Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(35): 11018-23, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26283364

RESUMEN

Toll-like receptors (TLRs) are crucial in innate recognition of invading micro-organisms and their subsequent clearance. Bacteria are not passive bystanders and have evolved complex evasion mechanisms. Staphylococcus aureus secretes a potent TLR2 antagonist, staphylococcal superantigen-like protein 3 (SSL3), which prevents receptor stimulation by pathogen-associated lipopeptides. Here, we present crystal structures of SSL3 and its complex with TLR2. The structure reveals that formation of the specific inhibitory complex is predominantly mediated by hydrophobic contacts between SSL3 and TLR2 and does not involve interaction of TLR2-glycans with the conserved Lewis(X) binding site of SSL3. In the complex, SSL3 partially covers the entrance to the lipopeptide binding pocket in TLR2, reducing its size by ∼50%. We show that this is sufficient to inhibit binding of agonist Pam2CSK4 effectively, yet allows SSL3 to bind to an already formed TLR2-Pam2CSK4 complex. The binding site of SSL3 overlaps those of TLR2 dimerization partners TLR1 and TLR6 extensively. Combined, our data reveal a robust dual mechanism in which SSL3 interferes with TLR2 activation at two stages: by binding to TLR2, it blocks ligand binding and thus inhibits activation. Second, by interacting with an already formed TLR2-lipopeptide complex, it prevents TLR heterodimerization and downstream signaling.


Asunto(s)
Endotoxinas/fisiología , Staphylococcus aureus/fisiología , Receptor Toll-Like 2/antagonistas & inhibidores , Dimerización , Endotoxinas/química , Endotoxinas/genética , Estructura Molecular , Mutagénesis , Unión Proteica , Receptor Toll-Like 2/química
2.
Proc Natl Acad Sci U S A ; 109(14): 5253-8, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22440751

RESUMEN

Fibrillar collagens, the most abundant proteins in the vertebrate body, are involved in a plethora of biological interactions. Plasma protein von Willebrand factor (VWF) mediates adhesion of blood platelets to fibrillar collagen types I, II, and III, which is essential for normal haemostasis. High affinity VWF-binding sequences have been identified in the homotrimeric collagen types II and III, however, it is unclear how VWF recognizes the heterotrimeric collagen type I, the superstructure of which is unknown. Here we present the crystal structure of VWF domain A3 bound to a collagen type III-derived homotrimeric peptide. Our structure reveals that VWF-A3 interacts with all three collagen chains and binds through conformational selection to a sequence that is one triplet longer than was previously appreciated from platelet and VWF binding studies. The VWF-binding site overlaps those of SPARC (also known as osteonectin) and discodin domain receptor 2, but is more extended and shifted toward the collagen amino terminus. The observed collagen-binding mode of VWF-A3 provides direct structural constraints on collagen I chain registry. A VWF-binding site can be generated from the sequences RGQAGVMF, present in the two α1(I) chains, and RGEOGNIGF, in the unique α2(I) chain, provided that α2(I) is in the middle or trailing position. Combining these data with previous structural data on integrin binding to collagen yields strong support for the trailing position of the α2(I) chain, shedding light on the fundamental and long-standing question of the collagen I chain registry.


Asunto(s)
Colágeno Tipo I/química , Factor de von Willebrand/química , Secuencia de Aminoácidos , Sitios de Unión , Colágeno Tipo I/metabolismo , Humanos , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido , Factor de von Willebrand/metabolismo
3.
Protein Expr Purif ; 99: 27-34, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24680733

RESUMEN

Transient transfection of the human HEK293-EBNA1 cell line using polyethyleneimine is widely adopted for recombinant protein production. Whereas high expression of many targets is achieved, purification yields of some highly expressed proteins remain low due to aggregation. We hypothesized that for these proteins the expression rates achieved at standard transfection conditions are too high, causing an overload of the protein folding machinery. Here we present plasmid titration as an efficient method to vary expression rates for the optimization of soluble protein expression. In plasmid titration a dilution series of expression vector mixed with dummy plasmid is transfected in small scale cultures. Application to GFP shows that plasmid titration achieves a wide range of expression levels while maintaining high transfection efficiencies even at 500-fold plasmid dilution. Application of plasmid titration to selected Nod-like receptors (NLRs), which at standard conditions are highly expressed but poorly soluble, delays the onset of NLR aggregation and improves cell viability and the buildup of biomass. The amount of soluble protein depends on the combination of dilution factor and harvest day in a protein specific manner. For NOD1 50-fold plasmid dilution increases the amount of soluble protein approximately 5-fold. Due to its association with chaperones at all dilution factors tested we were unable to purify NOD1 to homogeneity. For NLRC4, which did not associate with chaperones, 10-fold plasmid dilution increased the purification yield 2-fold. This improvement, obtained with minimal effort due to the simplicity of the method, shows that reducing total expression may increase soluble protein yield.


Asunto(s)
Células HEK293/metabolismo , Plásmidos/genética , Agregado de Proteínas/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Transfección/métodos , Proteínas Adaptadoras de Señalización CARD/química , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/aislamiento & purificación , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/aislamiento & purificación , Muerte Celular , Antígenos Nucleares del Virus de Epstein-Barr/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Polietileneimina , Pliegue de Proteína/efectos de los fármacos , Proteínas Recombinantes/genética , Solubilidad
4.
J Biol Chem ; 287(46): 38460-72, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23012363

RESUMEN

The NOD-like receptors NAIP5 and NLRC4 play an essential role in the innate immune response to the bacterial tail protein flagellin. Upon flagellin detection, NAIP5 and NLRC4 form a hetero-oligomeric inflammasome that induces caspase-1-dependent cell death. So far, both the mechanism of formation of the NAIP5-NLRC4 inflammasome and its structure are poorly understood. In this study we combine inflammasome reconstitution in HEK293 cells, purification of inflammasome components, and negative stain electron microscopy to address these issues. We find that a Salmonella typhimurium flagellin fragment comprising the D0 domain and the neighboring spoke region is able to co-precipitate NAIP5 and induce formation of the NAIP5-NLRC4 inflammasome. Comparison with smaller fragments indicates that flagellin recognition is mediated by its C-terminal residues as well as the spoke region. We reconstitute the inflammasome from purified flagellin, NAIP5, and NLRC4, thus proving that no other cellular components are required for its formation. Electron micrographs of the purified inflammasome provide unprecedented insight into its architecture, revealing disk-like complexes consisting of 11 or 12 protomers in which NAIP5 and NLRC4 appear to occupy equivalent positions. On the basis of our data, we propose a model for inflammasome formation wherein direct interaction of flagellin with a single NAIP5 induces the recruitment and progressive incorporation of NLRC4, resulting in the formation of a hetero-oligomeric inflammasome.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Proteínas Adaptadoras de Señalización CARD/química , Proteínas de Unión al Calcio/química , Flagelina/metabolismo , Inflamasomas/metabolismo , Proteína Inhibidora de la Apoptosis Neuronal/química , Salmonella typhimurium/metabolismo , Animales , Caspasa 1/química , ADN Complementario/metabolismo , Células HEK293 , Humanos , Ligandos , Ratones , Microscopía Electrónica/métodos , Plásmidos/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína
5.
Blood ; 115(7): 1364-73, 2010 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-20007810

RESUMEN

Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1), one of the most widely spread immune receptors, attenuates immune cell activation when bound to specific sites in collagen. The collagen-binding domain of LAIR-1 is homologous to that of glycoprotein VI (GPVI), a collagen receptor crucial for platelet activation. Because LAIR-1 and GPVI also display overlapping collagen-binding specificities, a common structural basis for collagen recognition would appear likely. Therefore, it is crucial to gain insight into the molecular interaction of both receptors with their ligand to prevent unwanted cross-reactions during therapeutic intervention. We determined the crystal structure of LAIR-1 and mapped its collagen-binding site by nuclear magnetic resonance (NMR) titrations and mutagenesis. Our data identify R59, E61, and W109 as key residues for collagen interaction. These residues are strictly conserved in LAIR-1 and GPVI alike; however, they are located outside the previously proposed GPVI collagen-binding site. Our data provide evidence for an unanticipated mechanism of collagen recognition common to LAIR-1 and GPVI. This fundamental insight will contribute to the exploration of specific means of intervention in collagen-induced signaling in immunity and hemostasis.


Asunto(s)
Colágeno/metabolismo , Glicoproteínas de Membrana Plaquetaria/química , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Inmunológicos/química , Receptores Inmunológicos/metabolismo , Sitios de Unión/fisiología , Cristalografía , Expresión Génica , Humanos , Células K562 , Mutagénesis , Resonancia Magnética Nuclear Biomolecular , Activación Plaquetaria/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores Inmunológicos/genética , Transducción de Señal/fisiología , Relación Estructura-Actividad
6.
Blood ; 112(7): 2803-9, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18559974

RESUMEN

Thrombin-activatable fibrinolysis inhibitor (TAFI) is a pro-metallocarboxypeptidase that can be proteolytically activated (TAFIa). TAFIa is unique among carboxypeptidases in that it spontaneously inactivates with a short half-life, a property that is crucial for its role in controlling blood clot lysis. We studied the intrinsic instability of TAFIa by solving crystal structures of TAFI, a TAFI inhibitor (GEMSA) complex and a quadruple TAFI mutant (70-fold more stable active enzyme). The crystal structures show that TAFIa stability is directly related to the dynamics of a 55-residue segment (residues 296-350) that includes residues of the active site wall. Dynamics of this flap are markedly reduced by the inhibitor GEMSA, a known stabilizer of TAFIa, and stabilizing mutations. Our data provide the structural basis for a model of TAFI auto-regulation: in zymogen TAFI the dynamic flap is stabilized by interactions with the activation peptide. Release of the activation peptide increases dynamic flap mobility and in time this leads to conformational changes that disrupt the catalytic site and expose a cryptic thrombin-cleavage site present at Arg302. This represents a novel mechanism of enzyme control that enables TAFI to regulate its activity in plasma in the absence of specific inhibitors.


Asunto(s)
Carboxipeptidasa B2/química , Carboxipeptidasas/química , Carboxipeptidasas/metabolismo , Línea Celular , Cristalografía por Rayos X , Activación Enzimática , Inhibidores Enzimáticos/metabolismo , Humanos , Modelos Biológicos , Mutación/genética , Precursores de Proteínas , Estructura Secundaria de Proteína
7.
Front Immunol ; 10: 2097, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31552043

RESUMEN

Properdin enhances complement-mediated opsonization of targeted cells and particles for immune clearance. Properdin occurs as dimers, trimers and tetramers in human plasma, which recognize C3b-deposited surfaces, promote formation, and prolong the lifetime of C3bBb-enzyme complexes that convert C3 into C3b, thereby enhancing the complement-amplification loop. Here, we report crystal structures of monomerized properdin, which was produced by co-expression of separate N- and C-terminal constructs that yielded monomer-sized properdin complexes that stabilized C3bBb. Consistent with previous low-resolution X-ray and EM data, the crystal structures revealed ring-shaped arrangements that are formed by interactions between thrombospondin type-I repeat (TSR) domains 4 and 6 of one protomer interacting with the N-terminal domain (which adopts a short transforming-growth factor B binding protein-like fold) and domain TSR1 of a second protomer, respectively. Next, a structure of monomerized properdin in complex with the C-terminal domain of C3b showed that properdin-domain TSR5 binds along the C-terminal α-helix of C3b, while two loops, one from domain TSR5 and one from TSR6, extend and fold around the C3b C-terminus like stirrups. This suggests a mechanistic model in which these TSR5 and TSR6 "stirrups" bridge interactions between C3b and factor B or its fragment Bb, and thereby enhance formation of C3bB pro-convertases and stabilize C3bBb convertases. In addition, properdin TSR6 would sterically block binding of the protease factor I to C3b, thus limiting C3b proteolytic degradation. The presence of a valine instead of a third tryptophan in the canonical Trp-ladder of TSR domains in TSR4 allows a remarkable ca. 60°-domain bending motion of TSR4. Together with variable positioning of TSR2 and, putatively, TSR3, this explains the conformational flexibility required for properdin to form dimers, trimers, and tetramers. In conclusion, the results indicate that binding avidity of oligomeric properdin is needed to distinguish surface-deposited C3b molecules from soluble C3b or C3 and suggest that properdin-mediated interactions bridging C3b-B and C3b-Bb enhance affinity, thus promoting convertase formation and stabilization. These mechanisms explain the enhancement of complement-mediated opsonization of targeted cells and particle for immune clearance.


Asunto(s)
Activación de Complemento , Complemento C3b/química , Factores Inmunológicos/química , Properdina/química , Complemento C3b/genética , Complemento C3b/inmunología , Glicosilación , Células HEK293 , Humanos , Factores Inmunológicos/inmunología , Properdina/genética , Properdina/inmunología , Dominios Proteicos , Proteínas Recombinantes/química
8.
Biochem J ; 379(Pt 1): 47-55, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-14674886

RESUMEN

Nap (periplasmic nitrate reductase) operons of many bacteria include four common, essential components, napD, napA, napB and napC (or a homologue of napC ). In Escherichia coli there are three additional genes, napF, napG and napH, none of which are essential for Nap activity. We now show that deletion of either napG or napH almost abolished Nap-dependent nitrate reduction by strains defective in naphthoquinone synthesis. The residual rate of nitrate reduction (approx. 1% of that of napG+ H+ strains) is sufficient to replace fumarate reduction in a redox-balancing role during growth by glucose fermentation. Western blotting combined with beta-galactosidase and alkaline phosphatase fusion experiments established that NapH is an integral membrane protein with four transmembrane helices. Both the N- and C-termini as well as the two non-haem iron-sulphur centres are located in the cytoplasm. An N-terminal twin arginine motif was shown to be essential for NapG function, consistent with the expectation that NapG is secreted into the periplasm by the twin arginine translocation pathway. A bacterial two-hybrid system was used to show that NapH interacts, presumably on the cytoplasmic side of, or within, the membrane, with NapC. As expected for a periplasmic protein, no NapG interactions with NapC or NapH were detected in the cytoplasm. An in vitro quinol dehydrogenase assay was developed to show that both NapG and NapH are essential for rapid electron transfer from menadiol to the terminal NapAB complex. These new in vivo and in vitro results establish that NapG and NapH form a quinol dehydrogenase that couples electron transfer from the high midpoint redox potential ubiquinone-ubiquinol couple via NapC and NapB to NapA.


Asunto(s)
Proteínas de Escherichia coli/fisiología , Escherichia coli/enzimología , Nitrato Reductasas/fisiología , Oxidorreductasas/fisiología , Proteínas Periplasmáticas/fisiología , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo , Arginina/química , Secuencia de Bases , Transporte de Electrón , Escherichia coli/ultraestructura , Proteínas de Escherichia coli/análisis , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Genes Bacterianos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Nitrato-Reductasa , Nitrato Reductasas/análisis , Nitrato Reductasas/metabolismo , Nitritos/metabolismo , Operón , Oxidación-Reducción , Oxidorreductasas/análisis , Oxidorreductasas/genética , Proteínas Periplasmáticas/análisis , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/análisis , Fracciones Subcelulares/enzimología , Técnicas del Sistema de Dos Híbridos
9.
PLoS One ; 8(7): e67923, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23844130

RESUMEN

BACKGROUND: RP105 (CD180) is TLR4 homologue lacking the intracellular TLR4 signaling domain and acts a TLR accessory molecule and physiological inhibitor of TLR4-signaling. The role of RP105 in vascular remodeling, in particular post-interventional remodeling is unknown. METHODS AND RESULTS: TLR4 and RP105 are expressed on vascular smooth muscle cells (VSMC) as well as in the media of murine femoral artery segments as detected by qPCR and immunohistochemistry. Furthermore, the response to the TLR4 ligand LPS was stronger in VSMC from RP105(-/-) mice resulting in a higher proliferation rate. In RP105(-/-) mice femoral artery cuff placement resulted in an increase in neointima formation as compared to WT mice (4982 ± 974 µm(2) vs.1947 ± 278 µm(2),p = 0.0014). Local LPS application augmented neointima formation in both groups, but in RP105(-/-) mice this effect was more pronounced (10316±1243 µm(2) vs.4208 ± 555 µm(2),p = 0.0002), suggesting a functional role for RP105. For additional functional studies, the extracellular domain of murine RP105 was expressed with or without its adaptor protein MD1 and purified. SEC-MALSanalysis showed a functional 2∶2 homodimer formation of the RP105-MD1 complex. This protein complex was able to block the TLR4 response in whole blood ex-vivo. In vivo gene transfer of plasmid vectors encoding the extracellular part of RP105 and its adaptor protein MD1 were performed to initiate a stable endogenous soluble protein production. Expression of soluble RP105-MD1 resulted in a significant reduction in neointima formation in hypercholesterolemic mice (2500 ± 573 vs.6581 ± 1894 µm(2),p<0.05), whereas expression of the single factors RP105 or MD1 had no effect. CONCLUSION: RP105 is a potent inhibitor of post-interventional neointima formation.


Asunto(s)
Antígenos CD/metabolismo , Vasos Sanguíneos/metabolismo , Neointima/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Antígenos CD/genética , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Arteria Femoral/metabolismo , Expresión Génica , Células HEK293 , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Inmunohistoquímica , Lipopolisacáridos/farmacología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Neointima/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Solubilidad , Receptor Toll-Like 4/genética
10.
J Biol Chem ; 282(13): 9302-9311, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17215253

RESUMEN

Yeast exopolyphosphatase (scPPX) processively splits off the terminal phosphate group from linear polyphosphates longer than pyrophosphate. scPPX belongs to the DHH phosphoesterase superfamily and is evolutionarily close to the well characterized family II pyrophosphatase (PPase). Here, we used steady-state kinetic and binding measurements to elucidate the metal cofactor requirement for scPPX catalysis over the pH range 4.2-9.5. A single tight binding site for Mg(2+) (K(d) of 24 microm) was detected by equilibrium dialysis. Steady-state kinetic analysis of tripolyphosphate hydrolysis revealed a second site that binds Mg(2+) in the millimolar range and modulates substrate binding. This step requires two protonated and two deprotonated enzyme groups with pK(a) values of 5.0-5.3 and 7.6-8.2, respectively. The catalytic step requiring two deprotonated groups (pK(a) of 4.6 and 5.6) is modulated by ionization of a third group (pK(a) of 8.7). Conservative mutations of Asp(127), His(148), His(149) (conserved in scPPX and PPase), and Asn(35) (His in PPase) reduced activity by a factor of 600-5000. N35H and D127E substitutions reduced the Mg(2+) affinity of the tight binding site by 25-60-fold. Contrary to expectations, the N35H variant was unable to hydrolyze pyrophosphate, but markedly altered metal cofactor specificity, displaying higher catalytic activity with Co(2+) bound to the weak binding site versus the Mg(2+)- or Mn(2+)-bound enzyme. These results provide an initial step toward understanding the dynamics of scPPX catalysis and reveal significant functional differences between structurally similar scPPX and family II PPase.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Ácido Anhídrido Hidrolasas/metabolismo , Sustitución de Aminoácidos/genética , Citosol/enzimología , Saccharomyces cerevisiae/enzimología , Análisis de Secuencia de Proteína , Ácido Anhídrido Hidrolasas/biosíntesis , Ácido Anhídrido Hidrolasas/química , Ácido Anhídrido Hidrolasas/aislamiento & purificación , Secuencia de Aminoácidos , Cinética , Saccharomyces cerevisiae/genética , Homología de Secuencia de Aminoácido
11.
J Biol Chem ; 282(50): 36434-43, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17911104

RESUMEN

Transhydrogenase couples the redox reaction between NADH and NADP+ to proton translocation across a membrane. The protein has three components: dI binds NADH, dIII binds NADP+, and dII spans the membrane. Transhydrogenase is a "dimer" of two dI-dII-dIII "monomers"; x-ray structures suggested that the two catalytic sites alternate during turnover. Invariant Tyr146 in recombinant dI of Rhodospirillum rubrum transhydrogenase was substituted with Phe and Ala (proteins designated dI.Y146F and dI.Y146A, respectively). Analytical ultracentrifuge experiments and differential scanning calorimetry show that dI.Y146A more readily dissociates into monomers than wild-type dI. Analytical ultracentrifuge and Trp fluorescence experiments indicate that the dI.Y146A monomers bind NADH much more weakly than dimers. Wild-type dI and dI.Y146F reconstituted activity to dI-depleted membranes with similar characteristics. However, dI.Y146A reconstituted activity in its dimeric form but not in its monomeric form, this despite monomers retaining their native fold and binding to the dI-depleted membranes. It is suggested that transhydrogenase reconstructed with monomers of dI.Y146A is catalytically compromised, at least partly as a consequence of the lowered affinity for NADH, and this results from lost interactions between the nucleotide binding site and the protein beta-hairpin upon dissociation of the dI dimer. The importance of these interactions and their coupling to dI domain rotation in the mechanism of action of transhydrogenase is emphasized. Two peaks in the 1H NMR spectrum of wild-type dI are broadened in dI.Y146A and are tentatively assigned to S-methyl groups of Met resonances in the beta-hairpin, consistent with the segmental mobility of this feature in the structure.


Asunto(s)
NADP Transhidrogenasas/química , Pliegue de Proteína , Rhodospirillum rubrum/enzimología , Sustitución de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Rastreo Diferencial de Calorimetría , Cristalografía por Rayos X , Dimerización , Activación Enzimática/genética , Mutación Missense , NADP Transhidrogenasas/genética , Resonancia Magnética Nuclear Biomolecular , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Rhodospirillum rubrum/genética , Relación Estructura-Actividad , Ultracentrifugación
12.
Microbiology (Reading) ; 152(Pt 11): 3227-3237, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17074894

RESUMEN

The periplasmic nitrate reductase of Escherichia coli is important during anaerobic growth in low-nitrate environments. The nap operon encoding this nitrate reductase comprises seven genes including a gene, napF, that encodes a putative cytoplasmic iron-sulphur protein of uncertain subcellular location and function. In this study, N-terminal sequence analysis, cell fractionation coupled with immunoblotting and construction of LacZ and PhoA fusion proteins were used together to establish that NapF is located in the E. coli cytoplasm. A bacterial two-hybrid protein-protein interaction system was used to demonstrate that NapF interacted in the cytoplasm with the terminal oxidoreductase NapA, but that it did not self-associate or interact with other electron-transport components of the Nap system, NapC, NapG or NapH, or with another cytoplasmic component, NapD. NapF, purified as a His(6)-tagged protein, exhibited spectral properties characteristic of an iron-sulphur protein. This protein was able to pull down NapA from soluble extracts of E. coli. A growth-based assay for NapF function in intact cell cultures was developed and applied to assess the effect of mutation of a number of conserved amino acids. It emerged that neither a highly conserved N-terminal double-arginine motif, nor a conserved proline motif, is essential for NapF-dependent growth. The combined data indicate that NapF plays one or more currently unidentified roles in the post-translational modification of NapA prior to the export of folded NapA via the twin-arginine translocation pathway into the periplasm.


Asunto(s)
Dominio Catalítico/fisiología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Proteínas Hierro-Azufre/fisiología , Nitrato-Reductasa/metabolismo , Sitios de Unión , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiología , Proteínas Hierro-Azufre/metabolismo , Operón , Proteínas Periplasmáticas/metabolismo , Unión Proteica , Fracciones Subcelulares/metabolismo , Técnicas del Sistema de Dos Híbridos
13.
J Biol Chem ; 281(19): 13345-13354, 2006 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16533815

RESUMEN

Transhydrogenase couples proton translocation across a membrane to hydride transfer between NADH and NADP+. Previous x-ray structures of complexes of the nucleotide-binding components of transhydrogenase ("dI2dIII1" complexes) indicate that the dihydronicotinamide ring of NADH can move from a distal position relative to the nicotinamide ring of NADP+ to a proximal position. The movement might be responsible for gating hydride transfer during proton translocation. We have mutated three invariant amino acids, Arg-127, Asp-135, and Ser-138, in the NAD(H)-binding site of Rhodospirillum rubrum transhydrogenase. In each mutant, turnover by the intact enzyme is strongly inhibited. Stopped-flow experiments using dI2dIII1 complexes show that inhibition results from a block in the steps associated with hydride transfer. Mutation of Asp-135 and Ser-138 had no effect on the binding affinity of either NAD+ or NADH, but mutation of Arg-127 led to much weaker binding of NADH and slightly weaker binding of NAD+. X-ray structures of dI2dIII1 complexes carrying the mutations showed that their effects were restricted to the locality of the bound NAD(H). The results are consistent with the suggestion that in wild-type protein movement of the Arg-127 side chain, and its hydrogen bonding to Asp-135 and Ser-138, stabilizes the dihydronicotinamide of NADH in the proximal position for hydride transfer.


Asunto(s)
Aminoácidos/metabolismo , NADP Transhidrogenasas/química , NADP Transhidrogenasas/metabolismo , Rhodospirillum rubrum/enzimología , Sitios de Unión , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , NAD/metabolismo , NADP/metabolismo , NADP Transhidrogenasas/genética , Unión Proteica , Conformación Proteica , Subunidades de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA