41 results on '"Ulrich Hassiepen"'
Search Results
2. Structure-Based Design and Preclinical Characterization of Selective and Orally Bioavailable Factor XIa Inhibitors: Demonstrating the Power of an Integrated S1 Protease Family Approach
- Author
-
Allan D'Arcy, Nikolaus Schiering, Cary Fridrich, Yu-Hsin Chiu, Donglei Liu, Micah Hollis-Symynkywicz, Edwige Lorthiois, Hyungchul Kim, Darija Dedic, Richard Sedrani, Rose Mo, Solene Dussauge, Calhoun Amy, Stefanie Harlfinger, Treeve Currie, Simon Ruedisser, Lionel Muller, Rohit Duvadie, Ulrich Hassiepen, Adelaide Druet, Louise Kirman, Jason Elliott, Kenji Namoto, Gabriela Monnet, Francesca Perruccio, Paul Ramage, Eva Altmann, David Louis Feldman, Fabien Tritsch, Xueming Huang, Joerg Berghausen, Tanzina Fazal, Richard Zessis, James Roache, Eric T Williams, Christopher M. Adams, Wilhelm A. Weihofen, Viktor Hornak, Peter Delgado, Peter Hoffmann, David Barnes-Seeman, Douglas Bevan, ShuangXi Wang, Hong Liu, Martin Renatus, Celine Dentel, Corinne Durand, Juergen Klaus Maibaum, Guiqing Liang, Kamal Fettis, Gordon Turner, Rajeshri Ganesh Karki, Loren Lindsley, Julie Lachal, and Andreas Hein
- Subjects
Drug ,Male ,Proteases ,Peptidomimetic ,medicine.medical_treatment ,media_common.quotation_subject ,Drug Evaluation, Preclinical ,Administration, Oral ,Biological Availability ,Pharmacology ,01 natural sciences ,Factor XIa ,Rats, Sprague-Dawley ,03 medical and health sciences ,Mice ,Structure-Activity Relationship ,Dogs ,Complement Factor D ,Drug Discovery ,medicine ,Animals ,Humans ,Amino Acid Sequence ,030304 developmental biology ,ADME ,media_common ,Serine protease ,0303 health sciences ,Protease ,biology ,Chemistry ,0104 chemical sciences ,Rats ,Mice, Inbred C57BL ,010404 medicinal & biomolecular chemistry ,Coagulation ,biology.protein ,Molecular Medicine ,Factor Xa Inhibitors - Abstract
The serine protease Factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anti-coagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement Factor D inhibitor and exhibited sub-micromolar FXIa activity and an encouraging ADME profile while being devoid of peptidomimetic architecture. Optimization of interactions in the S1, S1β, and S1` pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with sub-nanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a pre-clinical PK profile consistent with bid dosing in patients.
- Published
- 2020
3. Targeting of the Cholecystokinin-2 Receptor with the Minigastrin Analog 177Lu-DOTA-PP-F11N: Does the Use of Protease Inhibitors Further Improve In Vivo Distribution?
- Author
-
Tania Panigada, Rosalba Mansi, Damian Wild, Ulrich Hassiepen, Christof Rottenburger, Lionel Muller, Anna-Maria Wild, Melpomeni Fani, Alexander W. Sauter, Stefan Wiehr, Martin Béhé, and Susanne Geistlich
- Subjects
In vivo magnetic resonance spectroscopy ,0303 health sciences ,Chemotherapy ,Necrosis ,business.industry ,medicine.medical_treatment ,Medullary thyroid cancer ,medicine.disease ,Radiation therapy ,Meningioma ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Glioma ,medicine ,Radiology, Nuclear Medicine and imaging ,medicine.symptom ,Nuclear medicine ,business ,030304 developmental biology - Abstract
393 Objectives: The currently available PET tracers i.e. 18F-FET and 11C-methionine have proven high diagnostic efficacy in GBM. However, the development of SPECT based tracers is always viewed as a cost-effective alternate to PET imaging. The aim of the present study was to determine the diagnostic efficacy of 99mTc-labelled MDM (bis-methionine) SPECT in the diagnostic work up in glioma. Methods: The present study was conducted in 143 glioma patients (101M: 42F; mean age 41.97±11.9 years; range 18-71 years) who were newly diagnosed or previously treated or who were recruited for post-surgical radiotherapy/chemotherapy treatment from December 2014 to August 2018. Amongst, 143 patients, 29/143 (20.0%) were freshly diagnosed patients of glioma and the remaining 114/143 (80.0%) patients were on post-surgery follow-up (radiological/clinical) with chemotherapy/radiotherapy interventions. The patients were subjected to a detailed histopathological tumor analysis (including Ki-67 index), 99mTc-MDM-SPECT, conventional MRI, DSCE-MRI and MR spectroscopy for the disease evaluation at presentation and during the course of follow-up after surgery/chemo/radiotherapy. A total of 227 brain scans (99mTc-MDM-SPECT) and an equal number of conventional MRI scans were performed in 143 patients. The results of the two imaging modalities were compared and correlated with the clinical findings. In a sub-set of patients (n=43), a quantitative DSCE-MRI and MR spectroscopy analysis was done. The results of the later were compared with the 99mTc-MDM-SPECT quantitative results for validation of this technique for accuracy in the glioma detection and characterization. Results: On histopathological analysis, 26/29 patients (pre-surgery group) were diagnosed to have glioma ( G IV-13; G III-04; G II-09) and the remaining 3/29 patients were found to be meningioma. The mean target to non-target (T/NT) ratios of 99mTc-MDM in glioma grade II, grade III, and grade IV patients were estimated to be 2.46±2.3, 7.13±2.2 and 5.16±1.2 respectively. This ratio was much higher (15.9 ±6.8) in meningioma. The ROC curve analysis derived cut-off value of T/NT ratio of greater than 3.08 when used to discriminate low grade from high grade glioma provided 100% sensitivity, 87.5% specificity. In the post-surgery group, the final diagnosis could be made in 72/114 patients. Out of these, 47/72 showed tumor recurrent (Fig.1) or residual disease and the remaining 25 patients showed necrosis. The ROC curve analysis derived cut-off value of T/NT ratio of greater than 1.90 used to discriminate tumor recurrence from necrosis offered sensitivity and specificity of 97.9 % and 92.0% respectively. A similar analysis on DSCE-MRI quantitative data with derived nCBV cut of value of greater than 3.32 for discriminating tumor recurrence versus necrosis provided sensitivity and specificity of 84.6% and 93.0% respectively. MR spectroscopy data analysis estimated the cut-of ratios; sensitivity/specificity of different metabolites i.e. Cho/NAA, Cho/Cr, Cr/NAA, Cr/Cho and Cho/LL to be >1.57, 81.0%/73.0%; >1.64, 85.3%/73.7%; >1.06. 57.1%/ 63.6%; ≤ 0.60, 72.3%/81.0% and >0.90, 71.4%, /50% respectively. T/NT ratio showed a strongest linear correlation with nCBV(r = 0.775, P
- Published
- 2018
4. The Intrinsic Pepsin Resistance of Interleukin-8 Can Be Explained from a Combined Bioinformatical and Experimental Approach
- Author
-
Daniel Huster, Gerd Anders, Sergey A. Samsonov, Lionel Muller, Tania Panigada, Stephan Theisgen, Stephan Heymann, and Ulrich Hassiepen
- Subjects
0301 basic medicine ,Structural similarity ,medicine.medical_treatment ,Proteolysis ,Biology ,Protein Structure, Secondary ,Electronic mail ,Protein–protein interaction ,03 medical and health sciences ,Pepsin ,Genetics ,medicine ,Computer Simulation ,Nuclear Magnetic Resonance, Biomolecular ,Protein secondary structure ,Protease ,medicine.diagnostic_test ,Applied Mathematics ,Interleukin-8 ,Computational Biology ,Pepsin A ,030104 developmental biology ,Biochemistry ,biology.protein ,Heteronuclear single quantum coherence spectroscopy ,Protein Binding ,Biotechnology - Abstract
Interleukin-8 (IL-8, CXCL8) is a neutrophil chemotactic factor belonging to the family of chemokines. IL-8 was shown to resist pepsin cleavage displaying its high resistance to this protease. However, the molecular mechanisms underlying this resistance are not fully understood. Using our in-house database containing the data on three-dimensional arrangements of secondary structure elements from the whole Protein Data Bank, we found a striking structural similarity between IL-8 and pepsin inhibitor-3. Such similarity could play a key role in understanding IL-8 resistance to the protease pepsin. To support this hypothesis, we applied pepsin assays confirming that intact IL-8 is not degraded by pepsin in comparison to IL-8 in a denaturated state. Applying 1H-15N Heteronuclear Single Quantum Coherence NMR measurements, we determined the putative regions at IL-8 that are potentially responsible for interactions with the pepsin. The results obtained in this work contribute to the understanding of the resistance of IL-8 to pepsin proteolysis in terms of its structural properties.
- Published
- 2018
5. Azaindoles as Zinc‐Binding Small‐Molecule Inhibitors of the JAMM Protease CSN5
- Author
-
Eva Altmann, Paul Erbel, Martin Renatus, Michael Schaefer, Anita Schlierf, Adelaide Druet, Laurence Kieffer, Mickael Sorge, Keith Pfister, Ulrich Hassiepen, Matthew Jones, Simon Ruedisser, Daniela Ostermeier, Bruno Martoglio, Anne B. Jefferson, and Jean Quancard
- Subjects
General Medicine - Published
- 2016
6. MAA868, a novel FXI antibody with a unique binding mode, shows durable effects on markers of anticoagulation in humans
- Author
-
Meike Scharenberg, Serge Côté, Carsten Krantz, Craig T. Basson, Keith DiPetrillo, Stefan Ewert, Janeen M Salter, Brian Stoll, Geraldine Horny, Simone Schleeger, Yu-Hsin Chiu, Julie DeGagne, Peter Hoffmann, Andreas Hein, Jianying Yu, Peter McCormack, Zhiping Chen, Paul Ramage, Nikolaus Schiering, Frederic Villard, Yasser Khder, Samu Melkko, Jörg Eder, Ulrich Hassiepen, Yiming Zhang, Lionel Muller, Xueming Huang, Robert J. A. Frost, Kenneth Kulmatycki, Alexander W. Koch, and David Louis Feldman
- Subjects
0301 basic medicine ,Adult ,Male ,Adolescent ,medicine.drug_class ,Immunology ,030204 cardiovascular system & hematology ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Biochemistry ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Zymogen ,Antithrombotic ,Medicine ,Animals ,Humans ,Blood Coagulation ,Factor XI ,medicine.diagnostic_test ,business.industry ,Anticoagulant ,Anticoagulants ,Thrombosis ,Cell Biology ,Hematology ,Middle Aged ,Mice, Inbred C57BL ,Molecular Docking Simulation ,Macaca fascicularis ,030104 developmental biology ,Coagulation ,Hemostasis ,Immunoglobulin G ,Anticoagulant Agent ,Female ,business ,Fibrinolytic agent ,Partial thromboplastin time - Abstract
A large unmet medical need exists for safer antithrombotic drugs because all currently approved anticoagulant agents interfere with hemostasis, leading to an increased risk of bleeding. Genetic and pharmacologic evidence in humans and animals suggests that reducing factor XI (FXI) levels has the potential to effectively prevent and treat thrombosis with a minimal risk of bleeding. We generated a fully human antibody (MAA868) that binds the catalytic domain of both FXI (zymogen) and activated FXI. Our structural studies show that MAA868 traps FXI and activated FXI in an inactive, zymogen-like conformation, explaining its equally high binding affinity for both forms of the enzyme. This binding mode allows the enzyme to be neutralized before entering the coagulation process, revealing a particularly attractive anticoagulant profile of the antibody. MAA868 exhibited favorable anticoagulant activity in mice with a dose-dependent protection from carotid occlusion in a ferric chloride–induced thrombosis model. MAA868 also caused robust and sustained anticoagulant activity in cynomolgus monkeys as assessed by activated partial thromboplastin time without any evidence of bleeding. Based on these preclinical findings, we conducted a first-in-human study in healthy subjects and showed that single subcutaneous doses of MAA868 were safe and well tolerated. MAA868 resulted in dose- and time-dependent robust and sustained prolongation of activated partial thromboplastin time and FXI suppression for up to 4 weeks or longer, supporting further clinical investigation as a potential once-monthly subcutaneous anticoagulant therapy.
- Published
- 2018
7. Targeting of the Cholecystokinin-2 Receptor with the Minigastrin Analog
- Author
-
Alexander W, Sauter, Rosalba, Mansi, Ulrich, Hassiepen, Lionel, Muller, Tania, Panigada, Stefan, Wiehr, Anna-Maria, Wild, Susanne, Geistlich, Martin, Béhé, Christof, Rottenburger, Damian, Wild, and Melpomeni, Fani
- Subjects
Radioisotopes ,Single Photon Emission Computed Tomography Computed Tomography ,Biological Transport ,Lutetium ,Receptor, Cholecystokinin B ,Heterocyclic Compounds, 1-Ring ,Mice ,Drug Stability ,Cell Line, Tumor ,Positron Emission Tomography Computed Tomography ,Gastrins ,Animals ,Humans ,Female ,Protease Inhibitors ,Tissue Distribution ,Amino Acid Sequence - Abstract
Patients with metastatic medullary thyroid cancer (MTC) have limited systemic treatment options. The use of radiolabeled gastrin analogs targeting the cholecystokinin-2 receptor (CCK2R) is an attractive approach. However, their therapeutic efficacy is presumably decreased by their enzymatic degradation in vivo. We aimed to investigate whether the chemically stabilized analog
- Published
- 2018
8. Towards sensitive, high-throughput, biomolecular assays based on fluorescence lifetime
- Author
-
Wilfried Uhring, Anastasia Ioanna Skilitsi, Jérémie Léonard, Sophie Barre, Timothé Turko, Ulrich Hassiepen, Damien Cianfarani, Institut de Physique et Chimie des Matériaux de Strasbourg (IPCMS), Université de Strasbourg (UNISTRA)-Matériaux et nanosciences d'Alsace (FMNGE), Institut de Chimie du CNRS (INC)-Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Chimie du CNRS (INC)-Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre National de la Recherche Scientifique (CNRS)-Réseau nanophotonique et optique, Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA), Université Louis Pasteur - Strasbourg I-Centre National de la Recherche Scientifique (CNRS), Laboratoire des sciences de l'ingénieur, de l'informatique et de l'imagerie (ICube), Institut National des Sciences Appliquées - Strasbourg (INSA Strasbourg), Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-École Nationale du Génie de l'Eau et de l'Environnement de Strasbourg (ENGEES)-Réseau nanophotonique et optique, Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Matériaux et nanosciences d'Alsace (FMNGE), Institut de Chimie du CNRS (INC)-Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Chimie du CNRS (INC)-Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS)-Matériaux et Nanosciences Grand-Est (MNGE), Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Réseau nanophotonique et optique, Université de Strasbourg (UNISTRA)-Université de Haute-Alsace (UHA) Mulhouse - Colmar (Université de Haute-Alsace (UHA))-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)-Centre National de la Recherche Scientifique (CNRS), École Nationale du Génie de l'Eau et de l'Environnement de Strasbourg (ENGEES)-Université de Strasbourg (UNISTRA)-Institut National des Sciences Appliquées - Strasbourg (INSA Strasbourg), and Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-Institut National de Recherche en Informatique et en Automatique (Inria)-Les Hôpitaux Universitaires de Strasbourg (HUS)-Centre National de la Recherche Scientifique (CNRS)-Matériaux et Nanosciences Grand-Est (MNGE)
- Subjects
0301 basic medicine ,Negative control ,Context (language use) ,Nanotechnology ,01 natural sciences ,Fluorescence ,03 medical and health sciences ,General Materials Science ,Trypsin ,Droplet microfluidics ,Instrumentation ,Throughput (business) ,Spectroscopy ,Enzyme Assays ,Fluorescent Dyes ,chemistry.chemical_classification ,Biomolecule ,Microfluidic Analytical Techniques ,Chimie/Chimie théorique et/ou physique ,Atomic and Molecular Physics, and Optics ,Photon counting ,0104 chemical sciences ,[CHIM.THEO]Chemical Sciences/Theoretical and/or physical chemistry ,010404 medicinal & biomolecular chemistry ,030104 developmental biology ,chemistry ,Biological Assay ,Peptides - Abstract
Time-resolved fluorescence detection for robust sensing of biomolecular interactions is developed by implementing time-correlated single photon counting in high-throughput conditions. Droplet microfluidics is used as a promising platform for the very fast handling of low-volume samples. We illustrate the potential of this very sensitive and cost-effective technology in the context of an enzymatic activity assay based on fluorescently-labeled biomolecules. Fluorescence lifetime detection by time-correlated single photon counting is shown to enable reliable discrimination between positive and negative control samples at a throughput as high as several hundred samples per second.
- Published
- 2017
9. Structure of the DDB1-CRBN E3 ubiquitin ligase in complex with thalidomide
- Author
-
Fabrizio C. Serluca, Kerstin Böhm, Michael B. Stadler, Markus Schirle, Vincent Acker, Johannes Ottl, Eric S. Fischer, Nicolas H. Thomä, Marc Hild, Haidi Yang, John R. Lydeard, Gondichatnahalli M. Lingaraju, Jane Nagel, J. Wade Harper, Simone Cavadini, William C. Forrester, Jeremy L. Jenkins, Ulrich Hassiepen, Rohan Eric John Beckwith, Ritesh Bhanudasji Tichkule, and Michael Schebesta
- Subjects
Models, Molecular ,Ubiquitin-Protein Ligases ,Crystallography, X-Ray ,Article ,Substrate Specificity ,DDB1 ,Structure-Activity Relationship ,Ubiquitin ,medicine ,Humans ,Lenalidomide ,Adaptor Proteins, Signal Transducing ,chemistry.chemical_classification ,Homeodomain Proteins ,DNA ligase ,Multidisciplinary ,biology ,Cereblon ,Proteolysis targeting chimera ,Pomalidomide ,Molecular biology ,3. Good health ,Ubiquitin ligase ,Thalidomide ,DNA-Binding Proteins ,chemistry ,Multiprotein Complexes ,biology.protein ,Cancer research ,medicine.drug ,Peptide Hydrolases ,Protein Binding ,Transcription Factors - Abstract
In the 1950s, the drug thalidomide, administered as a sedative to pregnant women, led to the birth of thousands of children with multiple defects. Despite the teratogenicity of thalidomide and its derivatives lenalidomide and pomalidomide, these immunomodulatory drugs (IMiDs) recently emerged as effective treatments for multiple myeloma and 5q-deletion-associated dysplasia. IMiDs target the E3 ubiquitin ligase CUL4-RBX1-DDB1-CRBN (known as CRL4(CRBN)) and promote the ubiquitination of the IKAROS family transcription factors IKZF1 and IKZF3 by CRL4(CRBN). Here we present crystal structures of the DDB1-CRBN complex bound to thalidomide, lenalidomide and pomalidomide. The structure establishes that CRBN is a substrate receptor within CRL4(CRBN) and enantioselectively binds IMiDs. Using an unbiased screen, we identified the homeobox transcription factor MEIS2 as an endogenous substrate of CRL4(CRBN). Our studies suggest that IMiDs block endogenous substrates (MEIS2) from binding to CRL4(CRBN) while the ligase complex is recruiting IKZF1 or IKZF3 for degradation. This dual activity implies that small molecules can modulate an E3 ubiquitin ligase and thereby upregulate or downregulate the ubiquitination of proteins.
- Published
- 2014
10. Fluorescence Lifetime–Based Competitive Binding Assays for Measuring the Binding Potency of Protease Inhibitors In Vitro
- Author
-
Andreas Boettcher, Nathalie Gradoux, Frederic Cumin, Julian Woelcke, Trixi Brandl, David Orain, Edwige Lorthiois, Nikolaus Schiering, and Ulrich Hassiepen
- Subjects
Proteases ,medicine.medical_treatment ,Molecular Conformation ,Peptide ,Buffers ,Binding, Competitive ,Biochemistry ,Analytical Chemistry ,Dephosphorylation ,Inhibitory Concentration 50 ,chemistry.chemical_compound ,Catalytic Domain ,Drug Discovery ,Fluorescence Resonance Energy Transfer ,medicine ,Humans ,Protease Inhibitors ,Lung ,Fluorescent Dyes ,chemistry.chemical_classification ,Protease ,Drug discovery ,Hydrogen-Ion Concentration ,Recombinant Proteins ,Molecular Weight ,Acridone ,Kinetics ,Spectrometry, Fluorescence ,Förster resonance energy transfer ,chemistry ,Molecular Medicine ,Phosphorylation ,Tryptases ,Serine Proteases ,Peptides ,Acridones ,Protein Binding ,Biotechnology - Abstract
Fluorescence lifetime (FLT)-based assays have developed to become highly attractive tools in drug discovery. All recently published examples of FLT-based assays essentially describe their use for monitoring enzyme-mediated peptide modifications, such as proteolytic cleavage or phosphorylation/dephosphorylation. Here we report the development of competitive binding assays as novel, inhibitor-centric assays, principally employing the FLT of the acridone dye Puretime 14 (PT14) as the readout parameter. Exemplified with two case studies on human serine proteases, the details of the rationale for both the design and synthesis of probes (i.e., active site-directed low-molecular-weight inhibitors conjugated to PT14) are provided. Data obtained from testing inhibitors with the novel assay format match those obtained with alternative formats such as FLT-based protease activity and time-resolved fluorescence resonance energy transfer-based competitive binding assays.
- Published
- 2014
11. Structural Basis of BRCC36 Function in DNA Repair and Immune Regulation
- Author
-
W. Abdulrahman, Adel F. M. Ibrahim, Ronald T. Hay, Martin Renatus, Aimee H. Marceau, Ulrich Hassiepen, Simone Cavadini, Antoine H.F.M. Peters, Seth M. Rubin, Julius Rabl, Jacob D. Aguirre, Tewis Bouwmeester, Richard D. Bunker, Nicolas H. Thomä, H. van Attikum, Niels Mailand, M.E. Gill, Andreas D. Schenk, Emma Branigan, Claire Guérillon, A. Andres-Pons, L. Gelman, and Luijsterburg
- Subjects
Cytoplasm ,DNA Repair ,DNA repair ,Protein subunit ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Ubiquitin ,Nuclear Matrix-Associated Proteins ,Neoplasms ,DNA Repair Protein ,BRISC ,Humans ,DNA Breaks, Double-Stranded ,Histone Chaperones ,BRCA1-A ,Ubiquitin signaling ,Deubiquitination ,Regulation ,SHMT2 ,Binding site ,Molecular Biology ,deubiquitination ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,Immunity, Cellular ,Binding Sites ,biology ,Deubiquitinating Enzymes ,BRCA1 Protein ,Ubiquitination ,regulation ,Cell Biology ,ubiquitin signaling ,Cell biology ,DNA-Binding Proteins ,chemistry ,Multiprotein Complexes ,biology.protein ,Ubiquitin-Specific Proteases ,Carrier Proteins ,030217 neurology & neurosurgery ,Function (biology) ,DNA ,HeLa Cells ,Protein Binding - Abstract
Summary In mammals, ∼100 deubiquitinases act on ∼20,000 intracellular ubiquitination sites. Deubiquitinases are commonly regarded as constitutively active, with limited regulatory and targeting capacity. The BRCA1-A and BRISC complexes serve in DNA double-strand break repair and immune signaling and contain the lysine-63 linkage-specific BRCC36 subunit that is functionalized by scaffold subunits ABRAXAS and ABRO1, respectively. The molecular basis underlying BRCA1-A and BRISC function is currently unknown. Here we show that in the BRCA1-A complex structure, ABRAXAS integrates the DNA repair protein RAP80 and provides a high-affinity binding site that sequesters the tumor suppressor BRCA1 away from the break site. In the BRISC structure, ABRO1 binds SHMT2α, a metabolic enzyme enabling cancer growth in hypoxic environments, which we find prevents BRCC36 from binding and cleaving ubiquitin chains. Our work explains modularity in the BRCC36 DUB family, with different adaptor subunits conferring diversified targeting and regulatory functions., Graphical Abstract, Highlights • Detailed structural studies of the BRCA1-A and BRISC-SHMT2 protein complex • Complex-specific subunits functionalize BRCC36, regulating its activity and targeting • BRCA1-A sequesters BRCA1 with nanomolar affinity • Cancer-associated metabolic enzyme SHMT2 inhibits BRISC deubiquitination activity, Structural studies of the BRCA1-A and BRISC complexes reveal a common enzymatic core that is functionalized by complex-specific proteins. BRCA1-A localizes to DNA double-strand breaks, sequesters BRCA1, and suppresses DNA resection. BRISC regulates protein degradation by the endosomal-lysosomal pathway through deubiquitination and is inhibited by the metabolic enzyme SHMT2.
- Published
- 2019
12. A Novel Class of Oral Direct Renin Inhibitors: Highly Potent 3,5-Disubstituted Piperidines Bearing a Tricyclic P3–P1 Pharmacophore
- Author
-
Claus Ehrhardt, Takatoshi Kosaka, Jörg Trappe, Richard Sedrani, Bernd Gerhartz, Jürgen Maibaum, Simon Ruedisser, Sabine Geisse, Dean F. Rigel, Frederic Cumin, Johannes Ottl, Nils Ostermann, Trixie Wagner, Markus Krömer, Andreas Marzinzik, Eric Vangrevelinghe, Paul Richert, Edgar Jacoby, Martin Klumpp, Daniel K. Baeschlin, Eric Francotte, Randy L. Webb, J. Constanze D. Hartwieg, Werner Breitenstein, and Ulrich Hassiepen
- Subjects
Models, Molecular ,Protein Conformation ,Stereochemistry ,Peptidomimetic ,In silico ,Administration, Oral ,Biological Availability ,Inhibitory Concentration 50 ,chemistry.chemical_compound ,Piperidines ,Renin ,Drug Discovery ,Animals ,Protease Inhibitors ,chemistry.chemical_classification ,biology ,Active site ,Combinatorial chemistry ,Rats ,Enzyme ,chemistry ,Docking (molecular) ,Drug Design ,biology.protein ,Molecular Medicine ,Piperidine ,Pharmacophore ,Tricyclic - Abstract
A small library of fragments comprising putative recognition motifs for the catalytic dyad of aspartic proteases was generated by in silico similarity searches within the corporate compound deck based on rh-renin active site docking and scoring filters. Subsequent screening by NMR identified the low-affinity hits 3 and 4 as competitive active site binders, which could be shown by X-ray crystallography to bind to the hydrophobic S3-S1 pocket of rh-renin. As part of a parallel multiple hit-finding approach, the 3,5-disubstituted piperidine (rac)-5 was discovered by HTS using a enzymatic assay. X-ray crystallography demonstrated the eutomer (3S,5R)-5 to be a peptidomimetic inhibitor binding to a nonsubstrate topography of the rh-renin prime site. The design of the potent and selective (3S,5R)-12 bearing a P3(sp)-tethered tricyclic P3-P1 pharmacophore derived from 3 is described. (3S,5R)-12 showed oral bioavailability in rats and demonstrated blood pressure lowering activity in the double-transgenic rat model.
- Published
- 2013
13. The Discovery of Novel Potent trans-3,4-Disubstituted Pyrrolidine Inhibitors of the Human Aspartic Protease Renin from in Silico Three-Dimensional (3D) Pharmacophore Searches
- Author
-
Werner Breitenstein, Eric Francotte, Ulrich Hassiepen, Claus Ehrhardt, Takatoshi Kosaka, Nils Ostermann, Jürgen Maibaum, Holger Sellner, Paul Richert, Frederic Cumin, Trixie Wagner, Edgar Jacoby, Randy L. Webb, Edwige Lorthiois, and Dean F. Rigel
- Subjects
Models, Molecular ,Pyrrolidines ,Protein Conformation ,Stereochemistry ,In silico ,Administration, Oral ,Biological Availability ,Pyrrolidine ,law.invention ,Structure-Activity Relationship ,chemistry.chemical_compound ,law ,Renin ,Drug Discovery ,Animals ,Humans ,Protease Inhibitors ,chemistry.chemical_classification ,biology ,Diphenylamine ,Computational Biology ,Active site ,Rats ,Enzyme ,chemistry ,biology.protein ,Recombinant DNA ,Molecular Medicine ,Enantiomer ,Pharmacophore - Abstract
The small-molecule trans-3,4-disubstituted pyrrolidine 6 was identified from in silico three-dimensional (3D) pharmacophore searches based on known X-ray structures of renin-inhibitor complexes and demonstrated to be a weakly active inhibitor of the human enzyme. The unexpected binding mode of the more potent enantiomer (3S,4S)-6a in an extended conformation spanning the nonprime and S1' pockets of the recombinant human (rh)-renin active site was elucidated by X-ray crystallography. Initial structure-activity relationship work focused on modifications of the hydrophobic diphenylamine portion positioned in S1 and extending toward the S2 pocket. Replacement with an optimized P3-P1 pharmacophore interacting to the nonsubstrate S3(sp) cavity eventually resulted in significantly improved in vitro potency and selectivity. The prototype analogue (3S,4S)-12a of this new class of direct renin inhibitors exerted blood pressure lowering effects in a hypertensive double-transgenic rat model after oral administration.
- Published
- 2013
14. Targeted inhibition of the COP9 signalosome for treatment of cancer
- Author
-
Martin Renatus, Jean Quancard, Matthew Jones, Eva Altmann, Christian Wiesmann, Anita Schlierf, Michael Kiffe, Bruno Martoglio, Anne B. Jefferson, Rene Assenberg, Andreas Weiss, Richard Sedrani, Jörg Eder, Michael Schaefer, and Ulrich Hassiepen
- Subjects
0301 basic medicine ,NEDD8 Protein ,THP-1 Cells ,Ubiquitin-Protein Ligases ,Science ,General Physics and Astronomy ,Antineoplastic Agents ,Mice, SCID ,NEDD8 ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,Ubiquitin ,Animals ,Humans ,Molecular Targeted Therapy ,COP9 signalosome ,Regulation of gene expression ,Multidisciplinary ,biology ,COP9 Signalosome Complex ,Activator (genetics) ,Intracellular Signaling Peptides and Proteins ,Azepines ,General Chemistry ,HCT116 Cells ,Xenograft Model Antitumor Assays ,Molecular biology ,Tumor Burden ,Cell biology ,Gene Expression Regulation, Neoplastic ,Isoenzymes ,030104 developmental biology ,Apoptosis ,Proteolysis ,biology.protein ,Lymphoma, Large-Cell, Anaplastic ,Pyrazoles ,Female ,Protein Processing, Post-Translational ,Peptide Hydrolases - Abstract
The COP9 signalosome (CSN) is a central component of the activation and remodelling cycle of cullin-RING E3 ubiquitin ligases (CRLs), the largest enzyme family of the ubiquitin–proteasome system in humans. CRLs are implicated in the regulation of numerous cellular processes, including cell cycle progression and apoptosis, and aberrant CRL activity is frequently associated with cancer. Remodelling of CRLs is initiated by CSN-catalysed cleavage of the ubiquitin-like activator NEDD8 from CRLs. Here we describe CSN5i-3, a potent, selective and orally available inhibitor of CSN5, the proteolytic subunit of CSN. The compound traps CRLs in the neddylated state, which leads to inactivation of a subset of CRLs by inducing degradation of their substrate recognition module. CSN5i-3 differentially affects the viability of tumour cell lines and suppresses growth of a human xenograft in mice. Our results provide insights into how CSN regulates CRLs and suggest that CSN5 inhibition has potential for anti-tumour therapy., Dysregulation of protein degradation by the ubiquitin-proteasome system is a feature commonly associated with cancer. Here, the authors develop an orally available small molecule that inhibits CSN5, the proteolytic subunit of the COP9 signalosome, and blocks tumour growth in a xenograft model.
- Published
- 2016
15. Azaindoles as Zinc-Binding Small-Molecule Inhibitors of the JAMM Protease CSN5
- Author
-
Simon Ruedisser, Bruno Martoglio, Michael Schaefer, Paul Erbel, Laurence Kieffer, Ulrich Hassiepen, Anita Schlierf, Martin Renatus, Mickael Sorge, Daniela Ostermeier, Adelaide Druet, Jean Quancard, Matthew Jones, Anne B. Jefferson, Keith B. Pfister, and Eva Altmann
- Subjects
0301 basic medicine ,Indoles ,NEDD8 Protein ,Protein subunit ,medicine.medical_treatment ,Cleavage (embryo) ,Crystallography, X-Ray ,NEDD8 ,Catalysis ,03 medical and health sciences ,Ubiquitin ,Catalytic Domain ,medicine ,Fluorescence Resonance Energy Transfer ,Humans ,COP9 signalosome ,RNA, Small Interfering ,S-Phase Kinase-Associated Proteins ,Cell Proliferation ,Metalloproteinase ,Protease ,Binding Sites ,030102 biochemistry & molecular biology ,biology ,Chemistry ,COP9 Signalosome Complex ,General Chemistry ,HCT116 Cells ,Small molecule ,Molecular Docking Simulation ,Protein Subunits ,Zinc ,030104 developmental biology ,Biochemistry ,biology.protein ,RNA Interference - Abstract
CSN5 is the zinc metalloprotease subunit of the COP9 signalosome (CSN), which is an important regulator of cullin-RING E3 ubiquitin ligases (CRLs). CSN5 is responsible for the cleavage of NEDD8 from CRLs, and blocking deconjugation of NEDD8 traps the CRLs in a hyperactive state, thereby leading to auto-ubiquitination and ultimately degradation of the substrate recognition subunits. Herein, we describe the discovery of azaindoles as a new class of CSN5 inhibitors, which interact with the active-site zinc ion of CSN5 through an unprecedented binding mode. The best compounds inhibited CSN5 with nanomolar potency, led to degradation of the substrate recognition subunit Skp2 in cells, and reduced the viability of HCT116 cells.
- Published
- 2016
16. Cullin–RING ubiquitin E3 ligase regulation by the COP9 signalosome
- Author
-
Ulrich Hassiepen, Radosav S. Pantelic, Wassim Abdulrahman, Alessandro Potenza, Richard D. Bunker, Henning Stahlberg, Rohan Eric John Beckwith, Georg Petzold, Ritesh Bhanudasji Tichkule, Mahamadou Faty, Weaam I. Mohamed, Kaoru Sugasawa, Gondichatnahalli M. Lingaraju, Simone Cavadini, Kenneth N. Goldie, Eric S. Fischer, Syota Matsumoto, and Nicolas H. Thomä
- Subjects
0301 basic medicine ,Models, Molecular ,Crystallography, X-Ray ,NEDD8 ,03 medical and health sciences ,Ubiquitin ,Allosteric Regulation ,Humans ,COP9 signalosome ,Ubiquitins ,Multidisciplinary ,Binding Sites ,biology ,COP9 Signalosome Complex ,Cullin Proteins ,Cryoelectron Microscopy ,Ubiquitination ,Cell biology ,Ubiquitin ligase ,DNA-Binding Proteins ,Kinetics ,030104 developmental biology ,Multiprotein Complexes ,biology.protein ,Biocatalysis ,Apoproteins ,Carrier Proteins ,Cullin ,DNA Damage ,Peptide Hydrolases ,Protein Binding - Abstract
The cullin-RING ubiquitin E3 ligase (CRL) family comprises over 200 members in humans. The COP9 signalosome complex (CSN) regulates CRLs by removing their ubiquitin-like activator NEDD8. The CUL4A-RBX1-DDB1-DDB2 complex (CRL4A(DDB2)) monitors the genome for ultraviolet-light-induced DNA damage. CRL4A(DBB2) is inactive in the absence of damaged DNA and requires CSN to regulate the repair process. The structural basis of CSN binding to CRL4A(DDB2) and the principles of CSN activation are poorly understood. Here we present cryo-electron microscopy structures for CSN in complex with neddylated CRL4A ligases to 6.4 A resolution. The CSN conformers defined by cryo-electron microscopy and a novel apo-CSN crystal structure indicate an induced-fit mechanism that drives CSN activation by neddylated CRLs. We find that CSN and a substrate cannot bind simultaneously to CRL4A, favouring a deneddylated, inactive state for substrate-free CRL4 complexes. These architectural and regulatory principles appear conserved across CRL families, allowing global regulation by CSN.
- Published
- 2016
- Full Text
- View/download PDF
17. A Fluorescence Lifetime-Based Assay for Abelson Kinase
- Author
-
Gabriele Meder, Klaus Doering, Julian Woelcke, Peter Drueckes, Ulrich Hassiepen, Stephan Pritz, and Lorenz M. Mayr
- Subjects
Peptide ,Biochemistry ,Substrate Specificity ,Analytical Chemistry ,Inhibitory Concentration 50 ,Humans ,Amino Acid Sequence ,Phosphorylation ,Tyrosine ,Phosphotyrosine ,Proto-Oncogene Proteins c-abl ,Peptide sequence ,Fluorescent Dyes ,chemistry.chemical_classification ,Quenching (fluorescence) ,Kinase ,Spectrometry, Fluorescence ,chemistry ,Molecular Medicine ,Biological Assay ,Peptides ,Tyrosine kinase ,Biotechnology ,Cysteine - Abstract
We present a novel homogeneous in vitro assay format and apply it to the quantitative determination of the enzymatic activity of a tyrosine kinase. The assay employs a short peptidic substrate containing a single tyrosine and a single probe attached via a cysteine side chain. The structural flexibility of the peptide allows for the dynamic quenching of the probe by the nonphosphorylated tyrosine side chain. The probe responds with changes in its fluorescence lifetime depending on the phosphorylation state of the tyrosine. We use this effect to directly follow the enzymatic phosphorylation of the substrate, without having to resort to additional assay components such as an antibody against the phosphotyrosine. As an example for the application of this assay principle, we present results from the development of an assay for Abelson kinase (c-Abl) used for compound profiling. Adjustments in the peptide sequence would make this assay format suitable to a wide variety of other tyrosine kinases.
- Published
- 2011
18. Fragment-Based Screening by Biochemical Assays: Systematic Feasibility Studies with Trypsin and MMP12
- Author
-
Simon Ruedisser, Nikolaus Schiering, P. Erbel, Pascal Rigollier, Andreas Boettcher, D. Vinzenz, Julian Woelcke, Ulrich Hassiepen, and Lorenz M. Mayr
- Subjects
Magnetic Resonance Spectroscopy ,Light ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Biochemistry ,Fluorescence ,Mass Spectrometry ,Analytical Chemistry ,Matrix Metalloproteinase 12 ,medicine ,Animals ,Humans ,Scattering, Radiation ,False Positive Reactions ,Trypsin ,Electrophoretic mobility shift assay ,Surface plasmon resonance ,False Negative Reactions ,Serine protease ,Protease ,biology ,Chemistry ,Drug discovery ,Nuclear magnetic resonance spectroscopy ,Surface Plasmon Resonance ,Peptide Fragments ,Enzyme assay ,Kinetics ,Solubility ,biology.protein ,Feasibility Studies ,Molecular Medicine ,Biological Assay ,Cattle ,Chromatography, Liquid ,Biotechnology ,medicine.drug - Abstract
Fragment-based screening (FBS) has gained acceptance in the pharmaceutical industry as an attractive approach for the identification of new chemical starting points for drug discovery programs in addition to classical strategies such as high-throughput screening. There is the concern that screening of fragments at high µM concentrations in biochemical assays results in increased false-positive and false-negative rates. Here the authors systematically compare the data quality of FBS obtained by enzyme activity-based fluorescence intensity, fluorescence lifetime, and mobility shift assays with the data quality from surface plasmon resonance (SPR) and nuclear magnetic resonance (NMR) methods. The serine protease trypsin and the matrix metalloprotease MMP12 were selected as model systems. For both studies, 352 fragments were selected each. From the data generated, all 3 biochemical protease assay methods can be used for screening of fragments with low false-negative and low false-positive rates, comparable to those achieved with the SPR-based assays. It can also be concluded that only fragments with a solubility higher than the screening concentration determined by means of NMR should be used for FBS purposes. Extrapolated to 10,000 fragments, the biochemical assays speed up the primary FBS process by approximately a factor of 10 and reduce the protease consumption by approximately 10,000-fold compared to NMR protein observation experiments.
- Published
- 2010
19. Adverse effects of dipeptidyl peptidases 8 and 9 inhibition in rodents revisited
- Author
-
J. Trappe, M. Juedes, James E. Foley, Peter Hoffmann, Bryan Burkey, and Ulrich Hassiepen
- Subjects
Male ,Dipeptidases ,Pyrrolidines ,animal structures ,Endocrinology, Diabetes and Metabolism ,Adamantane ,Mice, Inbred Strains ,Pharmacology ,Dipeptidyl peptidase ,Mice ,Endocrinology ,In vivo ,Nitriles ,Toxicity Tests ,Internal Medicine ,medicine ,Animals ,Humans ,Vildagliptin ,Reticulocytopenia ,Dosing ,Rats, Wistar ,Dipeptidyl-Peptidases and Tripeptidyl-Peptidases ,Adverse effect ,Dipeptidyl-Peptidase IV Inhibitors ,business.industry ,medicine.disease ,Recombinant Proteins ,Rats ,Diabetes Mellitus, Type 2 ,Toxicity ,Female ,Lymph ,business ,medicine.drug - Abstract
Aim: To evaluate the association between inhibition of dipeptidyl peptidase (DPP)-8 and/or DPP-9 organ toxicities and mortality in rodents. Research Design and Methods: The relative selectivity of the DPP-4 inhibitor, vildagliptin, was determined by comparing its KI (concentration of compound yielding 50% inhibition of the enzyme) values for inhibition of recombinant human DPP-4, DPP-8 and DPP-9 assessed in vitro. In experiments performed in vivo, vildagliptin was administered by gavage for 13 weeks, at doses up to 1500 mg/kg/day in CD-1 mice and at doses up to 900 mg/kg/day in Wistar rats. Plasma concentrations of vildagliptin were assessed at week 12, and toxicities previously ascribed to inhibition of DPP-8 and/or DPP-9 were assessed at week 13. Results: The KI values for vildagliptin-induced inhibition of DPP-4, DPP-8 and DPP-9 were 3, 810 and 95 nM respectively. The mean plasma concentration 24 h after dose after 12-week daily dosing with 1500 mg/kg/day in mice was 2279 nM. The mean plasma drug level 24 h after dose after 12-week daily dosing with 900 mg/kg/day in rats was 5729 nM. These high doses maintained plasma drug levels well above the KI values for DPP-8 and DPP-9 throughout a 24-h period. At these high doses, the toxicities of a selective DPP-8/DPP-9 inhibitor that were reported previously (100% mortality in mice, alopecia, thrombocytopenia, reticulocytopenia, enlarged lymph nodes, splenomegaly and 20% mortality in rats) were not observed. Conclusions: Inhibition of DPP-8 and DPP-9 per se does not lead to organ toxicities and mortality in rodents. Thus, a mechanism other than DPP-8/DPP-9 inhibition likely underlies the toxicity previously reported to be associated with a selective DPP-8/DPP-9 inhibitor.
- Published
- 2008
20. Crystal structure of the human COP9 signalosome
- Author
-
Daniel Hess, Martin Renatus, Simone Cavadini, Gondichatnahalli M. Lingaraju, Eric S. Fischer, Richard D. Bunker, Ulrich Hassiepen, and Nicolas H. Thomä
- Subjects
Models, Molecular ,Protein subunit ,Plasma protein binding ,Crystallography, X-Ray ,NEDD8 ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Catalytic Domain ,Humans ,COP9 signalosome ,Transcription factor ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,COP9 Signalosome Complex ,Intracellular Signaling Peptides and Proteins ,Cell biology ,Ubiquitin ligase ,Protein Structure, Tertiary ,Enzyme Activation ,Proteasome ,030220 oncology & carcinogenesis ,Multiprotein Complexes ,biology.protein ,Peptide Hydrolases ,Protein Binding ,Transcription Factors - Abstract
Ubiquitination is a crucial cellular signalling process, and is controlled on multiple levels. Cullin–RING E3 ubiquitin ligases (CRLs) are regulated by the eight-subunit COP9 signalosome (CSN). CSN inactivates CRLs by removing their covalently attached activator, NEDD8. NEDD8 cleavage by CSN is catalysed by CSN5, a Zn2+-dependent isopeptidase that is inactive in isolation. Here we present the crystal structure of the entire ∼350-kDa human CSN holoenzyme at 3.8 A resolution, detailing the molecular architecture of the complex. CSN has two organizational centres: a horseshoe-shaped ring created by its six proteasome lid–CSN–initiation factor 3 (PCI) domain proteins, and a large bundle formed by the carboxy-terminal α-helices of every subunit. CSN5 and its dimerization partner, CSN6, are intricately embedded at the core of the helical bundle. In the substrate-free holoenzyme, CSN5 is autoinhibited, which precludes access to the active site. We find that neddylated CRL binding to CSN is sensed by CSN4, and communicated to CSN5 with the assistance of CSN6, resulting in activation of the deneddylase. The COP9 signalosome (CSN) complex regulates cullin–RING E3 ubiquitin ligases—the largest class of ubiquitin ligase enzymes, which are involved in a multitude of regulatory processes; here, the crystal structure of the entire human CSN holoenzyme is presented. The COP9 signalosome (CSN) is a large protein complex that functions in the ubiquitin–proteasome intracellular protein degradation pathway. First identified 20 years ago in developing Arabidopsis seedlings, it is now thought to be part of the regulatory machinery in all animals, plants and fungi. Here, Nicolas Thoma and co-workers present the crystal structure of the entire eight-subunit human COP9 signalosome at 3.8 A resolution, providing insights into its molecular architecture and mechanism of action. The structure reveals how the complex achieves such exquisite specificity for its substrates.
- Published
- 2014
21. Plasma contact system activation drives anaphylaxis in severe mast cell-mediated allergic reactions
- Author
-
Antonio Di Gennaro, Ulrich Hassiepen, Katrin F. Nickel, Tobias A. Fuchs, Olga Luengo, Andy T. Long, Evi X. Stavrou, Thomas Renné, Thorsten Krieger, Ellinor Kenne, Hartmut Schlüter, Keith R. McCrae, Stefanie Flohr, Riccardo Senter, Mar Guilarte, Moisés Labrador, Jenny Björkqvist, Lynn M. Butler, Anne Jämsä, Frederic Cumin, Anna Sala-Cunill, Victoria Cardona, Linda Labberton, Parvin Kumar, and Coen Maas
- Subjects
Male ,Time Factors ,Receptor, Bradykinin B2 ,High-molecular-weight kininogen ,Immunoglobulin E ,chemistry.chemical_compound ,Mice ,Bradykinin B2 ,Immunology and Allergy ,Mast Cells ,Non-U.S. Gov't ,Mice, Knockout ,Factor XII ,Kininogen ,biology ,Chemistry ,Research Support, Non-U.S. Gov't ,Middle Aged ,Mast cell ,medicine.anatomical_structure ,Female ,Hypotension ,Receptor ,circulatory and respiratory physiology ,Signal Transduction ,Adult ,Knockout ,Immunology ,tryptase ,Bradykinin ,Tryptase ,Research Support ,Young Adult ,medicine ,Journal Article ,Hypersensitivity ,Animals ,Humans ,mouse models ,cardiovascular diseases ,Anaphylaxis ,Aged ,Animal ,Kininogens ,Kallikrein ,contact system ,Disease Models, Animal ,Disease Models ,biology.protein ,bradykinin ,mast cell ,Biomarkers - Abstract
Background Anaphylaxis is an acute, potentially lethal, multisystem syndrome resulting from the sudden release of mast cell–derived mediators into the circulation. Objectives and Methods We report here that a plasma protease cascade, the factor XII–driven contact system, critically contributes to the pathogenesis of anaphylaxis in both murine models and human subjects. Results Deficiency in or pharmacologic inhibition of factor XII, plasma kallikrein, high-molecular-weight kininogen, or the bradykinin B2 receptor, but not the B1 receptor, largely attenuated allergen/IgE-mediated mast cell hyperresponsiveness in mice. Reconstitutions of factor XII null mice with human factor XII restored susceptibility for allergen/IgE-mediated hypotension. Activated mast cells systemically released heparin, which provided a negatively charged surface for factor XII autoactivation. Activated factor XII generates plasma kallikrein, which proteolyzes kininogen, leading to the liberation of bradykinin. We evaluated the contact system in patients with anaphylaxis. In all 10 plasma samples immunoblotting revealed activation of factor XII, plasma kallikrein, and kininogen during the acute phase of anaphylaxis but not at basal conditions or in healthy control subjects. The severity of anaphylaxis was associated with mast cell degranulation, increased plasma heparin levels, the intensity of contact system activation, and bradykinin formation. Conclusions In summary, the data collectively show a role of the contact system in patients with anaphylaxis and support the hypothesis that targeting bradykinin generation and signaling provides a novel and alternative treatment strategy for anaphylactic attacks.
- Published
- 2014
22. Analysis of protein self-association at constant concentration by fluorescence-energy transfer
- Author
-
Volker J. Lenz, Peter Krüger, Hans-Gregor Gattner, Thomas Mülders, Ulrich Hassiepen, Axel Wollmer, and Matthias Federwisch
- Subjects
Circular dichroism ,Circular Dichroism ,Dimer ,Intermolecular force ,Biochemistry ,Fluorescence ,Acceptor ,Dissociation (chemistry) ,chemistry.chemical_compound ,Crystallography ,Spectrometry, Fluorescence ,Monomer ,chemistry ,Insulin ,Determination of equilibrium constants - Abstract
Fluorescence-resonance-energy transfer from subunits labelled with a fluorescence donor group to subunits labelled with a fluorescence acceptor group can be used for quantitative analysis of protein self-association. The present approach evaluates fluorescence measurements on mixtures of equimolar solutions of donor-labelled and acceptor-labelled protein composed by systematic variation of the volume ratio. Its attractive feature is that it allows the determination of equilibrium constants at fixed total concentration. Problems encountered by most other methods, which require the equilibria to be followed to high dilution, are avoided. Conditions to be fulfilled are that a reactive site is available on the protein for specific introduction of the labels and that labelling neither affects the conformation nor interferes with the intermolecular interactions. It is desirable that the Forster distance of the donor/acceptor pair complies with its separation. While dimerisation constants can be determined exclusively by fluorescence measurements, the analysis of more complex cases of self-association depends on additional independent information. This communication reports on an application of the approach to the association/dissociation equilibrium between insulin monomers and dimers. Labelling of insulin at the epsilon-amino group of LysB29 does not disturb the conformation nor does it affect dimerisation. 2-Aminobenzoyl and 3-nitrotyrosyl residues served as the donor/acceptor pairs. Because they are less bulky than most other fluorescence labels and are of balanced polarity they do not alter the chemical nature of the protein. Their Forster distance of 29 A matches their 32-A separation in the insulin dimer. Energy transfer was measured as a function of the molar fractions of donor-insulin and acceptor-insulin at constant total concentration. Evaluation of this dependence resulted in a dimerisation constant, K12, of 0.72x10(5) M(-1). Its agreement with values obtained with other methods demonstrates that the present approach is a reliable alternative.
- Published
- 1998
23. Discovery of C-(1-aryl-cyclohexyl)-methylamines as selective, orally available inhibitors of dipeptidyl peptidase IV
- Author
-
Kenji Namoto, François Gessier, Garry Fenton, Mandy Beswick, Finton Sirockin, Alokesh Duttaroy, Suzie Ferreira, Ulrich Hassiepen, Stefanie Flohr, Bernd Gerhartz, Jörg Trappe, Jon Sutton, Nils Ostermann, David E. Clark, Daniel K. Baeschlin, and Richard Sedrani
- Subjects
animal structures ,Pyrrolidines ,Dipeptidyl Peptidase 4 ,Clinical Biochemistry ,Pharmaceutical Science ,Administration, Oral ,Adamantane ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Dipeptidyl peptidase ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Methylamines ,Pharmacokinetics ,Drug Discovery ,Nitriles ,Animals ,Humans ,Molecular Biology ,Vildagliptin ,Dipeptidyl-Peptidase IV Inhibitors ,Molecular Structure ,Chemistry ,Aryl ,Organic Chemistry ,Sitagliptin Phosphate ,Triazoles ,Rats ,Enzyme Activation ,Cyclization ,Pyrazines ,Molecular Medicine ,Caco-2 Cells - Abstract
The successful launches of dipeptidyl peptidase IV (DPP IV) inhibitors as oral anti-diabetics warrant and spur the further quest for additional chemical entities in this promising class of therapeutics. Numerous pharmaceutical companies have pursued their proprietary candidates towards the clinic, resulting in a large body of published chemical structures associated with DPP IV. Herein, we report the discovery of a novel chemotype for DPP IV inhibition based on the C-(1-aryl-cyclohexyl)-methylamine scaffold and its optimization to compounds which selectively inhibit DPP IV at low-nM potency and exhibit an excellent oral pharmacokinetic profile in the rat.
- Published
- 2013
24. Ubiquitin Specific Protease 2
- Author
-
Ulrich Hassiepen and Martin Renatus
- Subjects
Biochemistry ,Ubiquitin specific protease ,Biology ,Cell biology - Published
- 2013
25. Contributors
- Author
-
Catherine Anne Abbott, Carmela R. Abraham, Hideki Adachi, Osao Adachi, Zach Adam, Michael W.W. Adams, Michael J. Adang, Ibrahim M. Adham, Patrizia Aducci, David A. Agard, Alexey A. Agranovsky, Tetsuya Akamatsu, Yoshinori Akiyama, Reidar Albrechtsen, Alí Alejo, Sean M. Amberg, Alexander Y. Amerik, Piti Amparyup, Felipe Andrade, Germán Andrés, Daniel M. Andrews, Robert K. Andrews, Toni M. Antalis, Colin S. Anthony, Naoya Aoki, Suneel S. Apte, Kazunari Arima, Gérard Arlaud, Raghuvir Krishnaswamy Arni, Pascal Arnoux, Nathan N. Aronson, Michel Arthur, Yasuhisa Asano, Paolo Ascenzi, Marina T. Assakura, David S. Auld, Veridiana de Melo Rodrigues Ávila, Francesc X. Avilés, William M. Awad, Anand K. Bachhawat, Shan Bai, Teaster T. Baird, S. Paul Bajaj, Susan C. Baker, Agnieszka Banbula, Alan J. Barrett, Jemima Barrowman, John D. Bartlett, Jörg W. Bartsch, Nikola Baschuk, Isolda P. Baskova, Jyotsna Batra, Karl Bauer, Ulrich Baumann, Wolfgang Baumeister, Cédric Bauvois, Alex Bayés, Anne Beauvais, Christoph Becker-Pauly, Tadhg P. Begley, Miklós Békés, Robert Belas, Daniah Beleford, Teruhiko Beppu, Ernst M. Bergmann, Bruno A. Bernard, Dominique Bernard, Michael C. Berndt, Giovanna Berruti, Colin Berry, Greg P. Bertenshaw, Christian Betzel, Chetana Bhaskarla, Manoj Bhosale, Gabriele Bierbaum, B. Bjarnason Jón, Michael Blaber, Michael J. Blackman, Alexander Blinkovsky, Jef D. Boeke, Matthew Bogyo, Stefan Bohn, Guy Boileau, Mike Boland, Tové C. Bolken, Judith S. Bond, Jan Bondeson, Javier Bordallo, Claudia Borelli, Tiago O. Botelho, Richard R. Bott, David G. Bourne, Niels Bovenschen, Ralph A. Bradshaw, Klaus Breddam, Keith Brew, Paul J. Brindley, Diane L. Brinkman, Collette Britton, Jeff R. Broadbent, Anne Broadhurst, Dieter Brómme, Murray Broom, Jeremy S. Brown, Mark A. Brown, Iris Bruchhaus, Barbara A. Burleigh, Kristin E. Burns, James F. Burrows, Michael J. Butler, David J. Buttle, Chelsea M. Byrd, Tony Byun, Sandrine Cadel, Conor R. Caffrey, Santiago Cal, Javier Caldentey, Thomas Candela, Clemente Capasso, Daniel R. Capriogilio, Vincenzo Carginale, Adriana Karaoglanovic Carmona, Vern B. Carruthers, Francis J. Castellino, Joseph J. Catanese, Bruce Caterson, George H. Caughey, Naimh X. Cawley, Tim E. Cawston, Juan José Cazzulo, Jijie Chai, Karl X. Chai, Olga Meiri Chaim, L.S. Chang, Julie Chao, Marie-Pierre Chapot-Chartier, Jean-Louis Charli, Paulette Charlier, Karen J. Chave, Jian-Min Chen, Jinq-May Chen, Li-Mei Chen, Ya-Wen Chen, Yu-Yen Chen, Bernard Chevrier, Jean-François Chich, Jeremy Chien, Suneeta Chimalapati, Ki Joon Cho, Kwan Yong Choi, Woei-Jer Chuang, Chin Ha Chung, Ivy Yeuk Wah Chung, Christine Clamagirand, Ian M. Clark, Adrian K. Clarke, Nicola E. Clarke, Steven Gerard Clarke, Philippe Clauziat, Judith A. Clements, Catherine Coffinier, Paul Cohen, Alain Colige, Anne Collignon, Sean D. Colloms, Andreas Conzelmann, Graham H. Coombs, Jakki C. Cooney, Jonathan B. Cooper, Max D. Cooper, Nikki A. Copeland, Graeme S. Cottrell, Joseph T. Coyle, Charles S. Craik, John W.M. Creemers, Daniela Cretu, Jenifer Croce, Keith J. Cross, Rosario Cueva, Sheng Cui, Luis Cunha, Simon Cutting, Christophe d’Enfert, Hugues D’Orchymont, Björn Dahlbäck, Shujia Dai, Ross E. Dalbey, John P. Dalton, Pam M. Dando, R.M. Daniel, Sergei M. Danilov, Donna E. Davies, Heloisa S. De Araujo, Teresa De los Santos, Viviana de Luca, Ingrid De Meester, Ana Karina de Oliveira, Eduardo Brandt de Oliveira, Pedro Lagerblad De Oliveira, Sarah de Vos, Jeroen Declercq, Wim Declercq, Ala-Eddine Deghmane, Niek Dekker, Sonia Del Prete, Marina Del Rosal, Bernard Delmas, Robert DeLotto, Ilya V. Demidyuk, Mark R. Denison, Jan M. Deussing, Lakshmi A. Devi, Eleftherios P. Diamandis, Isabel Diaz, Araceli Díaz-Perales, Bauke W. Dijkstra, Yan Ding, Jack E. Dixon, Johannes Dodt, Terje Dokland, Iztok Dolenc, Ningzheng Dong, Tran Cat Dong, Ying Dong, Mitesh Dongre, Mark Donovan, Timothy M. Dore, Loretta Dorstyn, Hong Dou, Zhicheng Dou, Annette M. Dougall, Marcin Drag, Edward G. Dudley, Ben M. Dunn, Bruno Dupuy, Maria Conceicāo Duque-Magalhāes, M. Asunción Durá, Yves Eeckhout, Vincent Eijsink, Arthur Z. Eisen, Azza Eissa, Sandra Eklund, Ziad M. Eletr, Vincent Ellis, Wolfgang Engel, Ervin G. Erdös, Teresa Escalante, David A. Estell, Michael Etscheid, Herbert J. Evans, Roger D. Everett, Alex C. Faesen, Falk Fahrenholz, Miriam Fanjul-Fernández, Christopher J. Farady, Georges Feller, Hong Feng, Kurt M. Fenster, Claude Férec, Silvia Ferrari, Barbara Fingleton, Jed F. Fisher, Paula M. Fives-Taylor, Loren G. Fong, F. Forneris, Brian M. Forster, Friedrich Forster, Simon J. Foster, Thierry Foulon, Stephen I. Foundling, Jay William Fox, Bruno Franzetti, Alejandra P. Frasch, Hudson H. Freeze, Jean-Marie Frère, Teryl K. Frey, Beate Fricke, Lloyd D. Fricker, Rafael Fridman, Christopher J. Froelich, Camilla Fröhlich, Hsueh-Liang Fu, Cynthia N. Fuhrmann, Satoshi Fujimura, Hiroshi Fujiwara, Jun Fukushima, Keiichi Fukuyama, Robert S. Fuller, Martin Fusek, Christine Gaboriaud, Christian Gache, Oleksandr Gakh, Peter Gal, Junjun Gao, Adolfo García-Sastre, Donald L. Gardiner, John A. Gatehouse, G.M. Gaucher, Francis Gauthier, Jean-Marie Ghuysen, Wade Gibson, Jennifer Gillies, Elzbieta Glaser, Fabian Glaser, Michael H. Glickman, Peter Goettig, Colette Goffin, Eiichi Gohda, Alfred L. Goldberg, Daniel E. Goldberg, Gregory I. Goldberg, Nathan E. Goldfarb, F. Xavier Gomis-Rüth, B. Gopal, Alexander E. Gorbalenya, Stuart G. Gordon, Mark D. Gorrell, Friedrich Götz, Theodoros Goulas, Cécile Gouzy-Darmon, K. Govind, Lászlo Gráf, Robert R. Granados, Melissa Ann Gräwert, Douglas A. Gray, Thomas P. Graycar, Jonathan A. Green, Luiza Helena Gremski, Michael Groll, Tania Yu Gromova, P. Gros, Marvin J. Grubman, Amy M. Grunden, Ágústa Gudmundsdóttir, Micheline Guinand, Djamel Gully, Alla Gustchina, José María Gutiérrez, Byung Hak Ha, Jesper Z. Haeggström, James H. Hageman, Johanna Haiko, Stephan Hailfinger, Hans Michael Haitchi, Ji Seon Han, Chantal Hanquez, Minoru Harada, Ikuko Hara-Nishimura, Marianne Harboe, Torleif Härd, David A. Harris, Ulrich Hassiepen, Shoji Hata, Akira Hattori, Rong-Qiao He, Albert J.R. Heck, Dirk F. Hendricks, Bernhard Henrich, Patrick Henriet, Andrés Hernández-Arana, Irma Herrera-Camacho, Gerhard Heussipp, Toshihiko Hibino, P.M. Hicks, Bradley I. Hillman, B. Yukihiro Hiraoka, Jun Hiratake, Yohei Hizukuri, Heng-Chien Ho, Ngo Thi Hoa, Mark Hochstrasser, Kathryn M. Hodge, Theo Hofmann, Thomas Hohn, John R. Hoidal, Joachim-Volker Höltje, Koichi J. Homma, John F. Honek, Vivian Y.H. Hook, John D. Hooper, Nigel M. Hooper, Kazuo Hosoi, Christopher J. Howe, Dennis E. Hruby, James J.-D. Hseih, Chun-Chieh Hsu, Tony T. Huang, Tur-Fu Huang, Yoann Huet, Clare Hughes, Jean-Emmanuel Hugonnet, Adrienne L. Huston, Oumaïma Ibrahim-Granet, Eiji Ichishima, Yukio Ikehara, Tadashi Inagami, Jessica Ingram, R.E. Isaac, Grazia Isaya, Clara E. Isaza, Shin-ichi Ishii, Amandine Isnard, Kiyoshi Ito, Koreaki Ito, Yoshifumi Itoh, Xavier Iturrioz, Sadaaki Iwanaga, Ralph W. Jack, Mel C. Jackson, Michael N.G. James, Jiří Janata, Claire Janoir, Hanna Janska, Ken F. Jarrell, Mariusz Jaskolski, Sheila S. Jaswal, Ying Y. Jean, Dieter E. Jenne, Young Joo Jeon, Ping Jiang, John E. Johnson, Michael D. Johnson, James A. Johnston, Amanda Jones, Elizabeth W. Jones, Carine Joudiou, Luiz Juliano, Hea-Jin Jung, Ray Jupp, Todd F. Kagawa, Hubert Kalbacher, Yayoi Kamata, Shuichi Kaminogawa, Yoshiyuki Kamio, Makoto Kaneda, Sung Gyun Kang, Sung Hwan Kang, Mary Kania, Tomasz Kantyka, Nobuyuki Kanzawa, Abdulkarim Y. Karim, Takafumi Kasumi, Hiroaki Kataoka, Hardeep Kaur, Shun-Ichiro Kawabata, Mari Kawaguchi, John Kay, Murat Kaynar, Kenneth C. Keiler, R.M. Kelly, Nathaniel T. Kenton, Michael A. Kerr, Kristof Kersse, Jukka Kervinen, Benedikt M. Kessler, Efrat Kessler, Timo K. Khoronen, Simon Kidd, Marjolein Kikkert, Mogens Kilian, Do-Hyung Kim, Doyoun Kim, Eunice EunKyeong Kim, In Seop Kim, Jung-Gun Kim, Kyeong Kyu Kim, Kyung Hyun Kim, Matthew S. Kimber, Yukio Kimura, Heidrun Kirschke, Yoshiaki Kiso, Colin Kleanthous, Jürgen R. Klein, Michael Klemba, Beata Kmiec, Hideyuki Kobayashi, Hiroyuki Kodama, Gerald Koelsch, Jan Kok, P.E. Kolattukody, Fabrice A. Kolb, Harald Kolmar, Yumiko Komori, Jan Konvalinka, Brice Korkmaz, Sergey V. Kostrov, Hans-Georg Kräusslich, Gabi Krczal, Lawrence F. Kress, Magnüs Már Kristjánsson, Tomáš Kučera, Sayali S. Kukday, Hidehiko Kumagai, Sharad Kumar, Malika Kumarasiri, Takashi Kumazaki, Beate M. Kümmerer, Kouji Kuno, Markku Kurkinen, Eva Kutejová, Marie Kveiborg, Agnieszka Kwarciak, Liisa Laakkonen, Nikolaos E. Labrou, Gavin D. Laing, Gayle Lamppa, Thomas Langer, Richard A. Laursen, Richard A. Lawrenson, Matthew D. Layne, Bernard F. Le Bonniec, María C. Leal, Ronald M. Lechan, David H. Lee, Irene Lee, Jae Lee, Kye Joon Lee, Soohee Lee, Xiaobo Lei, Jonathan Leis, Ellen K. LeMosy, Thierry Lepage, Stephen H. Leppla, Adam Lesner, Ivan A.D. Lessard, Guy Lhomond, Huilin Li, Shu-Ming Li, Weiguo Li, Ta-Hsiu Liao, Robert C. Liddington, Toby Lieber, H.R. Lijnen, Christopher D. Lima, Chen-Yong Lin, Gang Lin, Ming T. Lin, Xinli Lin, Yee-Shin Lin, L.L. Lindsay, William N. Lipscomb, John W. Little, Ching-Chuan Liu, Chuan-ju Liu, Mark O. Lively, Nurit Livnat-Levanon, Per O. Ljungdahl, Catherine Llorens-Cortes, Peter Lobel, Y. Peng Loh, Jouko Lohi, G.P. Lomonossoff, Yvan Looze, Carlos López-Otin, Landys Lopez-Quezada, Alex Loukas, Long-Sheng Lu, Áke Lundwall, Liu-Ying Luo, Andrei Lupas, Dawn S. Luthe, Nicholas J. Lynch, Peter J. Lyons, Vivian L. MacKay, Jesica M. Levingston Macleod, Viktor Magdolen, Jean-Luc Mainardi, Kauko K. Mäkinen, Jeremy P. Mallari, Surya P. Manandhar, Fajga R. Mandelbaum, Anne M. Manicone, Johanna Mansfeld, Joseph Marcotrigiano, Michael Mares, Gemma Marfany, Francis S. Markland, Judith Marokházi, Hélène Marquis, Robert A. Marr, Enzo Martegani, Erik W. Martin, Manuel Martinez, L. Miguel Martins, Masato Maruyama, Masugi Maruyama, Sususmu Maruyama, Takeharu Masaki, Ramin Massoumi, Rency T. Mathew, Lynn M. Matrisian, Yoshihiro Matsuda, Osamu Matsushita, Marco Matuschek, Anna Matušková, Krisztina Matúz, Cornelia Mauch, Michael R. Maurizi, Lorenz M. Mayr, Dewey G. McCafferty, J. Ken McDonald, James H. McKerrow, David McMillan, Robert P. Mecham, Darshini P. Mehta, Chris Meisinger, Alan Mellors, Roger G. Melton, Jeffrey A. Melvin, Robert Ménard, Luis Menéndez-Arias, Milene C. Menezes, Andrew Mesecar, Stéphane Mesnage, Diane H. Meyer, Gregor Meyers, Susan Michaelis, Karolina Michalska, Wojciech P. Mielicki, Igor Mierau, Galina V. Mikoulinskaia, Charles G. Miller, Lydia K. Miller, John Mills, Kenneth V. Mills, Jinrong Min, Michel-Yves Mistou, Yoshio Misumi, Shin-ichi Miyoshi, Shigehiko Mizutani, Shahriar Mobashery, Satsuki Mochizuki, William L. Mock, Frank Möhrlen, Nathalie Moiré, Paul E. Monahan, Angela Moncada-Pazos, Véronique Monnet, Michel Monod, Cesare Montecucco, Laura Morelli, Sumiko Mori, Takashi Morita, James H. Morrissey, Richard J. Morse, John S. Mort, Uffe H. Mortensen, Rory E. Morty, Joel Moss, Hidemasa Motoshima, Jeremy C. Mottram, Ana M. Moura-da-Silva, Mary Beth Mudgett, Egbert Mundt, Kazuo Murakami, Mario Tyago Murakami, Kimiko MurakamiMurofoshi, Sawao Murao, Gillian Murphy, M.R.N. Murthy, Tatsushi Muta, Elmarie Myburgh, Nino Mzhavia, A.H.M. Nurun Nabi, Hideaki Nagase, Michael W. Nagle, Dorit K. Nägler, Rajesh R. Naik, Divya B. Nair, Toshiki Nakai, Yoshitaka Nakajima, Yukio Nakamura, Hitoshi Nakatogawa, Toru Nakayama, Natalia N. Nalivaeva, Dipankar Nandi, Maria Clara Leal Nascimento-Silva, Kim Nasmyth, Carl F. Nathan, Fernando Navarro-García, Dayane Lorena Naves, Danny D. Nedialkova, Keir C. Neuman, Jeffrey-Tri Nguyen, Ky-Anh Nguyen, Gabriela T. Niemirowicz, Toshiaki Nikai, Eiichiro Nishi, Wataru Nishii, Makoto Nishiyama, Yasuhiro Nishiyama, Masatoshi Noda, Seiji Nomura, Shigemi Norioka, Desire M.M. Nsangou, Amornrat O’Brien, Michael B. O’Connor, Kohei Oda, Irina V. Odinokova, Joyce Oetjen, Teru Ogura, Dennis E Ohman, Yoshinori Ohsumi, Mukti Ojha, Akinobu Okabe, Yasunori Okada, Keinosuke Okamoto, Kenji Okuda, Nobuaki Okumura, Takashi Okuno, Kjeld Oleson, Priscila Oliveira de Giuseppe, Martin Olivier, Yasuko Ono, Stephen Oroszlan, Nobuyuki Ota, Michael Ovadia, Jiyang O-Wang, Claus Oxvig, Jeremy C.L. Packer, Sergio Padilla-López, Mark Paetzel, Michael J. Page, Andrea Page-McCaw, Mark J.I. Paine, Byoung Chul Park, Eunyong Park, John E. Park, Pyong Woo Park, Sung Goo Park, Kirk L. Parkin, William C Parks, Thaysa Paschoalin, Annalisa Pastore, Alexander Nikolich Patananan, Sudhir Paul, Henry L. Paulson, Ulrich von Pawel-Rammingen, David A. Pearce, Mark S. Pearson, Duanqing Pei, Gunnar Pejler, Alan D. Pemberton, Jianhao Peng, Julien Pernier, Jan-Michael Peters, Thorsten Pfirrmann, Viet-Laï Pham, Iva Pichová, Darren Pickering, Christophe Piesse, David Pignol, Robert N. Pike, Lothaire Pinck, Hubert Pirkle, Henry C. Pitot, Andrew G. Plaut, Hidde Ploegh, László Polgár, Corrine Porter, Rolf Postina, Jan Potempa, Knud Poulsen, Scott D. Power, Rex. F. Pratt, Gerd Prehna, Gilles Prévost, Alexey V. Pshezhetsky, Mohammad A. Qasim, Feng Qian, Jiazhou Qiu, Víctor Quesada, Evette S. Radisky, Stephen D. Rader, Kavita Raman, Andrew J. Ramsay, Derrick E. Rancourt, Najju Ranjit, Narayanam V. Rao, Kiira Ratia, Neil D. Rawlings, Robert B. Rawson, Vijay Reddy, Colvin M. Redman, Maria Elena Regonesi, Andreas S. Reichert, Antonia P. Reichl, Han Remaut, S. James Remington, Martin Renatus, David Reverter, Eric C. Reynolds, Mohamed Rholam, Charles M. Rice, Todd W. Ridky, Howard Riezman, D.C. Rijken, Marie-Christine Rio, Alison Ritchie, Janine Robert-Baudouy, Mark W. Robinson, Michael Robinson, Adela Rodriguez-Romero, Renata Santos Rodriques, John C. Rogers, Camilo Rojas, Floyd E. Romesberg, David J. Roper, Nora Rosas-Murrieta, A.M. Rose, Philip J. Rosenthal, J. Rosing, Ornella Rossetto, Véronique Rossi, Richard A. Roth, Hanspeter Rottensteiner, Andrew D. Rowan, Mikhail Rozanov, Alexandra Rucavado, Andrea Ruecker, Françoise Rul, Till Rümenapf, Ilaria Russo, Martin D. Ryan, Elena Sacco, J. Evan Sadler, W. Saenger, Hans-Georg Sahl, Mohammed Sajid, Masayoshi Sakaguchi, Fumio Sakiyama, Maria L. Salas, Maria Cristina O. Salgado, Guy S. Salvesen, Edith Sánchez, Eladio F. Sanchez, Qing-Xiang Amy Sang, Krishnan Sankaran, Susanta K. Sarkar, Michael P. Sarras, Yoshikiyo Sasagawa, Araki Satohiko, Eric Sauvage, Loredana Saveanu, H.S. Savithri, Hitoshi Sawada, R. Gary Sawers, Isobel A. Scarisbrick, Andreas Schaller, Justin M. Scheer, Friedrich Scheiflinger, Cordelia Schiene-Fischer, Uwe Schlomann, Manfred Schlösser, Alvin H. Schmaier, Walter K. Schmidt, Anette Schneemann, Rick G. Schnellmann, Henning Scholze, Lutz Schomburg, Wilhelm J. Schwaeble, Christopher J. Scott, Rosaria Scudiero, Atsuko Sehara-Fujisawa, Nabil G. Seidah, Motoharu Seiki, Junichi Sekiguchi, Andrea Senff-Ribeiro, Ihn Sik Seong, Mihaela Serpe, Solange M.T. Serrano, Peter Setlow, Tina Shahian, M. Shanks, Feng Shao, Steven D. Shapiro, Navneet Sharma, Lindsey N. Shaw, Aimee Shen, Lei Shen, Roger F. Sherwood, Yun-Bo Shi, Hitoshi Shimoi, Yoichiro Shimura, A.D. Shirras, Viji Shridhar, Jinal K. Shukla, Ene Siigur, Jüri Siigur, Natalie C. Silmon de Monerri, Robert B. Sim, James P. Simmer, William H. Simmons, Jaspreet Singh, Alison Singleton, Tatiana D. Sirakova, Titia K. Sixma, Tim Skern, Randal A. Skidgel, Jeffrey Slack, David E. Sleat, Barbara S. Slusher, Janet L. Smith, Matthew A. Smith, Mark J. Smyth, Erik J. Snijder, Solmaz Sobhanifar, Kenneth Söderhaäll, Istvan Sohar, Peter Sonderegger, Marcos Henrique Ferreira Sorgine, Hiroyuki Sorimachi, Karen E. Soukhodolets, Tatiana de Arruda Campos Brasil de Souza, Tamás Sperka, Shiranee Sriskandan, Joseph W. St. Geme, Raymond J. St. Leger, Peter Staib, James L. Steele, Bjarki Stefansson, Christian Steinkühler, Leisa M. Stenberg, Johan Stenflo, Henning R. Stennicke, Valentin M. Stepanov, Olga A. Stepnaya, Frank Steven, Richard L. Stevens, Kenneth J. Stevenson, Mathieu St-Louis, Christopher C. Stobart, Walter Stöcker, Andrew C. Storer, Norbert Sträter, Ellen G. Strauss, James H. Strauss, Kvido Stříšovský, Natalie C.J. Strynadka, Edward D. Sturrock, Dan Su, Xiao-Dong Su, Paz Suárez-Rendueles, Traian Sulea, Venkatesh Sundararajan, Ryoji Suno, Carolyn K. Suzuki, Fumiaki Suzuki, Hideyuki Suzuki, Nobuhiro Suzuki, Stephen Swenson, Rose L. Szabady, Pal Bela Szecsi, Lászlo Szilágyi, Muhamed-Kheir Taha, Eizo Takahashi, Kenji Takahashi, Toshiro Takai, Atsushi Takeda, Soichi Takeda, Jeremy J.R.H. Tame, Tomohiro Tamura, Fulong Tan, Keiji Tanaka, Carmen Tanase, Jordan Tang, Martha M. Tanizaki, Egbert Tannich, Guido Tans, Anthony L. Tarentino, Anchalee Tassanakajon, Hiroki Tatsumi, Norbert Tautz, Erin Bassford Taylor, Pedro Filipe Teixeira, Bhanu Prakash V.L. Telugu, Markus F. Templin, Shigeyuki Terada, Uchikoba Tetsuya, C. Thacker, Maulik Thaker, Heinz-Jürgen Thiel, Nicole Thielens, Gonzales Thierry, Karine Thivierge, Mark D. Thomas, Margot Thome, Mary K. Thorsness, Peter E. Thorsness, Natalie J. Tigue, Sokol V. Todi, Birgitta Tomkinson, Fiorella Tonello, Liang Tong, H.S. Toogood, Paolo Tortora, József Tözsèr, Luiz Rodolpho Travassos, James Travis, Dilza Trevisan-Silva, Francesca Trinchella, Neil N. Trivedi, Carol M. Troy, Harald Tschesche, Yu-Lun Tseng, Masafumi Tsujimoto, Anthony T. Tu, Kathleen E. Tumelty, Boris Turk, Dusan Turk, Vito Turk, Anthony J. Turner, Tetsuya Uchikoba, Takayuki Ueno, Alejandro P. Ugalde, Veli-Jukka Uitto, Sinisa Urban, Olivier Valdenaire, Adrian Valli, Jozef Van Beeumen, Bertus Van den Burg, Renier A.L. Van der Hoorn, Jan Maarten van Dijl, Peter Van Endert, Bram J. Van Raam, Harold E. Van Wart, Tom Vanden Berghe, Peter Vandenabeele, Margo Vanoni, Silvio Sanches Veiga, William H. Velander, Gloria Velasco, Josep Vendrell, I. István Venekei, Vaclav Vetvicka, F.-Nora Vögtle, Waldemar Vollmer, Kei Wada, Fred W. Wagner, Sun Nyunt Wai, Timothy Wai, Shane Wainwright, Kenneth W. Walker, Stephen J. Walker, Jean Wallach, Linda L. Walling, Peter N. Walsh, Hai-Yan Wang, Hengbin Wang, Jianwei Wang, Peng Wang, Ping Wang, Michael Wassenegger, Kunihiko Watanabe, Helen Webb, Joseph M. Weber, Niklas Weber, Daniel R. Webster, Shuo Wei, Rodney A. Welch, James A. Wells, Herbert Wenzel, Ingrid E. Wertz, Ulla W. Wewer, Alison R. Whyteside, Sherwin Wilk, Jean-Marc Wilkin, Claudia Wilmes, Jakob R. Winther, David S. Wishart, Alexander Wlodawer, J. Fred Woessner, Michael S. Wolfe, Wilson Wong, Roger Woodgate, Gerry Wright, Jiunn-Jong Wu, Qingyu Wu, Magdalena Wysocka, Chao Xu, Zhenghong Xu, Kinnosuke Yahori, Shoji Yamada, Nozomi Yamaguchi, Shinji Yamaguchi, Yoshio Yamakawa, Hiroki Yamamoto, Ikao Yana, Maozhou Yang, Na Yang, Chenjuan Yao, Tingting Yao, Noriko Yasuda, Toshimasa Yasuhara, Shigeki Yasumasu, Edward T.H. Yeh, Irene Yiallouros, Jiang Yin, Hiroo Yonezawa, Soon Ji Yoo, Tadashi Yoshimoto, Michael W. Young, Stephen G. Young, Nousheen Zaidi, Ludmila L. Zavalova, Peter Zavodszky, Aidong Zhang, Xianming Zhang, Yi-Zheng Zhang, Dominick Zheng, Guangming Zhong, Rong Zhong, Yuan Zhou, Zhaohui Sunny Zhou, Michael Zick, Paola Zigrino, and Andrei A. Zimin
- Published
- 2013
26. Fluorescence lifetime assays: current advances and applications in drug discovery
- Author
-
Ulrich Hassiepen, Julian Woelcke, Klaus Doering, and Stephan Pritz
- Subjects
Enzyme inhibition ,Homogeneous ,Drug discovery ,Expert opinion ,Drug Discovery ,Compound profiling ,Signal modulation ,Nanotechnology ,Biology ,Fluorescence - Abstract
Fluorescence lifetime assays complement the portfolio of established assay formats available in drug discovery, particularly with the recent advances in microplate readers and the commercial availability of novel fluorescent labels. Fluorescence lifetime assists in lowering complexity of compound screening assays, affording a modular, toolbox-like approach to assay development and yielding robust homogeneous assays.To date, materials and procedures have been reported for biochemical assays on proteases, as well as on protein kinases and phosphatases. This article gives an overview of two assay families, distinguished by the origin of the fluorescence signal modulation.The pharmaceutical industry demands techniques with a robust, integrated compound profiling process and short turnaround times. Fluorescence lifetime assays have already helped the drug discovery field, in this sense, by enhancing productivity during the hit-to-lead and lead optimization phases. Future work will focus on covering other biochemical molecular modifications by investigating the detailed photo-physical mechanisms underlying the fluorescence signal.
- Published
- 2012
27. A macrocyclic HCV NS3/4A protease inhibitor interacts with protease and helicase residues in the complex with its full-length target
- Author
-
Gaby Monnet, Ulrich Hassiepen, Dmitri I. Svergun, Frederic Villard, Nikolaus Schiering, Jörg Eder, Allan D'Arcy, Marion Kamke, Ursula Bodendorf, Prakash Raman, Oliver Simic, and Ruth Pulfer
- Subjects
Models, Molecular ,Protein Conformation ,Hepatitis C virus ,medicine.medical_treatment ,viruses ,Biology ,Viral Nonstructural Proteins ,medicine.disease_cause ,Crystallography, X-Ray ,Inhibitory Concentration 50 ,Scattering, Small Angle ,medicine ,Escherichia coli ,Protease inhibitor (pharmacology) ,Protease Inhibitors ,Serine protease ,NS3 ,Multidisciplinary ,Protease ,Intracellular Signaling Peptides and Proteins ,virus diseases ,Biological Sciences ,Virology ,Hepatitis C ,digestive system diseases ,NS2-3 protease ,Biochemistry ,Viral replication ,biology.protein ,Chromatography, Gel ,Carrier Proteins ,Crystallization ,MASP1 - Abstract
Hepatitis C virus (HCV) infection is a global health burden with over 170 million people infected worldwide. In a significant portion of patients chronic hepatitis C infection leads to serious liver diseases, including fibrosis, cirrhosis, and hepatocellular carcinoma. The HCV NS3 protein is essential for viral polyprotein processing and RNA replication and hence viral replication. It is composed of an N-terminal serine protease domain and a C-terminal helicase/NTPase domain. For full activity, the protease requires the NS4A protein as a cofactor. HCV NS3/4A protease is a prime target for developing direct-acting antiviral agents. First-generation NS3/4A protease inhibitors have recently been introduced into clinical practice, markedly changing HCV treatment options. To date, crystal structures of HCV NS3/4A protease inhibitors have only been reported in complex with the protease domain alone. Here, we present a unique structure of an inhibitor bound to the full-length, bifunctional protease-helicase NS3/4A and show that parts of the P4 capping and P2 moieties of the inhibitor interact with both protease and helicase residues. The structure sheds light on inhibitor binding to the more physiologically relevant form of the enzyme and supports exploring inhibitor-helicase interactions in the design of the next generation of HCV NS3/4A protease inhibitors. In addition, small angle X-ray scattering confirmed the observed protease-helicase domain assembly in solution.
- Published
- 2011
28. Novel heterocyclic DPP-4 inhibitors for the treatment of type 2 diabetes
- Author
-
Christopher Higgs, Wiesia Maniara, Neil Victor Harris, Sussie L. Krintel, Kenji Namoto, Alokesh Duttaroy, Finton Sirockin, Christopher A. Hurley, Nils Ostermann, Daniel K. Baeschlin, Robert Edward Mackenzie, Eric Gangl, David E. Clark, Garry Fenton, Jörg Trappe, Amanda Fillmore, Ulrich Hassiepen, Stephen J. Dunsdon, Jon Sutton, Richard Sedrani, and Bernd Gerhartz
- Subjects
Male ,Models, Molecular ,animal structures ,Clinical Biochemistry ,Pharmaceutical Science ,Type 2 diabetes ,Pharmacology ,Crystallography, X-Ray ,Biochemistry ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Heterocyclic Compounds ,Drug Discovery ,Hydrolase ,medicine ,Animals ,Humans ,Molecular Biology ,IC50 ,Dipeptidyl-Peptidase IV Inhibitors ,Molecular Structure ,Chemistry ,DPP-4 Inhibitors ,Organic Chemistry ,medicine.disease ,Rats ,Enzyme Activation ,Diabetes Mellitus, Type 2 ,Molecular Medicine ,Caco-2 Cells - Abstract
Novel deazaxanthine-based DPP-4 inhibitors have been identified that are potent (IC(50)10nM) and highly selective versus other dipeptidyl peptidases. Their synthesis and SAR are reported, along with initial efforts to improve the PK profile through decoration of the deazaxanthine core. Optimisation of compound 3a resulted in the identification of compound (S)-4i, which displayed an improved in vitro and ADME profile. Further enhancements to the PK profile were possible by changing from the deazahypoxanthine to the deazaxanthine template, culminating in compound 12g, which displayed good ex vivo DPP-4 inhibition and a superior PK profile in rat, suggestive of once daily dosing in man.
- Published
- 2011
29. Fluorescence- Based Biochemical Protease Assay Formats
- Author
-
Julian Woelcke and Ulrich Hassiepen
- Subjects
Protease ,Chemistry ,medicine.medical_treatment ,medicine ,Fluorescence ,Molecular biology - Published
- 2009
30. The crystal structure of caspase-6, a selective effector of axonal degeneration
- Author
-
Renato Baumgartner, Allan D'Arcy, Richard Morse, Christophe Briand, Arnaud Decock, Gabriele Meder, Ulrich Hassiepen, and Martin Renatus
- Subjects
biology ,Caspase 6 ,Chemistry ,Effector ,Neurodegenerative Diseases ,Cell Biology ,Biochemistry ,Axons ,Protein Structure, Secondary ,Cell biology ,Structure-Activity Relationship ,Protein structure ,Drug development ,Hydrolase ,biology.protein ,Structure–activity relationship ,Humans ,Protein Multimerization ,Protein Structure, Quaternary ,Molecular Biology ,Linker ,Caspase - Abstract
Neurodegenerative diseases pose one of the most pressing unmet medical needs today. It has long been recognized that caspase-6 may play a role in several neurodegenerative diseases for which there are currently no disease-modifying therapies. Thus it is a potential target for neurodegenerative drug development. In the present study we report on the biochemistry and structure of caspase-6. As an effector caspase, caspase-6 is a constitutive dimer independent of the maturation state of the enzyme. The ligand-free structure shows caspase-6 in a partially mature but latent conformation. The cleaved inter-domain linker remains partially inserted in the central groove of the dimer, as observed in other caspases. However, in contrast with the structures of other caspases, not only is the catalytic machinery misaligned, but several structural elements required for substrate recognition are missing. Most importantly, residues forming a short anti-parallel β-sheet abutting the substrate in other caspase structures are part of an elongation of the central α-helix. Despite the dramatic structural changes that are required to adopt a canonical catalytically competent conformation, the pre-steady-state kinetics exhibit no lag phase in substrate turnover. This suggests that the observed conformation does not play a regulatory role in caspase-6 activity. However, targeting the latent conformation in search for specific and bio-available caspase-6 inhibitors might offer an alternative to active-site-directed approaches.
- Published
- 2009
31. Characterization of the catalytic activity of the membrane-anchored metalloproteinase ADAM15 in cell-based assays
- Author
-
Thorsten Maretzky, Ulrich Hassiepen, Susanne Worpenberg, Christopher M. Overall, Joerg Eder, Carl P. Blobel, Ouathek Ouerfelli, and Guangli Yang
- Subjects
ADAM15 ,Matrix metalloproteinase ,Biochemistry ,GM6001 ,Catalysis ,Cell Line ,Substrate Specificity ,chemistry.chemical_compound ,Mice ,Peptide Library ,Disintegrin ,Animals ,Receptor, Fibroblast Growth Factor, Type 2 ,Peptide library ,Molecular Biology ,Metalloproteinase ,biology ,Membrane Proteins ,Cell Biology ,ADAM Proteins ,Ectodomain ,chemistry ,Cell culture ,Mutation ,biology.protein - Abstract
ADAM15 (a disintegrin and metalloproteinase 15) is a membrane-anchored metalloproteinase, which is overexpressed in several human cancers and has been implicated in pathological neovascularization and prostate cancer metastasis. Yet, little is known about the catalytic properties of ADAM15. Here, we purified soluble recombinant ADAM15 to test for its ability to cleave a library of peptide substrates. However, we found no processing of any of the peptide substrates tested here, and therefore turned to cell-based assays to characterize the catalytic properties of ADAM15. Overexpression of full-length membrane-anchored ADAM15 or the catalytically inactive ADAM15E→A together with various membrane proteins resulted in increased release of the extracellular domain of the fibroblast growth factor receptor 2iiib (FGFR2iiib) by ADAM15, but not ADAM15E→A. This provided a robust assay for a characterization of the catalytic properties of ADAM15 in intact cells. We found that increased expression of ADAM15 resulted in increased FGFR2iiib shedding, but that ADAM15 was not stimulated by phorbol esters or calcium ionophores, two commonly used activators of ectodomain shedding. Moreover, ADAM15-dependent processing of FGFR2iiib was inhibited by the hydroxamate-based metalloproteinase inhibitors marimastat, TAPI-2 and GM6001, and by 50 nM TIMP-3 (tissue inhibitor of metalloproteinases 3), but not by 100 nM TIMP-1, and only weakly by 100 nM TIMP-2. These results define key catalytic properties of ADAM15 in cells and its response to stimulators and inhibitors of ectodomain shedding. A cell-based assay for the catalytic activity of ADAM15 could aid in identifying compounds, which could be used to block the function of ADAM15 in pathological neovascularization and cancer.
- Published
- 2009
32. A fluorescence lifetime-based assay for protease inhibitor profiling on human kallikrein 7
- Author
-
Lorenz M. Mayr, Manuela Hinnenberger, Gabriele Meder, Ulrich Hassiepen, Klaus Doering, and Julian Woelcke
- Subjects
chemistry.chemical_classification ,Fluorophore ,Time Factors ,High-throughput screening ,Molecular Sequence Data ,Tryptophan ,Peptide ,Fluorescence ,Amino acid ,Substrate Specificity ,Acridone ,chemistry.chemical_compound ,Scissile bond ,chemistry ,Biochemistry ,Luminescent Measurements ,Biophysics ,Humans ,Kallikreins ,Protease Inhibitors ,Amino Acid Sequence - Abstract
Fluorescence lifetime is an intrinsic parameter describing the fluorescence process. Changes in the fluorophore's physicochemical environment can lead to changes in the fluorescence lifetime. When used as the readout in biological assays, it is thought to deliver superior results to conventional optical readouts. Hence it has the potential to replace readout technologies currently established in drug discovery such as absorption, luminescence or fluorescence intensity. Here we report the development of an activity assay for human kallikrein 7, a serine protease involved in skin diseases. As a probe, we have selected a blue-fluorescent acridone dye, featuring a remarkably long lifetime that can be quenched by either of the 2 natural amino acids, tyrosine and tryptophan. Incorporating this probe and 1 of the quenching amino acids on either side of the scissile bond of the substrate peptide enables us to monitor the enzymatic activity by quantifying the increase in the fluorescence lifetime signal. A systematic investigation of substrate structures has led to a homogenous, microplate-based, compound profiling assay that yields inhibitory constants down into the single-digit nanomolar range. This type of assay has now been added to our standard portfolio of screening techniques, and is routinely used for compound profiling. ( Journal of Biomolecular Screening 2009:1-9)
- Published
- 2009
33. Multi-photon excitation of intrinsic protein fluorescence and its application to pharmaceutical drug screening
- Author
-
Alain Schilb, Kurt Mueller, Peter T. C. So, Kurt A. Stoeckli, Manfred Auer, Christof Buehler, Ulrich Hassiepen, and Joerg Dreessen
- Subjects
Drug Evaluation, Preclinical ,Nanotechnology ,medicine.disease_cause ,Light scattering ,symbols.namesake ,Drug Discovery ,Protein Interaction Mapping ,medicine ,Rayleigh scattering ,Spectroscopy ,Pharmacology ,Microscopy, Confocal ,Chemistry ,Proteins ,Equipment Design ,Fluorescence ,Photobleaching ,Equipment Failure Analysis ,Microscopy, Fluorescence, Multiphoton ,Spectrometry, Fluorescence ,Drug Design ,Femtosecond ,Biophysics ,symbols ,Molecular Medicine ,Computer-Aided Design ,Feasibility Studies ,Ultraviolet ,Excitation - Abstract
The majority of proteins contain intrinsic fluorophores as natural sensors of molecular structures, dynamics, and interactions. The intrinsic protein fluorescence signal allows for the label-free and, hence, undisturbed and rapid study of protein-ligand interactions. Ultraviolet-based drug screening is hampered by the background, photobleaching, light scattering, inner filter effects, and interfering assay compounds. Such problems can be overcome by means of molecular three-photon excitation (3PE) with infrared femtosecond light pulses since longer excitation wavelengths result in less Raleigh scattering, and the subfemtoliter (confocal-like) 3PE volume minimizes out-of-focus photobleaching, background generation, and inner filter effects. We demonstrate the general feasibility of 3PE for protein spectroscopy and illustrate the technique's excellent potential for high-throughput screening. By using the intrinsic fluorescence intensity of a protein-substrate, we were able to discriminate between ligands of different affinities in binding assays.
- Published
- 2005
34. Characterization and binding specificity of the monomeric STAT3-SH2 domain
- Author
-
Ulrike Hemmann, Serge Haan, Peter C. Heinrich, Joachim Grötzinger, Ulrich Hassiepen, Fred Schaper, Jens Schneider-Mergener, and Axel Wollmer
- Subjects
STAT3 Transcription Factor ,Conformational change ,Protein Folding ,Protein Conformation ,Enzyme-Linked Immunosorbent Assay ,SH2 domain ,Biochemistry ,src Homology Domains ,chemistry.chemical_compound ,Protein structure ,Escherichia coli ,Tyrosine ,Phosphotyrosine ,Molecular Biology ,Binding Sites ,biology ,Circular Dichroism ,Tyrosine phosphorylation ,Cell Biology ,DNA-Binding Proteins ,Spectrometry, Fluorescence ,chemistry ,biology.protein ,Trans-Activators ,GRB2 ,Phosphotyrosine-binding domain ,Binding domain - Abstract
Signal transducers and activators of transcription (STATs) are important mediators of cytokine signal transduction. STAT factors are recruited to phosphotyrosine-containing motifs of activated receptor chains via their SH2 domains. The subsequent tyrosine phosphorylation of the STATs leads to their dissociation from the receptor, dimerization, and translocation to the nucleus. Here we describe the expression, purification, and refolding of the STAT3-SH2 domain. Proper folding of the isolated protein was proven by circular dichroism and fluorescence spectroscopy. The STAT3-SH2 domain undergoes a conformational change upon dimerization. Using an enzyme-linked immunosorbent assay we demonstrate that the monomeric domain binds to specific phosphotyrosine peptides. The specificity of binding to phosphotyrosine peptides was assayed with the tyrosine motif encompassing Tyr705 of STAT3 and with all tyrosine motifs present in the cytoplasmic tail of the signal transducer gp130.
- Published
- 1999
35. The signal transducer gp130--bacterial expression, refolding and properties of the carboxy-terminal domain of the cytokine-binding module
- Author
-
Peter C. Heinrich, Gerhard Müller-Newen, Stefan Pflanz, Joachim Grötzinger, Jürgen Stahl, Axel Wollmer, and Ulrich Hassiepen
- Subjects
Circular dichroism ,Protein Folding ,Binding Sites ,Magnetic Resonance Spectroscopy ,Chemistry ,Interleukin-6 ,Circular Dichroism ,Protein domain ,Glycoprotein 130 ,Biochemistry ,Transmembrane protein ,Crystallography ,Spectrometry, Fluorescence ,EVH1 domain ,Cyclic nucleotide-binding domain ,Biophysics ,Escherichia coli ,Cytokines ,Humans ,B3 domain ,Cytokine binding ,Receptors, Cytokine ,Signal Transduction - Abstract
Gp130 is the signal transducing receptor subunit of the so-called interleukin-6-type cytokines. This transmembrane protein is a member of the cytokine-receptor superfamily predicted to consist of six fibronectin-type-III-like domains in its extracellular part. The second and the third domain constitute the so-called cytokine-binding module. Domain 2 is characterized by a set of four conserved Cys residues, domain 3 by a conserved WSXWS motif. As a first approach to a more detailed characterization of the cytokine-binding domains of human gp130, we have expressed in Escherichia coli two forms of domain 3 differing in length. Both proteins were purified and refolded in a single step applying size-exclusion chromatography. According to the rotational correlation times deduced from fluorescence anisotropy decay, they do not form aggregates. CD and fluorescence spectroscopy were used to study thermal unfolding and denaturation by guanidinium hydrochloride. It was shown that N- and C-terminal extension by residues of the adjacent hinge regions substantially increase the thermal stability of the domain, which is conceivable from a molecular model. These results are the basis for further structural investigation by NMR spectroscopy.
- Published
- 1997
36. Recombinant human O6-alkylguanine-DNA alkyltransferase (AGT), Cys145-alkylated AGT and Cys145 --> Met145 mutant AGT: comparison by isoelectric focusing, CD and time-resolved fluorescence spectroscopy
- Author
-
Ulrich Hassiepen, Kirsten Bender, Manfred F. Rajewsky, Matthias Federwisch, Axel Wollmer, and Manfred Dewor
- Subjects
Conformational change ,Time Factors ,Protein Conformation ,Mutant ,Biochemistry ,law.invention ,O(6)-Methylguanine-DNA Methyltransferase ,Protein structure ,Methionine ,law ,parasitic diseases ,Humans ,Point Mutation ,Amino Acid Sequence ,Cysteine ,Molecular Biology ,DNA Primers ,Isoelectric focusing ,Chemistry ,Circular Dichroism ,Mutagenesis ,Tryptophan ,Cell Biology ,Methyltransferases ,Recombinant Proteins ,Kinetics ,Spectrometry, Fluorescence ,Recombinant DNA ,Mutagenesis, Site-Directed ,Isoelectric Focusing ,Alkyltransferase ,Research Article - Abstract
Isoelectric focusing, CD, steady-state and time-resolved fluorescence spectroscopy were used to compare the native recombinant human DNA-repair protein O6-alkylguanine-DNA alkyltransferase (AGT) with AGT derivatives methylated or benzylated on Cys145 or modified by site-directed mutagenesis at the active centre (Met145 mutant). The AGT protein is approximately spherical with highly constrained Trp residues, but is not stabilized by disulphide bridges. In contrast with native AGT, alkylated AGT precipitated at 25 degrees C but remained monomeric at 4 degrees C. As revealed by isoelectric focusing, pI changed from 8.2 (AGT) to 8. 4 (Cys145-methylated AGT) and 8.6 (Cys145-benzylated AGT). The alpha-helical content of the Met145 mutant was decreased by approx. 5% and Trp residues were partially liberated. Although non-covalent binding of O6-benzylguanine did not alter the secondary structure of AGT, its alpha-helical content was increased by approx. 2% on methylation and by approx. 4% on benzylation, altogether indicating a small conformational change in AGT on undergoing alkylation. No signal sequences have been found in AGT that mark it for polyubiquitination. Therefore the signal for AGT degradation remains to be discovered.
- Published
- 1997
37. Erratum to 'Novel heterocyclic DPP-4 inhibitors for the treatment of type 2 diabetes' [Bioorg. Med. Chem. Lett. 22 (2012) 1464–1468]
- Author
-
Christopher A. Hurley, Robert Edward Mackenzie, Christopher Higgs, David E. Clark, Richard Sedrani, Nils Ostermann, Stephen J. Dunsdon, Sussie L. Krintel, Bernd Gerhartz, Neil Victor Harris, Daniel K. Baeschlin, Jon Sutton, Jörg Trappe, Wiesia Maniara, Kenji Namoto, Finton Sirockin, Eric Gangl, Garry Fenton, Amanda Fillmore, Ulrich Hassiepen, and Alokesh Duttaroy
- Subjects
Chemistry ,DPP-4 Inhibitors ,Organic Chemistry ,Clinical Biochemistry ,Drug Discovery ,medicine ,Pharmaceutical Science ,Molecular Medicine ,Type 2 diabetes ,Pharmacology ,medicine.disease ,Molecular Biology ,Biochemistry - Published
- 2012
38. Multi-Photon Excitation of Intrinsic Protein Fluorescence and Its Application to Pharmaceutical Drug Screening.
- Author
-
Christof Buehler, Joerg Dreessen, Kurt Mueller, Peter T.C. So, Alain Schilb, Ulrich Hassiepen, Kurt A. Stoeckli, and Manfred Auer
- Published
- 2005
39. Structural Basis of Ubiquitin Recognition by the Deubiquitinating Protease USP2
- Author
-
Ulf Eidhoff, Susanne Worpenberg, Benoit Pierrat, Ralph Riedl, Martin Renatus, M. Kroemer, Ulrich Hassiepen, Allan D'Arcy, Shirley Gil Parrado, Bernd Gerhartz, and D. Vinzenz
- Subjects
Models, Molecular ,Ubiquitin-Specific Proteases ,Molecular Sequence Data ,Ubiquitin-conjugating enzyme ,Article ,Deubiquitinating enzyme ,Ubiquitin ,Structural Biology ,Endopeptidases ,Humans ,Amino Acid Sequence ,Molecular Biology ,DNA Primers ,biology ,Water ,Cysteine protease ,Protein ubiquitination ,Protein Structure, Tertiary ,Ubiquitin ligase ,Kinetics ,Proteasome ,Biochemistry ,Metals ,biology.protein ,Crystallization ,Ubiquitin Thiolesterase - Abstract
Summary Deubiquitinating proteases reverse protein ubiquitination and rescue their target proteins from destruction by the proteasome. USP2, a cysteine protease and a member of the ubiquitin specific protease family, is overexpressed in prostate cancer and stabilizes fatty acid synthase, which has been associated with the malignancy of some aggressive prostate cancers. Here, we report the structure of the human USP2 catalytic domain in complex with ubiquitin. Ubiquitin uses two major sites for the interaction with the protease. Both sites are required simultaneously, as shown by USP2 inhibition assays with peptides and ubiquitin mutants. In addition, a layer of ordered water molecules mediates key interactions between ubiquitin and USP2. As several of those molecules are found at identical positions in the previously solved USP7/ubiquitin-aldehyde complex structure, we suggest a general mechanism of water-mediated ubiquitin recognition by USPs.
- Full Text
- View/download PDF
40. Recombinant human O6-alkylguanine-DNA alkyltransferase induces conformational change in bound DNA
- Author
-
Manfred F. Rajewsky, Ulrich Hassiepen, Axel Wollmer, Kirsten Bender, and Matthias Federwisch
- Subjects
Conformational change ,Circular dichroism ,Protein Conformation ,Molecular Sequence Data ,Biophysics ,In Vitro Techniques ,Biochemistry ,law.invention ,O6-Alkylguanine-DNA alkyltransferase ,chemistry.chemical_compound ,O(6)-Methylguanine-DNA Methyltransferase ,Protein structure ,Structural Biology ,law ,parasitic diseases ,Genetics ,Humans ,Protein–DNA interaction ,Amino Acid Sequence ,Molecular Biology ,Protein-DNA interaction ,O-6-methylguanine-DNA methyltransferase ,Time-resolved fluorescence ,Cell Biology ,DNA ,Methyltransferases ,Recombinant Proteins ,Spectrometry, Fluorescence ,chemistry ,Recombinant DNA ,Nucleic Acid Conformation ,Alkyltransferase - Abstract
Circular dichroism, and steady-state and time-resolved fluorescence spectroscopy were used to compare the native recombinant human DNA repair protein O6-alkylguanine-DNA alkyltransferase (AGT) with AGT bound to ds-DNA. Contrary to fluorescence, analysis of the far-UV CD spectra indicated a conformational change of AGT upon binding to DNA: its alpha-helical content is increased by approximately 12%. Analysis of near-UV CD spectra revealed that DNA was also affected, probably being separated into single strands locally.
- Full Text
- View/download PDF
41. Towards sensitive, high-throughput, biomolecular assays based on fluorescence lifetime.
- Author
-
Anastasia Ioanna Skilitsi, Timothé Turko, Damien Cianfarani, Sophie Barre, Wilfried Uhring, Ulrich Hassiepen, and Jérémie Léonard
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.