Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Protein Sci ; 24(4): 495-507, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25401264

RESUMO

Type II R67 dihydrofolate reductase (DHFR) is a bacterial plasmid-encoded enzyme that is intrinsically resistant to the widely-administered antibiotic trimethoprim. R67 DHFR is genetically and structurally unrelated to E. coli chromosomal DHFR and has an unusual architecture, in that four identical protomers form a single symmetrical active site tunnel that allows only one substrate binding/catalytic event at any given time. As a result, substitution of an active-site residue has as many as four distinct consequences on catalysis, constituting an atypical model of enzyme evolution. Although we previously demonstrated that no single residue of the native active site is indispensable for function, library selection here revealed a strong bias toward maintenance of two native protomers per mutated tetramer. A variety of such "half-native" tetramers were shown to procure native-like catalytic activity, with similar KM values but kcat values 5- to 33-fold lower, illustrating a high tolerance for active-site substitutions. The selected variants showed a reduced thermal stability (Tm ∼12°C lower), which appears to result from looser association of the protomers, but generally showed a marked increase in resilience to heat denaturation, recovering activity to a significantly greater extent than the variant with no active-site substitutions. Our results suggest that the presence of two native protomers in the R67 DHFR tetramer is sufficient to provide native-like catalytic rate and thus ensure cellular proliferation.


Assuntos
Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/genética , Domínio Catalítico/genética , Farmacorresistência Bacteriana , Estabilidade Enzimática/genética , Mutagênese Sítio-Dirigida , Plasmídeos , Desnaturação Proteica , Multimerização Proteica , Tetra-Hidrofolato Desidrogenase/metabolismo , Trimetoprima
2.
Protein Sci ; 24(4): 534-44, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25492589

RESUMO

Bacterial resistance to ß-lactam antibiotics is a global issue threatening the success of infectious disease treatments worldwide. Mycobacterium tuberculosis has been particularly resilient to ß-lactam treatment, primarily due to the chromosomally encoded BlaC ß-lactamase, a broad-spectrum hydrolase that renders ineffective the vast majority of relevant ß-lactam compounds currently in use. Recent laboratory and clinical studies have nevertheless shown that specific ß-lactam-BlaC inhibitor combinations can be used to inhibit the growth of extensively drug-resistant strains of M. tuberculosis, effectively offering new tools for combined treatment regimens against resistant strains. In the present work, we performed combinatorial active-site replacements in BlaC to demonstrate that specific inhibitor-resistant (IRT) substitutions at positions 69, 130, 220, and/or 234 can act synergistically to yield active-site variants with several thousand fold greater in vitro resistance to clavulanate, the most common clinical ß-lactamase inhibitor. While most single and double variants remain sensitive to clavulanate, double mutants R220S-K234R and S130G-K234R are substantially less affected by time-dependent clavulanate inactivation, showing residual ß-lactam hydrolytic activities of 46% and 83% after 24 h incubation with a clinically relevant inhibitor concentration (5 µg/ml, 25 µM). These results demonstrate that active-site alterations in BlaC yield resistant variants that remain active and stable over prolonged bacterial generation times compatible with mycobacterial proliferation. These results also emphasize the formidable adaptive potential of inhibitor-resistant substitutions in ß-lactamases, potentially casting a shadow on specific ß-lactam-BlaC inhibitor combination treatments against M. tuberculosis.


Assuntos
Ácido Clavulânico/farmacologia , Farmacorresistência Bacteriana/genética , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , beta-Lactamases/química , beta-Lactamases/genética , Cinética , Modelos Moleculares , Mutação/genética , beta-Lactamases/metabolismo
3.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 11): 1316-22, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22102224

RESUMO

Trimethoprim is an antibiotic that targets bacterial dihydrofolate reductase (DHFR). A plasmid-encoded DHFR known as R67 DHFR provides resistance to trimethoprim in bacteria. To better understand the mechanism of this homotetrameric enzyme, a tandem dimer construct was created that linked two monomeric R67 DHFR subunits together and mutated the sequence of residues 66-69 of the first subunit from VQIY to INSF. Using a modified crystallization protocol for this enzyme that included in situ proteolysis using chymotrypsin, the tandem dimer was crystallized and the structure was solved at 1.4 Å resolution. Surprisingly, only wild-type protomers were incorporated into the crystal. Further experiments demonstrated that the variant protomer was selectively degraded by chymotrypsin, although no canonical chymotrypsin cleavage site had been introduced by these mutations.


Assuntos
Tetra-Hidrofolato Desidrogenase/química , Cristalização , Cristalografia por Raios X , Modelos Moleculares , Mutação , Regiões Promotoras Genéticas , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Tetra-Hidrofolato Desidrogenase/genética
4.
J Mol Recognit ; 24(2): 188-98, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21360609

RESUMO

Ex vivo selection of transduced hematopoietic stem cells (HSC) with drug-resistance genes offers the possibility to enrich transduced cells prior to engraftment, toward increased reconstitution in transplant recipients. We evaluated the potential of highly methotrexate (MTX)-resistant variants of human dihydrofolate reductase (hDHFR) for this application. Two subsets of hDHFR variants with reduced affinity for MTX that had been previously identified in a bacterial system were considered: those with substitutions at positions 31, 34, and/or 35, and those with substitutions at position 115. The variants were characterized for their resistance to pemetrexed (PMTX), an antifolate that is related to MTX. We observed a strong correlation between decreased binding to both antifolates, although the identity of specific sequence variations modulated the correlation. We chose a subset of hDHFR variants for tests of ex vivo MTX resistance, taking into consideration their residual specific activity and their decrease in affinity for the related antifolates. Murine myeloid progenitors and other differentiated hematopoietic cells were transduced and exposed to MTX in a nucleotide-free medium. Bone marrow (BM) cells including 15% cells infected with F31R/Q35E were enriched to 98% transduced cells within 6 days of ex vivo selection. hDHFR variant F31R/Q35E allowed a strong ex vivo enrichment upon a short exposure to MTX relative to a less resistant variant of hDHFR, L22Y. We have thus demonstrated that bacterial selection of highly antifolate-resistant hDHFR variants can provide selectable markers for rapid ex vivo enrichment of hematopoietic cells.


Assuntos
Células da Medula Óssea/citologia , Metotrexato/metabolismo , Seleção Genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/virologia , Domínio Catalítico , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Resistência a Medicamentos/efeitos dos fármacos , Antagonistas do Ácido Fólico/química , Antagonistas do Ácido Fólico/metabolismo , Antagonistas do Ácido Fólico/farmacologia , Glutamatos/química , Glutamatos/metabolismo , Glutamatos/farmacologia , Guanina/análogos & derivados , Guanina/química , Guanina/metabolismo , Guanina/farmacologia , Humanos , Cinética , Ligantes , Metotrexato/química , Metotrexato/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mutantes/metabolismo , Pemetrexede , Especificidade por Substrato/efeitos dos fármacos , Tetra-Hidrofolato Desidrogenase/química , Fatores de Tempo , Transdução Genética
5.
J Biol Chem ; 284(30): 20079-89, 2009 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-19478082

RESUMO

Methotrexate is a slow, tight-binding, competitive inhibitor of human dihydrofolate reductase (hDHFR), an enzyme that provides key metabolites for nucleotide biosynthesis. In an effort to better characterize ligand binding in drug resistance, we have previously engineered hDHFR variant F31R/Q35E. This variant displays a >650-fold decrease in methotrexate affinity, while maintaining catalytic activity comparable to the native enzyme. To elucidate the molecular basis of decreased methotrexate affinity in the doubly substituted variant, we determined kinetic and inhibitory parameters for the simple variants F31R and Q35E. This demonstrated that the important decrease of methotrexate affinity in variant F31R/Q35E is a result of synergistic effects of the combined substitutions. To better understand the structural cause of this synergy, we obtained the crystal structure of hDHFR variant F31R/Q35E complexed with methotrexate at 1.7-A resolution. The mutated residue Arg-31 was observed in multiple conformers. In addition, seven native active-site residues were observed in more than one conformation, which is not characteristic of the wild-type enzyme. This suggests that increased residue disorder underlies the observed methotrexate resistance. We observe a considerable loss of van der Waals and polar contacts with the p-aminobenzoic acid and glutamate moieties. The multiple conformers of Arg-31 further suggest that the amino acid substitutions may decrease the isomerization step required for tight binding of methotrexate. Molecular docking with folate corroborates this hypothesis.


Assuntos
Inibidores Enzimáticos/metabolismo , Metotrexato/metabolismo , Mutação Puntual , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/genética , Domínio Catalítico , Cristalografia por Raios X , Resistência a Medicamentos , Inibidores Enzimáticos/química , Ácido Fólico/química , Ácido Fólico/metabolismo , Humanos , Metotrexato/química , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Tetra-Hidrofolato Desidrogenase/metabolismo
6.
Drug Resist Updat ; 12(1-2): 28-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19272832

RESUMO

Human dihydrofolate reductase (DHFR) is a primary target for antifolate drugs in cancer treatment, while DHFRs from Plasmodium falciparum, Plasmodium vivax and various bacterial species are primary targets in the treatment of malaria and bacterial infections. Mutations in each of these DHFRs can result in resistance towards clinically relevant antifolates. We review the structural and functional impact of active-site mutations with respect to enzyme activity and antifolate resistance of DHFRs from mammals, protozoa and bacteria. The high structural homology between DHFRs results in a number of cross-species, active-site 'hot-spots' for broad-based antifolate resistance. In addition, we identify mutations that confer species-specific resistance, or antifolate-specific resistance. This comparative review of antifolate binding in diverse species provides new insights into the relationship between antifolate design and the development of mutational resistance. It also presents avenues for designing antifolate-resistant mammalian DHFRs as chemoprotective agents.


Assuntos
Proteínas de Bactérias/metabolismo , Resistência a Medicamentos/genética , Antagonistas do Ácido Fólico/uso terapêutico , Mutação , Neoplasias/tratamento farmacológico , Proteínas de Protozoários/metabolismo , Tetra-Hidrofolato Desidrogenase/metabolismo , Sequência de Aminoácidos , Animais , Antibacterianos/uso terapêutico , Antimaláricos/uso terapêutico , Antineoplásicos/uso terapêutico , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Farmacorresistência Bacteriana/genética , Resistencia a Medicamentos Antineoplásicos/genética , Antagonistas do Ácido Fólico/química , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Neoplasias/enzimologia , Neoplasias/genética , Conformação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Especificidade da Espécie , Relação Estrutura-Atividade , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/genética
7.
J Biomol Screen ; 13(6): 504-14, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18566481

RESUMO

We report a rapid and reliable 2-tier selection and screen for detection of activity as well as drug-resistance in mutated variants of a clinically-relevant drug-target enzyme. Human dihydrofolate reductase point-mutant libraries were subjected to a 1st-tier bacterial complementation assay, such that bacterial propagation served as an indicator of enzyme activity. Alternatively, when selection was performed in the presence of the inhibitor methotrexate (MTX), propagation indicated MTX resistance. The selected variants were then subjected to a 2nd-tier in vitro screen in 96-well plate format using crude bacterial lysate. Conditions were defined to establish a threshold for activity or for MTX resistance. The 2nd-tier assay allowed rapid detection of the best variants among the leads and provided reliable estimates of relative reactivity, (k(cat)) and IC(50)(MTX). Screening saturation libraries of active-site positions 7, 15, 24, 70, and 115 revealed a variety of novel mutations compatible with reactivity as well as 2 novel MTX-resistant variants: V115A and V115C. Both variants displayed K(i)(MTX)=20 nM, a 600-fold increase relative to the wild-type. We also present preliminary results from screening against further antifolates following simple modifications of the protocol. The flexibility and robustness of this method will provide new insights into interactions between ligands and active-site residues of this clinically relevant human enzyme.


Assuntos
Resistência a Medicamentos , Antagonistas do Ácido Fólico/farmacologia , Metotrexato/farmacologia , Tetra-Hidrofolato Desidrogenase/genética , Sítios de Ligação , Simulação por Computador , Escherichia coli/genética , Biblioteca Gênica , Teste de Complementação Genética , Variação Genética , Humanos , Metotrexato/química , Modelos Biológicos , Modelos Moleculares , Mutação Puntual , Reprodutibilidade dos Testes , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/metabolismo , Fatores de Tempo
8.
J Mol Biol ; 373(3): 599-611, 2007 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-17868689

RESUMO

Methotrexate-resistant forms of human dihydrofolate reductase have the potential to protect healthy cells from the toxicity of methotrexate (MTX), to improve prognosis during cancer therapy. It has been shown that synergistic MTX-resistance can be obtained by combining two active-site mutations that independently confer weak MTX-resistance. In order to obtain more highly MTX-resistant human dihydrofolate reductase (hDHFR) variants for this application, we used a semi-rational approach to obtain combinatorial active-site mutants of hDHFR that are highly resistant towards MTX. We created a combinatorial mutant library encoding various amino acids at residues Phe31, Phe34 and Gln35. In vivo library selection was achieved in a bacterial system on medium containing high concentrations of MTX. We characterized ten novel MTX-resistant mutants with different amino acid combinations at residues 31, 34 and 35. Kinetic and inhibition parameters of the purified mutants revealed that higher MTX-resistance roughly correlated with a greater number of mutations, the most highly-resistant mutants containing three active site mutations (Ki(MTX)=59-180 nM; wild-type Ki(MTX)<0.03 nM). An inverse correlation was observed between resistance and catalytic efficiency, which decreased mostly as a result of increased KM toward the substrate dihydrofolate. We verified that the MTX-resistant hDHFRs can protect eukaryotic cells from MTX toxicity by transfecting the most resistant mutants into DHFR-knock-out CHO cells. The transfected variants conferred survival at concentrations of MTX between 100-fold and >4000-fold higher than the wild-type enzyme, the most resistant triple mutant offering protection beyond the maximal concentration of MTX that could be included in the medium. These highly resistant variants of hDHFR offer potential for myeloprotection during administration of MTX in cancer treatment.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Resistência a Medicamentos , Metotrexato/farmacologia , Mutação/genética , Tetra-Hidrofolato Desidrogenase/genética , Animais , Antimetabólitos Antineoplásicos/química , Sítios de Ligação , Células CHO/efeitos dos fármacos , Cricetinae , Cricetulus , Cristalografia por Raios X , Humanos , Metotrexato/química , Modelos Moleculares , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA