Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 375
Filtrar
Más filtros

Medicinas Complementárias
Tipo del documento
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 3390, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37296102

RESUMEN

Elucidating intracellular drug targets is a difficult problem. While machine learning analysis of omics data has been a promising approach, going from large-scale trends to specific targets remains a challenge. Here, we develop a hierarchic workflow to focus on specific targets based on analysis of metabolomics data and growth rescue experiments. We deploy this framework to understand the intracellular molecular interactions of the multi-valent dihydrofolate reductase-targeting antibiotic compound CD15-3. We analyse global metabolomics data utilizing machine learning, metabolic modelling, and protein structural similarity to prioritize candidate drug targets. Overexpression and in vitro activity assays confirm one of the predicted candidates, HPPK (folK), as a CD15-3 off-target. This study demonstrates how established machine learning methods can be combined with mechanistic analyses to improve the resolution of drug target finding workflows for discovering off-targets of a metabolic inhibitor.


Asunto(s)
Antibacterianos , Proteínas , Proteínas/química , Metabolómica , Tetrahidrofolato Deshidrogenasa/genética , Poder Psicológico
2.
Hum Mol Genet ; 31(7): 1151-1158, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-34788822

RESUMEN

BACKGROUND: Higher serum homocysteine is associated with cognitive decline in older people. But homocysteine-lowering trials including folic acid (FA) show inconsistent results on cognitive decline. The reduction of FA to dihydrofolate by dihydrofolate reductase (DHFR) is slow in humans. OBJECTIVE: We examined the effects of the DHFR 19-bp deletion/insertion (del/ins) polymorphism on FA-containing treatment on cognitive decline and brain atrophy in older people with mild cognitive impairment (MCI). METHODS: This study used pooled data from two randomized B-vitamin trials on 545 MCI subjects who received either FA-containing B vitamins or placebo for 24 months. Subjects were typed for the DHFR genotype. Primary outcome was the Clinical Dementia Rating scale-global score (CDR-global). Secondary outcomes were CDR-sum of boxes score (CDR-SOB), memory and executive Z-scores and whole brain atrophy rate by serial MRI. RESULTS: The proportions of subjects with del/del, del/ins and ins/ins genotype were 29.5, 44.3 and 26.1%, respectively. DHFR genotypes modified the effects of B vitamins on CDR-global, CDR-SOB and executive function Z-score (Pinteraction = 0.017, 0.014 and 0.052, respectively), with significant benefits being observed only in those with ins/ins genotype (Beta = -1.367, -0.614 and 0.315, P = 0.004, 0.014 and 0.012, respectively). The interaction was not significant for memory Z-score and whole brain atrophy rate. Notably, the supplements only slowed brain atrophy in members of the 'ins/ins' group who were not using aspirin. CONCLUSIONS: Our data indicate that the beneficial effects of B vitamins including FA on cognitive function are only apparent in those with ins/ins genotype, i.e. relatively better preserved DHFR activity.


Asunto(s)
Trastornos del Conocimiento , Disfunción Cognitiva , Complejo Vitamínico B , Anciano , Cognición , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/genética , Humanos , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/farmacología , Complejo Vitamínico B/uso terapéutico
3.
Microbiol Spectr ; 9(3): e0073921, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34937180

RESUMEN

Antimicrobial resistance (AMR) has become a serious public and economic threat. The rate of bacteria acquiring AMR surpasses the rate of new antibiotics discovery, projecting more deadly AMR infections in the future. The Pathogen Box is an open-source library of drug-like compounds that can be screened for antibiotic activity. We have screened molecules of the Pathogen Box against Vibrio cholerae, the cholera-causing pathogen, and successfully identified two compounds, MMV687807 and MMV675968, that inhibit growth. RNA-seq analyses of V. cholerae after incubation with each compound revealed that both compounds affect cellular functions on multiple levels including carbon metabolism, iron homeostasis, and biofilm formation. In addition, whole-genome sequencing analysis of spontaneous resistance mutants identified an efflux system that confers resistance to MMV687807. We also identified that the dihydrofolate reductase is the likely target of MMV675968 suggesting it acts as an analog of trimethoprim but with a MIC 14-fold lower than trimethoprim in molar concentration. In summary, these two compounds that effectively inhibit V. cholerae and other bacteria may lead to the development of new antibiotics for better treatment of the cholera disease. IMPORTANCE Cholera is a serious infectious disease in tropical regions causing millions of infections annually. Vibrio cholerae, the causative agent of cholera, has gained multi-antibiotic resistance over the years, posing greater threat to public health and current treatment strategies. Here we report two compounds that effectively target the growth of V. cholerae and have the potential to control cholera infection.


Asunto(s)
Antibacterianos/farmacología , Cólera/tratamiento farmacológico , Evaluación Preclínica de Medicamentos/métodos , Antagonistas del Ácido Fólico/farmacología , Vibrio cholerae/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Genoma Bacteriano/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Trimetoprim/análogos & derivados , Trimetoprim/farmacología , Vibrio cholerae/genética , Vibrio cholerae/crecimiento & desarrollo , Secuenciación Completa del Genoma
4.
Molecules ; 26(21)2021 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-34771128

RESUMEN

In this work, co-crystal screening was carried out for two important dihydrofolate reductase (DHFR) inhibitors, trimethoprim (TMP) and pyrimethamine (PMA), and for 2,4-diaminopyrimidine (DAP), which is the pharmacophore of these active pharmaceutical ingredients (API). The isomeric pyridinecarboxamides and two xanthines, theophylline (THEO) and caffeine (CAF), were used as co-formers in the same experimental conditions, in order to evaluate the potential for the pharmacophore to be used as a guide in the screening process. In silico co-crystal screening was carried out using BIOVIA COSMOquick and experimental screening was performed by mechanochemistry and supported by (solid + liquid) binary phase diagrams, infrared spectroscopy (FTIR) and X-ray powder diffraction (XRPD). The in silico prediction of low propensities for DAP, TMP and PMA to co-crystallize with pyridinecarboxamides was confirmed: a successful outcome was only observed for DAP + nicotinamide. Successful synthesis of multicomponent solid forms was achieved for all three target molecules with theophylline, with DAP co-crystals revealing a greater variety of stoichiometries. The crystalline structures of a (1:2) TMP:THEO co-crystal and of a (1:2:1) DAP:THEO:ethyl acetate solvate were solved. This work demonstrated the possible use of the pharmacophore of DHFR inhibitors as a guide for co-crystal screening, recognizing some similar trends in the outcome of association in the solid state and in the molecular aggregation in the co-crystals, characterized by the same supramolecular synthons.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Pirimetamina/farmacología , Pirimidinas/farmacología , Tetrahidrofolato Deshidrogenasa/metabolismo , Trimetoprim/farmacología , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Pirimetamina/química , Pirimidinas/química , Trimetoprim/química
5.
Nat Commun ; 12(1): 3486, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34108489

RESUMEN

The metabolome represents a complex network of biological events that reflects the physiologic state of the organism in health and disease. Additionally, specific metabolites and metabolic signaling pathways have been shown to modulate animal ageing, but whether there are convergent mechanisms uniting these processes remains elusive. Here, we used high resolution mass spectrometry to obtain the metabolomic profiles of canonical longevity pathways in C. elegans to identify metabolites regulating life span. By leveraging the metabolomic profiles across pathways, we found that one carbon metabolism and the folate cycle are pervasively regulated in common. We observed similar changes in long-lived mouse models of reduced insulin/IGF signaling. Genetic manipulation of pathway enzymes and supplementation with one carbon metabolites in C. elegans reveal that regulation of the folate cycle represents a shared causal mechanism of longevity and proteoprotection. Such interventions impact the methionine cycle, and reveal methionine restriction as an underlying mechanism. This comparative approach reveals key metabolic nodes to enhance healthy ageing.


Asunto(s)
Carbono/metabolismo , Ácido Fólico/metabolismo , Longevidad/fisiología , Redes y Vías Metabólicas , Animales , Caenorhabditis elegans , Insulina/metabolismo , Longevidad/genética , Redes y Vías Metabólicas/genética , Metaboloma , Metionina/metabolismo , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación , Péptidos/metabolismo , Transducción de Señal , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Tetrahidrofolatos/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo
6.
J Enzyme Inhib Med Chem ; 36(1): 198-206, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33530764

RESUMEN

In various malaria-endemic regions, the appearance of resistance has precluded the use of pyrimidine-based antifolate drugs. Here, a three-step fragment screening was used to identify new non-pyrimidine Plasmodium falciparum dihydrofolate reductase (PfDHFR) inhibitors. Starting from a 1163-fragment commercial library, a two-step differential scanning fluorimetry screen identified 75 primary fragment hits. Subsequent enzyme inhibition assay identified 11 fragments displaying IC50 in the 28-695 µM range and selectivity for PfDHFR. In addition to the known pyrimidine, three new anti-PfDHFR chemotypes were identified. Fragments from each chemotype were successfully co-crystallized with PfDHFR, revealing a binding in the active site, in the vicinity of catalytic residues, which was confirmed by molecular docking on all fragment hits. Finally, comparison with similar non-hit fragments provides preliminary input on available growth vectors for future drug development.


Asunto(s)
Antimaláricos/farmacología , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/antagonistas & inhibidores , Antimaláricos/síntesis química , Antimaláricos/química , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Plasmodium falciparum/enzimología , Proguanil/síntesis química , Proguanil/química , Proguanil/farmacología , Proteínas Protozoarias/aislamiento & purificación , Proteínas Protozoarias/metabolismo , Pirimetamina/síntesis química , Pirimetamina/química , Pirimetamina/farmacología , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/aislamiento & purificación , Tetrahidrofolato Deshidrogenasa/metabolismo , Triazinas/síntesis química , Triazinas/química , Triazinas/farmacología
7.
Curr Drug Discov Technol ; 18(4): 554-569, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32729419

RESUMEN

BACKGROUND: In a bid to come up with effective compounds as inhibitors for antimalarial treatment, we built a library of 2,000 traditional Chinese medicine(TCM)-derived compounds retrieved from TCM Database@Taiwan. METHODS: The active sites of both the wild type and mutant Plasmodium falciparum dihydrofolatereductase (pfDHFR) were explored using computational tools. pfDHFR, one of the prime drug targets in the prevention of malaria infection induced by the female anopheles mosquito has continued to offer resistance to drugs (antifolates) due to mutation in some of the key amino acid residues crucial for its inhibition. RESULTS: We utilized virtual throughput screening and glide XP docking to screen the compounds, and 8 compounds were found to have promising docking scores with both the wild type and mutant pfDHFR. They were further subjected to Induce Fit Docking (IFD) to affirm their inhibitory potency. The ADME properties and biological activity spectrum of the compounds were also considered. The inhibition profile of the compounds revealed that a number of compounds formed intermolecular interactions with ASP54, ILE14, LEU164, SER108/ASN108, ARG122 and ASP58. Most of the compounds can be considered as drug candidates due to their antiprotozoal activities and accordance with the Lipinski's Rule of Five (ROF). CONCLUSION: The outcome of the present study should further be investigated to attest the efficacy of these compounds as better drug candidates than the antifolates.


Asunto(s)
Antimaláricos/farmacología , Medicamentos Herbarios Chinos/farmacología , Malaria Falciparum/prevención & control , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Antimaláricos/química , Antimaláricos/uso terapéutico , Dominio Catalítico/efectos de los fármacos , Diseño de Fármacos/métodos , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/uso terapéutico , Humanos , Malaria Falciparum/parasitología , Simulación del Acoplamiento Molecular , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Tetrahidrofolato Deshidrogenasa/genética
8.
Angiogenesis ; 24(1): 97-110, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32935224

RESUMEN

Gene therapies that chronically suppress vascular endothelial growth factor (VEGF) represent a new approach for managing retinal vascular leakage and neovascularization. However, constitutive suppression of VEGF in the eye may have deleterious side effects. Here, we developed a novel strategy to introduce Flt23k, a decoy receptor that binds intracellular VEGF, fused to the destabilizing domain (DD) of Escherichia coli dihydrofolate reductase (DHFR) into the retina. The expressed DHFR(DD)-Flt23k fusion protein is degraded unless "switched on" by administering a stabilizer; in this case, the antibiotic trimethoprim (TMP). Cells transfected with the DHFR(DD)-Flt23k construct expressed the fusion protein at levels correlated with the TMP dose. Stabilization of the DHFR(DD)-Flt23k fusion protein by TMP was able to inhibit intracellular VEGF in hypoxic cells. Intravitreal injection of self-complementary adeno-associated viral vector (scAAV)-DHFR(DD)-Flt23k and subsequent administration of TMP resulted in tunable suppression of ischemia-induced retinal neovascularization in a rat model of oxygen-induced retinopathy (OIR). Hence, our study suggests a promising novel approach for the treatment of retinal neovascularization. Schematic diagram of the tunable system utilizing the DHFR(DD)-Flt23k approach to reduce VEGF secretion. a The schematic shows normal VEGF secretion. b Without the ligand TMP, the DHFR(DD)-Flt23k protein is destabilized and degraded by the proteasome. c In the presence of the ligand TMP, DHFR(DD)-Flt23k is stabilized and sequestered in the ER, thereby conditionally inhibiting VEGF. Green lines indicate the intracellular and extracellular distributions of VEGF. Blue lines indicate proteasomal degradation of the DHFR(DD)-Flt23k protein. Orange lines indicate the uptake of cell-permeable TMP. TMP, trimethoprim; VEGF, vascular endothelial growth factor; ER, endoplasmic reticulum.


Asunto(s)
Terapia Genética , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Neovascularización Retiniana/genética , Neovascularización Retiniana/terapia , Animales , Hipoxia de la Célula , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Células HEK293 , Humanos , Inyecciones Intravítreas , Dominios Proteicos , Ratas Sprague-Dawley , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo , Transgenes , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Sci Rep ; 10(1): 19844, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33199757

RESUMEN

This study aimed to investigate the influence of chronic ischemia on nitric oxide biosynthesis in the bladder and the effect of administering tetrahydrobiopterin (BH4), a cofactor for endothelial nitric oxide synthase (eNOS), on chronic ischemia-related lower urinary tract dysfunction (LUTD). This study divided male Sprague-Dawley rats into Control, chronic bladder ischemia (CBI) and CBI with oral BH4 supplementation (CBI/BH4) groups. In the CBI group, bladder capacity and bladder muscle strip contractility were significantly lower, and arterial wall was significantly thicker than in Controls. Significant improvements were seen in bladder capacity, muscle strip contractility and arterial wall thickening in the CBI/BH4 group as compared with the CBI group. Western blot analysis of bladder showed expressions of eNOS (p = 0.043), HIF-1α (p < 0.01) and dihydrofolate reductase (DHFR) (p < 0.01), which could regenerate BH4, were significantly higher in the CBI group than in Controls. In the CBI/BH4 group, HIF-1α (p = 0.012) and DHFR expressions (p = 0.018) were significantly decreased compared with the CBI group. Our results suggest that chronic ischemia increases eNOS and DHFR in the bladder to prevent atherosclerosis progression. However, DHFR could not synthesize sufficient BH4 relative to the increased eNOS, resulting in LUTD. BH4 supplementation protects lower urinary tract function by promoting eNOS activity.


Asunto(s)
Biopterinas/análogos & derivados , Isquemia/prevención & control , Óxido Nítrico/biosíntesis , Vejiga Urinaria/irrigación sanguínea , Animales , Disponibilidad Biológica , Biopterinas/administración & dosificación , Biopterinas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isquemia/etiología , Isquemia/metabolismo , Masculino , Contracción Muscular/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Tetrahidrofolato Deshidrogenasa/metabolismo , Vejiga Urinaria/efectos de los fármacos
10.
Pharmacol Res ; 161: 105123, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32822867

RESUMEN

Breast cancer (BC) is the most common cancer in women and, among different BC subtypes, triple negative (TN) and human epidermal growth factor receptor 2 (HER2)-positive BCs have the worst prognosis. In this study, we investigated the anticancer activity of the root ethanolic and hexane extracts from Lithospermum erythrorhizon, a traditional Chinese herbal medicine known also as tzu ts'ao or tzu-ken, against in vitro and in vivo models of TNBC and HER2-positive BC. Treatment with L. erythrorhizon root extracts resulted in a dose-dependent inhibition of BC cell viability and in a significant reduction of the growth of TNBC cells transplanted in syngeneic mice. Acetylshikonin, a naphthoquinone, was identified as the main bioactive component in extracts and was responsible for the observed antitumor activity, being able to decrease BC cell viability and to interfere with autochthonous mammary carcinogenesis in Δ16HER2 transgenic mice. Acetylshikonin anticancer effect depends on its ability to act as a potent inhibitor of dihydrofolate reductase (DHFR), to down-regulate key mediators governing cancer growth and progression, such as HER2, Src and STAT3, and to induce apoptosis by caspase-3 activation. The accumulation of acetylshikonin in blood samples as well as in brain, kidney, liver and tumor tissues was also investigated by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) highlighting that L. erythrorhizon treatment is effective in delivering the active compound into the target tissues. These results provide evidence that L. erythrorhizon extract and in particular its main component acetylshikonin are effective against aggressive BC subtypes and reveal new acetylshikonin mechanisms of action.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/prevención & control , Antagonistas del Ácido Fólico/farmacología , Lithospermum , Receptor ErbB-2/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo , Animales , Antraquinonas/aislamiento & purificación , Antraquinonas/farmacocinética , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Antagonistas del Ácido Fólico/aislamiento & purificación , Antagonistas del Ácido Fólico/farmacocinética , Humanos , Lithospermum/química , Ratones Transgénicos , Raíces de Plantas , Receptor ErbB-2/genética , Transducción de Señal , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
mSphere ; 5(3)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32581079

RESUMEN

While the folate biosynthetic pathway has provided a rich source of antibacterial, antiprotozoal, and anticancer therapies, it has not yet been exploited to develop uniquely antifungal agents. Although there have been attempts to develop fungal-specific inhibitors of dihydrofolate reductase (DHFR), the protein itself has not been unequivocally validated as essential for fungal growth or virulence. The purpose of this study was to establish dihydrofolate reductase as a valid antifungal target. Using a strain with doxycycline-repressible transcription of DFR1 (PTETO-DFR1 strain), we were able to demonstrate that Dfr1p is essential for growth in vitro Furthermore, nutritional supplements of most forms of folate are not sufficient to restore growth when Dfr1p expression is suppressed or when its activity is directly inhibited by methotrexate, indicating that Candida albicans has a limited capacity to acquire or utilize exogenous sources of folate. Finally, the PTETO-DFR1 strain was rendered avirulent in a mouse model of disseminated candidiasis upon doxycycline treatment. Collectively, these results confirm the validity of targeting dihydrofolate reductase and, by inference, other enzymes in the folate biosynthetic pathway as a strategy to devise new and efficacious therapies to combat life-threatening invasive fungal infections.IMPORTANCE The folate biosynthetic pathway is a promising and understudied source for novel antifungals. Even dihydrofolate reductase (DHFR), a well-characterized and historically important drug target, has not been conclusively validated as an antifungal target. Here, we demonstrate that repression of DHFR inhibits growth of Candida albicans, a major human fungal pathogen. Methotrexate, an antifolate, also inhibits growth but through pH-dependent activity. In addition, we show that C. albicans has a limited ability to take up or utilize exogenous folates as only the addition of high concentrations of folinic acid restored growth in the presence of methotrexate. Finally, we show that repression of DHFR in a mouse model of infection was sufficient to eliminate host mortality. Our work conclusively establishes DHFR as a valid antifungal target in C. albicans.


Asunto(s)
Antifúngicos/farmacología , Candida albicans/efectos de los fármacos , Candida albicans/enzimología , Proteínas Fúngicas/antagonistas & inhibidores , Tetrahidrofolato Deshidrogenasa/metabolismo , Animales , Vías Biosintéticas , Candida albicans/patogenicidad , Candidiasis/tratamiento farmacológico , Desarrollo de Medicamentos/métodos , Femenino , Ácido Fólico/biosíntesis , Antagonistas del Ácido Fólico/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Virulencia
12.
Mol Biochem Parasitol ; 238: 111292, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32505674

RESUMEN

Defining protein-protein interactions is fundamental to the understanding of gene function. Protein-fragment complementation assays have been used for the analysis of protein-protein interactions in various organisms. The split-dihydrofolate reductase (DHFR) protein-fragment complementation assay utilises two complementary fragments of the enzyme fused to a pair of potentially interacting proteins. If these proteins interact, the DHFR fragments associate, fold into their native structure, reconstitute their function and confer resistance to antifolate drugs. We show that murine DHFR fragments fused to interacting proteins reconstitute a functional enzyme and confer resistance to the antifolate drug WR99210 in Plasmodium falciparum. These data demonstrate that the split-DHFR method can be used to detect in vivo protein-protein interactions in the parasite. Additionally, we show that split-DHFR fragments can be used as selection markers, permitting simultaneous selection of two plasmids in the presence of a single antifolate drug. Taken together, these experiments show that split-DHFR represents a valuable tool for the characterisation of Plasmodium protein function and genetic manipulation of the parasite.


Asunto(s)
Plasmodium falciparum/genética , Mapeo de Interacción de Proteínas/métodos , Proteínas Protozoarias/genética , Tetrahidrofolato Deshidrogenasa/genética , Transfección/métodos , Antimaláricos/farmacología , Bioensayo , Eritrocitos/parasitología , Antagonistas del Ácido Fólico/farmacología , Expresión Génica , Genes Reporteros , Prueba de Complementación Genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Unión Proteica , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo , Triazinas/farmacología , Proteína Fluorescente Roja
13.
Anal Bioanal Chem ; 412(17): 4037-4043, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32328689

RESUMEN

A convenient analytical system for protein-ligand interactions under crude conditions was developed using native mass spectrometry (MS). As a model protein, Escherichia coli (E. coli) dihydrofolate reductase (DHFR) with and without a histidine tag was used for the study. First, overexpressed DHFR with a His-tag was roughly purified with a Ni-sepharose resin and subjected to native mass spectrometry with or without incubation with an inhibitor, Methotrexate (MTX). Even only with the minimum cleanup by the Ni-sepharose resin, intact ions of DHFR-nicotinamide adenine dinucleotide phosphate (NADPH) and DHFR-NADPH-ligand complexes were successfully observed. By optimizing the preparation procedures of the crude sample for native MS, e.g., avoiding sonication for cell lysis, we successfully observed intact ions of the specific DHFR-NADPH-MTX ternary complex starting with cultivation of E. coli in ≤ 25 mL medium. When the crude DHFR sample was mixed with two, four, or eight candidate compounds, only ions of the specific protein-ligand complex were observed. This indicates that the present system can be used as a rapid and convenient method for the rough determination of binding of specific ligands to the target protein without the time-consuming purification of protein samples. Moreover, it is important to rapidly determine specific interactions with target proteins under conditions similar to those in "real" biological systems. Graphical abstract.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Antagonistas del Ácido Fólico/farmacología , Metotrexato/farmacología , Tetrahidrofolato Deshidrogenasa/metabolismo , Sitios de Unión , Evaluación Preclínica de Medicamentos/métodos , Escherichia coli/química , Proteínas de Escherichia coli/química , NADP/química , NADP/metabolismo , Unión Proteica , Espectrometría de Masa por Ionización de Electrospray/métodos , Tetrahidrofolato Deshidrogenasa/química
14.
Nat Rev Rheumatol ; 16(3): 145-154, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32066940

RESUMEN

Despite the introduction of numerous biologic agents for the treatment of rheumatoid arthritis (RA) and other forms of inflammatory arthritis, low-dose methotrexate therapy remains the gold standard in RA therapy. Methotrexate is generally the first-line drug for the treatment of RA, psoriatic arthritis and other forms of inflammatory arthritis, and it enhances the effect of most biologic agents in RA. Understanding the mechanism of action of methotrexate could be instructive in the appropriate use of the drug and in the design of new regimens for the treatment of RA. Although methotrexate is one of the first examples of intelligent drug design, multiple mechanisms potentially contribute to the anti-inflammatory actions of methotrexate, including the inhibition of purine and pyrimidine synthesis, transmethylation reactions, translocation of nuclear factor-κB (NF-κB) to the nucleus, signalling via the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway and nitric oxide production, as well as the promotion of adenosine release and expression of certain long non-coding RNAs.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Artritis/tratamiento farmacológico , Inmunidad Celular/efectos de los fármacos , Metotrexato/uso terapéutico , Ribonucleótidos/antagonistas & inhibidores , Linfocitos T/inmunología , Tetrahidrofolato Deshidrogenasa/efectos de los fármacos , Aminoimidazol Carboxamida/antagonistas & inhibidores , Antirreumáticos/uso terapéutico , Artritis/inmunología , Artritis/metabolismo , Humanos , Linfocitos T/efectos de los fármacos , Tetrahidrofolato Deshidrogenasa/metabolismo
15.
Elife ; 82019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31573512

RESUMEN

The mechanisms of adaptation to inactivation of essential genes remain unknown. Here we inactivate E. coli dihydrofolate reductase (DHFR) by introducing D27G,N,F chromosomal mutations in a key catalytic residue with subsequent adaptation by an automated serial transfer protocol. The partial reversal G27- > C occurred in three evolutionary trajectories. Conversely, in one trajectory for D27G and in all trajectories for D27F,N strains adapted to grow at very low metabolic supplement (folAmix) concentrations but did not escape entirely from supplement auxotrophy. Major global shifts in metabolome and proteome occurred upon DHFR inactivation, which were partially reversed in adapted strains. Loss-of-function mutations in two genes, thyA and deoB, ensured adaptation to low folAmix by rerouting the 2-Deoxy-D-ribose-phosphate metabolism from glycolysis towards synthesis of dTMP. Multiple evolutionary pathways of adaptation converged to a suboptimal solution due to the high accessibility to loss-of-function mutations that block the path to the highest, yet least accessible, fitness peak.


Asunto(s)
Adaptación Biológica , Escherichia coli/enzimología , Escherichia coli/crecimiento & desarrollo , Genes Esenciales , Tetrahidrofolato Deshidrogenasa/deficiencia , Escherichia coli/genética , Evolución Molecular , Metaboloma , Mutación Missense , Proteoma , Pase Seriado , Tetrahidrofolato Deshidrogenasa/genética
16.
J Enzyme Inhib Med Chem ; 34(1): 1439-1450, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31409157

RESUMEN

Leishmaniasis is a tropical disease found in more than 90 countries. The drugs available to treat this disease have nonspecific action and high toxicity. In order to develop novel therapeutic alternatives to fight this ailment, pteridine reductase 1 (PTR1) and dihydrofolate reductase-thymidylate synthase (DHF-TS) have been targeted, once Leishmania is auxotrophic for folates. Although PTR1 and DHFR-TS from other protozoan parasites have been studied, their homologs in Leishmania chagasi have been poorly characterized. Hence, this work describes the optimal conditions to express the recombinant LcPTR1 and LcDHFR-TS enzymes, as well as balanced assay conditions for screening. Last but not the least, we show that 2,4 diaminopyrimidine derivatives are low-micromolar competitive inhibitors of both enzymes (LcPTR1 Ki = 1.50-2.30 µM and LcDHFR Ki = 0.28-3.00 µM) with poor selectivity index. On the other hand, compound 5 (2,4-diaminoquinazoline derivative) is a selective LcPTR1 inhibitor (Ki = 0.47 µM, selectivity index = 20).


Asunto(s)
Inhibidores Enzimáticos/farmacología , Leishmania infantum/enzimología , Complejos Multienzimáticos/antagonistas & inhibidores , Oxidorreductasas/antagonistas & inhibidores , Timidilato Sintasa/antagonistas & inhibidores , Catálisis , Cromatografía de Afinidad , Clonación Molecular , Evaluación Preclínica de Medicamentos , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Concentración 50 Inhibidora , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/aislamiento & purificación , Complejos Multienzimáticos/metabolismo , Oxidorreductasas/genética , Oxidorreductasas/aislamiento & purificación , Oxidorreductasas/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/aislamiento & purificación , Tetrahidrofolato Deshidrogenasa/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/aislamiento & purificación , Timidilato Sintasa/metabolismo
17.
Bioorg Med Chem Lett ; 29(11): 1413-1418, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30929953

RESUMEN

Protozoans of the genus Cryptosporidium are the causative agent of the gastrointestinal disease, cryptosporidiosis, which can be fatal in immunocompromised individuals. Cryptosporidium hominis (C. hominis) bifunctional thymidylate synthase-dihydrofolate reductase (TS-DHFR) is an essential enzyme in the folate biosynthesis pathway and a molecular target for inhibitor design. Previous studies have demonstrated the importance of the ChTS-DHFR linker region "crossover helix" to the enzymatic activity and stability of the ChDHFR domain. We conducted a virtual screen of a novel non-active site pocket located at the interface of the ChDHFR domain and crossover helix. From this screen we have identified and characterized a noncompetitive inhibitor, compound 15, a substituted diphenyl thiourea. Through subsequent structure activity relationship studies, we have identified a time-dependent inhibitor lead, compound 15D17, a thiol-substituted 2-hydroxy-N-phenylbenzamide, which is selective for ChTS-DHFR, and whose effects appear to be mediated by covalent bond formation with a non-catalytic cysteine residue adjacent to the non-active site pocket.


Asunto(s)
Benzamidas/farmacología , Cryptosporidium/enzimología , Inhibidores Enzimáticos/farmacología , Complejos Multienzimáticos/antagonistas & inhibidores , Tiourea/farmacología , Timidilato Sintasa/antagonistas & inhibidores , Regulación Alostérica/efectos de los fármacos , Benzamidas/química , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Complejos Multienzimáticos/metabolismo , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/metabolismo , Tiourea/química , Timidilato Sintasa/metabolismo
18.
Molecules ; 24(6)2019 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-30909399

RESUMEN

Dihydrofolate reductase inhibitors are an important class of drugs, as evidenced by their use as antibacterial, antimalarial, antifungal, and anticancer agents. Progress in understanding the biochemical basis of mechanisms responsible for enzyme selectivity and antiproliferative effects has renewed the interest in antifolates for cancer chemotherapy and prompted the medicinal chemistry community to develop novel and selective human DHFR inhibitors, thus leading to a new generation of DHFR inhibitors. This work summarizes the mechanism of action, chemical, and anticancer profile of the DHFR inhibitors discovered in the last six years. New strategies in DHFR drug discovery are also provided, in order to thoroughly delineate the current landscape for medicinal chemists interested in furthering this study in the anticancer field.


Asunto(s)
Antineoplásicos/farmacología , Antagonistas del Ácido Fólico/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo
19.
J Biomol Struct Dyn ; 37(16): 4181-4199, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30648473

RESUMEN

Plasmodium falciparum dihydrofolate reductase enzyme (PfDHFR) is counted as one of the attractive and validated antimalarial drug targets. However, the point mutations in the active site of wild-type PfDHFR have developed resistance against the well-known antifolates. Therefore, there is a dire need for the development of inhibitors that can inhibit both wild-type and mutant-type DHFR enzyme. In the present contribution, we have constructed the common feature pharmacophore models from the available PfDHFR. A representative hypothesis was prioritized and then employed for the screening of natural product library to search for the molecules with complementary features responsible for the inhibition. The screened candidates were processed via drug-likeness filters and molecular docking studies. The docking was carried out on the wild-type PfDHFR (3QGT); double-mutant PfDHFR (3UM5 and 1J3J) and quadruple-mutant PfDHFR (1J3K) enzymes. A total of eight common hits were obtained from the docking calculations that could be the potential inhibitors for both wild and mutant type DHFR enzymes. Eventually, the stability of these candidates with the selected proteins was evaluated via molecular dynamics simulations. Except for SPECS14, all the prioritized candidates were found to be stable throughout the simulation run. Overall, the strategy employed in the present work resulted in the retrieval of seven candidates that may show inhibitory activity against PfDHFR and could be further exploited as a scaffold to develop novel antimalarials. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Antimaláricos/química , Antagonistas del Ácido Fólico/química , Malaria Falciparum/tratamiento farmacológico , Proteínas Protozoarias/ultraestructura , Tetrahidrofolato Deshidrogenasa/ultraestructura , Animales , Antimaláricos/uso terapéutico , Dominio Catalítico/efectos de los fármacos , Antagonistas del Ácido Fólico/uso terapéutico , Humanos , Malaria Falciparum/parasitología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/química , Tetrahidrofolato Deshidrogenasa/química
20.
Toxicol Lett ; 301: 146-156, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30502384

RESUMEN

Methotrexate (MTX) is a folic acid antagonist and widely used for acute lymphoblastic leukemia (ALL) in children. MTX is associated with acute and chronic neurotoxicity during treatment, however the underlying mechanism is still poorly understood. In this study we investigate whether MTX is neurovirulent to astrocytes in the Central Nervous System (CNS) of adolescent mice. We demonstrated that MTX induced severe cytotoxicity in C6 astrocyte-like cell line and rat primary cultures of astrocytes in a dose-dependent manner. Moreover, GFAP-labeled astrocyte cells significantly decreased in the mouse spinal cord and brain. Furthermore, protein levels of PARP and pro-Caspase-3 were reduced by MTX, indicating MTX-induced apoptosis leads to the astrocytes loss. Notably, overexpression of dihydrofolate reductase (DHFR) or exogenous addition of folate markedly reversed the astrocytes toxicity induced by MTX through activating folate metabolism pathway. Taken together, our study provides evidence for neurotoxic effect of MTX-induced astrocytes apoptosis both in vitro and in vivo with disruption of folate metabolism, and additional supplement of folate may provide novel approaches for alleviating the astrocytes toxicity induced by MTX in the clinic.


Asunto(s)
Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Ácido Fólico/metabolismo , Metotrexato/toxicidad , Animales , Astrocitos/citología , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Sistema Nervioso Central/metabolismo , Relación Dosis-Respuesta a Droga , Antagonistas del Ácido Fólico/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Ratones , Ratones Endogámicos C57BL , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Ratas , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA