Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 369
Filtrar
1.
Nature ; 609(7929): 1056-1062, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36071163

RESUMEN

Folates are essential nutrients with important roles as cofactors in one-carbon transfer reactions, being heavily utilized in the synthesis of nucleic acids and the metabolism of amino acids during cell division1,2. Mammals lack de novo folate synthesis pathways and thus rely on folate uptake from the extracellular milieu3. The human reduced folate carrier (hRFC, also known as SLC19A1) is the major importer of folates into the cell1,3, as well as chemotherapeutic agents such as methotrexate4-6. As an anion exchanger, RFC couples the import of folates and antifolates to anion export across the cell membrane and it is a major determinant in methotrexate (antifolate) sensitivity, as genetic variants and its depletion result in drug resistance4-8. Despite its importance, the molecular basis of substrate specificity by hRFC remains unclear. Here we present cryo-electron microscopy structures of hRFC in the apo state and captured in complex with methotrexate. Combined with molecular dynamics simulations and functional experiments, our study uncovers key determinants of hRFC transport selectivity among folates and antifolate drugs while shedding light on important features of anion recognition by hRFC.


Asunto(s)
Microscopía por Crioelectrón , Antagonistas del Ácido Fólico , Metotrexato , Proteína Portadora de Folato Reducido , Aniones/metabolismo , Apoproteínas/genética , Apoproteínas/metabolismo , Transporte Biológico , Carbono/metabolismo , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Humanos , Metotrexato/química , Metotrexato/metabolismo , Simulación de Dinámica Molecular , Proteína Portadora de Folato Reducido/genética , Proteína Portadora de Folato Reducido/metabolismo , Proteína Portadora de Folato Reducido/ultraestructura , Especificidad por Sustrato
2.
Nature ; 595(7865): 130-134, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34040256

RESUMEN

Folates (also known as vitamin B9) have a critical role in cellular metabolism as the starting point in the synthesis of nucleic acids, amino acids and the universal methylating agent S-adenylsmethionine1,2. Folate deficiency is associated with a number of developmental, immune and neurological disorders3-5. Mammals cannot synthesize folates de novo; several systems have therefore evolved to take up folates from the diet and distribute them within the body3,6. The proton-coupled folate transporter (PCFT) (also known as SLC46A1) mediates folate uptake across the intestinal brush border membrane and the choroid plexus4,7, and is an important route for the delivery of antifolate drugs in cancer chemotherapy8-10. How PCFT recognizes folates or antifolate agents is currently unclear. Here we present cryo-electron microscopy structures of PCFT in a substrate-free state and in complex with a new-generation antifolate drug (pemetrexed). Our results provide a structural basis for understanding antifolate recognition and provide insights into the pH-regulated mechanism of folate transport mediated by PCFT.


Asunto(s)
Microscopía por Crioelectrón , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Pemetrexed/química , Pemetrexed/metabolismo , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Apoproteínas/ultraestructura , Transporte Biológico , Humanos , Modelos Moleculares , Transportador de Folato Acoplado a Protón/ultraestructura , Protones
3.
FASEB J ; 36(2): e22164, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35061292

RESUMEN

The human proton-coupled folate transporter (PCFT; SLC46A1) or hPCFT was identified in 2006 as the principal folate transporter involved in the intestinal absorption of dietary folates. A rare autosomal recessive hereditary folate malabsorption syndrome is attributable to human SLC46A1 variants. The recognition that hPCFT was highly expressed in many tumors stimulated substantial interest in its potential for cytotoxic drug targeting, taking advantage of its high-level transport activity under acidic pH conditions that characterize many tumors and its modest expression in most normal tissues. To better understand the basis for variations in hPCFT levels between tissues including human tumors, studies have examined the transcriptional regulation of hPCFT including the roles of CpG hypermethylation and critical transcription factors and cis elements. Additional focus involved identifying key structural and functional determinants of hPCFT transport that, combined with homology models based on structural homologies to the bacterial transporters GlpT and LacY, have enabled new structural and mechanistic insights. Recently, cryo-electron microscopy structures of chicken PCFT in a substrate-free state and in complex with the antifolate pemetrexed were reported, providing further structural insights into determinants of (anti)folate recognition and the mechanism of pH-regulated (anti)folate transport by PCFT. Like many major facilitator proteins, hPCFT exists as a homo-oligomer, and evidence suggests that homo-oligomerization of hPCFT monomeric proteins may be important for its intracellular trafficking and/or transport function. Better understanding of the structure, function and regulation of hPCFT should facilitate the rational development of new therapeutic strategies for conditions associated with folate deficiency, as well as cancer.


Asunto(s)
Ácido Fólico/metabolismo , Transportador de Folato Acoplado a Protón/metabolismo , Animales , Antagonistas del Ácido Fólico/metabolismo , Humanos , Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología
4.
Bioorg Med Chem ; 28(15): 115600, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32631571

RESUMEN

The enzyme dihydrofolate reductase from M.tuberculosis (MtDHFR) has a high unexploited potential to be a target for new drugs against tuberculosis (TB), due to its importance for pathogen survival. Preliminary studies have obtained fragment-like molecules with low affinity to MtDHFR which can potentially become lead compounds. Taking this into account, the fragment MB872 was used as a prototype for analogue development by bioisosterism/retro-bioisosterism, which resulted in 20 new substituted 3-benzoic acid derivatives. Compounds were active against MtDHFR, with IC50 values ranging from 7 to 40 µM, where compound 4e not only had the best inhibitory activity (IC50 = 7 µM), but also was 71-fold more active than the original fragment MB872. The 4e inhibition kinetics indicated an uncompetitive mechanism, which was supported by molecular modeling which suggested that the compounds can access an independent backpocket from the substrate and competitive inhibitors. Thus, based on these results, substituted 3-benzoic acid derivatives have strong potential to be developed as novel MtDHFR inhibitors and also anti-TB agents.


Asunto(s)
Antituberculosos/farmacología , Proteínas Bacterianas/metabolismo , Benzoatos/farmacología , Antagonistas del Ácido Fólico/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Tetrahidrofolato Deshidrogenasa/metabolismo , Antituberculosos/síntesis química , Antituberculosos/metabolismo , Proteínas Bacterianas/química , Benzoatos/síntesis química , Benzoatos/metabolismo , Dominio Catalítico , Diseño de Fármacos , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/metabolismo , Cinética , Simulación de Dinámica Molecular , Estructura Molecular , Unión Proteica , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/química
5.
Bioorg Med Chem ; 28(12): 115544, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32503687

RESUMEN

Tumor-targeted 6-substituted pyrrolo[2,3-d]pyrimidine benzoyl compounds based on 2 were isosterically modified at the 4-carbon bridge by replacing the vicinal (C11) carbon by heteroatoms N (4), O (5) or S (6), or with an N-substituted formyl (7), trifluoroacetyl (8) or acetyl (9). Replacement with sulfur (6) afforded the most potent KB tumor cell inhibitor, ~6-fold better than the parent 2. In addition, 6 retained tumor transport selectivity via folate receptor (FR) α and -ß over the ubiquitous reduced folate carrier (RFC). FRα-mediated cell inhibition for 6 was generally equivalent to 2, while the FRß-mediated activity was improved by 16-fold over 2. N (4) and O (5) substitutions afforded similar tumor cell inhibitions as 2, with selectivity for FRα and -ß over RFC. The N-substituted analogs 7-9 also preserved transport selectivity for FRα and -ß over RFC. For FRα-expressing CHO cells, potencies were in the order of 8 > 7 > 9. Whereas 8 and 9 showed similar results with FRß-expressing CHO cells, 7 was ~16-fold more active than 2. By nucleoside rescue experiments, all the compounds inhibited de novo purine biosynthesis, likely at the step catalyzed by glycinamide ribonucleotide formyltransferase. Thus, heteroatom replacements of the CH2 in the bridge of 2 afford analogs with increased tumor cell inhibition that could provide advantages over 2, as well as tumor transport selectivity over clinically used antifolates including methotrexate and pemetrexed.


Asunto(s)
Antineoplásicos/síntesis química , Diseño de Fármacos , Receptor 1 de Folato/metabolismo , Receptor 2 de Folato/metabolismo , Ácido Fólico/metabolismo , Pirimidinas/química , Pirroles/química , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Sitios de Unión , Células CHO , Dominio Catalítico , Línea Celular Tumoral , Cricetinae , Cricetulus , Receptor 1 de Folato/química , Receptor 1 de Folato/genética , Receptor 2 de Folato/química , Receptor 2 de Folato/genética , Ácido Fólico/química , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacología , Humanos , Simulación del Acoplamiento Molecular , Fosforribosilglicinamida-Formiltransferasa/química , Fosforribosilglicinamida-Formiltransferasa/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacología , Pirroles/metabolismo , Pirroles/farmacología , Relación Estructura-Actividad
6.
Molecules ; 25(15)2020 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-32752079

RESUMEN

Drug resistance is a core issue in cancer chemotherapy. A known folate antagonist, methotrexate (MTX) inhibits human dihydrofolate reductase (hDHFR), the enzyme responsible for the catalysis of 7,8-dihydrofolate reduction to 5,6,7,8-tetrahydrofolate, in biosynthesis and cell proliferation. Structural change in the DHFR enzyme is a significant cause of resistance and the subsequent loss of MTX. In the current study, wild type hDHFR and double mutant (engineered variant) F31R/Q35E (PDB ID: 3EIG) were subject to computational study. Structure-based pharmacophore modeling was carried out for wild type (WT) and mutant (MT) (variant F31R/Q35E) hDHFR structures by generating ten models for each. Two pharmacophore models, WT-pharma and MT-pharma, were selected for further computations, and showed excellent ROC curve quality. Additionally, the selected pharmacophore models were validated by the Guner-Henry decoy test method, which yielded high goodness of fit for WT-hDHFR and MT-hDHFR. Using a SMILES string of MTX in ZINC15 with the selections of 'clean', in vitro and in vivo options, 32 MTX-analogs were obtained. Eight analogs were filtered out due to their drug-like properties by applying absorption, distribution, metabolism, excretion, and toxicity (ADMET) assessment tests and Lipinski's Rule of five. WT-pharma and MT-pharma were further employed as a 3D query in virtual screening with drug-like MTX analogs. Subsequently, seven screening hits along with a reference compound (MTX) were subjected to molecular docking in the active site of WT- and MT-hDHFR. Through a clustering analysis and examination of protein-ligand interactions, one compound was found with a ChemPLP fitness score greater than that of MTX (reference compound). Finally, a simulation of molecular dynamics (MD) identified an MTX analog which exhibited strong affinity for WT- and MT-hDHFR, with stable RMSD, hydrogen bonds (H-bonds) in the binding site and the lowest MM/PBSA binding free energy. In conclusion, we report on an MTX analog which is capable of inhibiting hDHFR in wild type form, as well as in cases where the enzyme acquires resistance to drugs during chemotherapy treatment.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Metotrexato/farmacología , Tetrahidrofolato Deshidrogenasa/metabolismo , Área Bajo la Curva , Sitios de Unión , Dominio Catalítico , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacología , Antagonistas del Ácido Fólico/uso terapéutico , Humanos , Enlace de Hidrógeno , Ligandos , Metotrexato/análogos & derivados , Metotrexato/metabolismo , Metotrexato/uso terapéutico , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Curva ROC , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/genética , Termodinámica
7.
Mol Pharm ; 16(7): 3133-3144, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31198046

RESUMEN

The clinical translation of methotrexate (MTX) is limited because of low aqueous solubility, poor bioavailability, low uptake efficiency, and toxicity concerns. Herein, dual-acting MTX (not only targeting folate receptors but also killing cells via inhibition of intracellular folate metabolism) and hyaluronic acid (HA, targeting CD44 receptors) were selected to be covalently linked by the redox-responsive disulfide bond. The synthesized prodrug (HA-SS-MTX) as a molecular structural motif could self-assemble into simple yet multifunctional nanoparticles (HA-SS-MTX NPs) in aqueous solution. The HA-SS-MTX NPs displayed an average diameter of ∼110 nm with a uniformly spherical shape and maintained stability in different physiological media. Moreover, the HA-SS-MTX NPs could exhibit a sharp redox-dependent response for rapid structure disassembly and sequential MTX release compared to the redox-irresponsive group (HA-ADH-MTX NPs). Furthermore, the results of confocal microscopy and flow cytometry verified that the nanosystems were selectively uptaken by cancer cells via folate and CD44 receptor-mediated internalization through the dual-active targeting mechanism. In addition, HA-SS-MTX NPs could accumulate within tumor sites for a longer period. Notably, in vitro and in vivo antitumor results demonstrated that HA-SS-MTX NPs significantly promoted the death of cancer cells and enhanced the inhibition of tumor growth while reducing the toxicity as compared to MTX and HA-ADH-MTX NPs. Therefore, the smart HA-SS-MTX NPs as the simple and efficient platform have great potential in tumor-targeting drug delivery and therapy.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Antagonistas del Ácido Fólico/metabolismo , Ácido Hialurónico/metabolismo , Metotrexato/metabolismo , Nanopartículas/química , Profármacos/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Supervivencia Celular/efectos de los fármacos , Disulfuros/química , Liberación de Fármacos , Femenino , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/farmacología , Células HeLa , Humanos , Receptores de Hialuranos/antagonistas & inhibidores , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Metotrexato/química , Metotrexato/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oxidación-Reducción , Distribución Tisular , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Bioorg Chem ; 88: 102923, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30991189

RESUMEN

A new series of 6-substituted amido, azo or thioureido-quinazolin-4(3H)-one was synthesized and tested for their in-vitro antitumor activity. Compounds 21, 53 and 60 showed broad spectrum antitumor activity with average IC50 values of 6.7, 7.6 and 9.1 µM, respectively compared with methotrexate (1, IC50 19.26 µM). As an attempt to reveal the mechanism of the antitumor potency, cell cycle analysis and DHFR inhibition were performed. Compounds 59 and 61 induced their cytotoxicity in Hela (IC50 10.6 µM) and HCT-116 (IC50 15.5 µM) cell lines, respectively through Pre-G1 apoptosis, inhibiting cell growth at G2-M phase. Compounds 29, 33, 59 and 61 showed DHFR inhibitory potency at IC50 0.2, 0.2, 0.3 and 0.3 µM, respectively. The active DHFR inhibitors showed high affinity binding toward the amino acid residues Thr56, Ser59 and Ser118. The active compounds obeyed Lipinski's rule of five and could be used as template model for further optimization.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Azo/farmacología , Quinazolinonas/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Compuestos Azo/síntesis química , Compuestos Azo/metabolismo , Compuestos Azo/farmacocinética , Dominio Catalítico , Bovinos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Pruebas de Enzimas , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacocinética , Antagonistas del Ácido Fólico/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Quinazolinonas/síntesis química , Quinazolinonas/metabolismo , Quinazolinonas/farmacocinética , Relación Estructura-Actividad , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo , Tiourea/metabolismo , Tiourea/farmacocinética
9.
Biochemistry ; 57(49): 6780-6786, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30452231

RESUMEN

The folate antagonist methotrexate is a cytotoxic drug used in the treatment of several cancer types. The entry of methotrexate into the cell is mediated by two main transport systems: the reduced folate carrier and membrane-associated folate receptors. These transporters differ considerably in their mechanism of (anti)folate uptake, substrate specificity, and tissue specificity. Although the mechanism of action of the reduced folate carrier is fairly well-established, that of the folate receptor has remained unknown. The development of specific folate receptor-targeted antifolates would be accelerated if additional mechanistic data were to become available. In this work, we used two fluorescently labeled conjugates of methotrexate, differently linked at the terminal groups, to clarify the uptake mechanism by folate receptor-α. The results demonstrate the importance of methotrexate amino groups in the interaction with folate receptor-α.


Asunto(s)
Receptor 1 de Folato/metabolismo , Antagonistas del Ácido Fólico/metabolismo , Metotrexato/análogos & derivados , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/metabolismo , Transporte Biológico Activo , Línea Celular Tumoral , Endocitosis , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Receptor 1 de Folato/química , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/química , Humanos , Metotrexato/química , Metotrexato/metabolismo , Modelos Biológicos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteína Portadora de Folato Reducido/química , Proteína Portadora de Folato Reducido/metabolismo
10.
J Biol Chem ; 292(32): 13449-13458, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28634233

RESUMEN

Thymidylate synthase (TS) is the sole enzyme responsible for de novo biosynthesis of thymidylate (TMP) and is essential for cell proliferation and survival. Inhibition of human TS (hTS) has been extensively investigated for cancer chemotherapy, but several aspects of its activity and regulation are still uncertain. In this study, we performed comprehensive structural and biophysical studies of hTS using crystallography and thermal shift assay and provided the first detailed structural information on the conformational changes induced by ligand binding to the hTS active site. We found that upon binding of the antifolate agents raltitrexed and nolatrexed, the two insert regions in hTS, the functions of which are unclear, undergo positional shifts toward the catalytic center. We investigated the inactive conformation of hTS and found that the two insert regions are also involved in the conformational transition between the active and inactive state of hTS. Moreover, we identified a ligand-binding site in the dimer interface, suggesting that the cavity in the dimer interface could serve as an allosteric site of hTS to regulate the conformational switching between the active and inactive states. On the basis of these findings, we propose a regulatory mechanism of hTS activity that involves allosteric regulation of interactions of hTS with its own mRNA depending on cellular demands for TMP.


Asunto(s)
Modelos Moleculares , Timidilato Sintasa/metabolismo , Sitio Alostérico/efectos de los fármacos , Sustitución de Aminoácidos , Sitios de Unión , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Nucleótidos de Desoxiuracil/química , Nucleótidos de Desoxiuracil/metabolismo , Dimerización , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacología , Humanos , Ligandos , Mutagénesis Sitio-Dirigida , Mutación , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Quinazolinas/química , Quinazolinas/metabolismo , Quinazolinas/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tiofenos/química , Tiofenos/metabolismo , Tiofenos/farmacología , Timidilato Sintasa/antagonistas & inhibidores , Timidilato Sintasa/química , Timidilato Sintasa/genética
11.
Bioorg Med Chem ; 26(9): 2640-2650, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29691153

RESUMEN

To combine the potency of trimetrexate (TMQ) or piritrexim (PTX) with the species selectivity of trimethoprim (TMP), target based design was carried out with the X-ray crystal structure of human dihydrofolate reductase (hDHFR) and the homology model of Pneumocystis jirovecii DHFR (pjDHFR). Using variation of amino acids such as Met33/Phe31 (in pjDHFR/hDHFR) that affect the binding of inhibitors due to their distinct positive or negative steric effect at the active binding site of the inhibitor, we designed a series of substituted-pyrrolo[2,3-d]pyrimidines. The best analogs displayed better potency (IC50) than PTX and high selectivity for pjDHFR versus hDHFR, with 4 exhibiting a selectivity for pjDHFR of 24-fold.


Asunto(s)
Aminoácidos/química , Antibacterianos/farmacología , Antagonistas del Ácido Fólico/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Antibacterianos/síntesis química , Dominio Catalítico , Cristalografía por Rayos X , Diseño de Fármacos , Pruebas de Enzimas , Antagonistas del Ácido Fólico/síntesis química , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Simulación del Acoplamiento Molecular , Estructura Molecular , Pneumocystis carinii/enzimología , Unión Proteica , Pirimidinas/síntesis química , Pirimidinas/química , Pirimidinas/metabolismo , Pirroles/síntesis química , Pirroles/química , Pirroles/metabolismo , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo
12.
Phys Chem Chem Phys ; 19(18): 11416-11428, 2017 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-28422217

RESUMEN

Dihydrofolate reductase (DHFR) is a ubiquitous enzyme with an essential role in cell metabolism. DHFR catalyzes the reduction of dihydrofolate to tetrahydrofolate, which is a precursor for purine and thymidylate synthesis. Several DHFR targeting antifolate drugs including trimethoprim, a competitive antibacterial inhibitor, have therefore been developed and are clinically used. Evolution of resistance against antifolates is a common public health problem rendering these drugs ineffective. To combat the resistance problem, it is important to understand resistance-conferring changes in the DHFR structure and accordingly develop alternative strategies. Here, we structurally and dynamically characterize Escherichia coli DHFR in its wild type (WT) and trimethoprim resistant L28R mutant forms in the presence of the substrate and its inhibitor trimethoprim. We use molecular dynamics simulations to determine the conformational space, loop dynamics and hydrogen bond distributions at the active site of DHFR for the WT and the L28R mutant. We also report their experimental kcat, Km, and Ki values, accompanied by isothermal titration calorimetry measurements of DHFR that distinguish enthalpic and entropic contributions to trimethoprim binding. Although mutations that confer resistance to competitive inhibitors typically make enzymes more promiscuous and decrease affinity to both the substrate and the inhibitor, strikingly, we find that the L28R mutant has a unique resistance mechanism. While the binding affinity differences between the WT and the mutant for the inhibitor and the substrate are small, the newly formed extra hydrogen bonds with the aminobenzoyl glutamate tail of DHF in the L28R mutant leads to increased barriers for the dissociation of the substrate and the product. Therefore, the L28R mutant indirectly gains resistance by enjoying prolonged binding times in the enzyme-substrate complex. While this also leads to slower product release and decreases the catalytic rate of the L28R mutant, the overall effect is the maintenance of a sufficient product formation rate. Finally, the experimental and computational analyses together reveal the changes that occur in the energetic landscape of DHFR upon the resistance-conferring L28R mutation. We show that the negative entropy associated with the binding of trimethoprim in WT DHFR is due to water organization at the binding interface. Our study lays the framework to study structural changes in other trimethoprim resistant DHFR mutants.


Asunto(s)
Farmacorresistencia Bacteriana , Escherichia coli/enzimología , Antagonistas del Ácido Fólico/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo , Trimetoprim/metabolismo , Dominio Catalítico/genética , Farmacorresistencia Bacteriana/genética , Antagonistas del Ácido Fólico/química , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Mutación Puntual , Unión Proteica , Conformación Proteica , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/genética , Trimetoprim/química
13.
Biochemistry ; 55(7): 1107-19, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26848874

RESUMEN

Mycobacterium tuberculosis (Mtb) Rv2671 is annotated as a 5-amino-6-ribitylamino-2,4(1H,3H)-pyrimidinedione 5'-phosphate (AROPP) reductase (RibD) in the riboflavin biosynthetic pathway. Recently, a strain of Mtb with a mutation in the 5' untranslated region of Rv2671, which resulted in its overexpression, was found to be resistant to dihydrofolate reductase (DHFR) inhibitors including the anti-Mtb drug para-aminosalicylic acid (PAS). In this study, a biochemical analysis of Rv2671 showed that it was able to catalyze the reduction of dihydrofolate (DHF) to tetrahydrofolate (THF), which explained why the overexpression of Rv2671 was sufficient to confer PAS resistance. We solved the structure of Rv2671 in complex with the NADP(+) and tetrahydrofolate (THF), which revealed the structural basis for the DHFR activity. The structures of Rv2671 complexed with two DHFR inhibitors, trimethoprim and trimetrexate, provided additional details of the substrate binding pocket and elucidated the differences between their inhibitory activities. Finally, Rv2671 was unable to catalyze the reduction of AROPP, which indicated that Rv2671 and its closely related orthologues are not involved in riboflavin biosynthesis.


Asunto(s)
Proteínas Bacterianas/química , Modelos Moleculares , Mycobacterium tuberculosis/enzimología , NADP/química , Nucleótido Desaminasas/química , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolatos/química , Ácido Aminosalicílico/farmacología , Antituberculosos/química , Antituberculosos/metabolismo , Antituberculosos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Farmacorresistencia Bacteriana , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacología , Cinética , Ligandos , Conformación Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , NADP/metabolismo , Nucleótido Desaminasas/antagonistas & inhibidores , Nucleótido Desaminasas/genética , Nucleótido Desaminasas/metabolismo , Filogenia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Tetrahidrofolatos/metabolismo , Trimetoprim/química , Trimetoprim/metabolismo , Trimetoprim/farmacología , Trimetrexato/química , Trimetrexato/metabolismo , Trimetrexato/farmacología
14.
Mol Pharmacol ; 89(4): 425-34, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26837243

RESUMEN

Pemetrexed (PMX) is a 5-substituted pyrrolo[2,3-d]pyrimidine antifolate used for therapy of nonsquamous nonsmall cell lung cancer (NS-NSCLC). PMX is transported by the reduced folate carrier (RFC) and proton-coupled folate transporter (PCFT). Unlike RFC, PCFT is active at acidic pH levels characterizing the tumor microenvironment. By real-time reverse-transcription polymerase chain reaction (RT-PCR) and immunohistochemistry, PCFT transcripts and proteins were detected in primary NS-NSCLC specimens. In six NS-NSCLC cell lines (A549, H1437, H460, H1299, H1650, and H2030), PCFT transcripts and proteins were detected by real-time RT-PCR and western blots, respectively. 6-Substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates related to PMX [compound 1 (C1) and compound 2 (C2), respectively] are selective substrates for PCFT over RFC. In the NS-NSCLC cell lines, both [(3)H]PMX and [(3)H]C2 were transported by PCFT. C1 and C2 inhibited proliferation of the NS-NSCLC cell lines; A549, H460, and H2030 cells were more sensitive to C1 than to PMX. C1 and C2 inhibited glycinamide ribonucleotide formyltransferase in de novo purine nucleotide biosynthesis. When treated at pH 6.8, which favors PCFT uptake, C1 and C2 inhibited clonogenicity of H460 cells greater than PMX; PMX inhibited clonogenicity more than C1 or C2 at pH 7.2, which favors RFC transport over PCFT. Knockdown of PCFT in H460 cells resulted in decreased [(3)H]PMX and [(3)H]C2 transport and decreased growth inhibition by C1 and C2, and to a lesser extent by PMX. In vivo efficacy of C1 was seen toward H460 tumor xenografts in severe-combined immunodeficient mice. Our results suggest that 6-substituted pyrrolo[2,3-d]pyrimidine thienoyl antifolates offer significant promise for treating NS-NSCLC by selective uptake by PCFT.


Asunto(s)
Antineoplásicos/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Antagonistas del Ácido Fólico/metabolismo , Neoplasias Pulmonares/metabolismo , Transportador de Folato Acoplado a Protón/metabolismo , Animales , Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Antagonistas del Ácido Fólico/administración & dosificación , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Endogámicos ICR , Ratones SCID , Pirimidinas/administración & dosificación , Pirimidinas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
15.
Proc Natl Acad Sci U S A ; 110(38): 15180-8, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-23934049

RESUMEN

Antifolates, folate analogs that inhibit vitamin B9 (folic acid)-using cellular enzymes, have been used over several decades for the treatment of cancer and inflammatory diseases. Cellular uptake of the antifolates in clinical use occurs primarily via widely expressed facilitative membrane transporters. More recently, human folate receptors (FRs), high affinity receptors that transport folate via endocytosis, have been proposed as targets for the specific delivery of new classes of antifolates or folate conjugates to tumors or sites of inflammation. The development of specific, FR-targeted antifolates would be accelerated if additional biophysical data, particularly structural models of the receptors, were available. Here we describe six distinct crystallographic models that provide insight into biological trafficking of FRs and distinct binding modes of folate and antifolates to these receptors. From comparison of the structures, we delineate discrete structural conformations representative of key stages in the endocytic trafficking of FRs and propose models for pH-dependent conformational changes. Additionally, we describe the molecular details of human FR in complex with three clinically prevalent antifolates, pemetrexed (also Alimta), aminopterin, and methotrexate. On the whole, our data form the basis for rapid design and implementation of unique, FR-targeted, folate-based drugs for the treatment of cancer and inflammatory diseases.


Asunto(s)
Receptores de Folato Anclados a GPI/química , Antagonistas del Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Modelos Moleculares , Conformación Proteica , Animales , Células CHO , Cromatografía de Afinidad , Cricetinae , Cricetulus , Cristalización , Receptores de Folato Anclados a GPI/genética , Humanos , Estructura Molecular , Reacción en Cadena de la Polimerasa , Transporte de Proteínas/genética
16.
Proc Natl Acad Sci U S A ; 109(42): 16823-8, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23035243

RESUMEN

Malarial dihydrofolate reductase (DHFR) is the target of antifolate antimalarial drugs such as pyrimethamine and cycloguanil, the clinical efficacy of which have been compromised by resistance arising through mutations at various sites on the enzyme. Here, we describe the use of cocrystal structures with inhibitors and substrates, along with efficacy and pharmacokinetic profiling for the design, characterization, and preclinical development of a selective, highly efficacious, and orally available antimalarial drug candidate that potently inhibits both wild-type and clinically relevant mutated forms of Plasmodium falciparum (Pf) DHFR. Important structural characteristics of P218 include pyrimidine side-chain flexibility and a carboxylate group that makes charge-mediated hydrogen bonds with conserved Arg122 (PfDHFR-TS amino acid numbering). An analogous interaction of P218 with human DHFR is disfavored because of three species-dependent amino acid substitutions in the vicinity of the conserved Arg. Thus, P218 binds to the active site of PfDHFR in a substantially different fashion from the human enzyme, which is the basis for its high selectivity. Unlike pyrimethamine, P218 binds both wild-type and mutant PfDHFR in a slow-on/slow-off tight-binding mode, which prolongs the target residence time. P218, when bound to PfDHFR-TS, resides almost entirely within the envelope mapped out by the dihydrofolate substrate, which may make it less susceptible to resistance mutations. The high in vivo efficacy in a SCID mouse model of P. falciparum malaria, good oral bioavailability, favorable enzyme selectivity, and good safety characteristics of P218 make it a potential candidate for further development.


Asunto(s)
Antimaláricos/química , Antimaláricos/farmacología , Antagonistas del Ácido Fólico/metabolismo , Modelos Moleculares , Plasmodium falciparum/enzimología , Tetrahidrofolato Deshidrogenasa/química , Tetrahidrofolato Deshidrogenasa/metabolismo , Animales , Antimaláricos/farmacocinética , Dominio Catalítico/genética , Cristalografía por Rayos X , Diseño de Fármacos , Ratones , Ratones SCID , Estructura Molecular , Conformación Proteica
17.
Biochemistry ; 53(23): 3719-26, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24832237

RESUMEN

In the present study, we report the structure of the free and drug-bound Fab fragment of a high affinity anti-methotrexate antibody and perform a thermodynamic analysis of the binding process. The anti-methotrexate Fab fragment features a remarkably rigid tunnel-like binding site that extends into a water channel serving as a specialized route to move solvent out and into the site upon ligand binding and dissociation. This new finding in antibody structure-function relationships directly relates to the fast association (1 × 107 M⁻¹ s⁻¹) and slow dissociation (4 × 10⁻5 s⁻¹) rates determined for mAb ADD056, resulting in a very strong binding with a K(D) ~ 3.6 pM at 20 °C. As follows from the X-ray data analysis, the methotrexate-antibody complex is stabilized by an extended network of hydrogen bonds and stacking interactions. The analysis also shows structural involvement of the CDR H3 in formation of the water channel revealing another important role of this hypervariable region. This suggests a new direction in natural affinity maturation and opens a new possibility in antibody engineering. Methotrexate is a widely used therapeutic agent for many malignant diseases and inflammatory disorders. Unfortunately, it may also interfere with central aspects of metabolism and thereby cause inevitable side effects. Therefore, methotrexate therapy requires careful monitoring of drug blood levels, which is traditionally done by immunoassays. An understanding of the structure-function properties of antibodies selected for drug monitoring substantiates the performance and robustness of such tests.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Sitios de Unión de Anticuerpos , Antagonistas del Ácido Fólico/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Metotrexato/antagonistas & inhibidores , Modelos Moleculares , Agua/metabolismo , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Afinidad de Anticuerpos , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/metabolismo , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/metabolismo , Cristalografía por Rayos X , Antagonistas del Ácido Fólico/metabolismo , Hibridomas , Enlace de Hidrógeno , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Inmunosupresores/antagonistas & inhibidores , Inmunosupresores/química , Inmunosupresores/metabolismo , Ligandos , Metotrexato/química , Metotrexato/metabolismo , Ratones , Estabilidad Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Termodinámica , Agua/química
18.
Biol Pharm Bull ; 37(3): 439-46, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24583862

RESUMEN

The stereoselective transport of methotrexate (L-amethopterin, L-MTX) and its antipode (D-amethopterin, D-MTX) by human reduced folate carrier (hRFC) has been examined in HEK293 cells expressing H27-hRFC and R27-hRFC. The uptake of both L-MTX and D-MTX increased as the extracellular pH increased from 6.0 to 7.4. The initial uptake rate of L-MTX into the H27- and R27-hRFCs of the HEK293 cells followed Michaelis-Menten kinetics with Km values of approximately 0.24 and 0.47 µM, respectively. Dixon plots revealed that the [(3)H]-L-MTX uptake mediated by the H27- and R27-hRFCs was inhibited competitively by unlabeled L-MTX with Ki values of approximately 0.1 and 0.5 µM, respectively. D-MTX also competitively inhibited the H27- and R27-hRFC mediated uptake of [(3)H]-L-MTX with Ki values of approximately 3.4 and 3.2 µM, respectively. The RFC-mediated uptake clearance of L-MTX was approximately 15-fold greater than that of D-MTX in the H27-hRFC-HEK293 cells, and was more than 30-fold greater than that of D-MTX in the R27-hRFC-HEK293 cells. The results of the current study have revealed that the enantiomers of MTX enantiomers can be transported in a stereoselective manner by the H27- and R27-hRFCs because of significant differences in the affinities of the enantiomers to the hRFC.


Asunto(s)
Antagonistas del Ácido Fólico/metabolismo , Metotrexato/metabolismo , Proteína Portadora de Folato Reducido/genética , Adulto , Transporte Biológico/genética , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Cinética , Masculino , Proteína Portadora de Folato Reducido/metabolismo , Estereoisomerismo , Transfección , Adulto Joven
19.
ACS Infect Dis ; 10(4): 1185-1200, 2024 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-38499199

RESUMEN

New drugs with novel modes of action are needed to safeguard malaria treatment. In recent years, millions of compounds have been tested for their ability to inhibit the growth of asexual blood-stage Plasmodium falciparum parasites, resulting in the identification of thousands of compounds with antiplasmodial activity. Determining the mechanisms of action of antiplasmodial compounds informs their further development, but remains challenging. A relatively high proportion of compounds identified as killing asexual blood-stage parasites show evidence of targeting the parasite's plasma membrane Na+-extruding, H+-importing pump, PfATP4. Inhibitors of PfATP4 give rise to characteristic changes in the parasite's internal [Na+] and pH. Here, we designed a "pH fingerprint" assay that robustly identifies PfATP4 inhibitors while simultaneously allowing the detection of (and discrimination between) inhibitors of the lactate:H+ transporter PfFNT, which is a validated antimalarial drug target, and the V-type H+ ATPase, which was suggested as a possible target of the clinical candidate ZY19489. In our pH fingerprint assays and subsequent secondary assays, ZY19489 did not show evidence for the inhibition of pH regulation by the V-type H+ ATPase, suggesting that it has a different mode of action in the parasite. The pH fingerprint assay also has the potential to identify protonophores, inhibitors of the acid-loading Cl- transporter(s) (for which the molecular identity(ies) remain elusive), and compounds that act through inhibition of either the glucose transporter PfHT or glycolysis. The pH fingerprint assay therefore provides an efficient starting point to match a proportion of antiplasmodial compounds with their mechanisms of action.


Asunto(s)
Antimaláricos , Antagonistas del Ácido Fólico , Antimaláricos/farmacología , Antimaláricos/química , Plasmodium falciparum/metabolismo , Homeostasis , Proteínas de Transporte de Membrana/metabolismo , Iones/metabolismo , Antagonistas del Ácido Fólico/metabolismo , Concentración de Iones de Hidrógeno , ATPasas de Translocación de Protón/metabolismo
20.
Cell Chem Biol ; 31(2): 221-233.e14, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-37875111

RESUMEN

Methotrexate (MTX) is a tight-binding dihydrofolate reductase (DHFR) inhibitor, used as both an antineoplastic and immunosuppressant therapeutic. MTX, like folate undergoes folylpolyglutamate synthetase-mediated γ-glutamylation, which affects cellular retention and target specificity. Mechanisms of MTX resistance in cancers include a decrease in MTX poly-γ-glutamylation and an upregulation of DHFR. Here, we report a series of potent MTX-based proteolysis targeting chimeras (PROTACs) to investigate DHFR degradation pharmacology and one-carbon biochemistry. These on-target, cell-active PROTACs show proteasome- and E3 ligase-dependent activity, and selective degradation of DHFR in multiple cancer cell lines. By comparison, treatment with MTX increases cellular DHFR protein expression. Importantly, these PROTACs produced distinct, less-lethal phenotypes compared to MTX. The chemical probe set described here should complement conventional DHFR inhibitors and serve as useful tools for studying one-carbon biochemistry and dissecting complex polypharmacology of MTX and related drugs. Such compounds may also serve as leads for potential autoimmune and antineoplastic therapeutics.


Asunto(s)
Antineoplásicos , Antagonistas del Ácido Fólico , Neoplasias , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carbono , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacología , Antagonistas del Ácido Fólico/uso terapéutico , Metotrexato/farmacología , Metotrexato/metabolismo , Metotrexato/uso terapéutico , Neoplasias/tratamiento farmacológico , Quimera Dirigida a la Proteólisis , Tetrahidrofolato Deshidrogenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA