Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
BMC Microbiol ; 24(1): 52, 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38331716

RESUMO

Resistance mechanisms are a shelter for Acinetobacter baumannii to adapt to our environment which causes difficulty for the infections to be treated and WHO declares this organism on the top of pathogens priority for new drug development. The most common mechanism that develops drug resistance is the overexpression of the efflux pump, especially Resistance-nodulation-cell division (RND) family, to almost most antibiotics. The study is designed to detect RND efflux pump genes in A. baumannii, and its correlation to multidrug resistance, in particular, the carbapenems resistance Acinetobacter baumannii (CRAB), and using different inhibitors that restore the antibiotic susceptibility of imipenem. Clinical A. baumannii isolates were recovered from different Egyptian hospitals in Intensive care unit (ICU). The expression of genes in two strains was analyzed using RT-PCR before and after inhibitor treatment. About 100 clinical A. baumannii isolates were recovered and identified and recorded as MDR strains with 75% strains resistant to imipenem. adeB, adeC, adeK, and adeJ were detected in thirty- seven the carbapenems resistance Acinetobacter baumannii (CRAB) strains. Cinnamomum verum oil, Trimethoprim, and Omeprazole was promising inhibitor against 90% of the carbapenems resistance Acinetobacter baumannii (CRAB) strains with a 2-6-fold decrease in imipenem MIC. Downregulation of four genes was associated with the addition of those inhibitors to imipenem for two the carbapenems resistance Acinetobacter baumannii (CRAB) (ACN15 and ACN99) strains, and the effect was confirmed in 24 h killing kinetics. Our investigation points to the carbapenems resistance Acinetobacter baumannii (CRAB) strain's prevalence in Egyptian hospitals with the idea to revive the imipenem activity using natural and chemical drugs as inhibitors that possessed high synergistic activity.


Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii , Humanos , Trimetoprima/metabolismo , Trimetoprima/farmacologia , Trimetoprima/uso terapêutico , Cinnamomum zeylanicum/metabolismo , Proteínas de Bactérias/metabolismo , Infecções por Acinetobacter/tratamento farmacológico , Antibacterianos/uso terapêutico , Imipenem/farmacologia , Imipenem/uso terapêutico , Testes de Sensibilidade Microbiana , Farmacorresistência Bacteriana Múltipla/genética
2.
Am J Physiol Renal Physiol ; 326(1): F143-F151, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37942538

RESUMO

There is growing consensus that under physiological conditions, collecting duct H+ secretion is independent of epithelial Na+ channel (ENaC) activity. We have recently shown that the direct ENaC inhibitor benzamil acutely impairs H+ excretion by blocking renal H+-K+-ATPase. However, the question remains whether inhibition of ENaC per se causes alterations in renal H+ excretion. To revisit this question, we studied the effect of the antibiotic trimethoprim (TMP), which is well known to cause K+ retention by direct ENaC inhibition. The acute effect of TMP (5 µg/g body wt) was assessed in bladder-catheterized mice, allowing real-time measurement of urinary pH, electrolyte, and acid excretion. Dietary K+ depletion was used to increase renal H+-K+-ATPase activity. In addition, the effect of TMP was investigated in vitro using pig gastric H+-K+-ATPase-enriched membrane vesicles. TMP acutely increased natriuresis and decreased kaliuresis, confirming its ENaC-inhibiting property. Under control diet conditions, TMP had no effect on urinary pH or acid excretion. Interestingly, K+ depletion unmasked an acute urine alkalizing effect of TMP. This finding was corroborated by in vitro experiments showing that TMP inhibits H+-K+-ATPase activity, albeit at much higher concentrations than benzamil. In conclusion, under control diet conditions, TMP inhibited ENaC function without changing urinary H+ excretion. This finding further supports the hypothesis that the inhibition of ENaC per se does not impair H+ excretion in the collecting duct. Moreover, TMP-induced urinary alkalization in animals fed a low-K+ diet highlights the importance of renal H+-K+-ATPase-mediated H+ secretion in states of K+ depletion.NEW & NOTEWORTHY The antibiotic trimethoprim (TMP) often mediates K+ retention and metabolic acidosis. We suggest a revision of the underlying mechanism that causes metabolic acidosis. Our results indicate that TMP-induced metabolic acidosis is secondary to epithelial Na+ channel-dependent K+ retention. Under control dietary conditions, TMP does not per se inhibit collecting duct H+ secretion. These findings add further argument against a physiologically relevant voltage-dependent mechanism of collecting duct H+ excretion.


Assuntos
Acidose , Túbulos Renais Coletores , Camundongos , Animais , Suínos , Trimetoprima/farmacologia , Trimetoprima/metabolismo , Túbulos Renais Coletores/metabolismo , Canais Epiteliais de Sódio/metabolismo , Sódio/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Antibacterianos/farmacologia , Acidose/metabolismo
3.
ACS Chem Biol ; 18(4): 711-723, 2023 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-36215670

RESUMO

Opportunistic infections by Burkholderia cenocepacia are life threatening for patients suffering from cystic fibrosis and chronic granulomatous disease. These infections are often associated with variable clinical outcomes, prompting an interest in molecular investigations of phenotypes associated with disease severity. The production of the pyomelanin pigment is one such phenotype, which was recently linked to the ability of clinical strains to carry out biotransformation of the antibiotic trimethoprim. However, this biotransformation product was not identified, and differences in metabolite production associated with pyomelanin pigmentation are poorly understood. Here, we identify several key metabolites produced exclusively by the pyomelanin-producing strains. To provide insight into the structures and biosynthetic origin of these metabolites, we developed a mass spectrometry-based strategy coupling unsupervised in silico substructure prediction with stable isotope labeling referred to as MAS-SILAC (Metabolite Annotation assisted by Substructure discovery and Stable Isotope Labeling by Amino acids in Cell culture). This approach led to discovery of homogentisic acid as a precursor for biosynthesis of several natural products and for biotransformation of trimethoprim, representing a previously unknown mechanism of antibiotic tolerance. This work presents application of computational methods for analysis of untargeted metabolomic data to link the chemotype of pathogenic microorganisms with a specific phenotype. The observations made in this study provide insights into the clinical significance of the melanated phenotype.


Assuntos
Produtos Biológicos , Trimetoprima , Antibacterianos , Produtos Biológicos/metabolismo , Ácido Homogentísico/metabolismo , Metabolômica , Trimetoprima/química , Trimetoprima/metabolismo
4.
ACS Chem Biol ; 16(8): 1557-1565, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34339163

RESUMO

Manipulating subcellular protein localization using light is a powerful approach for controlling signaling processes with high spatiotemporal precision. The most widely used strategy for this is based on light-induced protein heterodimerization. The use of small synthetic molecules that can control the localization of target proteins in response to light without the need for a second protein has several advantages. However, such methods have not been well established. Herein, we present a chemo-optogenetic approach for controlling protein localization using a photoactivatable self-localizing ligand (paSL). We developed a paSL that can recruit tag-fused proteins of interest from the cytoplasm to the plasma membrane within seconds upon light illumination. This paSL-induced protein translocation (paSLIPT) is reversible and enables the spatiotemporal control of signaling processes in living cells, even in a local region. paSLIPT can also be used to implement simultaneous optical stimulation and multiplexed imaging of molecular processes in a single cell, offering an attractive and novel chemo-optogenetic platform for interrogating and engineering dynamic cellular functions.


Assuntos
Carbamatos/farmacologia , Transporte Proteico/efeitos dos fármacos , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetoprima/análogos & derivados , Trimetoprima/farmacologia , Animais , Carbamatos/metabolismo , Carbamatos/efeitos da radiação , Membrana Celular/metabolismo , Cisteína/análogos & derivados , Cisteína/metabolismo , Cisteína/farmacologia , Cisteína/efeitos da radiação , Células HeLa , Humanos , Ligantes , Luz , Camundongos , Células NIH 3T3 , Optogenética/métodos , Trimetoprima/metabolismo , Trimetoprima/efeitos da radiação
5.
J Vis Exp ; (170)2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33900288

RESUMO

Chromatin-associated condensates are implicated in many nuclear processes, but the underlying mechanisms remain elusive. This protocol describes a chemically-induced protein dimerization system to create condensates on telomeres. The chemical dimerizer consists of two linked ligands that can each bind to a protein: Halo ligand to Halo-enzyme and trimethoprim (TMP) to E. coli dihydrofolate reductase (eDHFR), respectively. Fusion of Halo enzyme to a telomere protein anchors dimerizers to telomeres through covalent Halo ligand-enzyme binding. Binding of TMP to eDHFR recruits eDHFR-fused phase separating proteins to telomeres and induces condensate formation. Because TMP-eDHFR interaction is non-covalent, condensation can be reversed by using excess free TMP to compete with the dimerizer for eDHFR binding. An example of inducing promyelocytic leukemia (PML) nuclear body formation on telomeres and determining condensate growth, dissolution, localization and composition is shown. This method can be easily adapted to induce condensates at other genomic locations by fusing Halo to a protein that directly binds to the local chromatin or to dCas9 that is targeted to the genomic locus with a guide RNA. By offering the temporal resolution required for single cell live imaging while maintaining phase separation in a population of cells for biochemical assays, this method is suitable for probing both the formation and function of chromatin-associated condensates.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Multimerização Proteica , Telômero/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetoprima/metabolismo , Proteínas de Escherichia coli/química , Humanos , Ligantes , Ligação Proteica , Tetra-Hidrofolato Desidrogenase/química , Trimetoprima/química
6.
ACS Chem Biol ; 15(4): 837-843, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32182034

RESUMO

Inducing protein translocation to the plasma membrane (PM) is an important approach for manipulating diverse signaling molecules/pathways in living cells. We previously devised a new chemogenetic system, in which a protein fused to Escherichia coli dihydrofolate reductase (eDHFR) can be rapidly translocated from the cytoplasm to the PM using a trimethoprim (TMP)-based self-localizing ligand (SL), mgcTMP. However, mgcTMP-induced protein translocation turned out to be transient and spontaneously reversed within 1 h, limiting its application. Here, we first demonstrated that the spontaneous reverse translocation was caused by cellular degradation of mgcTMP, presumably by proteases. To address this problem, we newly developed a proteolysis-resistant SL, mDcTMP. This mDcTMP now allows sustained PM localization of eDHFR-fusion proteins (over several hours to a day), and it was applicable to inducing prolonged signal activation and cell differentiation. mDcTMP also worked in live nematodes, making it an attractive new tool for probing and controlling living systems.


Assuntos
Cisteína/análogos & derivados , Cisteína/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Trimetoprima/análogos & derivados , Trimetoprima/farmacologia , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Cisteína/metabolismo , Escherichia coli/enzimologia , Proteínas de Escherichia coli/metabolismo , Complexo de Golgi/metabolismo , Humanos , Ligantes , Lipoilação , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/metabolismo , Ratos , Transdução de Sinais/fisiologia , Estereoisomerismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetoprima/metabolismo
7.
Angew Chem Int Ed Engl ; 58(19): 6285-6289, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30834641

RESUMO

Several genome engineering applications of CRISPR-Cas9, an RNA-guided DNA endonuclease, require precision control of Cas9 activity over dosage, timing, and targeted site in an organism. While some control of Cas9 activity over dose and time have been achieved using small molecules, and spatial control using light, no singular system with control over all the three attributes exists. Furthermore, the reported small-molecule systems lack wide dynamic range, have background activity in the absence of the small-molecule controller, and are not biologically inert, while the optogenetic systems require prolonged exposure to high-intensity light. We previously reported a small-molecule-controlled Cas9 system with some dosage and temporal control. By photocaging this Cas9 activator to render it biologically inert and photoactivatable, and employing next-generation protein engineering approaches, we have built a system with a wide dynamic range, low background, and fast photoactivation using a low-intensity light while rendering the small-molecule activator biologically inert. We anticipate these precision controls will propel the development of practical applications of Cas9.


Assuntos
Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Engenharia de Proteínas , Estrutura Terciária de Proteína , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Ativação Transcricional/efeitos dos fármacos , Trimetoprima/química , Trimetoprima/metabolismo , Trimetoprima/farmacologia , Raios Ultravioleta
8.
Water Sci Technol ; 2017(1): 144-155, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29698230

RESUMO

A pilot scale biological nutrient removal (BNR) process, batch experiments and modeling exercises were employed to investigate the removal and biotransformation of trimethoprim (TMP) in a BNR activated sludge process. The concentrations of the active microbial groups - ammonia oxidizing bacteria (AOB), ordinary heterotrophic organisms (OHOs) and polyphosphate accumulating organisms (PAOs) - in the BNR bioreactor were quantified through modeling of the pilot bioreactor. The overall TMP removal efficiency for the pilot BNR process was 64 ± 14% while the TMP biotransformation efficiencies in the anaerobic, anoxic and aerobic zones were 22 ± 20%, 27 ± 8% and 36 ± 5% respectively. Batch tests with and without nitrification inhibition showed that AOB played a role in the biotransformation of TMP in BNR activated sludge. A pseudo first order model which incorporated the contributions of PAOs, OHOs and AOB to the overall biodegradation of TMP was found to describe the biodegradation of TMP in batch tests with and without nitrification inhibition. This model showed that PAOs, OHOs and AOB contributed towards the biotransformation of TMP in aerobic BNR activated sludge with the biotransformation rate constants following the trend of kAOB > kOHOs > kPAOs.


Assuntos
Reatores Biológicos , Trimetoprima/química , Eliminação de Resíduos Líquidos , Anti-Infecciosos Urinários/química , Anti-Infecciosos Urinários/metabolismo , Biodegradação Ambiental , Biotransformação , Nitrogênio/metabolismo , Polifosfatos/metabolismo , Esgotos/microbiologia , Trimetoprima/metabolismo , Poluentes Químicos da Água/química , Poluentes Químicos da Água/metabolismo
9.
Proc Natl Acad Sci U S A ; 113(11): E1470-8, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929328

RESUMO

Fitness landscapes of drug resistance constitute powerful tools to elucidate mutational pathways of antibiotic escape. Here, we developed a predictive biophysics-based fitness landscape of trimethoprim (TMP) resistance for Escherichia coli dihydrofolate reductase (DHFR). We investigated the activity, binding, folding stability, and intracellular abundance for a complete set of combinatorial DHFR mutants made out of three key resistance mutations and extended this analysis to DHFR originated from Chlamydia muridarum and Listeria grayi We found that the acquisition of TMP resistance via decreased drug affinity is limited by a trade-off in catalytic efficiency. Protein stability is concurrently affected by the resistant mutants, which precludes a precise description of fitness from a single molecular trait. Application of the kinetic flux theory provided an accurate model to predict resistance phenotypes (IC50) quantitatively from a unique combination of the in vitro protein molecular properties. Further, we found that a controlled modulation of the GroEL/ES chaperonins and Lon protease levels affects the intracellular steady-state concentration of DHFR in a mutation-specific manner, whereas IC50 is changed proportionally, as indeed predicted by the model. This unveils a molecular rationale for the pleiotropic role of the protein quality control machinery on the evolution of antibiotic resistance, which, as we illustrate here, may drastically confound the evolutionary outcome. These results provide a comprehensive quantitative genotype-phenotype map for the essential enzyme that serves as an important target of antibiotic and anticancer therapies.


Assuntos
Farmacorresistência Bacteriana/genética , Escherichia coli/efeitos dos fármacos , Tetra-Hidrofolato Desidrogenase/genética , Trimetoprima/farmacologia , Sequência de Aminoácidos , Biofísica/métodos , Chlamydia muridarum/genética , Evolução Molecular Direcionada , Estabilidade Enzimática/genética , Epistasia Genética , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Concentração Inibidora 50 , Listeria/genética , Dados de Sequência Molecular , Mutação , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetoprima/metabolismo
10.
Glycobiology ; 21(2): 235-46, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20978009

RESUMO

Sulfation represents an essential modification for various molecules and regulates many biological processes. The sulfation of glycans requires a specific transporter for 3'-phosphoadenosine 5'-phosphosulfate (PAPS) on the Golgi apparatus. This study investigated the expression of PAPS transporter genes in colorectal carcinomas and the significance of Golgi-specific sulfation in the proliferation of colorectal carcinoma cells. The relative amount of PAPST1 transcripts was found to be higher than those of PAPST2 in colorectal cancerous tissues. Immunohistochemically, the enhanced expression of PAPST1 was observed in fibroblasts in the vicinity of invasive cancer cells, whereas the expression of PAPST2 was decreased in the epithelial cells. RNA interference of either of the two PAPS transporter genes reduced the extent of sulfation of cellular proteins and cellular proliferation of DLD-1 human colorectal carcinoma cells. Silencing the PAPS transporter genes reduced fibroblast growth factor signaling in DLD-1 cells. These findings indicate that PAPS transporters play a role in the proliferation of colorectal carcinoma cells themselves and take part in a desmoplastic reaction to support cancer growth by controlling their sulfation status.


Assuntos
Proteínas de Transporte de Ânions , Expressão Gênica , Proteínas de Membrana Transportadoras , Polissacarídeos , Sulfamonometoxina , Trimetoprima , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Transporte Biológico , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Combinação de Medicamentos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Imuno-Histoquímica , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Invasividade Neoplásica , Fosfoadenosina Fosfossulfato/metabolismo , Polissacarídeos/genética , Polissacarídeos/metabolismo , Interferência de RNA , RNA Mensageiro/biossíntese , Transdução de Sinais/genética , Sulfamonometoxina/metabolismo , Transportadores de Sulfato , Trimetoprima/metabolismo , Células Tumorais Cultivadas
11.
Rapid Commun Mass Spectrom ; 24(9): 1241-50, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20391594

RESUMO

Drug bioactivation leading to the formation of reactive species capable of covalent binding to proteins represents an important cause of drug-induced toxicity. Reactive metabolite detection using in vitro microsomal incubations is a crucial step in assessing potential toxicity of pharmaceutical compounds. The most common method for screening the formation of these unstable, electrophilic species is by trapping them with glutathione (GSH) followed by liquid chromatography/mass spectrometry (LC/MS) analysis. The present work describes the use of a brominated analog of glutathione, N-(2-bromocarbobenzyloxy)-GSH (GSH-Br), for the in vitro screening of reactive metabolites by LC/MS. This novel trapping agent was tested with four drug compounds known to form reactive metabolites, acetaminophen, fipexide, trimethoprim and clozapine. In vitro rat microsomal incubations were performed with GSH and GSH-Br for each drug with subsequent analysis by liquid chromatography/high-resolution mass spectrometry on an electrospray time-of-flight (ESI-TOF) instrument. A generic LC/MS method was used for data acquisition, followed by drug-specific processing of accurate mass data based on mass defect filtering and isotope pattern matching. GSH and GSH-Br incubations were compared to control samples using differential analysis (Mass Profiler) software to identify adducts formed via the formation of reactive metabolites. In all four cases, GSH-Br yielded improved results, with a decreased false positive rate, increased sensitivity and new adducts being identified in contrast to GSH alone. The combination of using this novel trapping agent with powerful processing routines for filtering accurate mass data and differential analysis represents a very reliable method for the identification of reactive metabolites formed in microsomal incubations.


Assuntos
Compostos de Bromo/química , Cromatografia Líquida/métodos , Glutationa/análogos & derivados , Preparações Farmacêuticas/química , Espectrometria de Massas por Ionização por Electrospray/métodos , Acetaminofen/química , Acetaminofen/metabolismo , Animais , Compostos de Bromo/metabolismo , Clozapina/química , Clozapina/metabolismo , Glutationa/química , Glutationa/metabolismo , Microssomos Hepáticos/metabolismo , Preparações Farmacêuticas/metabolismo , Piperazinas/química , Piperazinas/metabolismo , Ratos , Sensibilidade e Especificidade , Trimetoprima/química , Trimetoprima/metabolismo
12.
Proteins ; 76(3): 706-17, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19280600

RESUMO

Dihydrofolate reductase (DHFR) is the enzyme responsible for the NADPH-dependent reduction of 5,6-dihydrofolate to 5,6,7,8-tetrahydrofolate, an essential cofactor in the synthesis of purines, thymidylate, methionine, and other key metabolites. Because of its importance in multiple cellular functions, DHFR has been the subject of much research targeting the enzyme with anticancer, antibacterial, and antimicrobial agents. Clinically used compounds targeting DHFR include methotrexate for the treatment of cancer and diaminopyrimidines (DAPs) such as trimethoprim (TMP) for the treatment of bacterial infections. DAP inhibitors of DHFR have been used clinically for >30 years and resistance to these agents has become widespread. Methicillin-resistant Staphylococcus aureus (MRSA), the causative agent of many serious nosocomial and community acquired infections, and other gram-positive organisms can show resistance to DAPs through mutation of the chromosomal gene or acquisition of an alternative DHFR termed "S1 DHFR." To develop new therapies for health threats such as MRSA, it is important to understand the molecular basis of DAP resistance. Here, we report the crystal structure of the wild-type chromosomal DHFR from S. aureus in complex with NADPH and TMP. We have also solved the structure of the exogenous, TMP resistant S1 DHFR, apo and in complex with TMP. The structural and thermodynamic data point to important molecular differences between the two enzymes that lead to dramatically reduced affinity of DAPs to S1 DHFR. These differences in enzyme binding affinity translate into reduced antibacterial activity against strains of S. aureus that express S1 DHFR.


Assuntos
Cristalografia por Raios X/métodos , Staphylococcus aureus/enzimologia , Tetra-Hidrofolato Desidrogenase/química , Trimetoprima/química , Ligação de Hidrogênio , Mutação , NADP/química , Ligação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Termodinâmica , Trimetoprima/metabolismo
13.
Chem Res Toxicol ; 21(11): 2181-7, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18816075

RESUMO

Trimethoprim (TMP) is a widely used antibacterial agent that is usually considered as a safe drug. TMP has, however, been implicated in rare adverse drug reactions (ADRs) in humans. Bioactivation to a reactive iminoquinone methide intermediate has been proposed as a possible cause for the toxicity of the drug. However, little is known about the cytochrome P450s (P450s) involved in this bioactivation and in the metabolism of TMP in general. In this study, we have investigated the metabolism and bioactivation of TMP by human liver microsomes (HLM) and rat liver microsomes, by recombinant human cytochrome P450s, and by the bacterial P450 BM3 mutant M11(his). In addition to non GSH-dependent metabolites, five GSH adducts were identified in the HLM incubations. Next to two major GSH adducts probably originating from the iminoquinone methide intermediate described previously, three minor GSH adducts were also identified, indicating that other types of reactive intermediates are formed by HLM, such as ortho-quinones and para-quinone methide intermediates. The major GSH adducts were produced by P450 1A2 and P450 3A4, while the minor GSH adducts were mainly formed by P450 1A2, P450 3A4, and P450 2D6. Although preliminary, these results might implicate that genetic polymorphisms in P450 enzymes could play a role in the onset of TMP-related ADRs in humans.


Assuntos
Antibacterianos/metabolismo , Trimetoprima/metabolismo , Biotransformação , Cromatografia Líquida de Alta Pressão , Sistema Enzimático do Citocromo P-450/fisiologia , Glutationa/metabolismo , Humanos , Espectrometria de Massas , Microssomos Hepáticos/metabolismo , Trimetoprima/efeitos adversos
14.
Clin Pharmacol Ther ; 82(2): 197-203, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17361129

RESUMO

Metabolism of the antimalarial drug amodiaquine (AQ) into its primary metabolite, N-desethylamodiaquine, is mediated by CYP2C8. We studied the frequency of CYP2C8 variants in 275 malaria-infected patients in Burkina Faso, the metabolism of AQ by CYP2C8 variants, and the impact of other drugs on AQ metabolism. The allele frequencies of CYP2C8*2 and CYP2C8*3 were 0.155 and 0.003, respectively. No evidence was seen for influence of CYP2C8 genotype on AQ efficacy or toxicity, but sample size limited these assessments. The variant most common in Africans, CYP2C8(*)2, showed defective metabolism of AQ (threefold higher K(m) and sixfold lower intrinsic clearance), and CYP2C8(*)3 had markedly decreased activity. Considering drugs likely to be coadministered with AQ, the antiretroviral drugs efavirenz, saquinavir, lopinavir, and tipranavir were potent CYP2C8 inhibitors at clinically relevant concentrations. Variable CYP2C8 activity owing to genetic variation and drug interactions may have important clinical implications for the efficacy and toxicity of AQ.


Assuntos
Amodiaquina/metabolismo , Hidrocarboneto de Aril Hidroxilases/metabolismo , Malária Falciparum/tratamento farmacológico , Polimorfismo Genético , Alcinos , Amodiaquina/análogos & derivados , Amodiaquina/farmacologia , Antimaláricos/metabolismo , Antimaláricos/farmacologia , Hidrocarboneto de Aril Hidroxilases/genética , Benzoxazinas/metabolismo , Benzoxazinas/farmacologia , Burkina Faso , Cromatografia Líquida de Alta Pressão , Ciclopropanos , Citocromo P-450 CYP2C8 , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Genótipo , Inibidores da Protease de HIV/metabolismo , Inibidores da Protease de HIV/farmacologia , Humanos , Lopinavir , Malária Falciparum/genética , Malária Falciparum/metabolismo , Modelos Biológicos , Piridinas/metabolismo , Piridinas/farmacologia , Pirimidinonas/metabolismo , Pirimidinonas/farmacologia , Pironas/metabolismo , Pironas/farmacologia , Inibidores da Transcriptase Reversa/metabolismo , Inibidores da Transcriptase Reversa/farmacologia , Saquinavir/metabolismo , Saquinavir/farmacologia , Espectrofotometria Ultravioleta , Sulfonamidas , Resultado do Tratamento , Trimetoprima/metabolismo , Trimetoprima/farmacologia
15.
J Clin Invest ; 113(3): 464-73, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14755343

RESUMO

An antibiotic efflux gene cluster that confers resistance to chloramphenicol, trimethoprim, and ciprofloxacin has been identified in Burkholderia cenocepacia (genomovar III), an important cystic fibrosis pathogen. Five open reading frames have been identified in the cluster. There is apparently a single transcriptional unit, with llpE encoding a lipase-like protein, ceoA encoding a putative periplasmic linker protein, ceoB encoding a putative cytoplasmic membrane protein, and opcM encoding a previously described outer membrane protein. A putative LysR-type transcriptional regulatory gene, ceoR, is divergently transcribed upstream of the structural gene cluster. Experiments using radiolabeled chloramphenicol and salicylate demonstrated active efflux of both compounds in the presence of the gene cluster. Salicylate is an important siderophore produced by B. cepacia complex isolates, and both extrinsic salicylate and iron starvation appear to upregulate ceoR promoter activity, as does chloramphenicol. These results suggest that salicylate is a natural substrate for the efflux pump in B. cenocepacia and imply that the environment of low iron concentration in the cystic fibrosis lung can induce efflux-mediated resistance, even in the absence of antibiotic selective pressure.


Assuntos
Complexo Burkholderia cepacia/metabolismo , Farmacorresistência Bacteriana/fisiologia , Salicilatos/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Complexo Burkholderia cepacia/genética , Cloranfenicol/metabolismo , Ciprofloxacina/metabolismo , Farmacorresistência Bacteriana/genética , Perfilação da Expressão Gênica , Humanos , Lactente , Família Multigênica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Trimetoprima/metabolismo
16.
J Pharmacol Exp Ther ; 291(1): 292-9, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10490916

RESUMO

The antibacterial agent, trimethoprim, is normally used synergistically with sulfonamides. Its use is associated with idiosyncratic reactions including liver toxicity and agranulocytosis. In this study, we demonstrated that trimethoprim was oxidized by activated human neutrophils, as well as a combination of myeloperoxidase/hydrogen peroxide/chloride or hypochlorous acid, to a reactive pyrimidine iminoquinone methide intermediate with a protonated molecular ion of m/z 289 as detected by mass spectrometry. In the presence of N-acetyl-L-cysteine (NAC), the pyrimidine iminoquinone methide could be trapped as three NAC adducts. The three NAC adducts were separable on HPLC, but showed the same protonated molecular ion of m/z 452. The proton NMR spectrum of the major adduct showed that the NAC group was at the 6 position of the pyrimidine ring. The mass spectra of the two minor NAC adducts indicated that they were the two diastereomers in which NAC was attached to the exo-cyclic prechiral carbon of the pyrimidine iminoquinone methide. Incubation of trimethoprim with isolated hepatic microsomes, both human and rat, in presence of NAC gave the same set of trimethoprim-NAC adducts. We propose that the formation of this pyrimidine iminoquinone methide by both hepatic microsomes and neutrophils may be responsible for trimethoprim-induced idiosyncratic hepatotoxicity and agranulocytosis.


Assuntos
Microssomos Hepáticos/metabolismo , Neutrófilos/metabolismo , Trimetoprima/metabolismo , Acetilcisteína/farmacologia , Animais , Interações Medicamentosas , Sequestradores de Radicais Livres/farmacologia , Humanos , Ácido Hipocloroso/farmacologia , Técnicas In Vitro , Espectroscopia de Ressonância Magnética , Masculino , Neutrófilos/efeitos dos fármacos , Oxirredução , Peroxidase/metabolismo , Quinonas/metabolismo , Ratos , Ratos Sprague-Dawley , Trimetoprima/farmacologia , Trítio
17.
J Antimicrob Chemother ; 36(4): 607-18, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8591935

RESUMO

Sulphadiazine had little or no antibacterial effect against three strains of Enterococcus faecalis (MICs of above 3600 mg/L) but it caused approximately six-fold increases in bacterial uptake of trimethoprim, increased release of bacterial ATP and produced ultrastructural damage to both the trimethoprim resistant E. faecalis 463 and the trimethoprim sensitive NCTC 5957. MICs of trimethoprim were 0.6, 0.8 and 76.8 mg/L for E. faecalis NCTC 775, NCTC 5957 and 463 respectively. When log phase E. faecalis 463 and NCTC 5957 were grown for 4 h in trimethoprim 56 and 0.6 mg/L plus sulphadiazine 320 and 100 mg/L respectively there was an approximate ten-fold and nine-fold increase in uptake of sulphadiazine and a two-fold increase in leakage of ATP. Trimethoprim caused more damage to the cell wall and cytoplasmic membrane than sulphadiazine but the combination of sulphadiazine plus trimethoprim caused the most cell damage. The increased activity observed with the combination seems very likely to have resulted from the increased uptakes of the antibacterials, which in turn had resulted from the cell wall damage and consequent increased cell permeability caused by each antibacterial. It is proposed that a markedly subinhibitory concentration of sulphadiazine enhanced the antibacterial activity of trimethoprim against all three strains of E. faecalis by this mechanism.


Assuntos
Antibacterianos/farmacologia , Enterococcus faecalis/efeitos dos fármacos , Sulfadiazina/farmacologia , Trimetoprima/farmacologia , Trifosfato de Adenosina/metabolismo , Antibacterianos/metabolismo , Resistência Microbiana a Medicamentos , Sinergismo Farmacológico , Enterococcus faecalis/crescimento & desenvolvimento , Enterococcus faecalis/metabolismo , Enterococcus faecalis/ultraestrutura , Membranas/efeitos dos fármacos , Membranas/ultraestrutura , Testes de Sensibilidade Microbiana , Microscopia Eletrônica , Permeabilidade , Sulfadiazina/metabolismo , Trimetoprima/metabolismo
18.
J Chromatogr ; 616(1): 79-85, 1993 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-7690764

RESUMO

Reversed-phase ion-pair high-performance liquid chromatography has been investigated for the separation, detection, identification and quantitation of the isomeric 1N- and 3N-oxide metabolites of metoprine, pyrimethamine and trimethoprim. A rapid and sensitive analytical method for the simultaneous determination of the isomeric 1N- and 3N-oxides of metoprine and pyrimethamine was devised for in vitro metabolic studies by optimisation of mobile phase pH, pairing-ion concentration, secondary-ion concentration and percentage organic modifier.


Assuntos
Óxidos/análise , Pirimidinas/análise , Animais , Antineoplásicos/análise , Antineoplásicos/metabolismo , Cromatografia Líquida de Alta Pressão , Cricetinae , Ácido Dioctil Sulfossuccínico , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Indicadores e Reagentes , Isomerismo , Microssomos Hepáticos/química , Microssomos Hepáticos/metabolismo , Óxidos/metabolismo , Pirimetamina/análogos & derivados , Pirimetamina/análise , Pirimetamina/metabolismo , Pirimidinas/metabolismo , Tensoativos , Trimetoprima/análise , Trimetoprima/metabolismo
19.
J Biol Chem ; 268(6): 4556-66, 1993 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-8440739

RESUMO

We describe the characterization of an antitumor drug resistance following multiple step selection of hamster cells to the 2,4-diaminopyrimidines (DAP) metoprine, pyrimethamine (Pyr), and trimethoprim (Tmp). Pyr and Tmp are DAP lipophilic antifolates currently used as antiparasitic and antibacterial antibiotics, respectively. Dihydrofolate reductase (DHFR) from hamster cells bore a low or poor affinity to these DAP as compared to the hydrophilic folate antagonist methotrexate (MTX). Metoprine-resistant cells over-expressed DHFR enzyme and consequently displayed a high level of resistance to both hydrophilic and lipophilic antifolates including DAP but maintained wild type sensitivity to pleiotropic drugs involved in multi-drug resistance (MDR). In contrast, although Pyr- and Tmp-resistant cells expressed parental levels of wild type DHFR, they displayed a high degree of resistance to DAP and, surprisingly, to the lipophilic MTX analogs piritrexim (PTX) and trimetrexate (TMTX), while maintaining sensitivity to MTX. These drug-resistant cells maintained wild type mRNA levels of the MDR gene product P-glycoprotein and showed collateral hypersensitivity to pleiotropic drugs. To study the underlying mechanism of this apparently new resistance phenotype, we have employed fluorescein-methotrexate (F-MTX) labeling of cells and its displacement by different antifolates. Parental AA8 and Pyr-resistant cells showed a similar level of F-MTX labeling, however, while DAP, TMTX, and PTX showed an efficient competitive displacement of F-MTX from AA8 cells, Pyr-resistant cells displayed a persistent retention of F-MTX labeling in the presence of high concentrations of these lipophilic antifolates. Pyr-resistant cells showed a wild type displacement of F-MTX with MTX. This DAP resistance phenotype was unstable as it was rapidly lost upon growth under nonselective conditions. Furthermore, when the antifolate resistance levels of Pyr-resistant cells were plotted versus the ratios of the 50% F-MTX displacement values obtained with resistant and parental AA8 cells, a good correlation (r2 > 0.98) was obtained. We conclude that Pyr-resistant cells possess a novel phenotype that derives its resistance to lipophilic antifolates solely from a predominant decrease in the accumulation of DAP and lipid-soluble analogs of MTX.


Assuntos
Antagonistas do Ácido Fólico/farmacologia , Pirimetamina/farmacologia , Animais , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Resistência a Medicamentos , Citometria de Fluxo , Leucovorina/farmacologia , Pirimetamina/análogos & derivados , Pirimetamina/metabolismo , RNA Mensageiro/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetoprima/metabolismo , Trimetoprima/farmacologia
20.
FEBS Lett ; 218(1): 178-84, 1987 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-3595861

RESUMO

The structure of mouse L1210 dihydrofolate reductase (DHFR) complexed with NADPH and trimethoprim has been refined at 2.0 A resolution. The analogous complex with NADPH and methotrexate has been refined at 2.5 A resolution. These structures reveal for the first time details of drug interactions with a mammalian DHFR, which are compared with those observed from previous X-ray investigations of DHFR/inhibitor complexes. The refined L1210 structure has been used as the basis for the construction of a model of the human enzyme. There are only twenty-one sequence differences between mouse L1210 and human DHFRs, and all but two of these are located close to the molecular surface: a strong indication that the active sites are essentially identical in these two mammalian enzymes.


Assuntos
Leucemia L1210/enzimologia , NADP/metabolismo , Proteínas de Neoplasias/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetoprima/metabolismo , Animais , Sítios de Ligação , Humanos , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Especificidade da Espécie , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA