Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
Blood Cells Mol Dis ; 89: 102570, 2021 07.
Article in English | MEDLINE | ID: mdl-33962291

ABSTRACT

Hemophilia A and hemophilia B are X-linked inherited bleeding disorders caused by a deficiency of coagulation factor VIII and IX, respectively. Standard of care is prophylactic factor replacement therapy; however, the development of neutralizing antibodies against these factors represents serious complications underlining the need for alternative treatment approaches. Human coagulation factor X has a central role within the blood coagulation system making it an attractive target for the development of alternative treatment strategies for patients with hemophilia. This study focuses on a modified variant of the human coagulation factor X with enhanced hemostatic bypass activity due to insertion of a factor IX derived activation sequence. This molecule design leads to the direct activation of the modified factor X protein by factor XIa allowing it to bypass the need for coagulation factor VIIIa/factor IXa. The modified variant was able to correct in-vitro activated partial prothrombin time of human and murine factor VIII/factor IX deficient plasma. Furthermore, reduced blood loss in factor VIII knock-out mice was observed after intravenous application of the modified factor X variant. In conclusion, these data suggest that the factor X variant described here could potentially serve as a bypassing agent independent of the inhibitor status of hemophilia patients. However, more research is needed to further investigate the potential of this molecule.


Subject(s)
Blood Coagulation/drug effects , Factor X/pharmacology , Hemostatics/pharmacology , Animals , Factor X/therapeutic use , Female , Hemophilia A/blood , Hemophilia A/drug therapy , Hemorrhage/blood , Hemorrhage/drug therapy , Hemostatics/therapeutic use , Humans , Male , Mice , Partial Thromboplastin Time , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
2.
Methods Mol Biol ; 2178: 49-62, 2021.
Article in English | MEDLINE | ID: mdl-33128743

ABSTRACT

Monoclonal antibodies (mAbs) are the fastest-growing segment in the drug market with eight of the top 20 selling drugs being mAbs and combined sales of close to 60 billion US$/year. The development of new therapeutic mAbs requires the purification of a large number of candidate molecules during initial screenings, subsequent affinity maturation campaigns, and finally the engineering of variants to improve half-life, functionality, or biophysical properties of potential lead molecules. A successful strategy to purify this ever-increasing number of mAbs in a timely manner has been the miniaturization and automation of the purification process using automatic liquid handlers (ALHs) such as Tecan's Evo or PerkinElmer's Janus platforms. These systems can be equipped with miniaturized columns, which are available in a wide variety of sizes and affinity matrices to cater to the need of the respective application. Various publications have described the setup of ALHs including the respective purification procedure. However, despite being very precise regarding the overall approach, most publications do not focus on the technical optimization and potential pitfalls, which can be crucial to obtain a robust process. To fill this gap, the present publication is aiming to point at some technical difficulties and suggesting potential ways to overcome these problems in order to facilitate the setup of new ALH systems for the purification of antibodies.


Subject(s)
Antibodies, Monoclonal , Automation, Laboratory , Chromatography, Affinity , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Humans
3.
Blood Adv ; 4(9): 1870-1880, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32374879

ABSTRACT

A novel mechanism for extending the circulatory half-life of coagulation factor VIII (FVIII) has been established and evaluated preclinically. The FVIII binding domain of von Willebrand factor (D'D3) fused to human albumin (rD'D3-FP) dose dependently improved pharmacokinetics parameters of coadministered FVIII in all animal species tested, from mouse to cynomolgus monkey, after IV injection. At higher doses, the half-life of recombinant FVIII (rVIII-SingleChain) was calculated to be increased 2.6-fold to fivefold compared with rVIII-SingleChain administered alone in rats, rabbits, and cynomolgus monkeys, and it was increased 3.1-fold to 9.1-fold in mice. Sustained pharmacodynamics effects were observed (ie, activated partial thromboplastin time and thrombin generation measured ex vivo). No increased risk of thrombosis was observed with coadministration of rVIII-SingleChain and rD'D3-FP compared with rVIII-SingleChain alone. At concentrations beyond the anticipated therapeutic range, rD'D3-FP reduced the hemostatic efficacy of coadministered rVIII-SingleChain. This finding might be due to scavenging of activated FVIII by the excessive amount of rD'D3-FP which, in turn, might result in a reduced probability of the formation of the tenase complex. This observation underlines the importance of a fine-tuned balance between FVIII and its binding partner, von Willebrand factor, for hemostasis in general.


Subject(s)
Hemophilia A , Hemostatics , Albumins , Animals , Factor VIII , Half-Life , Life Expectancy , Macaca fascicularis , Mice , Rabbits , Rats
4.
Protein Expr Purif ; 159: 75-82, 2019 07.
Article in English | MEDLINE | ID: mdl-30917921

ABSTRACT

The ability to engineer monoclonal antibodies (mAbs) with high specificity made mAbs the fastest growing segment in the drug market. mAbs represent 8 of the top 20 selling drugs with combined sales of more than 57 billion US$ per year. The ability to purify large numbers of mAbs with sufficient yields for initial screening campaigns has direct impact on the timelines of a project. Automated liquid handling (ALH)-based mAb purification platforms have been used to facilitate the production of large numbers of mAbs. However, the ongoing pressure to de-risk potential lead molecules at an early development stage by including bio-physical characterization of mAbs has further increased the demand to produce sufficient quantities from limited sample volumes. A bottleneck so far has been the limited dynamic binding capacity of these systems, which is partly due to the binding properties of commonly used Protein A affinity matrices. The present publication suggests that by using a Protein A matrix optimized for continuous chromatography applications the yields of ALH-based but also standard lab-scale mAb purifications can be significantly increased without the need to change established protocols.


Subject(s)
Antibodies, Monoclonal/chemistry , Recombinant Fusion Proteins/chemistry , Antibodies, Monoclonal/genetics , Cells, Cultured , Chromatography, Affinity , High-Throughput Screening Assays/methods , Humans , Recombinant Fusion Proteins/genetics , Robotics , Staphylococcal Protein A/chemistry , Transfection
5.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 2): 89-97, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30713159

ABSTRACT

The influenza neuraminidase (NA) is a homotetramer with head, stalk, transmembrane and cytoplasmic regions. The structure of the NA head with a stalk has never been determined. The NA head from an N9 subtype influenza A virus, A/tern/Australia/G70C/1975 (H1N9), was expressed with an artificial stalk derived from the tetrabrachion (TB) tetramerization domain from Staphylothermus marinus. The NA was successfully crystallized both with and without the TB stalk, and the structures were determined to 2.6 and 2.3 Šresolution, respectively. Comparisons of the two NAs with the native N9 NA structure from egg-grown virus showed that the artificial TB stalk maintained the native NA head structure, supporting previous biological observations.


Subject(s)
Bacterial Proteins/chemistry , Influenza A Virus, H5N1 Subtype/enzymology , Neuraminidase/chemistry , Amino Acid Sequence , Bacterial Proteins/metabolism , Crystallization , Crystallography, X-Ray , Desulfurococcaceae/metabolism , Humans , Influenza, Human/virology , Models, Molecular , Neuraminidase/metabolism , Protein Conformation , Protein Domains
6.
BMC Biotechnol ; 18(1): 15, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29544494

ABSTRACT

BACKGROUND: Preclinical studies have evaluated haptoglobin (Hp) polymers from pooled human plasma as a therapeutic protein to attenuate toxic effects of cell-free hemoglobin (Hb). Proof of concept studies have demonstrated efficacy of Hp in hemolysis associated with transfusion and sickle cell anemia. However, phenotype-specific Hp products might be desirable to exploit phenotype specific activities of Hp 1-1 versus Hp 2-2, offering opportunities for recombinant therapeutics. Prohaptoglobin (proHp) is the primary translation product of the Hp mRNA. ProHp is proteolytically cleaved by complement C1r subcomponent-like protein (C1r-LP) in the endoplasmic reticulum. Two main allelic Hp variants, HP1 and HP2 exist. The larger HP2 is considered to be the ancestor variant of all human Hp alleles and is characterized by an α2-chain, which contains an extra cysteine residue that pairs with additional α-chains generating multimers with molecular weights of 200-900 kDa. The two human HP1 alleles (HP1F and HP1S) differ by a two-amino-acid substitution polymorphism within the α-chain and are derived from HP2 by recurring exon deletions. RESULTS: In the present study, we describe a process for the production of recombinant phenotype specific Hp polymers in mammalian FS293F cells. This approach demonstrates that efficient expression of mature and fully functional protein products requires co-expression of active C1r-LP. The functional characterization of our proteins, which included monomer/polymer distribution, binding affinities as well as NO-sparing and antioxidant functions, demonstrated that C1r-LP-processed recombinant Hp demonstrates equal protective functions as plasma derived Hp in vitro as well as in animal studies. CONCLUSIONS: We present a recombinant production process for fully functional phenotype-specific Hp therapeutics. The proposed process could accelerate the development of Hb scavengers to treat patients with cell-free Hb associated disease states, such as sickle cell disease and other hemolytic conditions.


Subject(s)
Haptoglobins/genetics , Haptoglobins/metabolism , Hemoglobins/metabolism , Protein Engineering/methods , Serine Endopeptidases/genetics , Animals , Coronary Vessels/drug effects , Guinea Pigs , Haptoglobins/pharmacology , Heme/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lipid Peroxidation/drug effects , Male , Nitric Oxide/metabolism , Phenotype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Serine Endopeptidases/metabolism , Swine
7.
J Chromatogr A ; 1455: 9-19, 2016 Jul 15.
Article in English | MEDLINE | ID: mdl-27283099

ABSTRACT

Monoclonal antibodies (mAbs) have become the fastest growing segment in the drug market with annual sales of more than 40 billion US$ in 2013. The selection of lead candidate molecules involves the generation of large repertoires of antibodies from which to choose a final therapeutic candidate. Improvements in the ability to rapidly produce and purify many antibodies in sufficient quantities reduces the lead time for selection which ultimately impacts on the speed with which an antibody may transition through the research stage and into product development. Miniaturization and automation of chromatography using micro columns (RoboColumns(®) from Atoll GmbH) coupled to an automated liquid handling instrument (ALH; Freedom EVO(®) from Tecan) has been a successful approach to establish high throughput process development platforms. Recent advances in transient gene expression (TGE) using the high-titre Expi293F™ system have enabled recombinant mAb titres of greater than 500mg/L. These relatively high protein titres reduce the volume required to generate several milligrams of individual antibodies for initial biochemical and biological downstream assays, making TGE in the Expi293F™ system ideally suited to high throughput chromatography on an ALH. The present publication describes a novel platform for purifying Expi293F™-expressed recombinant mAbs directly from cell-free culture supernatant on a Perkin Elmer JANUS-VariSpan ALH equipped with a plate shuttle device. The purification platform allows automated 2-step purification (Protein A-desalting/size exclusion chromatography) of several hundred mAbs per week. The new robotic method can purify mAbs with high recovery (>90%) at sub-milligram level with yields of up to 2mg from 4mL of cell-free culture supernatant.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Robotics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Automation , Chromatography, Affinity , Chromatography, Gel , Chromatography, High Pressure Liquid , HEK293 Cells , Humans , Miniaturization , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Staphylococcal Protein A/chemistry , Staphylococcal Protein A/metabolism
8.
Ann Neurol ; 79(6): 970-82, 2016 06.
Article in English | MEDLINE | ID: mdl-27043916

ABSTRACT

OBJECTIVE: Traumatic brain injury is a major global public health problem for which specific therapeutic interventions are lacking. There is, therefore, a pressing need to identify innovative pathomechanism-based effective therapies for this condition. Thrombus formation in the cerebral microcirculation has been proposed to contribute to secondary brain damage by causing pericontusional ischemia, but previous studies have failed to harness this finding for therapeutic use. The aim of this study was to obtain preclinical evidence supporting the hypothesis that targeting factor XII prevents thrombus formation and has a beneficial effect on outcome after traumatic brain injury. METHODS: We investigated the impact of genetic deficiency of factor XII and acute inhibition of activated factor XII with a single bolus injection of recombinant human albumin-fused infestin-4 (rHA-Infestin-4) on trauma-induced microvascular thrombus formation and the subsequent outcome in 2 mouse models of traumatic brain injury. RESULTS: Our study showed that both genetic deficiency of factor XII and an inhibition of activated factor XII in mice minimize trauma-induced microvascular thrombus formation and improve outcome, as reflected by better motor function, reduced brain lesion volume, and diminished neurodegeneration. Administration of human factor XII in factor XII-deficient mice fully restored injury-induced microvascular thrombus formation and brain damage. INTERPRETATION: The robust protective effect of rHA-Infestin-4 points to a novel treatment option that can decrease ischemic injury after traumatic brain injury without increasing bleeding tendencies. Ann Neurol 2016;79:970-982.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Factor XII/therapeutic use , Factor XIIa/antagonists & inhibitors , Insect Proteins/therapeutic use , Intracranial Thrombosis/drug therapy , Recombinant Fusion Proteins/therapeutic use , Serum Albumin/therapeutic use , Adult , Aged , Animals , Brain Injuries, Traumatic/physiopathology , Case-Control Studies , Disease Models, Animal , Factor XII/genetics , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Middle Aged , Neuroimaging , Platelet Aggregation/physiology , Serum Albumin, Human , Young Adult
9.
Methods Mol Biol ; 1020: 205-14, 2013.
Article in English | MEDLINE | ID: mdl-23709035

ABSTRACT

The characterization of the interaction between a ligand and its receptor is crucial for a broad variety of applications in academia as well as in the pharmaceutical industry. Although various sophisticated high-throughput technologies have been established to investigate the binding of ligands to their receptors, classical filtration-based receptor binding assays still have some advantages when smaller number of samples need to be tested. Here we describe a technically easy, cheap, and reliable receptor binding assay that was successfully applied to determine the binding constant of the NO-independent activator of soluble guanylate cyclase, cinaciguat, and the impact of other small molecules on its interaction with the enzyme.


Subject(s)
Benzoates/pharmacology , High-Throughput Screening Assays/methods , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Benzoates/metabolism , Guanylate Cyclase/metabolism , Ligands , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase
10.
PLoS One ; 7(6): e37779, 2012.
Article in English | MEDLINE | ID: mdl-22701579

ABSTRACT

In 1988 the preceding journal of Nature Biotechnology, Bio/Technology, reported a work by Hopp and co-workers about a new tag system for the identification and purification of recombinant proteins: the FLAG-tag. Beside the extensively used hexa-his tag system the FLAG-tag has gained broad popularity due to its small size, its high solubility, the presence of an internal Enterokinase cleavage site, and the commercial availability of high-affinity anti-FLAG antibodies. Surprisingly, considering the heavy use of FLAG in numerous laboratories world-wide, we identified in insect cells a post-translational modification (PTM) that abolishes the FLAG-anti-FLAG interaction rendering this tag system ineffectual for secreted proteins. The present publication shows that the tyrosine that is part of the crucial FLAG epitope DYK is highly susceptible to sulfation, a PTM catalysed by the enzyme family of Tyrosylprotein-Sulfo-transferases (TPSTs). We showed that this modification can result in less than 20% of secreted FLAG-tagged protein being accessible for purification questioning the universal applicability of this established tag system.


Subject(s)
Epitopes/genetics , Neuraminidase/metabolism , Peptides/genetics , Peptides/metabolism , Recombinant Proteins/metabolism , Sulfotransferases/metabolism , Animals , Antibodies/immunology , Blotting, Western , Cell Line , Chromatography, Affinity , Chromatography, Gel , Epitopes/immunology , Epitopes/metabolism , HEK293 Cells , Humans , Insecta , Mass Spectrometry , Neuraminidase/isolation & purification , Oligopeptides , Peptides/immunology , Peptides/isolation & purification
11.
PLoS One ; 6(8): e23596, 2011.
Article in English | MEDLINE | ID: mdl-21858179

ABSTRACT

In cardiovascular disease, the protective NO/sGC/cGMP signalling-pathway is impaired due to a decreased pool of NO-sensitive haem-containing sGC accompanied by a reciprocal increase in NO-insensitive haem-free sGC. However, no direct method to detect cellular haem-free sGC other than its activation by the new therapeutic class of haem mimetics, such as BAY 58-2667, is available. Here we show that fluorescence dequenching, based on the interaction of the optical active prosthetic haem group and the attached biarsenical fluorophor FlAsH can be used to detect changes in cellular sGC haem status. The partly overlap of the emission spectrum of haem and FlAsH allows energy transfer from the fluorophore to the haem which reduces the intensity of FlAsH fluorescence. Loss of the prosthetic group, e.g. by oxidative stress or by replacement with the haem mimetic BAY 58-2667, prevented the energy transfer resulting in increased fluorescence. Haem loss was corroborated by an observed decrease in NO-induced sGC activity, reduced sGC protein levels, and an increased effect of BAY 58-2667. The use of a haem-free sGC mutant and a biarsenical dye that was not quenched by haem as controls further validated that the increase in fluorescence was due to the loss of the prosthetic haem group. The present approach is based on the cellular expression of an engineered sGC variant limiting is applicability to recombinant expression systems. Nevertheless, it allows to monitor sGC's redox regulation in living cells and future enhancements might be able to extend this approach to in vivo conditions.


Subject(s)
Fluorescence , Guanylate Cyclase/metabolism , Heme/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Benzoates/pharmacology , CHO Cells , Cricetinae , Cricetulus , Cyclic GMP/metabolism , Cysteine/genetics , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Fluorescent Dyes/chemistry , Guanylate Cyclase/chemistry , Guanylate Cyclase/genetics , Humans , Molecular Sequence Data , Mutation , Nitric Oxide/metabolism , Oxadiazoles/pharmacology , Oxazines/pharmacology , Oxidation-Reduction/drug effects , Protein Engineering/methods , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Rotenone/pharmacology , Signal Transduction/drug effects , Soluble Guanylyl Cyclase , Spectrometry, Fluorescence
12.
PLoS One ; 6(8): e23627, 2011.
Article in English | MEDLINE | ID: mdl-21858186

ABSTRACT

The influenza neuraminidase (NA) inhibitors zanamivir, oseltamivir and peramivir were all designed based on the knowledge that the transition state analogue of the cleaved sialic acid, 2-deoxy,2,3-dehydro N-acetyl neuraminic acid (DANA) was a weak inhibitor of NA. While DANA bound rapidly to the NA, modifications leading to the improved potency of these new inhibitors also conferred a time dependent or slow binding phenotype. Many mutations in the NA leading to decreased susceptibility result in loss of slow binding, hence this is a phenotypic marker of many but not all resistant NAs. We present here a simplified approach to determine whether an inhibitor is fast or slow binding by extending the endpoint fluorescent enzyme inhibition assay to a real time assay and monitoring the changes in IC(50)s with time. We carried out two reactions, one with a 30 min preincubation with inhibitor and the second without. The enzymatic reaction was started via addition of substrate and IC(50)s were calculated after each 10 min interval up to 60 min. Results showed that without preincubation IC(50)s for the wild type viruses started high and although they decreased continuously over the 60 min reaction time the final IC(50)s remained higher than for pre-incubated samples. These results indicate a slow equilibrium of association and dissociation and are consistent with slow binding of the inhibitors. In contrast, for viruses with decreased susceptibility, preincubation had minimal effect on the IC(50)s, consistent with fast binding. Therefore this modified assay provides additional phenotypic information about the rate of inhibitor binding in addition to the IC(50), and critically demonstrates the differential effect of incubation times on the IC(50) and K(i) values of wild type and mutant viruses for each of the inhibitors.


Subject(s)
Enzyme Assays/methods , Enzyme Inhibitors/pharmacology , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae/drug effects , Viral Proteins/antagonists & inhibitors , Acids, Carbocyclic , Binding, Competitive , Cyclopentanes/metabolism , Cyclopentanes/pharmacology , Enzyme Inhibitors/metabolism , Guanidines/metabolism , Guanidines/pharmacology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/genetics , Inhibitory Concentration 50 , Kinetics , Mutation , N-Acetylneuraminic Acid/analogs & derivatives , N-Acetylneuraminic Acid/metabolism , N-Acetylneuraminic Acid/pharmacology , Neuraminidase/metabolism , Orthomyxoviridae/enzymology , Orthomyxoviridae/genetics , Oseltamivir/metabolism , Oseltamivir/pharmacology , Protein Binding , Substrate Specificity , Time Factors , Viral Proteins/metabolism , Zanamivir/metabolism , Zanamivir/pharmacology
13.
PLoS One ; 6(2): e16284, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21326879

ABSTRACT

The influenza surface glycoprotein neuraminidase (NA) is essential for the efficient spread of the virus. Antiviral drugs such as Tamiflu (oseltamivir) and Relenza (zanamivir) that inhibit NA enzyme activity have been shown to be effective in the treatment of influenza infections. The recent 'swine flu' pandemic and world-wide emergence of Tamiflu-resistant seasonal human influenza A(H1N1) H(274)Y have highlighted the need for the ongoing development of new anti-virals, efficient production of vaccine proteins and novel diagnostic tools. Each of these goals could benefit from the production of large quantities of highly pure and stable NA. This publication describes a generic expression system for NAs in a baculovirus Expression Vector System (BEVS) that is capable of expressing milligram amounts of recombinant NA. To construct NAs with increased stability, the natural influenza NA stalk was replaced by two different artificial tetramerization domains that drive the formation of catalytically active NA homotetramers: GCN4-pLI from yeast or the Tetrabrachion tetramerization domain from Staphylothermus marinus. Both recombinant NAs are secreted as FLAG-tagged proteins to allow for rapid and simple purification. The Tetrabrachion-based NA showed good solubility, increased stability and biochemical properties closer to the original viral NA than the GCN4-pLI based construct. The expressed quantities and high quality of the purified recombinant NA suggest that this expression system is capable of producing recombinant NA for a broad range of applications including high-throughput drug screening, protein crystallisation, or vaccine development.


Subject(s)
Cloning, Molecular/methods , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H1N1 Subtype/genetics , Neuraminidase/genetics , Neuraminidase/isolation & purification , Drug Resistance, Viral/genetics , Gene Expression , Genes, Reporter/genetics , Genes, Reporter/physiology , Genetic Vectors/analysis , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Neuraminidase/metabolism , Phylogeny , Protein Stability , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Solubility
14.
Circ Res ; 105(1): 33-41, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19478201

ABSTRACT

Nitric oxide (NO) is an essential vasodilator. In vascular diseases, oxidative stress attenuates NO signaling by both chemical scavenging of free NO and oxidation and downregulation of its major intracellular receptor, the alphabeta heterodimeric heme-containing soluble guanylate cyclase (sGC). Oxidation can also induce loss of the heme of sGC, as well as the responsiveness of sGC to NO. sGC activators such as BAY 58-2667 bind to oxidized/heme-free sGC and reactivate the enzyme to exert disease-specific vasodilation. Here, we show that oxidation-induced downregulation of sGC protein extends to isolated blood vessels. Mechanistically, degradation was triggered through sGC ubiquitination and proteasomal degradation. The heme-binding site ligand BAY 58-2667 prevented sGC ubiquitination and stabilized both alpha and beta subunits. Collectively, our data establish oxidation-ubiquitination of sGC as a modulator of NO/cGMP signaling and point to a new mechanism of action for sGC activating vasodilators by stabilizing their receptor, oxidized/heme-free sGC.


Subject(s)
Guanylate Cyclase/metabolism , Heme/metabolism , Nitric Oxide/pharmacology , Proteasome Endopeptidase Complex/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Vasodilator Agents/pharmacology , Blood Vessels , Cell Line , Cyclic GMP/metabolism , Humans , Oxidation-Reduction , Soluble Guanylyl Cyclase , Ubiquitination
15.
Handb Exp Pharmacol ; (191): 195-228, 2009.
Article in English | MEDLINE | ID: mdl-19089331

ABSTRACT

Cyclic guanosine monophosphate (cGMP), generated via the guanylate cyclase (GC)-catalyzed conversion from GTP, is unequivocally recognized as crucial second messenger, intimately involved in the regulation of a broad range of physiological processes such as long term potentiation, blood pressure regulation, or platelet aggregation (for review: Hobbs 2000). Since its first identification in rat urine by Ashman and co-workers (1963), various approaches have been conceived and established to quantify cGMP in biological samples, or to detect cGMP as the reaction product of enzymatic assays, allowing the determination of kinetic parameters. These approaches have evolved from laborious handling of small numbers of samples with average sensitivity to highly developed biochemical detection assays allowing the processing of very large numbers of samples. The present article focuses upon the history of biochemical cGMP detection from the pioneering work of the early years to the actual state-of-the-art approaches for the detection of this important biological messenger.


Subject(s)
Biological Assay/methods , Cyclic GMP/analysis , Guanylate Cyclase/metabolism , Animals , Biological Assay/history , Biological Assay/trends , History, 20th Century , History, 21st Century , Humans
16.
Handb Exp Pharmacol ; (191): 309-39, 2009.
Article in English | MEDLINE | ID: mdl-19089335

ABSTRACT

Oxidative stress, a risk factor for several cardiovascular disorders, interferes with the NO/sGC/cGMP signalling pathway through scavenging of NO and formation of the strong intermediate oxidant, peroxynitrite. Under these conditions, endothelial and vascular dysfunction develops, culminating in different cardio-renal and pulmonary-vascular diseases. Substituting NO with organic nitrates that release NO (NO donors) has been an important principle in cardiovascular therapy for more than a century. However, the development of nitrate tolerance limits their continuous clinical application and, under oxidative stress and increased formation of peroxynitrite foils the desired therapeutic effect. To overcome these obstacles of nitrate therapy, direct NO- and haem-independent sGC activators have been developed, such as BAY 58-2667 (cinaciguat) and HMR1766 (ataciguat), showing unique biochemical and pharmacological properties. Both compounds are capable of selectively activating the oxidized/haem-free enzyme via binding to the enzyme's haem pocket, causing pronounced vasodilatation. The potential importance of these new drugs resides in the fact that they selectively target a modified state of sGC that is prevalent under disease conditions as shown in several animal models and human disease. Activators of sGC may be beneficial in the treatment of a range of diseases including systemic and pulmonary hypertension (PH), heart failure, atherosclerosis, peripheral arterial occlusive disease (PAOD), thrombosis and renal fibrosis. The sGC activator HMR1766 is currently in clinical development as an oral therapy for patients with PAOD. The sGC activator BAY 58-2667 has demonstrated efficacy in a proof-of-concept study in patients with acute decompensated heart failure (ADHF), reducing pre- and afterload and increasing cardiac output from baseline. A phase IIb clinical study for the indication of ADHF is currently underway.


Subject(s)
Cardiovascular Diseases/drug therapy , Enzyme Activators/pharmacology , Guanylate Cyclase/drug effects , Receptors, Cytoplasmic and Nuclear/drug effects , Animals , Benzoates/pharmacology , Cardiovascular Diseases/physiopathology , Clinical Trials as Topic , Drug Delivery Systems , Guanylate Cyclase/metabolism , Heme/metabolism , Humans , Nitric Oxide/metabolism , Oxidative Stress , Receptors, Cytoplasmic and Nuclear/metabolism , Risk Factors , Soluble Guanylyl Cyclase , Sulfonamides/pharmacology , ortho-Aminobenzoates/pharmacology
17.
Mol Pharmacol ; 72(5): 1181-90, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17715400

ABSTRACT

The ubiquitously expressed nitric oxide (NO) receptor soluble guanylate cyclase (sGC) plays a key role in signal transduction. Binding of NO to the N-terminal prosthetic heme moiety of sGC results in approximately 200-fold activation of the enzyme and an increased conversion of GTP into the second messenger cGMP. sGC exists as a heterodimer the dimerization of which is mediated mainly by the central region of the enzyme. In the present work, we constructed deletion mutants within the predicted dimerization region of the sGC alpha(1)- and beta(1)-subunit to precisely map the sequence segments crucial for subunit dimerization. To track mutation-induced alterations of sGC dimerization, we used a bimolecular fluorescence complementation approach that allows visualizing sGC heterodimerization in a noninvasive manner in living cells. Our study suggests that segments spanning amino acids alpha(1)363-372, alpha(1)403-422, alpha(1)440-459, beta(1)212-222, beta(1)304-333, beta(1)344-363, and beta(1)381-400 within the predicted dimerization region are involved in the process of heterodimerization and therefore in the expression of functional sGC.


Subject(s)
Guanylate Cyclase/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Dimerization , Fluorescence , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Microscopy, Confocal , Molecular Sequence Data , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Deletion , Soluble Guanylyl Cyclase , Spectrometry, Fluorescence/methods
18.
Nat Rev Drug Discov ; 5(9): 755-68, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16955067

ABSTRACT

Soluble guanylate cyclase (sGC) is a key signal-transduction enzyme activated by nitric oxide (NO). Impaired bioavailability and/or responsiveness to endogenous NO has been implicated in the pathogenesis of cardiovascular and other diseases. Current therapies that involve the use of organic nitrates and other NO donors have limitations, including non-specific interactions of NO with various biomolecules, lack of response and the development of tolerance following prolonged administration. Compounds that activate sGC in an NO-independent manner might therefore provide considerable therapeutic advantages. Here we review the discovery, biochemistry, pharmacology and clinical potential of haem-dependent sGC stimulators (including YC-1, BAY 41-2272, BAY 41-8543, CFM-1571 and A-350619) and haem-independent sGC activators (including BAY 58-2667 and HMR-1766).


Subject(s)
Enzyme Activators/pharmacology , Guanylate Cyclase/metabolism , Nitric Oxide/metabolism , Animals , Drug Design , Enzyme Activation/drug effects , Enzyme Activators/chemistry , Enzyme Activators/therapeutic use , Humans , Molecular Structure
19.
J Clin Invest ; 116(9): 2552-61, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16955146

ABSTRACT

ROS are a risk factor of several cardiovascular disorders and interfere with NO/soluble guanylyl cyclase/cyclic GMP (NO/sGC/cGMP) signaling through scavenging of NO and formation of the strong oxidant peroxynitrite. Increased oxidative stress affects the heme-containing NO receptor sGC by both decreasing its expression levels and impairing NO-induced activation, making vasodilator therapy with NO donors less effective. Here we show in vivo that oxidative stress and related vascular disease states, including human diabetes mellitus, led to an sGC that was indistinguishable from the in vitro oxidized/heme-free enzyme. This sGC variant represents what we believe to be a novel cGMP signaling entity that is unresponsive to NO and prone to degradation. Whereas high-affinity ligands for the unoccupied heme pocket of sGC such as zinc-protoporphyrin IX and the novel NO-independent sGC activator 4-[((4-carboxybutyl){2-[(4-phenethylbenzyl)oxy]phenethyl}amino) methyl [benzoic]acid (BAY 58-2667) stabilized the enzyme, only the latter activated the NO-insensitive sGC variant. Importantly, in isolated cells, in blood vessels, and in vivo, BAY 58-2667 was more effective and potentiated under pathophysiological and oxidative stress conditions. This therapeutic principle preferentially dilates diseased versus normal blood vessels and may have far-reaching implications for the currently investigated clinical use of BAY 58-2667 as a unique diagnostic tool and highly innovative vascular therapy.


Subject(s)
Benzoates/pharmacology , Blood Vessels/physiology , Endothelium, Vascular/physiology , Guanylate Cyclase/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Benzoates/chemical synthesis , Blood Pressure/drug effects , Cell Culture Techniques , Cyclic GMP/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Guanylate Cyclase/drug effects , Heme , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pulmonary Artery , Rats , Rats, Inbred SHR , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Soluble Guanylyl Cyclase , Swine , Vasodilation
20.
FEBS Lett ; 580(17): 4205-13, 2006 Jul 24.
Article in English | MEDLINE | ID: mdl-16831427

ABSTRACT

The ubiquitous heterodimeric nitric oxide (NO) receptor soluble guanylate cyclase (sGC) plays a key role in various signal transduction pathways. Binding of NO takes place at the prosthetic heme moiety at the N-terminus of the beta(1)-subunit of sGC. The induced structural changes lead to an activation of the catalytic C-terminal domain of the enzyme and to an increased conversion of GTP into the second messenger cyclic GMP (cGMP). In the present work we selected and substituted different residues of the sGC heme-binding pocket based on a sGC homology model. The generated sGC variants were tested in a cGMP reporter cell for their effect on the enzyme activation by heme-dependent (NO, BAY 41-2272) stimulators and heme-independent (BAY 58-2667) activators. The use of these experimental tools allows the enzyme's heme content to be explored in a non-invasive manner. Asp(44), Asp(45) and Phe(74) of the beta(1)-subunit were identified as being crucially important for functional enzyme activation. beta(1)Asp(45) may serve as a switch between different conformational states of sGC and point to a possible mechanism of action of the heme dependent sGC stimulator BAY 41-2272. Furthermore, our data shows that the activation profile of beta(1)IIe(145) Tyr is unchanged compared to the native enzyme, suggesting that Tyr(145) does not confer the ability to distinguish between NO and O(2). In summary, the present work further elucidated intramolecular mechanisms underlying the NO- and BAY 41-2272-mediated sGC activation and raises questions regarding the postulated role of Tyr(145) for ligand discrimination.


Subject(s)
Cyclic GMP/chemistry , Guanylate Cyclase/chemistry , Models, Molecular , Animals , Cyclic GMP/metabolism , Enzyme Activation , Guanylate Cyclase/metabolism , Heme , Protein Structure, Tertiary , Rats , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity , Thermoanaerobacter/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL