Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharmacol ; 893: 173840, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33359649

ABSTRACT

Cancer stem cell (CSC) plays an important role in pancreatic cancer pathogenesis and treatment failure. CSCs are characterized by their ability to form tumor spheres in serum-free medium and expression of CSC related markers. In the present study, we investigated the effect atorvastatin, celecoxib and tipifarnib in combination on proliferation and apoptosis in Panc-1 sphere-forming cells. The sphere-forming cells were isolated from Panc-1 cells by sphere-forming method. These sphere-forming cells showed CSC properties. The levels of CD44, CD133 and ALDH1A1 in the sphere-forming cells were increased. Moreover, Panc-1 sphere-forming cells were resistant to chemotherapeutic drug gemcitabine. Combined atorvastatin with celecoxib and tipifarnib synergistically decreased the sphere forming ability of Panc-1 cells and the drug combination also strongly inhibited cell proliferation and promoted apoptosis in the sphere-forming cells. The effects of the drug combination on the Panc-1 sphere-forming cells were associated with decreases in the levels of CD44, CD133 and ALDH1A1, and suppression of Akt and NF-κB activation. Results of the present study indicate that the combination of atorvastatin, celecoxib and tipifarnib may represent an effective approach for inhibiting pancreatic CSCs.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Atorvastatin/pharmacology , Celecoxib/pharmacology , Cell Proliferation/drug effects , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/drug therapy , Quinolones/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction , Spheroids, Cellular
2.
Phytomedicine ; 78: 153309, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32890914

ABSTRACT

BACKGROUND: Combination drug therapy has become an effective strategy for inflammation control. The anti­inflammatory capacities of silibinin and thymol have each been investigated on its own, but little is known about the synergistic anti-inflammatory effects of these two compounds. PURPOSE: This study aims to investigate the synergistic anti-inflammatory effects of silibinin and thymol when administered in combination to lipopolysaccharide (LPS)-induced RAW264.7 cells. METHODS: RAW264.7 cells were pre-treated with silibinin and thymol individually or in combination for 2 h before LPS stimulation. Cell viability was detected by the MTT assay. Nitric oxide (NO) production was measured by Griess reagent. Reactive oxygen species (ROS) was evaluated by 2',7'-dichlorofluorescein-diacetate. ELISA was used to detect tumour necrosis factor-α (TNF-α), and interleukin-6 (IL-6). Western blot was performed to analyse the protein expression of LPS-induced RAW264.7 cells. RESULTS: We observed a synergistic anti-inflammatory effect of silibinin and thymol when administered in combination to LPS-induced RAW264.7 cells. Silibinin combined with thymol (40 µM and 120 µM respectively, with the molar ratio 1:3) had more potent effects on the inhibition of NO, TNF-α, and IL-6 than those exerted by individual administration of these compounds in LPS-induced RAW264.7 cells. The combination of silibinin and thymol (40 µM and 120 µM respectively, with the molar ratio 1:3) strongly inhibited ROS and cyclooxygenase-2 (COX-2). More importantly, the combination of silibinin and thymol (40 µM and 120 µM respectively, with the molar ratio 1:3) was also successful in inhibiting nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) activities. Our results suggest that the synergistic anti-inflammatory effects of silibinin with thymol were associated with the inhibition of NF-κB and MAPK signalling pathways. CONCLUSION: The combination of silibinin and thymol (40 µM and 120 µM, respectively, with the molar ratio 1:3) could inhibit inflammation by suppressing NF-κB and MAPK signalling pathways in LPS-induced RAW264.7 cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , NF-kappa B/metabolism , Silybin/pharmacology , Thymol/pharmacology , Animals , Cyclooxygenase 2/metabolism , Drug Synergism , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Eur J Med Chem ; 189: 112013, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31972390

ABSTRACT

In this study, two series of coumarin derivatives 5a∼i and 6a∼i were synthesized, and their inhibitory activity against α-glucosidase was determined. The results indicated that most of the synthesized derivatives exhibited prominent inhibitory activities against α-glucosidase. Among them, compounds 5a and 5b showed the strongest inhibition with the IC50 values of 19.64 µM and 12.98 µM, respectively. Enzyme kinetic studies of compounds 5a and 5b proved that their inhibition was reversible and a mixed type. The KI and KIS values of compound 5a were calculated to be 27.39 µM and 13.02 µM, respectively, and the corresponding values for compound 5b being 27.02 µM and 13.65 µM, respectively. The docking studies showed that compound 5b could be inserted into the active pocket of α-glucosidase and form hydrogen bonds with LYS293 to enhance the binding affinity.


Subject(s)
Coumarins/chemistry , Glycoside Hydrolase Inhibitors/chemical synthesis , Glycoside Hydrolase Inhibitors/pharmacology , alpha-Glucosidases/chemistry , Hydrogen Bonding , Kinetics , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
4.
Neural Regen Res ; 13(4): 742-752, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29722330

ABSTRACT

This review summarizes and describes the use of curcumin in diagnosis, prevention, and treatment of Alzheimer's disease. For diagnosis of Alzheimer's disease, amyloid-ß and highly phosphorylated tau protein are the major biomarkers. Curcumin was developed as an early diagnostic probe based on its natural fluorescence and high binding affinity to amyloid-ß. Because of its multi-target effects, curcumin has protective and preventive effects on many chronic diseases such as cerebrovascular disease, hypertension, and hyperlipidemia. For prevention and treatment of Alzheimer's disease, curcumin has been shown to effectively maintain the normal structure and function of cerebral vessels, mitochondria, and synapses, reduce risk factors for a variety of chronic diseases, and decrease the risk of Alzheimer's disease. The effect of curcumin on Alzheimer's disease involves multiple signaling pathways: anti-amyloid and metal iron chelating properties, antioxidation and anti-inflammatory activities. Indeed, there is a scientific basis for the rational application of curcumin in prevention and treatment of Alzheimer's disease.

5.
Mol Nutr Food Res ; 62(2)2018 01.
Article in English | MEDLINE | ID: mdl-28849618

ABSTRACT

SCOPE: Curcuma longa (turmeric) is a folk medicine in South and Southeast Asia, which has been widely used to alleviate chronic inflammation. Aromatic-turmerone is one of the main components abundant in turmeric essential oil. However, little information is available from controlled studies regarding its biological activities and underlying molecular mechanisms against chronic inflammation in the brain. In the current study, we employed a classical LPS model to study the effect and mechanism of aromatic-turmerone on neuroinflammation. METHODS AND RESULTS: The effects of aromatic-turmerone were studied in LPS-treated mice and BV2 cells. The cognitive function assays, protein analyses, and histological examination were performed. Oral administration of aromatic-turmerone could reverse LPS-induced memory disturbance and normalize glucose intake and metabolism in the brains of mice. Moreover, aromatic-turmerone significantly limited brain damage, through inhibiting the activation of microglia and generation of inflammatory cytokines. Further study in vitro revealed that aromatic-turmerone targeted Toll-like receptor 4 mediated downstream signaling, and lowered the release of inflammatory mediators. CONCLUSION: These observations indicate that aromatic-turmerone is effective in preventing brain damage caused by neuroinflammation and may be useful in the treatment of neuronal inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/drug therapy , Ketones/pharmacology , Memory Disorders/drug therapy , Sesquiterpenes/pharmacology , Toll-Like Receptor 4/metabolism , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Brain/drug effects , Brain/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Ketones/administration & dosage , Lipopolysaccharides/toxicity , Maze Learning/drug effects , Memory Disorders/chemically induced , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Sesquiterpenes/administration & dosage , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics
6.
Protein Expr Purif ; 37(1): 47-52, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15294280

ABSTRACT

Na+, K+-ATPase beta2 subunit (NKA1b2) is not only a regulator of Na+, K+-ATPase, but also functions in the interaction between neuron and glia cells as a Ca2+-dependent adhesion molecule. To further study the function of NKA1b2, the anti-NKA1b2 polyclonal antibody was prepared to recognize the outer-membrane carboxyl portion segment of NKA1b2. The coding region for amino acids 190-290 at the carboxyl portion of NKA1b2 (NKA1b2-CP) was sub-cloned into the vector pGEX-4T-2 and introduced into the Escherichia coli BL21(DE3) cell for efficient soluble expression. The amino acid sequence of expressed protein was determined using mass spectrometry following Mascot analysis. After purification, GST-NKA-beta2-CP was used to immunize the adult rabbits following standard protocols. The produced antiserum could detect the NKA1b2 protein expressed not only in the prokaryotic cells (E. coli) but also in the eukaryotic cells (COS7) transfected with NKA1b2 expression vector (pEGFP-NKA1b2). Furthermore, the antiserum was used for determining the localization of NKA1b2 in primary culture of neonatal rat neurons using immunohistochemical technique. Results demonstrated that NKA1b2 was localized both in the cytoplasm and cellular membrane. The preparation of anti-NKA-beta2-CP polyclonal antibody will facilitate further functional study on NKA1b2.


Subject(s)
Antibodies/metabolism , Prokaryotic Cells/physiology , Protein Subunits/metabolism , Recombinant Fusion Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Humans , Mice , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Protein Subunits/genetics , Protein Subunits/immunology , Rabbits , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Alignment , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...