Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
Exp Neurol ; 372: 114612, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37993080

RESUMO

Edema formation is one of the very first events to occur after spinal cord injury (SCI) leading to an increase of the intrathecal pressure and consequently to serious spinal tissue and functional impairments. Current edema treatments are still symptomatic and/or non-specific. Since edema formation mechanisms are mainly described as vasogenic and cytotoxic, it becomes crucial to understand the interplay between these two subtypes. Acting on key targets to inhibit edema formation may reduce secondary damage and related functional impairments. In this study, we characterize the edema kinetic after T9-10 spinal contusion. We use trifluoperazine (TFP) to block the expression and the functional subcellular localization of aquaporin-4 supposed to be implicated in the cytotoxic edema formation. We also use sodium cromoglycate (SCG) to deactivate mast cell degranulation known to be implicated in the vasogenic edema formation. Our results show a significant reduction of edema after TFP treatment and after TFP-SCG combined treatment compared to control. This reduction is correlated with limited onset of initial sensorimotor impairments particularly after combined treatment. Our results highlight the importance of potential synergetic targets in early edema therapy after SCI as part of tissue sparing strategies.


Assuntos
Traumatismos da Medula Espinal , Medula Espinal , Ratos , Animais , Medula Espinal/metabolismo , Cromolina Sódica/farmacologia , Cromolina Sódica/uso terapêutico , Cromolina Sódica/metabolismo , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico , Trifluoperazina/metabolismo , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo , Edema/tratamento farmacológico , Edema/etiologia
2.
Int J Med Sci ; 20(6): 797-809, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37213674

RESUMO

Currently, no specific and standard treatment for traumatic brain injury (TBI) has been developed. Therefore, studies on new therapeutic drugs for TBI treatment are urgently needed. Trifluoperazine (TFP) is a therapeutic agent for the treatment of psychiatric disorders that reduces edema of the central nervous system. However, the specific working mechanism of TFP is not fully understood in TBI. In this study, the immunofluorescence co-localization analysis revealed that the area and intensity covered by Aquaporin4 (AQP4) on the surface of brain cells (astrocyte endfeet) increased significantly after TBI. In contrast, TFP treatment reversed these phenomena. This finding showed that TFP inhibited AQP4 accumulation on the surface of brain cells (astrocyte endfeet). The tunel fluorescence intensity and fluorescence area were lower in the TBI+TFP group compared to the TBI group. Additionally, the brain edema, brain defect area, and modified neurological severity score (mNSS) were lower in the TBI+TFP. The RNA-seq was performed on the cortical tissues of rats in the Sham, TBI, and TBI+TFP groups. A total of 3774 genes differently expressed between the TBI and the Sham group were identified. Of these, 2940 genes were up-regulated and 834 genes were down-regulated. A total of 1845 differently expressed genes between the TBI+TFP and TBI group were also identified, in which 621 genes were up-regulated and 1224 genes were down-regulated. Analysis of the common differential genes in the three groups showed that TFP could reverse the expression of apoptosis and inflammation genes. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that the differentially expressed genes (DEGs) were highly enriched in the signaling pathways regulating inflammation. In conclusion, TFP alleviates brain edema after TBI by preventing the accumulation of AQP4 on the surface of brain cells. Generally, TFP alleviates apoptosis and inflammatory response induced by TBI, and promotes the recovery of nerve function in rats after TBI. Thus, TFP is a potential therapeutic agent for TBI treatment.


Assuntos
Edema Encefálico , Lesões Encefálicas Traumáticas , Animais , Ratos , Apoptose/genética , Aquaporina 4/antagonistas & inibidores , Aquaporina 4/genética , Aquaporina 4/metabolismo , Encéfalo , Edema Encefálico/etiologia , Edema Encefálico/genética , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/genética , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/metabolismo , Trifluoperazina/farmacologia , Trifluoperazina/uso terapêutico , Trifluoperazina/metabolismo
3.
PLoS Comput Biol ; 18(10): e1010583, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36206305

RESUMO

Calmodulin (CaM) is a calcium sensor which binds and regulates a wide range of target-proteins. This implicitly enables the concentration of calcium to influence many downstream physiological responses, including muscle contraction, learning and depression. The antipsychotic drug trifluoperazine (TFP) is a known CaM inhibitor. By binding to various sites, TFP prevents CaM from associating to target-proteins. However, the molecular and state-dependent mechanisms behind CaM inhibition by drugs such as TFP are largely unknown. Here, we build a Markov state model (MSM) from adaptively sampled molecular dynamics simulations and reveal the structural and dynamical features behind the inhibitory mechanism of TFP-binding to the C-terminal domain of CaM. We specifically identify three major TFP binding-modes from the MSM macrostates, and distinguish their effect on CaM conformation by using a systematic analysis protocol based on biophysical descriptors and tools from machine learning. The results show that depending on the binding orientation, TFP effectively stabilizes features of the calcium-unbound CaM, either affecting the CaM hydrophobic binding pocket, the calcium binding sites or the secondary structure content in the bound domain. The conclusions drawn from this work may in the future serve to formulate a complete model of pharmacological modulation of CaM, which furthers our understanding of how these drugs affect signaling pathways as well as associated diseases.


Assuntos
Antipsicóticos , Calmodulina , Calmodulina/metabolismo , Trifluoperazina/farmacologia , Trifluoperazina/química , Trifluoperazina/metabolismo , Antipsicóticos/química , Cálcio/metabolismo , Ligação Proteica , Sítios de Ligação
4.
J Appl Microbiol ; 133(4): 2631-2641, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35870147

RESUMO

AIMS: Calmodulin (CaM), acts as a kind of multifunctional Ca2+ sensing protein, which is ubiquitous in fungi, is highly conserved across eukaryotes and is involved in the regulation of a range of physiological processes, including morphogenesis, reproduction and secondary metabolites biosynthesis. Our aim was to understand the characteristics and functions of AaCaM in Alternaria alternata, the causal agent of pear black spot. METHODS AND RESULTS: A 450 bp cDNA sequence of AaCaM gene of A. alternata was cloned by the PCR homology method. Sequence analysis showed that this protein encoded by AaCaM was a stable hydrophilic protein and had a high similarity to Neurospora crassa (CAA50271.1) and other fungi. RT-qPCR analysis determined that AaCaM was differentially upregulated during infection structural differentiation of A. alternata both on hydrophobic and pear wax extract-coated surface, with a 3.37-fold upregulation during the hydrophobic induced appressorium formation period (6 h) and a 1.46-fold upregulation during the infection hyphae formation period (8 h) following pear wax induction. Pharmaceutical analysis showed that the CaM-specific inhibitor, trifluoperazine (TFP), inhibited spore germination and appressorium formation, and affected toxins and melanin biosynthesis in A. alternata. CONCLUSIONS: AaCaM plays an important role in regulating infection structure differentiation and secondary metabolism of A. alternata. SIGNIFICANCE AND IMPACT OF STUDY: Our study provides a theoretical basis for further in-depth investigation of the specific role of AaCaM in the calcium signalling pathway underlying hydrophobic and pear wax-induced infection structure differentiation and pathogenicity of A. alternata.


Assuntos
Pyrus , Alternaria/metabolismo , Cálcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , DNA Complementar/metabolismo , Melaninas/metabolismo , Preparações Farmacêuticas , Doenças das Plantas/microbiologia , Pyrus/genética , Pyrus/metabolismo , Pyrus/microbiologia , Trifluoperazina/metabolismo
5.
FEBS Open Bio ; 10(10): 2097-2106, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32810364

RESUMO

Multiple myeloma (MM) is the second most common hematologic malignancy of immunoglobulin-secreting plasma cells. Recent modern combination therapies have improved survival rates, but many patients develop resistance to novel drugs, leading to relapse. Trifluoperazine (TFP), a typical antipsychotic drug, has been reported to exert antitumor effects by targeting various pathways. Thus far, the role of TFP in MM has not been elucidated. In the current study, we demonstrated that TFP inhibited cell growth and autophagy activity but induced apoptosis of U266 and RPMI 8226 MM cells. Furthermore, cotreatment of these cell lines with TFP and rapamycin, a potent autophagy inducer, reduced cell apoptosis compared with TFP treatment alone. We also found that TFP inhibited nuclear protein 1 (NUPR1) expression. In the presence of TFP, cells stably overexpressing NUPR1 showed a higher viability than cells treated with the nonspecific control. Autophagy suppression and apoptosis induction caused by TFP were also reversed in MM cells upon NUPR1 overexpression. Overall, our results indicate that in the context of MM, TFP targets NUPR1, inhibiting cell growth and inducing apoptosis by autophagy inhibition. Our results could contribute toward efforts for the development of more effective therapies for MM to be tested in future clinical trials.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/metabolismo , Trifluoperazina/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Autofagia/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , China , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Proteínas de Neoplasias/genética , Sirolimo/farmacologia , Trifluoperazina/metabolismo
6.
Biochim Biophys Acta Proteins Proteom ; 1866(5-6): 617-623, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29555454

RESUMO

We compare the binding of an inhibitor with that of a natural peptide to Ca2+ saturated calmodulin (holo-CaM). As inhibitor we have chosen trifluoperazine (TFP) that is inducing a huge conformational change of holo-CaM from the open dumbbell-shaped to the closed globular conformation upon binding. On the other hand, melittin is used as model peptide, which is a well-known natural binding partner of holo-CaM. The experiments are carried out as a function of pressure to reveal the contribution of volume or packing effects to the stability of the calmodulin-ligand complexes. From high-pressure Fourier transform infrared (FTIR) spectroscopy, we find that the holo-CaM/TFP complex has a much higher pressure stability than the holo-CaM/melittin complex. Although the analysis of the secondary structure of holo-CaM (without and with ligand) indicates no major changes up to several kbar, pressure-induced exposure of α-helices to water is most pronounced for holo-CaM without ligand, followed by holo-CaM/melittin and then holo-CaM/TFP. Moreover, structural pressure resistance of the holo-CaM/TFP complex in comparison with the holo-CaM/melittin complex is also clearly visible by higher Ca2+ affinity. Förster resonance energy transfer (FRET) from the Tyr residues of holo-CaM to the Trp residue of melittin even suggests some partial dissociation of the complex under pressure which points to void volumes at the protein-ligand interface and to electrostatic binding. Thus, all results of this study show that the inhibitor TFP binds to holo-CaM with higher packing density than the peptide melittin enabling a favorable volume contribution to the inhibitor efficiency.


Assuntos
Calmodulina/metabolismo , Transferência Ressonante de Energia de Fluorescência , Meliteno/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Trifluoperazina/metabolismo , Animais , Sítios de Ligação , Calmodulina/antagonistas & inibidores , Calmodulina/química , Ligantes , Meliteno/química , Modelos Moleculares , Pressão , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Ratos , Relação Estrutura-Atividade , Trifluoperazina/química
7.
Mol Biochem Parasitol ; 221: 14-22, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29453993

RESUMO

Schistosoma mansoni, like other trematodes, expresses a number of unusual calcium binding proteins which consist of an EF-hand domain joined to a dynein light chain-like (DLC-like) domain by a flexible linker. These proteins have been implicated in host immune responses and drug binding. Three members of this protein family from S. mansoni (SmTAL1, SmTAL2 and SmTAL3) have been well characterised biochemically. Here we characterise the remaining family members from this species (SmTAL4-13). All of these proteins form homodimers and all except SmTAL5 bind to calcium and manganese ions. SmTAL9, 10 and 11 also bind to magnesium ions. The antischistosomal drug, praziquantel interacts with SmTAL4, 5 and 8. Some family members also bind to calmodulin antagonists such as chlorpromazine and trifluoperazine. Molecular modelling suggests that all ten proteins adopt similar overall folds with the EF-hand and DLC-like domains folding discretely. Bioinformatics analyses suggest that the proteins may fall into two main categories: (i) those which bind calcium ions reversibly at the second EF-hand and may play a role in signalling (SmTAL1, 2, 8 and 12) and (ii) those which bind calcium ions at the first EF-hand and may play either signalling or structural roles (SmTAL7, 9, 10 and 13). The remaining proteins include those which do not bind calcium ions (SmTAL3 and 5) and three other proteins (SmTAL4, 6 and 11). The roles of these proteins are less clear, but they may also have structural roles.


Assuntos
Alérgenos/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Helminto/metabolismo , Schistosoma mansoni/química , Alérgenos/química , Animais , Anti-Helmínticos/metabolismo , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/química , Cátions Bivalentes/metabolismo , Clorpromazina/metabolismo , Proteínas de Helminto/química , Manganês/metabolismo , Modelos Moleculares , Praziquantel/metabolismo , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Multimerização Proteica , Trifluoperazina/metabolismo
8.
Phys Chem Chem Phys ; 20(5): 3514-3522, 2018 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-29336441

RESUMO

Calmodulin (CaM) is a Ca2+ sensor and mediates Ca2+ signaling through binding of numerous target ligands. The binding of ligands by Ca2+-saturated CaM (holo-CaM) is governed by attractive hydrophobic and electrostatic interactions that are weakened under high pressure in aqueous solutions. Moreover, the potential formation of void volumes upon ligand binding creates a further source of pressure sensitivity. Hence, high pressure is a suitable thermodynamic variable to probe protein-ligand interactions. In this study, we compare the binding of two different ligands to holo-CaM as a function of pressure by using X-ray and neutron scattering techniques. The two ligands are the farnesylated hypervariable region (HVR) of the K-Ras4B protein, which is a natural binding partner of holo-CaM, and the antagonist trifluoperazine (TFP), which is known to inhibit holo-CaM activity. From small-angle X-ray scattering experiments performed up to 3000 bar, we observe a pressure-induced partial unfolding of the free holo-CaM in the absence of ligands, where the two lobes of the dumbbell-shaped protein are slightly swelled. In contrast, upon binding TFP, holo-CaM forms a closed globular conformation, which is pressure stable at least up to 3000 bar. The HVR of K-Ras4B shows a different binding behavior, and the data suggest the dissociation of the holo-CaM/HVR complex under high pressure, probably due to a less dense protein contact of the HVR as compared to TFP. The elastic incoherent neutron scattering experiments corroborate these findings. Below 2000 bar, pressure induces enhanced atomic fluctuations in both holo-CaM/ligand complexes, but those of the holo-CaM/HVR complex seem to be larger. Thus, the inhibition of holo-CaM by TFP is supported by a low-volume ligand binding, albeit this is not associated with a rigidification of the complex structure on the sub-ns Å-scale.


Assuntos
Calmodulina/química , Ligantes , Sequência de Aminoácidos , Cálcio/química , Cálcio/metabolismo , Calmodulina/metabolismo , Difração de Nêutrons , Pressão , Ligação Proteica , Espalhamento a Baixo Ângulo , Trifluoperazina/química , Trifluoperazina/metabolismo , Difração de Raios X
9.
Pharmacol Biochem Behav ; 165: 9-13, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29273456

RESUMO

Trifluoperazine, a typical antipsychotic drug, not only antagonizes dopamine D2 receptors but also enhances serotonin 5-HT2 receptor-mediated behavior. Moreover, trifluoperazine suppresses human purinergic receptor P2X7 responses and calmodulin. However, the effect of trifluoperazine on marble-burying behavior, which has been considered an animal model of obsessive-compulsive disorder (OCD), has not been studied. Here, we examined the effect of trifluoperazine on marble-burying behavior in mice. Oral administration of paroxetine, a selective serotonin reuptake inhibitor, significantly reduced marble-burying behavior without affecting total locomotor activity. Similar results were obtained for trifluoperazine (3mg/kg). The D2 receptor agonist, quinpirole (0.03mg/kg, intraperitoneal [i.p.]), and 5-HT2A receptor antagonist, ketanserin (0.3mg/kg, i.p.), significantly counteracted this reduction of marble-burying behavior by trifluoperazine. These results show that trifluoperazine reduces marble-burying behavior via D2 and 5-HT2A receptors, and may be a useful drug for the treatment of OCD.


Assuntos
Antipsicóticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Antagonistas dos Receptores de Dopamina D2/farmacologia , Transtorno Obsessivo-Compulsivo/tratamento farmacológico , Agonistas do Receptor 5-HT2 de Serotonina/farmacologia , Trifluoperazina/farmacologia , Animais , Antipsicóticos/metabolismo , Antipsicóticos/uso terapêutico , Modelos Animais de Doenças , Agonistas de Dopamina/farmacologia , Antagonistas dos Receptores de Dopamina D2/metabolismo , Antagonistas dos Receptores de Dopamina D2/uso terapêutico , Ketanserina/farmacologia , Masculino , Camundongos Endogâmicos ICR , Atividade Motora , Paroxetina/farmacologia , Quimpirol/farmacologia , Receptor 5-HT2A de Serotonina/metabolismo , Receptores de Dopamina D2/metabolismo , Agonistas do Receptor 5-HT2 de Serotonina/metabolismo , Agonistas do Receptor 5-HT2 de Serotonina/uso terapêutico , Antagonistas da Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Trifluoperazina/metabolismo , Trifluoperazina/uso terapêutico
10.
Curr Clin Pharmacol ; 12(1): 50-54, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28103775

RESUMO

BACKGROUND: In patients with the Congenital Sucrase-Isomaltase Deficiency (CSID), who lack intestinal sucrase-isomaltase enzyme, a suspension of yeast sucrase is applied as a drug to compensate the enzyme deficiency. While antipsychotic drugs are used for the treatment of schizophrenia, administering multiple drugs at the same time may counteract each other. METHODS: In this study, the interaction between trifluoperazine and haloperidol as antipsychotic drugs on oral drug yeast sucrase was investigated. In this regard, the kinetic parameters of enzyme were determined in the presence or absence of the drugs. The kinetic parameters of the drugs such as Ki and IC50 were also calculated. Lineweaver - Burk plot was used to reveal the type of inhibition. RESULTS: The results showed that both drugs could reduce sucrase activity and decrease the Vmax of the enzyme by non-competitive inhibition. The IC50 and Ki values of the drugs were determined to be 0.7 and 0.068 mM and 0.45 and 0.063 mM for haloperidol and trifluoperazine, respectively. The results suggested that trifluoperazine binds to the enzyme with higher affinity than haloperidol. Fluorescence measurement was used for conformational investigations of the drugs and sucrase interaction. It was shown that the drugs bind to free enzyme and enzyme-substrate complex which are accompanied with hyperchromicity. This suggests that tryptophan residues of the enzyme transferred to hydrophobic medium after binding of the drugs to the enzyme. CONCLUSION: The finding of this research revealed that both trifluoperazine and haloperidol could inhibit sucrase in non-competitive manner. The kinetic parameters and conformational changes due to binding of trifluoperazine to the enzyme were different from that of haloperidol.


Assuntos
Antipsicóticos/farmacologia , Inibidores Enzimáticos/farmacologia , Terapia de Reposição de Enzimas/métodos , Haloperidol/farmacologia , Sacarase/antagonistas & inibidores , Trifluoperazina/farmacologia , Regulação Alostérica , Antipsicóticos/química , Antipsicóticos/metabolismo , Sítios de Ligação , Interações Medicamentosas , Inibidores Enzimáticos/química , Terapia de Reposição de Enzimas/efeitos adversos , Haloperidol/química , Haloperidol/metabolismo , Humanos , Cinética , Ligação Proteica , Conformação Proteica , Medição de Risco , Relação Estrutura-Atividade , Sacarase/química , Sacarase/metabolismo , Sacarase/farmacologia , Trifluoperazina/química , Trifluoperazina/metabolismo
11.
Exp Parasitol ; 170: 109-115, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27693219

RESUMO

FH22 has been previously identified as a calcium-binding protein from the common liver fluke, Fasciola hepatica. It is part of a family of at least four proteins in this organism which combine an EF-hand containing N-terminal domain with a C-terminal dynein light chain-like domain. Here we report further biochemical properties of FH22, which we propose should be renamed FhCaBP1 for consistency with other family members. Molecular modelling predicted that the two domains are linked by a flexible region and that the second EF-hand in the N-terminal domain is most likely the calcium ion binding site. Native gel electrophoresis demonstrated that the protein binds both calcium and manganese ions, but not cadmium, magnesium, strontium, barium, cobalt, copper(II), iron (II), nickel, zinc, lead or potassium ions. Calcium ion binding alters the conformation of the protein and increases its stability towards thermal denaturation. FhCaBP1 is a dimer in solution and calcium ions have no detectable effect on the protein's ability to dimerise. FhCaBP1 binds to the calmodulin antagonists trifluoperazine and chlorpromazine. Overall, the FhCaBP1's biochemical properties are most similar to FhCaBP2 a fact consistent with the close sequence and predicted structural similarity between the two proteins.


Assuntos
Proteínas de Ligação ao Cálcio/química , Dineínas/química , Fasciola hepatica/metabolismo , Proteínas de Helminto/química , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/isolamento & purificação , Proteínas de Ligação ao Cálcio/metabolismo , Calmodulina/antagonistas & inibidores , Clorpromazina/metabolismo , Eletroforese em Gel de Poliacrilamida , Fasciola hepatica/genética , Expressão Gênica , Proteínas de Helminto/genética , Proteínas de Helminto/isolamento & purificação , Proteínas de Helminto/metabolismo , Manganês/metabolismo , Conformação Proteica , Multimerização Proteica , Trifluoperazina/metabolismo
12.
Pharmacol Res ; 110: 139-150, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27208893

RESUMO

The wide application of herbal medicines and foods containing steroids has resulted in the high risk of herb-drug interactions (HDIs). The present study aims to evaluate the inhibition potential of 43 natural steroids from herb medicines toward human UDP- glucuronosyltransferases (UGTs). A remarkable structure-dependent inhibition toward UGT1A4 was observed in vitro. Some natural steroids such as gitogenin, tigogenin, and solasodine were found to be the novel selective inhibitors of UGT1A4, and did not inhibit the activities of major human CYP isoforms. To clarify the possibility of the in vivo interaction of common steroids and clinical drugs, the kinetic inhibition type and related kinetic parameters (Ki) were measured. The target compounds 2-6 and 15, competitively inhibited the UGT1A4-catalyzed trifluoperazine glucuronidation reaction, with Ki values of 0.6, 0.18, 1.1, 0.7, 0.8, and 12.3µM, respectively. And this inhibition of steroids towards UGT1A4 was also verified in human primary hepatocytes. Furthermore, a quantitative structure-activity relationship (QSAR) of steroids with inhibitory effects toward human UGT1A4 isoform was established using the computational methods. Our findings elucidate the potential for in vivo HDI effects of steroids in herbal medicine and foods, with the clinical dr ugs eliminated by UGT1A4, and reveal the vital pharamcophoric requirement of natural steroids for UGT1A4 inhibition activity.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Hepatócitos/efeitos dos fármacos , Interações Ervas-Drogas , Fitosteróis/farmacologia , Preparações de Plantas/farmacologia , Simulação por Computador , Inibidores Enzimáticos/química , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Hepatócitos/enzimologia , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Inativação Metabólica , Cinética , Modelos Moleculares , Estrutura Molecular , Fitosteróis/química , Preparações de Plantas/química , Relação Quantitativa Estrutura-Atividade , Saponinas/química , Saponinas/farmacologia , Eletricidade Estática , Tamoxifeno/metabolismo , Trifluoperazina/metabolismo
13.
Drug Metab Dispos ; 43(11): 1769-72, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26329791

RESUMO

The P24T polymorphic variant of the human UDP-glucuronosyltransferase 1A4 (UGT1A4*2, 70C>A) occurs within the signal peptide, five amino acids upstream of the cleavage site and the start of the mature protein. Bioinformatic analysis of the variant suggested that the signal peptide of part of the translated protein is cleaved two residues upstream of the regular site, whereas the rest is cleaved as usual. To test this, recombinant UGT1A4-P24T, with a C-terminal His-tag, was expressed in sf9 insect cells and affinity-purified for N-terminal protein sequencing. The results were in agreement with the in silico prediction. About half of the mutant protein was cleaved at the regular site, between S28 and G29, whereas the other half was cleaved two amino acids upstream, between A26 and E27. The glucuronidation of two substrates, dexmedetomidine and trifluoperazine, was assayed using membrane-enriched UGT1A4-P24T and wild-type UGT1A4. The variant exhibited much lower glucuronidation rates, but kinetic analyses revealed large differences between them only in the Vmax values. The Km values for both substrates were not affected by the mutation and its consequences. This might suggest that the unusual signal peptide cleavage in UGT1A4-P24T somehow disturbs protein folding. Moreover, it raises the possibility that the effect of UGT1A4-P24T on the glucuronidation rate in mammalian expression systems would be mild since they contain more effective post-translation protein control systems in the endoplasmic reticulum. In summary, our results reveal the effect of a polymorphic mutation on the signal sequence cleavage and thereby also the mature UGT.


Assuntos
Variação Genética/genética , Glucuronosiltransferase/genética , Polimorfismo Genético/genética , Animais , Células HEK293 , Humanos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Trifluoperazina/metabolismo , Trifluoperazina/farmacologia
14.
Microb Cell Fact ; 14: 82, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-26062974

RESUMO

BACKGROUND: Getting access to authentic human drug metabolites is an important issue during the drug discovery and development process. Employing recombinant microorganisms as whole-cell biocatalysts constitutes an elegant alternative to organic synthesis to produce these compounds. The present work aimed for the generation of an efficient whole-cell catalyst based on the flavin monooxygenase isoform 2 (FMO2), which is part of the human phase I metabolism. RESULTS: We show for the first time the functional expression of human FMO2 in E. coli. Truncations of the C-terminal membrane anchor region did not result in soluble FMO2 protein, but had a significant effect on levels of recombinant protein. The FMO2 biocatalysts were employed for substrate screening purposes, revealing trifluoperazine and propranolol as FMO2 substrates. Biomass cultivation on the 100 L scale afforded active catalyst for biotransformations on preparative scale. The whole-cell conversion of trifluoperazine resulted in perfectly selective oxidation to 48 mg (46% yield) of the corresponding N (1)-oxide with a purity >98%. CONCLUSIONS: The generated FMO2 whole-cell catalysts are not only useful as screening tool for human metabolites of drug molecules but more importantly also for their chemo- and regioselective preparation on the multi-milligram scale.


Assuntos
Escherichia coli/genética , Oxigenases de Função Mista/química , Oxigenases de Função Mista/metabolismo , Preparações Farmacêuticas/metabolismo , Biocatálise , Dinitrocresóis/metabolismo , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Oxigenases de Função Mista/genética , Propranolol/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Trifluoperazina/metabolismo
15.
Pharmazie ; 70(4): 239-43, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26012253

RESUMO

UDP-glucuronosyltransferases (UGTs) are involved in the clearance of many important drugs and endogenous substances, and inhibition of UGTs' activity by herbal components might induce severe herb-drug interactions or metabolic disturbances of endogenous substances. The present study aims to determine the inhibition of UGTs' activity by podophyllotoxin derivatives, trying to indicate the potential herb-drug interaction or metabolic influence towards endogenous substances' metabolism. Recombinant UGT isoforms (except UGT1A4)-catalyzed 4-methylumbelliferone (4-MU) glucuronidation reaction and UGT1A4-catalyzed trifluoperazine (TFP) glucuronidation were employed to firstly screen the podophyllotoxin derivatives' inhibition potential. Structure-dependent inhibition behavior of podophyllotoxin derivatives towards UGT isoforms was detected. Inhibition kinetic type and parameter (Ki) were determined for the inhi- bition of podophyllotoxin towards UGT1A1, and competitive inhibition of podophyllotoxin towards UGT1A1 was observed with the inhibition kinetic parameter (Ki) to be 4.0 µM. Furthermore, podophyllotoxin was demonstrated to exert medium and weak inhibition potential towards human liver microsomes (HLMs)-catalyzed SN-38 glucuronidation and estradiol-3-glucuronidation. In conclusion, podophyllotoxin inhibited UGT1A1 activity, indicating potential herb-drug interactions between podophyllotoxin-containing herbs and drugs mainly undergoing UGT1A1-mediated metabolism.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Podofilotoxina/farmacologia , Ligação Competitiva/efeitos dos fármacos , Camptotecina/análogos & derivados , Camptotecina/metabolismo , Interações Medicamentosas , Inibidores Enzimáticos/química , Estradiol/metabolismo , Glucuronídeos/metabolismo , Humanos , Himecromona/metabolismo , Técnicas In Vitro , Irinotecano , Isoenzimas/antagonistas & inibidores , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Podofilotoxina/química , Relação Estrutura-Atividade , Especificidade por Substrato , Trifluoperazina/metabolismo
16.
J Lipid Res ; 54(12): 3334-44, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24115227

RESUMO

Increased levels of bile acids (BAs) due to the various hepatic diseases could interfere with the metabolism of xenobiotics, such as drugs, and endobiotics including steroid hormones. UDP-glucuronosyltransferases (UGTs) are involved in the conjugation and elimination of many xenobiotics and endogenous compounds. The present study sought to investigate the potential for inhibition of UGT enzymes by BAs. The results showed that taurolithocholic acid (TLCA) exhibited the strongest inhibition toward UGTs, followed by lithocholic acid. Structure-UGT inhibition relationships of BAs were examined and in vitro-in vivo extrapolation performed by using in vitro inhibition kinetic parameters (Ki) in combination with calculated in vivo levels of TLCA. Substitution of a hydrogen with a hydroxyl group in the R1, R3, R4, R5 sites of BAs significantly weakens their inhibition ability toward most UGTs. The in vivo inhibition by TLCA toward UGT forms was determined with following orders of potency: UGT1A4 > UGT2B7 > UGT1A3 > UGT1A1 ∼ UGT1A7 ∼ UGT1A10 ∼ UGT2B15. In conclusion, these studies suggest that disrupted homeostasis of BAs, notably taurolithocholic acid, found in various diseases such as cholestasis, could lead to altered metabolism of xenobiotics and endobiotics through inhibition of UGT enzymes.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Glucuronosiltransferase/metabolismo , Doenças Metabólicas/enzimologia , Ácido Taurolitocólico/farmacologia , Biocatálise/efeitos dos fármacos , Glucuronosiltransferase/química , Humanos , Himecromona/metabolismo , Intestinos/enzimologia , Cinética , Fígado/enzimologia , Modelos Moleculares , Conformação Proteica , Trifluoperazina/metabolismo
17.
Biochimie ; 94(11): 2398-406, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22727910

RESUMO

An 18.2 kDa protein from the liver fluke, Fasciola hepatica has been identified and characterised. The protein shows strongest sequence similarity to egg antigen proteins from Schistosoma mansoni, Schistosoma japonicum and Clonorchis sinensis. The protein is predicted to adopt a calmodulin-like fold; it thus represents the third calmodulin-like protein to be characterised in F. hepatica and has been named FhCaM3. Compared to the classical calmodulin structure there are some variations. Most noticeably, the central, linker helix is disrupted by a cysteine residue. Alkaline native gel electrophoresis showed that FhCaM3 binds calcium ions. This binding event increases the ability of the protein to bind the hydrophobic fluorescent probe 8-anilinonaphthalene-1-sulphonate, consistent with an increase in surface hydrophobicity as seen in other calmodulins. FhCaM3 binds to the calmodulin antagonists trifluoperazine and W7, but not to the myosin regulatory light chain binding compound praziquantel. Immunolocalisation demonstrated that the protein is found in eggs and vitelline cells. Given the critical role of calcium ions in egg formation and hatching this suggests that FhCaM3 may play a role in calcium signalling in these processes. Consequently the antagonism of FhCaM3 may, potentially, offer a method for inhibiting egg production and thus reducing the spread of infection.


Assuntos
Calmodulina/química , Calmodulina/metabolismo , Fasciola hepatica , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Calmodulina/antagonistas & inibidores , Calmodulina/isolamento & purificação , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Sulfonamidas/metabolismo , Sulfonamidas/farmacologia , Trifluoperazina/metabolismo , Trifluoperazina/farmacologia
18.
Drug Metab Dispos ; 39(9): 1522-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21632963

RESUMO

The aim of this study was to investigate the effect of commonly used botanicals on UDP-glucuronosyltransferase (UGT) 1A4, UGT1A6, and UGT1A9 activities in human liver microsomes. The extracts screened were black cohosh, cranberry, echinacea, garlic, ginkgo, ginseng, milk thistle, saw palmetto, and valerian in addition to the green tea catechin epigallocatechin gallate (EGCG). Formation of trifluoperazine glucuronide, serotonin glucuronide, and mycophenolic acid phenolic glucuronide was used as an index reaction for UGT1A4, UGT1A6, and UGT1A9 activities, respectively, in human liver microsomes. Inhibition potency was expressed as the concentration of the inhibitor at 50% activity (IC(50)) and the volume in which the dose could be diluted to generate an IC(50)-equivalent concentration [volume/dose index (VDI)]. Potential inhibitors were EGCG for UGT1A4, milk thistle for both UGT1A6 and UGT1A9, saw palmetto for UGT1A6, and cranberry for UGT1A9. EGCG inhibited UGT1A4 with an IC(50) value of (mean ± S.E.) 33.8 ± 3.1 µg/ml. Milk thistle inhibited both UGT1A6 and UGT1A9 with IC(50) values of 59.5 ± 3.6 and 33.6 ± 3.1 µg/ml, respectively. Saw palmetto and cranberry weakly inhibited UGT1A6 and UGT1A9, respectively, with IC(50) values >100 µg/ml. For each inhibition, VDI was calculated to determine the potential of achieving IC(50)-equivalent concentrations in vivo. VDI values for inhibitors indicate a potential for inhibition of first-pass glucuronidation of UGT1A4, UGT1A6, and UGT1A9 substrates. These results highlight the possibility of herb-drug interactions through modulation of UGT enzyme activities. Further clinical studies are warranted to investigate the in vivo extent of the observed interactions.


Assuntos
Glucuronídeos/metabolismo , Glucuronosiltransferase/antagonistas & inibidores , Microssomos Hepáticos/enzimologia , Extratos Vegetais/farmacologia , Trifluoperazina/metabolismo , Glucuronosiltransferase/metabolismo , Interações Ervas-Drogas , Humanos , Microssomos Hepáticos/efeitos dos fármacos
19.
Drug Metab Dispos ; 39(9): 1486-94, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21673130

RESUMO

Eslicarbazepine acetate (ESL) is a once-daily novel antiepileptic drug approved in Europe for use as adjunctive therapy for refractory partial-onset seizures with or without secondary generalization. Metabolism of ESL consists primarily of hydrolysis to eslicarbazepine, which is then subject to glucuronidation followed by renal excretion. In this study, we have identified that human liver microsomes (HLM) enriched with uridine 5'-diphosphoglucuronic acid give origin to a single Escherichia coli ß-glucuronidase-sensitive eslicarbazepine glucuronide (most likely the O-glucuronide). The kinetics of eslicarbazepine glucuronidation in HLM was investigated in the presence and absence of bovine serum albumin (BSA). The apparent K(m) were 412.2 ± 63.8 and 349.7 ± 74.3 µM in the presence and absence of BSA, respectively. Incubations with recombinant human UDP glucuronosyltransferases (UGTs) indicated that UGT1A4, UGT1A9, UGT2B4, UGT2B7, and UGT2B17 appear to be involved in eslicarbazepine conjugation. The UGT with the highest affinity for conjugation was UGT2B4 (K(m) = 157.0 ± 31.2 and 28.7 ± 10.1 µM, in the absence and presence of BSA, respectively). There was a significant correlation between eslicarbazepine glucuronidation and trifluoperazine glucuronidation, a typical UGT1A4 substrate; however, no correlation was found with typical substrates for UGT1A1 and UGT1A9. Diclofenac inhibited eslicarbazepine glucuronidation in HLM with an IC(50) value of 17 µM. In conclusion, glucuronidation of eslicarbazepine results from the contribution of UGT1A4, UGT1A9, UGT2B4, UGT2B7, and UGT2B17, but the high-affinity component of the UGT2B4 isozyme may play a major role at therapeutic plasma concentrations of unbound eslicarbazepine.


Assuntos
Dibenzazepinas/metabolismo , Glucuronosiltransferase/metabolismo , Fígado/metabolismo , Microssomos Hepáticos/metabolismo , Animais , Anticonvulsivantes/metabolismo , Escherichia coli/metabolismo , Glucuronidase/metabolismo , Glucuronídeos/metabolismo , Humanos , Cinética , Fígado/enzimologia , Camundongos , Microssomos Hepáticos/enzimologia , Soroalbumina Bovina/metabolismo , Trifluoperazina/metabolismo , Uridina Difosfato Ácido Glucurônico/metabolismo
20.
J Med Chem ; 54(11): 3875-84, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21495717

RESUMO

A novel, sensible, and specific fluorescent biosensor of human calmodulin (hCaM), namely hCaM L39C-mBBr/V91C-mBBr, was constructed. The biosensor was useful for detecting ligands with opposing fluorescent signals, calcium ions (Ca(2+)) and CaM inhibitors in solution. Thus, the device was successfully applied to analyze the allosteric effect of Ca(2+) on trifluoroperazine (TFP) binding to CaM (Ca(2+)K(d) = 0.24 µM ± 0.03 with a stoichiometry 4.10 ± 0.15; TFPK(d) ∼ 5.74-0.53 µM depending on the degree of saturation of Ca(2+), with a stoichiometry of 2:1). In addition, it was suitable for discovering additional xanthones (5, 6, and 8) with anti-CaM properties from the fungus Emericella 25379. The affinity of 1-5, 7, and 8 for the complex (Ca(2+))(4)-CaM was excellent because their experimental K(d)s were in the nM range (4-498 nM). Docking analysis predicted that 1-8 bind to CaM at sites I, III, and IV as does TFP.


Assuntos
Técnicas Biossensoriais , Cálcio/análise , Calmodulina/antagonistas & inibidores , Calmodulina/metabolismo , Emericella/química , Corantes Fluorescentes/síntese química , Xantonas/metabolismo , Xantonas/farmacologia , Antipsicóticos/análise , Antipsicóticos/química , Sítios de Ligação , Compostos Bicíclicos com Pontes/química , Calmodulina/síntese química , Calmodulina/química , Calmodulina/genética , Calmodulina/farmacologia , Emericella/crescimento & desenvolvimento , Emericella/metabolismo , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Corantes Fluorescentes/farmacologia , Humanos , Ligantes , Modelos Moleculares , Conformação Molecular , Patentes como Assunto , Trifluoperazina/metabolismo , Xantonas/análise , Xantonas/síntese química , Xantonas/química , Xantonas/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...