Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 286(26): 23150-9, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21550976

RESUMEN

Herein we measure the effect of four adaptive non-synonymous mutations to the glycerol kinase (glpK) gene on catalytic function and regulation, to identify changes that correlate to increased fitness in glycerol media. The mutations significantly reduce affinity for the allosteric inhibitor fructose-1,6-bisphosphate (FBP) and formation of the tetramer, which are structurally related, in a manner that correlates inversely with imparted fitness during growth on glycerol, which strongly suggests that these enzymatic parameters drive growth improvement. Counterintuitively, the glpK mutations also increase glycerol-induced auto-catabolite repression that reduces glpK transcription in a manner that correlates to fitness. This suggests that increased specific GlpK activity is attenuated by negative feedback on glpK expression via catabolite repression, possibly to prevent methylglyoxal toxicity. We additionally report that glpK mutations were fixed in 47 of 50 independent glycerol-adapted lineages. By far the most frequently mutated locus (nucleotide 218) was mutated in 20 lineages, strongly suggesting this position has an elevated mutation rate. This study demonstrates that fitness correlations can be used to interrogate adaptive processes at the protein level and to identify the regulatory constraints underlying selection and improved growth.


Asunto(s)
Escherichia coli K12/enzimología , Proteínas de Escherichia coli/metabolismo , Fructosadifosfatos/metabolismo , Glicerol Quinasa/metabolismo , Glicerol/metabolismo , Regulación Alostérica/fisiología , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Fructosadifosfatos/genética , Glicerol Quinasa/genética , Mutación
2.
Arch Biochem Biophys ; 492(1-2): 29-39, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19819219

RESUMEN

Unlike those for monomeric superfamily members, heterotropic allosteric effectors of the tetrameric Escherichia coli glycerol kinase (EGK) bind to only one of the two domains that define the catalytic cleft and far from the active site. An R369A amino acid substitution removes oligomeric interactions of a novel mini domain-swap loop of one subunit with the catalytic site of another subunit, and an A65T substitution perturbs oligomeric interactions in a second interface. Linked-functions enzyme kinetics, analytical ultracentrifugation, and FRET are used to assess effects of these substitutions on the allosteric control of catalysis. Inhibition by phosphotransferase system protein IIA(Glc) is reduced by the R369A substitution, and inhibition by fructose 1,6-bisphosphate is abolished by the A65T substitution. The oligomeric interactions enable the heterotropic allosteric effectors to act on both domains and modulate the catalytic cleft closure despite binding to only one domain.


Asunto(s)
Actinas/química , Proteínas de Escherichia coli/metabolismo , Glicerol Quinasa/antagonistas & inhibidores , Glicerol Quinasa/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Alanina/genética , Regulación Alostérica/genética , Sustitución de Aminoácidos/genética , Arginina/genética , Dominio Catalítico/genética , Dimerización , Interacciones Farmacológicas , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/química , Glicerol Quinasa/química , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/química , Familia de Multigenes , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Estereoisomerismo
3.
Arch Biochem Biophys ; 481(2): 151-6, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19056335

RESUMEN

IIA(Glc), the glucose-specific phosphocarrier protein of the phosphoenolpyruvate:glycose phosphotransferase system, is an allosteric inhibitor of Escherichia coli glycerol kinase. A linked-functions initial-velocity enzyme kinetics approach is used to define the MgATP-IIA(Glc) heterotropic allosteric interaction. The interaction is measured by the allosteric coupling constants Q and W, which describe the mutual effect of the ligands on binding affinity and the effect of the allosteric ligand on V(max), respectively. Allosteric interactions between these ligands display K-type activation and V-type inhibition. The allosteric coupling constant Q is about 3, showing cooperative coupling such that each ligand increases the affinity for binding of the other. The allosteric coupling constant W is about 0.1, showing that the allosteric inhibition is partial such that binding of IIA(Glc) at saturation does not reduce V(max) to zero. E. coli glycerol kinase is a member of the sugar kinase/heat shock protein 70/actin superfamily, and an element of the superfamily conserved ATPase catalytic core was identified as part of the IIA(Glc) inhibition network because it is required to transplant IIA(Glc) allosteric control into a non-allosteric glycerol kinase [A.C. Pawlyk, D.W. Pettigrew, Proc. Natl. Acad. Sci. USA 99 (2002) 11115-11120]. Two of the amino acids at this locus of E. coli glycerol kinase are replaced with those from the non-allosteric enzyme to enable determination of its contributions to MgATP-IIA(Glc) allosteric coupling. The substitutions reduce the affinity for IIA(Glc) by about 5-fold without changing significantly the allosteric coupling constants Q and W. The insensitivity of the allosteric coupling constants to the substitutions may indicate that the allosteric network is robust or the locus is not an element of that network. These possibilities may arise from differences of E. coli glycerol kinase relative to other superfamily members with respect to oligomeric structure and location of the allosteric site in a single domain far from the catalytic site.


Asunto(s)
Escherichia coli/enzimología , Glicerol Quinasa/genética , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Regulación Alostérica , Sustitución de Aminoácidos , Secuencia Conservada , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Glicerol Quinasa/química , Glicerol Quinasa/metabolismo , Cinética , L-Lactato Deshidrogenasa/metabolismo , Modelos Moleculares , Conformación Proteica , Piruvato Quinasa/metabolismo , Termodinámica
4.
Biochemistry ; 46(43): 12355-65, 2007 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-17924663

RESUMEN

Steady-state and time-resolved fluorescence anisotropy methods applied to an extrinsic fluorophore that is conjugated to non-native cysteine residues demonstrate that amino acids in an allosteric communication network within a protein subunit tune protein backbone motions at a distal site to enable allosteric binding and inhibition. The unphosphorylated form of the phosphocarrier protein IIAGlc is an allosteric inhibitor of Escherichia coli glycerol kinase, binding more than 25 A from the kinase active site. Crystal structures that showed a ligand-dependent conformational change and large temperature factors for the IIAGlc-binding site on E. coli glycerol kinase suggest that motions of the allosteric site have an important role in the inhibition. Three E. coli glycerol kinase amino acids that are located at least 15 A from the active site and the allosteric site were shown previously to be necessary for transplanting IIAGlc inhibition into the nonallosteric glycerol kinase from Haemophilus influenzae. These three amino acids are termed the coupling locus. The apparent allosteric site motions and the requirement for the distant coupling locus to transplant allosteric inhibition suggest that the coupling locus modulates the motions of the IIAGlc-binding site. To evaluate this possibility, variants of E. coli glycerol kinase and the chimeric, allosteric H. influenzae glycerol kinase were constructed with a non-native cysteine residue replacing one of the native residues in the IIAGlc-binding site. The extrinsic fluorophore Oregon Green 488 (2',7'-difluorofluorescein) was conjugated specifically to the non-native cysteine residue. Steady-state and time-resolved fluorescence anisotropy measurements show that the motions of the fluorophore reflect backbone motions of the IIAGlc-binding site and these motions are modulated by the amino acids at the coupling locus.


Asunto(s)
Proteínas de Escherichia coli/fisiología , Glicerol Quinasa/antagonistas & inhibidores , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/fisiología , Sitio Alostérico , Escherichia coli/enzimología , Proteínas de Escherichia coli/genética , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética
5.
Biochemistry ; 42(14): 4243-52, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12680779

RESUMEN

Escherichia coli glycerol kinase (EC 2.7.1.30; ATP-glycerol 3-phosphotransferase) is inhibited allosterically by fructose 1,6-bisphosphate (FBP), and this inhibition is a primary mechanism by which glucose controls glycerol utilization in vivo. Earlier work indicates that glycerol kinase displays a dimer-tetramer equilibrium in solution, FBP shifts the equilibrium toward the tetramer, and tetramer formation is required for FBP inhibition. However, equilibrium constants for FBP binding and dimer-tetramer assembly that describe the linkage between these processes are unknown. Here, decreased fluorescence anisotropy of extrinsic fluorophores fluorescein and 2',7'-difluorofluorescein due to homo fluorescence resonance energy transfer (homo-FRET) is used to quantitate tetramer assembly and FBP binding. Glycerol kinase is labeled with extrinsic fluorophores covalently attached to an engineered surface cysteine residue under conditions that prevent labeling of native cysteine residues. Tryptic peptide mapping and MALDI-MS verify labeling at the engineered site only. Initial velocity studies show the labeling does not alter the catalytic properties or FBP inhibition. The steady-state fluorescence anisotropy of enzyme with a labeling stoichiometry of approximately 0.1 mol of fluorophore/mol of subunit is not sensitive to increased protein concentration or binding of FBP, indicating the absence of homo-FRET. However, steady-state fluorescence anisotropy of enzyme with a labeling stoichiometry of approximately 0.4 mol of fluorophore/mol of subunit decreases with increasing protein concentration, which is consistent with depolarization due to homo-FRET. The protein concentration dependence of the decreased fluorescence anisotropy is described by a dimer-tetramer equilibrium with an apparent dissociation constant of 61 +/- 7 nM (subunits) at pH 7.0 and 25 degrees C. FBP binds to both the dimer and tetramer of glycerol kinase, and the FBP concentration dependence of the apparent dissociation constant for the dimer-tetramer equilibrium shows critical behavior. The apparent dissociation constant decreases and then increases with increasing FBP concentration, reaching a minimum at about 20 mM FBP. Critical behavior is seen also in the FBP dependence of the inhibition. The critical behavior arises because tetramer dissociation increases FBP stoichiometry from two sites per tetramer to four half-sites per two dimers. The phenomenological description of the coupling between tetramer assembly and FBP binding shows antagonistic binding of FBP to the two sites on the tetramer, indicating that the strong positive cooperativity observed for FBP inhibition of catalytic activity (Hill coefficient approximately 1.5) is due to the approximately 4000-fold higher affinity of the tetramer for FBP rather than to positive coupling between the two FBP sites.


Asunto(s)
Escherichia coli/enzimología , Fructosadifosfatos/metabolismo , Glicerol Quinasa/metabolismo , Biopolímeros , Glicerol Quinasa/química , Ligandos , Modelos Moleculares , Conformación Proteica , Espectrometría de Fluorescencia
6.
Proc Natl Acad Sci U S A ; 99(17): 11115-20, 2002 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-12161559

RESUMEN

Glycerol kinase from Escherichia coli, but not Haemophilus influenzae, is inhibited allosterically by phosphotransferase system protein IIA(Glc). The primary structures of these related kinases contain 501 amino acids, differing at 117. IIA(Glc) inhibition is transplanted from E. coli glycerol kinase into H. influenzae glycerol kinase by interconverting only 11 of the differences: 8 residues that interact with IIA(Glc) at the allosteric binding site and 3 residues in the conserved ATPase catalytic core that do not interact with IIA(Glc) but the solvent accessible surface of which decreases when it binds. The three core residues are crucial for coupling the allosteric site to the conserved catalytic core of the enzyme. The site of the coupling residues identifies a regulatory locus in the sugar kinase/heat shock protein 70/actin superfamily and suggests relations between allosteric regulation and the active site closure that characterizes the family. The location of the coupling residues provides empirical validation of a computational model that predicts a coupling pathway between the IIA(Glc)-binding site and the active site [Luque, I. & Freire, E. (2000) Proteins Struct. Funct. Genet. Suppl. 4, 63-71]. The requirement for changes in core residues to couple the allosteric and active sites and switching from inhibition to activation by a single amino acid change are consistent with a postulated mechanism for molecular evolution of allosteric regulation.


Asunto(s)
Enzimas/metabolismo , Glicerol Quinasa/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Enzimas/química , Escherichia coli/enzimología , Glicerol Quinasa/química , Haemophilus influenzae/enzimología , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Fosfotransferasas/metabolismo , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA