Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biotechnol Biofuels ; 10: 128, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28523076

RESUMO

BACKGROUND: Hitherto, the main goal of metaproteomic analyses has been to characterize the functional role of particular microorganisms in the microbial ecology of various microbial communities. Recently, it has been suggested that metaproteomics could be used for bioprospecting microbial communities to query for the most active enzymes to improve the selection process of industrially relevant enzymes. In the present study, to reduce the complexity of metaproteomic samples for targeted bioprospecting of novel enzymes, a microbial community capable of producing cellulases was maintained on a chemically defined medium in an enzyme suppressed metabolic steady state. From this state, it was possible to specifically and distinctively induce the desired cellulolytic activity. The extracellular fraction of the protein complement of the induced sample could thereby be purified and compared to a non-induced sample of the same community by differential gel electrophoresis to discriminate between constitutively expressed proteins and proteins upregulated in response to the inducing substance. RESULTS: Using the applied approach, downstream analysis by mass spectrometry could be limited to only proteins recognized as upregulated in the cellulase-induced sample. Of 39 selected proteins, the majority were found to be linked to the need to degrade, take up, and metabolize cellulose. In addition, 28 (72%) of the proteins were non-cytosolic and 17 (44%) were annotated as carbohydrate-active enzymes. The results demonstrated both the applicability of the proposed approach for identifying extracellular proteins and guiding the selection of proteins toward those specifically upregulated and targeted by the enzyme inducing substance. Further, because identification of interesting proteins was based on the regulation of enzyme expression in response to a need to hydrolyze and utilize a specific substance, other unexpected enzyme activities were able to be identified. CONCLUSIONS: The described approach created the conditions necessary to be able to select relevant extracellular enzymes that were extracted from the enzyme-induced microbial community. However, for the purpose of bioprospecting for enzymes to clone, produce, and characterize for practical applications, it was concluded that identification against public databases was not sufficient to identify the correct gene or protein sequence for cloning of the identified novel enzymes.

2.
Anal Biochem ; 516: 23-36, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27742212

RESUMO

Enzyme discovery in individual strains of microorganisms is compromised by the limitations of pure culturing. In principle, metaproteomics allows for fractionation and study of different parts of the protein complement but has hitherto mainly been used to identify intracellular proteins. However, the extracellular environment is also expected to comprise a wealth of information regarding important proteins. An absolute requirement for metaproteomic studies of protein expression, and irrespective of downstream methods for analysis, is that sample preparation methods provide clean, concentrated and representative samples of the protein complement. A battery of methods for concentration, extraction, precipitation and resolubilization of proteins in the extracellular environment of a constructed microbial community was assessed by means of 2D gel electrophoresis and image analysis to elucidate whether it is possible to make the extracellular protein complement available for metaproteomic analysis. Most methods failed to provide pure samples and therefore negatively influenced protein gel migration and gel background clarity. However, one direct precipitation method (TCA-DOC/acetone) and one extraction/precipitation method (phenol/methanol) provided complementary high quality 2D gels that allowed for high spot detection ability and thereby also spot detection of less abundant extracellular proteins.


Assuntos
Bactérias/química , Proteínas de Bactérias/química , Proteômica/métodos , Manejo de Espécimes/métodos , Eletroforese em Gel Bidimensional/métodos
3.
Sci Rep ; 6: 28386, 2016 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-27328749

RESUMO

Here, we study and compare the mechanisms of action of the GroEL/GroES and the TRiC chaperonin systems on MreB client protein variants extracted from E. coli. MreB is a homologue to actin in prokaryotes. Single-molecule fluorescence correlation spectroscopy (FCS) and time-resolved fluorescence polarization anisotropy report the binding interaction of folding MreB with GroEL, GroES and TRiC. Fluorescence resonance energy transfer (FRET) measurements on MreB variants quantified molecular distance changes occurring during conformational rearrangements within folding MreB bound to chaperonins. We observed that the MreB structure is rearranged by a binding-induced expansion mechanism in TRiC, GroEL and GroES. These results are quantitatively comparable to the structural rearrangements found during the interaction of ß-actin with GroEL and TRiC, indicating that the mechanism of chaperonins is conserved during evolution. The chaperonin-bound MreB is also significantly compacted after addition of AMP-PNP for both the GroEL/ES and TRiC systems. Most importantly, our results showed that GroES may act as an unfoldase by inducing a dramatic initial expansion of MreB (even more than for GroEL) implicating a role for MreB folding, allowing us to suggest a delivery mechanism for GroES to GroEL in prokaryotes.


Assuntos
Chaperonina 10/metabolismo , Chaperonina 60/metabolismo , Chaperonina com TCP-1/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Actinas/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Transferência Ressonante de Energia de Fluorescência , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Imagem Individual de Molécula
4.
PLoS One ; 8(2): e56874, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23526922

RESUMO

Circular Dichroism (CD) spectroscopy is a powerful method for investigating conformational changes in proteins and therefore has numerous applications in structural and molecular biology. Here a computational investigation of the CD spectrum of the Human Carbonic Anhydrase II (HCAII), with main focus on the near-UV CD spectra of the wild-type enzyme and it seven tryptophan mutant forms, is presented and compared to experimental studies. Multilevel computational methods (Molecular Dynamics, Semiempirical Quantum Mechanics, Time-Dependent Density Functional Theory) were applied in order to gain insight into the mechanisms of interaction between the aromatic chromophores within the protein environment and understand how the conformational flexibility of the protein influences these mechanisms. The analysis suggests that combining CD semi empirical calculations, crystal structures and molecular dynamics (MD) could help in achieving a better agreement between the computed and experimental protein spectra and provide some unique insight into the dynamic nature of the mechanisms of chromophore interactions.


Assuntos
Anidrase Carbônica II/química , Dicroísmo Circular/métodos , Biologia Computacional/métodos , Modelos Moleculares , Conformação Proteica , Cristalização , Humanos , Isoenzimas/química , Simulação de Dinâmica Molecular
5.
J Chem Biol ; 7(1): 1-15, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386013

RESUMO

The commonly accepted dogma of the bacterial GroE chaperonin system entails protein folding mediated by cycles of several ATP-dependent sequential steps where GroEL interacts with the folding client protein. In contrast, we herein report GroES-mediated dynamic remodeling (expansion and compression) of two different protein substrates during folding: the endogenous substrate MreB and carbonic anhydrase (HCAII), a well-characterized protein folding model. GroES was also found to influence GroEL binding induced unfolding and compression of the client protein underlining the synergistic activity of both chaperonins, even in the absence of ATP. This previously unidentified activity by GroES should have important implications for understanding the chaperonin mechanism and cellular stress response. Our findings necessitate a revision of the GroEL/ES mechanism.

6.
Biochem Biophys Res Commun ; 413(4): 545-9, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21924243

RESUMO

The apparent length of FVIIa in solution was estimated by a FRET analysis. Two fluorescent probes, fluorescein (Fl-FPR) and a rhodamine derivative (TMR), were covalently attached to FVIIa. The binding site of Fl-FPR was in the protease domain whereas TMR was positioned in the Gla domain, thus allowing a length measure over virtually the whole extension of the protein. From the FRET measurements, the distances between the two probes were determined to be 61.4 for free FVIIa and 65.5Å for FVIIa bound to soluble tissue factor (sTF). These seemingly short distances, compared to those anticipated based on the complex crystal structure, require that the probes stretch towards each other. Thus, the apparent distance from the FRET analysis was shown to increase with 4Å upon formation of a complex with sTF in solution. However, considering how protein dynamics, based on recent molecular dynamics simulations of FVIIa and sTF:FVIIa (Y.Z. Ohkubo, J.H. Morrissey, E. Tajkhorshid, J. Thromb. Haemost. 8 (2010) 1044-1053), can influence the apparent fluorescence signal our calculations indicated that the global average conformation of active-site inhibited FVIIa is nearly unaltered upon ligation to sTF. It is known from amidolytic activity measurements that Ca(2+) binding leads to activation of FVIIa, but we have for the first time directly demonstrated conformational changes in the environment of the active site upon Ca(2+) binding. Interestingly, this Ca(2+)-induced conformational change can be noted even in the presence of an inhibitor. Forming a complex with sTF further stabilized this conformational change, leading to a more inaccessible active-site located probe.


Assuntos
Cálcio/química , Fator VIIa/química , Tromboplastina/química , Substituição de Aminoácidos , Domínio Catalítico , Ativação Enzimática , Fator VIIa/genética , Transferência Ressonante de Energia de Fluorescência , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína
7.
Biochemistry ; 49(6): 1137-45, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20070121

RESUMO

The single-domain cyclophilin 18 (Cyp18) has long been known to function as a peptidyl-prolyl cis/trans isomerase (PPI) and was proposed by us to also function as a chaperone [Freskgard, P.-O., Bergenhem, N., Jonsson, B.-H., Svensson, M., and Carlsson, U. (1992) Science 258, 466-468]. Later several multidomain PPIs were demonstrated to work as both a peptidyl-prolyl cis/trans isomerase and a chaperone. However, the chaperone ability of Cyp18 has been debated. In this work, we add additional results that show that Cyp18 can both accelerate the rate of refolding and increase the yield of native protein during the folding reaction, i.e., function as both a folding catalyst and a chaperone. Refolding experiments were performed using severely destabilized mutants of human carbonic anhydrase II under conditions where the unfolding reaction is significant and a larger fraction of a more destabilized variant populates molten globule-like intermediates during refolding. A correlation of native state protein stability of the substrate protein versus Cyp18 chaperone activity was demonstrated. The induced correction of misfolded conformations by Cyp18 likely functions through rescue from misfolding of transient molten globule intermediates. ANS binding data suggest that the interaction by Cyp18 leads to an early stage condensation of accessible hydrophobic portions of the misfolding-prone protein substrate during folding. The opposite effect was observed for GroEL known as an unfoldase at early stages of refolding. The chaperone effect of Cyp18 was also demonstrated for citrate synthase, suggesting a general chaperone effect of this PPI.


Assuntos
Ciclofilinas/química , Ciclofilinas/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Dobramento de Proteína , Naftalenossulfonato de Anilina/química , Naftalenossulfonato de Anilina/metabolismo , Anidrase Carbônica II/química , Anidrase Carbônica II/genética , Anidrase Carbônica II/metabolismo , Ciclofilinas/fisiologia , Estabilidade Enzimática , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Chaperonas Moleculares/fisiologia , Peso Molecular , Mutagênese Sítio-Dirigida , Desnaturação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Regulação para Cima
8.
J Chem Biol ; 3(3): 127-39, 2010 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-21479077

RESUMO

The Hsp60-type chaperonin GroEL assists in the folding of the enzyme human carbonic anhydrase II (HCA II) and protects it from aggregation. This study was aimed to monitor conformational rearrangement of the substrate protein during the initial GroEL capture (in the absence of ATP) of the thermally unfolded HCA II molten-globule. Single- and double-cysteine mutants were specifically spin-labeled at a topological breakpoint in the ß-sheet rich core of HCA II, where the dominating antiparallel ß-sheet is broken and ß-strands 6 and 7 are parallel. Electron paramagnetic resonance (EPR) was used to monitor the GroEL-induced structural changes in this region of HCA II during thermal denaturation. Both qualitative analysis of the EPR spectra and refined inter-residue distance calculations based on magnetic dipolar interaction show that the spin-labeled positions F147C and K213C are in proximity in the native state of HCA II at 20 °C (as close as ∼8 Å), and that this local structure is virtually intact in the thermally induced molten-globule state that binds to GroEL. In the absence of GroEL, the molten globule of HCA II irreversibly aggregates. In contrast, a substantial increase in spin-spin distance (up to >20 Å) was observed within minutes, upon interaction with GroEL (at 50 and 60 °C), which demonstrates a GroEL-induced conformational change in HCA II. The GroEL binding-induced disentanglement of the substrate protein core at the topological break-point is likely a key event for rearrangement of this potent aggregation initiation site, and hence, this conformational change averts HCA II misfolding.

9.
Biochemistry ; 48(23): 5358-64, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19415900

RESUMO

Carbonic anhydrase II deficiency syndrome or Marble brain disease (MBD) is caused by autosomal recessive mutations in the human carbonic anhydrase II (HCA II) gene. Here we report a small-molecule stabilization study of the exceptionally destabilized HCA II mutant H107Y employing inhibitors based on p-aminobenzoylsulfonamide compounds and 1,3,4-thiadiazolylsulfonamides as well as amino acid activators. Protein stability assays showed a significant stabilization by the aromatic sulfonamide inhibitors when present at 10 microM concentration, providing shifts of the midpoint of thermal denaturation between 10 degrees C and 16 degrees C and increasing the free energies of denaturation 0.5-3.0 kcal/mol as deduced from GuHCl denaturation. This study could be used as a starting point for the design of small-molecule folding modulators and possibly autoactivatable molecules for suppression of misfolding of destabilized HCA II mutants.


Assuntos
Encefalopatias Metabólicas Congênitas/genética , Anidrase Carbônica II/química , Anidrase Carbônica II/genética , Inibidores da Anidrase Carbônica/farmacologia , Anidrase Carbônica II/antagonistas & inibidores , Humanos , Cinética , Modelos Moleculares , Conformação Proteica , Desnaturação Proteica , Dobramento de Proteína , Sulfonamidas/química , Sulfonamidas/farmacologia , Termodinâmica
10.
Protein Sci ; 18(2): 475-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19185003

RESUMO

The protein folding process is often in vitro rate-limited by slow cis-trans proline isomerization steps. Importantly, the rate of this process in vivo is accelerated by prolyl isomerases (PPIases). The archetypal PPIase is the human cyclophilin 18 (Cyp18 or CypA), and Arg 55 has been demonstrated to play a crucial role when studying short peptide substrates in the catalytic action of Cyp18 by stabilizing the transition state of isomerization. However, in this study we show that a R55A mutant of Cyp18 is as efficient as the wild type to accelerate the refolding reaction of human carbonic anhydrase II (HCA II). Thus, it is evident that the active-site located Arg 55 is not required for catalysis of the rate-limiting prolyl cis-trans isomerization steps during the folding of a protein substrate as HCA II. Nevertheless, catalysis of cis-trans proline isomerization in HCA II occurs in the active-site of Cyp18, since binding of the inhibitor cyclosporin A abolishes rate acceleration of the refolding reaction. Obviously, the catalytic mechanisms of Cyp18 can differ when acting upon a simple model peptide, four residues long, with easily accessible Pro residues compared with a large protein molecule undergoing folding with partly or completely buried Pro residues. In the latter case, the isomerization kinetics are significantly slower and simpler mechanistic factors such as desolvation and/or strain might operate during folding-assisted catalysis, since binding to the hydrophobic active site is still a prerequisite for catalysis.


Assuntos
Arginina/fisiologia , Ciclofilina A/química , Ciclofilina A/metabolismo , Dobramento de Proteína , Anidrase Carbônica II/metabolismo , Domínio Catalítico , Ciclofilina A/genética , Ciclosporina , Escherichia coli/metabolismo , Humanos , Isomerismo , Cinética , Mutação , Peptídeos/metabolismo , Engenharia de Proteínas
11.
Biochemistry ; 47(5): 1288-98, 2008 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-18189416

RESUMO

UNLABELLED: Marble brain disease (MBD) also known as Guibaud-Vainsel syndrome is caused by autosomal recessive mutations in the human carbonic anhydrase II (HCA II) gene. HCA II is a 259 amino acid single domain enzyme and is dominated by a 10-stranded beta-sheet. One mutation associated with MBD entails the H107Y substitution where H107 is a highly conserved residue in the carbonic anhydrase protein family. We have previously demonstrated that the H107Y mutation is a remarkably destabilizing folding mutation [Almstedt et al. (2004) J. Mol. Biol. 342, 619-633]. Here, the exceptional destabilization by the H107Y mutation has been further investigated. A mutational survey of position H107 and a neighboring conserved position E117 has been performed entailing the mutants H107A, H107F, H107N, E117A and the double mutants H107A/E117A and H107N/E117A. All mutants were severely destabilized versus GuHCl and heat denaturation. Thermal denaturation and GuHCl phase diagram and ANS analyses showed that the mutants shifted HCA II toward populating ensembles of intermediates of molten globule type under physiological conditions. The native state stability of the mutants was in the following order: wt > H107N > E117A > H107A > H107F > H107Y > H107N/E117A > H107A/E117A. IN CONCLUSION: (i) H107N is least destabilizing likely due to compensatory H-bonding ability of the introduced Asn residue. (ii) Double mutant cycles surprisingly reveal additive destabilization of H107N and E117A showing that H107 and E117 are independently stabilizing the folded protein. (iii) H107Y and H107F are exceptionally destabilizing due to bulkiness of the side chains whereas H107A is more accommodating, indicating long-range destabilizing effects of the natural pathogenic H107Y mutation.


Assuntos
Encefalopatias Metabólicas Congênitas/genética , Anidrase Carbônica II/genética , Acetazolamida/farmacologia , Substituição de Aminoácidos , Encefalopatias Metabólicas Congênitas/enzimologia , Encefalopatias Metabólicas Congênitas/metabolismo , Anidrase Carbônica II/antagonistas & inibidores , Inibidores da Anidrase Carbônica/farmacologia , Estabilidade Enzimática , Guanidina/farmacologia , Temperatura Alta , Humanos , Modelos Moleculares , Mutação , Desnaturação Proteica , Dobramento de Proteína , Estrutura Quaternária de Proteína , Espectrometria de Fluorescência , Termodinâmica
12.
Biochem Biophys Res Commun ; 349(3): 1111-6, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-16970919

RESUMO

Blood coagulation is triggered by the formation of a complex between factor VIIa (FVIIa) and its cofactor, tissue factor (TF). The gamma-carboxyglutamic acid-rich domain of FVIIa docks with the C-terminal domain of TF, the EGF1 domain of FVIIa contacts both domains of TF, and the EGF2 domain and protease domain (PD) form a continuous surface that sits on the N-terminal domain of TF. Our aim was to investigate the conformational changes that occur in the sTF.PD binding region when different types of inhibitors, i.e., one active-site inhibitor (FFR-chloromethyl ketone (FFR)), two different peptide exosite inhibitors (E-76 and A-183), and the natural inhibitor tissue factor pathway inhibitor (TFPI), were allowed to bind to FVIIa. For this purpose, we constructed two sTF mutants (Q37C and E91C). By the aid of site-directed labeling technique, a fluorescent label was attached to the free cysteine. The sTF.PD interface was affected in position 37 by the binding of FFR, TFPI, and E-76, i.e., a more compact structure was sensed by the probe, while for position 91 located in the same region no change in the surrounding structure was observed. Thus, the active site inhibitors FFR and TFPI, and the exosite inhibitor E-76 have similar effects on the probe in position 37 of sTF, despite their differences in size and inhibition mechanism. The allosteric changes at the active site caused by binding of the exosite inhibitor E-76 in turn induce similar conformational changes in the sTF.PD interface as does the binding of the active site inhibitors. A-183, on the other hand, did not affect position 37 in sTF, indicating that the A-183 inhibition mechanism is different from that of E-76.


Assuntos
Fator VIIa/química , Fator VIIa/metabolismo , Tromboplastina/química , Tromboplastina/metabolismo , Sítios de Ligação , Fator VIIa/antagonistas & inibidores , Modelos Moleculares , Estrutura Terciária de Proteína , Espectrometria de Fluorescência
13.
J Dermatol Sci ; 43(1): 49-56, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16621453

RESUMO

BACKGROUND: Hepatocyte growth factor (HGF) is a potent regenerative factor involved in wound healing. Previous studies have shown that mesenchymal cells produce HGF, stimulating epithelial cells in a paracrine fashion. OBJECTIVE: To examine whether autocrine HGF production by keratinocytes can occur upon skin injury. METHODS: A 31-year-old male patient sustained a burn affecting 80% of his total body surface area. Biopsies were taken from intact skin near the injured area, and skin keratinocytes were separated and cultured. Conditioned medium from keratinocytes was analyzed for HGF by ELISA, surface plasmon resonance (SPR), and dot blotting. Binding of HGF from conditioned medium to its receptor, c-Met, was compared with recombinant HGF by SPR. Finally, we examined the motogenic effect on mouse transformed skin epithelial cells (CCL-53.1) of HGF from conditioned medium. RESULTS: HGF was detected in the cultured keratinocyte medium. Similar to recombinant HGF, HGF from conditioned medium had a high affinity for dextran sulfate and albumin, and the same epitopes were engaged by the interaction of HGF with the c-Met receptor. The conditioned medium from keratinocytes obtained from the burn patient, but not medium from keratinocytes obtained from healthy volunteers, accelerated the motogenesis of CCL-53.1 cells. Unexpectedly, anti-HGF antibodies did not prevent this effect. However, anti-c-Met antibodies completely inhibited the motogenic effect. CONCLUSION: Upon injury, human skin keratinocytes might produce biologically active HGF in an autocrine fashion. This HGF might have different structural and/or biological properties from HGF produced by mesenchymal cells.


Assuntos
Fator de Crescimento de Hepatócito/biossíntese , Pele/lesões , Pele/metabolismo , Adulto , Animais , Comunicação Autócrina , Células Cultivadas , Meios de Cultivo Condicionados , Humanos , Técnicas In Vitro , Queratinócitos/metabolismo , Masculino , Camundongos , Modelos Biológicos , Proteínas Proto-Oncogênicas c-met/metabolismo
14.
Biochem Biophys Res Commun ; 337(4): 1276-82, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16236263

RESUMO

Vessel wall tissue factor (TF) is exposed to blood upon vascular damage which enables association with factor VIIa (FVIIa). This leads to initiation of the blood coagulation cascade through localization and allosteric induction of FVIIa procoagulant activity. To examine the docking pathway of the FVIIa-TF complex, various residues in the extracellular part of TF (sTF) that are known to interact with FVIIa were replaced with cysteines labelled with a fluorescent probe. By using stopped-flow fluorescence kinetic measurements in combination with surface plasmon resonance analysis, we studied the association of the resulting sTF variants with FVIIa. We found the docking trajectory to be a sequence of events in which the protease domain of FVIIa initiates contact with sTF. Thereafter, the two proteins are tethered via the first epidermal growth factor-like and finally the gamma-carboxyglutamic acid (Gla) domain. The two labelled sTF residues interacting with the protease domain of FVIIa bind or become eventually ordered at different rates, revealing kinetic details pertinent to the allosteric activation of FVIIa by sTF. Moreover, when the Gla domain of FVIIa is removed the difference in the rate of association for the remaining domains is much more pronounced.


Assuntos
Fator VIIa/química , Fator VIIa/metabolismo , Tromboplastina/metabolismo , Fator VIIa/genética , Humanos , Modelos Moleculares , Mutação/genética , Ligação Proteica , Estrutura Terciária de Proteína , Espectrometria de Fluorescência , Tromboplastina/química , Tromboplastina/genética
15.
Biochemistry ; 44(18): 6755-63, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15865421

RESUMO

Tissue factor (TF), a small transmembrane receptor, binds factor VIIa (FVIIa), and the formed complex initiates blood coagulation by proteolytic activation of substrate factors IX and X. A naturally occurring mutation in the human TF gene was recently reported, where a single-base substitution results in an R200W mutation in the TF extracellular domain [Zawadzki, C., Preudhomme, C., Gaveriaux, V., Amouyel, P., and Jude, B. (2002) Thromb. Haemost. 87, 540-541]. This mutation appears to be associated with low monocyte TF expression and may protect against thrombosis but has not been associated with any pathological condition, and individuals who present the heterozygous trait appear healthy. Here, we report the activity, folding, and aggregation behavior of the R200W mutant of the 219-residue soluble extracellular domain of TF (sTF(R200W)) compared to that of the wild-type protein (sTF(wt)). No differences in stability or FVIIa cofactor activity but an impaired ability to promote FX activation at physiological conditions between the sTF(R200W) mutant and sTF(wt) were evident. Increased binding of 1-anilino-8-naphthalene-sulfonic acid (ANS) to sTF(R200W) indicated a population of partially folded intermediates during denaturation. sTF(R200W) showed a dramatically increased propensity for aggregate formation compared to sTF(wt) at mildly acidic pHs, with an increased rate of aggregation during conditions, promoting the intermediate state. The lowered pH resistance could explain the loss of sTF(R200W) in vivo because of aggregation of the mutant. The intrinsic structure of the sTF aggregates appears reminiscent of amyloid fibrils, as revealed by thioflavin T fluorescence, atomic force microscopy, and transmission electron microscopy. We conclude that the lowered activity for FX activation and the propensity of the mutant protein to misfold and aggregate will both contribute to decreased coagulation activity in TF(R200W) carriers, which could protect from thrombotic disease.


Assuntos
Arginina/genética , Mutação , Dobramento de Proteína , Tromboplastina/química , Tromboplastina/genética , Triptofano/genética , Amiloide/genética , Amiloide/metabolismo , Amiloide/ultraestrutura , Naftalenossulfonato de Anilina/metabolismo , Benzotiazóis , Espaço Extracelular/química , Espaço Extracelular/genética , Espaço Extracelular/metabolismo , Fator VIIa/metabolismo , Triagem de Portadores Genéticos , Humanos , Cinética , Nefelometria e Turbidimetria , Ligação Proteica/genética , Desnaturação Proteica , Espectrometria de Fluorescência , Especificidade por Substrato/genética , Tiazóis/metabolismo , Tromboplastina/metabolismo , Tromboplastina/ultraestrutura , Triptofano/química
16.
J Biol Chem ; 280(27): 25558-64, 2005 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-15857836

RESUMO

The influence of protein stability on the adsorption and desorption behavior to surfaces with fundamentally different properties (negatively charged, positively charged, hydrophilic, and hydrophobic) was examined by surface plasmon resonance measurements. Three engineered variants of human carbonic anhydrase II were used that have unchanged surface properties but large differences in stability. The orientation and conformational state of the adsorbed protein could be elucidated by taking all of the following properties of the protein variants into account: stability, unfolding, adsorption, and desorption behavior. Regardless of the nature of the surface, there were correlation between (i) the protein stability and kinetics of adsorption, with an increased amplitude of the first kinetic phase of adsorption with increasing stability; (ii) the protein stability and the extent of maximally adsorbed protein to the actual surface, with an increased amount of adsorbed protein with increasing stability; (iii) the protein stability and the amount of protein desorbed upon washing with buffer, with an increased elutability of the adsorbed protein with increased stability. All of the above correlations could be explained by the rate of denaturation and the conformational state of the adsorbed protein. In conclusion, protein engineering for increased stability can be used as a strategy to decrease irreversible adsorption on surfaces at a liquid-solid interface.


Assuntos
Anidrase Carbônica II/química , Engenharia de Proteínas/métodos , Adsorção , Anidrase Carbônica II/farmacocinética , Eletroquímica , Humanos , Interações Hidrofóbicas e Hidrofílicas , Raios Infravermelhos , Cinética , Peso Molecular , Conformação Proteica , Dobramento de Proteína , Água/química
17.
Biochemistry ; 44(9): 3487-93, 2005 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-15736958

RESUMO

The engineered disulfide bridge A23C/L203C in human carbonic anhydrase II, inserted from homology modeling of Neisseria gonorrhoeae carbonic anhydrase, significantly stabilizes the native state of the protein. The inserted cysteine residues are placed in the interior of the structure, and because of the conformationally restrained localization, the protein is expressed in the reduced state and the cysteines are not readily oxidized. However, upon exposure to low concentrations of denaturant (0.6 M guanidine hydrochloride), corresponding to the lower part of the denaturation curve for the first unfolding transition, the oxidation rate of correctly formed disulfide bridges was markedly increased. By entropy estimations it appears that the increased flexibility, induced by the denaturant, enables the cysteines to find each other and hence to form the disulfide bridge. The outlined strategy of facilitating formation of disulfide bonds by addition of adjusted concentrations of a denaturant should be applicable to other proteins in which engineered cysteine residues are located in nonideal conformations. Moreover, a S99C/V242C variant was constructed, in which the cysteine residues are located on the surface. In this mutant the disulfide bridge was spontaneously formed and the native state was considerably stabilized (midpoint concentration of unfolding was increased from 1.0 to 1.4 M guanidine hydrochloride).


Assuntos
Anidrase Carbônica II/química , Anidrase Carbônica II/genética , Cisteína/genética , Dissulfetos/química , Guanidina/química , Engenharia de Proteínas/métodos , Alanina/genética , Anidrase Carbônica II/síntese química , Eletroforese em Gel de Poliacrilamida , Entropia , Estabilidade Enzimática/genética , Humanos , Leucina/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Neisseria gonorrhoeae/enzimologia , Neisseria gonorrhoeae/genética , Conformação Proteica , Desnaturação Proteica/genética , Dobramento de Proteína , Software , Relação Estrutura-Atividade
18.
Biophys J ; 88(5): 3536-44, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15731384

RESUMO

Little is known about the direction and specificity of protein adsorption to solid surfaces, a knowledge that is of great importance in many biotechnological applications. To resolve the direction in which a protein with known structure and surface potentials binds to negatively charged silica nanoparticles, fluorescent probes were attached to different areas on the surface of the protein human carbonic anhydrase II. By this approach it was clearly demonstrated that the adsorption of the native protein is specific to limited regions at the surface of the N-terminal domain of the protein. Furthermore, the adsorption direction is strongly pH-dependent. At pH 6.3, a histidine-rich area around position 10 is the dominating adsorption region. At higher pH values, when the histidines in this area are deprotonated, the protein is also adsorbed by a region close to position 37, which contains several lysines and arginines. Clearly the adsorption is directed by positively charged areas on the protein surface toward the negatively charged silica surface at conditions when specific binding occurs.


Assuntos
Anidrase Carbônica II/química , Corantes Fluorescentes/farmacologia , Silício/química , Acrilamida/farmacologia , Adsorção , Anisotropia , Arginina/química , Biotecnologia , Cromatografia em Gel , Dicroísmo Circular , Escherichia coli/metabolismo , Histidina/química , Humanos , Concentração de Íons de Hidrogênio , Lisina/química , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Naftalenossulfonatos/farmacologia , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas/química , Software , Espectrometria de Fluorescência , Espectrofotometria , Compostos de Sulfidrila/química
19.
Biochem Biophys Res Commun ; 327(3): 789-93, 2005 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-15649415

RESUMO

Injury of a blood vessel exposes membrane-bound tissue factor (TF) to blood, which allows binding of coagulation factor VIIa (FVIIa). This initiation of the coagulation cascade is dictated by a specific multi-domain interaction between FVIIa and TF. To examine the energies involved in the transition state of the FVIIa:TF complex, various residues in the extracellular part of TF (sTF) that are known to interact with FVIIa were replaced with a smaller cysteine residue. Determination of Phi values in each of the positions using surface plasmon resonance measurements enabled us to characterize the transition state complex between the resulting sTF variants and FVIIa. We found that the interactions in the transition state seemed to be most pronounced between the protease domain of FVIIa and sTF while detailed specific interactions between the Gla-domain and sTF were missing. Thus, the transition state energy data indicate a sequential binding event between these two macromolecules.


Assuntos
Coagulação Sanguínea/fisiologia , Fator VIIa/metabolismo , Tromboplastina/metabolismo , Transferência de Energia , Fator VIIa/química , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína/fisiologia , Termodinâmica , Tromboplastina/química , Fatores de Tempo
20.
J Mol Biol ; 342(2): 619-33, 2004 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15327960

RESUMO

Most loss-of-function diseases are caused by aberrant folding of important proteins. These proteins often misfold due to mutations. The disease marble brain syndrome (MBS), known also as carbonic anhydrase II deficiency syndrome (CADS), can manifest in carriers of point mutations in the human carbonic anhydrase II (HCA II) gene. One mutation associated with MBS entails the His107Tyr substitution. Here, we demonstrate that this mutation is a remarkably destabilizing folding mutation. The loss-of-function is clearly a folding defect, since the mutant shows 64% of CO(2) hydration activity compared to that of the wild-type at low temperature where the mutant is folded. On the contrary, its stability towards thermal and guanidine hydrochloride (GuHCl) denaturation is highly compromised. Using activity assays, CD, fluorescence, NMR, cross-linking, aggregation measurements and molecular modeling, we have mapped the properties of this remarkable mutant. Loss of enzymatic activity had a midpoint temperature of denaturation (T(m)) of 16 degrees C for the mutant compared to 55 degrees C for the wild-type protein. GuHCl-denaturation (at 4 degrees C) showed that the native state of the mutant was destabilized by 9.2kcal/mol. The mutant unfolds through at least two equilibrium intermediates; one novel intermediate that we have termed the molten globule light state and, after further denaturation, the classical molten globule state is populated. Under physiological conditions (neutral pH; 37 degrees C), the His107Tyr mutant will populate the molten globule light state, likely due to novel interactions between Tyr107 and the surroundings of the critical residue Ser29 that destabilize the native conformation. This intermediate binds the hydrophobic dye 8-anilino-1-naphthalene sulfonic acid (ANS) but not as strong as the molten globule state, and near-UV CD reveals the presence of significant tertiary structure. Notably, this intermediate is not as prone to aggregation as the classical molten globule. As a proof of concept for an intervention strategy with small molecules, we showed that binding of the CA inhibitor acetazolamide increases the stability of the native state of the mutant by 2.9kcal/mol in accordance with its strong affinity. Acetazolamide shifts the T(m) to 34 degrees C that protects from misfolding and will enable a substantial fraction of the enzyme pool to survive physiological conditions.


Assuntos
Encefalopatias Metabólicas Congênitas/metabolismo , Anidrase Carbônica II/metabolismo , Substituição de Aminoácidos , Encefalopatias Metabólicas Congênitas/genética , Anidrase Carbônica II/química , Anidrase Carbônica II/genética , Dicroísmo Circular , Guanidina/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Mutação , Dobramento de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA