Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Int J Mol Sci ; 24(4)2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36834962

ABSTRACT

Azacitidine (AZA) is commonly used hypomethylating agent for higher risk myelodysplastic syndromes and acute myeloid leukemia (AML). Although some patients achieve remission, eventually most patients fail AZA therapy. Comprehensive analysis of intracellular uptake and retention (IUR) of carbon-labeled AZA (14C-AZA), gene expression, transporter pump activity with or without inhibitors, and cytotoxicity in naïve and resistant cell lines provided insight into the mechanism of AZA resistance. AML cell lines were exposed to increasing concentrations of AZA to create resistant clones. 14C-AZA IUR was significantly lower in MOLM-13- (1.65 ± 0.08 ng vs. 5.79 ± 0.18 ng; p < 0.0001) and SKM-1- (1.10 ± 0.08 vs. 5.08 ± 0.26 ng; p < 0.0001) resistant cells compared to respective parental cells. Importantly, 14C-AZA IUR progressively reduced with downregulation of SLC29A1 expression in MOLM-13- and SKM-1-resistant cells. Furthermore, nitrobenzyl mercaptopurine riboside, an SLC29A inhibitor, reduced 14C-AZA IUR in MOLM-13 (5.79 ± 0.18 vs. 2.07 ± 0.23, p < 0.0001) and SKM-1-naive cells (5.08 ± 2.59 vs. 1.39 ± 0.19, p = 0.0002) and reduced efficacy of AZA. As the expression of cellular efflux pumps such as ABCB1 and ABCG2 did not change in AZA-resistant cells, they are unlikely contribute to AZA resistance. Therefore, the current study provides a causal link between in vitro AZA resistance and downregulation of cellular influx transporter SLC29A1.


Subject(s)
Azacitidine , Drug Resistance, Neoplasm , Equilibrative Nucleoside Transporter 1 , Leukemia, Myeloid, Acute , Humans , Azacitidine/pharmacology , Azacitidine/therapeutic use , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Down-Regulation , Drug Resistance, Neoplasm/genetics , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism
2.
Biomed Pharmacother ; 153: 113491, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36076585

ABSTRACT

Cordyceps militaris is rich in adenosine derivatives, including 3'-deoxyadenosine, also known as cordycepin. It has been reported for antitumor effects, but its underlying molecular mechanism has yet to be elucidated. We investigated how adenosine derivatives exerted antitumor effects against ovarian cancer using human ovarian cancer cells and a xenograft mouse model. Treatment with adenosine derivatives effectively resulted in cell death of ovarian cancer cells through AMPK activation and subsequently mTOR-mediated autophagic induction. Intriguingly, the effect required membrane transport of adenosine derivatives via ENT1, rather than ADORA-mediated cellular signaling. Our data suggest that adenosine derivatives may be an effective therapeutic intervention in ovarian cancer through induction of ENT1-AMPK-mTOR-mediated autophagic cell death.


Subject(s)
Adenosine , Autophagic Cell Death , Cordyceps , Ovarian Neoplasms , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Animals , Autophagic Cell Death/drug effects , Carcinoma, Ovarian Epithelial , Cordyceps/chemistry , Deoxyadenosines/pharmacology , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/metabolism , Female , Humans , Mice , Ovarian Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism
3.
Neuroreport ; 32(17): 1341-1348, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34718248

ABSTRACT

OBJECTIVES: Neuroinflammation and apoptosis are two key factors contributing to early brain injury (EBI) after subarachnoid hemorrhage (SAH) and are strongly associated with a poor prognosis. Recently, equilibrative nucleoside transporter 1 (ENT1) was emerged to accelerate the severity of inflammation and cell apoptosis in several nervous system diseases, including cerebral ischemia, neurodegeneration and epilepsy. However, no study has yet elaborated the expression levels and effects of ENT1 in EBI after SAH. METHODS: Sprague-Dawley rats were subjected to SAH by endovascular perforation. Nitrobenzylthioinosine (NBTI) was intranasally administered at 0.5 h after SAH. The protein expression levels of ENT1, NLRP3, Bcl2, Bax, ACS, Caspase-1, IL-1 were detected by western blot. The modified Garcia score and beam balance score were employed to evaluate the neurologic function of rats following SAH. In addition, hematoxylin-eosin, fluoro-jade C and TdT-mediated dUTP nick-end labeling staining were then used to evaluate brain tissue damage and neuronal apoptosis. RESULTS: Analysis indicated that endogenous levels of ENT1 were significantly upregulated at 24-hour post-SAH, accompanied by NLRP3 inflammasome activation and Bcl2 decline. The administration of NBTI, an inhibitor of ENT1, at a dose of 15 mg/kg, ameliorated neurologic deficits and morphologic lesions at both 24 and 72 h after SAH. Moreover, ENT1 inhibition efficiently mitigated neuronal degeneration and cell apoptosis. In addition, NBTI at 15 mg/kg observably increased Bcl2 content and decreased Bax level. Furthermore, suppression of ENT1 notably reduced the expression levels of NLRP3, apoptosis associated speck like protein containing CARD, caspase-1 and IL-1ß. CONCLUSIONS: NBTI relieved SAH-induced EBI partly through ENT1/NLRP3/Bcl2 pathway.


Subject(s)
Apoptosis/drug effects , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Neuroinflammatory Diseases/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Subarachnoid Hemorrhage/metabolism , Thioinosine/analogs & derivatives , Animals , Disease Models, Animal , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/metabolism , Inflammasomes/drug effects , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Thioinosine/pharmacology
4.
Eur J Pharmacol ; 885: 173504, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32858046

ABSTRACT

Leishmania infected macrophages have conditions to produce adenosine. Despite its known immunosuppressive effects, no studies have yet established whether adenosine alter Leishmania parasitic burden upon macrophage infection. This work aimed at investigating whether endogenous adenosine exerts an autocrine modulation of macrophage response towards Leishmania infection, identifying its origin and potential pharmacological targets for visceral leishmaniasis (VL), using THP-1 differentiated macrophages. Adenosine deaminase treatment of infected THP-1 cells reduced the parasitic burden (29.1 ± 2.2%, P < 0.05). Adenosine A2A and A2B receptor subtypes expression was confirmed by RT-qPCR and by immunocytochemistry and their blockade with selective adenosine A2A and A2B antagonists reduced the parasitic burden [14.5 ± 3.1% (P < 0.05) and 12.3 ± 3.1% (P < 0.05), respectively; and 24.9 ± 2.8% (P < 0.05), by the combination of the two antagonists)], suggesting that adenosine A2 receptors are tonically activated in infected THP-1 differentiated macrophages. The tonic activation of adenosine A2 receptors was dependent on the release of intracellular adenosine through equilibrative nucleoside transporters (ENT1/ENT2): NBTI or dipyridamole reduced (~25%) whereas, when ENTs were blocked, adenosine A2 receptor antagonists failed to reduce and A2 agonists increase parasitic burden. Effects of adenosine A2 receptors antagonists and ENT1/2 inhibitor were prevented by L-NAME, indicating that nitric oxide production inhibition prevents adenosine from increasing parasitic burden. Results suggest that intracellular adenosine, released through ENTs, elicits an autocrine increase in parasitic burden in THP-1 macrophages, through adenosine A2 receptors activation. These observations open the possibility to use well-established ENT inhibitors or adenosine A2 receptor antagonists as new therapeutic approaches in VL.


Subject(s)
Adenosine/metabolism , Autocrine Communication/drug effects , Leishmania infantum/drug effects , Leishmaniasis, Visceral/drug therapy , Macrophages/drug effects , Macrophages/parasitology , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2B/drug effects , THP-1 Cells/drug effects , Adenosine A2 Receptor Antagonists/pharmacology , Body Burden , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative-Nucleoside Transporter 2/drug effects , Humans , Leishmaniasis, Visceral/parasitology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors
5.
Clin Pharmacokinet ; 58(3): 283-297, 2019 03.
Article in English | MEDLINE | ID: mdl-29987837

ABSTRACT

Cladribine Tablets (MAVENCLAD®) are used to treat relapsing multiple sclerosis (MS). The recommended dose is 3.5 mg/kg, consisting of 2 annual courses, each comprising 2 treatment weeks 1 month apart. We reviewed the clinical pharmacology of Cladribine Tablets in patients with MS, including pharmacokinetic and pharmacometric data. Cladribine Tablets are rapidly absorbed, with a median time to reach maximum concentration (Tmax) of 0.5 h (range 0.5-1.5 h) in fasted patients. When administered with food, absorption is delayed (median Tmax 1.5 h, range 1-3 h), and maximum concentration (Cmax) is reduced by 29% (based on geometric mean). Area under the concentration-time curve (AUC) is essentially unchanged. Oral bioavailability of cladribine is approximately 40%, pharmacokinetics are linear and time-independent, and volume of distribution is 480-490 L. Plasma protein binding is 20%, independent of cladribine plasma concentration. Cladribine is rapidly distributed to lymphocytes and retained (either as parent drug or its phosphorylated metabolites), resulting in approximately 30- to 40-fold intracellular accumulation versus extracellular concentrations as early as 1 h after cladribine exposure. Cytochrome P450-mediated biotransformation of cladribine is of minor importance. Cladribine elimination is equally dependent on renal and non-renal routes. In vitro studies indicate that cladribine efflux is minimally P-glycoprotein (P-gp)-related, and clinically relevant interactions with P-gp inhibitors are not expected. Cladribine distribution across membranes is primarily facilitated by equilibrative nucleoside transporter (ENT) 1, concentrative nucleoside transporter (CNT) 3 and breast cancer resistance protein (BCRP), and there is no evidence of any cladribine-related effect on heart rate, atrioventricular conduction or cardiac repolarisation (QTc interval prolongation). Cladribine Tablets are associated with targeted lymphocyte reduction and durable efficacy, with the exposure-effect relationship showing the recommended dose is appropriate in reducing relapse risk.


Subject(s)
Cladribine/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Multiple Sclerosis, Relapsing-Remitting/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Administration, Oral , Adult , Aged , Biological Availability , Cladribine/administration & dosage , Cladribine/blood , Cladribine/therapeutic use , Cytochrome P-450 Enzyme System/metabolism , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/metabolism , Female , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , Immunosuppressive Agents/therapeutic use , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Middle Aged , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Pharmacology, Clinical , Protein Binding/drug effects
6.
Diabetes Obes Metab ; 20(12): 2748-2758, 2018 12.
Article in English | MEDLINE | ID: mdl-29962100

ABSTRACT

AIM: Recently we have observed differences in the ability of metformin and AICAR to repress glucose production from hepatocytes using 8CPT-cAMP. Previous results indicate that, in addition to activating protein kinase A, 8CPT-modified cAMP analogues suppress the nitrobenzylthioinosine (NBMPR)-sensitive equilibrative nucleoside transporter ENT1. We aimed to exploit 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR, which is highly selective for a high-affinity binding-site on ENT1, to investigate the role of ENT1 in the liver-specific glucose-lowering properties of AICAR and metformin. METHODS: Primary mouse hepatocytes were incubated with AICAR and metformin in combination with cAMP analogues, glucagon, forskolin and NBMPR. Hepatocyte glucose production (HGP) and AMPK signalling were measured, and a uridine uptake assay with supporting LC-MS was used to investigate nucleoside depletion from medium by cells. RESULTS: AICAR and metformin increased AMPK pathway phosphorylation and decreased HGP induced by dibutyryl cAMP and glucagon. HGP was also induced by 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR; however, in each case this was resistant to suppression by AICAR but not by metformin. Cross-validation of tracer and mass spectrometry studies indicates that 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR inhibited the effects of AICAR, at least in part, by impeding its uptake into hepatocytes. CONCLUSIONS: We report for the first time that suppression of ENT1 induces HGP. ENT1 inhibition also impedes uptake and the effects of AICAR, but not metformin, on HGP. Further investigation of nucleoside transport may illuminate a better understanding of how metformin and AICAR each regulate HGP.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Equilibrative Nucleoside Transporter 1/drug effects , Glucose/metabolism , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacokinetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacokinetics , Animals , Biological Transport/drug effects , Female , Liver/metabolism , Metformin/pharmacokinetics , Mice , Phosphorylation/drug effects , Ribonucleotides/pharmacokinetics , Signal Transduction/drug effects , Thioinosine/analogs & derivatives , Thioinosine/metabolism
7.
Cardiovasc Res ; 113(1): 61-69, 2017 01.
Article in English | MEDLINE | ID: mdl-28028070

ABSTRACT

AIMS: The P2Y12 antagonist ticagrelor reduces mortality in patients with acute coronary syndrome (ACS), compared with clopidogrel, and the mechanisms underlying this effect are not clearly understood. Arterial thrombosis is the key event in ACS; however, direct vascular effects of either ticagrelor or clopidogrel with focus on arterial thrombosis and its key trigger tissue factor have not been previously investigated. METHODS AND RESULTS: Human aortic endothelial cells were treated with ticagrelor or clopidogrel active metabolite (CAM) and stimulated with tumour necrosis factor-alpha (TNF-α); effects on procoagulant tissue factor (TF) expression and activity, its counter-player TF pathway inhibitor (TFPI) and the underlying mechanisms were determined. Further, arterial thrombosis by photochemical injury of the common carotid artery, and TF expression in the murine endothelium were examined in C57BL/6 mice treated with ticagrelor or clopidogrel. Ticagrelor, but not CAM, reduced TNF-α-induced TF expression via proteasomal degradation and TF activity, independently of the P2Y12 receptor and the equilibrative nucleoside transporter 1 (ENT1), an additional target of ticagrelor. In C57BL/6 mice, ticagrelor prolonged time to arterial occlusion, compared with clopidogrel, despite comparable antiplatelet effects. In line with our in vitro results, ticagrelor, but not clopidogrel, reduced TF expression in the endothelium of murine arteries. CONCLUSION: Ticagrelor, unlike clopidogrel, exhibits endothelial-specific antithrombotic properties and blunts arterial thrombus formation. The additional antithrombotic properties displayed by ticagrelor may explain its greater efficacy in reducing thrombotic events in clinical trials. These findings may provide the basis for new indications for ticagrelor.


Subject(s)
Adenosine/analogs & derivatives , Blood Coagulation/drug effects , Carotid Artery Injuries/drug therapy , Endothelial Cells/drug effects , Fibrinolytic Agents/pharmacology , Thromboplastin/metabolism , Thrombosis/prevention & control , Ticlopidine/analogs & derivatives , Adenosine/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Carotid Artery Injuries/blood , Carotid Artery Injuries/genetics , Cells, Cultured , Clopidogrel , Disease Models, Animal , Down-Regulation , Endothelial Cells/metabolism , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/metabolism , Humans , Male , Mice, Inbred C57BL , Platelet Aggregation Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/metabolism , Thromboplastin/genetics , Thrombosis/blood , Thrombosis/genetics , Ticagrelor , Ticlopidine/pharmacology , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
8.
Microvasc Res ; 95: 68-75, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24976360

ABSTRACT

Purine nucleosides and nucleobases play key roles in the physiological response to vascular ischemia/reperfusion events. The intra- and extracellular concentrations of these compounds are controlled, in part, by equilibrative nucleoside transporter subtype 1 (ENT1; SLC29A1) and by equilibrative nucleobase transporter subtype 1 (ENBT1). These transporters are expressed at the membranes of numerous cell types including microvascular endothelial cells. We studied the impact of reactive oxygen species on the function of ENT1 and ENBT1 in primary (CMVEC) and immortalized (HMEC-1) human microvascular endothelial cells. Both cell types displayed similar transporter expression profiles, with the majority (>90%) of 2-chloro[(3)H]adenosine (nucleoside) uptake mediated by ENT1 and [(3)H]hypoxanthine (nucleobase) uptake mediated by ENBT1. An in vitro mineral oil-overlay model of ischemia/reperfusion had no effect on ENT1 function, but significantly reduced ENBT1 Vmax in both cell types. This decrease in transport function was mimicked by the intracellular superoxide generator menadione and could be reversed by the superoxide dismutase mimetic MnTMPyP. In contrast, neither the extracellular peroxide donor TBHP nor the extracellular peroxynitrite donor 3-morpholinosydnonimine (SIN-1) affected ENBT1-mediated [(3)H]hypoxanthine uptake. SIN-1 did, however, enhance ENT1-mediated 2-chloro[(3)H]adenosine uptake. Our data establish HMEC-1 as an appropriate model for study of purine transport in CMVEC. Additionally, these data suggest that the generation of intracellular superoxide in ischemia/reperfusion leads to the down-regulation of ENBT1 function. Modification of purine transport by oxidant stress may contribute to ischemia/reperfusion induced vascular damage and should be considered in the development of therapeutic strategies.


Subject(s)
Endothelial Cells/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Equilibrative-Nucleoside Transporter 2/metabolism , Microvessels/metabolism , Oxidative Stress , Purines/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Antioxidants/pharmacology , Biological Transport , Cell Hypoxia , Cell Line , Endothelial Cells/drug effects , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative-Nucleoside Transporter 2/drug effects , Humans , Hypoxanthine/metabolism , Microvessels/drug effects , Oxidants/pharmacology , Oxidative Stress/drug effects , Reperfusion Injury/metabolism , Superoxides/metabolism , Time Factors
9.
Asian Pac J Trop Med ; 6(11): 908-11, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24083589

ABSTRACT

OBJECTIVE: To investigate the expression of hENTl and ERCC1 genes in tumor tissues non-small cell lung cancer (NSCLC). METHODS: Fresh non-small lung cancer specimens were transplanted into nude mice. Twenty mice were randomized into two groups: experimental group receiving gemcitabine plus cisplatin and control group receiving 0.9% physiological saline. The expressions of hENTl and ERCC1 mRNA in tumor tissue were detected by real-time fluorescent quantitative PCR. The volume of tumor, the weight of nude mice and tumor volume were respectively measured and calculated 2-3 times per week. Tissue samples were collected from NSCLC mice treated with gemcitabine plus carboplatin. RESULTS: The histological examination showed that many tumor cells were well preserved in nude mice. The rate of transplanted tumor cells was 86.7%. The concomitant treatment study showed that the rate of TV, RTV, T/C in GEM + DDP group was the lowest. LBP + DOC, DDP + DOC obviously influenced the body weight. Compared with NS group, DDP group, GEM group, the survival period and the level of hENTl of DDP+GEM group increased obviously, the level of ERCC1 decreased significantly (P<0.05). CONCLUSIONS: The expression of hENT1 and ERCC1 genes in tumor tissues were closely correlated with the response to chemotherapy and prognosis of patients with NSCLC treated with gemcitabine plus cisplatin.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Lung Neoplasms/pathology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/administration & dosage , DNA-Binding Proteins/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Endonucleases/drug effects , Equilibrative Nucleoside Transporter 1/drug effects , Lung Neoplasms/drug therapy , Mice , RNA, Messenger/metabolism , Survival Rate , Tumor Burden/drug effects , Gemcitabine
10.
Braz J Med Biol Res ; 39(11): 1417-23, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17146554

ABSTRACT

Infant acute lymphoblastic leukemia (IALL) is characterized by mixed lineage leukemia (MLL) gene rearrangements, unique gene expression profiles, poor prognosis, and drug resistance. One exception is cytosine arabinoside (Ara-C) to which IALL cells seem to be more sensitive. We quantified mRNA expression of Ara-C key enzymes in leukemic lymphoblasts from 64 Brazilian ALL children, 15 of them presenting MLL gene rearrangement, and correlated it with clinical and biological features. The diagnosis was based on morphological criteria and immunophenotyping using monoclonal antibodies. MLL gene rearrangements were detected by conventional cytogenetic analysis, RT-PCR and/or fluorescence in situ hybridization. The DCK and HENT1 expression levels were determined by real-time quantitative PCR using SYBR Green I. Relative quantification was made by the standard curve method. The results were analyzed by Mann-Whitney and Fisher exact tests. A P value of

Subject(s)
Antimetabolites, Antineoplastic/metabolism , Cytarabine/metabolism , Gene Rearrangement/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Antimetabolites, Antineoplastic/therapeutic use , Case-Control Studies , Child , Child, Preschool , Cytarabine/therapeutic use , Deoxycytidine Kinase/drug effects , Deoxycytidine Kinase/genetics , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Male , Myeloid-Lymphoid Leukemia Protein/drug effects , Neoplasm, Residual , Polymerase Chain Reaction/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , RNA, Messenger/analysis , Time Factors
11.
Braz. j. med. biol. res ; 39(11): 1417-1423, Nov. 2006. graf, tab
Article in English | LILACS | ID: lil-437833

ABSTRACT

Infant acute lymphoblastic leukemia (IALL) is characterized by mixed lineage leukemia (MLL) gene rearrangements, unique gene expression profiles, poor prognosis, and drug resistance. One exception is cytosine arabinoside (Ara-C) to which IALL cells seem to be more sensitive. We quantified mRNA expression of Ara-C key enzymes in leukemic lymphoblasts from 64 Brazilian ALL children, 15 of them presenting MLL gene rearrangement, and correlated it with clinical and biological features. The diagnosis was based on morphological criteria and immunophenotyping using monoclonal antibodies. MLL gene rearrangements were detected by conventional cytogenetic analysis, RT-PCR and/or fluorescence in situ hybridization. The DCK and HENT1 expression levels were determined by real-time quantitative PCR using SYBR Green I. Relative quantification was made by the standard curve method. The results were analyzed by Mann-Whitney and Fisher exact tests. A P value of ú0.05 was considered to be statistically significant. DCK and HENT1 expression levels were significantly lower in children with MLL gene-rearranged ALL compared to children with MLL germ line ALL (P = 0.0003 and 0.03, respectively). Our results differ from previous ones concerning HENT1 mRNA expression that observed a higher expression level in MLL gene-rearranged leukemias. In conclusion, the expression of the genes related to Ara-C metabolism was lower in MLL-positive children in the sample studied, suggesting the presence of population differences in the expression profile of these genes especially for HENT1.


Subject(s)
Humans , Male , Female , Infant , Child, Preschool , Child , Antimetabolites, Antineoplastic/therapeutic use , Cytarabine/therapeutic use , Gene Rearrangement/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Case-Control Studies , Deoxycytidine Kinase/drug effects , Deoxycytidine Kinase/genetics , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/genetics , Gene Expression Regulation, Neoplastic , Myeloid-Lymphoid Leukemia Protein/drug effects , Neoplasm, Residual , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Polymerase Chain Reaction/methods , RNA, Messenger/analysis , Time Factors
12.
Br J Cancer ; 95(3): 289-97, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16868547

ABSTRACT

The present study was performed to investigate the capability of gemcitabine and pemetrexed to synergistically interact with respect to cytotoxicity and apoptosis in T24 and J82 bladder cancer cells, and to establish a correlation between drug activity and gene expression of selected genes in tumour samples. The interaction between gemcitabine and pemetrexed was synergistic; indeed, pemetrexed favoured gemcitabine cytotoxicity by increasing cellular population in S-phase, reducing Akt phosphorylation as well as by inducing the expression of a major gemcitabine uptake system, the human equilibrative nucleoside transporter-1 (hENT1), and the key activating enzyme deoxycytidine kinase (dCK) in both cell lines. Bladder tumour specimens showed an heterogeneous gene expression pattern and patients with higher levels of dCK and hENT1 had better response. Moreover, human nucleoside concentrative transporter-1 was detectable only in 3/12 patients, two of whom presented a complete response to gemcitabine. These data provide evidence that the chemotherapeutic activity of the combination of gemcitabine and pemetrexed is synergistic against bladder cancer cells in vitro and that the assessment of the expression of genes involved in gemcitabine uptake and activation might be a possible determinant of bladder cancer response and may represent a new tool for treatment optimization.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Glutamates/administration & dosage , Guanine/analogs & derivatives , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Administration, Intravesical , Adult , Aged , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/antagonists & inhibitors , Deoxycytidine/pharmacology , Deoxycytidine Kinase/antagonists & inhibitors , Deoxycytidine Kinase/genetics , Dipyridamole/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative Nucleoside Transporter 1/genetics , Excitatory Amino Acid Antagonists , Female , Gene Expression Regulation, Neoplastic/drug effects , Glutamates/pharmacology , Guanine/administration & dosage , Guanine/antagonists & inhibitors , Guanine/pharmacology , Humans , Male , Middle Aged , Neoplasm Staging , Pemetrexed , Phosphoribosylglycinamide Formyltransferase/antagonists & inhibitors , Phosphoribosylglycinamide Formyltransferase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , S Phase/drug effects , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/genetics , Treatment Outcome , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...