Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Org Biomol Chem ; 15(45): 9685-9695, 2017 Nov 22.
Article in English | MEDLINE | ID: mdl-29119993

ABSTRACT

Sulfonyl fluorides (SFs) have recently emerged as a promising warhead for the targeted covalent modification of proteins. Despite numerous examples of the successful deployment of SFs as covalent probe compounds, a detailed exploration of the factors influencing the stability and reactivity of SFs has not yet appeared. In this work we present an extensive study on the influence of steric and electronic factors on the reactivity and stability of the SF and related SVI-F groups. While SFs react rapidly with N-acetylcysteine, the resulting adducts were found to be unstable, rendering SFs inappropriate for the durable covalent inhibition of cysteine residues. In contrast, SFs afforded stable adducts with both N-acetyltyrosine and N-acetyllysine; furthermore, we show that the reactivity of arylsulfonyl fluorides towards these nucleophilic amino acids can be predictably modulated by adjusting the electronic properties of the warhead. These trends were largely conserved when the covalent reaction occurred within a protein binding pocket. We have also obtained a crystal structure depicting covalent modification of the catalytic lysine of a tyrosine kinase (FGFR1) by the ATP analog 5'-O-3-((fluorosulfonyl)benzoyl)adenosine (m-FSBA). Highly reactive warheads were demonstrated to be unstable with respect to hydrolysis in buffered aqueous solutions, indicating that warhead reactivity must be carefully tuned to provide optimal rates of protein modification. Our results demonstrate that the reactivity of SFs complements that of more commonly studied acrylamides, and we hope that this work spurs the rational design of novel SF-containing covalent probe compounds and inhibitors, particularly in cases where a suitably positioned cysteine residue is not present.


Subject(s)
Amino Acids/chemistry , Sulfinic Acids/chemistry , Animals , Crystallography, X-Ray , Models, Molecular , Molecular Structure , Rats , Rats, Wistar , Sulfinic Acids/blood , Sulfinic Acids/chemical synthesis
2.
J Interferon Cytokine Res ; 21(11): 931-41, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11747625

ABSTRACT

The epitopes important for receptor binding and activation of human interferon-beta1a (IFN-beta1a) were mapped with monoclonal antibodies (mAb), grouped on the basis of their specificity and ability to neutralize biologic activity, and alanine scanning mutagenesis (ASM). The binding properties of nine mAb were defined, using ASM-IFN-beta mutants having alanine substituted at targeted, surface-exposed residues. The results were correlated with the mAb neutralizing potency. Of six mAb that bound either at or adjacent to the IFNAR-2 receptor chain binding site defined by the ASM epitopes, only three had measurable neutralizing activity. Two of these inhibited IFN-beta/IFNAR-2 complex formation, suggesting that steric hindrance of receptor binding constitutes their mechanism of neutralization. However, two mAb that bound to sites remote from the IFNAR-2 binding site on IFN-beta also inhibited IFN-beta/IFNAR-2 complex formation and demonstrated potent neutralizing activity. Thus, neutralizing mAb may employ mechanisms other than steric blockade to inhibit directly the binding of receptor by cytokine, limiting their usefulness as tools to define precise receptor-ligand interaction sites.


Subject(s)
Alanine/genetics , Antibodies, Monoclonal/immunology , Epitopes/immunology , Interferon-beta/chemistry , Interferon-beta/immunology , Receptors, Interferon/metabolism , Amino Acid Sequence , Amino Acid Substitution , Antibody Specificity , Binding, Competitive , Dose-Response Relationship, Drug , Epitope Mapping , Humans , Interferon beta-1a , Interferon-beta/genetics , Interferon-beta/metabolism , Membrane Proteins , Models, Molecular , Molecular Sequence Data , Neutralization Tests , Receptor, Interferon alpha-beta , Sequence Alignment
3.
Nat Immunol ; 2(12): 1159-66, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713465

ABSTRACT

According to the two-signal model of T cell activation, costimulatory molecules augment T cell receptor (TCR) signaling, whereas adhesion molecules enhance TCR-MHC-peptide recognition. The structure and binding properties of CD28 imply that it may perform both functions, blurring the distinction between adhesion and costimulatory molecules. Our results show that CD28 on naïve T cells does not support adhesion and has little or no capacity for directly enhancing TCR-MHC-peptide interactions. Instead of being dependent on costimulatory signaling, we propose that a key function of the immunological synapse is to generate a cellular microenvironment that favors the interactions of potent secondary signaling molecules, such as CD28.


Subject(s)
B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , B7-1 Antigen/genetics , CD28 Antigens/chemistry , Cell Adhesion , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Glycosylphosphatidylinositols/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism
4.
J Biol Chem ; 276(39): 36520-9, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11473127

ABSTRACT

We have used the highly selective alpha(4)beta(1) inhibitor 2S-[(1-benzenesulfonyl-pyrrolidine-2S-carbonyl)-amino]-4-[4-methyl-2S-(methyl-[2-[4-(3-o-tolyl-ureido)-phenyl]-acetyl]-amino)-pentanoylamino]-butyric acid (BIO7662) as a model ligand to study alpha(4)beta(1) integrin-ligand interactions on Jurkat cells. Binding of [(35)S]BIO7662 to Jurkat cells was dependent on the presence of divalent cations and could be blocked by treatment with an excess of unlabeled inhibitor or with EDTA. K(D) values for the binding of BIO7662 to Mn(2+)-activated alpha(4)beta(1) and to the nonactivated state of the integrin that exists in 1 mm Mg(2+), 1 mm Ca(2+) were <10 pm, indicating that it has a high affinity for both activated and nonactivated integrin. No binding was observed on alpha(4)beta(1) negative cells. Through an analysis of the metal ion dependences of ligand binding, several unexpected findings about alpha(4)beta(1) function were made. First, we observed that Ca(2+) binding to alpha(4)beta(1) was stimulated by the addition of BIO7662. From solution binding studies on purified alpha(4)beta(1), two types of Ca(2+)-binding sites were identified, one dependent upon and the other independent of BIO7662 binding. Second, we observed that the metal ion dependence of ligand binding was affected by the affinity of the ligand for alpha(4)beta(1). ED(50) values for the metal ion dependence of the binding of BIO7762 and the binding of a lower affinity ligand, BIO1211, differed by 2-fold for Mn(2+), 30-fold for Mg(2+), and >1000-fold for Ca(2+). Low Ca(2+) (ED(50) = 5-10 microm) stimulated the binding of BIO7662 to alpha(4)beta(1). The effects of microm Ca(2+) closely resembled the effects of Mn(2+) on alpha(4)beta(1) function. Third, we observed that the rate of BIO7662 binding was dependent on the metal ion concentration and that the ED(50) for the metal ion dependence of BIO7662 binding was affected by the concentration of the BIO7662. These studies point to an even more complex interplay between metal ion and ligand binding than previously appreciated and provide evidence for a three-component coupled equilibrium model for metal ion-dependent binding of ligands to alpha(4)beta(1).


Subject(s)
Integrins/chemistry , Integrins/metabolism , Ions , Ligands , Receptors, Lymphocyte Homing/chemistry , Receptors, Lymphocyte Homing/metabolism , Benzoates/pharmacology , Calcium/metabolism , Calcium/pharmacology , Cations , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Edetic Acid/pharmacology , Humans , Integrin alpha4beta1 , Integrins/antagonists & inhibitors , Jurkat Cells , Kinetics , Magnesium/pharmacology , Manganese/pharmacology , Models, Chemical , Phenylurea Compounds/pharmacology , Protein Binding , Receptors, Lymphocyte Homing/antagonists & inhibitors , Time Factors
5.
Biochemistry ; 40(14): 4359-71, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11284692

ABSTRACT

Post-translational modifications of the developmental signaling protein Sonic hedgehog (Shh) by a long-chain fatty acid at the N-terminus and cholesterol at the C-terminus greatly activate the protein in a cell-based signaling assay. To investigate the structural determinants of this activation phenomenon, hydrophobic and hydrophilic moieties have been introduced by chemical and mutagenic methods to the soluble N-terminal signaling domain of Shh and tested in both in vitro and in vivo assays. A wide variety of hydrophobic modifications increased the potency of Shh when added at the N-terminus of the protein, ranging from long-chain fatty acids to hydrophobic amino acids, with EC(50) values from 99 nM for the unmodified protein to 0.6 nM for the myristoylated form. The N-myristoylated Shh was as active as the natural form having both N- and C-terminal modifications. The degree of activation appears to correlate with the hydrophobicity of the modification rather than any specific chemical feature of the adduct; moreover, substitution with hydrophilic moieties decreased activity. Hydrophobic modifications at the C-terminus of Shh resulted in only a 2-3-fold increase in activity, and no activation was found with hydrophobic modification at other surface positions. The N-terminal modifications did not appear to alter the binding affinity of the Shh protein for the transfected receptor protein, Patched, and had no apparent effect on structure as measured by circular dichroism, thermal denaturation, and size determination. Activation of Desert Hh through modification of its N-terminus was also observed, suggesting that this is a common feature of Hh proteins.


Subject(s)
Proteins/chemistry , Proteins/physiology , Trans-Activators , Up-Regulation , Acyl Coenzyme A/chemistry , Amides , Amino Acid Substitution/genetics , Animals , Cell Line , Circular Dichroism , Cysteine/chemistry , Cysteine/genetics , Ethylmaleimide/chemistry , Fatty Acids/chemistry , Formaldehyde/chemistry , Hedgehog Proteins , Humans , Indicators and Reagents , Intracellular Signaling Peptides and Proteins , Iodoacetamide/analogs & derivatives , Iodoacetamide/chemistry , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Mice, Inbred C3H , Patched Receptors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/genetics , Protein Processing, Post-Translational/genetics , Proteins/genetics , Proteins/metabolism , Receptors, Cell Surface , Signal Transduction/genetics , Spectrometry, Mass, Electrospray Ionization , Sulfhydryl Compounds/chemistry , Thiazoles/chemistry , Thiazoles/metabolism , Thiazolidines , Up-Regulation/genetics
6.
FEBS Lett ; 475(1): 1-6, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10854846

ABSTRACT

Linear free energy relationships can be used to link the changes in rate constant for a reaction to changes in the equilibrium caused by alterations in structure. While they have most often been used in the analysis of chemical reactions, they have also been employed to resolve questions in enzymology and protein folding. Here we analyze the reaction of a serpin with a panel of six serine proteinases, and observe that a linear free energy relationship exists between the true second-order rate constant for reaction, k(inh), and the inhibition constant, K(I), indicating that formation of the covalent serpin-enzyme complex may be reversible.


Subject(s)
Endopeptidases/chemistry , Endopeptidases/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Serpins/chemistry , Serpins/metabolism , Animals , Energy Metabolism , Humans , Kinetics , Protein Folding , Structure-Activity Relationship , Substrate Specificity
7.
Biochemistry ; 39(10): 2538-51, 2000 Mar 14.
Article in English | MEDLINE | ID: mdl-10704203

ABSTRACT

A systematic mutational analysis of human interferon-beta-1a (IFN-beta) was performed to identify regions on the surface of the molecule that are important for receptor binding and for functional activity. The crystal structure of IFN-beta-1a was used to design a panel of 15 mutant proteins, in each of which a contiguous group of 2-8 surface residues was mutated, in most instances to alanine. The mutants were analyzed for activity in vitro in antiviral and in antiproliferation assays, and for their ability to bind to the type I IFN (ifnar1/ifnar2) receptor on Daudi cells and to a soluble ifnar2 fusion protein (ifnar2-Fc). Abolition of binding to ifnar2-Fc for mutants A2, AB1, AB2, and E established that the ifnar2 binding site on IFN-beta comprises parts of the A helix, the AB loop, and the E helix. Mutations in these areas, which together define a contiguous patch of the IFN-beta surface, also resulted in reduced affinity for binding to the receptor on cells and in reductions in activity of 5-50-fold in functional assays. A second receptor interaction site, concluded to be the ifnar1 binding site, was identified on the opposite face of the molecule. Mutations in this region, which encompasses parts of the B, C, and D helices and the DE loop, resulted in disparate effects on receptor binding and on functional activity. Analysis of antiproliferation activity as a function of the level of receptor occupancy allowed mutational effects on receptor activation to be distinguished from effects on receptor binding. The results suggest that the binding energy from interaction of IFN-beta with ifnar2 serves mainly to stabilize the bound IFN/receptor complex, whereas the binding energy generated by interaction of certain regions of IFN-beta with ifnar1 is not fully expressed in the observed affinity of binding but instead serves to selectively stabilize activated states of the receptor.


Subject(s)
Interferon-beta/genetics , Interferon-beta/metabolism , Receptors, Interferon/metabolism , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Antiviral Agents/metabolism , COS Cells , DNA Mutational Analysis , Genetic Vectors/metabolism , Growth Inhibitors/physiology , Humans , Interferon beta-1a , Interferon-beta/biosynthesis , Interferon-beta/physiology , Membrane Proteins , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Mapping , Protein Binding/genetics , Protein Structure, Tertiary , Receptor, Interferon alpha-beta , Structure-Activity Relationship , Tumor Cells, Cultured
8.
Biochemistry ; 38(45): 14868-80, 1999 Nov 09.
Article in English | MEDLINE | ID: mdl-10555969

ABSTRACT

The role of the zinc site in the N-terminal fragment of human Sonic hedgehog (ShhN) was explored by comparing the biophysical and functional properties of wild-type ShhN with those of mutants in which the zinc-coordinating residues H140, D147, and H182, or E176 which interacts with the metal ion via a bridging water molecule, were mutated to alanine. The wild-type and E176A mutant proteins retained 1 mol of zinc/mol of protein after extensive dialysis, whereas the H140A and D147A mutants retained only 0.03 and 0.05 mol of zinc/mol of protein, respectively. Assay of the wild-type and mutant proteins in two activity assays indicated that the wild-type and E176A mutant proteins had similar activity, whereas the H140A and D147A mutants were significantly less active. These assays also indicated that the H140A and D147A mutants were susceptible to proteolysis. CD, fluorescence, and (1)H NMR spectra of the H140A, D147A, and E176A mutants measured at 20 or 25 degrees C were very similar to those observed for wild-type ShhN. However, CD measurements at 37 degrees C showed evidence of some structural differences in the H140A and D147A mutants. Guanidine hydrochloride (GuHCl) denaturation studies revealed that the loss of zinc from the H140A and D147A mutants destabilized the folded proteins by approximately 3.5 kcal/mol, comparable to the effect of removing zinc from wild-type ShhN by treatment with EDTA. Thermal melting curves of wild-type ShhN gave a single unfolding transition with a midpoint T(m) of approximately 59 degrees C, whereas both the H140A and D147A mutants displayed two distinct transitions with T(m) values of 37-38 and 52-54 degrees C, similar to that observed for EDTA-treated wild-type ShhN. Addition of zinc to the H140A and D147A mutants resulted in a partial restoration of stability against thermal and GuHCl denaturation. The ability of these mutants to bind zinc was confirmed using a fluorescence-based binding assay that indicated that they bound zinc with K(d) values of approximately 1.6 and approximately 15 nM, respectively, as compared to a value of

Subject(s)
Proteins/chemistry , Trans-Activators , Zinc/chemistry , Alkaline Phosphatase/biosynthesis , Amino Acid Substitution , Animals , Chick Embryo , Circular Dichroism , Gene Expression Regulation, Enzymologic , Hedgehog Proteins , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C3H , Models, Chemical , Mutagenesis, Site-Directed , Protein Conformation , Proteins/genetics , Structure-Activity Relationship
9.
J Immunol Methods ; 227(1-2): 121-35, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10485260

ABSTRACT

Upon treatment with protein therapeutics, a subset of patients will typically develop antibodies against the drug. These anti-drug antibodies can be of concern because they have the potential to alter the drug's therapeutic activity. In the case of relapsing-remitting multiple sclerosis (RRMS) patients receiving recombinant interferon-beta (IFN-beta), those receiving BETASERON (IFN-beta-1b; E. coli expressed, non-glycosylated, des-Met-1, Cys17Ser recombinant IFN-beta) have a higher incidence of IFN-beta specific antibodies compared to those receiving AVONEX (IFN-beta-1a; mammalian cell-expressed, natural sequence, glycosylated recombinant IFN-beta). The current study reports the development and characterization of ELISAs that detect distinct components of the anti-IFN-beta response in patients' sera, and therefore can potentially be used to characterize the composition of the anti-IFN-beta antibody response. ELISAs were developed using a constant detecting reagent but a variety of IFN-beta-derived test antigens (e.g., native IFN-beta, biotinylated IFN-beta, IFN-beta peptides) and capture methods. Assays were characterized using serum samples from a small number of patients treated with recombinant IFN-beta (either BETASERON or AVONEX). Assays in which IFN-beta was captured via a specific mAb, or in which biotinylated IFN-beta was captured via streptavidin, detected serum antibodies that recognize IFN-beta in its native structural state. In contrast, assays in which IFN-beta was coated directly onto the assay plates detected antibodies that recognize forms of IFN-beta possessing a folded structure distinct from the native structure. Certain epitopes present on native IFN-beta were not represented in these assays in which the test antigen was directly coated on plastic. Antibodies specific for linear epitopes could be detected using linear peptides as test antigens; the locations of these epitopes were mapped by reference to the X-ray crystal structure of IFN-beta-1a. Together, these data show that the mode of antigen presentation employed in IFN-beta ELISAs determines which antibody specificities are detected, and can affect whether or not a given serum sample is identified as positive for anti-IFN-beta antibodies. As a consequence, screening samples in a single ELISA format presenting IFN-beta in a non-native form may lead to underestimation of the incidence of IFN-beta treated MS patients that have generated antibodies specific to the native, active form of the drug.


Subject(s)
Antibodies/blood , Interferon-beta/immunology , Multiple Sclerosis/therapy , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitopes , Humans , Interferon beta-1a , Interferon beta-1b , Interferon-beta/therapeutic use , Multiple Sclerosis/immunology , Recombinant Proteins/immunology
10.
J Biol Chem ; 274(19): 13167-75, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224072

ABSTRACT

We have used the highly specific alpha4beta1 inhibitor 4-((N'-2-methylphenyl)ureido)-phenylacetyl-leucine-aspartic acid-valine-proline (BIO1211) as a model LDV-containing ligand to study alpha4beta1 integrin-ligand interactions on Jurkat cells under diverse conditions that affect the activation state of alpha4beta1. Observed KD values for BIO1211 binding ranged from a value of 20-40 nM in the non-activated state of the integrin that exists in 1 mM Mg2+, 1 mM Ca2+ to 100 pM in the activated state seen in 2 mM Mn2+ to 18 pM when binding was measured after co-activation by 2 mM Mn2+ plus 10 microgram/ml of the integrin-activating monoclonal antibody TS2/16. The large range in KD values was governed almost exclusively by differences in the dissociation rates of the integrin-BIO1211 complex, which ranged from 0.17 x 10(-4) s-1 to >140 x 10(-4) s-1. Association rate constants varied only slightly under the same conditions, all falling in the narrow range from 0.9 to 2.7 x 10(6) M-1 s-1. The further increase in affinity observed upon co-activation by divalent cations and TS2/16 compared with that observed at saturating concentrations of metal ions or TS2/16 alone indicates that the mechanism by which these factors bring about activation are distinct and identified a previously unrecognized high affinity state on alpha4beta1 that had not been detected by conventional assay methods. Similar changes in affinity were observed when the binding properties of vascular cell adhesion molecule-1 and CS1 to alpha4beta1 were studied, indicating that the different affinity states detected with BIO1211 are an inherent property of the integrin.


Subject(s)
Integrins/metabolism , Oligopeptides/metabolism , Receptors, Lymphocyte Homing/metabolism , Cations, Divalent , Humans , Integrin alpha4beta1 , Integrins/antagonists & inhibitors , Jurkat Cells , Kinetics , Ligands , Protein Binding , Receptors, Lymphocyte Homing/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/metabolism
11.
Chem Biol ; 6(4): R107-18, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10099129

ABSTRACT

A number of reports describe small peptides, and even bona fide small organic molecules, that activate homodimeric cytokine receptors and show cytokine-like activity in vitro and in vivo. These cases can be examined in light of the mechanistic and thermodynamic principles that govern cytokine-receptor activation.


Subject(s)
Cytokines/metabolism , Molecular Mimicry , Receptors, Cytokine/metabolism , Cytokines/chemistry , Dimerization , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Peptides/pharmacology , Receptors, Cytokine/agonists , Receptors, Cytokine/antagonists & inhibitors
12.
Biochemistry ; 38(9): 2849-59, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10052957

ABSTRACT

We have investigated the mechanism by which the complement protease, Factor D, achieves its high specificity for the cleavage of Factor B in complex with C3(H2O). Kinetic experiments showed that Factor B and C3(H2O) associate with a KD of >/=2.5 microM and that Factor D acts on this complex with a second-order rate constant of kcat/KM >/= 2 x 10(6) M-1 s-1, close to the rate of a diffusion-controlled reaction for proteins of this size. In contrast, Factor D, which is a member of the trypsin family of serine proteases, was 10(3)-10(4)-fold less active than trypsin toward both thioester and p-nitroanilide substrates containing an arginine at P1. Furthermore, peptides spanning the Factor B cleavage site were not detectably cleaved by Factor D (kcat/KM /=9 kcal/mol of binding energy to stabilize the transition state for reaction. In support of this, we demonstrate that chemical modification of Factor D at a single lysine residue that is distant from the active site abolishes the activity of the enzyme toward Factor B while not affecting activity toward small synthetic substrates. We propose that Factor D may exemplify a special case of the induced fit mechanism in which the requirement for conformational activation of the enzyme results in a substantial increase in substrate specificity.


Subject(s)
Complement Factor D/chemistry , Serine Endopeptidases/chemistry , Acetylation , Anilides/chemistry , Arginine/analogs & derivatives , Arginine/chemistry , Binding Sites , Biotinylation , Chromogenic Compounds/chemistry , Complement Factor B/antagonists & inhibitors , Complement Factor B/chemistry , Complement Factor D/metabolism , Enzyme Activation , Humans , Hydrolysis , Models, Molecular , Oligopeptides/chemistry , Serine Endopeptidases/metabolism , Substrate Specificity
13.
Cell Adhes Commun ; 7(3): 267-79, 1999.
Article in English | MEDLINE | ID: mdl-10626910

ABSTRACT

Quantitative analysis of binding of the bivalent recombinant soluble fusion protein, LFA-3/IgG1, shows that the fusion protein binds to human CD2+ PBLs primarily through low affinity (KD approximately 140 microM) but also through high avidity (90 nM) interactions. The concentration dependence for LFA-3/IgG1 PBL binding took the form of two overlapping bell-shaped curves separated by a clear and reproducible minimum. This was accounted for in part by minor heterogeneity in the LFA-3/IgG1 preparations, and potentially by the ability of the ligand to bind to both CD2 and Fc receptors (FcR), best evidenced by the distinct binding properties of the fusion protein to NK and T cells. The low affinity LFA-3/ IgG1 binding to T cells is consistent with binding to CD2 only, and is in agreement with the low affinity reported for interactions between soluble forms of LFA-3 and CD2 by surface plasmon resonance technology. Moreover, as the low affinity determinations are similar for CD2 on resting and activated T cells, although the CD2 molecule has been reported to be altered to reveal new epitopes upon T cell activation, the binding data argue against multiple cell activation-dependent affinity states of CD2 for LFA-3 binding. This is distinct from that observed with other adhesion partners, and suggests that the different adhesion pathways utilize distinct mechanisms to mediate cell adhesion.


Subject(s)
CD2 Antigens/metabolism , CD58 Antigens/metabolism , Immunoglobulin G/metabolism , Lymphocyte Activation , T-Lymphocyte Subsets/metabolism , Animals , Flow Cytometry , Humans , Jurkat Cells , Kinetics , Ligands , Macaca fascicularis , Mice , Mice, Transgenic , Papio , Protein Binding , Receptors, Fc/metabolism , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/immunology
14.
Cell Mol Life Sci ; 54(11): 1203-16, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9849615

ABSTRACT

Interferons (IFNs) are potent extracellular protein mediators of host defence and homoeostasis. This article reviews the structure of human IFN-beta (HuIFN-beta), in particular in relation to its activity. The recently determined crystal structure of HuIFN-beta provides a framework for understanding of the mechanism of differentiation of type I IFNs by their common receptor. Insights are generated by comparison with the structures of other type I IFNs and from the interpretation of existing mutagenesis data. The details of the observed carbohydrate structure, together with biochemical data, implicate the glycosylation of HuIFN-beta, which is uncommon among type I IFNs, as an important factor in the solubility, stability and, consequently, activity of the protein. Finally, these structural implications are discussed in the context of the clinical use of HuIFN-beta.


Subject(s)
Interferon-beta/chemistry , Crystallography, X-Ray , Dimerization , Glycosylation , Humans , Interferon-beta/therapeutic use , Models, Molecular , Protein Binding , Protein Structure, Secondary , Receptors, Interferon/chemistry
15.
Proc Natl Acad Sci U S A ; 95(22): 13165-70, 1998 Oct 27.
Article in English | MEDLINE | ID: mdl-9789059

ABSTRACT

The anti-common gamma chain (gammac) mAb CP.B8 is shown to inhibit interleukin 4 (IL-4)-dependent proliferation of phytohemagglutinin (PHA) activated T cells noncompetitively with respect to cytokine by blocking the IL-4-induced heterodimerization of IL-4Ralpha and gammac receptor chains. Affinities for the binding of IL-4 to Cos-7 cells transfected with huIL-4Ralpha, and to PHA blasts expressing both IL-4Ralpha and gammac, were used to estimate the affinity of the key interaction between gammac and the binary IL-4Ralpha.IL-4 complex on the cell surface. This affinity was defined in terms of the dimensionless ratio [IL-4Ralpha.IL-4.gammac]/[IL-4Ralpha.IL-4], which we designate KR. The results show that on PHA blasts this interaction is relatively weak; KR approximately 9, implying that approximately 10% of the limiting IL-4Ralpha chain remains free of gammac even at saturating concentrations of IL-4. This quantitative treatment establishes KR as a key measure of the coupling between ligand binding and receptor activation, providing a basis for functional distinctions between different receptors that are activated by ligand-induced receptor dimerization.


Subject(s)
Lymphocyte Activation , Receptor Cross-Talk , Receptors, Cytokine/physiology , Receptors, Interleukin-4/physiology , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/pharmacology , Cell Membrane/immunology , Cells, Cultured , Humans , Interleukin-4/pharmacology , Interleukin-4/physiology , Kinetics , Mice , Models, Immunological , T-Lymphocytes/drug effects
16.
Biochemistry ; 37(41): 14337-49, 1998 Oct 13.
Article in English | MEDLINE | ID: mdl-9772159

ABSTRACT

A murine monoclonal antibody, CP.B8, specific for the extracellular portion of the human common gamma (gammac) chain, and its Fab fragment are shown to block the binding of IL-2 to COS-7 cells transfected with the cDNA for the full-length IL-2 receptor beta (IL-2Rbeta) and gammac chains, components which together comprise the intermediate affinity IL-2 receptor (IL-2R) expressed on the surface of resting T cells, NK cells, and on certain intestinal epithelial cells. To investigate the mechanism of this inhibition, the extracellular portions of the IL-2Rbeta and gammac chains were expressed and purified, and their interactions with each other and with IL-2 were studied by gel filtration and by surface plasmon resonance (SPR). By gel filtration, a stable ternary complex was formed by association of the three proteins, while no stable binary complexes were detected between any two of the three proteins. By SPR analysis, IL-2 was shown to associate rapidly with IL-2Rbeta, forming a binary complex with an equilibrium dissociation constant (Kd) of 800 nM, which permitted subsequent association of the gammac chain. Dissociation of the IL-2/IL-2Rbeta/gammac chain complex was significantly slower than dissociation of the IL-2/IL-2Rbeta complex. Using these model systems, we tested the ability of mAb CP.B8 to inhibit the association of the gammac chain with IL-2 and IL-2Rbeta. By gel filtration, mAb CP.B8 formed a stable complex with the gammac chain, preventing its association with IL-2 and IL-2Rbeta. MAb CP.B8 was also capable of dissociating the gammac chain already complexed with IL-2 and IL-2Rbeta. SPR analysis confirmed these findings and showed, in addition, that the Fab fragment of CP.B8 was also capable of inhibiting the association of the gammac chain with the IL-2/IL-2Rbeta complex. We conclude that mAb CP.B8 blocks the second step in the formation of the intermediate affinity IL-2R on the surface of transfected COS-7 cells by binding at or close to a region on the gammac chain that is involved in contact with IL-2 and/or IL-2Rbeta.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Receptors, Interleukin-2/antagonists & inhibitors , Receptors, Interleukin-2/immunology , Amino Acid Sequence , Animals , Biosensing Techniques , Chromatography, Gel , Histidine/genetics , Humans , Immunoglobulin Fab Fragments/pharmacology , Interleukin-2/antagonists & inhibitors , Interleukin-2/immunology , Interleukin-2/metabolism , Macromolecular Substances , Mice , Molecular Sequence Data , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Solubility , Transfection
17.
J Biol Chem ; 273(33): 20982-91, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9694848

ABSTRACT

SERP-1 is a myxoma virus-encoded serpin, secreted from infected cells, that is required for virulence and has anti-inflammatory activity. We report that purified recombinant SERP-1 forms SDS-stable complexes with urokinase-type plasminogen activator (uPA), tissue-type plasminogen activator (tPA), plasmin, thrombin, and factor Xa. N-terminal sequencing confirmed Arg319-Asn320 as the site of reaction. Mutation of these residues to Ala-Ala abolished inhibitory activity but had no effect on the specific cleavage at Thr315-Leu316 seen with elastase and with cathepsin G. Kinetic analysis of the reactions with uPA, tPA, plasmin, thrombin, Xa, and C1s showed second-order rate constants to vary over 3 logs, from kinh = 3 x 10(5) M-1 s-1 with thrombin to approximately 600 M-1 s-1 with C1s, while steady-state inhibition constants ranged from KI = 10 pM with thrombin to approximately 100 nM with C1s. Stoichiometries of inhibition varied between SI = 1.4 +/- 0.1 for uPA to SI = 13 +/- 3 for thrombin. Analysis of the variations in inhibition kinetics shows that when serpins act at low concentrations, comparable with the target protease or with KI (as appears likely for SERP-1 in vivo), inhibitory specificity becomes less dominated by kinh and is increasingly dependent on partitioning within the branched reaction mechanism and on the lifetime of the inhibited complex.


Subject(s)
Myxoma virus/metabolism , Serpins/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cell Line , DNA Primers , Factor Xa/metabolism , Fibrinolysin/metabolism , Inflammation , Mutagenesis, Site-Directed , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serpins/chemistry , Serpins/genetics , Thrombin/metabolism , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics
18.
Pharm Res ; 15(4): 641-9, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9587963

ABSTRACT

PURPOSE: Two recombinant IFN-beta products have been approved for the treatment of multiple sclerosis, a glycosylated form with the predicted natural amino acid sequence (IFN-beta-1a) and a non-glycosylated form that has a Met-1 deletion and a Cys-17 to Ser mutation (IFN-beta-1b). The structural basis for activity differences between IFN-beta-1a and IFN-beta-1b, is determined. METHODS: In vitro antiviral, antiproliferative and immunomodulatory assays were used to directly compare the two IFN-beta products. Size exclusion chromatography (SEC), SDS-PAGE, thermal denaturation, and X-ray crystallography were used to examine structural differences. RESULTS: IFN-beta-1a was 10 times more active than IFN-beta-1b with specific activities in a standard antiviral assay of 20 x 10(7) IU/mg for IFN-beta-1a and 2 x 10(7) IU/mg for IFN-beta-1b. Of the known structural differences between IFN-beta-1a and IFN-beta-1b, only glycosylation affected in vitro activity. Deglycosylation of IFN-beta-1a produced a decrease in total activity that was primarily caused by the formation of an insoluble disulfide-linked IFN precipitate. Deglycosylation also resulted in an increased sensitivity to thermal denaturation. SEC data for IFN-beta-1b revealed large, soluble aggregates that had reduced antiviral activity (approximated at 0.7 x 10(7) IU/mg). Crystallographic data for IFN-beta-1a revealed that the glycan formed H-bonds with the peptide backbone and shielded an uncharged surface from solvent exposure. CONCLUSIONS: Together these results suggest that the greater biological activity of IFN-beta-1a is due to a stabilizing effect of the carbohydrate on structure.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antiviral Agents/pharmacology , Interferon-beta/pharmacology , Recombinant Proteins/pharmacology , Adjuvants, Immunologic/chemistry , Base Sequence , Cardiovirus Infections/drug therapy , Cell Division/drug effects , Cell Line , Cysteine/chemistry , Encephalomyocarditis virus/drug effects , Glycosylation , Humans , Interferon beta-1a , Interferon beta-1b , Interferon-beta/chemistry , Major Histocompatibility Complex/drug effects , Molecular Sequence Data , Mutation , Recombinant Proteins/chemistry , Serine/chemistry
19.
Proc Natl Acad Sci U S A ; 94(12): 6238-43, 1997 Jun 10.
Article in English | MEDLINE | ID: mdl-9177201

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF)-dependent activation of the tyrosine kinase receptor RET is necessary for kidney and enteric neuron development, and mutations in RET are associated with human diseases. Activation of RET by GDNF has been shown to require an accessory component, GDNFR-alpha (RETL1). We report the isolation and characterization of rat and human cDNAs for a novel cell-surface associated accessory protein, RETL2, that shares 49% identity with RETL1. Both RETL1 and RETL2 can mediate GDNF dependent phosphorylation of RET, but they exhibit different patterns of expression in fetal and adult tissues. The most striking differences in expression observed were in the adult central and peripheral nervous systems. In addition, the mechanisms by which the two accessory proteins facilitate the activation of RET by GDNF are quite distinct. In vitro binding experiments with soluble forms of RET, RETL1 and RETL2 demonstrate that while RETL1 binds GDNF tightly to form a membrane-associated complex which can then interact with RET, RETL2 only forms a high affinity complex with GDNF in the presence of RET. This strong RET dependence of the binding of RETL2 to GDNF was confirmed by FACS analysis on RETL1 and RETL2 expressing cells. Together with the recent discovery of a GDNF related protein, neurturin, these data raise the possibility that RETL1 and RETL2 have distinctive roles during development and in the nervous system of the adult. RETL1 and RETL2 represent new candidate susceptibility genes and/or modifier loci for RET-associated diseases.


Subject(s)
Brain/metabolism , Drosophila Proteins , Nerve Tissue Proteins/pharmacology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Spinal Cord/metabolism , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Embryo, Mammalian , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Kidney/metabolism , Kinetics , Molecular Sequence Data , Nerve Growth Factors/pharmacology , Organ Specificity , Protein Binding , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-ret , Rats , Rats, Wistar , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Sequence Alignment
20.
J Med Chem ; 40(7): 1130-5, 1997 Mar 28.
Article in English | MEDLINE | ID: mdl-9089334

ABSTRACT

We report the use of structure-based drug design to create a selective erbB-1 (a.k.a. epidermal growth factor receptor) and erbB-2 (a.k.a. neu/her2 growth factor receptor) tyrosine kinase inhibitor. Using the X-ray crystal structure of the ternary complex of the cAMP-dependent Ser/Thr kinase together with a sequence alignment of the catalytic domains of a representative set of Ser/Thr and Tyr protein kinases, we have examined the nucleotide binding site for potential positions to attach an irreversible inhibitor. This information, combined with homology modeling of the erbB-1 and erbB-2 tyrosine kinase catalytic domains, has led to the identification of Cys797 of erbB1 and Cys805 of erbB2, which are structurally equivalent to Glu127 in the cAMP dependant Ser/Thr kinase as potential target residues. The X-ray structure of the cAMP Ser/Thr kinase shows Glu127 to be involved in a hydrogen-bonding interaction with the 2'-OH of the ribose portion of ATP. Using molecular modeling, it was predicted that the Cys side chains in erbB-1 and erbB-2 performed an analogous role, and it was postulated that the replacement of the 2'-OH of adenosine with a thiol might allow for a covalent bond to form. Since only erbB-1 and erbB-2 have a Cys at this position, the inhibitor should be selective. This model was subsequently tested experimentally by chemical synthesis of 2'-thioadenosine and assayed against the full length erbB-1 receptor and the catalytic domains of erbB-2, insulin receptor, beta-PDGF receptor, and the FGF receptor. Our results show that thioadenosine covalently inactivates erbB-1 with a second-order rate constant of k(max)/K(S) = 2000 +/- 500 M(-1) s(-1). Inactivation is fully reversed by 1 mM dithiothreitol, suggesting that inactivation involves the modification of a cysteine residue at the active site, presumably Cys797. The rate of inactivation saturates with increasing thioadenosine concentrations, suggesting that inactivation occurs through initial formation of a noncovalent complex with K(D) = 1.0 +/- 0.3 microM, followed by the slow formation of a disulfide bond with a rate constant of k(max) = (2.3 +/- 0.2) x 10(-3) s(-1). This approach may have application in the design of selective irreversible inhibitors against other members of the kinase family.


Subject(s)
Enzyme Inhibitors/chemical synthesis , ErbB Receptors/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Drug Design , Enzyme Inhibitors/pharmacology , Humans , Molecular Sequence Data , Protein-Tyrosine Kinases/chemistry , Sequence Homology, Amino Acid , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...