Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Language
Publication year range
1.
Genet Mol Res ; 14(2): 5793-803, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26125778

ABSTRACT

We observed the influence of different concentrations of Rhizoma paridis total saponins (RPTS) on the apoptosis of colorectal cancer cells and explored the internal mechanism involved. We determined whether RPTS influences the interleukin-6 (IL-6)/Janus kinase (JAK)-signal transducer and activator of transcription-3 (STAT3) apoptosis molecular pathway and looked for colon cancer-related signal transduction pathways or targets inducing apoptosis. We also cultured SW480 colorectal cancer cells using different concentrations of RPTS (10, 20, 40, and 80 µg/ mL), and observed the effect of RPTS on SW480 cell morphology under a fluorescence inverted microscope. We detected serum IL-6 using the polymerase chain reaction and the expression of JAK-STAT3 protein by western blot. After treating SW480 with RPTS and Hoechst 33258 dyeing, we found that the typical apoptosis morphology had changed. Secretion of IL-6 in the serum decreased significantly (P < 0.05), and STAT3 levels were reduced. RPTS can significantly promote apoptosis in SW480 colorectal cancer cells. The mechanism may be that it suppresses the secretion of IL-6 and inhibits the IL-6/JAK-STAT3 protein signaling pathway.


Subject(s)
Colorectal Neoplasms/drug therapy , Interleukin-6/biosynthesis , Janus Kinases/biosynthesis , Saponins/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/genetics , Janus Kinases/genetics , Phosphorylation , Plant Extracts/administration & dosage , Plant Extracts/chemistry , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Saponins/chemistry , Signal Transduction/drug effects
2.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;48(5): 401-407, 05/2015. graf
Article in English | LILACS | ID: lil-744380

ABSTRACT

Recent studies have revealed that an intrinsic apoptotic signaling cascade is involved in vascular hyperpermeability and endothelial barrier dysfunction. Propofol (2,6-diisopropylphenol) has also been reported to inhibit apoptotic signaling by regulating mitochondrial permeability transition pore (mPTP) opening and caspase-3 activation. Here, we investigated whether propofol could alleviate burn serum-induced endothelial hyperpermeability through the inhibition of the intrinsic apoptotic signaling cascade. Rat lung microvascular endothelial cells (RLMVECs) were pretreated with propofol at various concentrations, followed by stimulation with burn serum, obtained from burn-injury rats. Monolayer permeability was determined by transendothelial electrical resistance. Mitochondrial release of cytochrome C was measured by ELISA. Bax and Bcl-2 expression and mitochondrial release of second mitochondrial-derived activator of caspases (smac) were detected by Western blotting. Caspase-3 activity was assessed by fluorometric assay; mitochondrial membrane potential (Δψm) was determined with JC-1 (a potential-sensitive fluorescent dye). Intracellular ATP content was assayed using a commercial kit, and reactive oxygen species (ROS) were measured by dichlorodihydrofluorescein diacetate (DCFH-DA). Burn serum significantly increased monolayer permeability (P<0.05), and this effect could be inhibited by propofol (P<0.05). Compared with a sham treatment group, intrinsic apoptotic signaling activation - indicated by Bax overexpression, Bcl-2 downregulation, Δψm reduction, decreased intracellular ATP level, increased cytosolic cytochrome C and smac, and caspase-3 activation - was observed in the vehicle group. Propofol not only attenuated these alterations (P<0.05 for all), but also significantly decreased burn-induced ROS production (P<0.05). Propofol attenuated burn-induced RLMVEC monolayer hyperpermeability by regulating the intrinsic apoptotic signaling pathway.


Subject(s)
Humans , Cross Infection/epidemiology , Cross Infection/etiology , Equipment Contamination/statistics & numerical data , Brazil/epidemiology , Hospitals/statistics & numerical data , Intensive Care Units , Sentinel Surveillance
3.
Braz J Med Biol Res ; 48(5): 401-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25760023

ABSTRACT

Recent studies have revealed that an intrinsic apoptotic signaling cascade is involved in vascular hyperpermeability and endothelial barrier dysfunction. Propofol (2,6-diisopropylphenol) has also been reported to inhibit apoptotic signaling by regulating mitochondrial permeability transition pore (mPTP) opening and caspase-3 activation. Here, we investigated whether propofol could alleviate burn serum-induced endothelial hyperpermeability through the inhibition of the intrinsic apoptotic signaling cascade. Rat lung microvascular endothelial cells (RLMVECs) were pretreated with propofol at various concentrations, followed by stimulation with burn serum, obtained from burn-injury rats. Monolayer permeability was determined by transendothelial electrical resistance. Mitochondrial release of cytochrome C was measured by ELISA. Bax and Bcl-2 expression and mitochondrial release of second mitochondrial-derived activator of caspases (smac) were detected by Western blotting. Caspase-3 activity was assessed by fluorometric assay; mitochondrial membrane potential (Δψm) was determined with JC-1 (a potential-sensitive fluorescent dye). Intracellular ATP content was assayed using a commercial kit, and reactive oxygen species (ROS) were measured by dichlorodihydrofluorescein diacetate (DCFH-DA). Burn serum significantly increased monolayer permeability (P<0.05), and this effect could be inhibited by propofol (P<0.05). Compared with a sham treatment group, intrinsic apoptotic signaling activation - indicated by Bax overexpression, Bcl-2 downregulation, Δψm reduction, decreased intracellular ATP level, increased cytosolic cytochrome C and smac, and caspase-3 activation - was observed in the vehicle group. Propofol not only attenuated these alterations (P<0.05 for all), but also significantly decreased burn-induced ROS production (P<0.05). Propofol attenuated burn-induced RLMVEC monolayer hyperpermeability by regulating the intrinsic apoptotic signaling pathway.


Subject(s)
Apoptosis/drug effects , Burns/blood , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Propofol/pharmacology , Serum , Adenosine Triphosphate , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins , Caspase 3/drug effects , Caspase 3/metabolism , Cell Line , Cyclin D1/metabolism , Cytochromes c/analysis , Cytochromes c/metabolism , Electric Impedance , Endothelial Cells/enzymology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation/drug effects , Genes, bcl-2/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Microvessels/cytology , Microvessels/metabolism , Mitochondrial Proteins/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/blood , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL