Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 60(7): 3980-7, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27090174

RESUMO

Amixicile is a promising derivative of nitazoxanide (an antiparasitic therapeutic) developed to treat systemic infections caused by anaerobic bacteria, anaerobic parasites, and members of the Epsilonproteobacteria (Campylobacter and Helicobacter). Amixicile selectively inhibits pyruvate-ferredoxin oxidoreductase (PFOR) and related enzymes by inhibiting the function of the vitamin B1 cofactor (thiamine pyrophosphate) by a novel mechanism. Here, we interrogate the amixicile scaffold, guided by docking simulations, direct PFOR inhibition assays, and MIC tests against Clostridium difficile, Campylobacter jejuni, and Helicobacter pylori Docking simulations revealed that the nitro group present in nitazoxanide interacts with the protonated N4'-aminopyrimidine of thiamine pyrophosphate (TPP). The ortho-propylamine on the benzene ring formed an electrostatic interaction with an aspartic acid moiety (B456) of PFOR that correlated with improved PFOR-inhibitory activity and potency by MIC tests. Aryl substitution with electron-withdrawing groups and substitutions of the propylamine with other alkyl amines or nitrogen-containing heterocycles both improved PFOR inhibition and, in many cases, biological activity against C. difficile Docking simulation results correlate well with mechanistic enzymology and nuclear magnetic resonance (NMR) studies that show members of this class of antimicrobials to be specific inhibitors of vitamin B1 function by proton abstraction, which is both novel and likely to limit mutation-based drug resistance.


Assuntos
Antibacterianos/síntese química , Antibacterianos/farmacologia , Bactérias Anaeróbias/efeitos dos fármacos , Benzamidas/síntese química , Benzamidas/farmacologia , Inibidores Enzimáticos/farmacologia , Epsilonproteobacteria/efeitos dos fármacos , Ferredoxinas/metabolismo , Oxirredutases/antagonistas & inibidores , Ácido Pirúvico/metabolismo , Tiazóis/síntese química , Tiazóis/farmacologia , Antibacterianos/química , Bactérias Anaeróbias/metabolismo , Benzamidas/química , Campylobacter jejuni/efeitos dos fármacos , Campylobacter jejuni/metabolismo , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Epsilonproteobacteria/metabolismo , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/metabolismo , Oxirredutases/metabolismo , Tiazóis/química
2.
Antimicrob Agents Chemother ; 58(8): 4703-12, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24890599

RESUMO

Amixicile shows efficacy in the treatment of Clostridium difficile infections (CDI) in a mouse model, with no recurrence of CDI. Since amixicile selectively inhibits the action of a B vitamin (thiamine pyrophosphate) cofactor of pyruvate:ferredoxin oxidoreductase (PFOR), it may both escape mutation-based drug resistance and spare beneficial probiotic gut bacteria that do not express this enzyme. Amixicile is a water-soluble derivative of nitazoxanide (NTZ), an antiparasitic therapeutic that also shows efficacy against CDI in humans. In comparative studies, amixicile showed no toxicity to hepatocytes at 200 µM (NTZ was toxic above 10 µM); was not metabolized by human, dog, or rat liver microsomes; showed equivalence or superiority to NTZ in cytochrome P450 assays; and did not activate efflux pumps (breast cancer resistance protein, P glycoprotein). A maximum dose (300 mg/kg) of amixicile given by the oral or intraperitoneal route was well tolerated by mice and rats. Plasma exposure (rats) based on the area under the plasma concentration-time curve was 79.3 h · µg/ml (30 mg/kg dose) to 328 h · µg/ml (100 mg/kg dose), the maximum concentration of the drug in serum was 20 µg/ml, the time to the maximum concentration of the drug in serum was 0.5 to 1 h, and the half-life was 5.6 h. Amixicile did not concentrate in mouse feces or adversely affect gut populations of Bacteroides species, Firmicutes, segmented filamentous bacteria, or Lactobacillus species. Systemic bioavailability was demonstrated through eradication of Helicobacter pylori in a mouse infection model. In summary, the efficacy of amixicile in treating CDI and other infections, together with low toxicity, an absence of mutation-based drug resistance, and excellent drug metabolism and pharmacokinetic metrics, suggests a potential for broad application in the treatment of infections caused by PFOR-expressing microbial pathogens in addition to CDI.


Assuntos
Antibacterianos/farmacocinética , Benzamidas/farmacocinética , Infecções por Helicobacter/tratamento farmacológico , Helicobacter pylori/efeitos dos fármacos , Tiazóis/farmacocinética , Animais , Antibacterianos/sangue , Antibacterianos/farmacologia , Área Sob a Curva , Benzamidas/sangue , Benzamidas/farmacologia , Disponibilidade Biológica , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cães , Avaliação Pré-Clínica de Medicamentos , Meia-Vida , Infecções por Helicobacter/sangue , Infecções por Helicobacter/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Masculino , Testes de Sensibilidade Microbiana , Microbiota/efeitos dos fármacos , Microbiota/fisiologia , Microssomos Hepáticos/efeitos dos fármacos , Piruvato Sintase/metabolismo , Ratos , Tiamina Pirofosfato/metabolismo , Tiazóis/sangue , Tiazóis/farmacologia
3.
Antimicrob Agents Chemother ; 56(8): 4103-11, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22585229

RESUMO

Clostridium difficile infection (CDI) is a serious diarrheal disease that often develops following prior antibiotic usage. One of the major problems with current therapies (oral vancomycin and metronidazole) is the high rate of recurrence. Nitazoxanide (NTZ), an inhibitor of pyruvate:ferredoxin oxidoreductase (PFOR) in anaerobic bacteria, parasites, Helicobacter pylori, and Campylobacter jejuni, also shows clinical efficacy against CDI. From a library of ∼250 analogues of NTZ, we identified leads with increased potency for PFOR. MIC screens indicated in vitro activity in the 0.05- to 2-µg/ml range against C. difficile. To improve solubility, we replaced the 2-acetoxy group with propylamine, producing amixicile, a soluble (10 mg/ml), nontoxic (cell-based assay) lead that produced no adverse effects in mice by oral or intraperitoneal (i.p.) routes at 200 mg/kg of body weight/day. In initial efficacy testing in mice treated (20 mg/kg/day, 5 days each) 1 day after receiving a lethal inoculum of C. difficile, amixicile showed slightly less protection than did vancomycin by day 5. However, in an optimized CDI model, amixicile showed equivalence to vancomycin and fidaxomicin at day 5 and there was significantly greater survival produced by amixicile than by the other drugs on day 12. All three drugs were comparable by measures of weight loss/gain and severity of disease. Recurrence of CDI was common for mice treated with vancomycin or fidaxomicin but not for mice receiving amixicile or NTZ. These results suggest that gut repopulation with beneficial (non-PFOR) bacteria, considered essential for protection against CDI, rebounds much sooner with amixicile therapy than with vancomycin or fidaxomicin. If the mouse model is indeed predictive of human CDI disease, then amixicile, a novel PFOR inhibitor, appears to be a very promising new candidate for treatment of CDI.


Assuntos
Antibacterianos/farmacologia , Benzamidas/farmacologia , Clostridioides difficile/efeitos dos fármacos , Infecções por Clostridium/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Piruvato Sintase/antagonistas & inibidores , Tiazóis/farmacologia , Aminoglicosídeos/farmacologia , Animais , Antibacterianos/uso terapêutico , Benzamidas/uso terapêutico , Clostridioides difficile/enzimologia , Infecções por Clostridium/microbiologia , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Fidaxomicina , Camundongos , Testes de Sensibilidade Microbiana , Nitrocompostos , Tiazóis/química , Tiazóis/uso terapêutico , Resultado do Tratamento , Vancomicina/farmacologia
4.
Biochem J ; 440(3): 345-53, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21848514

RESUMO

S1P (sphingosine 1-phosphate) is a signalling molecule involved in a host of cellular and physiological functions, most notably cell survival and migration. S1P, which signals via a set of five G-protein-coupled receptors (S1P1-S1P5), is formed by the action of two SphKs (sphingosine kinases) from Sph (sphingosine). Interfering RNA strategies and SphK1 (sphingosine kinase type 1)-null (Sphk1-/-) mouse studies implicate SphK1 in multiple signalling cascades, yet there is a paucity of potent and selective SphK1 inhibitors necessary to evaluate the effects of rapid onset inhibition of this enzyme. We have identified a set of submicromolar amidine-based SphK1 inhibitors and report using a pair of these compounds to probe the cellular and physiological functions of SphK1. In so doing, we demonstrate that our inhibitors effectively lower S1P levels in cell-based assays, but we have been unable to correlate SphK1 inhibition with changes in cell survival. However, SphK1 inhibition did diminish EGF (epidermal growth factor)-driven increases in S1P levels and Akt (also known as protein kinase B)/ERK (extracellular-signal-regulated kinase) phosphorylation. Finally, administration of the SphK1 inhibitor to wild-type, but not Sphk1-/-, mice resulted in a rapid decrease in blood S1P levels indicating that circulating S1P is rapidly turned over.


Assuntos
Amidinas/farmacologia , Lisofosfolipídeos/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Pirrolidinas/farmacologia , Esfingosina/análogos & derivados , Amidinas/farmacocinética , Animais , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Lisofosfolipídeos/sangue , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirrolidinas/farmacocinética , Ratos , Esfingolipídeos/metabolismo , Esfingosina/sangue , Esfingosina/metabolismo , Estereoisomerismo
5.
J Pharmacol Exp Ther ; 338(3): 879-89, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21632869

RESUMO

Sphingosine 1-phosphate (S1P) is a phospholipid that binds to a set of G protein-coupled receptors (S1P(1)-S1P(5)) to initiate an array of signaling cascades that affect cell survival, differentiation, proliferation, and migration. On a larger physiological scale, the effects of S1P on immune cell trafficking, vascular barrier integrity, angiogenesis, and heart rate have also been observed. An impetus for the characterization of S1P-initiated signaling effects came with the discovery that FTY720 [fingolimod; 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol] modulates the immune system by acting as an agonist at S1P(1). In the course of structure-activity relationship studies to better understand the functional chemical space around FTY720, we discovered conformationally constrained FTY720 analogs that behave as S1P receptor type-selective antagonists. Here, we present a pharmacological profile of a lead S1P(1/3) antagonist prodrug, 1-(hydroxymethyl)-3-(3-octylphenyl)cyclobutane (VPC03090). VPC03090 is phosphorylated by sphingosine kinase 2 to form the competitive antagonist species 3-(3-octylphenyl)-1-(phosphonooxymethyl)cyclobutane (VPC03090-P) as observed in guanosine 5'-O-(3-[(35)S]thio)triphosphate binding assays, with effects on downstream S1P receptor signaling confirmed by Western blot and calcium mobilization assays. Oral dosing of VPC03090 results in an approximate 1:1 phosphorylated/alcohol species ratio with a half-life of 30 h in mice. Because aberrant S1P signaling has been implicated in carcinogenesis, we applied VPC03090 in an immunocompetent mouse mammary cancer model to assess its antineoplastic potential. Treatment with VPC03090 significantly inhibited the growth of 4T1 primary tumors in mice. This result calls to attention the value of S1P receptor antagonists as not only research tools but also potential therapeutic agents.


Assuntos
Derivados de Benzeno/farmacologia , Ciclobutanos/farmacologia , Pró-Fármacos/farmacologia , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Esfingosina/análogos & derivados , Animais , Derivados de Benzeno/farmacocinética , Western Blotting , Células CHO , Cálcio/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Cricetinae , Cricetulus , Ciclobutanos/farmacocinética , Feminino , Cloridrato de Fingolimode , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Contagem de Linfócitos , Linfopenia/sangue , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pró-Fármacos/farmacocinética , Propilenoglicóis/farmacocinética , Conformação Proteica , Ensaio Radioligante , Esfingosina/farmacocinética , Esfingosina/farmacologia , Relação Estrutura-Atividade
6.
Anal Biochem ; 411(2): 230-5, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21216217

RESUMO

Sphingosine kinases (SphKs) catalyze the transfer of phosphate from adenosine triphosphate (ATP) to sphingosine to generate sphingosine 1-phosphate (S1P), an important bioactive lipid molecule that mediates a diverse range of cell signaling processes. The conventional assay of SphK enzymatic activity uses [γ-(32)P]ATP and sphingosine as substrates, with the radiolabeled S1P product recovered by organic extraction, displayed by thin layer chromatography, and quantified by liquid scintillation counting. Although this assay is sensitive and accurate, it is slow and labor-intensive; thus, it precludes the simultaneous screening of more than a few inhibitor compounds. Here we describe a 96-well assay for SphKs that is rapid and reproducible. Our method, which takes advantage of the limited solubility of S1P, detects radioactive S1P adhering to the plate by scintillation proximity counting. Our procedure obviates extraction into organic solvents, postreaction transfers, and chromatography. Furthermore, our assay enables assessment of both inhibitors and substrates, and it can detect endogenous SphK activity in cell and tissue extracts. The SphK kinetic parameter, K(m), and the K(i) values of inhibitors determined with our assay and the conventional assay were indistinguishable. These results document that our assay is well-suited for the screening of chemical libraries of SphK inhibitors.


Assuntos
Inibidores Enzimáticos/análise , Ensaios de Triagem em Larga Escala , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia em Camada Fina , Inibidores Enzimáticos/isolamento & purificação , Cinética , Lisofosfolipídeos/metabolismo , Radioisótopos de Fósforo/química , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Contagem de Cintilação , Bibliotecas de Moléculas Pequenas/química , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Especificidade por Substrato
7.
J Am Soc Nephrol ; 21(6): 955-65, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20338995

RESUMO

Agonists of the sphingosine-1-phosphate receptor (S1PR) attenuate kidney ischemia-reperfusion injury (IRI). Previous studies suggested that S1P1R-induced lymphopenia mediates this protective effect, but lymphocyte-independent mechanisms could also contribute. Here, we investigated the effects of S1PR agonists on kidney IRI in mice that lack T and B lymphocytes (Rag-1 knockout mice). Administration of the nonselective S1PR agonist FTY720 or the selective S1P1R agonist SEW2871 reduced injury in both Rag-1 knockout and wild-type mice. In vitro, SEW2871 significantly attenuated LPS- or hypoxia/reoxygenation-induced apoptosis in cultured mouse proximal tubule epithelial cells, supporting a direct protective effect of S1P1R agonists via mitogen-activated protein kinase and/or Akt pathways. S1P1Rs in the proximal tubule mediated IRI in vivo as well: Mice deficient in proximal tubule S1P1Rs experienced a greater decline in renal function after IRI than control mice and their kidneys were no longer protected by SEW2871 administration. In summary, S1PRs in the proximal tubule are necessary for stress-induced cell survival, and S1P1R agonists are renoprotective via direct effects on the tubule cells. Selective agonists of S1P1Rs may hold therapeutic potential for the prevention and treatment of acute kidney injury.


Assuntos
Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/prevenção & controle , Células Epiteliais/metabolismo , Túbulos Renais Proximais/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células Epiteliais/patologia , Cloridrato de Fingolimode , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Túbulos Renais Proximais/patologia , Leucócitos/efeitos dos fármacos , Leucócitos/patologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Oxidiazóis/farmacologia , Propilenoglicóis/farmacologia , RNA Mensageiro/metabolismo , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Tiofenos/farmacologia
8.
Antimicrob Agents Chemother ; 54(7): 2767-74, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20404119

RESUMO

Coagulase-negative species of Staphylococcus are often associated with opportunistic hospital-acquired infections that arise from the colonization of indwelling catheters. Here we show that the antiparasitic drug nitazoxanide (NTZ) and its active metabolite, tizoxanide (TIZ), are inhibitory to the growth of Staphylococcus epidermidis and other staphylococci, including methicillin-resistant Staphylococcus aureus strains, under aerobic and microaerobic conditions (MICs, 8 to 16 microg/ml). At sub-MIC levels, NTZ and TIZ also inhibited biofilm production under static conditions by strains of S. epidermidis and Staphylococcus haemolyticus with a 50% inhibitory concentration of approximately 2.5 microg/ml (8 microM). The 5-nitro group was required for biological activity, and a hydrophilic derivative of NTZ (AMIX) also inhibited biofilm formation. NTZ did not disperse the existing biofilm but did block further accumulation. Sub-MICs of NTZ had no effect on primary attachment to surfaces at either 4 or 37 degrees C. The inhibitory action of NTZ and TIZ, but not vancomycin, on biofilm production could be reversed by the addition of zinc salts (2.5 to 40 microM) but not other metals, suggesting that NTZ might target the zinc-dependent accumulation-associated protein (Aap) that mediates accumulation on surfaces. However, neither NTZ nor TIZ formed chelation complexes with zinc salts, based on spectrophotometric and nuclear magnetic resonance analyses, and addition of excess zinc to NTZ-grown bacteria (apo-Aap) did not restore the accumulation phenotype. Our studies suggest that sub-MIC levels of NTZ may affect the assembly or function of cell structures associated with the biofilm phenotype.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Staphylococcus epidermidis/efeitos dos fármacos , Tiazóis/farmacologia , Espectroscopia de Ressonância Magnética , Testes de Sensibilidade Microbiana , Estrutura Molecular , Nitrocompostos , Vancomicina/farmacologia , Zinco/farmacologia
9.
Chem Res Toxicol ; 23(11): 1691-700, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20939553

RESUMO

Nimesulide, a widely used nonsteroidal anti-inflammatory drug (NSAID), has been associated with rare idiosyncratic hepatotoxicity. The chemical mechanisms underlying the liver injury remain unknown. We have undertaken the detailed study of the metabolic pathways of nimesulide in an effort to identify potential reactive metabolites. A previous report from this laboratory has demonstrated that one of the known nimesulide metabolites, termed reduced nimesulide (M1), is further bioactivated by human liver microsomes (HLMs) to form a reactive diiminoquinone species M2. The formation of M2 was confirmed indirectly by trapping with N-acetylcysteine (NAC). The aim of this study was to explore the fate of M1 in an inflammatory environment created by the recruitment of leukocytes. Leukocytes upon activation produce hydrogen peroxide (H(2)O(2)) and other myeloperoxidase (MPO) products, such as hypochlorous acid (HOCl), that are capable of metabolite oxidation. We demonstrate here that the reduced nimesulide, M1, undergoes a facile oxidation with activated neutrophils or with MPO in the presence of H(2)O(2) or HOCl to produce a variety of reactive as well as stable metabolites. One major metabolite, M3, was also produced by HLM as determined by trapping with NAC. Other metabolites, for example, M6, M8, and M9, were unique to the myeloperoxidase, because of their mode of formation from activation of the amino group of reduced nimesulide. The structures of some of these reactive metabolites were proposed on the basis of liquid chromatography-tandem mass spectrometry analyses and established by their comparison with synthetic standards. Metabolite M6 is interesting because it provides clear evidence of amine activation and indicates the potential of the reactive intermediate of M1 to conjugate with protein nucleophiles. In summary, our results demonstrate that a known nimesulide metabolite could be bioactivated by MPO through a pathway distinct from HLM-mediated pathways and that the generation of reactive species by the MPO-mediated bioactivation pathway at the site of inflammation may contribute to the toxicity associated with nimesulide.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Neutrófilos/metabolismo , Peroxidase/metabolismo , Sulfonamidas/metabolismo , Acetilcisteína/química , Anti-Inflamatórios não Esteroides/toxicidade , Cromatografia Líquida , Humanos , Peróxido de Hidrogênio/metabolismo , Ácido Hipocloroso/metabolismo , Microssomos Hepáticos/metabolismo , Neutrófilos/imunologia , Oxirredução , Peroxidase/antagonistas & inibidores , Sulfonamidas/toxicidade , Espectrometria de Massas em Tandem
10.
Bioorg Med Chem Lett ; 20(12): 3537-9, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20488706

RESUMO

Head group analogues of the antibacterial and antiparasitic drug nitazoxanide (NTZ) are presented. A library of 39 analogues was synthesized and assayed for their ability to suppress growth of Helicobacter pylori, Campylobacter jejuni, Clostridium difficile and inhibit NTZ target pyruvate:ferredoxin oxidoreductase (PFOR). Two head groups assayed recapitulated NTZ activity and possessed improved activity over their 2-amino-5-nitrothiazole counterparts, demonstrating that head group modification is a viable route for the synthesis of NTZ-related antibacterial analogues.


Assuntos
Antibacterianos/síntese química , Antiparasitários/síntese química , Tiazóis/síntese química , Antibacterianos/farmacologia , Antiparasitários/farmacologia , Campylobacter jejuni/efeitos dos fármacos , Clostridioides difficile/efeitos dos fármacos , Helicobacter pylori/efeitos dos fármacos , Nitrocompostos , Piruvato Sintase/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/síntese química , Tiazóis/farmacologia
11.
Bioorg Med Chem Lett ; 20(23): 7132-6, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20951039

RESUMO

Autotaxin (ATX) is a secreted soluble enzyme that generates lysophosphatidic acid (LPA) through its lysophospholipase D activity. Because of LPA's role in neoplastic diseases, ATX is an attractive therapeutic target due to its involvement in LPA biosynthesis. Here we describe the SAR of ATX inhibitor, VPC8a202, and apply this SAR knowledge towards developing a high potency inhibitor. We found that electron density in the pyridine region greatly influences activity of our inhibitors at ATX.


Assuntos
Complexos Multienzimáticos/antagonistas & inibidores , Fosfodiesterase I/antagonistas & inibidores , Pirofosfatases/antagonistas & inibidores , Tirosina/análogos & derivados , Antineoplásicos/química , Inibidores Enzimáticos/química , Humanos , Lisofosfolipídeos/biossíntese , Diester Fosfórico Hidrolases/efeitos dos fármacos , Piridinas/química , Relação Estrutura-Atividade
12.
Bioorg Med Chem Lett ; 20(7): 2264-9, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20188554
13.
Biochem J ; 423(3): 315-21, 2009 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-19723024

RESUMO

Dietary ITCs (isothiocyanates) prevent cancer and show other bioactivities in vivo. As electrophiles, ITCs may covalently modify cellular proteins. Using a novel proteomics screen, we identified MIF (macrophage migration inhibitory factor) as the principal target of nutrient ITCs in intact cells. ITCs covalently modify the N-terminal proline residue of MIF and extinguish its catalytic tautomerase activity. MIF deficiency does not prevent induction of Phase 2 gene expression, a hallmark of many cancer chemopreventives, including ITCs. Due to the emerging role of MIF in the control of malignant cell growth and its clear involvement in inflammation, inhibition of MIF by nutrient ITCs suggests therapeutic strategies for inflammatory diseases and cancer.


Assuntos
Oxirredutases Intramoleculares/metabolismo , Isotiocianatos/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Processamento de Proteína Pós-Traducional , Células HeLa , Humanos , Inflamação/genética , Inflamação/metabolismo , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/genética , Neoplasias/genética , Neoplasias/metabolismo
14.
Biochim Biophys Acta ; 1781(9): 508-12, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18638568

RESUMO

A dozen years ago, the term 'S1P' (sphingosine 1-phosphate) was not in the lexicons of scientific literature databases. By early 2008, this query term retrieved well over 1000 citations from PubMed - about 225 of these appeared in 2007. Indeed, S1P is arguably the most heavily studied lipid molecule at present. What happened to distinguish S1P among many other signaling lipids? We believe that the seminal event was the linking of the investigational drug, FTY720 (fingolimod), to S1P signaling. This realization profoundly altered understanding of S1P biology, revealing both that S1P is prominent in lymphocyte trafficking and that mimicking S1P signaling with an agonist drug can modulate the immune system to considerable therapeutic benefit. Neither fact was known prior to FTY720; indeed, this molecule is testament to the power of chemical biology. In this communication, we attempt to summarize progress to date in S1P chemical biology.


Assuntos
Lisofosfolipídeos/química , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Animais , Fenômenos Bioquímicos , Bioquímica , Inibidores Enzimáticos/farmacologia , Cloridrato de Fingolimode , Humanos , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/química , Esfingosina/metabolismo , Esfingosina/farmacologia
15.
J Neurochem ; 110(4): 1191-202, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19493165

RESUMO

Sphingosine-1-phosphate (S1P) is a ubiquitous, lipophilic cellular mediator that acts in part by activation of G-protein-coupled receptor. Modulation of S1P signaling is an emerging pharmacotherapeutic target for immunomodulatory drugs. Although multiple S1P receptor types exist in the CNS, little is known about their function. Here, we report that S1P stimulated G-protein activity in the CNS, and results from [(35)S]GTPgammaS autoradiography using the S1P(1)-selective agonist SEW2871 and the S1P(1/3)-selective antagonist VPC44116 show that in several regions a majority of this activity is mediated by S1P(1) receptors. S1P receptor activation inhibited glutamatergic neurotransmission as determined by electrophysiological recordings in cortical neurons in vitro, and this effect was mimicked by SEW2871 and inhibited by VPC44116. Moreover, central administration of S1P produced in vivo effects resembling the actions of cannabinoids, including thermal antinociception, hypothermia, catalepsy and hypolocomotion, but these actions were independent of CB(1) receptors. At least one of the central effects of S1P, thermal antinociception, is also at least partly S1P(1) receptor mediated because it was produced by SEW2871 and attenuated by VPC44116. These results indicate that CNS S1P receptors are part of a physiologically relevant and widespread neuromodulatory system, and that the S1P(1) receptor contributes to S1P-mediated antinociception.


Assuntos
Córtex Cerebral/metabolismo , Lisofosfolipídeos/metabolismo , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Receptores de Neurotransmissores/metabolismo , Esfingosina/análogos & derivados , Animais , Animais Recém-Nascidos , Ligação Competitiva/fisiologia , Canabinoides/farmacologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Técnicas de Cocultura , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Oxidiazóis/farmacologia , Técnicas de Patch-Clamp , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Neurotransmissores/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Esfingosina/metabolismo , Radioisótopos de Enxofre/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Tiofenos/farmacologia
16.
Mol Carcinog ; 48(9): 801-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19204929

RESUMO

Increased expression of autotaxin in tumors including glioblastoma, breast, renal, ovarian, lung, and thyroid cancers is associated with increased tumor aggressiveness. Autotaxin promotes metastasis as well as cell growth, survival, and migration of cancer cells. These actions could depend on the noncatalytic effects of autotaxin on cell adhesion, or the catalytic activity of autotaxin, which converts lysophosphatidylcholine into lysophosphatidate in the extracellular fluid surrounding the tumor. Both lysophosphatidylcholine (LPC) and lysophosphatidate have been reported to stimulate migration through their respective G-protein coupled receptors. The present study determines the roles of autotaxin, LPC, and lysophosphatidate in controlling the migration of two cancer cell lines: MDA-MB-231 breast cancer cells, which produce little autotaxin and MDA-MB-435 melanoma cells that secrete significant levels of autotaxin. LPC alone was unable to stimulate the migration of either cell type unless autotaxin was present. Knocking down autotaxin secretion, or inhibiting its catalytic activity, blocked cell migration by preventing lysophosphatidate production and the subsequent activation of LPA(1/3) receptors. We conclude that inhibiting autotaxin production or activity could provide a beneficial adjuvant to chemotherapy for preventing tumor growth and metastasis in patients with high autotaxin expression in their tumors.


Assuntos
Movimento Celular/efeitos dos fármacos , Lisofosfatidilcolinas/farmacologia , Complexos Multienzimáticos/metabolismo , Fosfodiesterase I/metabolismo , Pirofosfatases/metabolismo , Anilidas/farmacologia , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Catálise/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Cinética , Lisofosfatidilcolinas/metabolismo , Lisofosfolipídeos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/genética , Organofosfatos/farmacologia , Organofosfonatos/farmacologia , Fosfodiesterase I/antagonistas & inibidores , Fosfodiesterase I/genética , Diester Fosfórico Hidrolases , Piridinas/farmacologia , Pirofosfatases/antagonistas & inibidores , Pirofosfatases/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
17.
Chem Res Toxicol ; 22(1): 72-80, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19053182

RESUMO

Nimesulide is a nonsteroidal anti-inflammatory drug (NSAID) marketed in more than 50 countries. This drug has caused rare and idiosyncratic but severe hepatotoxicity. The mechanisms associated with and factors responsible for this toxicity remain unknown. One of the nimesulide metabolites identified in human urine is 4-amino-2-phenoxy-methanesulfonanilide (M1). In the current study, we demonstrate that M1 is a stable metabolite that is highly susceptible to facile oxidation by cytochrome P450 enzymes (P450s) to form a reactive diiminoquinone intermediate (M2). Direct detection of M2 was difficult by LC-MS. However, its formation was confirmed indirectly by identification of N-acetyl-cysteine (NAC) adducts of M2. The formation of diiminoquinone M2 was P450 mediated with 2C19 and 1A2 as the two principal P450 enzymes catalyzing M1 oxidation. M1 metabolism irreversibly inhibited 2C19 but activated 1A2 in a time-dependent manner. P450 2C19 exclusively mediated further metabolism of M1 to the amino hydroxynimesulide M3 and its diiminoquinone M4. Similar to M2, M4 is also reactive and can be observed indirectly as its NAC adduct. Nucleophilic addition to diiminoquinone M2 occurs with low regioselectivity, yielding three adducts (the peak area ratio 1:0.08:12). The three regioisomers have the same m/z for [M + H](+), presumably due to nucleophilic addition at the three possible electrophilic sites (C-3, -5, and -6 positions of the sulfonaniline ring). The primary adduct, R, was derived from the attack of the nucleophile at the C-5 position of the sulfonaniline ring and was determined by MS/MS and (1)H and (13)C NMR analyses. The structural assignments were confirmed by chemical synthesis of the adduct R. M2 demonstrated its electrophilic reactivity by selectively alkylating human serum albumin (HSA) at the only free thiol, Cys-34. This suggests the possibility that other proteins may undergo a similar conjugation to form irreversible adducts. Under oxidizing conditions in the presence of cumene hydroperoxide (CHP), the formation of M2 was enhanced, indicating that oxidative stress may accelerate the production of reactive diiminoquinone species (M2 and M4).


Assuntos
Anti-Inflamatórios não Esteroides/química , Doença Hepática Induzida por Substâncias e Drogas , Iminas/química , Sulfonamidas/química , Anilidas/toxicidade , Anilidas/urina , Anti-Inflamatórios não Esteroides/toxicidade , Derivados de Benzeno/química , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Cromatografia Líquida , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Iminas/toxicidade , Espectrometria de Massas , Sulfonamidas/toxicidade
18.
Arterioscler Thromb Vasc Biol ; 28(8): 1454-61, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18535287

RESUMO

OBJECTIVE: The role of sphingosine-1-phosphate (S1P) receptors in acute vascular injury and smooth muscle cell (SMC) phenotypic modulation is not completely resolved. METHODS AND RESULTS: S1P receptor antagonists were used to test the hypothesis that specific S1P receptor subtypes differentially regulate SMC phenotypic modulation. In response to acute balloon injury of the rat carotid artery, S1P1/S1P3 receptor mRNA levels were transiently increased at 48 hours whereas S1P2 receptor expression was decreased. S1P2 expression was reinduced and increased at 7 to 10 days postinjury. Daily intraperitoneal injection of the S1P1/S1P3 antagonist VPC44116 decreased neointimal hyperplasia by approximately 50%. In vitro, pharmacological inhibition of S1P1/S1P3 receptors with VPC25239 attenuated S1P-induced proliferation of rat aortic SMCs. Conversely, inhibition of S1P2 with JTE013 potentiated S1P-induced proliferation. Inhibition of S1P1/S1P3 resulted in S1P-induced activation of the SMC differentiation marker genes SMalpha-actin and SMMHC, whereas inhibition of S1P2 attenuated this response. S1P2-dependent activation of SMalpha-actin and SMMHC was shown to be mediated by L-type voltage-gated Ca(2+) channels and subsequent RhoA/Rho kinase-dependent SRF enrichment of CArG box promoter regions. CONCLUSIONS: Results provide evidence that S1P1/S1P3 receptors promote, whereas S1P2 receptors antagonize, SMC proliferation and phenotypic modulation in vitro in response to S1P, or in vivo after vascular injury.


Assuntos
Lisofosfolipídeos/fisiologia , Miócitos de Músculo Liso/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Animais , Masculino , Miócitos de Músculo Liso/classificação , Fenótipo , Ratos , Receptores de Lisoesfingolipídeo/classificação , Esfingosina/fisiologia
19.
Bioorg Med Chem ; 17(16): 6123-36, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19632123

RESUMO

In the search for bioactive sphingosine 1-phosphate (S1P) receptor ligands, a series of 2-amino-2-heterocyclic-propanols were synthesized. These molecules were discovered to be substrates of human-sphingosine kinases 1 and 2 (SPHK1 and SPHK2). When phosphorylated, the resultant phosphates showed varied activities at the five sphingosine-1-phosphate (S1P) receptors (S1P(1-5)). Agonism at S1P(1) was displayed in vivo by induction of lymphopenia. A stereochemical preference of the quaternary carbon was crucial for phosphorylation by the kinases and alters binding affinities at the S1P receptors. Oxazole and oxadiazole compounds are superior kinase substrates to FTY720, the prototypical prodrug immunomodulator, fingolimod (FTY720). The oxazole-derived structure was the most active for human SPHK2. Imidazole analogues were less active substrates for SPHKs, but more potent and selective agonists of the S1P(1) receptor; additionally, the imidazole class of compounds rendered mice lymphopenic.


Assuntos
Imunossupressores/síntese química , Pró-Fármacos/síntese química , Propanóis/síntese química , Propilenoglicóis/química , Receptores de Lisoesfingolipídeo/agonistas , Esfingosina/análogos & derivados , Animais , Cloridrato de Fingolimode , Humanos , Imunossupressores/química , Imunossupressores/farmacologia , Linfopenia/induzido quimicamente , Camundongos , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Propanóis/química , Propilenoglicóis/síntese química , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/síntese química , Esfingosina/química , Esfingosina/farmacologia , Relação Estrutura-Atividade
20.
Circ Res ; 99(7): 731-9, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16960101

RESUMO

Monocyte recruitment and adhesion to vascular endothelium are key early events in atherosclerosis. We examined the role of sphingosine-1-phosphate (S1P) on modulating monocyte/endothelial interactions in the NOD/LtJ (NOD) mouse model of type 1 diabetes. Aortas from nondiabetic and diabetic NOD mice were incubated in the absence or presence of 100 nmol/L S1P. Fluorescently labeled monocytes were incubated with the aortas. Aortas from NOD diabetic mice bound 7-fold more monocytes than nondiabetic littermates (10+/-1 monocytes bound/field for nondiabetic mice vs 74+/-12 monocytes bound/field for diabetic mice, P<0.0001). Incubation of diabetic aortas with 100 nmol/L S1P reduced monocyte adhesion to endothelium by 90%. We found expression of S1P1, S1P2, and S1P3 receptors on NOD aortic endothelial cells. The S1P1 receptor-specific agonist SEW2871 inhibited monocyte adhesion to diabetic aortas. Studies in diabetic S1P3-deficient mice revealed that the S1P3 receptor did not play a pivotal role in this process. S1P reduced endothelial VCAM-1 induction in type 1 diabetic NOD mice, most likely through inhibition of nuclear factor kappaB translocation to the nucleus. Thus, S1P activation of the S1P1 receptor functions in an antiinflammatory manner in type 1 diabetic vascular endothelium to prevent monocyte/endothelial interactions. S1P may play an important role in the prevention of vascular complications of type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Endotélio Vascular/fisiopatologia , Lisofosfolipídeos/metabolismo , Monócitos , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Anti-Inflamatórios/farmacologia , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/fisiopatologia , Transporte Biológico/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Quimiocina CCL2/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-6/metabolismo , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Esfingosina/metabolismo , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Molécula 1 de Adesão de Célula Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA