Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Pharm Sci ; 113(4): 1047-1053, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37844758

ABSTRACT

The purpose of this study was to elucidate and compare styrene maleic acid copolymer (SMA)-coated lipid emulsions (SMA emulsions) uptake pathway in vascular endothelial cells and surrounding cancer cells under not only neutral but also acidic pH, which is often observed in tumor microenvironment. DiI-labeled SMA emulsions were prepared using 1-palmitoyl-2-oleoyl-sn­glycero-3-phosphocholine and triolein. In murine melanoma B16-BL6 (B16) cells and human umbilical vein endothelial cells (HUVEC), DiI-labeled SMA emulsions uptake under near-neutral (pH 7.4) and acidic (pH 6.0) conditions was determined by fluorescent analysis. SMA emulsions were taken up more efficiently into HUVEC than B16 cells under acidic condition in a temperature-dependent manner. Uptake study using endocytosis inhibitors showed that SMA emulsions were taken up by macropinocytosis and clathrin-mediated endocytosis in B16 cells. In HUVEC, however, they were taken up by clathrin- and caveolae-independent, but dynamin-dependent pathway. SMA emulsions would be internalized efficiently into vascular endothelial cells as well as cancer cells under acidic microenvironment via different endocytosis pathways. SMA emulsions could be a promising drug delivery carrier for anti-angiogenic drugs.


Subject(s)
Endothelial Cells , Tumor Microenvironment , Mice , Humans , Animals , Emulsions , Polystyrenes , Maleates , Drug Carriers , Clathrin
2.
Biol Pharm Bull ; 46(10): 1479-1483, 2023.
Article in English | MEDLINE | ID: mdl-37779050

ABSTRACT

Niosomes are non-ionic surfactant (NIS)-based bilayer vesicles and, like liposomes, have great potential as drug-delivery systems. Our previous study revealed that polyethylene glycol (PEG) niosomes using different sorbitan ester (Span) surfactants (sorbitan monoester, Span 20, 40, 60, 80; sorbitan triester, Span 65) distributed within tumors similarly to PEG liposomes. The aim of this study was to encapsulate efficiently an anti-cancer drug, paclitaxel (PTX) into Span PEG niosomes, and evaluate PTX release profiles and anti-tumor efficacy of PTX-loaded Span PEG niosomes. Niosome sizes ranged between 100-150 nm, and the PTX encapsulation efficiency was more than 50%. All niosomes examined, in the presence of serum, yielded sustained PTX-release profiles. PTX release at 24 and 48 h from Span 80 PEG niosomes was significantly the highest among the other Span PEG niosomes examined. In C26 tumor-bearing mice, PTX-loaded Span 40 PEG niosomes (the lowest PTX release in vitro) suppressed tumor growth while PTX-loaded Span 80 PEG niosomes (the highest PTX release in vitro) did not. Thus, we succeeded in the control of PTX release from Span PEG niosomes by modifying the component of niosomes, and it influenced the effects of drugs loaded into niosomes. This demonstrates that the excellent NIS physicochemical properties of Spans make them an ideal candidate for anti-cancer drug-carrier niosomes.


Subject(s)
Antineoplastic Agents , Liposomes , Mice , Animals , Liposomes/chemistry , Paclitaxel/pharmacology , Polyethylene Glycols/chemistry , Antineoplastic Agents/pharmacology , Drug Carriers , Surface-Active Agents
3.
Biol Pharm Bull ; 46(9): 1347-1351, 2023.
Article in English | MEDLINE | ID: mdl-37661414

ABSTRACT

Macrophages selectively infiltrate the lesion sites of several diseases, including cancers, and, thus, have attracted attention as a biomimetic drug delivery carrier. To achieve the efficient drug loading of macrophages with minimal cytotoxicity, drugs are preferably encapsulated into nanoparticles, such as liposomes, and modified on the surface of macrophages rather than being incorporated into cells. However, liposomes are rapidly taken up by macrophages after binding to the cell surface because of their strong phagocytic activity. To overcome this, we herein attempted to modify the surface of macrophages with liposomes by suppressing their phagocytic activity using a pretreatment with anionic liposomes. We confirmed that 1,2-distearoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (DSPG)- and cholesterol-rich anionic liposomes were efficiently taken up by RAW264.7 murine macrophage-like cells. Furthermore, the cellular uptake of anionic liposomes by RAW264.7 cells was higher in the absence of fetal bovine serum (FBS) than in its presence. Moreover, the viability of RAW264.7 cells was maintained above 90% when cells were incubated with anionic liposomes for 3 h, whereas viability was markedly decreased after a 24-h incubation. Based on these results, we pretreated RAW264.7 cells by an incubation with DSPG- and cholesterol-rich liposomes for 3 h in the absence of FBS. This pretreatment significantly inhibited the internalization of other liposomes, which subsequently bound to the cell surface. Therefore, we succeeded in modifying the surface of macrophages with liposomes, and liposome-modified macrophages have potential as a biomimetic active drug delivery carrier.


Subject(s)
Biomimetics , Liposomes , Animals , Mice , Macrophages , Phagocytes , Drug Delivery Systems , Drug Carriers , Excipients
4.
Biochem Biophys Res Commun ; 677: 26-30, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37542772

ABSTRACT

One of common characteristics of solid tumors is low O2 level, so-called hypoxia, which plays a critical role in chemoresistance. Epigenetic mechanism such as DNA methylation and histone modification is involved in cancer development and progression. There is ample evidence that epigenetic drugs reversed acquired chemoresistance in cancer cells under normal O2 level, normoxia. However, it remains unknown whether epigenetic drugs improve acquired chemoresistance under hypoxia. The aim of our study was to investigate whether epigenetic drugs can improve the chemoresistance induced under hypoxia in cancer cells. In murine melanoma B16-BL6 (B16) cells, the culture under hypoxia, 1%O2 caused the elevated expression of hypoxia-inducible factor-1α (HIF-1α) and its target genes. The chemoresistance to 7-ethyl-10-hydroxycamptothecin (SN-38, the active metabolite of irinotecan) was also acquired under hypoxia in B16 cells. In addition, as epigenetic mechanisms, the protein expression of the enhancer of zeste homolog 2 (EZH2), histone methyltransferase and its target histone H3 trimethylation at lysine 27 (H3K27Me3) level increased under hypoxia. The induction of H3K27Me3 under hypoxia was suppressed by EZH2 siRNA and 3-deazaneplanocin A (DZNep), an EZH2 inhibitor. Furthermore, both EZH2 siRNA and DZNep significantly reduced the cell viability after SN-38 treatment and improved the chemoresistance to SN-38 under hypoxia. These results indicated that the chemoresistance to SN-38 under hypoxia would arise from epigenetic mechanism, H3K27Me3 elevation due to EZH2 induction. In conclusion, a histone methyltransferase EZH2 inhibitor, DZNep was capable of tackling acquired chemoresistance via the suppression of histone methylation induced under hypoxic tumor microenvironment.


Subject(s)
Histones , Melanoma , Humans , Animals , Mice , Histones/metabolism , Histone Methyltransferases/genetics , Irinotecan , Drug Resistance, Neoplasm , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/metabolism , Enzyme Inhibitors/pharmacology , DNA Methylation , RNA, Small Interfering/metabolism , Melanoma/genetics , Tumor Microenvironment
5.
Biomedicines ; 11(2)2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36831094

ABSTRACT

Mesenchymal stem cells (MSCs) have a tumor-homing capacity; therefore, MSCs are a promising drug delivery carrier for cancer therapy. To maintain the viability and activity of MSCs, anti-cancer drugs are preferably loaded on the surface of MSCs, rather than directly introduced into MSCs. In this study, we attempted to load liposomes on the surface of MSCs by using the magnetic anionic liposome/atelocollagen complexes that we previously developed and assessed the characters of liposome-loaded MSCs as drug carriers. We observed that large-sized magnetic anionic liposome/atelocollagen complexes were abundantly associated with MSCs via electrostatic interactions under a magnetic field, and its cellular internalization was lower than that of the small-sized complexes. Moreover, the complexes with higher atelocollagen concentrations showed lower cellular internalization than the complexes with lower atelocollagen concentrations. Based on these results, we succeeded in the efficient loading of liposomes on the surface of MSCs by using large-sized magnetic anionic liposomes complexed with a high concentration of atelocollagen. The constructed liposome-loaded MSCs showed a comparable proliferation rate and differentiation potential to non-loaded MSCs. Furthermore, the liposome-loaded MSCs efficiently adhered to vascular endothelial cells and migrated toward the conditioned medium from cancer cells in vitro and solid tumor tissue in vivo. These findings suggest that liposome-loaded MSCs could serve as an efficient cell-based drug carrier for tumor-targeted delivery.

6.
Int J Hematol ; 118(1): 146-150, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36797397

ABSTRACT

BACKGROUND: Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is caused by anti-HPA alloantibody, and anti-HPA-4b is the most common cause in Japanese. Anti-HPA-5b is frequently detected in pregnant women, but it is still controversial whether anti-HPA-5b causes severe FNAIT. CASE PRESENTATION: A Japanese woman with anti-HPA-4b and anti-HPA-5b alloantibodies delivered a baby with severe FNAIT who was both HPA-4b and HPA-5b incompatible. We carefully monitored the patient's following three pregnancies (the second and the fourth siblings were HPA-4b incompatible and HPA-5b compatible; the third sibling was both HPA-4b and HPA-5b compatible). FNAIT was not observed in all three siblings, although a modest decrease in cord blood platelet count was observed in the HPA-4b incompatible siblings compared to the HPA-4b compatible sibling. Serial monitoring of anti-HPA titer showed that anti-HPA-4b markedly decreased in late pregnancy and recovered after delivery of the HPA-4b incompatible siblings, but these decreases were not observed during the mother's pregnancy with the HPA-4b compatible sibling. In contrast, anti-HPA-5b remained at a high titer during pregnancy with all three siblings. CONCLUSION: Our data indicate that dynamic changes of anti-HPA-4b occur during pregnancy and strongly suggest that anti-HPA-5b was mainly responsible for severe FNAIT in this case.


Subject(s)
Antigens, Human Platelet , Thrombocytopenia, Neonatal Alloimmune , Infant, Newborn , Humans , Pregnancy , Female , Thrombocytopenia, Neonatal Alloimmune/diagnosis , Isoantibodies , Siblings , Platelet Count
7.
Biochem Biophys Res Commun ; 607: 9-14, 2022 06 04.
Article in English | MEDLINE | ID: mdl-35358872

ABSTRACT

Vorinostat (suberoylanilide hydroxamic acid: SAHA), a histone deacetylase inhibitor, has potential benefit of improving the resistance to conventional other anti-cancer drugs. This study was aimed to clarify whether SAHA improves the resistance to oxaliplatin (L-OHP), a platinum-based anticancer drug using L-OHP-resistant HCT116 cells (HCT116/OxR), established from colorectal cancer (CRC) cell line HCT116. HCT116/OxR cells showed cross-resistance to other platinum-based drugs. Pre-treatment with SAHA improved the sensitivity of both L-OHP and its metabolite in HCT116/OxR cells, but not in parental HCT116 cells. However, pre-treatment with SAHA did not affect the sensitivity of other platinum-based drugs. These results indicated that SAHA specifically improved the sensitivity of L-OHP in HCT116/OxR cells. Focusing on NF-E2 p45-related factor 2-Kelch-like ECH-associated protein 1 pathway (Nrf2-Keap1) pathway, which is activated by oxidative stress such as the treatment with anti-cancer drugs, mechanisms behind these observations were elucidated. In HCT116/OxR cells transfected with Nrf2 siRNA, the improving effects on L-OHP resistance by SAHA were abolished, suggesting that Nrf2-Keap1 pathway was involved in L-OHP-resistance. In addition, L-OHP metabolite significantly induced the expression of the nuclear protein Nrf2 and its target gene mRNA expression in HCT116/OxR cells. Pre-treatment with SAHA suppressed these changes observed in HCT116/OxR cells. In conclusion, this study demonstrated that SAHA improved L-OHP resistance by inhibiting Nrf2-Keap1 activation via Nrf2 nuclear translocation by L-OHP metabolite.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Vorinostat/pharmacology
8.
Biochem Biophys Res Commun ; 578: 157-162, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34571370

ABSTRACT

Decitabine (DAC), a DNA methyltransferase (DNMT) inhibitor is a novel anti-cancer drug regulating epigenetic mechanisms. Similar to conventional anti-cancer drugs, drug resistance to DAC also has been reported, resulting in tumor recurrence. Our previous study using colorectal cancer HCT116 cells found the decrease in deoxycytidine kinase (dCK) (activation enzyme of DAC) and the increase in cytidine deaminase (inactivation enzyme of DAC) in acquired DAC-resistant HCT116 (HCT116/DAC) cells. The aim of our study was to clarify the involvement of dCK and CDA in DAC resistance. In order to tackle DAC resistance, it was also examined whether other DNMT inhibitors such as azacytidine (AC) and polyphenols are effective in DAC-resistant cancer cells. When dCK siRNA was transfected into HCT116 cells, IC50 value of DAC increased by about 74-fold and reached that of HCT116/DAC cells with attenuated dCK. dCK siRNA to HCT116 cells also abolished DNA demethylation effects of DAC. In contrast, CDA siRNA to HCT116 cells did not influence the efficacy of DAC. In addition, CDA siRNA to HCT116/DAC cells with increased CDA did not restore the compromised effects of DAC. These results suggested that attenuated dCK but not increased CDA mainly contributed to DAC resistance. Regarding dCK in HCT116/DAC cells, a point mutation with amino acid substitution was observed while the product size and expression of mRNA coding region did not change, suggesting that dCK protein was decreased by post-transcriptional regulation. AC and polyphenols showed no cross-resistance in HCT116/DAC cells. AC but not polyphenols exerted DNA demethylation effect. Among polyphenols, curcumin (Cur) showed the most synergistic cytotoxicity in combination with AC while DNA demethylation effect of AC was partly maintained. Taken together, combination of AC and Cur would be a promising alternative to tackle DAC resistance mainly due to attenuated dCK.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azacitidine/pharmacology , Colorectal Neoplasms/drug therapy , Curcumin/pharmacology , Decitabine/pharmacology , Deoxycytidine Kinase/deficiency , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/administration & dosage , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Curcumin/administration & dosage , Cytidine Deaminase/metabolism , DNA Methylation , Decitabine/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Humans
9.
Biochem Biophys Rep ; 26: 100996, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33898768

ABSTRACT

microRNAs (miRNAs) contained in small extracellular vesicles (sEVs) are candidates for non-invasive biomarkers. Oxaliplatin (L-OHP) has been approved for advanced colorectal cancer (CRC) chemotherapy. However, the response to L-OHP differs among CRC patients. In addition, CRC cells often acquire the resistance to L-OHP. This study aimed at the prediction of L-OHP sensitivity by measuring extracellular miRNAs levels. Firstly, we compared intracellular miRNAs expressions in L-OHP-sensitive CRC cells (SW620 and HCT116 cells) with those in acquired and intrinsic L-OHP-resistant cells. In microarray and real-time RT-PCR analyses, the intracellular miR-33a-5p, miR-210-3p, and miR-224-5p expressions were lower in acquired and intrinsic L-OHP-resistant CRC cells than sensitive cells. Furthermore, in SW620 cells, L-OHP sensitivity was decreased by miR-33a-5p inhibitor. On the other hand, miR-210-3p or miR-224-5p inhibitor did not affect L-OHP sensitivity in SW620 cells. Secondly, the amount of miR-33a-5p, miR-210-3p, and miR-224-5p in sEVs was compared. The amount of miR-33a-5p and miR-210-3p in sEVs secreted from acquired and intrinsic L-OHP-resistant cells tended to be small. miR-224-5p was not detected in sEVs secreted from three types of CRC cells examined. To the best of our knowledge, this is the first study demonstrating that miR-33a-5p and/or miR-210-3p in sEVs would be candidates for biomarkers of L-OHP sensitivity. In particular, miR-33a-5p is a promising candidate because it would be directly involved in L-OHP sensitivity.

10.
Chem Pharm Bull (Tokyo) ; 68(12): 1233-1237, 2020.
Article in English | MEDLINE | ID: mdl-33268655

ABSTRACT

The aim of this study was to investigate appropriate analytical conditions for hydrophilic nucleosides and nucleotides (monophosphates and triphosphates) by HPLC methods using a mixed-mode AX-C18 column with anion-exchange and hydrophobic interactions by quaternary ammonium and C18, respectively, and a reversed-phase pentabromobenzyl (PBr) column with dispersion force and hydrophobic interactions by PBr group. The higher compound polarity led to stronger retention on AX-C18 (triphosphates > monophosphates > nucleosides). AX-C18 demonstrated feasible retention of nucleotides via anion-exchange interaction by increasing the salt and methanol concentrations. In contrast, on PBr, the lower compound polarity led to stronger retention. On PBr, feasible retention of both nucleosides and nucleotides was obtained via dispersion interactions with purine and pyrimidine rings by increasing the methanol concentration. Regarding the pH of phosphate buffer used as the mobile phase, pH 7.0 should be used in measuring nucleoside triphosphates on AX-C18, whereas pH 2.5 is better suited for measuring nucleotides on PBr. In terms of selectivity to highly hydrophilic nucleotides, the mixed-mode AX-C18 column had an advantage over the reverse-phase PBr column. In contrast, PBr column was more versatile than the AX-C18 column. Taken together, HPLC analyses of nucleosides and nucleotides should be carried out by optimizing the interactions between the stationary phase and nucleic acids.


Subject(s)
Nucleic Acids/analysis , Phosphates/analysis , Chromatography, High Pressure Liquid , Hydrophobic and Hydrophilic Interactions
11.
Drug Metab Pharmacokinet ; 35(1): 124-130, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31964620

ABSTRACT

Decitabine (DAC), a DNA methylation inhibitor, is transported into cancer cells mainly via equilibrative nucleoside transporter 1 (ENT1) and subsequently phosphorylated by deoxycytidine kinase (dCK). We previously reported that apparent DAC uptake into cells may be described using a simple compartment model with clearance for facilitated diffusion (PS) and subsequent phosphorylation (CLmet). In the present study, time course of apparent intracellular [3H]-DAC uptake was analyzed numerically, and PS and CLmet values were calculated using the compartment model in human colon cancer HCT116 cells. PS at 0.1 µM [3H]-DAC was markedly decreased in the presence of 100 µM irinotecan or etoposide, while CLmet was markedly decreased in the presence of 100 µM cytarabine or gemcitabine. CLmet at 0.1-10 µM [3H]-DAC varied in a concentration-dependent manner and was described by Michaelis-Menten parameters Km,met and Vmax,met. In conclusion, DAC uptake mainly via ENT1 may be described by a bidirectional first-order kinetic parameter, while phosphorylation by dCK may be described by Michaelis-Menten parameters.


Subject(s)
Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacokinetics , Decitabine/metabolism , Decitabine/pharmacokinetics , Equilibrative Nucleoside Transporter 1/metabolism , Nonlinear Dynamics , HCT116 Cells , Humans , Kinetics , Phosphorylation , Tritium/chemistry
12.
Yakugaku Zasshi ; 140(1): 107-111, 2020.
Article in Japanese | MEDLINE | ID: mdl-31902878

ABSTRACT

The purpose of this study was to examine how students prepare for the pharmaceutical technical English course "Yakugaku-Eigo Nyumon" by qualitative analysis. A sub-text, supplemental material was used to assist students with class preparation. Qualitative questionnaires on understanding and approaches for class preparation as well as review of class were analyzed in comparison with different academic performance levels on the final exam. The results of qualitative analysis of class preparation based on coding revealed that high-academic-performing students understood and adopted deep-processing approaches for the preparation of "English words" and "understanding of content" more often than low-academic-performing students. High-performing students attempted to not literally translate English sentences into Japanese while checking the English words with thinking and ingenuity, and to understand English sentences by drawing figures and thinking of relationships using previously learned knowledge. These approaches were not adopted by low-performing students. Furthermore, sub-text was one of the means for understanding by high-performing students, whereas it was essential for low-performing students to understand the content. Coding results on the review of class also showed that low-performing students were dependent mainly on sub-text for understanding. These results suggest that deep-processing approaches to both English and content of materials are necessary for deep understanding in "Yakugaku-Eigo Nyumon".


Subject(s)
Comprehension , Curriculum , Education, Pharmacy , Knowledge , Language , Learning , Students, Pharmacy/psychology , Academic Success , Aptitude Tests , Female , Humans , Male , Surveys and Questionnaires
14.
Biochem Biophys Res Commun ; 509(1): 249-254, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30581001

ABSTRACT

Epigenetic modifiers such as decitabine (DAC), a DNA methyltransferase inhibitor, have the potential benefit of combination therapy with conventional anti-cancer drugs. This study was aimed to clarify whether DAC at the low concentration without cytotoxic effects exerts synergistic effects with conventional anti-cancer drugs in human colorectal cancer cell line, HT29 cells low-sensitive to DAC. DAC at very low concentration below its IC20 synergistically enhanced cytotoxicity of oxaliplatin (L-OHP), and the enhancement was the most remarkable for L-OHP among several anti-cancer drugs tested. DAC was found to be metabolized and incorporated into DNA in HT29 cells. Combination of L-OHP with DAC did not increase the protein expression of γH2A.X, the earliest indicator of DNA damage, in HT29 cells. On the other hand, although DAC alone did not produce reactive oxygen species (ROS), DAC significantly enhanced ROS production by L-OHP in HT29 cells. Furthermore, enhanced cytotoxicity of L-OHP by DAC was cancelled with the presence of N-acetyl-l-cysteine, a ROS scavenger. The mRNA expression of ROS-generating enzymes was significantly increased by combination in HT29 cells. Taken together, indirect enhancement of ROS production by DAC at the low concentration with negligible cytotoxicity should be at least involved in synergistic effects with L-OHP in HT29 cells with intrinsic resistance to DAC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Decitabine/pharmacology , Drug Resistance, Neoplasm/drug effects , Oxaliplatin/pharmacology , Apoptosis , Colorectal Neoplasms/metabolism , Drug Synergism , HCT116 Cells , HT29 Cells , Humans , Reactive Oxygen Species/metabolism
15.
Transfusion ; 58(12): 3003-3013, 2018 12.
Article in English | MEDLINE | ID: mdl-30267414

ABSTRACT

BACKGROUND: There is an increasing demand for daratumumab (DARA), an immunoglobulin (Ig)G1κ monoclonal antibody (MoAb) that recognizes CD38, to manage relapsed or refractory multiple myeloma (MM) patients. However, DARA leads to positive and panreactive agglutination reactions in indirect antiglobulin tests (IATs) in vitro (the DARA interference). In addition, effects of DARA on red blood cells (RBCs) in vivo remains elusive. STUDY DESIGN AND METHODS: To develop a new method to negate the DARA interference, the effects of various concentrations of dithiothreitol (DTT) on RBC CD38 and Kell antigenicity in combination with an automatic blood cell washing centrifuge were compared with the AABB standard procedure in parallel. Moreover, direct antiglobulin tests (DATs) for RBCs in DARA-treated MM patients were examined. RESULTS: A quantity of 0.01 mol/L DTT as well as the AABB procedure (equivalent to 0.15 mol/L DTT in our procedure) markedly reduced the reactivity of phycoerythrin-mouse anti-CD38 MoAb and DARA with RBCs. In sharp contrast to the AABB procedure, 0.01 mol/L DTT partially preserved K antigenicity and allowed the determination of phenotype of K antigen even in the presence of the DARA interference. In contrast, DAT for RBCs obtained from MM patients showed a weak positive or negative reaction. Immunoblotting further indicated that DARA induced loss of CD38 in vivo. CONCLUSION: A simple and reliable method to negate the DARA interference with partially preserving Kell antigenicity is proposed (Osaka method). CD38 antigenicity is susceptible to 0.01 mol/L DTT treatment even in the presence of DARA. Our data also demonstrate distinct effects of DARA on IAT in vitro and DAT in vivo.


Subject(s)
Antibodies, Monoclonal , Antigens, Bacterial/blood , Antigens, Surface/blood , Coombs Test/methods , Dithiothreitol/chemistry , Erythrocytes/metabolism , Multiple Myeloma , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Female , Humans , Male , Multiple Myeloma/blood , Multiple Myeloma/drug therapy
16.
Drug Metab Pharmacokinet ; 32(6): 301-310, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29174536

ABSTRACT

Decitabine (DAC), a nucleoside-related DNA methylation inhibitor, is taken up into cancer cells via equilibrative nucleoside transporter 1 (ENT1), and is then monophosphorylated by deoxycytidine kinase (dCK). In the present study, we examined the contribution of dCK to the uptake of DAC in HCT116 colon cancer cells. Irinotecan and etoposide inhibited the uptake of [3H]-uridine and [3H]-DAC at 10 s and 5 min, while cytarabine and gemcitabine only inhibited that of [3H]-DAC at 5 min. Irinotecan and etoposide inhibited [3H]-DAC uptake in negative control small interfering RNA (siRNA)- or dCK siRNA-transfected cells at 10 s, whereas cytarabine and gemcitabine did not. Cytarabine and gemcitabine inhibited DAC monophosphate generation by the cytosolic proteins of HCT116 cells and recombinant human dCK protein, assessed using polyethylenimine cellulose thin-layered chromatography. Simulations using simple kinetic models showed that apparent DAC uptake in dCK and ENT1 siRNA-treated cells was attributed to its conversion to monophosphates or a decrease in the cellular flux, respectively, and that the apparent uptake of DAC in dCK-knockdown and ENT1-knockdown cells was similar at longer times, but differed at a very short time. These results suggest that the apparent uptake of DAC is affected by ENT1 and dCK in HCT116 cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Azacitidine/analogs & derivatives , Colonic Neoplasms/drug therapy , Deoxycytidine Kinase/metabolism , Antimetabolites, Antineoplastic/metabolism , Azacitidine/antagonists & inhibitors , Azacitidine/metabolism , Azacitidine/pharmacokinetics , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Colonic Neoplasms/pathology , Cytarabine/pharmacology , Decitabine , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Equilibrative Nucleoside Transporter 1/metabolism , Etoposide/pharmacology , HCT116 Cells , Humans , Irinotecan , Phosphorylation , Gemcitabine
17.
Biol Pharm Bull ; 40(12): 2199-2204, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28954936

ABSTRACT

Limited information is currently available on how to apply epigenetic modifiers to current colorectal cancer (CRC) chemotherapy. The purpose of this study is to clarify the schedule-dependent effects of combined treatment with conventional anticancer drugs and epigenetic modifiers in human CRC cells. Cytotoxicity in 4 CRC cell lines (SW480, HT29, SW48, and HCT116) was measured using the WST-8 assay. As epigenetic modifiers, 3 DNA methyltransferase (DNMT) inhibitors such as decitabine (DAC), azacytidine (AC), and zebularine (Zeb), and 3 histone deacetylase (HDAC) inhibitors including trichostatin A (TSA), suberoylanilide hydroxamic acid (SAHA), and valproic acid (VPA) were used. Combination effects were analyzed by the isobologram method. SW480 cells showed the lowest sensitivity to the anticancer drugs 5-fluorouracil, SN-38 (the active form of irinotecan), and oxaliplatin. In SW480 cells, epigenetic modifiers other than VPA showed the most significant synergistic effects when used before anticancer drugs, while VPA showed synergistic effects in co- or post-treatment. In the 3 other CRC cells, synergistic effects were less frequent and weaker. The dose of anticancer drugs may be reduced by combining epigenetic modifiers in SW480 cells, which are less sensitive to anticancer drugs, unlike the more sensitive HT29, SW48, and HCT116 cell lines. These results provide useful information for understanding how to incorporate epigenetic modifiers into current CRC chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA Modification Methylases/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Synergism , Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Inhibitory Concentration 50
18.
Biol Pharm Bull ; 40(8): 1320-1325, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28539527

ABSTRACT

The effects of zebularine, a DNA methyltransferase inhibitor, on the invasion activity as well as intracellular expression level of let-7b, tumor suppressor microRNA, were examined in three human colorectal cancer (CRC) cell lines: SW480, SW620, and oxaliplatin-resistant SW620 (SW620/OxR). Zebularine suppressed the invasion activity of SW620 and SW620/OxR cells. The intracellular expression level of let-7b was up-regulated by zebularine in SW620 and SW620/OxR cells. The overexpression of let-7b by the transfection of let-7b mimic suppressed invasion activity in SW620 and SW620/OxR cells. These results suggest that zebularine may inhibit invasion activity by up-regulating the intracellular expression level of let-7b in high-invasive CRC cells.


Subject(s)
Colorectal Neoplasms/drug therapy , Cytidine/analogs & derivatives , Enzyme Inhibitors/pharmacology , MicroRNAs/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Cytidine/pharmacology , Cytidine/therapeutic use , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Drug Resistance, Neoplasm , Enzyme Inhibitors/therapeutic use , Exosomes/drug effects , Exosomes/metabolism , Humans , Neoplasm Invasiveness/prevention & control , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Transfection , Up-Regulation
19.
Oncol Lett ; 10(2): 761-767, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26622566

ABSTRACT

The aim of the present study was to determine the effects of long-term exposure of decitabine (DAC) to HCT116 colorectal cancer (CRC) cells on the acquisition of resistance to DAC as well as cross-resistance to anticancer drugs used for CRC or other epigenetic modifiers. In the present study, DAC-resistant HCT116 CRC cells were established through long-term treatment with increasing concentrations of DAC (10 to 540 nM); and the cross-resistance to other drugs was subsequently examined. DAC-resistant HCT116 cells were obtained following a 104-day treatment with DAC, including DAC-free intervals. The results demonstrated that the IC50 value of DAC was increased ~100-fold in DAC-resistant HCT116 cells. Messenger (m)RNA expression of secreted frizzed-related protein 1 (SFRP1), which is regulated by DNA methylation, was not detected in DAC-resistant cells; however, SFRP1 mRNA was present in HCT116 cells treated with DAC for 52 days. DNA methyltransferase 1 (DNMT1) protein levels were slightly decreased until day 81 and then returned to control levels in DAC-resistant cells. Further experiments using DAC-resistant HCT116 cells revealed that these cells exhibited cross-resistance to gemcitabine (Gem); however, cross-resistance was not observed for other DNMT inhibitors (azacitidine and zebularine), histone deacetylase inhibitors (trichostatin A, vorinostat and valproic acid) or anticancer drugs for CRC (5-fluorouracil, irinotecan and oxaliplatin). Furthermore, the protein expression levels of cytidine deaminase (CDA) were increased, while those of deoxycytidine kinase (dCK) were decreased in DAC-resistant HCT116 cells; by contrast, the mRNA expression levels for these proteins were not significantly altered. In conclusion, the results of the present study indicated that the long-term treatment of HCT116 cells with DAC led to the acquisition of resistance to both DAC and Gem. In addition, these results may be partly attributed to changes in CDA and/or dCK, which are involved in metabolic pathways common to these two drugs.

20.
Biol Pharm Bull ; 38(8): 1113-9, 2015.
Article in English | MEDLINE | ID: mdl-26235575

ABSTRACT

DNA hypermethylation, an epigenetic change that silences gene expression without altering nucleotide sequences, plays a critical role in the formation and progression of colorectal cancers as well as in the acquisition of drug resistance. Decitabine (DAC), a DNA methyltransferase 1 inhibitor of nucleoside analogues, has been shown to restore gene expression silenced by hypermethylation. In the present study, the mechanisms underlying both uridine and DAC uptake were examined in the human colon cancer cell line HCT116. Real-time polymerase chain reaction analysis revealed that ENT1 mRNA was the most abundant among the nucleoside transporters examined in HCT116 cells. The ENT1 protein was detected in the membrane fraction, as determined by Western blotting. The uptake of uridine or DAC was time- and concentration-dependent, but also Na(+)-independent. The uptake of these agents was inhibited by S-(4-nitrobenzyl)-6-thioinosine (NBMPR), an inhibitor of equilibrative nucleoside transporters (ENTs), and was also decreased in cells treated with ENT1 small interfering RNA. The uptake of both uridine and DAC was inhibited by uridine, cytidine, adenosine, or inosine, while that of DAC was also inhibited by thymidine. The expression of MAGEA1 mRNA, the DNA of which was methylated in HCT116 cells, was increased by DAC treatment, and this increment was attenuated by concomitant treatment with NBMPR. The IC50 value of DAC was also increased in the presence of NBMPR. These results suggest that DAC is mainly taken up by ENT1 and that this uptake is one of the key determinants of the activity of DAC in HCT116 cells.


Subject(s)
Azacitidine/analogs & derivatives , Colorectal Neoplasms/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/metabolism , Azacitidine/pharmacology , Azacitidine/therapeutic use , Carrier Proteins/metabolism , Colorectal Neoplasms/drug therapy , DNA Methylation , Decitabine , Gene Expression/drug effects , HCT116 Cells , Humans , Melanoma-Specific Antigens/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Uridine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...