Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biopolymers ; 98(5): 485-500, 2012.
Article in English | MEDLINE | ID: mdl-23203693

ABSTRACT

To conduct biophysical analyses on large domains of GPCRs, multimilligram quantities of highly homogeneous proteins are necessary. This communication discusses the biosynthesis of four transmembrane and five transmembrane-containing fragments of Ste2p, a GPCR recognizing the Saccharomyces cerevisiae tridecapeptide pheromone α-factor. The target fragments contained the predicted four N-terminal Ste2p[G(31) -A(198) ] (4TMN), four C-terminal Ste2p[T(155) -L(340) ] (4TMC), or five C-terminal Ste2p[I(120) -L(340) ] (5TMC) transmembrane segments of Ste2p. 4TMN was expressed as a fusion protein using a modified pMMHa vector in L-arabinose-induced Escherichia coli BL21-AI, and cleaved with cyanogen bromide. 4TMC and 5TMC were obtained by direct expression using a pET21a vector in IPTG-induced E. coli BL21(DE3) cells. 4TMC and 5TMC were biosynthesized on a preparative scale, isolated in multimilligram amounts, characterized by MS and investigated by biophysical methods. CD spectroscopy indicated the expected highly α-helical content for 4TMC and 5TMC in membrane mimetic environments. Tryptophan fluorescence showed that 5TMC integrated into the nonpolar region of 1-stearoyl-2-hydroxy-sn-glycero-3-phospho-(1'-rac-glycerol) micelles. HSQC-TROSY investigations revealed that [(15) N]-labeled 5TMC in 50% trifluoroethanol-d(2) /H(2) O/0.05%-trifluoroacetic acid was stable enough to conduct long multidimensional NMR measurements. The entire Ste2p GPCR was not readily reconstituted from the first two and last five or first three and last four transmembrane domains.


Subject(s)
Peptide Fragments/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/isolation & purification , Receptors, Mating Factor/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Amino Acid Sequence , Biomimetics , Biophysical Phenomena , Cell Membrane/chemistry , Cyanogen Bromide/chemistry , Electrophoresis, Polyacrylamide Gel , Escherichia coli/chemistry , Genetic Vectors/chemistry , Inclusion Bodies/chemistry , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Plasmids/chemistry , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Receptors, G-Protein-Coupled/chemistry , Recombinant Fusion Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Tryptophan/chemistry
2.
J Biol Chem ; 284(31): 20989-1000, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19483088

ABSTRACT

Flagella of the bacteria Helicobacter pylori and Campylobacter jejuni are important virulence determinants, whose proper assembly and function are dependent upon glycosylation at multiple positions by sialic acid-like sugars, such as 5,7-diacetamido-3,5,7,9-tetradeoxy-l-glycero-l-manno-nonulosonic acid (pseudaminic acid (Pse)). The fourth enzymatic step in the pseudaminic acid pathway, the hydrolysis of UDP-2,4-diacetamido-2,4,6-trideoxy-beta-l-altropyranose to generate 2,4-diacetamido-2,4,6-trideoxy-l-altropyranose, is performed by the nucleotide sugar hydrolase PseG. To better understand the molecular basis of the PseG catalytic reaction, we have determined the crystal structures of C. jejuni PseG in apo-form and as a complex with its UDP product at 1.8 and 1.85 A resolution, respectively. In addition, molecular modeling was utilized to provide insight into the structure of the PseG-substrate complex. This modeling identifies a His(17)-coordinated water molecule as the putative nucleophile and suggests the UDP-sugar substrate adopts a twist-boat conformation upon binding to PseG, enhancing the exposure of the anomeric bond cleaved and favoring inversion at C-1. Furthermore, based on these structures a series of amino acid substitution derivatives were constructed, altering residues within the active site, and each was kinetically characterized to examine its contribution to PseG catalysis. In conjunction with structural comparisons, the almost complete inactivation of the PseG H17F and H17L derivatives suggests that His(17) functions as an active site base, thereby activating the nucleophilic water molecule for attack of the anomeric C-O bond of the UDP-sugar. As the PseG structure reveals similarity to those of glycosyltransferase family-28 members, in particular that of Escherichia coli MurG, these findings may also be of relevance for the mechanistic understanding of this important enzyme family.


Subject(s)
Biosynthetic Pathways , Campylobacter jejuni/enzymology , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Sugar Acids/metabolism , Uridine Diphosphate Sugars/metabolism , Bacterial Outer Membrane Proteins/chemistry , Binding Sites , Biocatalysis , Computer Simulation , Crystallography, X-Ray , Helicobacter pylori/enzymology , Kinetics , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , N-Acetylglucosaminyltransferases/chemistry , Protein Structure, Secondary , Sequence Homology, Amino Acid , Structure-Activity Relationship
6.
Arch Biochem Biophys ; 460(1): 10-6, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17353004

ABSTRACT

Glycosaminoglycans (GAGs) are believed to be associated with prion disease pathology and also with metabolism of the prion protein. Fluorescence polarization assay (FPA) of binding between bovine recombinant prion protein (brecPrP) and heparin labelled with AlexaFluor488 was used in model experiments to study glycosaminoglycan-prion protein interaction. Heparin binding to brecPrP was a rapid reversible event which occurred under defined conditions. The interaction of brecPrP with fluorophore-labelled heparin was inhibited by the presence of Cu(2+) ions and was sensitive to competition with heparin, heparan sulphate, and dextran. The dissociation constant of the heparin-brecPrP complex was 73.4+/-3.7 nM. Circular dichroism (CD) experiments indicated that the structure of brecPrP was less helical in the presence of heparin.


Subject(s)
Heparin/metabolism , Prions/metabolism , Animals , Binding Sites , Cattle , Circular Dichroism , Endopeptidase K/metabolism , Fluorescence Polarization , Glycosaminoglycans/metabolism , Heparin/chemistry , Hydrogen-Ion Concentration , Kinetics , Osmolar Concentration , Prions/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sheep
7.
Biochemistry ; 45(37): 11113-21, 2006 Sep 19.
Article in English | MEDLINE | ID: mdl-16964972

ABSTRACT

We have studied the effects of C-terminal group modifications (amide, methylamide, dimethylamide, aldehyde, and alcohol) on the conformation, adenylyl cyclase stimulation (AC), or binding of parathyroid hormone (hPTH) analogues, hPTH(1-28)NH(2) and hPTH(1-31)NH(2). hPTH(1-31)NH(2) has a C-terminal alpha-helix bounded by residues 17-29 [Chen, Z., et al. (2000) Biochemistry 39, 12766]. In both cases, relative to the natural analogue with a carboxyl C-terminus, the amide and methylamide had increased helix content whereas the dimethylamide forms had CD spectra more similar to the carboxyl one. Conformational effects were more pronounced with hPTH(1-28) than with hPTH(1-31), with increases in helix content of approximately 30% in contrast to 10%. Stabilization of the C-terminal helix of residues 1-28 seemed to correlate with an ability of the C-terminal function to H-bond appropriately. None of the analogues affected the AC stimulating activity significantly, but there was an up to 15-fold decrease in the level of apparent binding of the carboxyl hPTH(1-28) analogue compared to that of the methylamide and a 4-fold decrease in the level of binding of the aldehyde or dimethylamide. There was no significant change in binding activities for the 1-31 analogues. These observations are consistent with previous studies that imply the importance of a region of the hormone's C-terminal alpha-helix for tight binding to the receptor. They also show that modulation of helix stability does have an effect on the binding of the hormone, but only when the C-terminus is at the putative end of the helix. The similarity of AC stimulation even when binding changed 10-fold can be explained by assuming greater efficacy of the weaker binding PTH-receptor complexes in stimulating AC.


Subject(s)
Parathyroid Hormone/chemistry , Peptide Fragments/chemistry , Animals , Cell Line , Chromatography, High Pressure Liquid , Circular Dichroism , Macaca mulatta , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Thermodynamics
8.
J Biol Chem ; 281(43): 32485-95, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16923805

ABSTRACT

The principal receptor-binding domain (Ser(17)-Val(31)) of parathyroid hormone (PTH) is predicted to form an amphiphilic alpha-helix and to interact primarily with the N-terminal extracellular domain (N domain) of the PTH receptor (PTHR). We explored these hypotheses by introducing a variety of substitutions in region 17-31 of PTH-(1-31) and assessing, via competition assays, their effects on binding to the wild-type PTHR and to PTHR-delNt, which lacks most of the N domain. Substitutions at Arg(20) reduced affinity for the intact PTHR by 200-fold or more, but altered affinity for PTHR-delNt by 4-fold or less. Similar effects were observed for Glu substitutions at Trp(23), Leu(24), and Leu(28), which together form the hydrophobic face of the predicted amphiphilic alpha-helix. Glu substitutions at Arg(25), Lys(26), and Lys(27) (which forms the hydrophilic face of the helix) caused 4-10-fold reductions in affinity for both receptors. Thus, the side chains of Arg(20), together with those composing the hydrophobic face of the ligand's putative amphiphilic alpha-helix, contribute strongly to PTHR-binding affinity by interacting specifically with the N domain of the receptor. The side chains projecting from the opposite helical face contribute weakly to binding affinity by different mechanisms, possibly involving interactions with the extracellular loop/transmembrane domain region of the receptor. The data help define the roles that side chains in the binding domain of PTH play in the PTH-PTHR interaction process and provide new clues for understanding the overall topology of the bimolecular complex.


Subject(s)
Parathyroid Hormone/chemistry , Peptide Fragments/chemistry , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/metabolism , Alanine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Amino Acids/chemistry , Cell Culture Techniques , Cell Line , Circular Dichroism , Glutamic Acid/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Parathyroid Hormone/metabolism , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
9.
J Biol Chem ; 280(25): 23771-7, 2005 Jun 24.
Article in English | MEDLINE | ID: mdl-15826940

ABSTRACT

We have used backbone N-methylations of parathyroid hormone (PTH) to study the role of these NH groups in the C-terminal amphiphilic alpha-helix of PTH (1-31) in binding to and activating the PTH receptor (P1R). The circular dichroism (CD) spectra indicated the structure of the C-terminal alpha-helix was locally disrupted around the methylation site. The CD spectra differences were explained by assuming a helix disruption for four residues on each side of the site of methylation and taking into account the known dependence of CD on the length of an alpha-helix. Binding and adenylyl cyclase-stimulating data showed that outside of the alpha-helix, methylation of residues Asp30 and Val31 had little effect on structure or activities. Within the alpha-helix, disruption of the structure was associated with increased loss of activity, but for specific residues Val21, Leu24, Arg25, and Leu28 there was a dramatic loss of activities, thus suggesting a more direct role of these NH groups in correct P1R binding and activation. Activity analyses with P1R-delNT, a mutant with its long N-terminal region deleted, gave a different pattern of effects and implicated Ser17, Trp23, and Lys26 as important for its PTH activation. These two groups of residues are located on opposite sides of the helix. These results are compatible with the C-terminal helix binding to both the N-terminal segment and also to the looped-out extracellular region. These data thus provide direct evidence for important roles of the C-terminal domain of PTH in determining high affinity binding and activation of the P1R receptor.


Subject(s)
Parathyroid Hormone/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Circular Dichroism , Humans , Methylation , Molecular Sequence Data , Parathyroid Hormone/chemistry , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...