Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
PLoS Negl Trop Dis ; 14(10): e0008767, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33044962

RESUMO

Schistosomiasis is one of the most devastating neglected tropical parasitic diseases caused by trematodes of the genus Schistosoma. Praziquantel (PZQ) is today the only drug used in humans and animals for the treatment of schistosomiasis but unfortunately it is poorly effective on larval and juvenile stages of the parasite. Therefore, it is urgent the discovery of new drug targets and compounds. We have recently showed that the anti-anginal drug perhexiline maleate (PHX) is very active on multiple developmental stages of Schistosoma mansoni in vitro. It is well known that PHX impacts the lipid metabolism in mammals, but the final target on schistosomes still remains unknown. The aim of this study was to evaluate the ability of 1H nuclear magnetic resonance (NMR) spectroscopy in revealing metabolic perturbations due to PHX treatment of S. mansoni adult male worms. The effects of PHX were compared with the ones induced by vehicle and gambogic acid, in order to detect different metabolic profiles and specificity of the PHX action. Remarkably a list of metabolites associated to PHX-treatment was identified with enrichment in several connected metabolic pathways including also the Kennedy pathway mediating the glycerophospholipid metabolism. Our study represents the first 1H-NMR metabolomic approach to characterize the response of S. mansoni to drug treatment. The obtained "metabolic fingerprint" associated to PHX treatment could represent a strategy for displaying cellular metabolic changes for any given drug and to compare compounds targeting similar or distinct biochemical pathways.


Assuntos
Anti-Helmínticos/administração & dosagem , Monitoramento de Medicamentos/métodos , Espectroscopia de Prótons por Ressonância Magnética/métodos , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/tratamento farmacológico , Adulto , Animais , Feminino , Humanos , Masculino , Metaboloma/efeitos dos fármacos , Camundongos Endogâmicos ICR , Perexilina/administração & dosagem , Perexilina/análogos & derivados , Praziquantel/administração & dosagem , Schistosoma mansoni/crescimento & desenvolvimento , Esquistossomose mansoni/parasitologia
2.
Bioorg Chem ; 102: 104067, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32663671

RESUMO

Schistosomiasis is a neglected tropical disease mainly affecting the poorest tropical and subtropical areas of the world with the impressive number of roughly 200 million infections per year. Schistosomes are blood trematode flukes of the genus Schistosoma causing symptoms in humans and animals. Organ morbidity is caused by the accumulation of parasite eggs and subsequent development of fibrosis. If left untreated, schistosomiasis can result in substantial morbidity and even mortality. Praziquantel (PZQ) is the most effective and widely used compound for the treatment of the disease, in prevention and control programs in the last 30 years. Unfortunately, it has no effect on juvenile immature schistosomes and cannot prevent reinfection or interfere with the schistosome life cycle; moreover drug-resistance represents a serious threat. The search for an alternative or complementary treatment is urgent and drug repurposing could accelerate a solution. The anti-anginal drug perhexiline maleate (PHX) has been previously shown to be effective on larval, juvenile, and adult stages of S. mansoni and to impact egg production in vitro. Since PHX is a racemic mixture of R-(+)- and S-(-)-enantiomers, we designed and realized a stereoselective synthesis of both PHX enantiomers and developed an analytical procedure for the direct quantification of the enantiomeric excess also suitable for semipreparative separation of PHX enantiomers. We next investigated the impact of each enantiomer on viability of newly transformed schistosomula (NTS) and worm pairs of S. mansoni as well as on egg production and vitellarium morphology by in vitro studies. Our results indicate that the R-(+)-PHX is mainly driving the anti-schistosomal activity but that also the S-(-)-PHX possesses a significant activity towards S. mansoni in vitro.


Assuntos
Perexilina/análogos & derivados , Schistosoma mansoni/efeitos dos fármacos , Animais , Larva , Estrutura Molecular , Perexilina/uso terapêutico , Estereoisomerismo , Relação Estrutura-Atividade
3.
J Med Chem ; 60(7): 2780-2789, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28277663

RESUMO

We designed and synthesized perhexiline analogues that have the same therapeutic profile as the parent cardiovascular drug but lacking its metabolic liability associated with CYP2D6 metabolism. Cycloalkyl perhexiline analogues 6a-j were found to be unsuitable for further development, as they retained a pharmacokinetic profile very similar to that shown by the parent compound. Multistep synthesis of perhexiline analogues incorporating fluorine atoms onto the cyclohexyl ring(s) provided a range of different fluoroperhexiline analogues. Of these, analogues 50 (4,4-gem-difluoro) and 62 (4,4,4',4'-tetrafluoro) were highly stable and showed greatly reduced susceptibility to CYP2D6-mediated metabolism. In vitro efficacy studies demonstrated that a number of derivatives retained acceptable potency against CPT-1. Having the best balance of properties, 50 was selected for further evaluation. Like perhexiline, it was shown to be selectively concentrated in the myocardium and, using the Langendorff model, to be effective in improving both cardiac contractility and relaxation when challenged with high fat buffer.


Assuntos
Fármacos Cardiovasculares/química , Fármacos Cardiovasculares/farmacocinética , Perexilina/análogos & derivados , Perexilina/farmacocinética , Animais , Fármacos Cardiovasculares/metabolismo , Fármacos Cardiovasculares/farmacologia , Citocromo P-450 CYP2D6/metabolismo , Halogenação , Coração/efeitos dos fármacos , Coração/fisiologia , Humanos , Masculino , Camundongos Endogâmicos BALB C , Contração Miocárdica/efeitos dos fármacos , Miocárdio/metabolismo , Perexilina/metabolismo , Perexilina/farmacologia
4.
PLoS Negl Trop Dis ; 10(8): e0004928, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27518281

RESUMO

BACKGROUND: Schistosomiasis, one of the world's greatest human neglected tropical diseases, is caused by parasitic trematodes of the genus Schistosoma. A unique feature of schistosome biology is that the induction of sexual maturation as well as the maintenance of the differentiation status of female reproductive organs and egg production, necessary for both disease transmission and pathogenesis, are strictly dependent on the male. The treatment and most control initiatives of schistosomiasis rely today on the long-term application of a single drug, praziquantel (PZQ), mostly by campaigns of mass drug administration. PZQ, while very active on adult parasites, has much lower activity against juvenile worms. Monotherapy also favors the selection of drug resistance and, therefore, new drugs are urgently needed. METHODS AND FINDINGS: Following the screening of a small compound library with an ATP-based luminescent assay on Schistosoma mansoni schistosomula, we here report the identification and characterization of novel antischistosomal properties of the anti-anginal drug perhexiline maleate (PHX). By phenotypic worm survival assays and confocal microscopy studies we show that PHX, in vitro, has a marked lethal effect on all S. mansoni parasite life stages (newly transformed schistosomula, juvenile and adult worms) of the definitive host. We further demonstrate that sub-lethal doses of PHX significantly impair egg production and lipid depletion within the vitellarium of adult female worms. Moreover, we highlighted tegumental damage in adult male worms and remarkable reproductive system alterations in both female and male adult parasites. The in vivo study in S. mansoni-patent mice showed a notable variability of worm burdens in the individual experiments, with an overall minimal schistosomicidal effect upon PHX treatment. The short PHX half-life in mice, together with its very high rodent plasma proteins binding could be the cause of the modest efficacy of PHX in the schistosomiasis murine model. CONCLUSIONS/SIGNIFICANCE: Overall, our data indicate that PHX could represent a promising starting point for novel schistosomicidal drug discovery programmes.


Assuntos
Genitália/efeitos dos fármacos , Perexilina/análogos & derivados , Schistosoma mansoni/efeitos dos fármacos , Schistosoma mansoni/ultraestrutura , Esquistossomose mansoni/tratamento farmacológico , Esquistossomicidas/farmacologia , Animais , Modelos Animais de Doenças , Resistência a Medicamentos , Feminino , Meia-Vida , Humanos , Estágios do Ciclo de Vida/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Perexilina/farmacologia , Praziquantel/farmacologia
5.
Cardiovasc Drugs Ther ; 30(4): 399-405, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27106834

RESUMO

Approaches to the pharmacotherapy of angina pectoris have previously centred on the concept that a transient imbalance between myocardial oxygen "demand" and supply within the myocardium can best be addressed by reducing demand (for example, with ß-adrenoceptor antagonist) or by increasing availability of blood (via coronary vasomotor reactivity adjustment or coronary revascularization). However, this principle is potentially challenged by the emergence of cases of angina unsuitable for such therapies (for example because of concomitant severe systolic heart failure) and by the recognition that impaired myocardial energetics may precipitate angina in the absence of fixed or variable coronary obstruction (for example in hypertrophic cardiomyopathy). The past 20 years have seen the re-emergence of a class of anti-anginal agents which act primarily by improving efficiency of myocardial oxygen utilization, and thus can correct impaired energetics, simultaneously treating angina and heart failure symptoms. We review the principles underlying the safe use of such agents, beginning with the prototype drug perhexiline maleate, which despite complex pharmacokinetics and potential hepato- or neuro-toxicity has emerged as an attractive management option in many "complicated" cases of angina pectoris.


Assuntos
Angina Pectoris/tratamento farmacológico , Fármacos Cardiovasculares/uso terapêutico , Miocárdio/metabolismo , Perexilina/análogos & derivados , Angina Pectoris/metabolismo , Animais , Fármacos Cardiovasculares/farmacologia , Ácidos Graxos/metabolismo , Glucose/metabolismo , Humanos , Mitocôndrias/metabolismo , Perexilina/farmacologia , Perexilina/uso terapêutico
6.
Sci Rep ; 5: 18144, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26674674

RESUMO

High Risk Neuroblastoma (HR-NB) is a pediatric cancer characterized by high malignancy and remarkable cell heterogeneity within the tumour nodules. In a recent study, we demonstrated that in vitro and in vivo over-expression of the non-coding RNA NDM29 (neuroblastoma differentiation marker 29) induces NB cell differentiation, dramatically reducing their malignancy. Among gene expression changes, differentiated phenotype induced by NDM29 is characterized by decrease of the expression of ABC transporters responsible for anticancer drug resistance. Thus, the pharmacological induction of NDM29, in principle, might represent a possible novel strategy to increase cytotoxic drug responses. In this work, we identify a small molecule able to induce the expression of NDM29 in NB cells, conferring to malignant cells increased susceptibility to cisplatin cytotoxic effects. We demonstrate that the pharmacological induction of NDM29 expression in vivo enhances the antitumoral effects of chemotherapy specifically on tumour initiating/cancer stem cells sub-population, usually refractory to therapies and responsible for tumour relapse. In summary, we suggest a novel therapeutical approach possibly useful to treat very aggressive NB cases with poor prognosis. This novel pharmacological strategy aims to promote differentiation of "stem-like" cells to render them more susceptible to the killing action of cytotoxic anticancer drugs.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , RNA não Traduzido/genética , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Sinergismo Farmacológico , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Neuroblastoma/patologia , Perexilina/administração & dosagem , Perexilina/análogos & derivados , Perexilina/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Ther Drug Monit ; 37(6): 821-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25774703

RESUMO

BACKGROUND: The polymorphic nature of cytochrome P450 2D6 has made therapeutic drug monitoring of the anti-anginal agent perhexiline a compulsory step in reducing adverse events associated with plasma concentrations above the therapeutic range (0.15-0.60 mg/L). The aim of this study was to develop a high-performance liquid chromatography-mass spectrometry/mass spectrometry method for the determination of plasma perhexiline concentrations and its major metabolite cis-hydroxy-perhexiline to reduce sample extraction procedures and improve sample turnaround times. METHODS: The method was validated by determining the precision and accuracy of calibrators and quality control material, comparing quality assurance program samples and patient samples measured by a previously reported liquid-liquid extraction fluorescence (FL) detection high-performance liquid chromatography method and performing matrix effects investigations. RESULTS: Replicates of calibrators at concentrations of 3.00 and 0.05 mg/L demonstrated imprecision of <10.8% and inaccuracy of <8.2% for perhexiline and <10.1% and <4.5% for cis-hydroxy-perhexiline, respectively. All samples measured by the 2 methods (n = 102) demonstrated Deming regression of perhexiline = 1.20 FL + 0.00 (Sy.x = 0.08, 1/slope = 0.67); cis-hydroxy-perhexiline = 1.48 FL - 0.20 (Sy.x = 0.40, 1/slope = 0.67). CONCLUSIONS: The assay performance was deemed acceptable and integrated into the routine therapeutic drug monitoring program of the department.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Monitoramento de Medicamentos/métodos , Perexilina/análogos & derivados , Espectrometria de Massas em Tandem/métodos , Calibragem , Citocromo P-450 CYP2D6/genética , Humanos , Extração Líquido-Líquido , Perexilina/sangue , Polimorfismo Genético , Reprodutibilidade dos Testes
8.
J Antimicrob Chemother ; 69(4): 1035-44, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24284780

RESUMO

OBJECTIVES: Biofilms of Candida species, often formed on medical devices, are generally resistant to currently available antifungal drugs. The aim of this study was to identify compounds that increase the activity of amphotericin B and caspofungin, commonly used antifungal agents, against Candida biofilms. METHODS: A library containing off-patent drugs was screened for compounds, termed enhancers, that increase the in vitro activity of amphotericin B against Candida albicans biofilms. Biofilms were grown in 96-well plates and growth was determined by the cell titre blue assay. Synergy between identified enhancers and antifungal agents was further characterized in vitro using fractional inhibitory concentration index (FICI) values and in vivo using a worm biofilm infection model. In light of the application of these enhancers onto implants, their possible effect on the growth potential of MG63 osteoblast-like cells was assessed. RESULTS: Pre-incubation of C. albicans biofilms with subinhibitory concentrations of the enhancers drospirenone, perhexiline maleate or toremifene citrate significantly increased the activity of amphotericin B or caspofungin (FICI  < 0.5) against C. albicans and Candida glabrata biofilms. Moreover, these enhancers did not affect the growth potential of osteoblasts. Interestingly, toremifene citrate also enhanced the in vitro activity of caspofungin in a mixed biofilm consisting of C. albicans and Staphylococcus epidermidis. Furthermore, we demonstrate synergy between toremifene citrate and caspofungin in an in vivo worm C. albicans biofilm infection model. CONCLUSIONS: Our data demonstrate an in vitro and in vivo enhancement of the antibiofilm activity of caspofungin by toremifene citrate. Furthermore, our results pave the way for implant-related applications of the identified enhancers.


Assuntos
Anfotericina B/farmacologia , Antifúngicos/farmacologia , Biofilmes/efeitos dos fármacos , Candida albicans/efeitos dos fármacos , Reposicionamento de Medicamentos , Sinergismo Farmacológico , Equinocandinas/farmacologia , Androstenos/farmacologia , Animais , Caenorhabditis elegans/microbiologia , Candida albicans/fisiologia , Caspofungina , Lipopeptídeos , Testes de Sensibilidade Microbiana , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Perexilina/análogos & derivados , Perexilina/farmacologia , Toremifeno/farmacologia
9.
Orphanet J Rare Dis ; 8: 163, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24131551

RESUMO

BACKGROUND: Currently, there are no effective medical treatment options to prevent the formation of heterotopic bones in fibrodysplasia ossificans progressiva (FOP). By the drug repositioning strategy, we confirmed that perhexiline maleate (Pex) potentially ameliorates heterotopic ossification in model cells and mice. Here, we conducted a prospective study to assess the efficacy and safety of Pex in the treatment of FOP patients. METHODS: FOP patients in this open-label single-center study were treated with Pex for a total of 12 months, and followed up for 12 consecutive months after medication discontinuation. The safety of the treatment was assessed regularly by physical and blood examinations. The efficacy of Pex for preventing heterotopic ossifications was evaluated by the presence of flare-ups, measurements of serum bone markers, and changes in the total bone volume calculated by the three-dimensional computed tomography (3D-CT) images. RESULTS: Five patients with an average age of 23.4 years were enrolled. Within safe doses of Pex administration in each individual, there were no drug-induced adverse effects during the medication phase. Three patients showed no intense inflammatory reactions during the study period, while two patients had acute flare-ups around the hip joint without evidence of trauma during the medication phase. In addition, one of them became progressively incapable of opening her mouth over the discontinuation phase. Serum levels of alkaline phosphatase (ALP) and bone specific ALP (BAP) were significantly and synchronously increased with the occurrence of flare-ups. Volumetric 3D-CT analysis demonstrated a significant increase in the total bone volume of Case 2 (378 cm(3)) and Case 3 (833 cm(3)) during the two-year study period. CONCLUSIONS: We could not prove the efficacy of oral Pex administration in the prevention of heterotopic ossifications in FOP. Serum levels of ALP and BAP appear to be promising biomarkers for monitoring the development of ectopic ossifications and efficacy of the therapy. Quantification of change in the total bone volume by whole body CT scanning could be a reliable evaluation tool for disease progression in forthcoming clinical trials of FOP.


Assuntos
Miosite Ossificante/tratamento farmacológico , Perexilina/análogos & derivados , Adolescente , Adulto , Fosfatase Alcalina/sangue , Feminino , Humanos , Masculino , Miosite Ossificante/sangue , Miosite Ossificante/diagnóstico por imagem , Osteocalcina/sangue , Perexilina/uso terapêutico , Radiografia , Adulto Jovem
10.
J Bone Miner Metab ; 31(1): 26-33, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23011467

RESUMO

Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant disorder characterized by progressive heterotopic ossification. FOP is caused by a gain-of-function mutation in ACVR1 encoding the bone morphogenetic protein type II receptor, ACVR1/ALK2. The mutant receptor causes upregulation of a transcriptional factor, Id1. No therapy is available to prevent the progressive heterotopic ossification in FOP. In an effort to search for clinically applicable drugs for FOP, we screened 1,040 FDA-approved drugs for suppression of the Id1 promoter activated by the mutant ACVR1/ALK2 in C2C12 cells. We found that that two antianginal agents, fendiline hydrochloride and perhexiline maleate, suppressed the Id1 promoter in a dose-dependent manner. The drugs also suppressed the expression of native Id1 mRNA and alkaline phosphatase in a dose-dependent manner. Perhexiline but not fendiline downregulated phosphorylation of Smad 1/5/8 driven by bone morphogenetic protein (BMP)-2. We implanted crude BMPs in muscles of ddY mice and fed them fendiline or perhexiline for 30 days. Mice taking perhexiline showed a 38.0 % reduction in the volume of heterotopic ossification compared to controls, whereas mice taking fendiline showed a slight reduction of heterotopic ossification. Fendiline, perhexiline, and their possible derivatives are potentially applicable to clinical practice to prevent devastating heterotopic ossification in FOP.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Fendilina/farmacologia , Células Musculares/metabolismo , Miosite Ossificante/tratamento farmacológico , Ossificação Heterotópica/tratamento farmacológico , Osteoblastos/metabolismo , Perexilina/análogos & derivados , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/metabolismo , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Proteína 1 Inibidora de Diferenciação/biossíntese , Proteína 1 Inibidora de Diferenciação/genética , Camundongos , Camundongos Mutantes , Células Musculares/patologia , Mutação , Miosite Ossificante/genética , Miosite Ossificante/metabolismo , Miosite Ossificante/patologia , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/patologia , Osteoblastos/patologia , Perexilina/farmacologia , Regiões Promotoras Genéticas/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo
11.
Ther Drug Monit ; 34(2): 227-31, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22322401

RESUMO

BACKGROUND: Concomitant treatment with amiodarone and perhsexiline has been considered to be relatively contraindicated because of the hypothetical risk of potentiated adverse effects mediated by additive inhibition of carnitine palmitoyl transferase 1. AIM: To study the prevalence of adverse effects associated with the concomitant use of perhexiline and amiodarone. METHODS: A retrospective analysis of a single hospital database of patients receiving perhexiline and amiodarone between July 2009 and April 2011. Files were reviewed for short- and long-term adverse effects requiring drug cessation. Glucose concentration, gamma glutamyl transferase activity. and perhexiline blood concentrations were recorded. RESULTS: We identified 26 patients concomitantly treated with perhexiline and amiodarone, 20 on a long-term basis. In 6 cases, amiodarone was introduced on top of preceding perhexiline. In none of the cases were drugs stopped because of adverse effects. Although blood glucose concentrations fell significantly 48 hours postadmission to hospital, this seems to reflect the resolution of "admission hyperglycemia" rather than onset of hypoglycemia; the latter was rare (5 patients), mild, and clinically silent. In 4 patients, gamma glutamyl transferase approximately doubled. CONCLUSIONS: Traditionally, concomitant treatment with amiodarone and perhexiline has been considered to be relatively contraindicated on the basis of the theoretical potential for synergistic toxicity. This cohort of 26 patients received this concomitant treatment without any excess of major adverse reactions. Our findings suggest that concomitant treatment with perhexiline and amiodarone may be safe in the setting of (1) previous tolerance of either agent, and (2) titration of plasma perhexiline concentrations to guide therapy.


Assuntos
Amiodarona/efeitos adversos , Perexilina/análogos & derivados , Vasodilatadores/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Amiodarona/administração & dosagem , Amiodarona/uso terapêutico , Glicemia/efeitos dos fármacos , Carnitina O-Palmitoiltransferase/antagonistas & inibidores , Bases de Dados Factuais , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Perexilina/administração & dosagem , Perexilina/efeitos adversos , Perexilina/uso terapêutico , Prevalência , Estudos Retrospectivos , Vasodilatadores/administração & dosagem , Vasodilatadores/uso terapêutico , gama-Glutamiltransferase/efeitos dos fármacos , gama-Glutamiltransferase/metabolismo
13.
Cell Physiol Biochem ; 24(3-4): 167-76, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19710531

RESUMO

Peptidoglycans (PGN) from bacterial cell walls may modify the course of an infection with bacterial pathogens. The present study explored the effect of PGN on cytosolic Ca2+ activity, cytokine production and phagocytosis of mouse dendritic cells (DCs), essential cells in the initiation and direction of antigen-specific T cell responses. Exposure of DCs to PGN was followed by a rapid increase in cytosolic Ca2+ activity ([Ca2+]i), which was due to Ca2+ release from intracellular stores and influx of extracellular Ca2+ across the cell membrane. In DCs isolated from Toll-like receptor 2 (TLR2) deficient mice the effect of PGN on [Ca2+]i was dramatically impaired. The PGN-induced increase of [Ca2+]i was dependent on voltage-gated K+ (Kv) channel activity. PGN-induced increase of [Ca2+]i was significantly blunted by margatoxin (MgTx) and perhexiline maleate (PM), inhibitors of Kv1.3 and Kv1.5, respectively. PGN further stimulated the release of tumour necrosis factor alpha (TNFalpha), interleukin-12 (IL-12) and interleukin-10 (IL-10), an effect significantly blunted by PM and the specific blocker of store-operated Ca2+ channels SKF-96365. Moreover, phagocytic capacity was dramatically increased in PGN-stimulated DCs in the presence of either Kv channel inhibitors or SKF-96365. The observations disclose Ca2+ and Kv channel-dependent cytokine production and phagocytosis in PGN-stimulated DCs.


Assuntos
Cálcio/metabolismo , Células Dendríticas/efeitos dos fármacos , Peptidoglicano/farmacologia , Staphylococcus aureus/química , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Citocinas/análise , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Eletrofisiologia , Feminino , Fêmur/citologia , Corantes Fluorescentes/metabolismo , Fura-2/metabolismo , Imidazóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Neurotoxinas/farmacologia , Técnicas de Patch-Clamp , Perexilina/análogos & derivados , Perexilina/farmacologia , Fagocitose/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Venenos de Escorpião/farmacologia , Tíbia/citologia , Receptor 2 Toll-Like/imunologia
14.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(27): 3025-30, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19646935

RESUMO

Perhexiline is a drug that is used for treatment of moderate to severe angina pectoris that has not responded to other treatment. It has a low therapeutic index, and saturable metabolism that is also subject to genetic polymorphism (CYP2D6). Concentration monitoring of the parent drug and its major metabolite is considered necessary to optimise efficacy and reduce the risk of hepatotoxicity and neuropathy. A rapid, simple and sensitive liquid chromatography/tandem mass spectrometry (LC-MS/MS) assay was developed for the determination of perhexiline and its metabolite cis-hydroxyperhexiline in human plasma. After proteins were precipitated with acetonitrile, perhexiline, the major metabolite cis-hydroxyperhexiline and nordoxepin as the internal standard were resolved on a phenyl-hexyl column using gradient elution of 0.05% formic acid and methanol. The three compounds were detected using electrospray ionisation in the positive mode. Standard curves were linear over the concentration range 10-2000microg/L (r>0.999), bias was

Assuntos
Cromatografia Líquida/métodos , Perexilina/análogos & derivados , Perexilina/sangue , Espectrometria de Massas em Tandem/métodos , Doxepina/análogos & derivados , Doxepina/química , Humanos , Modelos Lineares , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
15.
Toxicol In Vitro ; 22(6): 1511-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18603402

RESUMO

Mallory-Denk bodies (MDB) are hepatocyte inclusions containing cytokeratin 8 (CK8) which can develop, along with other steatohepatitis lesions, in patients treated with amiodarone, perhexiline maleate or 4,4'-diethylaminoethoxyhexestrol. These drugs accumulate lipids, whose subsequent peroxidation liberates reactive by-products, like malondialdehyde (MDA). The formation of MDB has been previously reproduced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine or griseofulvin administration which cross-link CK8 by tissue transglutaminase, thus forming an entangled network, from which MDB progressively arise. The present study depicts the mechanisms initiating MDB formation by steatohepatitis-inducing drugs. Short incubation of hepatocytes with amiodarone (50 microM), 4,4'-diethylaminoethoxyhexestrol (50 microM) or perhexiline maleate (25 microM) increased the pool of CK8 monomers and increased cell calcium to activate Ca(++)-dependent transglutaminases which cross-linked the CK8 monomers into CK8-containing oligomers. The present study also provides the first evidence that MDA might directly participate in MDB formation, as this reactive agent cross-linked purified CK8 or albumin in vitro, disrupted the cytokeratin network of isolated hepatocytes, and bridged CK8 molecules. In conclusion, steatohepatitis-inducing drugs increase cell calcium and activate tissue transglutaminase, which cross-links CK8 to form a molecular scaffold, from which MDB might secondarily arise. Malondialdehyde also cross-links CK8, albeit through a different mechanism, and might also contribute to MDB formation.


Assuntos
Hepatócitos/efeitos dos fármacos , Corpos de Inclusão/efeitos dos fármacos , Queratina-8/efeitos dos fármacos , Malondialdeído/metabolismo , Amiodarona/toxicidade , Animais , Cálcio/metabolismo , Fígado Gorduroso/induzido quimicamente , Proteínas de Ligação ao GTP/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Hepatócitos/metabolismo , Hexestrol/análogos & derivados , Hexestrol/toxicidade , Corpos de Inclusão/metabolismo , Queratina-8/metabolismo , Masculino , Perexilina/análogos & derivados , Perexilina/toxicidade , Proteína 2 Glutamina gama-Glutamiltransferase , Proteínas , Ratos , Ratos Sprague-Dawley , Transglutaminases/efeitos dos fármacos , Transglutaminases/metabolismo
16.
Pharmacogenet Genomics ; 17(5): 305-12, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17429312

RESUMO

AIMS: This study investigated the effects of increasing doses of rac-perhexiline maleate and CYP2D6 phenotype and genotype on the pharmacokinetics of (+) and (-)-perhexiline. METHODS: In a prospective study, steady-state plasma concentrations of (+) and (-)-perhexiline were quantified in 10 CYP2D6 genotyped patients following dosing with 100 mg/day rac-perhexiline maleate, and following a subsequent dosage increase to 150 or 200 mg/day. In a retrospective study, steady-state plasma concentrations of (+) and (-)-perhexiline were obtained from 111 CYP2D6 phenotyped patients receiving rac-perhexiline maleate. RESULTS: In the prospective study, comprising one poor and nine extensive/intermediate metabolizers, the apparent oral clearance (CL/F) of both enantiomers increased with the number of functional CYP2D6 genes. In the nine extensive/intermediate metabolizers receiving the 100 mg/day dose, the median CL/F of (+)-perhexiline was lower than that of (-)-perhexiline (352.5 versus 440.6 l/day, P<0.01). Following the dosage increase, the median CL/F of both enantiomers decreased by 45.4 and 41.4%, respectively. In the retrospective study, the median (+)-/(-)-perhexiline plasma concentration ratio was lower (P<0.0001) in phenotypic extensive/intermediate (1.41) versus poor metabolizers (2.29). Median CL/F of (+) and (-)-perhexiline was 10.6 and 24.2 l/day (P<0.05), respectively, in poor metabolizers, and 184.1 and 272.0 l/day (P<0.001), respectively, in extensive/intermediate metabolizers. CONCLUSIONS: Perhexiline's pharmacokinetics exhibit significant enantioselectivity in CYP2D6 extensive/intermediate and poor metabolizers, with both enantiomers displaying polymorphic and saturable metabolism via CYP2D6. Clinical use of rac-perhexiline may be improved by developing specific enantiomer target plasma concentration ranges.


Assuntos
Fármacos Cardiovasculares/farmacocinética , Citocromo P-450 CYP2D6/genética , Isquemia Miocárdica/metabolismo , Perexilina/análogos & derivados , Disponibilidade Biológica , Fármacos Cardiovasculares/química , Genótipo , Humanos , Taxa de Depuração Metabólica , Isquemia Miocárdica/genética , Perexilina/química , Perexilina/farmacocinética , Fenótipo , Polimorfismo Genético , Estudos Prospectivos , Estudos Retrospectivos , Sensibilidade e Especificidade , Estereoisomerismo
17.
Artigo em Inglês | MEDLINE | ID: mdl-16837252

RESUMO

The use of perhexiline (PHX) is limited by hepatic and neurological toxicity associated with elevated concentrations in plasma that are the result of polymorphism of the cytochrome P450 2D6 isoform (CYP2D6). PHX is cleared by hepatic oxidation that produces three 4-monohydroxy metabolites: cis-OH-PHX, trans1-OH-PHX and trans2-OH-PHX. The current study describes an HPLC-fluorescent method utilising pre-column derivatization with dansyl chloride. Following derivatization, the metabolites were resolved on a C18 column with a gradient elution using a mobile phase composed of methanol and water. The method described is suitable for the quantification of the metabolites in human plasma and urine following clinical doses and for kinetic studies using human liver microsomes. The method demonstrates sufficient sensitivity, accuracy and precision between 5.0 and 0.01, 50.0 and 0.2 and 1.0 and 0.005 mg/l in human plasma, urine and liver microsomes, respectively, with intra-assay coefficients of variation and bias <15%, except at the lowest limit of quantification (<20%). The inter-assay coefficients of variation and bias were <15%. The application of this method to plasma and urine samples of five CYP2D6 extensive metaboliser (EM) patients at steady state with respect to PHX dosing determined that the mean (+/-S.D.) renal clearances of trans1-OH-PHX and cis-OH-PHX were 1.58+/-0.35 and 0.16+/-0.06l/h, respectively. The mean (+/-S.D.) dose recovered in urine as free and glucuronidated 4-monohydroxy PHX metabolites was 20.6+/-11.6%.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Microssomos Hepáticos/química , Perexilina/análogos & derivados , Perexilina/metabolismo , Humanos
18.
Eur J Cardiovasc Nurs ; 5(2): 175-84, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16469541

RESUMO

The ever-increasing burden of ischaemic heart disease and its common manifestation chronic angina pectoris calls for the exploration of other treatment options for those patients who despite the maximum conventional pharmacological and surgical interventions continue to suffer. Such exploration has led to the increasing use of new metabolically acting antianginal agents and the re-emergence of an old and somewhat forgotten pharmacological agent, perhexiline maleate. This review aims to update the cardiac nurse with knowledge to manage the care a patient receiving perhexiline maleate treatment and provide a brief review of three new metabolic agents: trimetazidine, ranolazine and etomoxir.


Assuntos
Angina Pectoris/tratamento farmacológico , Angina Pectoris/metabolismo , Perexilina/análogos & derivados , Acetanilidas , Angina Pectoris/enfermagem , Bloqueadores dos Canais de Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/uso terapêutico , Fármacos Cardiovasculares/metabolismo , Fármacos Cardiovasculares/uso terapêutico , Carnitina O-Palmitoiltransferase/antagonistas & inibidores , Monitoramento de Medicamentos , Compostos de Epóxi/uso terapêutico , Ácidos Graxos/metabolismo , Meia-Vida , Humanos , Taxa de Depuração Metabólica , Papel do Profissional de Enfermagem , Avaliação em Enfermagem , Educação de Pacientes como Assunto , Seleção de Pacientes , Perexilina/metabolismo , Perexilina/farmacologia , Perexilina/uso terapêutico , Piperazinas/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Ranolazina , Resultado do Tratamento , Trimetazidina/uso terapêutico , Vasodilatadores/metabolismo , Vasodilatadores/uso terapêutico
19.
Artigo em Inglês | MEDLINE | ID: mdl-15113543

RESUMO

A method for the quantitative determination of perhexiline and its main hydroxylated metabolites in human plasma, based on liquid chromatography-mass spectrometry (LC-MS), was developed. The method used protein precipitation with acetonitrile followed by dilution with water and subsequent direct injection of the extract into the LC-MS system. Hexadiline was used as internal standard and the intra-assay coefficients of variation were

Assuntos
Fármacos Cardiovasculares/sangue , Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas/métodos , Perexilina/análogos & derivados , Perexilina/sangue , Humanos , Isomerismo , Padrões de Referência
20.
J Interferon Cytokine Res ; 22(3): 343-50, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12034042

RESUMO

The novel colony-stimulating factor (CSF) inducer leustroducsin B (LSN-B), which was isolated from Streptomyces platensis, has been shown to have potent cytokine-inducing activities in clonal human bone marrow-derived stromal cell line KM-102 and in primary human bone marrow-derived stromal cells. In this study, we investigated the signal transduction pathway of LSN-B using luciferase expression plasmids linked to the 5'-flanking region of interleukin-8 (IL-8) and that of the IL-11 gene. In KM-102 cells, LSN-B induced luciferase activity both in the wild-type and in the activated protein 1 (AP-1) site point-mutated IL-8 promoter. The mutation in the nuclear factor-kappaB (NF-kappaB) site abrogated LSN-B-stimulated induction of the reporter gene. LSN-B-inducing activity was inhibited by (1) N-acetyl-L-cysteine, a well-characterized antioxidant, (2) cationic amphiphilic drugs, inhibitors of acidic sphingomyelinase (A-SMase), and (3) D609, an inhibitor of phosphatidylcholine-specific phospholipase C (PC-PLC). These observations suggest that LSN-B potentiates the A-SMase-mediated signaling pathway to stimulate NF-kappaB. In contrast, LSN-B did not induce IL-11 promoter-driven luciferase activity. The observed increase in IL-11 mRNA stability by LSN-B indicates that the inducible production of IL-11 by LSN-B is regulated at the posttranscriptional level. In addition, inhibition of LSN-B-mediated induction of IL-11 production by cationic amphiphilic drugs and D609 in KM-102 cells demonstrates that increased IL-11 mRNA stability by LSN-B might be mediated via NF-kappaB activation. From these results, we suggest that LSN-B induces cytokine production through at least two separate mechanisms, at the transcriptional level and at the posttranscriptional level via NF-kappaB activation.


Assuntos
Células da Medula Óssea/metabolismo , Lactonas/farmacologia , NF-kappa B/metabolismo , Compostos Organofosforados/farmacologia , Perexilina/análogos & derivados , Esfingomielina Fosfodiesterase/metabolismo , Células Estromais/metabolismo , Acetilcisteína/farmacologia , Antioxidantes/farmacologia , Células da Medula Óssea/citologia , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Linhagem Celular , Células Clonais , Desipramina/farmacologia , Genes Reporter , Humanos , Interleucina-11/análise , Interleucina-11/biossíntese , Interleucina-11/genética , Interleucina-8/análise , Interleucina-8/genética , Mutagênese Sítio-Dirigida , NF-kappa B/genética , Norbornanos , Perexilina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Mutação Puntual , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Pironas , Transdução de Sinais , Células Estromais/citologia , Tiocarbamatos , Tionas/farmacologia , Ativação Transcricional/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA