23 results on '"Christian U. Stirnimann"'
Search Results
2. Drug screening and genome editing in human pancreatic cancer organoids identifies drug-gene interactions and candidates for off-label therapy
- Author
-
Christian K. Hirt, Tijmen H. Booij, Linda Grob, Patrik Simmler, Nora C. Toussaint, David Keller, Doreen Taube, Vanessa Ludwig, Alexander Goryachkin, Chantal Pauli, Daniela Lenggenhager, Daniel J. Stekhoven, Christian U. Stirnimann, Katharina Endhardt, Femke Ringnalda, Lukas Villiger, Alexander Siebenhüner, Sofia Karkampouna, Marta De Menna, Janette Beshay, Hagen Klett, Marianna Kruithof-de Julio, Julia Schüler, and Gerald Schwank
- Subjects
pancreatic cancer ,organoids ,drug screening ,PDX ,BRCA2 ,ARID1A ,Genetics ,QH426-470 ,Internal medicine ,RC31-1245 - Abstract
Summary: 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 further 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 that are approved for non-cancer indications and that perturb the ability of PDAC organoids to respond to hypoxia. Our study provides proof-of-concept for advancing precision oncology and for identifying candidates for drug repurposing via genome editing and drug screening in tumor organoid biobanks.
- Published
- 2022
- Full Text
- View/download PDF
3. Structural basis for the oligomerization-state switch from a dimer to a trimer of an engineered cortexillin-1 coiled-coil variant.
- Author
-
Saša Bjelić, Mara Wieser, Daniel Frey, Christian U Stirnimann, Mark R Chance, Rolf Jaussi, Michel O Steinmetz, and Richard A Kammerer
- Subjects
Medicine ,Science - 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.
- Published
- 2013
- Full Text
- View/download PDF
4. Patient-derived xenografts and organoids model therapy response in prostate cancer
- Author
-
Joel Grosjean, Peter Clark Gray, Jean-Philippe Theurillat, Sofia Karkampouna, Andrea Sboner, M. De Menna, M.R. De Filippo, George N. Thalmann, Andrea Garofoli, Eugenio Zoni, Andrej Benjak, Marco Bolis, Vera Genitsch, David Keller, Charlotte K.Y. Ng, Salvatore Piscuoglio, Irena Klima, Mark A. Rubin, Kenneth Eng, Arianna Vallerga, Tijmen H. Booij, Julio Marianna Kruithof-de, F. La Manna, Christian U. Stirnimann, and Mirjam Kiener
- Subjects
Oncology ,medicine.medical_specialty ,Prostate cancer ,Therapy response ,business.industry ,Urology ,Internal medicine ,Organoid ,medicine ,medicine.disease ,business - Published
- 2021
5. PD59-07 PERSONALISED ORGANOID DRUG TREATMENT AND THERAPY RESISTANCE ON NOVEL EARLY ONSET METASTASIS XENOGRAFT MODEL
- Author
-
Charlotte K.Y. Ng, Marta De Menna, Andrea Garofoli, Maria R. De Filippo, Salvatore Piscuoglio, Federico La Manna, Eugenio Zoni, Marianna Kruithof-de Julio, Andrea Sboner, Christian U. Stirnimann, Irena Klima, Joel Grosjean, Mark A. Rubin, Martin Spahn, Vera Genitsch, Tijmen H. Booij, Sofia Karkampouna, David Keller, and George N. Thalmann
- Subjects
Oncology ,medicine.medical_specialty ,Drug treatment ,business.industry ,Urology ,Internal medicine ,medicine ,Organoid ,Treatment resistance ,business ,medicine.disease ,Early onset ,Metastasis - Published
- 2020
6. Patient-derived xenografts and organoids model therapy response in prostate cancer
- Author
-
Mirjam Kiener, Marianna Kruithof-de Julio, George N. Thalmann, Salvatore Piscuoglio, Marta De Menna, Maria R. De Filippo, Federico La Manna, Peter C. Gray, Andrea Sboner, Eugenio Zoni, Charlotte K.Y. Ng, Jo eumll Grosjean, David Keller, Christian U. Stirnimann, Sofia Karkampouna, Marco Bolis, Tijmen H. Booij, Martin Spahn, Irena Klima, Mark A. Rubin, Kenneth Eng, Andrea Garofoli, Jean-Philippe Theurillat, and Vera Genitsch
- Subjects
Sorafenib ,Sunitinib ,Ponatinib ,Microsatellite instability ,SPOP ,Biology ,medicine.disease ,Primary tumor ,chemistry.chemical_compound ,Prostate cancer ,chemistry ,medicine ,Organoid ,Cancer research ,medicine.drug - 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 a novel 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 harboursBRCA2 and CHD1somatic mutations, shows anSPOP/FOXA1-like transcriptomic signature and microsatellite instability, which occurs in 3% of advanced PCa and has never been modelledin 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.
- Published
- 2020
7. Patient-derived xenografts and organoids model therapy response in prostate cancer
- Author
-
Marco Bolis, Arianna Vallerga, Salvatore Piscuoglio, Andrej Benjak, Andrea Sboner, Federico La Manna, Mirjam Kiener, Joel Grosjean, Tijmen H. Booij, David Keller, Charlotte K.Y. Ng, Eugenio Zoni, Martin Spahn, Vera Genitsch, Sofia Karkampouna, Irena Klima, Mark A. Rubin, Kenneth Eng, Marianna Kruithof-de Julio, Christian U. Stirnimann, Jean-Philippe Theurillat, Andrea Garofoli, Maria R. De Filippo, Marta De Menna, George N. Thalmann, and Peter C. Gray
- Subjects
0301 basic medicine ,Sorafenib ,Male ,Science ,Medizin ,General Physics and Astronomy ,610 Medicine & health ,Antineoplastic Agents ,SPOP ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cancer screening ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,medicine ,Organoid ,Humans ,Neoplasm Metastasis ,Cancer models ,Multidisciplinary ,Sunitinib ,business.industry ,Genome, Human ,Cancer stem cells ,Ponatinib ,Microsatellite instability ,Prostatic Neoplasms ,General Chemistry ,medicine.disease ,Primary tumor ,Xenograft Model Antitumor Assays ,3. Good health ,Organoids ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Androgens ,570 Life sciences ,biology ,business ,Transcriptome ,medicine.drug - 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., To date, patients still succumb to cancer, due to tumors not responding to therapy or ultimately acquiring resistance. Here the authors show that by exploiting patient derived organoids and a treatment-naïve patient derived xenograft, patient therapy can be personalized.
- Published
- 2020
8. Personalised organoid drug treatment and therapy resistance characterization based on novel BRCA2 prostate cancer xenograft of SPOP-like phenotype and microsatellite instability
- Author
-
Joel Grosjean, M.R. De Filippo, M. De Menna, Salvatore Piscuoglio, F. La Manna, G.N. Thalmann, M. Kruithof-De Julio, Tijmen H. Booij, Christian U. Stirnimann, Sofia Karkampouna, Eugenio Zoni, Charlotte K.Y. Ng, David Keller, Irena Klima, Andrea Sboner, Andrea Garofoli, Martin Spahn, Vera Genitsch, and A.R. Mark
- Subjects
business.industry ,Urology ,Microsatellite instability ,SPOP ,lcsh:Diseases of the genitourinary system. Urology ,lcsh:RC870-923 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Phenotype ,Drug treatment ,Prostate cancer ,Cancer research ,Organoid ,Medicine ,Treatment resistance ,business - Published
- 2020
9. Complex Interdependence Regulates Heterotypic Transcription Factor Distribution and Coordinates Cardiogenesis
- Author
-
Daniel He, Florence Baudin, Eric M. Small, Alisha K. Holloway, Maria Mileikovsky, Benoit G. Bruneau, Christoph W. Müller, Abul Hassan Samee, Sebastian Glatt, Sean Thomas, Andras Nagy, Luis Luna-Zurita, Bogac Kaynak, Katherine S. Pollard, and Christian U. Stirnimann
- Subjects
0301 basic medicine ,Models, Molecular ,Cellular differentiation ,Organogenesis ,Cardiovascular ,Crystallography, X-Ray ,Medical and Health Sciences ,Transgenic ,Mice ,Models ,Transcription (biology) ,Gene expression ,Promoter Regions, Genetic ,Genetics ,Zinc finger ,Crystallography ,GATA4 ,Cell Differentiation ,Biological Sciences ,Cell biology ,Heart Disease ,Embryo ,Homeobox Protein Nkx-2.5 ,1.1 Normal biological development and functioning ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Promoter Regions ,03 medical and health sciences ,Genetic ,Underpinning research ,Animals ,Protein Interaction Domains and Motifs ,Gene ,Transcription factor ,Homeodomain Proteins ,Biochemistry, Genetics and Molecular Biology(all) ,Mammalian ,Myocardium ,Molecular ,Embryo, Mammalian ,GATA4 Transcription Factor ,030104 developmental biology ,X-Ray ,Homeobox ,Generic health relevance ,T-Box Domain Proteins ,Developmental Biology ,Transcription Factors - 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 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.
- Published
- 2016
- Full Text
- View/download PDF
10. Abstract B18: Patient-derived xenograft and organoids models of prostate cancer
- Author
-
Andrea Garofoli, Marianna Kruithof-de Julio, David Keller, Booij Tijmen, Jean-Philippe Theurillat, Marco Bolis, Christian U. Stirnimann, Charlotte K.Y. Ng, Irena Klima, Marta De Menna, Mirjam Kiener, Mark A. Rubin, George N. Thalmann, Martin Spahn, Vera Genitsch, Joel Grosjean, Maria R. De Filippo, Andrea Sboner, Sofia Karkampouna, Federico La Manna, Eugenio Zoni, and Salvatore Piscuoglio
- Subjects
Cancer Research ,medicine.drug_class ,business.industry ,Microsatellite instability ,Cancer ,medicine.disease ,Androgen ,Primary tumor ,Drug repositioning ,Prostate cancer ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Organoid ,Bone marrow ,business - Abstract
Therapy resistance and metastatic processes in prostate cancer (PCa) remain undefined, mainly due to the lack of experimental models representing different disease stages. We aim to provide functional experimental and preclinical drug tools applicable for direct use with patient-derived material. Biopsies from metastatic PCa were used for the establishment of patient-derived xenografts (PDXs) by subcutaneous implantation. In vivo tumor growth kinetic in response to androgens was assessed by surgical castration and testosterone supplementation. RNA and whole-exome sequencing (WES) and organoid drug screens were used to characterize the genomic, transcriptomic, and functional properties. Organoid culture methodology was adapted in order to set up an automated medium-throughput organoid drug screen (Nexus Theragnostics automated platform) for screening standard-of-care compounds and candidates for drug repurposing. A novel case of PCa xenograft model derived from soft tissue metastasis (PNPCa) was established. RNAseq and WES confirmed transcriptomic and genomic similarity to the primary tumor. PNPCa harbors BRAC2 and CHD1 mutations and shows SPOP-like and FOXA1-like transcriptomic signature with microsatellite instability. PNPCa tumor growth is inhibited after androgen deprivation by castration, while androgen replacement leads to tumor reformation. No spontaneous tumor growth occurred after prolonged period of castration; however, scarce micrometastases were found in the bone marrow and lymph nodes. The treatment-naive and androgen-dependent properties of the PNPCa made it a candidate model for identification of potent drug compounds. PNPCa-derived organoid screens on standard-of-care and 74 FDA-approved compounds showed that mTOR pathway and multi-tyrosine kinase inhibitors, used for clinical treatment of other cancer types, have high impact on PCa organoid viability. Application of the organoid drug screen panel on two additional metastatic PDXs of advanced disease has allowed shortlisting of compounds for repurposing use in patient-derived organoid screens that can be routinely performed in a timeframe of few weeks, and thus, provide information on treatment decision. Treatment response of a treatment-naive, early metastatic PCa case (PNPCa PDX) highlights the potential of organoid screens to assess therapy response with extended applications for personalized screens. Citation Format: Sofia Karkampouna, Federico la Manna, Maria R. De Filippo, Mirjam Kiener, Marta De Menna, Eugenio Zoni, Joel Grosjean, Irena Klima, Andrea Garofoli, Marco Bolis, Jean-Philippe Theurillat, Vera Genitsch, David Keller, Booij Tijmen, Christian U. Stirnimann, Andrea Sboner, Charlotte K.Y. Ng, Salvatore Piscuoglio, Martin Spahn, Mark A. Rubin, George N. Thalmann, Marianna Kruithof-de Julio. Patient-derived xenograft and organoids models of prostate cancer [abstract]. In: Proceedings of the AACR Special Conference on the Evolving Landscape of Cancer Modeling; 2020 Mar 2-5; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2020;80(11 Suppl):Abstract nr B18.
- Published
- 2020
11. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding
- Author
-
Marcus Vetter, Ilona Krol, Christian U. Stirnimann, Francesc Castro-Giner, Christian Kurzeder, Christian Beisel, Nicola Aceto, Sofia Gkountela, Walter P. Weber, Ramona Scherrer, Julia Landin, Manuel C. Scheidmann, Christoph Rochlitz, Viola Heinzelmann-Schwarz, and Barbara Maria Szczerba
- Subjects
Homeobox protein NANOG ,Cellular differentiation ,Bisulfite sequencing ,Breast Neoplasms ,Biology ,circulating tumor cells ,circulating tumor cell clusters ,General Biochemistry, Genetics and Molecular Biology ,Article ,single cell sequencing ,drug screen ,Metastasis ,Metastasis Suppression ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Circulating tumor cell ,SOX2 ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,030304 developmental biology ,Cell Proliferation ,0303 health sciences ,proliferation-associated transcription factors ,stemness-associated transcription factors ,SOXB1 Transcription Factors ,RNA sequencing ,Cell Differentiation ,Nanog Homeobox Protein ,DNA Methylation ,medicine.disease ,Neoplastic Cells, Circulating ,3. Good health ,Repressor Proteins ,Disease Models, Animal ,Sin3 Histone Deacetylase and Corepressor Complex ,DNA methylation ,Cancer research ,bisulfite sequencing ,Female ,Octamer Transcription Factor-3 ,030217 neurology & neurosurgery - Abstract
Summary 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., Graphical Abstract, Highlights • Binding sites for OCT4, SOX2, NANOG, and SIN3A are hypomethylated in CTC clusters • CTC cluster hypomethylation profile correlates with a poor prognosis in breast cancer • Treatment with FDA-approved Na+/K+-ATPase inhibitors dissociates CTC clusters • Dissociation reverts the methylation profile of CTC clusters and suppresses metastasis, A comparative analysis of the methylation landscape of single and clusters of circulating tumor cells reveals patterns of similarity to embryonic stem cells and identifies pharmacological agents that can target clustering, suppress stemness, and blunt metastatic spreading.
- Published
- 2019
12. Chromatin-modifying Complex Component Nurf55/p55 Associates with Histones H3 and H4 and Polycomb Repressive Complex 2 Subunit Su(z)12 through Partially Overlapping Binding Sites
- Author
-
Christoph W. Müller, Florence Baudin, Doris Lindner, Christian U. Stirnimann, Nga Ly-Hartig, Vladimir Rybin, Claudio Alfieri, and Agnieszka J. Nowak
- Subjects
Stereochemistry ,Polycomb-Group Proteins ,macromolecular substances ,DNA and Chromosomes ,Biology ,Crystallography, X-Ray ,Biochemistry ,Protein Structure, Secondary ,Histones ,Histone H4 ,Histone H3 ,Histone H1 ,Animals ,Drosophila Proteins ,Histone octamer ,Binding site ,Molecular Biology ,Binding Sites ,Polycomb Repressive Complex 2 ,Histone-Lysine N-Methyltransferase ,Cell Biology ,Molecular biology ,Chromatin ,Repressor Proteins ,Drosophila melanogaster ,Histone ,biology.protein ,Retinoblastoma-Binding Protein 4 ,PRC2 - 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 β-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.
- Published
- 2011
13. Structural Basis of TBX5–DNA Recognition: The T-Box Domain in Its DNA-Bound and -Unbound Form
- Author
-
Christoph W. Müller, Christian U. Stirnimann, Denis Ptchelkine, and Clemens Grimm
- Subjects
Fetal Proteins ,Heart Defects, Congenital ,Models, Molecular ,HMG-box ,Molecular Sequence Data ,Mutant ,Limb Deformities, Congenital ,Biology ,chemistry.chemical_compound ,Protein structure ,Structural Biology ,Animals ,Humans ,Point Mutation ,Abnormalities, Multiple ,Protein–DNA interaction ,Amino Acid Sequence ,Molecular Biology ,Transcription factor ,Base Sequence ,Circular Dichroism ,Point mutation ,DNA ,Molecular biology ,Protein Structure, Tertiary ,chemistry ,Biophysics ,Nucleic Acid Conformation ,T-Box Domain Proteins ,Sequence Alignment ,Binding domain - 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.
- Published
- 2010
14. Evidence for Proton Shuffling in a Thioredoxin-Like Protein during Catalysis
- Author
-
Christian U. Stirnimann, Shirley W. I. Siu, Rudi Glockshuber, Guido Capitani, John P.A. Grimshaw, Rainer A. Böckmann, Daniele Narzi, University of Zurich, and Böckmann, R A
- Subjects
Models, Molecular ,Stereochemistry ,Lysine ,Protein Disulfide-Isomerases ,Protonation ,Thioredoxin fold ,Catalysis ,Thioredoxins ,1315 Structural Biology ,Structural Biology ,Oxidoreductase ,10019 Department of Biochemistry ,1312 Molecular Biology ,Computer Simulation ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,biology ,Escherichia coli Proteins ,Active site ,Protein Structure, Tertiary ,Crystallography ,chemistry ,Intramolecular force ,biology.protein ,570 Life sciences ,Protons ,Thioredoxin ,Oxidation-Reduction ,Cysteine - 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.
- Published
- 2008
15. A Type IV Translocated Legionella Cysteine Phytase Counteracts Intracellular Growth Restriction by Phytate
- Author
-
Xiao-Dan Li, Stephen Weber, Mara M. Wieser, Sabrina Engelhardt, Roger Meier, Guido Capitani, Richard A. Kammerer, Hubert Hilbi, Christian U. Stirnimann, Daniel Frey, University of Zurich, and Hilbi, Hubert
- Subjects
1303 Biochemistry ,Phytic Acid ,Phosphatase ,Gene Expression ,610 Medicine & health ,Biology ,Arginine ,Biochemistry ,Legionella pneumophila ,Microbiology ,Protein Structure, Secondary ,1307 Cell Biology ,Bacterial Proteins ,Catalytic Domain ,1312 Molecular Biology ,Escherichia coli ,Secretion ,Dictyostelium ,Cysteine ,Molecular Biology ,Bacterial Secretion Systems ,6-Phytase ,Acanthamoeba castellanii ,10179 Institute of Medical Microbiology ,Effector ,Intracellular parasite ,Genetic Complementation Test ,Cell Biology ,biology.organism_classification ,Bacterial effector protein ,Recombinant Proteins ,respiratory tract diseases ,Kinetics ,Host-Pathogen Interactions ,570 Life sciences ,biology ,Intracellular ,Bacteria - 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 A resolution revealed a mainly α-helical globular protein stabilized by four antiparallel β-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.
- Published
- 2014
16. ChipX: A Novel Microfluidic Chip for Counter-Diffusion Crystallization of Biomolecules and in Situ Crystal Analysis at Room Temperature
- Author
-
Pierre Morin, Anne-Laure Deman, Claude Sauter, Nicolas Terrier, Jean-François Chateaux, Christian U. Stirnimann, Bernard Lorber, Rosaria Ferrigno, Vincent Olieric, M. Brun, Franziska Pinker, Architecture et Réactivité de l'ARN (ARN), Institut de biologie moléculaire et cellulaire (IBMC), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Centre National de la Recherche Scientifique (CNRS), INL - Lab-On-Chip et Instrumentation (INL - LOCI), Institut des Nanotechnologies de Lyon (INL), Centre National de la Recherche Scientifique (CNRS)-Institut National des Sciences Appliquées de Lyon (INSA Lyon), Institut National des Sciences Appliquées (INSA)-Université de Lyon-Institut National des Sciences Appliquées (INSA)-Université de Lyon-École Centrale de Lyon (ECL), Université de Lyon-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-École supérieure de Chimie Physique Electronique de Lyon (CPE)-Centre National de la Recherche Scientifique (CNRS)-Institut National des Sciences Appliquées de Lyon (INSA Lyon), Université de Lyon-École supérieure de Chimie Physique Electronique de Lyon (CPE), Bureau d'Économie Théorique et Appliquée (BETA), Université de Lorraine (UL)-Université de Strasbourg (UNISTRA)-Institut National de la Recherche Agronomique (INRA)-Centre National de la Recherche Scientifique (CNRS), Architecture et réactivité de l'ARN (ARN), Université Louis Pasteur - Strasbourg I-Centre National de la Recherche Scientifique (CNRS), École Centrale de Lyon (ECL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National des Sciences Appliquées de Lyon (INSA Lyon), Université de Lyon-Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-École supérieure de Chimie Physique Electronique de Lyon (CPE)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-École Centrale de Lyon (ECL), Université de Lyon, Institut National de la Recherche Agronomique (INRA)-Université de Strasbourg (UNISTRA)-Université de Lorraine (UL)-Centre National de la Recherche Scientifique (CNRS), Université de Lyon-Université de Lyon-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-École supérieure de Chimie Physique Electronique de Lyon (CPE)-Institut National des Sciences Appliquées de Lyon (INSA Lyon), Institut National des Sciences Appliquées (INSA)-Université de Lyon-Institut National des Sciences Appliquées (INSA)-Centre National de la Recherche Scientifique (CNRS)-École Centrale de Lyon (ECL), Institut National des Sciences Appliquées (INSA)-Université de Lyon-Institut National des Sciences Appliquées (INSA)-Centre National de la Recherche Scientifique (CNRS), and INL - Plateforme Technologique Nanolyon (INL - Nanolyon)
- Subjects
Materials science ,Microfluidics ,Crystal growth ,Nanotechnology ,Cyclic olefin copolymer ,010402 general chemistry ,01 natural sciences ,law.invention ,Crystal ,03 medical and health sciences ,chemistry.chemical_compound ,[SPI]Engineering Sciences [physics] ,law ,Miniaturization ,General Materials Science ,Crystallization ,[SPI.NANO]Engineering Sciences [physics]/Micro and nanotechnologies/Microelectronics ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Biomolecule ,General Chemistry ,Condensed Matter Physics ,Chip ,0104 chemical sciences ,chemistry - Abstract
Microfluidic technology has opened new possibilities for the crystallization of biological macromolecules during the past decade. Microfluidic systems offer numerous advantages over conventional crystal growth methods. They enable easy handling of nanovolumes of solutions, extreme miniaturization, and parallelization of crystallization assays, especially for high-throughput screening applications. Our goal was to design a versatile, low cost, and easy-to-use crystallization chip based on counter-diffusion that is compatible with on-chip crystallographic characterization. The ChipX is a microfluidic chip made of cyclic olefin copolymer. It was used to grow crystals of biomolecules and perform complete X-ray diffraction analyses on synchrotron sources. Our results demonstrate that accurate crystallographic data can be collected at room temperature directly from ChipX microfluidic devices for both experimental single-wavelength anomalous dispersion phasing and structure refinement.
- Published
- 2013
17. Structural basis for the oligomerization-state switch from a dimer to a trimer of an engineered cortexillin-1 coiled-coil variant
- Author
-
Daniel Frey, Michel O. Steinmetz, Saša Bjelić, Mark R. Chance, Rolf Jaussi, Mara M. Wieser, Christian U. Stirnimann, and Richard A. Kammerer
- Subjects
Macromolecular Assemblies ,Models, Molecular ,Protein Structure ,Protein Folding ,Circular dichroism ,Stereochemistry ,Dimer ,Molecular Sequence Data ,Biophysics ,lcsh:Medicine ,Trimer ,Protein Engineering ,Biochemistry ,Protein Structure, Secondary ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Protein sequencing ,Macromolecular Structure Analysis ,Amino Acid Sequence ,Protein Interactions ,Protein Structure, Quaternary ,lcsh:Science ,Biology ,Condensed-Matter Physics ,Peptide sequence ,030304 developmental biology ,Coiled coil ,0303 health sciences ,Crystallography ,Multidisciplinary ,Protein Stability ,Chemistry ,Physics ,Microfilament Proteins ,lcsh:R ,Proteins ,Computational Biology ,Protein engineering ,Recombinant Proteins ,3. Good health ,A-site ,lcsh:Q ,Protein Multimerization ,030217 neurology & neurosurgery ,Research Article - 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. ISSN:1932-6203
- Published
- 2013
18. A series of Fas receptor agonist antibodies that demonstrate an inverse correlation between affinity and potency
- Author
-
Markus G. Grütter, Sara Züger, Ralph Minter, Lutz Jermutus, Matthieu Chodorge, Christophe Briand, Christian U. Stirnimann, University of Zurich, and Minter, R R
- Subjects
Agonist ,Fas Ligand Protein ,medicine.drug_class ,agonist antibody ,Apoptosis ,Biology ,Crystallography, X-Ray ,Protein Engineering ,Partial agonist ,Epitope ,Fas ligand ,Antibodies ,scFv ,1307 Cell Biology ,Jurkat Cells ,medicine ,10019 Department of Biochemistry ,1312 Molecular Biology ,Humans ,fas Receptor ,Receptor ,Molecular Biology ,affinity maturation ,Original Paper ,Binding Sites ,Cell Biology ,Fas ,Ligand (biochemistry) ,Fas receptor ,Molecular biology ,Cell biology ,Protein Structure, Tertiary ,Kinetics ,Mutagenesis ,570 Life sciences ,biology ,Signal transduction ,phage display ,HeLa Cells ,Signal Transduction ,Single-Chain Antibodies - Abstract
Receptor agonism remains poorly understood at the molecular and mechanistic level. In this study, we identified a fully human anti-Fas antibody that could efficiently trigger apoptosis and therefore function as a potent agonist. Protein engineering and crystallography were used to mechanistically understand the agonistic activity of the antibody. The crystal structure of the complex was determined at 1.9 A resolution and provided insights into epitope recognition and comparisons with the natural ligand FasL (Fas ligand). When we affinity-matured the agonist antibody, we observed that, surprisingly, the higher-affinity antibodies demonstrated a significant reduction, rather than an increase, in agonist activity at the Fas receptor. We propose and experimentally demonstrate a model to explain this non-intuitive impact of affinity on agonist antibody signalling and explore the implications for the discovery of therapeutic agonists in general.
- Published
- 2012
19. Oxidoreductase activity of oligosaccharyltransferase subunits Ost3p and Ost6p defines site-specific glycosylation efficiency
- Author
-
Markus Aebi, John P.A. Grimshaw, Rudi Glockshuber, Benjamin L. Schulz, Markus G. Grütter, Guido Capitani, Maurice S. Brozzo, Elisabeth Mohorko, Fabienne Fritsch, and Christian U. Stirnimann
- Subjects
Models, Molecular ,Multiprotein complex ,Glycosylation ,Saccharomyces cerevisiae Proteins ,Protein subunit ,Amino Acid Motifs ,Saccharomyces cerevisiae ,Biology ,Models, Biological ,Protein Structure, Secondary ,chemistry.chemical_compound ,N-linked glycosylation ,Oxidoreductase ,Catalytic Domain ,Sulfhydryl Compounds ,chemistry.chemical_classification ,Multidisciplinary ,Oligosaccharyltransferase ,Membrane Proteins ,Biological Sciences ,Protein Subunits ,chemistry ,Oligosaccharyltransferase complex ,Biochemistry ,Hexosyltransferases ,Protein folding ,Oxidoreductases ,Peptides ,Protein Binding - 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.
- Published
- 2009
20. DsbL and DsbI form a specific dithiol oxidase system for periplasmic arylsulfate sulfotransferase in uropathogenic Escherichia coli
- Author
-
Guido Capitani, John P.A. Grimshaw, Maurice S. Brozzo, Markus G. Grütter, Goran Malojčić, Rudi Glockshuber, and Christian U. Stirnimann
- Subjects
Models, Molecular ,Operon ,Protein Conformation ,Molecular Sequence Data ,medicine.disease_cause ,Crystallography, X-Ray ,chemistry.chemical_compound ,Structural Biology ,medicine ,Escherichia coli ,Insulin ,Amino Acid Sequence ,Disulfides ,Molecular Biology ,Oxidase test ,Binding Sites ,biology ,Chemistry ,Escherichia coli Proteins ,Genetic Complementation Test ,Dithiol ,Hydrogen Bonding ,Periplasmic space ,Ribonuclease, Pancreatic ,Arylsulfotransferase ,Glutathione ,Protein Structure, Tertiary ,DsbA ,Membrane protein ,Biochemistry ,Periplasm ,biology.protein ,Oxidoreductases ,Oxidation-Reduction ,Sequence Alignment ,Cysteine - 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.
- Published
- 2008
21. nDsbD: a redox interaction hub in the Escherichia coli periplasm
- Author
-
Christian U. Stirnimann, Guido Capitani, Markus G. Grütter, Rudi Glockshuber, University of Zurich, and Grütter, M G
- Subjects
Models, Molecular ,Protein Conformation ,Amino Acid Motifs ,2804 Cellular and Molecular Neuroscience ,medicine.disease_cause ,1307 Cell Biology ,Thioredoxins ,Protein Interaction Mapping ,biology ,Chemistry ,Cytochrome c ,Escherichia coli Proteins ,Disulfide bond ,3004 Pharmacology ,Biochemistry ,Disulfide isomerization ,Cytochrome c maturation ,DsbD ,DsbC ,DsbG ,CcmG ,Periplasm ,Molecular Medicine ,Cystine ,Periplasmic Proteins ,Oxidoreductases ,Dimerization ,Oxidation-Reduction ,Protein Disulfide-Isomerases ,Redox ,142-005 142-005 ,Electron Transport ,Cellular and Molecular Neuroscience ,Structure-Activity Relationship ,medicine ,1312 Molecular Biology ,Escherichia coli ,Cysteine ,Molecular Biology ,Pharmacology ,Hydrogen Bonding ,Protein Disulfide Reductase (Glutathione) ,Cell Biology ,Periplasmic space ,Protein Structure, Tertiary ,Enzyme Activation ,1313 Molecular Medicine ,biology.protein ,570 Life sciences ,NADP - Abstract
Cellular and Molecular Life Sciences, 63 (14), ISSN:1420-682X, ISSN:1420-9071
- Published
- 2006
22. High-resolution structures of Escherichia coli cDsbD in different redox states: A combined crystallographic, biochemical and computational study
- Author
-
Ulla Grauschopf, Markus G. Grütter, Rainer A. Böckmann, Christian U. Stirnimann, Anna Rozhkova, Rudi Glockshuber, and Guido Capitani
- Subjects
Models, Molecular ,Conformational change ,Protein Denaturation ,Molecular Sequence Data ,Crystallography, X-Ray ,Computing Methodologies ,Molecular dynamics ,Thioredoxins ,Structural Biology ,Oxidoreductase ,Escherichia coli ,Humans ,Amino Acid Sequence ,Cysteine ,Disulfides ,Molecular Biology ,Conserved Sequence ,chemistry.chemical_classification ,Binding Sites ,biology ,Chemistry ,Escherichia coli Proteins ,Titrimetry ,Active site ,Membrane Proteins ,Periplasmic space ,Electron transport chain ,Protein Structure, Tertiary ,Crystallography ,Transmembrane domain ,Structural Homology, Protein ,biology.protein ,Thermodynamics ,Thioredoxin ,Oxidoreductases ,Oxidation-Reduction ,Sequence Alignment - 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 p K a 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 p K a 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.
- Published
- 2005
23. Structural Basis and Kinetics of DsbD-Dependent Cytochrome c Maturation
- Author
-
Christian U. Stirnimann, Ulla Grauschopf, Guido Capitani, Anna Rozhkova, Markus G. Grütter, and Rudi Glockshuber
- Subjects
Models, Molecular ,Cytoplasm ,Time Factors ,Protein Conformation ,Kinetics ,Electrons ,medicine.disease_cause ,Crystallography, X-Ray ,Models, Biological ,Electron Transport ,Thioredoxins ,Structural Biology ,medicine ,Escherichia coli ,Disulfides ,Molecular Biology ,Chromatography, High Pressure Liquid ,Binding Sites ,biology ,Chemistry ,Cytochrome c ,Escherichia coli Proteins ,Cytochromes c ,Membrane Proteins ,Protein Disulfide Reductase (Glutathione) ,Periplasmic space ,Electron transport chain ,Protein Structure, Tertiary ,Oxygen ,Transmembrane domain ,Biochemistry ,biology.protein ,Thioredoxin ,Oxidoreductases ,Dimerization ,Oxidation-Reduction ,Plasmids - Abstract
Summary 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 × 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.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.