Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
mBio ; 15(2): e0207323, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38179948

RESUMO

Tuberculosis (TB) is a significant global public health threat. Despite the long-standing use of para-aminosalicylic acid (PAS) as a second-line anti-TB drug, its resistance mechanism remains unclear. In this study, we isolated 90 mutants of PAS-resistant Mycobacterium tuberculosis (MTB) H37Ra in 7H11 solid medium and performed whole-genome sequencing, gene overexpression, transcription level comparison and amino acid level determination in MTB, and promoter activity by ß-galactosidase assays in Mycobacterium smegmatis to elucidate the mechanism of PAS resistance. Herein, we found that 47 of 90 (52.2%) PAS-resistant mutants had nine different mutations in the intergenic region of metM (Rv3253c) and Rv3254. Beta-galactosidase assays confirmed that mutations increased promoter activity only for metM but not Rv3254. Interestingly, overexpression of MetM or its M. smegmatis homolog (MSMEI_1796) either by its promoter in metM's direction or by exogenous expression in MTB induced PAS resistance in a methionine-dependent manner. Therefore, drug susceptibility results for the metM promoter mutants can be misleading when using standard 7H10 or 7H9 medium, which lacks methionine. At the metabolism level, PAS treatment led to higher intracellular methionine levels in the mutants than the wild type, antagonizing PAS and conferring resistance. Furthermore, 12 different mutations in the metM promoter were identified in clinical MTB strains. In summary, we found a novel mechanism of PAS resistance in MTB. Mutations in the metM (Rv3253c) promoter upregulate metM transcription and elevate intracellular methionine, which antagonize PAS. Our findings shed new light on the mechanism of PAS resistance in MTB and highlight issues with the current PAS susceptibility culture medium.IMPORTANCEAlthough para-aminosalicylic acid (PAS) has been used to treat TB for more than 70 years, the understanding of PAS resistance mechanisms is still vague, living gaps in our ability to predict resistance and apply PAS effectively in clinical practice. This study aimed to address this knowledge gap by inducing in vitro PAS resistance in Mycobacterium tuberculosis (MTB) using 7H11 medium and discovering a new PAS resistance mechanism. Our research revealed that spontaneous mutations occurring in the promoter region of the methionine transporting gene, metM, can upregulate the expression of metM, resulting in increased intracellular transport of methionine and consequently high-level resistance of Mycobacterium tuberculosis to PAS. Notably, this resistance phenotype cannot be observed when using the commonly recommended 7H10 medium, possibly due to the lack of additional methionine supply compared with that when using the 7H11 medium. Mutations on the regulatory region of metM were also found in some clinical MTB strains. These findings may have important implications for the unexplained PAS resistance observed in clinical settings and provide insight into the failures of PAS treatment. Additionally, they underscore the importance of considering the choice of culture media when conducting drug susceptibility testing for MTB.


Assuntos
Ácido Aminossalicílico , Mycobacterium tuberculosis , Ácido Aminossalicílico/farmacologia , Ácido Aminossalicílico/metabolismo , Testes de Sensibilidade Microbiana , Farmacorresistência Bacteriana/genética , Antituberculosos/farmacologia , Mutação , Metionina/metabolismo , beta-Galactosidase/genética
2.
Sci Rep ; 12(1): 5635, 2022 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-35379873

RESUMO

Para-aminosalicylic acid (PAS) is an antibiotic that was largely used for the multi-therapy of tuberculosis in the twentieth century. To try to overcome the inconvenience of its low efficacy and poor tolerance, we searched for novel chemical entities able to synergize with PAS using a combination screening against growing axenic Mycobacterium tuberculosis. The screening was performed at a sub-inhibitory concentration of PAS on a library of about 100,000 small molecules. Selected hit compounds were analyzed by dose-response and further probed with an intracellular macrophage assay. Scaffolds with potential additive effect with PAS are reported, opening interesting prospects for mechanism of action studies. We also report here evidence of a yet unknown bio-activation mechanism, involving activation of pyrido[1,2-a]pyrimidin-4-one (PP) derivatives through the Rv3087 protein.


Assuntos
Ácido Aminossalicílico , Mycobacterium tuberculosis , Tuberculose dos Linfonodos , Ácido Aminossalicílico/metabolismo , Ácido Aminossalicílico/farmacologia , Antituberculosos/química , Humanos
3.
Antimicrob Agents Chemother ; 66(1): e0146521, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34780266

RESUMO

Tuberculosis (TB), caused by Mycobacterium tuberculosis, is one of the most fatal diseases in the world. Methylenetetrahydrofolate reductase (MTHFR) catalyzes the production of 5-methyltetrahydrofolate (5-CH3-THF), which is required for the de novo biosynthesis of methionine in bacteria. Here, we identified Rv2172c as an MTHFR in M. tuberculosis through in vitro and in vivo analyses and determined that the protein is essential for the in vitro growth of the bacterium. Subsequently, we constructed rv2172c R159N and L214A mutants in M. tuberculosis and found that these mutants were more sensitive to the antifolates para-aminosalicylic acid (PAS) and sulfamethoxazole (SMX). Combining biochemical and genetic methods, we found that rv2172c R159N or L214A mutation impaired methionine production, leading to increased susceptibility of M. tuberculosis to PAS, which was largely restored by adding exogenous methionine. Moreover, overexpression of rv2172c in M. tuberculosis could increase methionine production and lead to PAS resistance. This research is the first to identify an MTHFR in M. tuberculosis and reveals that the activity of this enzyme is associated with susceptibility to antifolates. These findings have particular value for antitubercular drug design for the treatment of drug-resistant TB.


Assuntos
Ácido Aminossalicílico , Mycobacterium tuberculosis , Ácido Aminossalicílico/metabolismo , Ácido Aminossalicílico/farmacologia , Antituberculosos/metabolismo , Antituberculosos/farmacologia , Proteínas de Bactérias/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/metabolismo
4.
Plant Cell Rep ; 39(9): 1129-1141, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32405654

RESUMO

KEY MESSAGE: 4-Coumarate coenzyme A ligase and 4-hydroxybenzoate coenzyme A ligase from the hornwort Anthoceros agrestis expressed in E. coli were characterized on biochemical and molecular levels and showed interesting substrate specificities. Acyl-activating enzymes are associated with the biosynthesis or degradation of various metabolic products such as lipids, amino acids, sugars, and natural compounds. In this work, cDNA sequences encoding 4-coumarate coenzyme A ligase (4CL) and 4-hydroxybenzoate coenzyme A ligase (4HBCL) were amplified from the hornwort Anthoceros agrestis. The coding sequences were expressed in E. coli and purified by Ni-chelate chromatography. The CoA ligases exhibited different substrate specificities. 4CL catalyzed the activation of 4-coumaric acid, 3-coumaric acid, 2-coumaric acid, caffeic acid, isoferulic acid, ferulic acid, and cinnamic acid but lacked activities towards sinapic acid and benzoic acids. In contrast, 4HBCL preferred 4-hydroxybenzoic acid and benzoic acid, but also accepted other benzoic acid derivatives except salicylic acid and 3-aminosalicylic acid. Furthermore, 4HBCL also activated isoferulic acid, cinnamic acid, 2-coumaric acid, 3-coumaric acid, 4-coumaric acid and caffeic acid, but lacked affinity for ferulic acid and sinapic acid. These substrate specificities could be related to the phenolic compounds identified in Anthoceros agrestis.


Assuntos
Anthocerotophyta/metabolismo , Coenzima A Ligases/metabolismo , Fenóis/metabolismo , Ácido Aminossalicílico/química , Ácido Aminossalicílico/metabolismo , Anthocerotophyta/genética , Ácidos Cafeicos/metabolismo , Cinamatos/metabolismo , Coenzima A Ligases/genética , Ácidos Cumáricos/metabolismo , Escherichia coli/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
5.
Chem Biol Drug Des ; 95(1): 104-112, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31562690

RESUMO

Previous studies have reported that genome-wide DNA methylation and differentially expressed genes and proteins are closely associated with drug resistance in Mycobacterium tuberculosis (M. tuberculosis). However, no reports have explored such associations in para-aminosalicylic acid (PAS)-resistant M. tuberculosis H37Rv. Here, we investigated genome-wide methylation and transcriptome and proteome changes to explore the associations between specific genes and PAS resistance in M. tuberculosis H37Rv. The results revealed that 1,388 differentially methylated (1,161 hypermethylated and 227 hypomethylated) genes, 214 significantly differentially expressed (103 up- and 111 down-regulated) genes and 137 differentially expressed (48 up- and 89 down-regulated) proteins were regulated by PAS in M. tuberculosis H37Rv. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that metabolic pathways and ABC transporters were closely associated with differentially methylated and expressed genes, respectively. In addition, correlation analysis revealed that differentially methylated genes were negatively correlated with their transcriptional levels in PAS-resistant M. tuberculosis H37Rv. Furthermore, the existence of five hypermethylated candidate genes (esxC, fabG3, fbpB, papA1 and pks2) in PAS-resistant M. tuberculosis H37Rv was verified using protein-protein interaction analysis in the STRING database. The integrated DNA methylation and transcriptome and proteome analysis could provide valuable resources for epigenetics studies in PAS-resistant M. tuberculosis H37Rv.


Assuntos
Ácido Aminossalicílico/metabolismo , Proteínas de Bactérias/genética , Metilação de DNA , Mycobacterium tuberculosis/genética , Proteínas de Bactérias/metabolismo , Bases de Dados de Proteínas , Farmacorresistência Bacteriana , Regulação da Expressão Gênica , Genes Bacterianos , Mapeamento de Interação de Proteínas , Proteoma , Transdução de Sinais , Transcriptoma
6.
Mucosal Immunol ; 12(1): 188-199, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30279515

RESUMO

Conjugated linoleic acid (CLA) has been shown to activate the nuclear receptor PPAR-γ and modulate metabolic and immune functions. Despite the worldwide use of CLA dietary supplementation, strong scientific evidence for its proposed beneficial actions are missing. We found that CLA-supplemented diet reduced mucosal damage and inflammatory infiltrate in the dextran sodium sulfate (DSS)-induced colitis model. Conditional deletion of PPAR-γ in macrophages from mice supplemented with CLA diet resulted in loss of this protective effect of CLA, suggesting a PPAR-γ-dependent mechanism mediated by macrophages. However, CLA supplementation significantly worsened colorectal tumor formation induced by azoxymethane and DSS by inducing macrophage and T-cell-producing TGF-ß via PPAR-γ activation. Accordingly, either macrophage-specific deletion of PPAR-γ or in vivo neutralization of latency-associated peptide (LAP, a membrane-bound TGF-ß)-expressing cells abrogated the protumorigenic effect of CLA. Thus, the anti-inflammatory properties of CLA are associated with prevention of colitis but also with development of colorectal cancer.


Assuntos
Colite/imunologia , Neoplasias Colorretais/imunologia , Doenças Inflamatórias Intestinais/imunologia , Ácidos Linoleicos Conjugados/metabolismo , Macrófagos/imunologia , PPAR gama/metabolismo , Linfócitos T/imunologia , Ácido Aminossalicílico/metabolismo , Animais , Carcinogênese , Células Cultivadas , Colite/induzido quimicamente , Neoplasias Colorretais/induzido quimicamente , Sulfato de Dextrana , Suplementos Nutricionais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR gama/genética , Fator de Crescimento Transformador beta/metabolismo
7.
Sci Rep ; 7(1): 6471, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28743871

RESUMO

MarR family proteins are transcriptional regulators that control expression of bacterial proteins involved in metabolism, virulence, stress responses and multi-drug resistance, mainly via ligand-mediated attenuation of DNA binding. Greater understanding of their underlying regulatory mechanism may open up new avenues for the effective treatment of bacterial infections. To gain molecular insight into the mechanism of Rv2887, a MarR family protein in M. tuberculosis, we first showed that it binds salicylate (SA) and para-aminosalicylic acid (PAS), its structural analogue and an antitubercular drug, in a 1:1 stoichiometry with high affinity. Subsequent determination and analysis of Rv2887 crystal structures in apo form, and in complex with SA, PAS and DNA showed that SA and PAS bind to Rv2887 at similar sites, and that Rv2887 interacts with DNA mainly by insertion of helix α4 into the major groove. Ligand binding triggers rotation of the wHTH domain of Rv2887 toward the dimerization domain, causing changes in protein conformation such that it can no longer bind to a 27 bp recognition sequence in the upstream region of gene Rv0560c. The structures provided here lay a foundation for the design of small molecules that target Rv2887, a potential new approach for the development of anti-mycobacterials.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Mycobacterium tuberculosis/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Ácido Aminossalicílico/metabolismo , Proteínas de Bactérias/genética , Sítios de Ligação , Cristalografia por Raios X , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Genfibrozila/metabolismo , Modelos Moleculares , Regiões Promotoras Genéticas , Conformação Proteica , Salicilatos/metabolismo , Fatores de Transcrição/genética
8.
Artigo em Inglês | MEDLINE | ID: mdl-28223391

RESUMO

para-Aminosalicylic acid (PAS) is a second-line antituberculosis drug that has been used to treat multidrug-resistant and extensively drug-resistant tuberculosis for more than 60 years. Renal secretion and glomerular filtration are the major pathways for the elimination of PAS. We comprehensively studied PAS transport by using cell lines that overexpressed various transporters and found that PAS acts as a novel substrate of an organic anionic polypeptide (OATP1B1), organic cationic transporters (OCT1 and OCT2), and organic anion transporters (OAT1 and OAT3) but is not a substrate of any ATP-binding cassette (ABC) transporters. Net PAS uptake was measured, and the transport affinities (Km values) for OATP1B1, OCT1, OCT2, OAT1, and OAT3 were found to be 50.0, 20.3, 28.7, 78.1, and 100.1 µM, respectively. The net uptake rates suggested that renal OAT1 and OAT3 play relatively major roles in PAS elimination. The representative inhibitors rifampin for OATP1B1, probenecid for OAT1 and OAT3, and verapamil for OCT1 and OCT2 greatly inhibited PAS uptake, suggesting that PAS is dependent on multiple transporters for uptake. We also evaluated nonsteroidal anti-inflammatory drugs (NSAIDs), proton pump inhibitors (PPIs), and metformin for the inhibition of PAS uptake via these transporters. Half-maximal (50%) inhibitory concentrations (IC50s) were kinetically determined and used to predict the drug-drug interactions (DDIs) affecting these transporters' activity toward PAS. We found that rifampin, probenecid, ibuprofen, naproxen, cimetidine, and quinidine each exhibited a significant potential for in vivo DDIs with PAS. In this study, PAS was found to be a novel substrate of several transporters, and drugs that inhibit these transporters can reduce PAS elimination.


Assuntos
Ácido Aminossalicílico/metabolismo , Ácido Aminossalicílico/farmacocinética , Anti-Inflamatórios não Esteroides/farmacologia , Antituberculosos/farmacocinética , Transportador 1 de Ânion Orgânico Específico do Fígado/metabolismo , Fator 1 de Transcrição de Octâmero/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Transportador 2 de Cátion Orgânico/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Ácido Aminossalicílico/farmacologia , Antituberculosos/metabolismo , Antituberculosos/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Linhagem Celular , Cimetidina/farmacologia , Interações Medicamentosas/fisiologia , Células HEK293 , Humanos , Ibuprofeno/farmacologia , Transportador 1 de Ânion Orgânico Específico do Fígado/antagonistas & inibidores , Naproxeno/farmacologia , Fator 1 de Transcrição de Octâmero/antagonistas & inibidores , Proteína 1 Transportadora de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos Sódio-Independentes/antagonistas & inibidores , Transportador 2 de Cátion Orgânico/antagonistas & inibidores , Probenecid/farmacologia , Inibidores da Bomba de Prótons/farmacologia , Quinidina/farmacologia , Rifampina/farmacologia , Verapamil/farmacologia
9.
Antimicrob Agents Chemother ; 60(12): 7505-7508, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27671064

RESUMO

Mycobacterium tuberculosis arylamine N-acetyltransferase (TBNAT) is able to acetylate para-aminosalicylic acid (PAS) both in vitro and in vivo as determined by high-performance liquid chromatography (HPLC) and electrospray ionization-mass spectrometry (ESI-MS) techniques. The antituberculosis activity of the acetylated PAS is significantly reduced. As a result, overexpression of TBNAT in M. tuberculosis results in PAS resistance, as determined by MIC tests and drug exposure experiments. Taken together, our results suggest that TBNAT from M. tuberculosis is able to inactivate PAS by acetylating the compound.


Assuntos
Ácido Aminossalicílico/metabolismo , Antituberculosos/metabolismo , Arilamina N-Acetiltransferase/metabolismo , Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/enzimologia , Acetilação , Ácido Aminossalicílico/química , Ácido Aminossalicílico/isolamento & purificação , Ácido Aminossalicílico/farmacologia , Antituberculosos/química , Antituberculosos/isolamento & purificação , Antituberculosos/farmacologia , Arilamina N-Acetiltransferase/genética , Proteínas de Bactérias/genética , Cromatografia Líquida de Alta Pressão , Expressão Gênica , Inativação Metabólica , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , Espectrometria de Massas por Ionização por Electrospray
10.
Science ; 339(6115): 88-91, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23118010

RESUMO

Folate biosynthesis is an established anti-infective target, and the antifolate para-aminosalicylic acid (PAS) was one of the first anti-infectives introduced into clinical practice on the basis of target-based drug discovery. Fifty years later, PAS continues to be used to treat tuberculosis. PAS is assumed to inhibit dihydropteroate synthase (DHPS) in Mycobacterium tuberculosis by mimicking the substrate p-aminobenzoate (PABA). However, we found that sulfonamide inhibitors of DHPS inhibited growth of M. tuberculosis only weakly because of their intracellular metabolism. In contrast, PAS served as a replacement substrate for DHPS. Products of PAS metabolism at this and subsequent steps in folate metabolism inhibited those enzymes, competing with their substrates. PAS is thus a prodrug that blocks growth of M. tuberculosis when its active forms are generated by enzymes in the pathway they poison.


Assuntos
Ácido Aminossalicílico/metabolismo , Antituberculosos/metabolismo , Di-Hidropteroato Sintase/metabolismo , Ácido Fólico/metabolismo , Mycobacterium tuberculosis/metabolismo , Pró-Fármacos/metabolismo , Ácido Aminossalicílico/farmacologia , Antituberculosos/farmacologia , Di-Hidropteroato Sintase/antagonistas & inibidores , Mimetismo Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Pró-Fármacos/farmacologia , Especificidade por Substrato
11.
Drug Metab Dispos ; 39(5): 882-90, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21317369

RESUMO

Human arylamine N-acetyltransferase 2 (NAT2) mediates the biotransformation of arylamine drugs and procarcinogens into either innocuous or reactive DNA-damaging metabolites and is expressed predominantly in liver. Interspecies differences and incongruous results between in vitro, in vivo, and epidemiological studies make it difficult to extrapolate animal results to human risk. We have generated human NAT2 transgenic mice on both C57BL/6 (hNAT2(tg)) and Nat1/2 null backgrounds [hNAT2(tg)Nat1/2(-/-)], in which liver-selective expression of human NAT2 is driven by the mouse albumin promoter. We detected expression of the human NAT2 transcript and protein in mouse liver by real-time PCR and Western blot analysis. NAT2 enzyme activity, measured using the human NAT2-selective substrate sulfamethazine (SMZ), was 40- to 80-fold higher in liver cytosols from hNAT2(tg)Nat1/2(-/-) mice than in wild-type mice. An unexpected gender difference was observed, with males displaying 2-fold higher activity than females. Transgenic mice also had an increased in vivo plasma clearance of SMZ and higher levels of N-acetylated SMZ than wild-type mice. Liver expression of human NAT2 did not affect the disposition of the human NAT1-selective substrate p-aminosalicylic acid (PAS), because hNAT2(tg)Nat1/2(-/-) mice displayed in vivo PAS pharmacokinetic profiles similar to those of Nat1/2(-/-) mice. The metabolism of 4-aminobiphenyl was similar between hNAT2(tg)Nat1/2(-/-) and wild-type mice with the exception of a more liver-restricted pattern in hNAT2(tg)Nat1/2(-/-) mice and lower activity in females. Overall, the hNAT2(tg)Nat1/2(-/-) mouse mimics human expression of NAT2 and may thus be of value in clarifying the role of human NAT2 in arylamine clearance, detoxification, and bioactivation.


Assuntos
Ácido Aminossalicílico/metabolismo , Anti-Infecciosos/metabolismo , Antituberculosos/metabolismo , Arilamina N-Acetiltransferase/biossíntese , Fígado/enzimologia , Sulfametazina/metabolismo , Ácido Aminossalicílico/sangue , Ácido Aminossalicílico/farmacocinética , Animais , Anti-Infecciosos/sangue , Anti-Infecciosos/farmacocinética , Antituberculosos/sangue , Antituberculosos/farmacocinética , Arilamina N-Acetiltransferase/sangue , Arilamina N-Acetiltransferase/genética , Citosol/metabolismo , Feminino , Genótipo , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Sulfametazina/farmacocinética
12.
J Pharm Biomed Anal ; 54(5): 1101-9, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21159459

RESUMO

Para-aminosalicylic acid (PAS), an approved drug for treatment of tuberculosis, is a promising therapeutic agent for treatment of manganese (Mn)-induced parkinsonian syndromes. Lack of a quantifying method, however, has hindered the clinical evaluation of its efficacy and there upon new drug development. This study was aimed at developing a simple and effective method to quantify PAS and its major metabolite, N-acetyl-para-aminosalicylic acid (AcPAS), in plasma, cerebrospinal fluid (CSF) and tissues. Biological samples underwent one-step protein precipitation. The supernatant was fractionated on a reversed-phase C18 column with a gradient mobile system, followed by on-line fluorescence detection. The lower limits of quantification for both PAS and AcPAS were 50 ng/ml of plasma and 17 ng/g of tissues. The intra-day and inter-day precision values did not exceed 5% and 8%, respectively, in all three matrices. The method was used to quantify PAS and AcPAS in rat plasma and brain following a single iv injection of PAS. Data showed a greater amount of PAS than AcPAS in plasma, while a greater amount of AcPAS than PAS was found in brain tissues. The method has been proven to be sensitive, reproducible, and practically useful for laboratory and clinical investigations of PAS in treatment of Mn Parkinsonism.


Assuntos
Ácido Aminossalicílico/análise , Ácido Aminossalicílico/metabolismo , Encéfalo/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Monitoramento de Medicamentos/métodos , Ácido Aminossalicílico/sangue , Ácido Aminossalicílico/líquido cefalorraquidiano , Animais , Calibragem , Estabilidade de Medicamentos , Injeções Intravenosas , Ratos , Ratos Sprague-Dawley , Padrões de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual
13.
Redox Rep ; 15(6): 259-67, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21208525

RESUMO

Sulfasalazine is a prodrug composed by a molecule of 5-aminosalicylic acid (5-ASA) and sulfapyridine (SP), linked by an azo bond, which has been shown to be effective in the therapy of inflammatory bowel diseases (IBD) such as ulcerative colitis and Crohn's disease, as well as of rheumatic diseases, such as rheumatoid arthritis and ankylosing spondylitis. The precise mechanism of action of sulfasalazine and/or its metabolites has not been completely elucidated, though its antioxidant effects are well established and are probably due to its scavenging effects against reactive oxygen and nitrogen species (ROS and RNS), as well as metal chelating properties, in association to its inhibitory effects over neutrophil oxidative burst. The present work was focused on screening and comparing the potential scavenging activity for an array of ROS (O(2)(•-), H(2)O(2), (1)O(2), ROO(•) and HOCl) and RNS ((•)NO and ONOO(-)), mediated by sulfasalazine and its metabolites 5-ASA and SP, using validated in vitro screening systems. The results showed that both 5-ASA and sulfasalazine were able to scavenge all the tested ROS while SP was practically ineffective in all the assays. For HOCl, (1)O(2), and ROO(•), 5-ASA showed the best scavenging effects. A new and important finding of the present study was the strong scavenging effect of 5-ASA against (1)O(2). 5-ASA was shown to be a strong scavenger of (•)NO and ONOO(-). Sulfasalazine was also able to scavenge these RNS, although with a much lower potency than 5-ASA. SP was unable to scavenge (•)NO in the tested concentrations but was shown to scavenge ONOO(-), with a higher strength when the assay was performed in the presence of 25 mM bicarbonate, suggesting further scavenging of oxidizing carbonate radical. In conclusion, the ROS- and RNS-scavenging effects of sulfasalazine and its metabolites shown in this study may contribute to the anti-inflammatory effects mediated by sulfasalazine through the prevention of the oxidative/nitrative/nitrosative damages caused by these species.


Assuntos
Ácido Aminossalicílico/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sulfapiridina/metabolismo , Sulfassalazina/metabolismo , Ácido Aminossalicílico/química , Peróxido de Hidrogênio/química , Estrutura Molecular , Espécies Reativas de Nitrogênio/química , Espécies Reativas de Oxigênio/química , Sulfapiridina/química , Sulfassalazina/química , Superóxidos/química
14.
APMIS ; 116(4): 326-32, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18397469

RESUMO

We report the clinical and histopathological characteristics of two cases of signet ring cell carcinoma of the eye lids, and discuss the histogenesis of this neoplasm. Two 72-year-old Caucasian males both presented with slowly growing tumours of the eyelids. The tumours were excised and specimens were examined using light- and transmission electron microscopic techniques. Clinically, the tumours infiltrated both eyelids on one side of the face with swelling and periocular inflammation, creating a monocle-like appearance. Extensive clinical work-up excluded periocular metastases. Histopathologically, the tumours were composed of rather bland cells with mainly histiocytoid morphology. A minor proportion had a signet ring cell appearance. The cytoplasmic inclusions giving the signet ring morphology were PAS- and colloidal iron positive. The tumour cells reacted with antibodies against cytokeratins, carcinoembryonic antigen, epithelial membrane antigen, gross cystic disease fluid protein-15 and lysozyme. Transmission electron microscopy demonstrated tumour cells containing intracytoplasmic vacuoles lined by microvilli. The tumour cells aggregated in duct-like clusters. A diagnosis of primary signet ring cell carcinoma was made in both cases. Histopathological, immunohistological and ultrastructural findings indicated that the tumours were of sweat gland origin.


Assuntos
Carcinoma de Células em Anel de Sinete/metabolismo , Carcinoma de Células em Anel de Sinete/patologia , Neoplasias Palpebrais/metabolismo , Neoplasias Palpebrais/patologia , Neoplasias das Glândulas Sudoríparas/metabolismo , Neoplasias das Glândulas Sudoríparas/patologia , Idoso , Ácido Aminossalicílico/metabolismo , Antígeno Carcinoembrionário/metabolismo , Proteínas de Transporte/metabolismo , Glicoproteínas/metabolismo , Histocitoquímica , Humanos , Imuno-Histoquímica , Corpos de Inclusão Intranuclear/patologia , Corpos de Inclusão Intranuclear/ultraestrutura , Compostos de Ferro/metabolismo , Queratinas/metabolismo , Masculino , Proteínas de Membrana Transportadoras , Microvilosidades/ultraestrutura , Mucina-1/metabolismo , Muramidase/metabolismo
15.
Prog Brain Res ; 164: 3-22, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17920423

RESUMO

The development of cerebral cortex includes highly organized, elaborate and long-lasting series of events, which do not come to an end by the time of birth. Indeed, many developmental events continue after the 40th postconceptual week resulting in a long morphological, behavioral and cognitive development of children. Premature birth causes an untimely dramatic change in the environment of the human fetus and often results in serious threats for life. Cognitive abilities of prematurely born children vary, but a correlation between cognitive impairment and the time of birth is evident. In this study we review the morphological evidence of cortical maturation in preterm and full-term infants. Various aspects of postnatal cortical development including cell proliferation and maturation of neurons in the temporal archi- and neocortex are discussed and compared in preterm infants and age-matched full-term controls. Our results suggest that cell proliferation and maturation are not influenced by the preterm delivery. In contrast, the perinatal decrease of the number of Cajal-Retzius cells might be regulated by a mechanism that is affected by preterm birth. We demonstrate that cognitive deficiencies of the prematurely born infants cannot be explained with light microscopically observed alteration of proliferation and maturation of neurons.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Diferenciação Celular/fisiologia , Proliferação de Células , Neurônios/fisiologia , Nascimento Prematuro/patologia , Ácido Aminossalicílico/metabolismo , Bromodesoxiuridina/metabolismo , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Antígeno Ki-67/metabolismo , Masculino , Proteínas do Tecido Nervoso/metabolismo , Mudanças Depois da Morte
16.
Proc Natl Acad Sci U S A ; 104(18): 7391-6, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17463090

RESUMO

S3I-201 (NSC 74859) is a chemical probe inhibitor of Stat3 activity, which was identified from the National Cancer Institute chemical libraries by using structure-based virtual screening with a computer model of the Stat3 SH2 domain bound to its Stat3 phosphotyrosine peptide derived from the x-ray crystal structure of the Stat3beta homodimer. S3I-201 inhibits Stat3.Stat3 complex formation and Stat3 DNA-binding and transcriptional activities. Furthermore, S3I-201 inhibits growth and induces apoptosis preferentially in tumor cells that contain persistently activated Stat3. Constitutively dimerized and active Stat3C and Stat3 SH2 domain rescue tumor cells from S3I-201-induced apoptosis. Finally, S3I-201 inhibits the expression of the Stat3-regulated genes encoding cyclin D1, Bcl-xL, and survivin and inhibits the growth of human breast tumors in vivo. These findings strongly suggest that the antitumor activity of S3I-201 is mediated in part through inhibition of aberrant Stat3 activation and provide the proof-of-concept for the potential clinical use of Stat3 inhibitors such as S3I-201 in tumors harboring aberrant Stat3.


Assuntos
Ácidos Aminossalicílicos , Antineoplásicos/química , Antineoplásicos/metabolismo , Benzenossulfonatos/química , Benzenossulfonatos/metabolismo , Avaliação Pré-Clínica de Medicamentos , Fator de Transcrição STAT3/química , Fator de Transcrição STAT3/metabolismo , Ácido Aminossalicílico/química , Ácido Aminossalicílico/metabolismo , Ácido Aminossalicílico/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Apoptose , Benzenossulfonatos/uso terapêutico , Linhagem Celular , Biologia Computacional , DNA/química , DNA/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosfotirosina/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Transcrição Gênica/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Drug Metab Dispos ; 35(7): 1064-70, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17403913

RESUMO

Arylamine N-acetyltransferases (NAT) catalyze the biotransformation of many important arylamine drugs and procarcinogens. NAT can either detoxify or activate procarcinogens, complicating the manner in which these enzymes may participate in enhancing or preventing toxic responses to particular agents. Mice possess three NAT isoenzymes: Nat1, Nat2, and Nat3. Whereas Nat1 and Nat2 can efficiently acetylate many arylamines, few substrates appear to be appreciably metabolized by Nat3. We generated a Nat3 knockout mouse strain and used it along with our double Nat1/2(-/-) knockout strain to further investigate the functional role of Nat3. Nat3(-/-) mice showed normal viability and reproductive capacity. Nat3 expression was very low in wild-type animals and completely undetectable in Nat3(-/-) mice. In contrast, greatly elevated expression of Nat3 transcript was observed in Nat1/2(-/-) mice. We used a transcribed marker polymorphism approach to establish that the increased expression of Nat3 in Nat1/2(-/-) mice is a positional artifact of insertion of the phosphoglycerate kinase-neomycin resistance cassette in place of the Nat1/Nat2 gene region and upstream of the intact Nat3 gene, rather than a biological compensatory mechanism. Despite the increase in Nat3 transcript, the N-acetylation of p-aminosalicylate, sulfamethazine, 2-aminofluorene, and 4-aminobiphenyl was undetectable either in vivo or in vitro in Nat1/2(-/-) animals. In parallel, no difference was observed in the in vivo clearance or in vitro metabolism of any of these substrates between wild-type and Nat3(-/-) mice. Thus, Nat3 is unlikely to play a significant role in the N-acetylation of arylamines either in wild-type mice or in mice lacking Nat1 and Nat2 activities.


Assuntos
Artefatos , Arilamina N-Acetiltransferase/metabolismo , Regulação Enzimológica da Expressão Gênica , Isoenzimas/metabolismo , Acetilação , Compostos de Aminobifenil/metabolismo , Ácido Aminossalicílico/metabolismo , Animais , Arilamina N-Acetiltransferase/deficiência , Arilamina N-Acetiltransferase/genética , Feminino , Fluorenos/metabolismo , Isoenzimas/deficiência , Isoenzimas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Fatores Sexuais , Especificidade por Substrato , Sulfametazina/metabolismo
18.
Int J Syst Evol Microbiol ; 54(Pt 5): 1723-1728, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15388735

RESUMO

A group of moderately halophilic bacteria able to degrade aromatic organic compounds contaminating hypersaline habitats in southern Spain have been isolated and characterized. The taxonomic position of these strains was determined using phenotypic, phylogenetic and genotypic methods. The G + C content of their DNA ranged from 61.0 to 62.9 mol%. DNA-DNA hybridization studies showed that they constitute a genospecies, having DNA-DNA hybridization values of 90-100 %. Analysis of the complete 16S rRNA gene sequence revealed a high level of similarity with members of the genus Halomonas, sharing 98 % 16S rRNA gene sequence similarity with the type strains of Halomonas salina and Halomonas halophila. However, phenotypic differences and the low level of DNA-DNA hybridization suggest the placement of these strains as a novel species within the genus Halomonas. The name Halomonas organivorans sp. nov. is proposed, with strain G-16.1T (= CECT 5995T = CCM 7142T) as the type strain. This novel species of Halomonas is characterized by its ability to use a wide range of organic compounds (benzoic acid, p-hydroxybenzoic acid, cinnamic acid, salicylic acid, phenylacetic acid, phenylpropionic acid, phenol, p-coumaric acid, ferulic acid and p-aminosalicylic acid), and it could be useful for the decontamination of polluted saline habitats.


Assuntos
Halomonas/classificação , Halomonas/metabolismo , Hidrocarbonetos Aromáticos/metabolismo , Ácido Aminossalicílico/metabolismo , Técnicas de Tipagem Bacteriana , Composição de Bases , Ácido Benzoico/metabolismo , Biodegradação Ambiental , Cinamatos/metabolismo , Ácidos Cumáricos/metabolismo , DNA Bacteriano/química , DNA Bacteriano/isolamento & purificação , DNA Ribossômico/química , DNA Ribossômico/isolamento & purificação , Genes de RNAr/genética , Halomonas/genética , Halomonas/isolamento & purificação , Dados de Sequência Molecular , Hibridização de Ácido Nucleico , Parabenos/metabolismo , Fenol/metabolismo , Fenilacetatos/metabolismo , Filogenia , Propionatos , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Ácido Salicílico/metabolismo , Análise de Sequência de DNA , Microbiologia do Solo , Espanha
19.
Neuromuscul Disord ; 14(4): 253-60, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15019703

RESUMO

Glycogen storage disease type IV or Andersen disease is an autosomal recessive disorder due to deficiency of glycogen branching enzyme. Typically, glycogen storage disease type IV presents with rapidly progressive liver cirrhosis and death in childhood. Variants include a cardiopathic form of childhood, a relatively benign myopathic form of young adults, and a late-onset neurodegenerative disorder (adult polyglucosan body disease). A severe neuromuscular variant resembling Werdnig-Hoffmann disease has also been described in two patients. The objective was to describe two additional infants with the neuromuscular variant and novel mutations in the GBE1 gene. Branching enzyme assay, Western blot, RT-PCR and sequencing were performed in muscle biopsies from both patients. The cDNA of patient 1 was subcloned and sequenced to define the mutations. Muscle biopsies showed accumulation of periodic acid Schiff-positive, diastase-resistant storage material in both patients and increased lysosomal enzyme activity in patient 1. Branching enzyme activity in muscle was negligible in both patients, and Western blot showed decreased branching enzyme protein. Patient 1 had two single base pair deletions, one in exon 10 (1238delT) and the other in exon 12 (1467delC), and each parent was heterozygous for one of the deletions. Patient 2 had a large homozygous deletion that spanned 627 bp and included exons 8-12. Patient 1, who died at 41 days, had neurophysiological and neuropathological features of Spinal Muscular Atrophy. Patient 2, who died at 5(1/2) weeks, had a predominantly myopathic process. The infantile neuromuscular form of glycogen storage disease type IV is considered extremely rare, but our encountering two patients in close succession suggests that the disease may be underdiagnosed.


Assuntos
Doença de Depósito de Glicogênio Tipo IV , Músculo Esquelético/patologia , Doenças Neuromusculares , Enzima Ramificadora de 1,4-alfa-Glucana/metabolismo , Ácido Aminossalicílico/metabolismo , Biópsia/métodos , Western Blotting/métodos , Tronco Encefálico/enzimologia , Tronco Encefálico/patologia , Análise Mutacional de DNA/métodos , Éxons , Feminino , Doença de Depósito de Glicogênio Tipo IV/complicações , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Humanos , Lactente , Oxirredutases Intramoleculares , Lisossomos/enzimologia , Microscopia Eletrônica/métodos , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestrutura , Mutação , Doenças Neuromusculares/enzimologia , Doenças Neuromusculares/patologia , Prostaglandina-E Sintases , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Atrofias Musculares Espinais da Infância/complicações , Atrofias Musculares Espinais da Infância/patologia , Transativadores/genética
20.
J Exp Clin Cancer Res ; 23(4): 661-7, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15743037

RESUMO

The purpose of the study was to establish the subcutaneous model of human extrahepatic bile duct carcinoma in nude mice so as to provide a suitable model for the study of extrahepatic bile duct carcinoma. Surgical specimens of the patient with extrahepatic bile duct carcinoma were transplanted into the subcutaneous layer of nude mice. Growth curve of transplanted tumors was drawn and its morphological and biological characteristics, as well as choromosome were observed. A well differentiated mucinous adenocarcinoma model of human bile duct carcinoma in nude mice, designated as HBDCM1-ZSH (Human Bile Duct Carcinoma Model No. 1 established by Zhong Shan Hospital in April, 2001), was established via subcutaneous transplantation of the surgically resected tumor from a 56-year-old Chinese man. HBDCM1-ZSH has been maintained for 13 passages and exhibited 98.1% transplantability. Mean latent periods were 26 days. Transplanted tumors exhibited the characteristics of the original tumor in morphology and biology. Chromosomal analysis revealed numerical abnormalities ranging from 67 to 84. HBDCM1-ZSH expressed carcinoembryonic antigen (CEA), carbohydrate antigen (CA)19-9, cytokeratin (CK7, CK19, CK20), PCNA, AB and PAS. In conclusion, HBDCM1-ZSH is similar to human extrahepatic bile duct carcinoma and provides an applicable animal model for research on extrahepatic bile duct carcinoma.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Carcinoma/patologia , Modelos Animais de Doenças , Transplante de Neoplasias/métodos , Ácido Aminossalicílico/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Antígeno CA-19-9/metabolismo , Antígeno Carcinoembrionário/metabolismo , Bandeamento Cromossômico , Mapeamento Cromossômico , DNA/metabolismo , Humanos , Imuno-Histoquímica , Proteínas de Filamentos Intermediários/metabolismo , Cariotipagem , Queratina-20 , Queratina-7 , Queratinas/metabolismo , Cinética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Eletrônica , Pessoa de Meia-Idade , Antígeno Nuclear de Célula em Proliferação/metabolismo , Radioimunoensaio , Fatores de Tempo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...