Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochemistry ; 62(16): 2442-2449, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37526986

RESUMO

The interplay between active-site chemistry and functionally relevant enzyme motions can provide useful insights into selective enzyme modulation. Modulation of the hypoxia-sensing function of factor-inhibiting-HIF-1 (FIH) enzyme is a potential therapeutic strategy in disease states such as ischemia and cancer. The hypoxia-sensing function of FIH relies in major part on the tight coupling of the first half of the catalytic mechanism which involves O2 activation and eventual succinate production to the second half which involves HIF-1α/CTAD substrate hydroxylation. In this study, we demonstrate the role of a loop hinge domain in FIH (FIH102-118) called the 100s loop in maintaining this particular tight coupling. Molecular dynamics patterns from Gaussian Network Model (iGNM) database analysis of FIH identified the 100s loop as one dynamic domain containing a hinge residue (Tyr102) with a potential substrate positioning role. Enzymological and biophysical studies of the 100s loop point mutants revealed altered enzyme kinetics with the exception of the conservative FIH mutant Y102F, which suggests a sterics-related role for this residue. Removal of the bulk of Tyr102 (Y102A) resulted in succinate production, autohydroxylation, and an O2 binding environment comparable to wild-type FIH. However, the HIF-1α/CTAD substrate hydroxylation of this mutant was significantly reduced which implies that (1) the FIH loop hinge residue Tyr102 does not affect O2 activation, (2) the stacking steric interaction of Tyr102 is important in substrate positioning for productive hydroxylation, and (3) Tyr102 is important for the synchronization of O2 activation and substrate hydroxylation.

2.
Methods Enzymol ; 679: 363-380, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36682871

RESUMO

Primary kinetic isotope effects (KIEs) provide unique insight into enzymatic reactions, as they can reveal rate-limiting steps and detailed chemical mechanisms. HIF hydroxylases, part of a family of 2-oxoglutarate (2OG) oxygenases are central to the regulation of many crucial biological processes through O2-sensing, but present a challenge to monitor due to the large size of the protein substrate and the similarity between native and hydroxylated substrate. MALDI-TOF MS is a convenient tool to measure peptide masses, which can also be used to measure the discontinuous kinetics of peptide hydroxylation for Factor Inhibiting HIF (FIH). Using this technique, rate data can be observed from the mole-fraction of CTAD and CTAD-OH in small volumes, allowing noncompetitive H/D KIEs to be measured. Slow dCTAD substrate leads to extensive uncoupling of O2 consumption from peptide hydroxylation, leading to enzyme autohydroxylation, which is observed using UV-vis spectroscopy. Simultaneously measuring both the normal product, CTAD-OH, and the uncoupled product, autohydroxylated enzyme, the KIE on the microscopic step of hydrogen atom transfer (HAT) can be estimated. MALDI-MS analysis is a strong method for monitoring reactions that hydroxylate peptides, and can be generalized to other similar reactions, and simultaneous kinetic detection of branched products can provide valuable insight on microscopic KIEs at intermediate mechanistic steps.


Assuntos
Peptídeos , Proteínas Repressoras , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Proteínas Repressoras/química , Cinética , Hidroxilação , Peptídeos/metabolismo
3.
Inorg Chem ; 57(20): 12588-12595, 2018 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-30252455

RESUMO

α-Ketoglutarate (αKG) dependent oxygenases comprise a large superfamily of enzymes that activate O2 for varied reactions. While most of these enzymes contain a nonheme Fe bound by a His2(Asp/Glu) facial triad, a small number of αKG-dependent halogenases require only the two His ligands to bind Fe and activate O2. The enzyme "factor inhibiting HIF" (FIH) contains a His2Asp facial triad and selectively hydroxylates polypeptides; however, removal of the Asp ligand in the Asp201→Gly variant leads to a highly active enzyme, seemingly without a complete facial triad. Herein, we report on the formation of an Fe-Cl cofactor structure for the Asp201→Gly FIH variant using X-ray absorption spectroscopy (XAS), which provides insight into the structure of the His2Cl facial triad found in halogenases. The Asp201→Gly variant supports anion dependent peptide hydroxylation, demonstrating the requirement for a complete His2X facial triad to support O2 reactivity. Our results indicated that exogenous ligand binding to form a complete His2X facial triad was essential for O2 activation and provides a structural model for the His2Cl-bound nonheme Fe found in halogenases.


Assuntos
Cloretos/química , Ferro/metabolismo , Oxigenases de Função Mista/metabolismo , Oxigênio/metabolismo , Proteínas Repressoras/metabolismo , Absorciometria de Fóton , Substituição de Aminoácidos , Cloretos/metabolismo , Ferro/química , Ligantes , Oxigenases de Função Mista/química , Ligação Proteica , Conformação Proteica , Proteínas Repressoras/química
4.
Biochemistry ; 55(2): 277-86, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26727884

RESUMO

The Fe(2+)/α-ketoglutarate (αKG)-dependent oxygenases use molecular oxygen to conduct a wide variety of reactions with important biological implications, such as DNA base excision repair, histone demethylation, and the cellular hypoxia response. These enzymes follow a sequential mechanism in which O2 binds and reacts after the primary substrate binds, making those structural factors that promote productive O2 binding central to their chemistry. A large challenge in this field is to identify strategies that engender productive turnover. Factor inhibiting HIF (FIH) is a Fe(2+)/αKG-dependent oxygenase that forms part of the O2 sensing machinery in human cells by hydroxylating the C-terminal transactivation domain (CTAD) found within the HIF-1α protein. The structure of FIH was determined with the O2 analogue NO bound to Fe, offering the first direct insight into the gas binding geometry in this enzyme. Through a combination of density functional theory calculations, {FeNO}(7) electron paramagnetic resonance spectroscopy, and ultraviolet-visible absorption spectroscopy, we demonstrate that CTAD binding stimulates O2 reactivity by altering the orientation of the bound gas molecule. Although unliganded FIH binds NO with moderate affinity, the bound gas can adopt either of two orientations with similar stability; upon CTAD binding, NO adopts a single preferred orientation that is appropriate for supporting oxidative decarboxylation. Combined with other studies of related enzymes, our data suggest that substrate-induced reorientation of bound O2 is the mechanism utilized by the αKG oxygenases to tightly couple O2 activation to substrate hydroxylation.


Assuntos
Ácidos Cetoglutáricos/metabolismo , Oxigenases de Função Mista/metabolismo , Oxigenases/metabolismo , Proteínas Repressoras/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Humanos , Ferro/metabolismo , Óxido Nítrico/metabolismo , Oxigênio/metabolismo , Estrutura Secundária de Proteína , Especificidade por Substrato
6.
J Inorg Biochem ; 133: 63-72, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24468676

RESUMO

The ability to sense and adapt to changes in pO2 is crucial for basic metabolism in most organisms, leading to elaborate pathways for sensing hypoxia (low pO2). This review focuses on the mechanisms utilized by mammals and bacteria to sense hypoxia. While responses to acute hypoxia in mammalian tissues lead to altered vascular tension, the molecular mechanism of signal transduction is not well understood. In contrast, chronic hypoxia evokes cellular responses that lead to transcriptional changes mediated by the hypoxia inducible factor (HIF), which is directly controlled by post-translational hydroxylation of HIF by the non-heme Fe(II)/αKG-dependent enzymes FIH and PHD2. Research on PHD2 and FIH is focused on developing inhibitors and understanding the links between HIF binding and the O2 reaction in these enzymes. Sulfur speciation is a putative mechanism for acute O2-sensing, with special focus on the role of H2S. This sulfur-centered model is discussed, as are some of the directions for further refinement of this model. In contrast to mammals, bacterial O2-sensing relies on protein cofactors that either bind O2 or oxidatively decompose. The sensing modality for bacterial O2-sensors is either via altered DNA binding affinity of the sensory protein, or else due to the actions of a two-component signaling cascade. Emerging data suggests that proteins containing a hemerythrin-domain, such as FBXL5, may serve to connect iron sensing to O2-sensing in both bacteria and humans. As specific molecular machinery becomes identified, these hypoxia sensing pathways present therapeutic targets for diseases including ischemia, cancer, or bacterial infection.


Assuntos
Hipóxia/metabolismo , Oxigênio/metabolismo , Transdução de Sinais/genética , Animais , Bactérias/metabolismo , Heme/metabolismo , Humanos , Hipóxia/genética , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Mamíferos/metabolismo
7.
Metallomics ; 5(4): 287-301, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23446356

RESUMO

The Fe(ii)/αketoglutarate (αKG) dependent oxygenases catalyze a diverse range of reactions significant in biological processes such as antibiotic biosynthesis, lipid metabolism, oxygen sensing, and DNA and RNA repair. Although functionally diverse, the eight-stranded ß-barrel (cupin) and HX(D/E)XnH facial triad motifs are conserved in this super-family of enzymes. Crystal structure analysis of 25 αKG oxygenases reveals two stereoisomers of the Fe cofactor, Anti and Clock, which differ in the relative position of the exchangeable ligand position and the primary substrate. Herein, we discuss the relationship between the chemical mechanism and the secondary coordination sphere of the αKG oxygenases, within the constraints of the stereochemistry of the Fe cofactor. Sequence analysis of the cupin barrel indicates that a small subset of positions constitute the second coordination sphere, which has significant ramifications for the structure of the ferryl intermediate. The competence of both Anti and Clock stereoisomers of Fe points to a ferryl intermediate that is 5 coordinate. The small number of conserved close contacts within the active sites of αKG oxygenases can be extended to chemically related enzymes, such as the αKG-dependent halogenases SyrB2 and CytC3, and the non-αKG dependent dioxygenases isopenicillin N synthase (IPNS) and cysteine dioxygenase (CDO).


Assuntos
Metais/química , Oxigenases/química , Oxigenases/metabolismo , Sequência de Aminoácidos , Ácidos Cetoglutáricos , Metais/metabolismo , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Estereoisomerismo , Especificidade por Substrato
8.
Biochemistry ; 52(9): 1594-602, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23351038

RESUMO

Oxygen homeostasis plays a critical role in angiogenesis, erythropoiesis, and cell metabolism. Oxygen homeostasis is set by the hypoxia inducible factor-1α (HIF-1α) pathway, which is controlled by factor inhibiting HIF-1α (FIH). FIH is a non-heme Fe(II), α-ketoglutarate (αKG)-dependent dioxygenase that inhibits HIF-1α by hydroxylating the C-terminal transactivation domain (CTAD) of HIF-1α at HIF-Asn(803). A tight coupling between CTAD binding and O2 activation is essential for hypoxia sensing, making changes in the coordination geometry of Fe(II) upon CTAD encounter a crucial feature of this enzyme. Although the consensus chemical mechanism for FIH proposes that CTAD binding triggers O2 activation by causing the Fe(II) cofactor to release an aquo ligand, experimental evidence of this has been absent. More broadly, this proposed coordination change at Fe(II) has not been observed during steady-state turnover in any αKG oxygenase to date. In this work, solvent isotope effects (SIEs) were used as a direct mechanistic probe of substrate-triggered aquo release in FIH, as inverse SIEs (SIE < 1) are signatures for pre-equilibrium aquo release from metal ions. Our mechanistic studies of FIH have revealed inverse solvent isotope effects in the steady-state rate constants at limiting concentrations of CTAD or αKG [(D2O)kcat/KM(CTAD) = 0.40 ± 0.07, and (D2O)kcat/KM(αKG) = 0.32 ± 0.08], providing direct evidence of aquo release during steady-state turnover. Furthermore, the SIE at saturating concentrations of CTAD and αKG was inverse ((D2O)kcat = 0.51 ± 0.07), indicating that aquo release occurs after CTAD binds. The inverse kinetic SIEs observed in the steady state for FIH can be explained by a strong Fe-OH2 bond. The stable Fe-OH2 bond plays an important part in FIH's regulatory role over O2 homeostasis in humans and points toward a strategy for tightly coupling O2 activation with CTAD hydroxylation that relies on substrate triggering.


Assuntos
Óxido de Deutério/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Ácidos Cetoglutáricos/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Repressoras/metabolismo , Sequência de Aminoácidos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/química , Cinética , Oxigenases de Função Mista/química , Dados de Sequência Molecular , Oxigênio/metabolismo , Estrutura Terciária de Proteína , Proteínas Repressoras/química , Especificidade por Substrato
9.
J Inorg Biochem ; 113: 25-30, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22687491

RESUMO

Two primary O(2)-sensors for humans are the HIF-hydroxylases, enzymes that hydroxylate specific residues of the hypoxia inducible factor-α (HIF). These enzymes are factor inhibiting HIF (FIH) and prolyl hydroxylase-2 (PHD2), each an α-ketoglutarate (αKG) dependent, non-heme Fe(II) dioxygenase. Although the two enzymes have similar active sites, FIH hydroxylates Asn(803) of HIF-1α while PHD2 hydroxylates Pro(402) and/or Pro(564) of HIF-1α. The similar structures but unique functions of FIH and PHD2 make them prime targets for selective inhibition leading to regulatory control of diseases such as cancer and stroke. Three classes of iron chelators were tested as inhibitors for FIH and PHD2: pyridines, hydroxypyrones/hydroxypyridinones and catechols. An initial screen of the ten small molecule inhibitors at varied [αKG] revealed a non-overlapping set of inhibitors for PHD2 and FIH. Dose response curves at moderate [αKG] ([αKG]~K(M)) showed that the hydroxypyrones/hydroxypyridinones were selective inhibitors, with IC(50) in the µM range, and that the catechols were generally strong inhibitors of both FIH and PHD2, with IC(50) in the low µM range. As support for binding at the active site of each enzyme as the mode of inhibition, electron paramagnetic resonance (EPR) spectroscopy were used to demonstrate inhibitor binding to the metal center of each enzyme. This work shows some selective inhibition between FIH and PHD2, primarily through the use of simple aromatic or pseudo-aromatic chelators, and suggests that hydroxypyrones and hydroxypyridones may be promising chelates for FIH or PHD2 inhibition.


Assuntos
Quelantes de Ferro/química , Ferro/química , Oxigenases de Função Mista/química , Pró-Colágeno-Prolina Dioxigenase/química , Proteínas Repressoras/química , Sequência de Aminoácidos , Sítios de Ligação , Catecóis/química , Espectroscopia de Ressonância de Spin Eletrônica , Escherichia coli , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/química , Prolina Dioxigenases do Fator Induzível por Hipóxia , Ácidos Cetoglutáricos/química , Cinética , Oxigenases de Função Mista/antagonistas & inibidores , Dados de Sequência Molecular , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Ligação Proteica , Piridinas/química , Piridonas/química , Pironas/química , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Repressoras/antagonistas & inibidores , Especificidade por Substrato
10.
Inorg Chem ; 37(23): 6065-6070, 1998 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-11670744

RESUMO

The use of the bis(bipyridine) ligand L (1,2-bis(2,2'-bipyridyl-6-yl)ethane) has yielded new dinuclear and hexanuclear complexes. The FeCl(3)/NaO(2)CPh/L (4:4:1) reaction system in MeCN gives red-brown [Fe(6)O(4)Cl(4)(O(2)CPh)(4)L(2)][FeCl(4)](2) (1). The same reaction system in a 3:3:1 ratio in MeOH gives orange [Fe(2)(OMe)(2)Cl(2)(O(2)CPh)L][FeCl(4)] (2). Complex 1.2MeCN: monoclinic, P2(1)/a, a = 15.317(2) Å, b = 18.303(3) Å, c = 16.168(3) Å, beta = 108.91(1) degrees, and Z = 2. Complex 2: triclinic, P&onemacr;, a = 14.099(6) Å, b = 18.510(7) Å, c = 7.108(3) Å, alpha = 96.77(2) degrees, beta = 99.45(2) degrees, gamma = 81.16(2) degrees, and Z = 2. The cation of 1 consists of a near-planar [Fe(6)(&mgr;(3)-O)(4)](10+) core that can be described as three edge-fused [Fe(2)O(2)] rhombs to which are attached two additional Fe atoms. The cation of 2 contains a [Fe(2)(&mgr;-OMe)(2)(&mgr;-O(2)CPh)](3+) core. In both cations, the L group acts as a bridging ligand across an Fe(2) unit, with the bpy rings essentially parallel. Variable-temperature solid-state magnetic-susceptibility studies of 1 and 2 in the 2.00-300 K range reveal that for both complexes the data are consistent with an S = 0 cation and S = (5)/(2) [FeCl(4)](-) anions. These conclusions were confirmed by magnetization vs field studies in the 2.00-4.00 K and 10.0-50.0 kG ranges. Fitting of the data for 2 to the appropriate theoretical equation for an equimolar composition of Fe(2) cations and [FeCl(4)](-) anions allowed the exchange interaction in the cation to be determined as J = -10.5 cm(-)(1) (H = -2JS(1)S(2)) with g held at 2.00. The obtained J value is consistent with that predicted by a previously published magnetostructural relationship.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA