Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Environ Sci Pollut Res Int ; 30(29): 74386-74397, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37209333

ABSTRACT

In the context of the COVID-19 pandemic, antiviral drugs (AVDs) were heavily excreted into wastewater and subsequently enriched in sewage sludge due to their widespread use. The potential ecological risks of AVDs have attracted increasing attention, but information on the effects of AVDs on sludge anaerobic digestion (AD) is limited. In this study, two typical AVDs (lamivudine and ritonavir) were selected to investigate the responses of AD to AVDs by biochemical methane potential tests. The results indicated that the effects of AVDs on methane production from sludge AD were dose- and type-dependent. The increased ritonavir concentration (0.05-50 mg/kg TS) contributed to an 11.27-49.43% increase in methane production compared with the control. However, methane production was significantly decreased at high lamivudine doses (50 mg/kg TS). Correspondingly, bacteria related to acidification were affected when exposed to lamivudine and ritonavir. Acetoclastic and hydrotropic methanogens were inhibited at a high lamivudine dose, while ritonavir enriched methylotrophic and hydrotropic methanogens. Based on the analysis of intermediate metabolites, the inhibition of lamivudine and the promotion of ritonavir on acidification and methanation were confirmed. In addition, the existence of AVDs could affect sludge properties. Sludge solubilization was inhibited when exposed to lamivudine and enhanced by ritonavir, perhaps caused by their different structures and physicochemical properties. Moreover, lamivudine and ritonavir could be partially degraded by AD, but 50.2-68.8% of AVDs remained in digested sludge, implying environmental risks.


Subject(s)
COVID-19 , Sewage , Humans , Sewage/chemistry , Anaerobiosis , Biofuels , Waste Disposal, Fluid/methods , Antiviral Agents/pharmacology , Ritonavir , Lamivudine/metabolism , Pandemics , Methane/metabolism , Bioreactors
2.
Cell Death Dis ; 13(4): 336, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35410316

ABSTRACT

Retinal endothelial cells (RECs) are the primary target cells for diabetes-induced vascular damage. The P2X7/NLRP3 pathway plays an essential role in amplifying inflammation via an ATP feedback loop, promoting the inflammatory response, pyroptosis, and apoptosis of RECs in the early stages of diabetic retinopathy induced by hyperglycemia and inflammation. 3TC, a type of nucleoside reverse transcriptase inhibitor, is effective against inflammation, as it can targeting formation of the P2X7 large pore formation. Hence, our aim was to evaluated the anti-inflammatory effects and potential mechanisms of action of 3TC in vitro in retinal microvascular endothelial cells treated with high-glucose (HG) and lipopolysaccharide (LPS), as well as in vivo in the retinas of C57BL/6J male mice with streptozotocin-induced diabetes. The expression of inflammasome-related proteins P2X7 and NLRP3, and apoptosis in the retinas of 3TC-treated diabetic mice were compared to those of untreated diabetic mice. Furthermore, the anti-inflammatory, anti-apoptotic, and anti-pyroptotic effects of 3TC were evaluated in vitro in cultured mice retinal endothelial cells. Co-application of HG and LPS significantly increased the secretion of IL-6, IL-1ß, and TNF-α, and ATP levels, whereas 3TC decreased cell inflammation, apoptosis, and pyroptosis. Inhibition of P2X7R and NLRP3 inflammasome activation decreased NLRP3 inflammasome-mediated injury. 3TC prevented cytokine and ATP release following co-application of HG and LPS/BzATP. Our findings provide new insights regarding the mechanisms of action of 3TC in diabetic environment-induced retinal injury, including apoptosis and pyroptosis.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Adenosine Triphosphate/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Apoptosis , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Lamivudine/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , Signal Transduction
3.
Ann Pharm Fr ; 80(6): 864-875, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35231396

ABSTRACT

BACKGROUND: Lamivudine and tenofovir disoproxil fumarate act against the replication of hepatitis B and human immunodeficiency viruses via inhibition of the reverse transcriptase enzyme activity, thereby preventing the synthesis of viral DNA. Chronic administration of these drugs has been associated with toxicities, including senescence, oxidative stress and premature death. A study of these toxicities in Drosophila melanogaster, which share 75% genomic similarity with humans could help to develop a pharmacologic intervention. METHODS: Susceptibility of D. melanogaster for lamivudine and tenofovir-induced toxicities were investigated. First, flies (≤3 days old) were fed with drugs-supplemented diet at varying concentrations (1mg to 300mg/10-gram diet) or distilled water for seven days to determine LD50. Secondly, five groups of 60 flies were fed with four concentrations of test drugs: 2.9mg, 5.82mg, 11.64mg and 23.28mg each per 10-gram diet for 28 days survival and lifespan assays. Then 5-day treatment plan was utilized to determine drugs toxicities on climbing ability and some biomarkers of oxidative stress. Finally, molecular docking was carried out using the Auto-dock vina mode to predict the biological interactions between the test drugs and D. melanogaster acetylcholinesterase (AChE) or glutathione-S-transferase (GST). RESULTS: The LD50 of lamivudine or tenofovir was 47.07 or 43.95mg/10g diet, respectively. Each drug significantly (P<0.05) reduced the survival rate, longevity and climbing performance of the flies dose-dependently. These drugs also altered levels of biochemical parameters: AChE, GST, superoxide dismutase (SOD), catalase (CAT), total thiol (T-SH), and malondialdehyde (MDA) of the flies significantly (P<0.05). In silico molecular analysis showed that the test drugs interacted with significantly (P<0.05) higher binding affinities at the same catalytic sites of D. melanogaster GST and AChE compared with substrates (glutathione or acetylcholine). CONCLUSION: The significant lamivudine and tenofovir-induced toxicities observed as increased mortality, climbing deficits and compromised antioxidant defence in D. melanogaster demands further research for possible pharmacological intervention.


Subject(s)
Antioxidants , Drosophila melanogaster , Animals , Humans , Acetylcholine/metabolism , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Antioxidants/pharmacology , Biomarkers , Catalase/genetics , Catalase/metabolism , DNA, Viral/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Glutathione , Glutathione Transferase/metabolism , Lamivudine/toxicity , Lamivudine/metabolism , Malondialdehyde/metabolism , Molecular Docking Simulation , Oxidative Stress , RNA-Directed DNA Polymerase/metabolism , Sulfhydryl Compounds , Superoxide Dismutase/metabolism , Tenofovir/toxicity , Tenofovir/metabolism
4.
BMC Infect Dis ; 21(1): 595, 2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34157984

ABSTRACT

BACKGROUND: We aimed to assess the overall cardiovascular and metabolic effect of the switch to three different single tablet regimens (STRs) [tenofovir alafenamide/emtricitabine/rilpivirine (TAF/FTC/RPV), TAF/FTC/elvitegravir/cobi (TAF/FTC/EVG/cobi) and ABC/lamivudine/dolutegravir (ABC/3TC/DTG)] in a cohort of people living with HIV/AIDS (PLWH) under effective ART. METHODS: All PLWH aged above 18 years on antiretroviral treatment with an HIV-RNA < 50 cp/mL at the time of the switch to TAF/FTC/RPV, TAF/FTC/EVG/cobi and ABC/3TC/DTG were retrospectively included in the analysis. Framingham risk score modification after 12 months from the switch such as lipid profile and body weight modification were assessed. The change from baseline to 12 months in mean cardiovascular risk and body weight in each of the STR's group were assessed by means of Wilcoxon signed-rank test whereas a mixed regression model was used to assess variation in lipid levels. RESULTS: Five-hundred and sixty PLWH were switched to an STR regimen of whom 170 (30.4%) to TAF/FTC/EVG/cobi, 191 (34.1%) to TAF/FTC/RPV and 199 (35.5%) to ABC/3TC/DTG. No difference in the Framingham cardiovascular risk score was observed after 12 months from the switch in each of the STR's groups. No significant overtime variation in mean total cholesterol levels from baseline to 12 months was observed for PLWH switched to ABC/3TC/DTG [200 (SD 38) mg/dl vs 201 (SD 35) mg/dl; p = 0.610] whereas a significant increment was observed in PLWH switched to TAF/FTC/EVG/cobi [192 (SD 34) mg/dl vs 208 (SD 40) mg/dl; p < 0.0001] and TAF/FTC/RPV [187 (SD 34) mg/dl vs 195 (SD 35) mg/dl; p = 0.027]. In addition, a significant variation in the mean body weight from baseline to 12 months was observed in PLWH switched to TAF/FTC/EVG/cobi [72.2 (SD 13.5) kilograms vs 74.6 (SD 14.3) kilograms; p < 0.0001] and TAF/FTC/RPV [73.4 (SD 11.6) kilograms vs 75.6 (SD 11.8) kilograms; p < 0.0001] whereas no difference was observed in those switched to ABC/3TC/DTG [71.5 (SD 12.8) kilograms vs 72.1 (SD 12.6) kilograms; p = 0.478]. CONCLUSION: No difference in the cardiovascular risk after 1 year from the switch to these STRs were observed. PLWH switched to TAF/FTC/EVG/cobi and TAF/FTC/RPV showed an increase in total cholesterol levels and body weight 12 months after the switch.


Subject(s)
Anti-HIV Agents/therapeutic use , Dideoxynucleosides/therapeutic use , Elvitegravir, Cobicistat, Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination/therapeutic use , Emtricitabine, Rilpivirine, Tenofovir Drug Combination/therapeutic use , HIV Infections/drug therapy , Heterocyclic Compounds, 3-Ring/therapeutic use , Lamivudine/therapeutic use , Oxazines/therapeutic use , Piperazines/therapeutic use , Pyridones/therapeutic use , Adult , Anti-HIV Agents/metabolism , Body Weight/drug effects , Cohort Studies , Dideoxynucleosides/metabolism , Drug Combinations , Elvitegravir, Cobicistat, Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination/metabolism , Emtricitabine, Rilpivirine, Tenofovir Drug Combination/metabolism , Female , Heart Disease Risk Factors , Heterocyclic Compounds, 3-Ring/metabolism , Humans , Italy/epidemiology , Lamivudine/metabolism , Lipid Metabolism/drug effects , Lipids/blood , Male , Middle Aged , Oxazines/metabolism , Piperazines/metabolism , Pyridones/metabolism , Retrospective Studies , Tablets/therapeutic use
5.
J Virol ; 95(16): e0240120, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34076480

ABSTRACT

Entecavir (ETV) is a widely used anti-hepatitis B virus (HBV) drug. However, the emergence of resistant mutations in HBV reverse transcriptase (RT) results in treatment failure. To understand the mechanism underlying the development of ETV resistance by HBV RT, we analyzed the L180M, M204V, and L180M/M204V mutants using a combination of biochemical and structural techniques. ETV-triphosphate (ETV-TP) exhibited competitive inhibition with dGTP in both wild-type (wt) RT and M204V RT, as observed using Lineweaver-Burk plots. In contrast, RT L180M or L180M/M204V did not fit either competitive, uncompetitive, noncompetitive, or typical mixed inhibition, although ETV-TP was a competitive inhibitor of dGTP. Crystallography of HIV RTY115F/F116Y/Q151M/F160M/M184V, mimicking HBV RT L180M/M204V, showed that the F115 bulge (F88 in HBV RT) caused by the F160M mutation induced deviated binding of dCTP from its normal tight binding position. Modeling of ETV-TP on the deviated dCTP indicated that a steric clash could occur between ETV-TP methylene and the 3'-end nucleoside ribose. ETV-TP is likely to interact primarily with HBV RT M171 prior to final accommodation at the deoxynucleoside triphosphate (dNTP) binding site (Y. Yasutake, S. Hattori, H. Hayashi, K. Matsuda, et al., Sci Rep 8:1624, 2018, https://doi.org/10.1038/s41598-018-19602-9). Therefore, in HBV RT L180M/M204V, ETV-TP may be stuck at M171, a residue that is conserved in almost all HBV isolates, leading to the strange inhibition pattern observed in the kinetic analysis. Collectively, our results provide novel insights into the mechanism of ETV resistance of HBV RT caused by L180M and M204V mutations. IMPORTANCE HBV infects 257 million people in the world, who suffer from elevated risks of liver cirrhosis and cancer. ETV is one of the most potent anti-HBV drugs, and ETV resistance mutations in HBV RT have been extensively studied. Nevertheless, the mechanisms underlying ETV resistance have remained elusive. We propose an attractive hypothesis to explain ETV resistance and effectiveness using a combination of kinetic and structural analyses. ETV is likely to have an additional interaction site, M171, beside the dNTP pocket of HBV RT; this finding indicates that nucleos(t)ide analogues (NAs) recognizing multiple interaction sites within RT may effectively inhibit the enzyme. Modification of ETV may render it more effective and enable the rational design of efficient NA inhibitors.


Subject(s)
Drug Resistance, Viral/genetics , Guanine/analogs & derivatives , Hepatitis B virus/drug effects , RNA-Directed DNA Polymerase/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Binding Sites , Crystallography, X-Ray , Deoxycytosine Nucleotides/metabolism , Deoxyguanine Nucleotides/metabolism , Guanine/metabolism , Guanine/pharmacology , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Hepatitis B virus/chemistry , Hepatitis B virus/enzymology , Inhibitory Concentration 50 , Kinetics , Lamivudine/metabolism , Lamivudine/pharmacology , Mutation , RNA-Directed DNA Polymerase/genetics , RNA-Directed DNA Polymerase/metabolism , Reverse Transcriptase Inhibitors/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
6.
Chin J Nat Med ; 17(9): 682-689, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31526503

ABSTRACT

Stellera chamaejasme L. is a traditional Chinese medicine with a long history to treat stubborn skin ulcer, and it also has antiviral and antitumor effects. Neochamaejasmine B (NCB), Neochamaejasmine A (NCA) and Chamaechromone (CMC) are the major components in dried roots of Stellera chamaejasme L.. Our studies suggested that NCB, NCA and CMC are inhibitors of Organic anion transporter 1 (OAT1). OAT1 is encoded by solute carrier family 22 member 6 gene (SLC22A6) in humans and plays a critical role in the organic anion drug uptake and excretion in the kidney. Lamivudine is the typical substrate of OAT1 and is frequently used in combination with other antiviral drugs in clinical antiviral treatments. The aim of this study is to investigate the interaction and its mechanism between these bi-flavone components in Stellera chamaejasme L. and lamivudine via OAT1 both in vitro and in vivo. In vitro, the uptake studies in Madin-Darby canine kidney (MDCK) cells overexpressing OAT1 suggested that NCB inhibited the uptake of 6-CFL and lamivudine.Similar results were obtained for NCA and CMC. NCB was a noncompetitive and competitive inhibitor interaction with OAT1. IC50 values of NCB, NCA and CMC for inhibiting OAT1-mediated lamivudine transport were 2.46, 8.35 and 0.61 µmol·L-1, respectively. In vivo, the pharmacokinetic results of lamivudine in rats showed that the mean area under the plasma concentration-time curve (AUC0-∞) and maximal plasma concentration (Cmax) of lamivudine after co-administration is increased 2.94-fold and 1.87-fold, respectively, compared to lamivudine administration alone. The results of interactions between lamivudine and these bi-flavone components in Stellera chamaejasme L. extracts via OAT1 in vivo are consistent with studies in vitro. The inhibition of OAT1-mediated uptake of lamivudine by NCB, NCA and CMC is the possible mechanism for Stellera chamaejasme L. extracts improving the oral bioavailability of lamivudine in rats.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Flavonoids/pharmacology , Lamivudine/metabolism , Organic Anion Transport Protein 1/antagonists & inhibitors , Thymelaeaceae/chemistry , Animals , Biflavonoids/pharmacology , Biological Availability , Biological Transport/drug effects , Dogs , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/chemistry , Female , Flavones/pharmacology , Flavonoids/chemistry , Humans , Inhibitory Concentration 50 , Lamivudine/pharmacokinetics , Madin Darby Canine Kidney Cells , Male , Organic Anion Transport Protein 1/metabolism , Rats, Sprague-Dawley
7.
Article in English | MEDLINE | ID: mdl-28696229

ABSTRACT

Rilpivirine (TMC278) is a highly potent nonnucleoside reverse transcriptase inhibitor (NNRTI) representing an effective component of combination antiretroviral therapy (cART) in the treatment of HIV-positive patients. Many antiretroviral drugs commonly used in cART are substrates of ATP-binding cassette (ABC) and/or solute carrier (SLC) drug transporters and, therefore, are prone to pharmacokinetic drug-drug interactions (DDIs). The aim of our study was to evaluate rilpivirine interactions with abacavir and lamivudine on selected ABC and SLC transporters in vitro and assess its importance for pharmacokinetics in vivo Using accumulation assays in MDCK cells overexpressing selected ABC or SLC drug transporters, we revealed rilpivirine as a potent inhibitor of MDR1 and BCRP, but not MRP2, OCT1, OCT2, or MATE1. Subsequent transport experiments across monolayers of MDCKII-MDR1, MDCKII-BCRP, and Caco-2 cells demonstrated that rilpivirine inhibits MDR1- and BCRP-mediated efflux of abacavir and increases its transmembrane transport. In vivo experiments in male Wistar rats confirmed inhibition of MDR1/BCRP in the small intestine, leading to a significant increase in oral bioavailability of abacavir. In conclusion, rilpivirine inhibits MDR1 and BCRP transporters and may affect pharmacokinetic behavior of concomitantly administered substrates of these transporters, such as abacavir.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Dideoxynucleosides/metabolism , Drug Interactions/physiology , Intestinal Absorption/physiology , Membrane Transport Proteins/metabolism , Rilpivirine/metabolism , Animals , Biological Transport/physiology , Caco-2 Cells , Cell Line , Cell Line, Tumor , Dideoxynucleosides/pharmacology , Dogs , Humans , Lamivudine/metabolism , Lamivudine/pharmacology , Madin Darby Canine Kidney Cells , Male , Rats , Rats, Wistar , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Rilpivirine/pharmacology
8.
Nucleic Acids Res ; 45(10): 6228-6237, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28402499

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) with L-stereochemistry have long been an effective treatment for viral infections because of the strong D-stereoselectivity exhibited by human DNA polymerases relative to viral reverse transcriptases. The D-stereoselectivity of DNA polymerases has only recently been explored structurally and all three DNA polymerases studied to date have demonstrated unique stereochemical selection mechanisms. Here, we have solved structures of human DNA polymerase ß (hPolß), in complex with single-nucleotide gapped DNA and L-nucleotides and performed pre-steady-state kinetic analysis to determine the D-stereoselectivity mechanism of hPolß. Beyond a similar 180° rotation of the L-nucleotide ribose ring seen in other studies, the pre-catalytic ternary crystal structures of hPolß, DNA and L-dCTP or the triphosphate forms of antiviral drugs lamivudine ((-)3TC-TP) and emtricitabine ((-)FTC-TP) provide little structural evidence to suggest that hPolß follows the previously characterized mechanisms of D-stereoselectivity. Instead, hPolß discriminates against L-stereochemistry through accumulation of several active site rearrangements that lead to a decreased nucleotide binding affinity and incorporation rate. The two NRTIs escape some of the active site selection through the base and sugar modifications but are selected against through the inability of hPolß to complete thumb domain closure.


Subject(s)
DNA Polymerase beta/metabolism , Reverse Transcriptase Inhibitors/metabolism , Catalysis , Catalytic Domain , Crystallography, X-Ray , DNA Polymerase beta/chemistry , DNA Polymerase beta/genetics , Deoxycytosine Nucleotides/metabolism , Emtricitabine/chemistry , Emtricitabine/metabolism , Humans , Kinetics , Lamivudine/chemistry , Lamivudine/metabolism , Models, Molecular , Molecular Conformation , Mutation, Missense , Protein Binding , Protein Conformation , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Stereoisomerism , Structure-Activity Relationship , Substrate Specificity
9.
J Am Chem Soc ; 139(1): 465-471, 2017 01 11.
Article in English | MEDLINE | ID: mdl-27959534

ABSTRACT

DNA polymerases are essential enzymes that faithfully and efficiently replicate genomic information.1-3 The mechanism of nucleotide incorporation by DNA polymerases has been extensively studied structurally and kinetically, but several key steps following phosphodiester bond formation remain structurally uncharacterized due to utilization of natural nucleotides. It is thought that the release of pyrophosphate (PPi) triggers reverse conformational changes in a polymerase in order to complete a full catalytic cycle as well as prepare for DNA translocation and subsequent incorporation events. Here, by using the triphosphates of chain-terminating antiviral drugs lamivudine ((-)3TC-TP) and emtricitabine ((-)FTC-TP), we structurally reveal the correct sequence of post-chemistry steps during nucleotide incorporation by human DNA polymerase ß (hPolß) and provide a structural basis for PPi release. These post-catalytic structures reveal hPolß in an open conformation with PPi bound in the active site, thereby strongly suggesting that the reverse conformational changes occur prior to PPi release. The results also help to refine the role of the newly discovered third divalent metal ion for DNA polymerase-catalyzed nucleotide incorporation. Furthermore, a post-chemistry structure of hPolß in the open conformation, following incorporation of (-)3TC-MP, with a second (-)3TC-TP molecule bound to the active site in the absence of PPi, suggests that nucleotide binding stimulates PPi dissociation and occurs before polymerase translocation. Our structural characterization defines the order of the elusive post-chemistry steps in the canonical mechanism of a DNA polymerase.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Nucleotides/metabolism , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Biocatalysis , DNA-Directed DNA Polymerase/chemistry , Emtricitabine/chemistry , Emtricitabine/metabolism , Humans , Lamivudine/chemistry , Lamivudine/metabolism , Nucleotides/chemistry , Protein Conformation
10.
DNA Repair (Amst) ; 49: 51-59, 2017 01.
Article in English | MEDLINE | ID: mdl-27989484

ABSTRACT

Human PrimPol is a recently discovered bifunctional enzyme that displays DNA template-directed primase and polymerase activities. PrimPol has been implicated in nuclear and mitochondrial DNA replication fork progression and restart as well as DNA lesion bypass. Published evidence suggests that PrimPol is a Mn2+-dependent enzyme as it shows significantly improved primase and polymerase activities when binding Mn2+, rather than Mg2+, as a divalent metal ion cofactor. Consistently, our fluorescence anisotropy assays determined that PrimPol binds to a primer/template DNA substrate with affinities of 29 and 979nM in the presence of Mn2+ and Mg2+, respectively. Our pre-steady-state kinetic analysis revealed that PrimPol incorporates correct dNTPs with 100-fold higher efficiency with Mn2+ than with Mg2+. Notably, the substitution fidelity of PrimPol in the presence of Mn2+ was determined to be in the range of 3.4×10-2 to 3.8×10-1, indicating that PrimPol is an error-prone polymerase. Furthermore, we kinetically determined the sugar selectivity of PrimPol to be 57-1800 with Mn2+ and 150-4500 with Mg2+, and found that PrimPol was able to incorporate the triphosphates of two anticancer drugs (cytarabine and gemcitabine), but not two antiviral drugs (emtricitabine and lamivudine).


Subject(s)
Coenzymes/metabolism , DNA Primase/metabolism , DNA Replication , DNA-Directed DNA Polymerase/metabolism , DNA/metabolism , Magnesium/metabolism , Manganese/metabolism , Multifunctional Enzymes/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Arabinofuranosylcytosine Triphosphate/metabolism , Arabinofuranosylcytosine Triphosphate/therapeutic use , Cations, Divalent/metabolism , Cytidine Triphosphate/analogs & derivatives , Cytidine Triphosphate/metabolism , Cytidine Triphosphate/therapeutic use , Deoxyribonucleotides/metabolism , Dideoxynucleotides/metabolism , Dideoxynucleotides/therapeutic use , Emtricitabine/analogs & derivatives , Emtricitabine/metabolism , Emtricitabine/therapeutic use , Humans , Kinetics , Lamivudine/analogs & derivatives , Lamivudine/metabolism , Lamivudine/therapeutic use
11.
Antimicrob Agents Chemother ; 60(9): 5563-72, 2016 09.
Article in English | MEDLINE | ID: mdl-27401571

ABSTRACT

Lamivudine is one of the antiretroviral drugs of choice for the prevention of mother-to-child transmission (MTCT) in HIV-positive women. In this study, we investigated the relevance of drug efflux transporters P-glycoprotein (P-gp) (MDR1 [ABCB1]), BCRP (ABCG2), MRP2 (ABCC2), and MATE1 (SLC47A1) for the transmembrane transport and transplacental transfer of lamivudine. We employed in vitro accumulation and transport experiments on MDCK cells overexpressing drug efflux transporters, in situ-perfused rat term placenta, and vesicular uptake in microvillous plasma membrane (MVM) vesicles isolated from human term placenta. MATE1 significantly accelerated lamivudine transport in MATE1-expressing MDCK cells, whereas no transporter-driven efflux of lamivudine was observed in MDCK-MDR1, MDCK-MRP2, and MDCK-BCRP monolayers. MATE1-mediated efflux of lamivudine appeared to be a low-affinity process (apparent Km of 4.21 mM and Vmax of 5.18 nmol/mg protein/min in MDCK-MATE1 cells). Consistent with in vitro transport studies, the transplacental clearance of lamivudine was not affected by P-gp, BCRP, or MRP2. However, lamivudine transfer across dually perfused rat placenta and the uptake of lamivudine into human placental MVM vesicles revealed pH dependency, indicating possible involvement of MATE1 in the fetal-to-maternal efflux of the drug. To conclude, placental transport of lamivudine does not seem to be affected by P-gp, MRP2, or BCRP, but a pH-dependent mechanism mediates transport of lamivudine in the fetal-to-maternal direction. We suggest that MATE1 might be, at least partly, responsible for this transport.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Lamivudine/metabolism , Placenta/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Biological Transport/physiology , Cell Line , Dogs , Female , Humans , Madin Darby Canine Kidney Cells , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Organic Cation Transport Proteins/metabolism , Pregnancy , Rats , Rats, Wistar
12.
Chem Biol Drug Des ; 83(5): 576-82, 2014 May.
Article in English | MEDLINE | ID: mdl-24325603

ABSTRACT

Ribavirin and lamivudine are representatives of antiviral drugs that are widely used to treat viral infections, especially chronic liver disease. To compare binding mechanism and behavior of antiviral drugs with human serum albumin (HSA), we performed fluorescence spectroscopy and X-ray crystallography to investigate the interactions of ribavirin and lamivudine with HSA. Fluorescence spectroscopy showed ribavirin and lamivudine inhibit binding affinity each other. Our results further demonstrated that ribavirin and lamivdudine interaction with HSA could be affected by the presence of other compounds, including the non-steroidal anti-inflammatory drugs, indometacin. X-ray structures revealed that ribavirin and lamivudine bind in IIA subdomain of HSA mainly by forming hydrogen bond and hydrophobic interactions forces. The carboxamido of ribavirin forms hydrogen bonds with Arg222; Hydroxyl group (6) of ribavirin forms hydrogen bond with Arg257. Hydroxyl group (15) of lamivudine forms hydrogen bond with Arg222; amino group (4) of lamivudine forms hydrogen bond with carbonyl of Arg257. Our results reveal the key biochemical and structural characteristics of the HSA interaction with ribavirin and lamivudine, providing guidance for future development of ribavirin- and lamivudine-based compounds and a drug-HSA delivery system.


Subject(s)
Antiviral Agents/metabolism , Lamivudine/metabolism , Ribavirin/metabolism , Serum Albumin/metabolism , Antiviral Agents/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Hydrogen Bonding , Lamivudine/chemistry , Molecular Dynamics Simulation , Protein Binding , Protein Structure, Tertiary , Ribavirin/chemistry , Serum Albumin/chemistry
13.
Natl Toxicol Program Tech Rep Ser ; (569): 1-212, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23385634

ABSTRACT

BACKGROUND: Antiretroviral drugs are used to treat patients positive for the human immunovirus HIV-1, and increasingly treatments include a combination of such drugs. The noninfected children of women who are pregnant and receiving such treatment may also be exposed to the drugs by transplacental exposure. We studied the long-term effects of such transplacental exposure in mice by exposing pregnant mice to combinations of four such antiretroviral drugs for seven days and then observing their pups for two years following birth. The four drugs studied were 3'-azido-3'-deoxythymidine (AZT), lamivudine (3TC), nevirapine (NVP), and nelfinavir mesylate (NFV). METHODS: Four different sets of exposure studies were performed: exposure to AZT; to AZT plus 3TC; to AZT, 3TC, and NVP; or to AZT, 3TC, and NFV. In each of these studies, groups of pregnant females were given one of three concentrations of the drug combinations seven times though a tube directly into their stomachs, and after birth their pups were maintained with no further exposure for two years. The offspring of another group of pregnant females not treated with the drugs served as controls. At the end of the study, tissues from more than 40 sites were examined for every animal. RESULTS: Survival of pups whose mothers were exposed to AZT or AZT plus 3TC was similar to their controls, while the survival rates for offspring of mice exposed to AZT, 3TC, and NVP or AZT, 3TC, and NFP were lower than for controls. In most cases the body weights of pups from mothers exposed were slightly less than those of the controls. There were slight increases in the incidences of thyroid gland tumors and skin tumors in the female pups of mothers exposed to AZT alone and of lung tumors in female pups of mothers exposed to AZT plus 3TC. For offspring of mothers exposed to AZT, 3TC, and NVP there were increased incidences of skin tumors in both male and female pups, and more so in the males. CONCLUSIONS: We conclude that exposure to the combination of AZT, 3TC, and NVP during pregnancy caused an increase in skin tumors in the male offspring and possibly also to the female offspring. Exposure to AZT alone during pregnancy may have been related to thyroid gland or skin tumors in female offspring, and exposure to AZT plus 3TC may have been related to lung tumors in female offspring.


Subject(s)
Anti-Retroviral Agents/toxicity , Lamivudine/toxicity , Nelfinavir/toxicity , Nevirapine/toxicity , Zidovudine/toxicity , Administration, Oral , Animals , Anti-Retroviral Agents/metabolism , DNA/drug effects , Drug Therapy, Combination , Escherichia coli/drug effects , Escherichia coli/genetics , Female , Lamivudine/metabolism , Longevity/drug effects , Male , Maternal Exposure , Mice , Mice, Inbred Strains , Nelfinavir/metabolism , Nevirapine/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Zidovudine/metabolism
14.
Eur J Pharm Sci ; 48(4-5): 781-9, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23298578

ABSTRACT

In order to reach the bloodstream and thus the target receptor, an orally-administered drug must first cross the intestinal barrier, which can occur via a paracellular, passive transcellular, or carrier-mediated uptake and/or efflux process (active or concentration gradient-driven). Our work aimed to explore the transport mechanism of the antiretroviral lamivudine (deoxycytidine nucleoside analogue), using a three-part strategy: in vitro, an ex vivo and an in situ method, represented by PAMPA, rat jejunum patches and rat Single Pass Intestinal Perfusion (SPIP), respectively. The determined permeability coefficients were compared with those from a published Caco-2 and MDCK study. Computational prediction of human jejunal permeability was explored, using various non-human permeability coefficients as descriptors. The ex vivo technique was performed in Franz-type diffusion cells, mounted with male Wistar rat jejunum segment patches. PAMPA was performed with an acceptor solution simulating the binding of serum proteins, an artificial membrane impregnated with egg lecithin/cholesterol and a gradient of pH between donor and acceptor solutions. The SPIP was conducted by proximal jejunum cannulation and drug perfusion in a constant flow rate of 0.2 mL/min. The outcomes of our studies showed the following predicted pattern for lamivudine effective jejunal permeability: P(eff)(exvivoA>B)>P(eff)(SPIP)>P(eff)(exvivo B>A)>P(eff)(Caco-2)≈P(eff)(MDCK)≈P(eff)(PAMPA), strongly suggesting that this compound has carrier-mediated uptake as its dominant transport mechanism. Notwithstanding, Caco-2 cells may indicate an under-expression of uptake transporters and possibly an over-expression efflux transporters, compared to that found in the rat jejunum.


Subject(s)
Anti-HIV Agents/metabolism , Jejunum/metabolism , Lamivudine/metabolism , Models, Biological , Animals , Biological Transport , Humans , In Vitro Techniques , Male , Perfusion , Permeability , Rats , Rats, Wistar
15.
Mol Pharm ; 9(11): 3259-65, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23009653

ABSTRACT

Owing to advantageous biochemical and pharmacological properties of human serum albumin (HSA), HSA-based drug carrier is playing an increasing role in the clinical setting. Since the IIA subdomain of HSA is a big hydrophobic cavity, we proposed that HSA delivers multiple drugs simultaneously docking on the IIA subdomain and that drugs may influence each other's binding affinity to albumin when cobinding the HSA IIA subdomain. Therefore, we studied the interactive association of drugs with the IIA subdomain of HSA by fluorescence spectroscopy and X-ray crystallography, in order to elucidate the molecular mechanism and behavior of drugs cobinding the IIA subdomain of HSA, and develop a universal structure-based model for HSA carrier design. We solved HSA-fatty acid-cinnamic acid, HSA-fatty acid-cinnamic acid-indometacin and HSA-fatty acid-cinnamic acid-indometacin-lamivudine complex structures, respectively. HSA complex structures and fluorescence quenching of HSA revealed that different drugs can regulate binding sites, binding mode and binding affinity of each other. For example, indometacin renders cinnamic acid to make reposition, and decreases binding affinity of HSA for cinnamic acid, but increases binding affinity of itself to HSA. Lamivudine makes cinnamic acid and indometacin to bind new subsites. Cinnamic acid-indometacin enhances binding affinity of lamivudine, and cinnamic acid-lamivudine increases binding affinity of indometacin, but indometacin-lamivudine decreases binding affinity of cinnamic acid to HSA. The study provided a biochemical basis for structure-guided development of HSA delivery system.


Subject(s)
Cinnamates/metabolism , Fatty Acids/metabolism , Indomethacin/metabolism , Lamivudine/metabolism , Serum Albumin/metabolism , Binding Sites , Cinnamates/chemistry , Crystallography, X-Ray , Fatty Acids/chemistry , Humans , Indomethacin/chemistry , Lamivudine/chemistry , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Serum Albumin/chemistry , Spectrometry, Fluorescence
16.
Biopharm Drug Dispos ; 33(3): 170-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22415520

ABSTRACT

The study sought to investigate the effect of genetic variants of OCT1 (OCT1-P283L and -P341L) and OCT2 (OCT2-T199I, -T201M and -A270S), which were identified in a Korean population, on the transport of lamivudine in vitro and to compare the substrate dependent effects of OCT1 and OCT2 variants with 1-methyl-4-phenylpyridinium (MPP+), tetraethyl ammonium (TEA), metformin and lamivudine as substrates for these transporters. When the transport kinetics of lamivudine uptake in oocytes overexpressing OCT1 and OCT2 wild-type (WT) and variant proteins were measured, lamivudine uptake mediated by OCT1-WT was saturable, and uptake was decreased in oocytes expressing OCT1-P283L and -P341L variants compared with that in OCT1-WT. The Clint of lamivudine in oocytes expressing OCT1-P283L was decreased by 85.1% compared with OCT1-WT, whereas it was decreased by 48.7% in oocytes expressing OCT1-P341L. The Clint of lamivudine in oocytes expressing OCT2-T199I, -T201M and -A270S was decreased by 86.2%, 88.9% and 73.6%, respectively, compared with OCT2-WT. When comparing various substrates such as MPP+, TEA, metformin and lamivudine, the effects of the OCT1 genetic polymorphisms on their uptake were not identical. However, contrary to the case of OCT1, the uptake of MPP+, TEA, metformin and lamivudine in oocytes expressing OCT2-T199I, -T201M and -A270S variants was decreased significantly compared with that in oocytes expressing OCT2-WT. In conclusion, the effect of genetic variations of OCT1 and OCT2 on the uptake of MPP+, TEA, metformin and lamivudine was substrate-dependent.


Subject(s)
Lamivudine/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 1/genetics , 1-Methyl-4-phenylpyridinium/metabolism , Amino Acid Substitution , Asian People/genetics , Humans , Metformin/metabolism , Oocytes/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/metabolism , Organic Cation Transporter 2 , Polymorphism, Single Nucleotide , Tetraethylammonium/metabolism
17.
Antimicrob Agents Chemother ; 56(6): 3011-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22391541

ABSTRACT

The cellular pharmacology of zidovudine (ZDV) and lamivudine (3TC) in vivo is not completely understood. This prospective longitudinal study investigated the relationship between HIV-1 serostatus, sex, race, and time on therapy with intracellular and plasma ZDV and 3TC concentrations. Of 20 HIV-seronegative and 23 HIV-seropositive volunteers enrolled, 16 (8 women) and 21 (5 women) completed all 12 study days, respectively. Volunteers began ZDV-3TC therapy (plus a third active drug in HIV-seropositive volunteers), and steady-state concentrations (C(ss)) were determined after days 1, 3, 7, and 12. A repeated-measures mixed model was utilized. HIV-seronegative status was associated with 22% (95% confidence interval [CI], 0%, 50%) and 37% (15%, 67%) higher C(ss) estimates compared to those of HIV-seropositive individuals for intracellular ZDV-TP and 3TC-TP levels, respectively. African-Americans had 36% (8%, 72%) higher ZDV-TP estimates than non-African-Americans. Sex was not associated with ZDV-TP or 3TC-TP (P > 0.19). Women had 36% (4%, 78%) higher plasma ZDV, but the effect was lessened when normalized by lean body weight (5% [-19%, 38%]; P = 0.68). Plasma 3TC was 19% (0%, 41%) higher in HIV-seropositive volunteers and 22% (0%, 48%) higher in African American volunteers, but these effects were not significant when corrected for creatinine clearance (7% [-9%, 20%] and -5% [-26%, 12%] for HIV serostatus and race, respectively; P > 0.35). These results suggest that HIV-seropositive status decreases and African American race elevates the cellular triphosphates of ZDV and 3TC. This information extends knowledge of ZDV and 3TC cellular pharmacology in vivo and provides new leads for future cellular pharmacology studies aimed at optimizing HIV prevention/treatment with these agents.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Lamivudine/pharmacology , Lamivudine/therapeutic use , Zidovudine/pharmacokinetics , Zidovudine/therapeutic use , Adult , Black or African American , Cytidine Triphosphate/analogs & derivatives , Cytidine Triphosphate/metabolism , Dideoxynucleotides/metabolism , Female , Humans , Lamivudine/analogs & derivatives , Lamivudine/metabolism , Longitudinal Studies , Male , Prospective Studies , Sex Factors , Thymine Nucleotides/metabolism , Zidovudine/analogs & derivatives , Zidovudine/metabolism
18.
DNA Cell Biol ; 31(1): 122-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21721935

ABSTRACT

One approach to accelerate the availability of new cancer drugs is to test drugs approved for other conditions as anticancer agents. In recent years, some researchers have shown that antiviral drugs, such as ritonavir, saquinavir, and nelfinavir, inhibit the growth of over 60 cancer cell lines derived from nine different tumor types. This article studied the anticancer potential of an antiviral drug, lamivudine (LA). The interaction of LA and calf thymus DNA (CT-DNA) was studied using emission, absorption, circular dichroism (CD), and viscosity techniques. The binding constants evaluated from fluorescence data at different temperatures revealed that fluorescence enhancement is a static process that involves complex-DNA formation in the ground state. Further, the enthalpy and entropy of the reaction between the drug and CT-DNA showed ΔH<0 (-126.38±0.61 kJ mol(-1)) and ΔS<0 (-352.17±2.1 J mol(-1) K(-1)); therefore, van der Waals interactions or hydrogen bonds are the main forces in the binding of LA to CT-DNA. The values of K(f) clearly underscore the high affinity of LA to DNA. In addition, detectable changes in the CD spectrum of CT-DNA in the presence of LA indicated conformational changes. All these results showed that groove binding is the binding mode of this drug and CT-DNA.


Subject(s)
Anti-HIV Agents/chemistry , DNA/chemistry , Lamivudine/chemistry , Algorithms , Anti-HIV Agents/metabolism , Binding Sites , Circular Dichroism , DNA/metabolism , Entropy , Humans , Lamivudine/metabolism , Models, Chemical , Molecular Structure , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Thermodynamics , Viscosity
19.
Eur J Pharm Sci ; 45(1-2): 110-5, 2012 Jan 23.
Article in English | MEDLINE | ID: mdl-22085635

ABSTRACT

A novel codrug (LMX) was obtained from lamivudine (LMV) and ursolic acid (UA) coupled with ethyl chloroacetate through an amide and ester linkage. The structure of LMX was confirmed by ¹H NMR, ¹³C NMR, IR and HRMS. Herein, the in vitro non-enzymatic and enzymatic hydrolysis and in vivo pharmacological activities of LMX were studied. The kinetics of hydrolysis of LMX was studied in aqueous solution of pH 1-10, 80% buffered human plasma and in the presence of lipase from Porcine pancreas (EC 3.1.1.3) at 37°C. It is found that LMX hydrolysis rate was significantly faster in lipase with half-life of 1.4h compared to pH 7.4 phosphate buffer (t(1/2) 11.2h) and buffered human plasma (t(1/2) 5.4h). The decomposition rates in aqueous solution (pH 1-10) showed a U-shaped curve. LMX was comparatively stable between pH 3 and 6 (half-life >40 h). Pharmacological studies indicated that LMX had the dual action of anti-hepatitis B virus activity and hepatoprotective effects against acute liver injury. These findings suggest that LMX could be a promising candidate agent for the treatment of hepatitis.


Subject(s)
Antiviral Agents/pharmacology , Lamivudine/chemistry , Lamivudine/pharmacology , Prodrugs/chemistry , Protective Agents/pharmacology , Triterpenes/chemistry , Triterpenes/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Drug Combinations , Drug Stability , Ducks , Half-Life , Hepadnaviridae Infections/drug therapy , Hepatitis B Virus, Duck/drug effects , Hepatitis, Viral, Animal/drug therapy , Humans , Hydrogen-Ion Concentration , Hydrolysis , Lamivudine/metabolism , Lamivudine/therapeutic use , Lipase/metabolism , Mice , Mice, Inbred ICR , Plasma/chemistry , Plasma/enzymology , Prodrugs/metabolism , Prodrugs/pharmacology , Prodrugs/therapeutic use , Protective Agents/chemistry , Protective Agents/metabolism , Protective Agents/therapeutic use , Random Allocation , Triterpenes/metabolism , Triterpenes/therapeutic use , Ursolic Acid
20.
Antimicrob Agents Chemother ; 55(1): 276-83, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21078938

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) are an important class of antiviral drugs used to manage infections by human immunodeficiency virus, which causes AIDS. Unfortunately, these drugs cause unwanted side effects, and the molecular basis of NRTI toxicity is not fully understood. Putative routes of NRTI toxicity include the inhibition of human nuclear and mitochondrial DNA polymerases. A strong correlation between mitochondrial toxicity and NRTI incorporation catalyzed by human mitochondrial DNA polymerase has been established both in vitro and in vivo. However, it remains to be determined whether NRTIs are substrates for the recently discovered human X- and Y-family DNA polymerases, which participate in DNA repair and DNA lesion bypass in vivo. Using pre-steady-state kinetic techniques, we measured the substrate specificity constants for human DNA polymerases ß, λ, η, ι, κ, and Rev1 incorporating the active, 5'-phosphorylated forms of tenofovir, lamivudine, emtricitabine, and zidovudine. For the six enzymes, all of the drug analogs were incorporated less efficiently (40- to >110,000-fold) than the corresponding natural nucleotides, usually due to a weaker binding affinity and a slower rate of incorporation for the incoming nucleotide analog. In general, the 5'-triphosphate forms of lamivudine and zidovudine were better substrates than emtricitabine and tenofovir for the six human enzymes, although the substrate specificity profile depended on the DNA polymerase. Our kinetic results suggest NRTI insertion catalyzed by human X- and Y-family DNA polymerases is a potential mechanism of NRTI drug toxicity, and we have established a structure-function relationship for designing improved NRTIs.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/metabolism , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/metabolism , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Humans , Lamivudine/chemistry , Lamivudine/metabolism , Molecular Structure , Organophosphonates/chemistry , Organophosphonates/metabolism , Structure-Activity Relationship , Tenofovir , Zidovudine/chemistry , Zidovudine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...