Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Phytother Res ; 35(3): 1658-1668, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33141989

ABSTRACT

Puerarin is an isoflavone isolated from Pueraria lobata (Willd.) Ohwi. In the present study, reversal effect and underlying mechanisms of puerarin on multidrug resistance (MDR) were investigated in K562/ADR cells. K562/ADR cells exhibited adriamycin (ADR) resistance and higher levels of MDR1 expression compared with K562 cells. Puerarin enhanced the chemosensitivity of K562/ADR cells and increased the ADR accumulation in K562/ADR cells. The expression levels of MDR1 were down-regulated by puerarin in K562/ADR cells. Luciferase reporter assay further demonstrated the inhibitory effect of puerarin on TNF-α-induced NF-κB activation. The phosphorylation of IκB-α was significantly suppressed by puerarin. In silico docking analyses suggested that puerarin well matched with the active sites of IκB-α. Moreover, a large number of autophagosomes were found in the cytoplasm of K562/ADR cells after puerarin treatment. The significant increase in LC3-II and beclin-1 was also observed, indicating autophagy induction by puerarin in K562/ADR cells. Puerarin induced cell cycle arrest and apoptosis in K562/ADR cells. Finally, puerarin inhibited phosphorylation of Akt and JNK. In conclusion, puerarin-sensitized K562/ADR cells by downregulating MDR1 expression via inhibition of NF-κB pathway and autophagy induction via Akt inhibition.


Subject(s)
Autophagy/drug effects , Isoflavones/therapeutic use , K562 Cells/metabolism , NF-kappa B/metabolism , Vasodilator Agents/therapeutic use , Humans , Isoflavones/pharmacology , Transfection , Vasodilator Agents/pharmacology
2.
Exp Hematol ; 41(2): 155-166.e17, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23261964

ABSTRACT

Red cell production is primarily determined by the action of erythropoietin. Additional erythropoiesis-regulatory factors include molecules and cellular interactions occurring within the bone marrow (BM) microenvironment. Sotatercept (ACE-011) is an activin receptor ligand trap that binds several members of the TGF-ß superfamily. Treatment with ACE-011 reverses bone loss and reduces the degree of osteoporosis, but it is accompanied by elevated hemoglobin and hematocrit levels. The mechanisms underlying the beneficial effects of ACE-011 on red cell production remain unknown. This study explores the means by which ACE-011 promotes erythropoiesis. We showed that ACE-011 does not directly affect erythroid differentiation of human CD34(+) cells in vitro. We next tested whether ACE-011 acts indirectly by affecting BM accessory cells. Conditioned media produced by BM stromal cells (SCs) inhibited erythroid differentiation of CD34(+) cells while maintained their ability to proliferate. However, conditioned media from SCs treated with ACE-011 partially restored erythropoiesis, coinciding with changes in the molecular and secretory profile of SCs, including the expression and secretion of erythropoiesis-modulatory factors. We conclude that inhibitory factors produced by BM SCs in vitro might control erythropoiesis in vivo and that agents that reverse these microenvironmental signals could provide an approach to attenuate anemia in clinical conditions.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Erythroid Precursor Cells/drug effects , Erythropoiesis/drug effects , Stromal Cells/drug effects , Adult , Blood Cells/cytology , Blood Cells/drug effects , Blood Cells/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Colony-Forming Units Assay , Culture Media, Conditioned/chemistry , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Drug Evaluation, Preclinical , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoiesis/physiology , Fetal Blood/cytology , Gene Expression Profiling , Humans , In Vitro Techniques , K562 Cells/cytology , K562 Cells/drug effects , K562 Cells/metabolism , Ligands , Organ Specificity , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins , Stromal Cells/physiology
3.
Toxicol In Vitro ; 22(5): 1264-72, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18515041

ABSTRACT

The total oligomers flavonoids (TOF), chloroform, petroleum ether and aqueous extracts from Acacia salicina, were investigated for the antioxidative, cytotoxic, antimutagenic and antigenotoxic activities. The viability of K562 cells were affected by all extracts after 48 h exposure. Our results showed that A. salicina extracts have antigenotoxic and/or antimutagenic activities. TOF and chloroform extracts exhibit antioxidant properties, expressed by the capacity of these extracts to inhibit xanthine oxidase activity. To further explore the mechanism of action of A. salicina extracts, we characterized expression profiles of genes involved in antioxidant protection and DNA repair in the human lymphoblastic cell line K562 exposed to H2O2. Transcription of several genes related to the thioredoxin antioxidant system and to the DNA base-excision repair pathway was up-regulated after incubation with chloroform, TOF and petroleum ether extracts. Moreover genes involved in the nucleotide-excision repair pathway and genes coding for catalase and Mn-superoxide-dismutase, two important antioxidant enzymes, were induced after incubation with the chloroform extract. Taken together, these observations provide evidence that the chloroform and TOF extracts of A. salicina leaves contain bioactive compounds that are able to protect cells against the consequences of an oxidative stress.


Subject(s)
Acacia/chemistry , Antimutagenic Agents/pharmacology , Antioxidants/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Medicine, Traditional , Oxidants/pharmacology , Animals , Cell Line, Tumor , Comet Assay , DNA/drug effects , Drug Combinations , Flavonoids/chemistry , Formazans/metabolism , Gene Expression Profiling , Genes, Bacterial/drug effects , Humans , K562 Cells/drug effects , K562 Cells/metabolism , Oligonucleotide Array Sequence Analysis , Plant Extracts/pharmacology , Rats , Ribosomal Protein S9 , Ribosomal Proteins/drug effects , Ribosomal Proteins/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Tetrazolium Salts/metabolism
4.
Ann N Y Acad Sci ; 1054: 228-37, 2005.
Article in English | MEDLINE | ID: mdl-16339670

ABSTRACT

The first studies of the pharmacological induction of fetal hemoglobin were conducted in patients with sickle cell disease and thalassemia. Although hydroxyurea was approved by the FDA for the treatment of sickle cell disease in 1996, no similar pharmacological agent(s) has been approved for the treatment of patients with thalassemic disorders. The small-scale studies of the induction of fetal hemoglobin in thalassemia have been generally disappointing. The aim of this report is to provide a critical analysis of the factors that may be responsible for our failure to develop an effective fetal hemoglobin induction therapy for patients with thalassemia. We also describe several areas for future investigation that may be critically important for the development of an effective therapy for thalassemia.


Subject(s)
Azacitidine/therapeutic use , Fetal Hemoglobin/biosynthesis , Gene Expression/drug effects , Globins/biosynthesis , Hydroxyurea/therapeutic use , Thalassemia/drug therapy , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Azacitidine/adverse effects , Azacitidine/pharmacology , Combined Modality Therapy , Drug Approval , Drug Evaluation , Drug Evaluation, Preclinical , Erythropoiesis/drug effects , Fetal Hemoglobin/genetics , Gene Expression Regulation , Globins/genetics , Humans , Hydroxyurea/adverse effects , Hydroxyurea/pharmacology , Iron/metabolism , K562 Cells/drug effects , K562 Cells/metabolism , Models, Animal , Papio , Thalassemia/blood , Thalassemia/genetics , Thalassemia/therapy , Transfusion Reaction , United States , United States Food and Drug Administration
5.
J Biomed Sci ; 12(1): 113-21, 2005.
Article in English | MEDLINE | ID: mdl-15864744

ABSTRACT

Denbinobin (5-hydroxy-3,7-dimethoxy-1,4-phenanthraquinone) has been reported to exhibit anti-tumor and anti-inflammatory activity. Nevertheless, the anti-tumor mechanism of denbinobin remains unclear. In the present study, we evaluated the anticancer activity of denbinobin in human myelogenous K562 leukemia cells. In accordance with the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) assay, we demonstrated that denbinobin inhibited cell viability in a concentration-dependent manner with an IC50 value of 1.84 microM. Cell cycle analysis illustrated that exposure of denbinobin caused a G2/M phase accumulation in a time-dependent manner. Tubulin polymerization in cells was apparently enhanced by denbinobin, implying that denbinobin might have a regulatory role in tubulin/microtubule. Furthermore, denbinobin significantly suppressed the expression of Bcr-Abl and phosphorylation of CrkL, a crucial tyrosine kinase and an adaptor protein in chronic myeloid leukemia, respectively. Denbinobin also markedly enhanced CD11b expression after a long-term treatment, suggesting that denbinobin might play a role in facilitating differentiation in K562 cells. In summary, we have demonstrated that denbinobin displays anticancer effects in K562 cells through the increase of levels of tubulin polymerization and deregulation of Bcr-Abl signaling. Our data demonstrate that denbinobin could be a potential anticancer lead compound for further development.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , K562 Cells/drug effects , Phenanthrenes/pharmacology , Protein-Tyrosine Kinases/metabolism , Tubulin/metabolism , Anthraquinones/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , CD11b Antigen/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Fusion Proteins, bcr-abl , Humans , K562 Cells/metabolism , Medicine, Chinese Traditional , Molecular Structure , Phenanthrenes/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Polymers , Protein-Tyrosine Kinases/genetics
6.
Can J Physiol Pharmacol ; 83(3): 309-18, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15870845

ABSTRACT

With the aim of enhancing the efficacy of chemotherapeutic agents, we investigated the antitumor actions and reversal effect on drug resistance of proanthocyanidin plus doxorubicin. The results showed that proanthocyanidin 12.5-200 mg/L significantly inhibited proliferation of K562, K562/DOX, SPC-A-1, and Lewis cells in vitro in a time- and concentration-dependent manner, as determined by microculture tetrazolium assay. A combination of proanthocyani din 12.5, or 25 mg/L and doxorubicin treatment synergistically inhibited cell proliferation with decreased IC50 values. Proanthocyanidin reverses drug resistance in doxorubicin-resistant K562/DOX cells, and IC50 values were decreased by 9.19 (3.64-23.19), 2.56 (1.48-.44), and 0.94 (0.81-1.09) mg/L, respectively, after 24 h treatment with doxorubicin 0.1-9.0 mg/L alone or in combination with proanthocyanidin 12.5 or 25 mg/L; the proanthocyanidin reversal fold was 3.6 and 9.8, respectively. Under confocal laser scanning microscope, the combination of proanthocyanidin 25 or 50 mg/L with doxorubicin 3 mg/L significantly increased the accumulation of intracellular doxorubicin, Ca2+, and Mg2+, and reduced the pH value and mitochondrial membrane potential in K562/DOX cells as compared with doxorubicin alone (p < 0.01). Additionally, the apoptosis rate was increased by 11.3% +/- 3.3%, 14.2% +/- 5.4%, and 23.8% +/- 2.8%, respectively, for doxorubicin 3 mg/L alone or with proanthocyanidin 12.5 or 25 mg/L, as compared with controls (3.0% +/- 1.4%), as demonstrated by flow cytometry. In vivo experiments demonstrated that i.p. administration of proanthocyanidin 10 mg/kg with doxorubicin 2 mg/kg had an inhibitory effect on the growth of transplantation tumor sarcoma 180 and hepatoma 22 in mice as compared with doxorubicin alone (p < 0.05). These results suggest that proanthocyanidin enhances doxorubicin-induced antitumor effect and reverses drug resistance, and its mechanism is attributed partially to the promotion of doxorubicin-induced apoptosis through an elevation of intracellular doxorubicin, and Ca2+, Mg2+ concentration, and a reduction of pH value and mitochondrial membrane potential.


Subject(s)
Antibiotics, Antineoplastic/agonists , Doxorubicin/agonists , K562 Cells/drug effects , Proanthocyanidins/pharmacology , Vitis , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Cell Line , Cell Proliferation , Doxorubicin/pharmacology , Drug Resistance/drug effects , Drug Synergism , Grape Seed Extract , Humans , Hydrogen-Ion Concentration , K562 Cells/metabolism , Magnesium/metabolism , Membrane Potentials/drug effects , Mice , Plant Extracts/pharmacology , Xenograft Model Antitumor Assays
7.
Oncogene ; 21(6): 861-6, 2002 Jan 31.
Article in English | MEDLINE | ID: mdl-11840331

ABSTRACT

We previously demonstrated that RB18A, a member of TRAP220/DRIP205/PBP family, in vivo acted as a cofactor of transcription by differently regulating p53wt transactivating activity on physiological promoters. Using p53-negative cells transfected with different constructs, we herein demonstrated that RB18A down-regulated p53wt-dependent apoptosis. This biological regulation was due to a specific diminution of p53wt protein level, as level of p53mut and GAPDH proteins was not modified. This p53wt diminution was dependent on proteasome activity, as inhibited by MG-132 inhibitor. This specific p53wt degradation was correlated with an increase in expression of MDM2, which promoted p53wt degradation into proteasome. RB18A up-regulated MDM2 expression by activating MDM2 promoter, even in absence of p53wt. Altogether, these data emphasized that RB18A could regulate p53wt function not only by direct interaction between both proteins, but also by up-regulating promoter activity of MDM2, a p53-regulating partner.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Gene Expression Regulation , Nuclear Proteins , Transcription Factors , Tumor Suppressor Protein p53/physiology , Apoptosis/genetics , Carcinoma, Embryonal/pathology , DNA, Complementary/genetics , Genes, p53 , Humans , K562 Cells/cytology , K562 Cells/metabolism , Lung Neoplasms/pathology , Mediator Complex Subunit 1 , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-mdm2 , Recombinant Fusion Proteins/physiology , Transfection , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism , Tumor Suppressor Protein p53/biosynthesis
8.
Anticancer Res ; 22(6A): 3349-56, 2002.
Article in English | MEDLINE | ID: mdl-12530086

ABSTRACT

AIMS: Radiolabeled antisense oligonucleotide to target the mRNA of the hmdr1 gene for diagnostic purposes is a new concept for evaluating the chemoresistance of tumors in vivo. METHODS AND RESULTS: An 18 mer complementary to the zone which contains the translation initiation codon of the hmdr1 gene was modified using one phosphoramidate group and one dimethoxytrityle group at the 5' and 3'ends. It permitted probe radiolabeling by 125I. Chemical modifications made to the antisense probe ensured the stability in biological media tested by incubation with human serum at 37 degrees C from 5 minutes to 24 hours. These modifications did not interfere with recognition of the target. Retention of the antisense probe followed the expression level of the target transcript in in vitro and in vivo studies. In vitro, after a 2-hour incubation in the presence of K562--sensitive (S) and- resistant (R) cell lines, uptake was respectively 4.27 +/- 0.96% ID/mg protein and 7.78 +/- 0.46% ID/mg protein (p < 0.001). In vivo, the ratios between radioactivity found in the tumor and that found in the striated muscle and in the blood were, respectively, 20 and 3 for IGR OV1 resistant tumor and 1 and 0.3 for the sensitive one. CONCLUSION: In our study, resistant cell lines and tumor showed greater retention of the specific probe than the sensitive ones. This constitutes a further advance towards non invasive imaging of resistant genes involved in chemoresistance. These results are encouraging: the current trend in innovative cancer therapy is moving towards targeting the genes of interest.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Genes, MDR , Iodine Radioisotopes , Oligonucleotides, Antisense/genetics , RNA, Messenger/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Dose-Response Relationship, Drug , Drug Stability , Female , Humans , K562 Cells/drug effects , K562 Cells/metabolism , Kinetics , Mice , Mice, Nude , Middle Aged , Nucleic Acid Hybridization , Oligonucleotides, Antisense/pharmacokinetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Polyethyleneimine/pharmacology , RNA, Messenger/biosynthesis , Tissue Distribution , Tumor Cells, Cultured
9.
Cancer Lett ; 160(1): 21-8, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11098080

ABSTRACT

We investigated the effects of the ethyl acetate extract of grapefruit juice (GFJ), that of orange juice (OJ) and their components on the uptake of [(3)H]vincristine into adriamycin-resistant human myelogenous leukemia cells. Its uptake was increased by the extracts of GFJ and OJ up to 7- and 3-fold, respectively, as well as verapamil and cyclosporin A. OJ components, i.e. 3,3',4',5,6,7,8-heptamethoxyflavone, nobiletin and tangeretin, also increased the uptake of [(3)H]vincristine in a concentration-dependent manner. Although GFJ components, dihydroxybergamottin and bergamottin, significantly increased the uptake, their potencies were considerably weaker than those of OJ components. These data suggest that OJ components inhibit P-gp-mediated efflux of [(3)H]vincristine, resulting in the intracellular accumulation of chemotherapeutic drugs. These components may become candidates of multi-drug resistance reversing agents in cancer chemotherapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Citrus/chemistry , Doxorubicin/pharmacology , Flavones , Flavonoids/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Acetates , Beverages , Blotting, Western , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Furocoumarins/chemistry , Furocoumarins/pharmacology , Humans , K562 Cells/drug effects , K562 Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Tritium , Verapamil/pharmacology , Vincristine/pharmacokinetics
10.
Photochem Photobiol ; 70(6): 858-67, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10628299

ABSTRACT

Singlet oxygen (1O2) can be quenched by water, lipids, proteins, nucleic acids and other small molecules. Polyunsaturated fatty acids (PUFA) of cells principally quench 1O2 by chemical mechanisms, producing lipid hydroperoxides, while proteins physically and chemically quench 1O2. Because cell lines can have different PUFA and protein levels, we hypothesized that 1O2 toxicity will vary between cell lines. We used Photofrin as a source of 1O2. Exposure of nine different leukemia cell lines (CEM, HEL, HL-60, K-562, KG-1, L1210, Molt-4, THP-1 and U-937) to Photofrin and light results in changes in membrane permeability (trypan blue) that vary with cell line. The greater the lipid content of the cell line, the less susceptible they are to membrane damage. When the cell media was supplemented with docosahexaenoic acid (DHA, 22:6), the overall unsaturation of cellular lipids increased. Photofrin and light resulted in increased radical formation in these supplemented cells compared to controls; however, there was no difference in membrane permeability between DHA-supplemented and control cells. Lipid-derived radical formation (electron paramagnetic resonance spin trapping) was cell line dependent; but no correlation between lipid content of cells and radical formation was found. However, we found that the greater the protein content of cells the more they were protected against membrane damage induced by Photofrin photosensitization. This suggests that cellular proteins are a key target for 1O2-mediated toxicity. A remarkable observation is that cell size correlates inversely with ability of cells to cope with a given flux of 1O2.


Subject(s)
Leukemia, Experimental/metabolism , Lipid Peroxidation/drug effects , Oxygen/toxicity , Animals , HL-60 Cells/drug effects , HL-60 Cells/metabolism , Humans , K562 Cells/drug effects , K562 Cells/metabolism , Leukemia L1210/drug therapy , Leukemia L1210/metabolism , Leukemia, Experimental/drug therapy , Mice , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL