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










Publication year range
1.
Metab Eng ; 81: 157-166, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38081506

ABSTRACT

Rare diseases are, despite their name, collectively common and millions of people are affected daily of conditions where treatment often is unavailable. Sulfatases are a large family of activating enzymes related to several of these diseases. Heritable genetic variations in sulfatases may lead to impaired activity and a reduced macromolecular breakdown within the lysosome, with several severe and lethal conditions as a consequence. While therapeutic options are scarce, treatment for some sulfatase deficiencies by recombinant enzyme replacement are available. The recombinant production of such sulfatases suffers greatly from both low product activity and yield, further limiting accessibility for patient groups. To mitigate the low product activity, we have investigated cellular properties through computational evaluation of cultures with varying media conditions and comparison of two CHO clones with different levels of one active sulfatase variant. Transcriptome analysis identified 18 genes in secretory pathways correlating with increased sulfatase production. Experimental validation by upregulation of a set of three key genes improved the specific enzymatic activity at varying degree up to 150-fold in another sulfatase variant, broadcasting general production benefits. We also identified a correlation between product mRNA levels and sulfatase activity that generated an increase in sulfatase activity when expressed with a weaker promoter. Furthermore, we suggest that our proposed workflow for resolving bottlenecks in cellular machineries, to be useful for improvements of cell factories for other biologics as well.


Subject(s)
Sulfatases , Humans , Sulfatases/genetics , Sulfatases/metabolism
2.
Discov Immunol ; 1(1): kyac005, 2022.
Article in English | MEDLINE | ID: mdl-38566906

ABSTRACT

Inflammasomes and the interleukin (IL)-1 family of cytokines are key mediators of both inflammation and immunothrombosis. Inflammasomes are responsible for the release of the pro-inflammatory cytokines IL-1ß and IL-18, as well as releasing tissue factor (TF), a pivotal initiator of the extrinsic coagulation cascade. Uncontrolled production of inflammatory cytokines results in what is known as a "cytokine storm" leading to hyperinflammatory disease. Cytokine storms can complicate a variety of diseases and results in hypercytokinemia, coagulopathies, tissue damage, multiorgan failure, and death. Patients presenting with cytokine storm syndromes have a high mortality rate, driven in part by disseminated intravascular coagulation (DIC). While our knowledge on the factors propagating cytokine storms is increasing, how cytokine storm influences DIC remains unknown, and therefore treatments for diseases, where these aspects are a key feature are limited, with most targeting specific cytokines. Currently, no therapies target the immunothrombosis aspect of hyperinflammatory syndromes. Here we discuss how targeting the inflammasome and pyroptosis may be a novel therapeutic strategy for the treatment of hyperinflammation and its associated pathologies.

3.
Brain Behav Immun Health ; 16: 100307, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34589799

ABSTRACT

The proinflammatory cytokine Interleukin-1 (IL-1), with its two isoforms α and ß, has important roles in multiple pathogenic processes in the central nervous system. The present study aimed to evaluate and compare the blood-to-brain distribution of anakinra (IL-1 receptor antagonist), bermekimab (IL-1α antagonist) and canakinumab (IL-1ß antagonist). A human in vitro model of the blood-brain barrier derived from human umbilical cord blood stem cells was used, where isolated CD34+ cells co-cultured with bovine pericytes were matured into polarized brain-like endothelial cells. Transport rates of the three test items were evaluated after 180 â€‹min incubation at concentrations 50, 250 and 1250 â€‹nM in a transwell system. We report herein that anakinra passes the human brain-like endothelial monolayer at a 4-7-fold higher rate than the monoclonal antibodies tested. Both antibodies had similar transport rates at all concentrations. No dose-dependent effects in transport rates were observed, nor any saturation effects at supraphysiological concentrations. The larger propensity of anakinra to pass this model of the human blood-brain barrier supports existing data and confirms that anakinra can reach the brain compartment at clinically relevant concentrations. As anakinra inhibits the actions of both IL-1α and IL-1ß, it blocks all effects of IL-1 downstream signaling. The results herein further add to the growing body of evidence of the potential utility of anakinra to treat neuroinflammatory disorders.

4.
iScience ; 24(3): 102154, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33665572

ABSTRACT

We show the successful application of ancestral sequence reconstruction to enhance the activity of iduronate-2-sulfatase (IDS), thereby increasing its therapeutic potential for the treatment of Hunter syndrome-a lysosomal storage disease caused by impaired function of IDS. Current treatment, enzyme replacement therapy with recombinant human IDS, does not alleviate all symptoms, and an unmet medical need remains. We reconstructed putative ancestral sequences of mammalian IDS and compared them with extant IDS. Some ancestral variants displayed up to 2-fold higher activity than human IDS in in vitro assays and cleared more substrate in ex vivo experiments in patient fibroblasts. This could potentially allow for lower dosage or enhanced therapeutic effect in enzyme replacement therapy, thereby improving treatment outcomes and cost efficiency, as well as reducing treatment burden. In summary, we showed that ancestral sequence reconstruction can be applied to lysosomal enzymes that function in concert with modern enzymes and receptors in cells.

5.
Sci Rep ; 10(1): 1315, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992763

ABSTRACT

Phenylalanine/tyrosine ammonia-lyases (PAL/TALs) have been approved by the FDA for treatment of phenylketonuria and may harbour potential for complementary treatment of hereditary tyrosinemia Type I. Herein, we explore ancestral sequence reconstruction as an enzyme engineering tool to enhance the therapeutic potential of PAL/TALs. We reconstructed putative ancestors from fungi and compared their catalytic activity and stability to two modern fungal PAL/TALs. Surprisingly, most putative ancestors could be expressed as functional tetramers in Escherichia coli and thus retained their ability to oligomerize. All ancestral enzymes displayed increased thermostability compared to both modern enzymes, however, the increase in thermostability was accompanied by a loss in catalytic turnover. One reconstructed ancestral enzyme in particular could be interesting for further drug development, as its ratio of specific activities is more favourable towards tyrosine and it is more thermostable than both modern enzymes. Moreover, long-term stability assessment showed that this variant retained substantially more activity after prolonged incubation at 25 °C and 37 °C, as well as an increased resistance to incubation at 60 °C. Both of these factors are indicative of an extended shelf-life of biopharmaceuticals. We believe that ancestral sequence reconstruction has potential for enhancing the properties of enzyme therapeutics, especially with respect to stability. This work further illustrates that resurrection of putative ancestral oligomeric proteins is feasible and provides insight into the extent of conservation of a functional oligomerization surface area from ancestor to modern enzyme.


Subject(s)
Dietary Supplements , Enzyme Replacement Therapy , Phenylalanine Ammonia-Lyase/therapeutic use , Tyrosinemias/therapy , Animals , Enzyme Activation , Enzyme Replacement Therapy/methods , Enzyme Stability , Fungi/classification , Fungi/enzymology , Fungi/genetics , Humans , Kinetics , Models, Molecular , Phenylalanine Ammonia-Lyase/administration & dosage , Phenylalanine Ammonia-Lyase/chemistry , Phenylalanine Ammonia-Lyase/classification , Protein Conformation , Recombinant Proteins , Structure-Activity Relationship , Thermodynamics , Tyrosinemias/etiology
6.
FEBS J ; 285(24): 4660-4673, 2018 12.
Article in English | MEDLINE | ID: mdl-30369053

ABSTRACT

Bacterial diterpene cyclases are receiving increasing attention in biocatalysis and synthetic biology for the sustainable generation of complex multicyclic building blocks. Herein, we explore the potential of ancestral sequence reconstruction (ASR) to generate remodeled cyclases with enhanced stability, activity, and promiscuity. Putative ancestors of spiroviolene synthase, a bacterial class I diterpene cyclase, display an increased yield of soluble protein of up to fourfold upon expression in the model organism Escherichia coli. Two of the resurrected enzymes, with an estimated age of approximately 1.7 million years, display an upward shift in thermostability of 7-13 °C. Ancestral spiroviolene synthases catalyze cyclization of the natural C20 -substrate geranylgeranyl diphosphate (GGPP) and also accept C15 farnesyl diphosphate (FPP), which is not converted by the extant enzyme. In contrast, the consensus sequence generated from the corresponding multiple sequence alignment was found to be inactive toward both substrates. Mutation of a nonconserved position within the aspartate-rich motif of the reconstructed ancestral cyclases was associated with modest effects on activity and relative substrate specificity (i.e., kcat /KM for GGPP over kcat /KM for FPP). Kinetic analyses performed at different temperatures reveal a loss of substrate saturation, when going from the ancestor with highest thermostability to the modern enzyme. The kinetics data also illustrate how an increase in temperature optimum of biocatalysis is reflected in altered entropy and enthalpy of activation. Our findings further highlight the potential and limitations of applying ASR to biosynthetic machineries in secondary metabolism.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Alkyl and Aryl Transferases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Diterpenes/chemistry , Polyisoprenyl Phosphates/metabolism , Sesquiterpenes/metabolism , Amino Acid Sequence , Enzyme Stability , Kinetics , Phylogeny , Sequence Homology , Streptomyces/enzymology , Substrate Specificity , Temperature
7.
Sci Rep ; 6: 31365, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27509843

ABSTRACT

The complement component 5 (C5)-binding antibody eculizumab is used to treat patients with paroxysmal nocturnal hemoglobinuria (PNH) and atypical haemolytic uremic syndrome (aHUS). As recently reported there is a need for a precise classification of eculizumab responsive patients to allow for a safe and cost-effective treatment. To allow for such stratification, knowledge of the precise binding site of the drug on its target is crucial. Using a structural epitope mapping strategy based on bacterial surface display, flow cytometric sorting and validation via haemolytic activity testing, we identified six residues essential for binding of eculizumab to C5. This epitope co-localizes with the contact area recently identified by crystallography and includes positions in C5 mutated in non-responders. The identified epitope also includes residue W917, which is unique for human C5 and explains the observed lack of cross-reactivity for eculizumab with other primates. We could demonstrate that Ornithodorus moubata complement inhibitor (OmCI), in contrast to eculizumab, maintained anti-haemolytic function for mutations in any of the six epitope residues, thus representing a possible alternative treatment for patients non-responsive to eculizumab. The method for stratification of patients described here allows for precision medicine and should be applicable to several other diseases and therapeutics.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Complement C5/chemistry , Complement C5/genetics , Epitope Mapping/methods , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Complement C5/metabolism , Complement Inactivating Agents/pharmacology , Cricetulus , Crystallography, X-Ray , Humans , Models, Molecular , Mutation , Protein Domains
8.
Comput Biol Chem ; 39: 29-34, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22885776

ABSTRACT

Amyloidosis is characterized by misfolding of proteins. The clinical gastrointestinal manifestations of amyloidosis may mimic other disease, such as inflammatory bowel disease or colonic cancer. As these patients have a high risk for bleeding and poor wound healing following surgery it is important to diagnose them correctly and do a careful preoperative assessment. The most common form of colonic amyloidosis is caused by Serum Amyloid A (SAA), an acute phase protein of unknown function. It is expressed in response to inflammation and the increased levels may lead to amyloidosis. The main treatment is to suppress the acute phase response and thereby reduce production of SAA. As no structure for SAA is available we aim to perform an in silico assessment of its structural and fibrillation properties. In the paper we propose an ab initio model of the structure of SAA, which consists of a five membered helical bundle with a fold related to the tetratricopeptide repeat domain. As there are uncertainties relating to the packing of the helices, each helical region is subjected to triplicate molecular dynamics simulations to assess the integrity of the structural region. The first helix, stretching from residues 1 to 13, is the least stable according to the simulations; almost all of the helical conformation is lost during the 10 ns simulations, whereas the other helices maintain portions that remain in an helical conformation in at least 80% of the simulations. All helices are also subjected to a single 100 ns simulation to investigate how the secondary structure develops over time. In them helix 1 adopts a ß-hairpin structure similar to other fibril forming proteins. The ß-hairpin can in turn multimerise and form a mature fibril structure. The mechanism behind the conformational transition appears to be driven by interactions of side chains of charged residues, particularly Arginine 1. It exchanges interaction partners in the simulation and stabilizes intermediate conformations on the folding pathway to the final ß-hairpin.


Subject(s)
Amyloidosis/pathology , Colonic Diseases/pathology , Molecular Dynamics Simulation , Serum Amyloid A Protein/genetics , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Protein Folding , Protein Structure, Secondary , Serum Amyloid A Protein/chemistry , Serum Amyloid A Protein/metabolism
9.
Biochem Pharmacol ; 81(3): 402-11, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21095183

ABSTRACT

The neutrophil formyl peptide receptors (FPR1 and FPR2) are G-protein coupled receptors that can induce pro-inflammatory as well as anti-inflammatory activities when activated. Accordingly, these receptors may become therapeutic targets for the development of novel drugs to be used for reducing the inflammation induced injuries in asthma, rheumatoid arthritis, Alzheimer's disease, cardiovascular diseases and traumatic shock. We screened a library of more then 50K small compounds for an ability of the compounds to induce a transient rise in intracellular Ca(2+) in cells transfected to express FPR2 (earlier called FPRL1 or the lipoxin A(4) receptor). Ten agonist hits were selected for further analysis representing different chemical series and five new together with five earlier described molecules were further profiled. Compounds 1-10 gave rise to a calcium response in the FPR2 transfectants with EC(50) values ranging from 4×10(-9)M to 2×10(-7)M. All 10 compounds activated human neutrophils to release superoxide, and based on the potency of their activity, the three most potent activators of the neutrophil NADPH-oxidase were further characterized. These three agonists were largely resistant to inactivation by neutrophil produced reactive oxygen species and shown to trigger the same functional repertoire in neutrophils as earlier described peptide agonists. Accordingly they induced chemotaxis, granule mobilization and secretion of superoxide. Interestingly, the oxidase activity was largely inhibited by cyclosporine H, an FPR1 selective antagonist, but not by PBP10, an FPR2 selective inhibitor, suggesting that FPR1 is the preferred receptor in neutrophils for all three agonists.


Subject(s)
Enzyme Activators/pharmacology , NADPH Oxidases/metabolism , Neutrophils/drug effects , Receptors, Formyl Peptide/agonists , Small Molecule Libraries , Adult , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Enzyme Activation , Enzyme Activators/chemistry , Humans , Neutrophils/enzymology , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Superoxides/metabolism
10.
BMC Res Notes ; 3: 270, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20979597

ABSTRACT

BACKGROUND: We aim to propose interactions between the parathyroid hormone-2 receptor (PTH2R) and its ligand the tuberoinfundibular peptide of 39 residues (TIP39) by constructing a homology model of their complex. The two related peptides parathyroid hormone (PTH) and parathyroid hormone related protein (PTHrP) are compared with the complex to examine their interactions. FINDINGS: In the model, the hydrophobic N-terminus of TIP39 is buried in a hydrophobic part of the central cavity between helices 3 and 7. Comparison of the peptide sequences indicates that the main discriminator between the agonistic peptides TIP39 and PTH and the inactive PTHrP is a tryptophan-phenylalanine replacement. The model indicates that the smaller phenylalanine in PTHrP does not completely occupy the binding site of the larger tryptophan residue in the other peptides. As only TIP39 causes internalisation of the receptor and the primary difference being an aspartic acid in position 7 of TIP39 that interacts with histidine 396 in the receptor, versus isoleucine/histidine residues in the related hormones, this might be a trigger interaction for the events that cause internalisation. CONCLUSIONS: A model is constructed for the complex and a trigger interaction for full agonistic activation between aspartic acid 7 of TIP39 and histidine 396 in the receptor is proposed.

11.
J Comput Aided Mol Des ; 22(1): 53-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18058241

ABSTRACT

Diseases associated with protein fibril-formation, such as the prion diseases and Alzheimer's disease, are gaining increased attention due to their medical importance and complex origins. Using molecular dynamics (MD) simulations in an aqueous environment, we have studied the stability of the alpha-helix covering positions 15-25 of the amyloid beta-peptide (A beta) involved in Alzheimer's disease. The effects of residue replacements, including the effects of A beta disease related mutations, were also investigated. The MD simulations show a very early (2 ns) loss of alpha-helical structure for the Flemish (A beta(A21G)), Italian (A beta(E22K)), and Iowa (A beta(D23N)) forms associated with hereditary Alzheimer's disease. Similarly, an early (5 ns) loss of alpha-helical structure was observed for the Dutch (A beta(E22Q)) variant. MD here provides a possible explanation for the structural changes. Two variants of A beta, A beta(K16A,L17A,F20A) and A beta(V18A,F19A,F20A), that do not produce fibrils in vitro were also investigated. The A beta(V18A,F19A,F20A) initially loses its helical conformation but refolds into helix several times and spends most of the simulation time in helical conformation. However, the A beta(K16A,L17A,F20A) loses the alpha-helical structure after 5 ns and does not refold. For the wildtype A beta(1-40) and A beta(1-42), the helical conformation is lost after 5 ns or after 40 ns, respectively, while for the "familial" (A beta(A42T)) variant, the MD simulations suggest that a C-terminal beta-strand is stabilised, which could explain the fibrillation. The simulations for the Arctic (A beta(E22G)) variant indicate that the alpha-helix is kept for 2 ns, but reappears 2 ns later, whereafter it disappears after 10 ns. The MD results are in several cases compatible with known experimental data, but the correlation is not perfect, indicating that multimerisation tendency and other factors might also be important for fibril formation.


Subject(s)
Peptides/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary
12.
Infect Immun ; 73(10): 6332-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16177304

ABSTRACT

Granulysin and NK-lysin are homologous bactericidal proteins with a moderate residue identity (35%), both of which have antimycobacterial activity. Short loop peptides derived from the antimycobacterial domains of granulysin, NK-lysin, and a putative chicken NK-lysin were examined and shown to have comparable antimycobacterial but variable Escherichia coli activities. The known structure of the NK-lysin loop peptide was used to predict the structure of the equivalent peptides of granulysin and chicken NK-lysin by homology modeling. The last two adopted a secondary structure almost identical to that of NK-lysin. All three peptides form very similar three-dimensional (3-D) architectures in which the important basic residues assume the same positions in space. The basic residues in granulysin are arginine, while those in NK-lysin and chicken NK-lysin are a mixture of arginine and lysine. We altered the ratio of arginine to lysine in the granulysin fragment to examine the importance of basic residues for antimycobacterial activity. The alteration of the amino acids reduced the activity against E. coli to a larger extent than that against Mycobacterium smegmatis. In granulysin, the arginines in the loop structure are not crucial for antimycobacterial activity but are important for cytotoxicity. We suggest that the antibacterial domains of the related proteins granulysin, NK-lysin, and chicken NK-lysin have conserved their 3-D structure and their function against mycobacteria.


Subject(s)
Anti-Infective Agents/chemistry , Antigens, Differentiation, T-Lymphocyte/chemistry , Peptides/chemistry , Proteolipids/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Anti-Infective Agents/pharmacology , Antigens, Differentiation, T-Lymphocyte/pharmacology , Arginine/genetics , Cells, Cultured , Conserved Sequence , Escherichia coli/drug effects , Humans , Lysine/genetics , Molecular Sequence Data , Mycobacterium marinum/drug effects , Mycobacterium smegmatis/drug effects , Peptides/pharmacology , Protein Conformation , Proteolipids/pharmacology
13.
J Chem Inf Comput Sci ; 44(6): 2207-15, 2004.
Article in English | MEDLINE | ID: mdl-15554691

ABSTRACT

Herein a general concept for the design of targeted libraries for proteins with binding sites that are divided into subsites is laid out, including several practical aspects and their solutions. The design is based on a chemogenomic classification of the subsites followed by collection of bioactive molecular fragments and virtual library generation. The general process is outlined and applied to the assembly of a library of 500 molecules targeting the serotonin type 7 (5-HT7) receptor, a class A G-Protein Coupled Receptor (GPCR). Utilizing commercially available building blocks of similar size and composition, a reference library was created. Control sets of known ligands for the 5-HT7 receptor, other GPCRs, and nuclear receptors were collected from literature sources. Principal component analysis of molecular descriptors for the two libraries and the literature sets, displayed a focusing of the targeted library to the region in the chemical space defined by the literature actives, suggesting a denser coverage of the bioactive region than for the more diverse reference library. Additional computational validations, including PCA class predictions, 3D pharmacophore modeling, and docking calculations all indicated an enrichment factor of 5-HT7 ligand-like molecules in the range of 2-4 for the targeted library compared to the reference library.


Subject(s)
Receptors, Serotonin/chemistry , Serotonin Antagonists/chemistry , Serotonin Receptor Agonists/chemistry , Binding Sites , Databases, Factual , Humans , Models, Molecular , Phylogeny , Protein Conformation , Receptors, Serotonin/genetics
14.
Protein Sci ; 13(5): 1251-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15096631

ABSTRACT

Several proteins and peptides that can convert from alpha-helical to beta-sheet conformation and form amyloid fibrils, including the amyloid beta-peptide (Abeta) and the prion protein, contain a discordant alpha-helix that is composed of residues that strongly favor beta-strand formation. In their native states, 37 of 38 discordant helices are now found to interact with other protein segments or with lipid membranes, but Abeta apparently lacks such interactions. The helical propensity of the Abeta discordant region (K16LVFFAED23) is increased by introducing V18A/F19A/F20A replacements, and this is associated with reduced fibril formation. Addition of the tripeptide KAD or phospho-L-serine likewise increases the alpha-helical content of Abeta(12-28) and reduces aggregation and fibril formation of Abeta(1-40), Abeta(12-28), Abeta(12-24), and Abeta(14-23). In contrast, tripeptides with all-neutral, all-acidic or all-basic side chains, as well as phosphoethanolamine, phosphocholine, and phosphoglycerol have no significant effects on Abeta secondary structure or fibril formation. These data suggest that in free Abeta, the discordant alpha-helix lacks stabilizing interactions (likely as a consequence of proteolytic removal from a membrane-associated precursor protein) and that stabilization of this helix can reduce fibril formation.


Subject(s)
Amyloid beta-Peptides/chemistry , Amino Acid Sequence , Amino Acid Substitution , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/ultrastructure , Molecular Sequence Data , Oligopeptides/chemistry , Organophosphorus Compounds/chemistry , Protein Structure, Secondary
15.
Biochem J ; 376(Pt 1): 49-60, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12917011

ABSTRACT

17beta-hydroxysteroid dehydrogenases (17beta-HSDs) catalyse the conversion of 17beta-OH (-hydroxy)/17-oxo groups of steroids, and are essential in mammalian hormone physiology. At present, eleven 17beta-HSD isoforms have been defined in mammals, with different tissue-expression and substrate-conversion patterns. We analysed 17beta-HSD type 10 (17beta-HSD10) from humans and Drosophila, the latter known to be essential in development. In addition to the known hydroxyacyl-CoA dehydrogenase, and 3alpha-OH and 17beta-OH activities with sex steroids, we here demonstrate novel activities of 17beta-HSD10. Both species variants oxidize the 20beta-OH and 21-OH groups in C21 steroids, and act as 7beta-OH dehydrogenases of ursodeoxycholic or isoursodeoxycholic acid (also known as 7beta-hydroxylithocholic acid or 7beta-hydroxyisolithocholic acid respectively). Additionally, the human orthologue oxidizes the 7alpha-OH of chenodeoxycholic acid (5beta-cholanic acid, 3alpha,7alpha-diol) and cholic acid (5beta-cholanic acid). These novel substrate specificities are explained by homology models based on the orthologous rat crystal structure, showing a wide hydrophobic cleft, capable of accommodating steroids in different orientations. These properties suggest that the human enzyme is involved in glucocorticoid and gestagen catabolism, and participates in bile acid isomerization. Confocal microscopy and electron microscopy studies reveal that the human form is localized to mitochondria, whereas Drosophila 17beta-HSD10 shows a cytosolic localization pattern, possibly due to an N-terminal sequence difference that in human 17beta-HSD10 constitutes a mitochondrial targeting signal, extending into the Rossmann-fold motif.


Subject(s)
17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxyacyl CoA Dehydrogenases , Bile Acids and Salts/metabolism , Gonadal Steroid Hormones/metabolism , 17-Hydroxysteroid Dehydrogenases/chemistry , 17-Hydroxysteroid Dehydrogenases/physiology , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Drosophila melanogaster/enzymology , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Isoenzymes/physiology , Kinetics , Mitochondria/chemistry , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Steroids/metabolism , Substrate Specificity
16.
Chem Biol Interact ; 143-144: 247-53, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12604210

ABSTRACT

Short-chain dehydrogenases/reductases (SDR) form a large, functionally heterogeneous protein family presently with about 3000 primary and about 30 3D structures deposited in databases. Despite low sequence identities between different forms (about 15-30%), the 3D structures display highly similar alpha/beta folding patterns with a central beta-sheet, typical of the Rossmann-fold. Based on distinct sequence motifs functional assignments and classifications are possible, making it possible to build a general nomenclature system. Recent mutagenetic and structural studies considerably extend the knowledge on the general reaction mechanism, thereby establishing a catalytic tetrad of Asn-Ser-Tyr-Lys residues, which presumably form the framework for a proton relay system including the 2'-OH of the nicotinamide ribose, similar to the mechanism found in horse liver ADH. Based on their cellular functions, several SDR enzymes appear as possible and promising pharmacological targets with application areas spanning hormone-dependent cancer forms or metabolic diseases such as obesity and diabetes, and infectious diseases.


Subject(s)
Oxidoreductases/metabolism , Crystallography, X-Ray , Models, Molecular , Oxidoreductases/chemistry , Oxidoreductases/drug effects , Protein Conformation
17.
Chem Biol Interact ; 143-144: 255-61, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12604211

ABSTRACT

Eukaryotic genomes code for at least eight medium-chain dehydrogenases/reductases (MDR) enzyme families of two types, with and without Zn(2+) at the active site. Four families have Zn(2+): 'Dimeric alcohol dehydrogenases (ADHs)' (including liver ADHs), 'Tetrameric ADHs' (including the yeast ADHs), 'Cinnamyl ADHs' and 'Polyol DHs'. In the human genome, there are minimally 23 MDR genes, but the list is still growing from further interpretations. Of these, seven genes on chromosome 4 (and three pseudogenes) represent the ADH classes in the gene order IV, Igamma, Ibeta, Ialpha, V, II and III. The lineages leading to human ADH establish five levels of divergence, with nodes at the MDR/short-chain dehydrogenases/reductases (SDR), dimer/tetramer, class III/non-III, further class, and intraclass levels of divergence. These multiplicities allow conclusions on pathways of function for ADHs and suggest this activity to have two roles in addition to its function in metabolism, one of a basic defence nature, the other of regulatory value in higher eukaryotes.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Oxidoreductases/metabolism , Humans
18.
Eur J Biochem ; 269(17): 4267-76, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12199705

ABSTRACT

Completed eukaryotic genomes were screened for medium-chain dehydrogenases/reductases (MDR). In the human genome, 23 MDR forms were found, a number that probably will increase, because the genome is not yet fully interpreted. Partial sequences already indicate that at least three further members exist. Within the MDR superfamily, at least eight families were distinguished. Three families are formed by dimeric alcohol dehydrogenases (ADH; originally detected in animals/plants), cinnamyl alcohol dehydrogenases (originally detected in plants) and tetrameric alcohol dehydrogenases (originally detected in yeast). Three further families are centred around forms initially detected as mitochondrial respiratory function proteins, acetyl-CoA reductases of fatty acid synthases, and leukotriene B4 dehydrogenases. The two remaining families with polyol dehydrogenases (originally detected as sorbitol dehydrogenase) and quinone reductases (originally detected as zeta-crystallin) are also distinct but with variable sequences. The most abundant families in the human genome are the dimeric ADH forms and the quinone oxidoreductases. The eukaryotic patterns are different from those of Escherichia coli. The different families were further evaluated by molecular modelling of their active sites as to geometry, hydrophobicity and volume of substrate-binding pockets. Finally, sequence patterns were derived that are diagnostic for the different families and can be used in genome annotations.


Subject(s)
Binding Sites/physiology , Genome, Human , Oxidoreductases/genetics , Amino Acid Motifs/physiology , Crystallins/genetics , Evolution, Molecular , Humans , Models, Molecular , Phylogeny , Quinone Reductases/genetics
19.
Chem Res Toxicol ; 15(6): 825-31, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12067250

ABSTRACT

In this study, human glutathione transferases (GSTs) of alpha class have been assayed with the ultimate carcinogenic (-)-anti- and (+)-syn-diol epoxides (DEs) derived from the nonplanar dibenzo[a,l]pyrene (DBPDE) and the (+)-anti-diol epoxide of the planar benzo[a]pyrene [(+)-anti-BPDE] in the presence of glutathione (GSH). In all DEs, the benzylic oxirane carbon reacting with GSH, possess R-absolute configuration. GSTA1-1 demonstrated activity with all DEs tested whereas A2-2 and A3-3 only were active with the DBPDE enantiomers. With GSTA4-4, no detectable activity was observed. GSTA1-1 was found to be the most efficient enzyme and demonstrated a catalytic efficiency (k(cat)/K(m)) of 464 mM(-)(1) s(-)(1) with (+)-syn-DBPDE. This activity was about 7-fold higher than that observed with (-)-anti-DBPDE and more than 65-fold higher than previously observed with less complex fjord-region DEs. GSTA3-3 also demonstrated high k(cat)/K(m) with the DEs of DBP and a high preference for the (+)-syn-DBPDE enantiomer [190 vs 16.2 mM(-)(1) s(-)(1) for (-)-anti-DBPDE]. Lowest k(cat)/K(m) value of the active enzymes was observed with GSTA2-2. In this case, 30.4 mM(-)(1) s(-)(1) was estimated for (+)-syn-DBPDE and 3.4 mM(-)(1) s(-)(1) with (-)-anti-DBPDE. Comparing the activity of the alpha class GSTs with (-)-anti-DBPDE and (+)-anti-BPDE revealed that GSTA1-1 was considerable more active with the former substrate (about 25-fold). Molecular modeling studies showed that the H-site of GSTA1-1 is deeper and wider than that of GSTA4-4. This is mainly due to the changes of Ser212-->Tyr212 and Ala216-->Val216, which cause a shallower active site, which cannot accommodate large substrates such as DBPDE. The higher activity of GSTA1-1 with (+)-syn-DBPDE relative to (-)-anti-DBPDE is explained by the formation of more favorable interactions between the substrate and the enzyme-GSH complex. The presence of GSTA1-1 in significant amounts in human lung, a primary target tissue for PAH carcinogenesis, may be an important factor for the protection against the harmful action of this type of potent carcinogenic intermediates.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/pharmacokinetics , Benzopyrenes/pharmacokinetics , Carcinogens/pharmacokinetics , Epoxy Compounds/pharmacokinetics , Glutathione Transferase/metabolism , Isoenzymes/metabolism , 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/chemistry , Benzopyrenes/chemistry , Carcinogens/chemistry , Epoxy Compounds/chemistry , Glutathione/chemistry , Glutathione/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Humans , Inactivation, Metabolic , Isoenzymes/chemistry , Isoenzymes/genetics , Kinetics , Liver/enzymology , Lung/enzymology , Models, Molecular , Molecular Conformation , Stereoisomerism , Substrate Specificity , Thermodynamics
20.
J Biol Chem ; 277(28): 25677-84, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-11976334

ABSTRACT

Short-chain dehydrogenases/reductases form a large, evolutionarily old family of NAD(P)(H)-dependent enzymes with over 60 genes found in the human genome. Despite low levels of sequence identity (often 10-30%), the three-dimensional structures display a highly similar alpha/beta folding pattern. We have analyzed the role of several conserved residues regarding folding, stability, steady-state kinetics, and coenzyme binding using bacterial 3beta/17beta-hydroxysteroid dehydrogenase and selected mutants. Structure determination of the wild-type enzyme at 1.2-A resolution by x-ray crystallography and docking analysis was used to interpret the biochemical data. Enzyme kinetic data from mutagenetic replacements emphasize the critical role of residues Thr-12, Asp-60, Asn-86, Asn-87, and Ala-88 in coenzyme binding and catalysis. The data also demonstrate essential interactions of Asn-111 with active site residues. A general role of its side chain interactions for maintenance of the active site configuration to build up a proton relay system is proposed. This extends the previously recognized catalytic triad of Ser-Tyr-Lys residues to form a tetrad of Asn-Ser-Tyr-Lys in the majority of characterized short-chain dehydrogenases/reductase enzymes.


Subject(s)
Oxidoreductases/metabolism , Amino Acid Sequence , Base Sequence , Catalysis , Crystallography, X-Ray , DNA Primers , Electrophoresis, Polyacrylamide Gel , Enzyme Stability , Kinetics , Models, Molecular , Molecular Sequence Data , Oxidoreductases/chemistry , Protein Folding , Sequence Homology, Amino Acid , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...