups with sixty patients each: Group A received 50 mg 0.5% bupivacaine subcutaneously at trocar sites before incision closure; Group B received intravenous parecoxib (40 mg) after entering the recovery room; Group C did not receive postoperative analgesia unless needed and was served as control. The postoperative pain at 1, 2, 4, 8, 12, and 24 h after the operation was assessed using a visual analog scale (VAS). Secondary outcomes, including intraoperative and postoperative complications, the incidence of shoulder pain, pethidine requirements, postoperative nausea and vomiting, and hospital stay were also recorded. RESULTS: At 1, 2, and 4 hours after surgery, VAS pain scores were significantly lower in group A and B compared with group C (P < 0.05 for all). There was no significant difference among the three groups at 8, 12, and 24 hours after the procedure (P > 0.05 for all). A repeated-measures ANOVA analysis revealed that VAS pain scores over the first 24 hours after LC were significantly lower in group A and B compared with group C (P = 0.014 and P = 0.029 for between-group comparison, respectively). Furthermore, the percentage of patients requiring postoperative rescue analgesics was significantly higher in group C as compared with group A and group B (P = 0.018). CONCLUSION: Local anesthesia with bupivacaine and intravenous parecoxib are both effective at decreasing postoperative pain and pethidine requirements after LC.

8.
Stem Cells Int ; 2015: 306836, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25810724

RESUMEN

Acute pancreatitis (AP), a common acute abdominal disease, 10%-20% of which can evolve into severe acute pancreatitis (SAP), is of significant morbidity and mortality. Bone marrow-derived mesenchymal stem cells (BMSCs) have been reported to have a potential therapeutic role on SAP, but the specific mechanism is unclear. Therefore, we conducted this experiment to shed light on the probable mechanism. We validated that SDF-1α significantly stimulated the expressions of VEGF, ANG-1, HGF, TGF-ß, and CXCR4 in BMSCs, which were inhibited by its receptor agonist, AMD3100. The capacities of proliferation, migration, and repair of human umbilical vein endothelial cells were enhanced by BMSCs supernatant. Meanwhile, BMSCs supernatant could also promote angiogenesis, especially after the stimulation with SDF-1α. In vivo, the migration of BMSCs was regulated by SDF-1α/CXCR4 axis. Moreover, transplanted BMSCs could significantly alleviate SAP, reduce the systematic inflammation (TNF-α↓, IL-1ß↓, IL-6↓, IL-4↑, IL-10↑, and TGF-ß↑), and promote tissue repair and angiogenesis (VEGF↑, ANG-1↑, HGF↑, TGF-ß↑, and CD31↑), compared with the SAP and anti-CXCR4 groups. Taken together, the results showed that BMSCs ameliorated SAP and the SDF-1α/CXCR4 axis was involved in the repair and regeneration process.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...