Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biotechnol Bioeng ; 121(5): 1716-1728, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38454640

RESUMEN

Host cell proteins (HCPs) are process-related impurities of therapeutic proteins produced in for example, Chinese hamster ovary (CHO) cells. Protein A affinity chromatography is the initial capture step to purify monoclonal antibodies or Fc-based proteins and is most effective for HCP removal. Previously proposed mechanisms that contribute to co-purification of HCPs with the therapeutic protein are either HCP-drug association or leaching from chromatin heteroaggregates. In this study, we analyzed protein A eluates of 23 Fc-based proteins by LC-MS/MS to determine their HCP content. The analysis revealed a high degree of heterogeneity in the number of HCPs identified in the different protein A eluates. Among all identified HCPs, the majority co-eluted with less than three Fc-based proteins indicating a drug-specific co-purification for most HCPs. Only ten HCPs co-purified with over 50% of the 23 Fc-based proteins. A correlation analysis of HCPs identified across multiple protein A eluates revealed their co-elution as HCP groups. Functional annotation and protein interaction analysis confirmed that some HCP groups are associated with protein-protein interaction networks. Here, we propose an additional mechanism for HCP co-elution involving protein-protein interactions within functional networks. Our findings may help to guide cell line development and to refine downstream purification strategies.


Asunto(s)
Proteína Estafilocócica A , Espectrometría de Masas en Tándem , Cricetinae , Animales , Cricetulus , Cromatografía Liquida , Células CHO , Proteína Estafilocócica A/química , Anticuerpos Monoclonales/química
2.
Biotechnol Bioeng ; 120(11): 3299-3310, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37526307

RESUMEN

Quality by Design (QbD) principles play an increasingly important role in the pharmaceutical industry. Here, we used an analytical QbD (AQbD) approach to develop a capillary electrophoresis sodium dodecyl sulfate under reducing conditions (rCE-SDS), with the aim of replacing SDS-polyacrylamide gel electrophoresis (SDS-PAGE) as release and stability test method for a commercialized monoclonal antibody product. Method development started with defining analytical method performance requirements as part of an analytical target profile, followed by a systematic risk assessment of method input parameters and their relation to defined method outputs. Based on this, design of experiments studies were performed to identify a method operable design region (MODR). The MODR could be leveraged to improve method robustness. In a bridging study, it was demonstrated that the rCE-SDS method is more sensitive than the legacy SDS-PAGE method, and a conversion factor could be established to compensate for an off-set due to the higher sensitivity, without losing the correlation to the historical data acquired with the former method. Overall, systematic application of analytical Quality by Design principles for designing and developing a new analytical method helped to elucidate the complex dependency of method outputs on its input parameters. The link of the method to product quality attributes and the definition of method performance requirements were found to be most relevant for derisking the analytical method switch, regarding impact on the control strategy.

3.
Biotechnol Bioeng ; 118(10): 3633-3639, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34241893

RESUMEN

The performance of immunoassays for the detection and quantification of host-cell proteins (HCPs) in biopharmaceuticals depends on the quality of the critical assay reagents. Not only their preparation, but also a stringent life-cycle management, including reagent qualification, requalification, and replacement, plays a crucial role in ensuring consistent and reliable results. To provide a cross-industry perspective on HCP reagent management, we conducted a survey on common practices among several pharmaceutical and biotech companies. Based on its outcome, as well as informed by a corresponding roundtable session ("Managing critical reagents over time") at the BioPharmaceutical Emerging Best Practices Association HCP conference in 2019, this study presents specific recommendations and proven concepts to support immunoassay reagent management for monitoring HCPs.


Asunto(s)
Anticuerpos Monoclonales/química , Productos Biológicos , Animales , Productos Biológicos/análisis , Productos Biológicos/química , Células CHO , Cricetulus , Ensayo de Inmunoadsorción Enzimática
4.
Anal Chem ; 90(19): 11240-11247, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30048127

RESUMEN

Biopharmaceuticals contain residual host cell protein (HCP) impurities, a complex mixture of endogenous proteins from production cell lines such as Chinese hamster ovary (CHO) cells. The composition of HCP impurities at harvest hinges on multiple factors, e.g., identity of cell line, cell density and viability at harvest, or other process parameters. Two-dimensional differential gel electrophoresis (2-D DIGE) was used to compare HCP in 15 null cell culture harvest supernatants, which are representative for a wide range of manufacturing processes of therapeutic antibodies, using five different CHO cell lines. Numerical metrics were developed to quantitatively compare HCP composition, which may be used to assess the suitability of a platform HCP assay standard for a new product or to assess the impact of process changes. A very similar HCP composition was found for the 15 analyzed CHO null cell culture harvests, demonstrating that even the wide range of applied manufacturing processes did not have a strong influence on the HCP impurities.


Asunto(s)
Proteínas/aislamiento & purificación , Animales , Células CHO , Técnicas de Cultivo de Célula , Cricetulus , Electroforesis en Gel Bidimensional , Proteínas/análisis , Proteínas/uso terapéutico
5.
Biotechnol Prog ; 30(6): 1311-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25082738

RESUMEN

Antigen-binding fragments (Fabs) are novel formats in the growing pipeline of biotherapeutics. Sharing similar features to monoclonal antibodies (mAbs) with regard to expression, Fabs are considered as unchallenging for upstream development. Yet for downstream processing, the mature mAb downstream purification platform is not directly applicable. New approaches need to be found to achieve a lean purification process that maintains quality, productivity, and timelines while being generically applicable independent of the expression system. In a successful collaboration, BAC BV, GE Healthcare, and Novartis Pharma AG have developed a new affinity chromatography medium (resin) suitable to support cGMP manufacturing of lambda Fabs. We show that using this novel chromatography medium for the capture step, a purification platform for lambda Fabs can be established.


Asunto(s)
Cromatografía de Afinidad/instrumentación , Cromatografía de Afinidad/métodos , Fragmentos Fab de Inmunoglobulinas , Químicos de Laboratorio , Animales , Células CHO , Cricetinae , Cricetulus , Escherichia coli , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fab de Inmunoglobulinas/metabolismo , Químicos de Laboratorio/química , Químicos de Laboratorio/metabolismo , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
6.
J Biol Chem ; 285(34): 26628-40, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20547770

RESUMEN

Fibroblast growth factor (FGF) signaling regulates mammalian development and metabolism, and its dysregulation is implicated in many inherited and acquired diseases, including cancer. Heparan sulfate glycosaminoglycans (HSGAGs) are essential for FGF signaling as they promote FGF.FGF receptor (FGFR) binding and dimerization. Using novel organic synthesis protocols to prepare homogeneously sulfated heparin mimetics (HM), including hexasaccharide (HM(6)), octasaccharide (HM(8)), and decasaccharide (HM(10)), we tested the ability of these HM to support FGF1 and FGF2 signaling through FGFR4. Biological assays show that both HM(8) and HM(10) are significantly more potent than HM(6) in promoting FGF2-mediated FGFR4 signaling. In contrast, all three HM have comparable activity in promoting FGF1.FGFR4 signaling. To understand the molecular basis for these differential activities in FGF1/2.FGFR4 signaling, we used NMR spectroscopy, isothermal titration calorimetry, and size-exclusion chromatography to characterize binding interactions of FGF1/2 with the isolated Ig-domain 2 (D2) of FGFR4 in the presence of HM, and binary interactions of FGFs and D2 with HM. Our data confirm the existence of both a secondary FGF1.FGFR4 interaction site and a direct FGFR4.FGFR4 interaction site thus supporting the formation of the symmetric mode of FGF.FGFR dimerization in solution. Moreover, our results show that the observed higher activity of HM(8) relative to HM(6) in stimulating FGF2.FGFR4 signaling correlates with the higher affinity of HM(8) to bind and dimerize FGF2. Notably FGF2.HM(8) exhibits pronounced positive binding cooperativity. Based on our findings we propose a refined symmetric FGF.FGFR dimerization model, which incorporates the differential ability of HM to dimerize FGFs.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Heparina/análogos & derivados , Oligosacáridos/farmacología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Sitios de Unión , Línea Celular , Humanos , Ratones , Complejos Multiproteicos/biosíntesis , Oligosacáridos/química , Unión Proteica , Multimerización de Proteína , Relación Estructura-Actividad
7.
J Biol Chem ; 285(1): 502-10, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19892700

RESUMEN

We previously proposed that the dimeric cytochrome bc(1) complex exhibits half-of-the-sites reactivity for ubiquinol oxidation and rapid electron transfer between bc(1) monomers (Covian, R., Kleinschroth, T., Ludwig, B., and Trumpower, B. L. (2007) J. Biol. Chem. 282, 22289-22297). Here, we demonstrate the previously proposed half-of-the-sites reactivity and intermonomeric electron transfer by characterizing the kinetics of ubiquinol oxidation in the dimeric bc(1) complex from Paracoccus denitrificans that contains an inactivating Y147S mutation in one or both cytochrome b subunits. The enzyme with a Y147S mutation in one cytochrome b subunit was catalytically fully active, whereas the activity of the enzyme with a Y147S mutation in both cytochrome b subunits was only 10-16% of that of the enzyme with fully wild-type or heterodimeric cytochrome b subunits. Enzyme with one inactive cytochrome b subunit was also indistinguishable from the dimer with two wild-type cytochrome b subunits in rate and extent of reduction of cytochromes b and c(1) by ubiquinol under pre-steady-state conditions in the presence of antimycin. However, the enzyme with only one mutated cytochrome b subunit did not show the stimulation in the steady-state rate that was observed in the wild-type dimeric enzyme at low concentrations of antimycin, confirming that the half-of-the-sites reactivity for ubiquinol oxidation can be regulated in the wild-type dimer by binding of inhibitor to one ubiquinone reduction site.


Asunto(s)
Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/metabolismo , Multimerización de Proteína , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo , Animales , Antimicina A/análogos & derivados , Antimicina A/farmacología , Sitios de Unión , Cromatografía de Afinidad , Activación Enzimática/efectos de los fármacos , Caballos , Cinética , Ligandos , Mutagénesis/efectos de los fármacos , Mutagénesis/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Operón/genética , Oxidación-Reducción/efectos de los fármacos , Paracoccus denitrificans/efectos de los fármacos , Paracoccus denitrificans/enzimología , Paracoccus denitrificans/genética , Multimerización de Proteína/efectos de los fármacos , Volumetría
8.
J Biol Chem ; 284(38): 25735-41, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19625251

RESUMEN

Biogenesis of mitochondrial cytochrome c oxidase (COX) relies on a large number of assembly factors, among them the transmembrane protein Surf1. The loss of human Surf1 function is associated with Leigh syndrome, a fatal neurodegenerative disorder caused by severe COX deficiency. In the bacterium Paracoccus denitrificans, two homologous proteins, Surf1c and Surf1q, were identified, which we characterize in the present study. When coexpressed in Escherichia coli together with enzymes for heme a synthesis, the bacterial Surf1 proteins bind heme a in vivo. Using redox difference spectroscopy and isothermal titration calorimetry, the binding of the heme cofactor to purified apo-Surf1c and apo-Surf1q is quantified: Each of the Paracoccus proteins binds heme a in a 1:1 stoichiometry and with Kd values in the submicromolar range. In addition, we identify a conserved histidine as a residue crucial for heme binding. Contrary to most earlier concepts, these data support a direct role of Surf1 in heme a cofactor insertion into COX subunit I by providing a protein-bound heme a pool.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/química , Complejo IV de Transporte de Electrones/biosíntesis , Complejo IV de Transporte de Electrones/química , Hemo/química , Hemo/metabolismo , Enfermedad de Leigh/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Paracoccus denitrificans/enzimología , Humanos , Enfermedad de Leigh/genética , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Paracoccus denitrificans/genética , Unión Proteica/genética
9.
J Biol Chem ; 283(46): 31333-40, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18809676

RESUMEN

Glucokinase (GK) is a key enzyme of glucose metabolism in liver and pancreatic beta-cells, and small molecule activators of GK (GKAs) are under evaluation for the treatment of type 2 diabetes. In liver, GK activity is controlled by the GK regulatory protein (GKRP), which forms an inhibitory complex with the enzyme. Here, we performed isothermal titration calorimetry and surface plasmon resonance experiments to characterize GK-GKRP binding and to study the influence that physiological and pharmacological effectors of GK have on the protein-protein interaction. In the presence of fructose-6-phosphate, GK-GKRP complex formation displayed a strong entropic driving force opposed by a large positive enthalpy; a negative change in heat capacity was observed (Kd = 45 nm, DeltaH = 15.6 kcal/mol, TDeltaS = 25.7 kcal/mol, DeltaCp = -354 cal mol(-1) K(-1)). With k(off) = 1.3 x 10(-2) s(-1), the complex dissociated quickly. The thermodynamic profile suggested a largely hydrophobic interaction. In addition, effects of pH and buffer demonstrated the coupled uptake of one proton and indicated an ionic contribution to binding. Glucose decreased the binding affinity between GK and GKRP. This decrease was potentiated by an ATP analogue. Prototypical GKAs of the amino-heteroaryl-amide type bound to GK in a glucose-dependent manner and impaired the association of GK with GKRP. This mechanism might contribute to the antidiabetic effects of GKAs.


Asunto(s)
Biofisica , Proteínas Portadoras/metabolismo , Glucoquinasa/metabolismo , Hígado/enzimología , Animales , Tampones (Química) , Proteínas Portadoras/genética , Proteínas Portadoras/aislamiento & purificación , Glucoquinasa/genética , Glucoquinasa/aislamiento & purificación , Humanos , Concentración de Iones de Hidrógeno , Cinética , Unión Proteica , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie , Temperatura , Termodinámica
10.
FEBS J ; 275(19): 4773-85, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18721136

RESUMEN

The protonation state of residues around the Q(o) binding site of the cytochrome bc(1) complex from Paracoccus denitrificans and their interaction with bound quinone(s) was studied by a combined electrochemical and FTIR difference spectroscopic approach. Site-directed mutations of two groups of conserved residues were investigated: (a) acidic side chains located close to the surface and thought to participate in a water chain leading up to the heme b(L) edge, and (b) residues located in the vicinity of this site. Interestingly, most of the mutants retain a high degree of catalytic activity. E295Q, E81Q and Y297F showed reduced stigmatellin affinity. On the basis of electrochemically induced FTIR difference spectra, we suggest that E295 and D278 are protonated in the oxidized form or that their mutation perturbs protonated residues. Mutations Y302, Y297, E81 and E295, directly perturb signals from the oxidized quinone and of the protein backbone. By monitoring the interaction with the inhibitor stigmatellin for the wild-type enzyme at various redox states, interactions of the bound stigmatellin with amino acid side chains such as protonated acidic residues and the backbone were observed, as well as difference signals arising from the redox active inhibitor itself and the replaced quinone. The infrared difference spectra of the above Q(o) site mutations in the presence of stigmatellin confirm the previously established role of E295 as a direct interaction partner in the enzyme from P.denitrificans as well. The protonated residue E295 is proposed to change the hydrogen-bonding environment upon stigmatellin binding in the oxidized form, and is deprotonated in the reduced form. Of the residues located close to the surface, D278 remains protonated and unperturbed in the oxidized form but its frequency shifts in the reduced form. The mechanistic implications of our observations are discussed, together with previous inhibitor binding data, and referred to the published X-ray structures.


Asunto(s)
Complejo III de Transporte de Electrones/genética , Paracoccus denitrificans/metabolismo , Sitios de Unión/genética , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Polienos/farmacología , Espectroscopía Infrarroja por Transformada de Fourier , Tirosina/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
11.
Biochemistry ; 47(16): 4683-91, 2008 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-18373353

RESUMEN

Glycogen phosphorylase (GP) is a validated target for the treatment of type 2 diabetes. Here we describe highly potent GP inhibitors, AVE5688, AVE2865, and AVE9423. The first two compounds are optimized members of the acyl urea series. The latter represents a novel quinolone class of GP inhibitors, which is introduced in this study. In the enzyme assay, both inhibitor types compete with the physiological activator AMP and act synergistically with glucose. Isothermal titration calorimetry (ITC) shows that the compounds strongly bind to nonphosphorylated, inactive GP (GPb). Binding to phosphorylated, active GP (GPa) is substantially weaker, and the thermodynamic profile reflects a coupled transition to the inactive (tense) conformation. Crystal structures confirm that the three inhibitors bind to the AMP site of tense state GP. These data provide the first direct evidence that acyl urea and quinolone compounds are allosteric inhibitors that selectively bind to and stabilize the inactive conformation of the enzyme. Furthermore, ITC reveals markedly different thermodynamic contributions to inhibitor potency that can be related to the binding modes observed in the cocrystal structures. For AVE5688, which occupies only the lower part of the bifurcated AMP site, binding to GPb (Kd = 170 nM) is exclusively enthalpic (Delta H = -9.0 kcal/mol, TDelta S = 0.3 kcal/mol). The inhibitors AVE2865 (Kd = 9 nM, Delta H = -6.8 kcal/mol, TDelta S = 4.2 kcal/mol) and AVE9423 (Kd = 24 nM, Delta H = -5.9 kcal/mol, TDelta S = 4.6 kcal/mol) fully exploit the volume of the binding pocket. Their pronounced binding entropy can be attributed to the extensive displacement of solvent molecules as well as to ionic interactions with the phosphate recognition site.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glucógeno Fosforilasa/antagonistas & inhibidores , Glucógeno Fosforilasa/metabolismo , Termodinámica , Regulación Alostérica/efectos de los fármacos , Animales , Tampones (Química) , Calorimetría , Glucosa/metabolismo , Glucógeno Fosforilasa/química , Cinética , Modelos Moleculares , Estructura Molecular , Unión Proteica , Conejos , Temperatura , Volumetría
12.
FEBS J ; 273(21): 4817-30, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16987314

RESUMEN

The Rieske [2Fe-2S] protein (ISP) is an essential subunit of cytochrome bc(1) complexes in mitochondrial and bacterial respiratory chains. Based on the presence of two consecutive arginines, it was argued that the ISP of Paracoccus denitrificans, a Gram-negative soil bacterium, is inserted into the cytoplasmic membrane via the twin-arginine translocation (Tat) pathway. Here, we provide experimental evidence that membrane integration of the bacterial ISP indeed relies on the Tat translocon. We show that targeting of the ISP depends on the twin-arginine motif. A strict requirement is established particularly for the second arginine residue (R16); conservative replacement of the first arginine (R15K) still permits substantial ISP transport. Comparative sequence analysis reveals characteristics common to Tat signal peptides in several bacterial ISPs; however, there are distinctive features relating to the fact that the presumed ISP Tat signal simultaneously serves as a membrane anchor. These differences include an elevated hydrophobicity of the h-region compared with generic Tat signals and the absence of an otherwise well-conserved '+5'-consensus motif lysine residue. Substitution of the +5 lysine (Y20K) compromises ISP export and/or cytochrome bc(1) stability to some extent and points to a specific role for this deviation from the canonical Tat motif. EPR spectroscopy confirms cytosolic insertion of the [2Fe-2S] cofactor. Mutation of an essential cofactor binding residue (C152S) decreases the ISP membrane levels, possibly indicating that cofactor insertion is a prerequisite for efficient translocation along the Tat pathway.


Asunto(s)
Arginina/metabolismo , Proteínas Bacterianas/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Membranas Intracelulares/metabolismo , Proteínas Hierro-Azufre/metabolismo , Proteínas de Transporte de Membrana/fisiología , Paracoccus denitrificans/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Complejo III de Transporte de Electrones/genética , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas Hierro-Azufre/genética , Datos de Secuencia Molecular , Señales de Clasificación de Proteína , Transporte de Proteínas , Esferoplastos/metabolismo
13.
J Biol Chem ; 279(6): 5000-7, 2004 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-14610094

RESUMEN

Stable supercomplexes of bacterial respiratory chain complexes III (ubiquinol:cytochrome c oxidoreductase) and IV (cytochrome c oxidase) have been isolated as early as 1985 (Berry, E. A., and Trumpower, B. L. (1985) J. Biol. Chem. 260, 2458-2467). However, these assemblies did not comprise complex I (NADH:ubiquinone oxidoreductase). Using the mild detergent digitonin for solubilization of Paracoccus denitrificans membranes we could isolate NADH oxidase, assembled from complexes I, III, and IV in a 1:4:4 stoichiometry. This is the first chromatographic isolation of a complete "respirasome." Inactivation of the gene for tightly bound cytochrome c552 did not prevent formation of this supercomplex, indicating that this electron carrier protein is not essential for structurally linking complexes III and IV. Complex I activity was also found in the membranes of mutant strains lacking complexes III or IV. However, no assembled complex I but only dissociated subunits were observed following the same protocols used for electrophoretic separation or chromatographic isolation of the supercomplex from the wild-type strain. This indicates that the P. denitrificans complex I is stabilized by assembly into the NADH oxidase supercomplex. In addition to substrate channeling, structural stabilization of a membrane protein complex thus appears as one of the major functions of respiratory chain supercomplexes.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/química , Paracoccus denitrificans/enzimología , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/aislamiento & purificación , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Estabilidad de Enzimas , Genes Bacterianos , Cinética , Peso Molecular , Complejos Multienzimáticos/química , Complejos Multienzimáticos/metabolismo , NADH NADPH Oxidorreductasas/química , NADH NADPH Oxidorreductasas/metabolismo , Paracoccus denitrificans/genética
14.
Biochemistry ; 42(42): 12391-9, 2003 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-14567700

RESUMEN

The cytochrome bc(1) complex from Paracoccus denitrificans and soluble fragments of its cytochrome c(1) and Rieske ISP subunits are characterized by a combined approach of protein electrochemistry and FTIR difference spectroscopy. The FTIR difference spectra provide information about alterations in the protein upon redox reactions: signals from the polypeptide backbone, from the cofactors, and from amino acid side chains. We describe typical modes for conformational changes in the polypeptide and contributions of different secondary structure elements. Signals attributed to the different cofactors can be presented on the basis of selected potential steps. Modes associated with bound quinone are identified by comparison with spectra of quinone in solution at 1656, 1642, and 1610 cm(-1) and between 1494 and 1388 cm(-1), as well as at 1288 and 1262 cm(-1). Signals originating from the quinone bound at the Q(o) site can be distinguished. On the basis of the infrared data, the total quinone concentration is determined to be 2.6-3.3 quinones per monomer, depending on preparation conditions. The balance of evidence supports the double-occupancy model. Interestingly, the amplitude of the band at 1746 cm(-1) increases with quinone content, reflecting a protonation reaction of acidic groups. In this context, the involvement of glutamates and/or aspartates in the vicinity of the Q(o) site is discussed on the basis of recently determined crystal structures.


Asunto(s)
Benzoquinonas/metabolismo , Electroquímica/métodos , Complejo III de Transporte de Electrones/metabolismo , Paracoccus denitrificans/enzimología , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Complejo III de Transporte de Electrones/química , Protones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...