Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Genom ; 2(2): 100095, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35187519

ABSTRACT

Pancreatic cancer (PDAC) is a highly aggressive malignancy for which the identification of novel therapies is urgently needed. Here, we establish a human PDAC organoid biobank from 31 genetically distinct lines, covering a representative range of tumor subtypes, and demonstrate that these reflect the molecular and phenotypic heterogeneity of primary PDAC tissue. We use CRISPR-Cas9 genome editing and drug screening to characterize drug-gene interactions with ARID1A and BRCA2. We find that missense- but not frameshift mutations in the PDAC driver gene ARID1A are associated with increased sensitivity to the kinase inhibitors dasatinib (p < 0.0001) and VE-821 (p < 0.0001). We conduct an automated drug-repurposing screen with 1,172 FDA-approved compounds, identifying 26 compounds that effectively kill PDAC organoids, including 19 chemotherapy drugs currently approved for other cancer types. We validate the activity of these compounds in vitro and in vivo. The in vivo validated hits include emetine and ouabain, compounds which are approved for non-cancer indications and which perturb the ability of PDAC organoids to respond to hypoxia. Our study provides proof-of-concept for advancing precision oncology and identifying candidates for drug repurposing via genome editing and drug screening in tumor organoid biobanks.

2.
Nat Commun ; 12(1): 1117, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33602919

ABSTRACT

Therapy resistance and metastatic processes in prostate cancer (PCa) remain undefined, due to lack of experimental models that mimic different disease stages. We describe an androgen-dependent PCa patient-derived xenograft (PDX) model from treatment-naïve, soft tissue metastasis (PNPCa). RNA and whole-exome sequencing of the PDX tissue and organoids confirmed transcriptomic and genomic similarity to primary tumor. PNPCa harbors BRCA2 and CHD1 somatic mutations, shows an SPOP/FOXA1-like transcriptomic signature and microsatellite instability, which occurs in 3% of advanced PCa and has never been modeled in vivo. Comparison of the treatment-naïve PNPCa with additional metastatic PDXs (BM18, LAPC9), in a medium-throughput organoid screen of FDA-approved compounds, revealed differential drug sensitivities. Multikinase inhibitors (ponatinib, sunitinib, sorafenib) were broadly effective on all PDX- and patient-derived organoids from advanced cases with acquired resistance to standard-of-care compounds. This proof-of-principle study may provide a preclinical tool to screen drug responses to standard-of-care and newly identified, repurposed compounds.


Subject(s)
Models, Biological , Organoids/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Xenograft Model Antitumor Assays , Androgens/metabolism , Antineoplastic Agents/therapeutic use , Genome, Human , Humans , Male , Mutation/genetics , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Transcriptome/genetics
3.
Cell ; 176(1-2): 98-112.e14, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30633912

ABSTRACT

The ability of circulating tumor cells (CTCs) to form clusters has been linked to increased metastatic potential. Yet biological features and vulnerabilities of CTC clusters remain largely unknown. Here, we profile the DNA methylation landscape of single CTCs and CTC clusters from breast cancer patients and mouse models on a genome-wide scale. We find that binding sites for stemness- and proliferation-associated transcription factors are specifically hypomethylated in CTC clusters, including binding sites for OCT4, NANOG, SOX2, and SIN3A, paralleling embryonic stem cell biology. Among 2,486 FDA-approved compounds, we identify Na+/K+ ATPase inhibitors that enable the dissociation of CTC clusters into single cells, leading to DNA methylation remodeling at critical sites and metastasis suppression. Thus, our results link CTC clustering to specific changes in DNA methylation that promote stemness and metastasis and point to cluster-targeting compounds to suppress the spread of cancer.


Subject(s)
Breast Neoplasms/genetics , Neoplasm Metastasis/genetics , Neoplastic Cells, Circulating/pathology , Animals , Breast Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , DNA Methylation/physiology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred NOD , Nanog Homeobox Protein/metabolism , Neoplasm Metastasis/physiopathology , Neoplastic Cells, Circulating/metabolism , Octamer Transcription Factor-3/metabolism , Repressor Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
4.
Cell ; 164(5): 999-1014, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26875865

ABSTRACT

Transcription factors (TFs) are thought to function with partners to achieve specificity and precise quantitative outputs. In the developing heart, heterotypic TF interactions, such as between the T-box TF TBX5 and the homeodomain TF NKX2-5, have been proposed as a mechanism for human congenital heart defects. We report extensive and complex interdependent genomic occupancy of TBX5, NKX2-5, and the zinc finger TF GATA4 coordinately controlling cardiac gene expression, differentiation, and morphogenesis. Interdependent binding serves not only to co-regulate gene expression but also to prevent TFs from distributing to ectopic loci and activate lineage-inappropriate genes. We define preferential motif arrangements for TBX5 and NKX2-5 cooperative binding sites, supported at the atomic level by their co-crystal structure bound to DNA, revealing a direct interaction between the two factors and induced DNA bending. Complex interdependent binding mechanisms reveal tightly regulated TF genomic distribution and define a combinatorial logic for heterotypic TF regulation of differentiation.


Subject(s)
GATA4 Transcription Factor/metabolism , Homeodomain Proteins/metabolism , Myocardium/cytology , Organogenesis , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Crystallography, X-Ray , Embryo, Mammalian/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Models, Molecular , Myocardium/metabolism , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , T-Box Domain Proteins/genetics , Transcription Factors/genetics
5.
J Biol Chem ; 289(49): 34175-88, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25339170

ABSTRACT

The causative agent of Legionnaires' pneumonia, Legionella pneumophila, colonizes diverse environmental niches, including biofilms, plant material, and protozoa. In these habitats, myo-inositol hexakisphosphate (phytate) is prevalent and used as a phosphate storage compound or as a siderophore. L. pneumophila replicates in protozoa and mammalian phagocytes within a unique "Legionella-containing vacuole." The bacteria govern host cell interactions through the Icm/Dot type IV secretion system (T4SS) and ∼300 different "effector" proteins. Here we characterize a hitherto unrecognized Icm/Dot substrate, LppA, as a phytate phosphatase (phytase). Phytase activity of recombinant LppA required catalytically essential cysteine (Cys(231)) and arginine (Arg(237)) residues. The structure of LppA at 1.4 Å resolution revealed a mainly α-helical globular protein stabilized by four antiparallel ß-sheets that binds two phosphate moieties. The phosphates localize to a P-loop active site characteristic of dual specificity phosphatases or to a non-catalytic site, respectively. Phytate reversibly abolished growth of L. pneumophila in broth, and growth inhibition was relieved by overproduction of LppA or by metal ion titration. L. pneumophila lacking lppA replicated less efficiently in phytate-loaded Acanthamoeba castellanii or Dictyostelium discoideum, and the intracellular growth defect was complemented by the phytase gene. These findings identify the chelator phytate as an intracellular bacteriostatic component of cell-autonomous host immunity and reveal a T4SS-translocated L. pneumophila phytase that counteracts intracellular bacterial growth restriction by phytate. Thus, bacterial phytases might represent therapeutic targets to combat intracellular pathogens.


Subject(s)
6-Phytase/chemistry , Bacterial Proteins/chemistry , Bacterial Secretion Systems/genetics , Legionella pneumophila/enzymology , Phytic Acid/metabolism , 6-Phytase/genetics , 6-Phytase/metabolism , Acanthamoeba castellanii/metabolism , Acanthamoeba castellanii/microbiology , Arginine/chemistry , Arginine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cysteine/chemistry , Cysteine/metabolism , Dictyostelium/metabolism , Dictyostelium/microbiology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Complementation Test , Host-Pathogen Interactions , Kinetics , Legionella pneumophila/drug effects , Legionella pneumophila/genetics , Phytic Acid/chemistry , Phytic Acid/pharmacology , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
Nature ; 505(7481): 108-11, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24240280

ABSTRACT

Botulinum neurotoxin A (BoNT/A) belongs to the most dangerous class of bioweapons. Despite this, BoNT/A is used to treat a wide range of common medical conditions such as migraines and a variety of ocular motility and movement disorders. BoNT/A is probably best known for its use as an antiwrinkle agent in cosmetic applications (including Botox and Dysport). BoNT/A application causes long-lasting flaccid paralysis of muscles through inhibiting the release of the neurotransmitter acetylcholine by cleaving synaptosomal-associated protein 25 (SNAP-25) within presynaptic nerve terminals. Two types of BoNT/A receptor have been identified, both of which are required for BoNT/A toxicity and are therefore likely to cooperate with each other: gangliosides and members of the synaptic vesicle glycoprotein 2 (SV2) family, which are putative transporter proteins that are predicted to have 12 transmembrane domains, associate with the receptor-binding domain of the toxin. Recently, fibroblast growth factor receptor 3 (FGFR3) has also been reported to be a potential BoNT/A receptor. In SV2 proteins, the BoNT/A-binding site has been mapped to the luminal domain, but the molecular details of the interaction between BoNT/A and SV2 are unknown. Here we determined the high-resolution crystal structure of the BoNT/A receptor-binding domain (BoNT/A-RBD) in complex with the SV2C luminal domain (SV2C-LD). SV2C-LD consists of a right-handed, quadrilateral ß-helix that associates with BoNT/A-RBD mainly through backbone-to-backbone interactions at open ß-strand edges, in a manner that resembles the inter-strand interactions in amyloid structures. Competition experiments identified a peptide that inhibits the formation of the complex. Our findings provide a strong platform for the development of novel antitoxin agents and for the rational design of BoNT/A variants with improved therapeutic properties.


Subject(s)
Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Endocytosis/drug effects , HEK293 Cells , Humans , Models, Molecular , Neostriatum/cytology , Neurons/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Structure-Activity Relationship
7.
PLoS One ; 8(5): e63370, 2013.
Article in English | MEDLINE | ID: mdl-23691037

ABSTRACT

Coiled coils are well suited to drive subunit oligomerization and are widely used in applications ranging from basic research to medicine. The optimization of these applications requires a detailed understanding of the molecular determinants that control of coiled-coil formation. Although many of these determinants have been identified and characterized in great detail, a puzzling observation is that their presence does not necessarily correlate with the oligomerization state of a given coiled-coil structure. Thus, other determinants must play a key role. To address this issue, we recently investigated the unrelated coiled-coil domains from GCN4, ATF1 and cortexillin-1 as model systems. We found that well-known trimer-specific oligomerization-state determinants, such as the distribution of isoleucine residues at heptad-repeat core positions or the trimerization motif Arg-h-x-x-h-Glu (where h = hydrophobic amino acid; x = any amino acid), switch the peptide's topology from a dimer to a trimer only when inserted into the trigger sequence, a site indispensable for coiled-coil formation. Because high-resolution structural information could not be obtained for the full-length, three-stranded cortexillin-1 coiled coil, we here report the detailed biophysical and structural characterization of a shorter variant spanning the trigger sequence using circular dichroism, anatytical ultracentrifugation and x-ray crystallography. We show that the peptide forms a stable α-helical trimer in solution. We further determined the crystal structure of an optimised variant at a resolution of 1.65 Å, revealing that the peptide folds into a parallel, three-stranded coiled coil. The two complemented R-IxxIE trimerization motifs and the additional hydrophobic core isoleucine residue adopt the conformations seen in other extensively characterized parallel, three-stranded coiled coils. These findings not only confirm the structural basis for the switch in oligomerization state from a dimer to a trimer observed for the full-length cortexillin-1 coiled-coil domain, but also provide further evidence for a general link between oligomerization-state specificity and trigger-sequence function.


Subject(s)
Microfilament Proteins/chemistry , Protein Engineering , Protein Multimerization , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Protein Stability , Protein Structure, Quaternary , Protein Structure, Secondary
8.
J Biol Chem ; 286(26): 23388-96, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21550984

ABSTRACT

Drosophila Nurf55 is a component of different chromatin-modifying complexes, including the PRC2 (Polycomb repressive complex 2). Based on the 1.75-Å crystal structure of Nurf55 bound to histone H4 helix 1, we analyzed interactions of Nurf55 (Nurf55 or p55 in fly and RbAp48/46 in human) with the N-terminal tail of histone H3, the first helix of histone H4, and an N-terminal fragment of the PRC2 subunit Su(z)12 using isothermal calorimetry and pulldown experiments. Site-directed mutagenesis identified the binding site of histone H3 at the top of the Nurf55 WD40 propeller. Unmodified or K9me3- or K27me3-containing H3 peptides were bound with similar affinities, whereas the affinity for K4me3-containing H3 peptides was reduced. Helix 1 of histone H4 and Su(z)12 bound to the edge of the ß-propeller using overlapping binding sites. Our results show similarities in the recognition of histone H4 and Su(z)12 and identify Nurf55 as a versatile interactor that simultaneously contacts multiple partners.


Subject(s)
Drosophila Proteins/chemistry , Histone-Lysine N-Methyltransferase/chemistry , Histones/chemistry , Repressor Proteins/chemistry , Retinoblastoma-Binding Protein 4/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Protein Structure, Secondary , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 4/genetics , Retinoblastoma-Binding Protein 4/metabolism
9.
J Mol Biol ; 400(1): 71-81, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20450920

ABSTRACT

TBX5, a member of the T-box transcription factor family, plays an important role in heart and limb development. More than 60 single point or deletion mutations of human TBX5 are associated with Holt-Oram syndrome that manifests itself as heart and limb malformations in 1 out of 100,000 live births. The majority of these mutations are located in the TBX5 T-box domain. We solved the crystal structures of the human TBX5 T-box domain in its DNA-unbound form and in complex with a natural DNA target site allowing for the first time the comparison between unbound and DNA-bound forms. Our analysis identifies a 3(10)-helix at the C-terminus of the T-box domain as an inducible recognition element, critically required for the interaction with DNA, as it only forms upon DNA binding and is unstructured in the DNA-unbound form. Using circular dichroism, we characterized the thermal stability of six TBX5 mutants containing single point mutations in the T-box domain (M74V, G80R, W121G, G169R, T223M, and R237W) and compared them with wild-type protein. Mutants G80R and W121G show drastically reduced thermal stability, while the other mutants only show a marginal stability decrease. For all TBX5 mutants, binding affinities to specific and nonspecific DNA sequences were determined using isothermal titration calorimetry. All TBX5 mutants show reduced binding affinities to a specific DNA target site, although to various degrees. Interestingly, all tested TBX5 mutants differ in their ability to bind unspecific DNA, indicating that both sequence-specific and unspecific binding might contribute to the misregulation of target gene expression.


Subject(s)
DNA/chemistry , DNA/metabolism , Protein Structure, Tertiary , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Amino Acid Sequence , Animals , Base Sequence , Circular Dichroism , DNA/genetics , Fetal Proteins/chemistry , Fetal Proteins/genetics , Fetal Proteins/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Humans , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/metabolism , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Point Mutation , Sequence Alignment , T-Box Domain Proteins/genetics
10.
Trends Biochem Sci ; 35(10): 565-74, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20451393

ABSTRACT

Recent findings indicate that WD40 domains play central roles in biological processes by acting as hubs in cellular networks; however, they have been studied less intensely than other common domains, such as the kinase, PDZ or SH3 domains. As suggested by various interactome studies, they are among the most promiscuous interactors. Structural studies suggest that this property stems from their ability, as scaffolds, to interact with diverse proteins, peptides or nucleic acids using multiple surfaces or modes of interaction. A general scaffolding role is supported by the fact that no WD40 domain has been found with intrinsic enzymatic activity despite often being part of large molecular machines. We discuss the WD40 domain distributions in protein networks and structures of WD40-containing assemblies to demonstrate their versatility in mediating critical cellular functions.


Subject(s)
Proteins/metabolism , Repetitive Sequences, Amino Acid , Animals , DNA Damage , Humans , Protein Binding , Proteins/chemistry
11.
Proc Natl Acad Sci U S A ; 106(27): 11061-6, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19549845

ABSTRACT

Asparagine-linked glycosylation is a common posttranslational modification of diverse secretory and membrane proteins in eukaryotes, where it is catalyzed by the multiprotein complex oligosaccharyltransferase. The functions of the protein subunits of oligoasccharyltransferase, apart from the catalytic Stt3p, are ill defined. Here we describe functional and structural investigations of the Ost3/6p components of the yeast enzyme. Genetic, biochemical and structural analyses of the lumenal domain of Ost6p revealed oxidoreductase activity mediated by a thioredoxin-like fold with a distinctive active-site loop that changed conformation with redox state. We found that mutation of the active-site cysteine residues of Ost6p and its paralogue Ost3p affected the glycosylation efficiency of a subset of glycosylation sites. Our results show that eukaryotic oligosaccharyltransferase is a multifunctional enzyme that acts at the crossroads of protein modification and protein folding.


Subject(s)
Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Oxidoreductases/metabolism , Protein Subunits/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Amino Acid Motifs , Catalytic Domain , Glycosylation , Models, Biological , Models, Molecular , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Sulfhydryl Compounds/metabolism
12.
J Mol Biol ; 382(4): 978-86, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18692066

ABSTRACT

Proteins of the thioredoxin (Trx) superfamily catalyze disulfide-bond formation, reduction and isomerization in substrate proteins both in prokaryotic and in eukaryotic cells. All members of the Trx family with thiol-disulfide oxidoreductase activity contain the characteristic Cys-X-X-Cys motif in their active site. Here, using Poisson-Boltzmann-based protonation-state calculations based on 100-ns molecular dynamics simulations, we investigate the catalytic mechanism of DsbL, the most oxidizing Trx-like protein known to date. We observed several correlated transitions in the protonation states of the buried active-site cysteine and a neighboring lysine coupled to the exposure of the active-site thiolate. These results support the view of an internal proton shuffling mechanism during oxidation crucial for the uptake of two electrons from the substrate protein. Intramolecular disulfide-bond formation is probably steered by the conformational switch facilitating interaction with the active-site thiolate. A consistent catalytic mechanism for DsbL, probably conferrable to other proteins of the same class, is presented. Our results suggest a functional role of hydration entropy of active-site groups.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Protein Structure, Tertiary , Protons , Thioredoxins/chemistry , Thioredoxins/metabolism , Binding Sites , Catalysis , Computer Simulation , Escherichia coli Proteins/genetics , Models, Molecular , Oxidation-Reduction , Protein Disulfide-Isomerases/genetics , Thioredoxins/genetics
13.
J Mol Biol ; 380(4): 667-80, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18565543

ABSTRACT

Disulfide bond formation in the Escherichia coli periplasm requires the transfer of electrons from substrate proteins to DsbA, which is recycled as an oxidant by the membrane protein DsbB. The highly virulent, uropathogenic E. coli strain CFT073 contains a second, homologous pair of proteins, DsbL and DsbI, which are encoded in a tri-cistronic operon together with a periplasmic, uropathogen-specific arylsulfate sulfotransferase (ASST). We show that DsbL and DsbI form a functional redox pair, and that ASST is a substrate of DsbL/DsbI in vivo. DsbL is the most reactive oxidizing thioredoxin-like protein known to date. In contrast to DsbA, however, DsbL oxidizes reduced RNaseA with a much lower rate and prevents unspecific aggregation of reduced insulin. The 1.55 A resolution crystal structure of reduced DsbL provides insight into the reduced state of thioredoxin-like dithiol oxidases at high resolution, and reveals an unusual cluster of basic residues stabilizing the thiolate anion of the nucleophilic active-site cysteine. We propose that the DsbL/DsbI pair of uropathogenic E. coli was acquired as an additional, specific redox couple that guarantees biological activity of ASST.


Subject(s)
Arylsulfotransferase/metabolism , Disulfides/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Oxidoreductases/metabolism , Periplasm/enzymology , Amino Acid Sequence , Arylsulfotransferase/chemistry , Arylsulfotransferase/genetics , Binding Sites , Crystallography, X-Ray , Disulfides/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Genetic Complementation Test , Glutathione/metabolism , Hydrogen Bonding , Insulin/chemistry , Insulin/metabolism , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Oxidoreductases/chemistry , Oxidoreductases/genetics , Protein Conformation , Protein Structure, Tertiary , Ribonuclease, Pancreatic/chemistry , Ribonuclease, Pancreatic/metabolism , Sequence Alignment
14.
J Mol Biol ; 358(3): 829-45, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16545842

ABSTRACT

Escherichia coli DsbD transports electrons from cytoplasmic thioredoxin to periplasmic target proteins. DsbD is composed of an N-terminal (nDsbD) and a C-terminal (cDsbD) periplasmic domain, connected by a central transmembrane domain. Each domain possesses two cysteine residues essential for electron transport. The transport proceeds via disulfide exchange reactions from cytoplasmic thioredoxin to the central transmembrane domain and via cDsbD to nDsbD, which then reduces the periplasmic target proteins. We determined four high-resolution structures of cDsbD: oxidized (1.65 A resolution), chemically reduced (1.3 A), photo-reduced (1.1 A) and chemically reduced at pH increased from 4.6 to 7. The latter structure was refined at 0.99 A resolution, the highest achieved so far for a thioredoxin superfamily member. The data reveal unprecedented structural details of cDsbD, demonstrating that the domain is very rigid and undergoes hardly any conformational change upon disulfide reduction or interaction with nDsbD. In full agreement with the crystallographic results, guanidinium chloride-induced unfolding and refolding experiments indicate that oxidized and reduced cDsbD are equally stable. We confirmed the structural rigidity of cDsbD by molecular dynamics simulations. A remarkable feature of cDsbD is the pKa of 9.3 for the active site Cys461: this value, determined using two different experimental methods, surprisingly was around 2.5 units higher than expected on the basis of the redox potential. Additionally, taking advantage of the very high quality of the cDsbD structures, we carried out pKa calculations, which gave results in agreement with the experimental findings. In conclusion, our wide-scope analysis of cDsbD, encompassing atomic-resolution crystallography, computational chemistry and biophysical measurements, highlighted two so far unrecognized key aspects of this domain: its unusual redox properties and extreme rigidity. Both are likely to be correlated to the role of cDsbD as a covalently linked electron shuttle between the membrane domain and the N-terminal periplasmic domain of DsbD.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Amino Acid Sequence , Binding Sites , Computing Methodologies , Conserved Sequence , Crystallography, X-Ray , Cysteine/genetics , Cysteine/metabolism , Disulfides/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Humans , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Oxidoreductases , Protein Denaturation , Protein Structure, Tertiary , Sequence Alignment , Structural Homology, Protein , Thermodynamics , Thioredoxins/chemistry , Thioredoxins/metabolism , Titrimetry
15.
Structure ; 13(7): 985-93, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16004871

ABSTRACT

DsbD from Escherichia coli transports two electrons from cytoplasmic thioredoxin to the periplasmic substrate proteins DsbC, DsbG and CcmG. DsbD consists of an N-terminal periplasmic domain (nDsbD), a C-terminal periplasmic domain, and a central transmembrane domain. Each domain possesses two cysteines required for electron transport. Herein, we demonstrate fast (3.9 x 10(5) M(-1)s(-1)) and direct disulfide exchange between nDsbD and CcmG, a highly specific disulfide reductase essential for cytochrome c maturation. We determined the crystal structure of the disulfide-linked complex between nDsbD and the soluble part of CcmG at 1.94 A resolution. In contrast to the other two known complexes of nDsbD with target proteins, the N-terminal segment of nDsbD contributes to specific recognition of CcmG. This and other features, like the possibility of using an additional interaction surface, constitute the structural basis for the adaptability of nDsbD to different protein substrates.


Subject(s)
Cytochromes c/chemistry , Escherichia coli Proteins/physiology , Membrane Proteins/physiology , Binding Sites , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Cytoplasm/metabolism , Dimerization , Disulfides/chemistry , Electron Transport , Electrons , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Kinetics , Membrane Proteins/chemistry , Models, Biological , Models, Molecular , Oxidation-Reduction , Oxidoreductases/chemistry , Oxygen/chemistry , Plasmids/metabolism , Protein Conformation , Protein Disulfide Reductase (Glutathione)/chemistry , Protein Structure, Tertiary , Thioredoxins/chemistry , Time Factors
16.
EMBO J ; 23(8): 1709-19, 2004 Apr 21.
Article in English | MEDLINE | ID: mdl-15057279

ABSTRACT

DsbD from Escherichia coli catalyzes the transport of electrons from cytoplasmic thioredoxin to the periplasmic disulfide isomerase DsbC. DsbD contains two periplasmically oriented domains at the N- and C-terminus (nDsbD and cDsbD) that are connected by a central transmembrane (TM) domain. Each domain contains a pair of cysteines that are essential for catalysis. Here, we show that Cys109 and Cys461 form a transient interdomain disulfide bond between nDsbD and cDsbD in the reaction cycle of DsbD. We solved the crystal structure of this catalytic intermediate at 2.85 A resolution, which revealed large relative domain movements in DsbD as a consequence of a strong overlap between the surface areas of nDsbD that interact with DsbC and cDsbD. In addition, we have measured the kinetics of all functional and nonfunctional disulfide exchange reactions between redox-active, periplasmic proteins and protein domains from the oxidative DsbA/B and the reductive DsbC/D pathway. We show that both pathways are separated by large kinetic barriers for nonfunctional disulfide exchange between components from different pathways.


Subject(s)
Disulfides/chemistry , Disulfides/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Catalysis , Crystallography, X-Ray , Cysteine/metabolism , Electron Transport , Escherichia coli/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Kinetics , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Models, Molecular , Molecular Weight , Oxidation-Reduction , Oxidoreductases , Periplasm/metabolism , Protein Binding , Protein Structure, Quaternary
SELECTION OF CITATIONS
SEARCH DETAIL