Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Struct Biol ; 185(2): 203-14, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23542582

ABSTRACT

Modern strategies in radio-immuno therapy and in vivo imaging require robust, small, and specific ligand-binding proteins. In this context we have previously developed artificial lipocalins, so-called Anticalins, with high binding activity toward rare-earth metal-chelate complexes using combinatorial protein design. Here we describe further improvement of the Anticalin C26 via in vitro affinity maturation to yield CL31, which has a fourfold slower dissociation half-life above 2h. Also, we present the crystallographic analyses of both the initial and the improved Anticalin, providing insight into the molecular mechanism of chelated metal binding and the role of amino acid substitutions during the step-wise affinity maturation. Notably, one of the four structurally variable loops that form the ligand pocket in the lipocalin scaffold undergoes a significant conformational change from C26 to CL31, acting as a lid that closes over the accommodated metal-chelate ligand. A systematic mutational study indicated that further improvement of ligand affinity is difficult to achieve while providing clues on the contribution of relevant side chains in the engineered binding pocket. Unexpectedly, some of the amino acid replacements led to strong increases - more then 10-fold - in the yield of soluble protein from periplasmic secretion in Escherichia coli.


Subject(s)
Lipocalins/chemistry , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Chelating Agents/chemistry , Crystallography, X-Ray , Directed Molecular Evolution , Lipocalins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Pentetic Acid/chemistry , Protein Binding , Protein Stability , Radiopharmaceuticals/chemistry , Solubility , Yttrium/chemistry
3.
Br J Pharmacol ; 134(6): 1113-36, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11704631

ABSTRACT

The cloning of the so-called 'parathyroid hormone-related protein' (PTHrP) in 1987 was the result of a long quest for the factor which, by mimicking the actions of PTH in bone and kidney, is responsible for the hypercalcemic paraneoplastic syndrome, humoral calcemia of malignancy. PTHrP is distinct from PTH in a number of ways. First, PTHrP is the product of a separate gene. Second, with the exception of a short N-terminal region, the structure of PTHrP is not closely related to that of PTH. Third, in contrast to PTH, PTHrP is a paracrine factor expressed throughout the body. Finally, most of the functions of PTHrP have nothing in common with those of PTH. PTHrP is a poly-hormone which comprises a family of distinct peptide hormones arising from post-translational endoproteolytic cleavage of the initial PTHrP translation products. Mature N-terminal, mid-region and C-terminal secretory forms of PTHrP are thus generated, each of them having their own physiologic functions and probably their own receptors. The type 1 PTHrP receptor, binding both PTH(1-34) and PTHrP(1-36), is the only cloned receptor so far. PTHrP is a PTH-like calciotropic hormone, a myorelaxant, a growth factor and a developmental regulatory molecule. The present review reports recent aspects of PTHrP pharmacology and physiology, including: (a) the identification of new peptides and receptors of the PTH/PTHrP system; (b) the recently discovered nuclear functions of PTHrP and the role of PTHrP as an intracrine regulator of cell growth and cell death; (c) the physiological and developmental actions of PTHrP in the cardiovascular and the renal glomerulo-vascular systems; (d) the role of PTHrP as a regulator of pancreatic beta cell growth and functions, and, (e) the interactions of PTHrP and calcium-sensing receptors for the control of the growth of placental trophoblasts. These new advances have contributed to a better understanding of the pathophysiological role of PTHrP, and will help to identify its therapeutic potential in a number of diseases.


Subject(s)
Islets of Langerhans/metabolism , Parathyroid Hormone/physiology , Proteins/physiology , Receptors, Parathyroid Hormone/physiology , Animals , Apoptosis , Calcium-Binding Proteins/metabolism , Cardiovascular System/metabolism , Cell Nucleus/metabolism , Female , Humans , Kidney/metabolism , Mice , Nuclear Localization Signals , Parathyroid Hormone/genetics , Parathyroid Hormone-Related Protein , Placenta/metabolism , Pregnancy , Proteins/genetics , Rats , Receptors, Parathyroid Hormone/genetics , Trophoblasts/metabolism
4.
FASEB J ; 15(3): 707-18, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11259389

ABSTRACT

In previous studies, added parathyroid hormone-related protein (PTHrP) inhibits whereas transfected PTHrP stimulates the proliferation of A10 aortic smooth muscle cells by nuclear translocation of the peptide. In the present studies, we asked whether these paradoxical trophic actions of PTHrP occur in smooth muscle cells (SMC) cultured from small intrarenal arteries of, and whether they are altered in, 12-wk-old spontaneously hypertensive rats (SHR) as compared to normotensive Wistar-Kyoto (WKY) rats. SHR cells grew faster than WKY cells. PTHrP transcript was increased in SHR-derived cells whereas PTH1 receptor (PTH1R) transcripts were similar in both cell lines. In both strains of cells, stable transfection with human PTHrP(1-139) cDNA did not further induce proliferation, suggesting maximal effect of endogenous PTHrP in wild cells. In contrast, transfection with antisense hPTHrP(1-139) cDNA, which abolished PTHrP mRNA, decreased WKY but increased SHR cell proliferation. Added PTHrP(1-36) (1-100 pM) decreased WKY and increased SHR cell proliferation. Additional studies indicated that the preferential coupling of PTH1-R to G-protein Gi was responsible for the proliferative effect of exogenous PTHrP in SHR cells. Moreover, PTHrP was detected in the nucleolus of a fraction of WKY and SHR renal SMC, in vitro as well as in situ, suggesting that the nucleolar translocation of PTHrP might be involved in the proliferative effects of endogenous PTHrP. In renovascular SMC, added PTHrP is antimitogenic, whereas endogenously produced PTHrP is mitogenic. These paradoxical effects of PTHrP on renovascular SMC proliferation appear to be reversed in the SHR model of genetic hypertension. A new concept emerges from these results, according to which a single molecule may have opposite effects on VSMC proliferation under physiological and pathophysiological conditions.


Subject(s)
Cell Division/drug effects , Hypertension/pathology , Kidney/blood supply , Muscle, Smooth, Vascular/drug effects , Proteins/pharmacology , Receptors, Parathyroid Hormone/metabolism , Animals , Arteries/anatomy & histology , Blotting, Western , Cells, Cultured , Cholera Toxin/pharmacology , Cloning, Molecular , Disease Models, Animal , Humans , Immunohistochemistry , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Parathyroid Hormone-Related Protein , Proteins/genetics , Proteins/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Virulence Factors, Bordetella/pharmacology
5.
J Mol Biol ; 293(3): 613-27, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10543954

ABSTRACT

Substitution of the native variable region-1 (VR1/37-loop) of thrombin by the corresponding VR1 of tissue-type plasminogen activator (thrombin-VR1(tPA)) increases the rate of inhibition by plasminogen activator inhibitor type 1 (PAI-1) by three orders of magnitude, and is thus sufficient to confer PAI-1 specificity to a heterologous serine protease. A structural and kinetical approach to establish the function of the VR1 loop of t-PA in the context of the thrombin-VR1(tPA) variant is described. The crystal structure of thrombin-VR1(tPA) was resolved and showed a conserved overall alpha-thrombin structure, but a partially disordered VR1 loop as also reported for t-PA. The contribution of a prominent charge substitution close to the active site was studied using charge neutralization variants thrombin-E39Q(c39) and thrombin-VR1(tPA)-R304Q(c39), resulting in only fourfold changes in the PAI-1 inhibition rate. Surface plasmon resonance revealed that the affinity of initial reversible complex formation between PAI-1 and catalytically inactive Ser195-->Ala variants of thrombin and thrombin-VR1(tPA) is only increased fivefold, i.e. KD is 652 and 128 nM for thrombin-S195A and thrombin-S195A-VR1(tPA), respectively. We established that the partition ratio of the suicide substrate reaction between the proteases and PAI-1 was largely unaffected in any variant studied. Hirugen allosterically decreases the rate of thrombin inhibition by PAI-1 2.5-fold and of thrombin-VR1(tPA) 20-fold, by interfering with a unimolecular step in the reaction, not by decreasing initial complex formation or by altering the stoichiometry. Finally, kinetic modeling demonstrated that acylation is the rate-limiting step in thrombin inhibition by PAI-1 (k approximately 10(-3) s(-1)) and this kinetic block is alleviated by the introduction of the tPA-VR1 into thrombin (k>1 s(-1)). We propose that the length, flexibility and different charge architecture of the VR1 loop of t-PA invoke an induced fit of the reactive center loop of PAI-1, thereby enhancing the rate of acylation in the Michaelis complex between thrombin-VR1(t-PA) and PAI-1 by more than two orders of magnitude.


Subject(s)
Plasminogen Activator Inhibitor 1/metabolism , Recombinant Fusion Proteins/metabolism , Thrombin/antagonists & inhibitors , Thrombin/metabolism , Tissue Plasminogen Activator/chemistry , Tissue Plasminogen Activator/metabolism , Acylation , Allosteric Regulation/drug effects , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Catalysis , Catalytic Domain , Crystallization , Crystallography, X-Ray , Hirudins/analogs & derivatives , Hirudins/metabolism , Hirudins/pharmacology , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Conformation , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Surface Plasmon Resonance , Thrombin/chemistry , Thrombin/genetics , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/genetics
6.
Eur J Biochem ; 265(2): 598-605, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10504391

ABSTRACT

Two bivalent thrombin inhibitors were synthesized, which consist of a benzamidine-based active-site-blocking segment, a fibrinogen recognition exosite inhibitor and a peptidic linker connecting these fragments. BZA-1 hirulog contains an Nalpha-(2-naphthylsulfonyl)-S-3-amidinophenylalanyl-is onipecotic acid residue connected via the carboxyl group to the linker segment. The active-site-directed moiety of BZA-2 hirulog [Nalpha-(2-naphthylsulfonyl-glutamyl)-R-4-amidinophenylal anyl-piperid ide] was coupled to the linker via the side chain of the glutamic acid. Both BZA-hirulogs contain almost identical linker-exo site inhibitor parts, except for the substitution of a glycine as the first linker residue in BZA-1 hirulog by a gamma-amino butyric acid in BZA-2 hirulog, thus increasing flexibility and linker length by two additional atoms. BZA-1 hirulog showed moderate potency (Ki = 0. 50 +/- 0.14 nM), while BZA-2 hirulog was characterized as a slow, tight binding inhibitor of thrombin (Ki = 0.29 +/- 0.08 pM). The stability in human plasma of both analogs was strongly improved compared with hirulog-1. For BZA-2 hirulog a significantly reduced plasma clearance was observed after intravenous injection in rats compared with BZA-1 hirulog and hirulog-1. The X-ray structure of the BZA-2 hirulog in complex with human alpha-thrombin was solved and confirmed the expected bivalent binding mode.


Subject(s)
Antithrombins/chemistry , Benzamidines/chemistry , Enzyme Inhibitors/chemistry , Guanidines/chemistry , Phenylalanine/analogs & derivatives , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/pharmacology , Kinetics , Models, Molecular , Molecular Sequence Data , Molecular Structure , Peptides/chemical synthesis , Rats
7.
EMBO J ; 18(20): 5453-62, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10523290

ABSTRACT

Gingipains are cysteine proteinases acting as key virulence factors of the bacterium Porphyromonas gingivalis, the major pathogen in periodontal disease. The 1.5 and 2.0 A crystal structures of free and D-Phe-Phe-Arg-chloromethylketone-inhibited gingipain R reveal a 435-residue, single-polypeptide chain organized into a catalytic and an immunoglobulin-like domain. The catalytic domain is subdivided into two subdomains comprising four- and six-stranded beta-sheets sandwiched by alpha-helices. Each subdomain bears topological similarities to the p20-p10 heterodimer of caspase-1. The second subdomain harbours the Cys-His catalytic diad and a nearby Glu arranged around the S1 specificity pocket, which carries an Asp residue to enforce preference for Arg-P1 residues. This gingipain R structure is an excellent template for the rational design of drugs with a potential to cure and prevent periodontitis. Here we show the binding mode of an arginine-containing inhibitor in the active-site, thus identifying major interaction sites defining a suitable pharmacophor.


Subject(s)
Cysteine Endopeptidases/chemistry , Hemagglutinins/chemistry , Adhesins, Bacterial , Amino Acid Sequence , Caspases/chemistry , Catalytic Domain , Crystallography, X-Ray , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Gingipain Cysteine Endopeptidases , Hemagglutinins/genetics , Hemagglutinins/metabolism , Immunoglobulins/chemistry , Models, Molecular , Molecular Sequence Data , Periodontal Diseases/etiology , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/pathogenicity , Protein Folding , Protein Structure, Secondary , Sequence Homology, Amino Acid , Virulence
8.
Proc Natl Acad Sci U S A ; 96(7): 3600-5, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10097083

ABSTRACT

Most known archaeal DNA polymerases belong to the type B family, which also includes the DNA replication polymerases of eukaryotes, but maintain high fidelity at extreme conditions. We describe here the 2.5 A resolution crystal structure of a DNA polymerase from the Archaea Thermococcus gorgonarius and identify structural features of the fold and the active site that are likely responsible for its thermostable function. Comparison with the mesophilic B type DNA polymerase gp43 of the bacteriophage RB69 highlights thermophilic adaptations, which include the presence of two disulfide bonds and an enhanced electrostatic complementarity at the DNA-protein interface. In contrast to gp43, several loops in the exonuclease and thumb domains are more closely packed; this apparently blocks primer binding to the exonuclease active site. A physiological role of this "closed" conformation is unknown but may represent a polymerase mode, in contrast to an editing mode with an open exonuclease site. This archaeal B DNA polymerase structure provides a starting point for structure-based design of polymerases or ligands with applications in biotechnology and the development of antiviral or anticancer agents.


Subject(s)
DNA Polymerase I/chemistry , Protein Structure, Secondary , Thermococcus/enzymology , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Computer Graphics , Conserved Sequence , Crystallography, X-Ray/methods , DNA Polymerase I/metabolism , Enzyme Stability , Hot Temperature , Models, Molecular , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics
9.
Z Naturforsch C J Biosci ; 53(3-4): 273-8, 1998.
Article in English | MEDLINE | ID: mdl-9618941

ABSTRACT

Under certain pathological conditions such as cerebral ischemia and reperfusion the occurrence of free radicals is remarkably increased. However, only very little information is available on their quantitative relevance for the pathophysiology and final outcome of diseases. The aim of the present study was to evaluate the contribution of oxygen radicals in the pathogenesis of a stroke. For this purpose a rat model for stroke was used. Two of three vitamin E deficient groups were repleted with different dosages of DL-alpha-tocopherylacetate. No signs of vitamin E deficiency could be observed. However, the weight gain during repletion was increased in the vitamin E repleted groups. Brain infarction was created by occlusion of the right middle cerebral artery (MCAO) for two hours. After 24 hours the measurements of infarct volumes were taken. The infarct volume of the group with the highest repletion dosage was significantly reduced by 81%. This was also expressed in a higher rate of gait disturbances after MCAO of the deficient animals. The control of vitamin E status exhibited a similar repletion-dependent level in plasma and brain. These results strongly support the hypothesis that the generation of oxygen radicals occurring during reperfusion is an important aspect of the pathophysiological mechanism in brain infarction.


Subject(s)
Brain/pathology , Ischemic Attack, Transient/prevention & control , Ischemic Attack, Transient/physiopathology , Vitamin E Deficiency/physiopathology , Vitamin E/pharmacology , Animals , Blood Pressure , Body Temperature , Body Weight , Brain/drug effects , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Male , Rats , Rats, Inbred Strains , Vitamin E/blood
10.
Int J Clin Pharmacol Ther ; 36(4): 231-8, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9587051

ABSTRACT

The bioequivalence of 2 formulations containing spironolactone and furosemide was determined. The test preparation was Spironolacton 50 plus Heumann tablets, a new generic spironolactone preparation, developed by Heumann Pharma GmbH, the reference preparation was Osyrol 50-Lasix capsules, Hoechst AG. The study was designed as a randomized 2-period, 2-sequence, crossover study. A daily dose of 50 mg spironolactone and 20 mg furosemide was administered over 5 days to 24 healthy volunteers in the fasting state. Plasma samples were assayed for spironolactone, its 2 active metabolites canrenone and 7alpha-thiomethylspirolactone, and furosemide by HPLC. Statistical analysis was performed by ANOVA and by nonparametric methods. Because spironolactone was rapidly eliminated from plasma, its pharmacokinetics could only be evaluated with regard to maximum plasma levels. This parameter did slightly miss the criteria for bioequivalence. For canrenone and 7alpha-thiomethylspirolactone bioequivalence was given. For furosemide the test formulation was found to be equivalent concerning the extent of bioavailability. Bioequivalence with regard to maximum concentrations could not be shown. However, from the point of view of pharmacodynamics, this finding may not necessarily be of clinical relevance.


Subject(s)
Diuretics/pharmacokinetics , Furosemide/pharmacokinetics , Mineralocorticoid Receptor Antagonists/pharmacokinetics , Spironolactone/pharmacokinetics , Adult , Area Under Curve , Canrenone/blood , Chromatography, High Pressure Liquid , Diuretics/administration & dosage , Diuretics/blood , Furosemide/administration & dosage , Furosemide/blood , Humans , Male , Mineralocorticoid Receptor Antagonists/administration & dosage , Mineralocorticoid Receptor Antagonists/blood , Spironolactone/administration & dosage , Spironolactone/analogs & derivatives , Spironolactone/blood , Therapeutic Equivalency
11.
Int J Clin Pharmacol Ther ; 35(8): 334-40, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9266289

ABSTRACT

The aldosterone antagonist spironolactone undergoes extensive and complex biotransformation. For investigation of bioequivalence of 2 oral spironolactone formulations, Spironolacton 50 Heumann and Aldactone 50, the pharmacokinetics and bioequivalence of the parent drug and 2 predominant active metabolites, canrenone and 7 alpha-thiomethylspirolactone, were determined in a 2-way crossover study in 24 young healthy male volunteers after multiple oral dosing of 100 mg once daily. Plasma samples were measured by a newly developed HPLC assay and individual pharmacokinetic parameters of the 3 compounds were calculated by use of noncompartmental techniques. Statistical analysis was performed by ANOVA and nonparametric methods. Spironolactone was rapidly cleared from plasma. Therefore, only Css,max and tss,max were determined. Concerning Css,max bioequivalence was found with 90% classical shortest confidence interval ranging from 80.7-112.4%. The intrasubject variability for Css,max was determined to be 28.1%. Higher and persisting concentrations were observed for the metabolites. For canrenone 90% classical shortest confidence intervals were calculated as 95.4-105.0% for AUCss,tau, as 92.9-105.8% for Css,max, and as 89.1-106.3% for peak trough fluctuation (PTF). In the case of 7 alpha-thiomethylspirolactone the values were 84.2-103.0% for AUCss,tau, 77.0-98.6% for Css,max, and 85.0-100.4% for PTF. For tss,max nonparametric 90% confidence intervals were determined as 0.00 to 1.50 h for spironolactone and canrenone and as -0.50 to 1.00 h for 7 alpha-thiomethylspirolactone. The intraindividual variability was below 30% for all pharmacokinetic parameters in the case of the metabolites. Thus, bioequivalence of the test and the reference formulation can be concluded. The study suggests the inclusion of parent compound and metabolites for bioequivalence testing of spironolactone formulations. Intraindividual subject variability was clearly diminished by investigating bioequivalence under steady-state conditions.


Subject(s)
Mineralocorticoid Receptor Antagonists/metabolism , Mineralocorticoid Receptor Antagonists/pharmacokinetics , Spironolactone/metabolism , Spironolactone/pharmacokinetics , Administration, Oral , Adult , Analysis of Variance , Canrenone/blood , Cross-Over Studies , Humans , Male , Mineralocorticoid Receptor Antagonists/blood , Spironolactone/analogs & derivatives , Spironolactone/blood , Therapeutic Equivalency , Time Factors
12.
Int J Clin Pharmacol Ther ; 35(3): 112-6, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9089000

ABSTRACT

Eighteen male volunteers have been treated with 3 different oral formulations of chlormezanone according to a randomized 3-way change-over design. The test preparation was a tablet (Krewel), reference preparation 1 was a suspension (Krewel), and reference preparation 2 was a tablet (Muskel Trancopal, Sanofi Winthrop GmbH). All preparations contained 200 mg of chlormezanone. Divided in 3 periods the volunteers received single doses of the test and the 2 reference formulations, respectively. Blood samples have been drawn immediately prior to each administration and at 21 sampling points within 144 h after dosing. A wash-out period of 2 weeks was maintained between successive drug doses. Plasma concentrations of chlormezanone were determined by a validated reversed-phase HPLC method with UV detection, with a lower limit of quantification of 0.1 microgram/ml. The following mean values have been obtained for the test preparation: AUC0-infinity 121 micrograms x h/ml, Cmax of 2.9 micrograms/ml at 1.5 h, t1/2 38 h, after administration of the suspension: AUC0-infinity 111 micrograms x h/ml, Cmax 2.6 micrograms/ml, tmax 1.5 h, t1/2 40 h, and after administration of the reference tablet: AUC0-infinity 121 micrograms x h/ml, Cmax 3.0 micrograms/ml, tmax 1.6 h, t1/2 38 h. The test preparation shows a relative bioavailability of 109% compared to the suspension and has been proven to be bioequivalent to the reference tablet with regard to extent and rate of absorption.


Subject(s)
Chlormezanone/pharmacokinetics , Muscle Relaxants, Central/pharmacokinetics , Administration, Oral , Adolescent , Adult , Analysis of Variance , Biological Availability , Chlormezanone/administration & dosage , Chlormezanone/blood , Chromatography, High Pressure Liquid , Cross-Over Studies , Humans , Male , Muscle Relaxants, Central/administration & dosage , Muscle Relaxants, Central/blood , Reference Standards , Regression Analysis , Spectrophotometry, Ultraviolet , Therapeutic Equivalency
13.
Int J Clin Pharmacol Ther ; 35(3): 117-22, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9089001

ABSTRACT

Eighteen volunteers have been treated with different oral formulations of butamirate citrate according to 2 randomized 2-way crossover designs. In the first study (study I) the test preparation was a syrup (Demotussol Hustensirup, Demopharm), and the reference preparation was a syrup already marketed (Sinecod Sirup, Zyma SA). A test preparation (Demotussol Tabletten) was compared to a solution (Demotussol Hustentropfen) in the second study (study II). Within the 2 study periods the volunteers received single 45 mg doses of the test and the reference formulation, respectively. Blood samples have been drawn immediately prior to each administration and at 17 sampling points within 96 h after dosing. A wash-out period of 1 week was maintained between successive drug doses. The plasma concentration of one of the main metabolites, 2-phenylbutyric acid, was determined by a validated reversed-phase HPLC method with UV detection, with a lower limit of quantification of 50 ng/ml. The following mean values have been obtained in study I (syrup preparations) for the test: AUC0-infinity 46.9 micrograms x h/ml, Cmax of 1.77 micrograms/ml at 1.1 h, t1/2 28 h and after administration of the reference: AUC0-infinity 50.4 micrograms x h/ml, Cmax 1.86 micrograms/ml, tmax 1.5 h, t1/2 26 h. In study II the following mean values have been obtained for the test preparation (tablet): AUC0-infinity 54.7 micrograms x h/ml, Cmax of 1.88 micrograms/ml at 1.1 h, t1/2 27 h and for the reference (solution): AUC0-infinity 54.5 micrograms x h/ml, Cmax 1.94 micrograms/ml, tmax 1.1 h, t1/2 26 h. Both preparations have been proven to be bioequivalent to their corresponding references regarding extent and rate of absorption.


Subject(s)
Antitussive Agents/pharmacokinetics , Phenylbutyrates/pharmacokinetics , Administration, Oral , Adolescent , Adult , Antitussive Agents/administration & dosage , Antitussive Agents/blood , Area Under Curve , Biological Availability , Chromatography, High Pressure Liquid , Cross-Over Studies , Female , Half-Life , Humans , Male , Phenylbutyrates/administration & dosage , Phenylbutyrates/blood , Pilot Projects , Reference Standards , Spectrophotometry, Ultraviolet , Therapeutic Equivalency
14.
Structure ; 4(11): 1353-62, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8939759

ABSTRACT

BACKGROUND: The explosive growth in the rate of X-ray determination of protein structures is fuelled largely by the expectation that structural information will be useful for pharmacological and biotechnological applications. For example, there have been intensive efforts to develop orally administrable antithrombotic drugs using information about the crystal structures of blood coagulation factors, including thrombin. Most of the low molecular weight thrombin inhibitors studied so far are based on arginine and benzamidine. We sought to expand the database of information on thrombin-inhibitor binding by studying new classes of inhibitors. RESULTS: We report the structures of three new inhibitors complexed with thrombin, two based on 4-aminopyridine and one based on naphthamidine. We observe several geometry changes in the protein main chain and side chains which accompany inhibitor binding. The two inhibitors based on 4-aminopyridine bind in notably different ways: one forms a water-mediated hydrogen bond to the active site Ser195, the other induces a rotation of the Ser214-Trp215 peptide plane that is unprecedented in thrombin structures. These binding modes also differ in their 'weak' interactions, including CH-O hydrogen bonds and interactions between water molecules and aromatic pi-clouds. Induced-fit structural changes were also seen in the structure of the naphthamidine inhibitor complex. CONCLUSIONS: Protein flexibility and variable water structures are essential elements in protein-ligand interactions. Ligand design strategies that fail to take this into account may overlook or underestimate the potential of lead structures. Further, the significance of 'weak' interactions must be considered both in crystallographic refinement and in analysis of binding mechanisms.


Subject(s)
Antithrombins/chemistry , Serine Proteinase Inhibitors/chemistry , Thrombin/chemistry , 4-Aminopyridine/analogs & derivatives , 4-Aminopyridine/chemistry , Animals , Binding Sites , Cattle , Computer Simulation , Crystallography, X-Ray , Drug Design , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Naphthalenes/chemistry , Structure-Activity Relationship
15.
Arch Pharm (Weinheim) ; 329(2): 73-82, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8851470

ABSTRACT

Biphenyl nitriles 5a-c, terphenyl dinitriles 11a-d, and naphthalene-bis(benzonitrile) 11c were prepared by palladium-catalyzed cross coupling reactions and subsequently converted to biphenyl amidines 8a-c and bis(benzamidines) 4a-e. Among the biphenyl amidines 8 only the meta-derivative 8b inhibits factor Xa and trypsin (Ki = 10 microM). The terphenyl bisamidine 4c does not inhibit factor Xa, trypsin, thrombin, and plasmin, while 4a and 4d are almost equipotent inhibitors of these enzymes (Ki 1-6 microM), and 4b and 4e are selective for trypsin (Ki = 0.2 and 0.3 microM; but Ki > 1 microM for factor Xa, thrombin, and plasmin). X-ray analysis of crystals of 4b complexed with bovine trypsin revealed a unique binding mode: one benzamidino group binds in the S1 site to the side chain carboxylate of Arg189. The central phenyl group is twisted away from the S2/S3 sites and the second amidino group contacts the Asn143 side chain.


Subject(s)
Serine Proteinase Inhibitors/chemical synthesis , Amidines/chemical synthesis , Amidines/pharmacology , Factor Xa Inhibitors , Humans , Serine Proteinase Inhibitors/pharmacology
17.
J Neurochem ; 48(4): 1109-14, 1987 Apr.
Article in English | MEDLINE | ID: mdl-3029327

ABSTRACT

When rat brain membranes were incubated with the benzodiazepine agonist [3H]flunitrazepam or the partial inverse benzodiazepine agonist [3H]Ro 15-4513 in the presence of ultraviolet light one protein (P51) was specifically and irreversibly labeled in cerebellum and at least two proteins (P51 and P55) were labeled in hippocampus. After digestion of the membranes with trypsin, protein P51 was degraded into several peptides. When P51 was photolabeled with [3H]Ro 15-4513, four peptides with apparent molecular weights of 39,000, 29,000, 21,000, and 17,000 were observed. When P51 was labeled with [3H]flunitrazepam, only two peptides with apparent molecular weights of 39,000 and 25,000 were obtained. Protein P55 was only partially degraded by trypsin, and whether it was labeled with [3H]flunitrazepam or [3H]Ro 15-4513 it yielded the same two proteolytic peptides with apparent molecular weights of 42,000 and 45,000. These results support the existence of at least two different benzodiazepine receptor subtypes associated with proteins P51 and P55. The different receptors seem to be differentially protected against treatment with trypsin. In addition, these results indicate that in the benzodiazepine receptor subtype associated with P51 benzodiazepine agonists and partial inverse benzodiazepine agonists irreversibly bind to different parts of the molecule.


Subject(s)
Azides/metabolism , Benzodiazepines/metabolism , Brain/metabolism , Flunitrazepam/metabolism , Peptide Fragments/metabolism , Receptors, GABA-A/metabolism , Trypsin , Animals , Cell Membrane/metabolism , Cerebellum/metabolism , Hippocampus/metabolism , Molecular Weight , Photochemistry , Rats
18.
J Neurochem ; 48(1): 46-52, 1987 Jan.
Article in English | MEDLINE | ID: mdl-3025369

ABSTRACT

Photolabeling of the benzodiazepine receptor, which to date has been done with benzodiazepine agonists such as flunitrazepam, can also be achieved with Ro 15-4513, a partial inverse agonist of the benzodiazepine receptor. [3H]Ro 15-4513 specifically and irreversibly labeled a protein with an apparent molecular weight of 51,000 (P51) in cerebellum and at least two proteins with apparent molecular weights of 51,000 (P51) and 55,000 (P55) in hippocampus. Photolabeling was inhibited by 10 microM diazepam but not by 10 microM Ro 5-4864. The BZ1 receptor-selective ligands CL 218872 and beta-carboline-3-carboxylate ethyl ester preferentially inhibited irreversible binding of [3H]Ro 15-4513 to protein P51. Not only these biochemical results but also the distribution and density of [3H]Ro 15-4513 binding sites in rat brain sections were similar to the findings with [3H]flunitrazepam. Thus, the binding sites for agonists and inverse agonists appear to be located on the same proteins. In contrast, whereas [3H]flunitrazepam is known to label only 25% of the benzodiazepine binding sites in brain membranes, all binding sites are photolabeled by [3H]Ro 15-4513. Thus, all benzodiazepine receptor sites are associated with photolabeled proteins with apparent molecular weights of 51,000 and/or 55,000. In cerebellum, an additional protein (MW 57,000) unrelated to the benzodiazepine receptor was labeled by [3H]Ro 15-4513 but not by [3H]flunitrazepam. In brain sections, this component contributed to higher labeling by [3H]Ro 15-4513 in the granular than the molecular layer.


Subject(s)
Azides/metabolism , Benzodiazepines/metabolism , Brain/metabolism , Receptors, GABA-A/metabolism , Affinity Labels/metabolism , Animals , Autoradiography , Carbolines/pharmacology , Cell Membrane/metabolism , Cerebellum/metabolism , Flunitrazepam/metabolism , Hippocampus/metabolism , Molecular Weight , Photochemistry , Pyridazines/pharmacology , Rats , Receptors, GABA-A/drug effects
19.
J Immunol Methods ; 95(1): 117-22, 1986 Dec 04.
Article in English | MEDLINE | ID: mdl-3023494

ABSTRACT

Bio-Beads SM-2 have previously been used for the removal of non-ionic detergents from protein solutions. Addition of Bio-Beads SM-2 to detergent solubilized antigen significantly enhanced the immobilization of antigen to microtiter wells. Depending on the incubation time used 35-45% of the applied antigen could be immobilized to the microtiter wells. Using this method and a subsequent ELISA procedure it was possible to detect monoclonal antibodies in hybridoma supernatants after coating microtiter wells with 100 microliters of a solution containing 16 ng antigen/ml in the presence of 0.01% Triton X-100.


Subject(s)
Antigens , Polyethylene Glycols , Allergy and Immunology/instrumentation , Antibodies, Monoclonal , Enzyme-Linked Immunosorbent Assay , Octoxynol , Receptors, GABA-A/immunology
20.
J Neurochem ; 46(1): 173-80, 1986 Jan.
Article in English | MEDLINE | ID: mdl-2999333

ABSTRACT

The postnatal development of several proteins irreversibly labeled by [3H]flunitrazepam in membranes from rat cerebral cortex was investigated. It was demonstrated that in the early postnatal days proteins with apparent molecular weights 55,000 and 59,000 were predominantly labeled whereas irreversible labeling of a protein with apparent molecular weight 51,000 started to predominate only in the second postnatal week. Irreversible labeling of another protein with apparent molecular weight 62,000 was weak throughout development. All these proteins seem to be associated with central benzodiazepine receptors. Irreversible labeling at various time points after birth seems to parallel the postnatal development of these proteins, and the different time course of development and different binding properties of the individual proteins support the hypothesis that these proteins are associated with separate and distinct benzodiazepine receptor subtypes. The pharmacological properties of the individual receptor subtypes seem to be fully developed in the early postnatal days, and therefore newborn animals seem to be a good model system for the investigation of properties and function of these various benzodiazepine receptor subtypes.


Subject(s)
Cerebral Cortex/growth & development , Receptors, GABA-A/physiology , Age Factors , Animals , Azides/metabolism , Benzodiazepines/metabolism , Cerebral Cortex/metabolism , Electrophoresis, Polyacrylamide Gel , Flunitrazepam/metabolism , Membrane Proteins/metabolism , Molecular Weight , Pyridazines , Rats , Receptors, GABA-A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...