139 results on '"Piet Gros"'
Search Results
2. A method for intuitively extracting macromolecular dynamics from structural disorder
- Author
-
Nicholas M. Pearce and Piet Gros
- Subjects
Science - Abstract
Here, the authors present a hierarchical disorder model for the analysis of disorder in both crystal and cryo-EM structures. They apply their approach to several structures of three proteins, including SARS-CoV-2 proteins, and discuss mechanistic and dynamical implications.
- Published
- 2021
- Full Text
- View/download PDF
3. Crystal structure of the second extracellular domain of human tetraspanin CD9: twinning and diffuse scattering
- Author
-
Viviana Neviani, Martin Lutz, Wout Oosterheert, Piet Gros, and Loes Kroon-Batenburg
- Subjects
twinning ,diffuse scattering ,tetraspanin cd9ec2 ,raw data ,Crystallography ,QD901-999 - Abstract
Remarkable features are reported in the diffraction pattern produced by a crystal of the second extracellular domain of tetraspanin CD9 (deemed CD9EC2), the structure of which has been described previously [Oosterheert et al. (2020), Life Sci. Alliance, 3, e202000883]. CD9EC2 crystallized in space group P1 and was twinned. Two types of diffuse streaks are observed. The stronger diffuse streaks are related to the twinning and occur in the direction perpendicular to the twinning interface. It is concluded that the twin domains scatter coherently as both Bragg reflections and diffuse streaks are seen. The weaker streaks along c* are unrelated to the twinning but are caused by intermittent layers of non-crystallographic symmetry related molecules. It is envisaged that the raw diffraction images could be very useful for methods developers trying to remove the diffuse scattering to extract accurate Bragg intensities or using it to model the effect of packing disorder on the molecular structure.
- Published
- 2022
- Full Text
- View/download PDF
4. Anti-LRP5/6 VHHs promote differentiation of Wnt-hypersensitive intestinal stem cells
- Author
-
Nicola Fenderico, Revina C. van Scherpenzeel, Michael Goldflam, Davide Proverbio, Ingrid Jordens, Tomica Kralj, Sarah Stryeck, Tarek Z. Bass, Guy Hermans, Christopher Ullman, Teodor Aastrup, Piet Gros, and Madelon M. Maurice
- Subjects
Science - Abstract
Enhanced Wnt receptor activity is a major cause of cancer development. Here the authors identify camelid single-domain antibody fragments (VHHs) that bind to the Wnt receptor LRP5/6 ectodomain, determine the crystal structures and show that these VHHs selectively inhibit Wnt3- mediated cellular responses and block the growth of mutant Wnt-hypersensitive intestinal tumor organoids.
- Published
- 2019
- Full Text
- View/download PDF
5. Cryo-EM structures of human STEAP4 reveal mechanism of iron(III) reduction
- Author
-
Wout Oosterheert, Laura S. van Bezouwen, Remco N. P. Rodenburg, Joke Granneman, Friedrich Förster, Andrea Mattevi, and Piet Gros
- Subjects
Science - Abstract
Enzymes of the six-transmembrane epithelial antigen of the prostate (STEAP) family reduce Fe3+ and Cu2+ ions to facilitate metal-ion uptake by mammalian cells. Here, authors employ single-particle cryo-EM to gain insights into the molecular principles of iron reduction by human STEAP4 .
- Published
- 2018
- Full Text
- View/download PDF
6. Stochastic palmitoylation of accessible cysteines in membrane proteins revealed by native mass spectrometry
- Author
-
Remco N. P. Rodenburg, Joost Snijder, Michiel van de Waterbeemd, Arie Schouten, Joke Granneman, Albert J. R. Heck, and Piet Gros
- Subjects
Science - Abstract
Cysteine palmitoylation affects the localization and function of membrane proteins, but its stoichiometry and specificity are not well understood. Here, the authors show that palmitoylation is a stochastic process that depends on the accessibility of cysteines rather than a defined substrate motif.
- Published
- 2017
- Full Text
- View/download PDF
7. Insights Into Enhanced Complement Activation by Structures of Properdin and Its Complex With the C-Terminal Domain of C3b
- Author
-
Ramon M. van den Bos, Nicholas M. Pearce, Joke Granneman, T. Harma C. Brondijk, and Piet Gros
- Subjects
complement system ,alternative pathway ,properdin ,convertase ,C3b ,factor B ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Properdin enhances complement-mediated opsonization of targeted cells and particles for immune clearance. Properdin occurs as dimers, trimers and tetramers in human plasma, which recognize C3b-deposited surfaces, promote formation, and prolong the lifetime of C3bBb-enzyme complexes that convert C3 into C3b, thereby enhancing the complement-amplification loop. Here, we report crystal structures of monomerized properdin, which was produced by co-expression of separate N- and C-terminal constructs that yielded monomer-sized properdin complexes that stabilized C3bBb. Consistent with previous low-resolution X-ray and EM data, the crystal structures revealed ring-shaped arrangements that are formed by interactions between thrombospondin type-I repeat (TSR) domains 4 and 6 of one protomer interacting with the N-terminal domain (which adopts a short transforming-growth factor B binding protein-like fold) and domain TSR1 of a second protomer, respectively. Next, a structure of monomerized properdin in complex with the C-terminal domain of C3b showed that properdin-domain TSR5 binds along the C-terminal α-helix of C3b, while two loops, one from domain TSR5 and one from TSR6, extend and fold around the C3b C-terminus like stirrups. This suggests a mechanistic model in which these TSR5 and TSR6 “stirrups” bridge interactions between C3b and factor B or its fragment Bb, and thereby enhance formation of C3bB pro-convertases and stabilize C3bBb convertases. In addition, properdin TSR6 would sterically block binding of the protease factor I to C3b, thus limiting C3b proteolytic degradation. The presence of a valine instead of a third tryptophan in the canonical Trp-ladder of TSR domains in TSR4 allows a remarkable ca. 60°-domain bending motion of TSR4. Together with variable positioning of TSR2 and, putatively, TSR3, this explains the conformational flexibility required for properdin to form dimers, trimers, and tetramers. In conclusion, the results indicate that binding avidity of oligomeric properdin is needed to distinguish surface-deposited C3b molecules from soluble C3b or C3 and suggest that properdin-mediated interactions bridging C3b-B and C3b-Bb enhance affinity, thus promoting convertase formation and stabilization. These mechanisms explain the enhancement of complement-mediated opsonization of targeted cells and particle for immune clearance.
- Published
- 2019
- Full Text
- View/download PDF
8. Heterogeneous MAC Initiator and Pore Structures in a Lipid Bilayer by Phase-Plate Cryo-electron Tomography
- Author
-
Thomas H. Sharp, Abraham J. Koster, and Piet Gros
- Subjects
phase plate ,membrane attack complex ,complement ,electron tomography ,subtomogram average ,membrane pore ,Biology (General) ,QH301-705.5 - Abstract
Pore formation in membranes is important for mammalian immune defense against invading bacteria. Induced by complement activation, the membrane attack complex (MAC) forms through sequential binding and membrane insertion of C5b6, C7, C8, and C9. Using cryo-electron tomography with a Volta phase plate and subtomogram averaging, we imaged C5b-7, C5b-8, and C5b-9 complexes and determined the C5b-9 pore structure in lipid bilayers. The in situ C5b-9 pore structure at 2.3-nm resolution reveals a 10- to 11.5-nm cone-shaped pore starting with C5b678 and multiple copies of C9 that is poorly closed, yielding a seam between C9 and C6 substituting for the shorter β strands in C6 and C7. However, large variations of composite pore complexes are apparent in subtomograms. Oligomerized initiator complexes C5b-7 and C5b-8 show stages of membrane binding, deformation, and perforation that yield ∼3.5-nm-wide pores. These data indicate a dynamic process of pore formation that likely adapts to biological membranes under attack.
- Published
- 2016
- Full Text
- View/download PDF
9. Analysis of C3 Gene Variants in Patients With Idiopathic Recurrent Spontaneous Pregnancy Loss
- Author
-
Frida C. Mohlin, Piet Gros, Eric Mercier, Jean-Christophe Raymond Gris, and Anna M. Blom
- Subjects
reproductive immunology ,miscarriage ,complement system ,C3 ,mutation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Miscarriage is the most common complication of pregnancy. Approximately 1% of couples trying to conceive will experience recurrent miscarriages, defined as three or more consecutive pregnancy losses and many of these cases remain idiopathic. Complement is implicated both in the physiology and pathology of pregnancy. Therefore, we hypothesized that alterations in the C3 gene could potentially predispose to this disorder. We performed full Sanger sequencing of all exons of C3, in 192 childless women, with at least two miscarriages and without any known risk factors. All exons carrying non-synonymous alterations found in the patients were then sequenced in a control group of 192 women. None of the identified alterations were significantly associated with the disorder. Thirteen identified non-synonymous alterations (R102G, K155Q, L302P, P314L, Y325H, V326A, S327P, V330I, K633R, R735W, R1591G, G1606D, and S1619R) were expressed recombinantly, upon which C3 expression and secretion were determined. The L302P and S327P were not secreted from the cells, likely due to misfolding and intracellular degradation. Y325H, V326A, V3301I, R1591G, and G1606D yielded approximately half C3 concentration in the cell media compared with wild type (WT). We analyzed the hemolytic activity of the secreted C3 variants by reconstituting C3-depleted serum. In this assay, R1591G had impaired hemolytic activity while majority of remaining mutants instead had increased activity. R1591G also yielded more factor B activation in solution compared with WT. R1591G and G1606D showed impaired degradation by factor I, irrespectively if factor H, CD46, or C4b-binding protein were used as cofactors. These two C3 mutants showed impaired binding of the cofactors and/or factor I. Taken together, several alterations in C3 were identified and some of these affected the secretion and/or the function of the protein, which might contribute to the disorder but the degree of association must be evaluated in larger cohorts.
- Published
- 2018
- Full Text
- View/download PDF
10. Structure of Stem Cell Growth Factor R-spondin 1 in Complex with the Ectodomain of Its Receptor LGR5
- Author
-
Weng Chuan Peng, Wim de Lau, Federico Forneris, Joke C.M. Granneman, Meritxell Huch, Hans Clevers, and Piet Gros
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Leucine-rich repeat-containing G protein-coupled receptors 4–6 (LGR4–LGR6) are receptors for R-spondins, potent Wnt agonists that exert profound trophic effects on Wnt-driven stem cells compartments. We present crystal structures of a signaling-competent fragment of R-spondin 1 (Rspo1) at a resolution of 2.0 Å and its complex with the LGR5 ectodomain at a resolution of 3.2 Å. Ecto-LGR5 binds Rspo1 at its concave leucine-rich-repeat (LRR) surface, forming a dimeric 2:2 complex. Fully conserved residues on LGR4–LGR6 explain promiscuous binding of R-spondins. A phenylalanine clamp formed by Rspo1 Phe106 and Phe110 pinches Ala190 of LGR5 and is critical for binding. Mutations related to congenital anonychia reduce signaling, but not binding of Rspo1 to LGR5. Furthermore, antibody binding to the extended loop of the C-terminal LRR cap of LGR5 activates signaling in a ligand-independent manner. Thus, our data reveal binding of R-spondins to conserved sites on LGR4–LGR6 and, in analogy to FSHR and related receptors, suggest a direct signaling role for LGR4–LGR6 in addition to its formation of Wnt receptor and coreceptor complexes.
- Published
- 2013
- Full Text
- View/download PDF
11. Assembly and Regulation of the Membrane Attack Complex Based on Structures of C5b6 and sC5b9
- Author
-
Michael A. Hadders, Doryen Bubeck, Pietro Roversi, Svetlana Hakobyan, Federico Forneris, B. Paul Morgan, Michael K. Pangburn, Oscar Llorca, Susan M. Lea, and Piet Gros
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Activation of the complement system results in formation of membrane attack complexes (MACs), pores that disrupt lipid bilayers and lyse bacteria and other pathogens. Here, we present the crystal structure of the first assembly intermediate, C5b6, together with a cryo-electron microscopy reconstruction of a soluble, regulated form of the pore, sC5b9. Cleavage of C5 to C5b results in marked conformational changes, distinct from those observed in the homologous C3-to-C3b transition. C6 captures this conformation, which is preserved in the larger sC5b9 assembly. Together with antibody labeling, these structures reveal that complement components associate through sideways alignment of the central MAC-perforin (MACPF) domains, resulting in a C5b6-C7-C8β-C8α-C9 arc. Soluble regulatory proteins below the arc indicate a potential dual mechanism in protection from pore formation. These results provide a structural framework for understanding MAC pore formation and regulation, processes important for fighting infections and preventing complement-mediated tissue damage.
- Published
- 2012
- Full Text
- View/download PDF
12. Structures of Wnt-antagonist ZNRF3 and its complex with R-spondin 1 and implications for signaling.
- Author
-
Weng Chuan Peng, Wim de Lau, Pramod K Madoori, Federico Forneris, Joke C M Granneman, Hans Clevers, and Piet Gros
- Subjects
Medicine ,Science - Abstract
Zinc RING finger 3 (ZNRF3) and its homolog RING finger 43 (RNF43) antagonize Wnt signaling in adult stem cells by ubiquitinating Frizzled receptors (FZD), which leads to endocytosis of the Wnt receptor. Conversely, binding of ZNRF3/RNF43 to LGR4-6 - R-spondin blocks Frizzled ubiquitination and enhances Wnt signaling. Here, we present crystal structures of the ZNRF3 ectodomain and its complex with R-spondin 1 (RSPO1). ZNRF3 binds RSPO1 and LGR5-RSPO1 with micromolar affinity via RSPO1 furin-like 1 (Fu1) domain. Anonychia-related mutations in RSPO4 support the importance of the observed interface. The ZNRF3-RSPO1 structure resembles that of LGR5-RSPO1-RNF43, though Fu2 of RSPO1 is variably oriented. The ZNRF3-binding site overlaps with trans-interactions observed in 2:2 LGR5-RSPO1 complexes, thus binding of ZNRF3/RNF43 would disrupt such an arrangement. Sequence conservation suggests a single ligand-binding site on ZNRF3, consistent with the proposed competing binding role of ZNRF3/RNF43 in Wnt signaling.
- Published
- 2013
- Full Text
- View/download PDF
13. Modelling dynamics in protein crystal structures by ensemble refinement
- Author
-
B Tom Burnley, Pavel V Afonine, Paul D Adams, and Piet Gros
- Subjects
protein ,crystallography ,structure ,function ,dynamics ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Single-structure models derived from X-ray data do not adequately account for the inherent, functionally important dynamics of protein molecules. We generated ensembles of structures by time-averaged refinement, where local molecular vibrations were sampled by molecular-dynamics (MD) simulation whilst global disorder was partitioned into an underlying overall translation–libration–screw (TLS) model. Modeling of 20 protein datasets at 1.1–3.1 Å resolution reduced cross-validated Rfree values by 0.3–4.9%, indicating that ensemble models fit the X-ray data better than single structures. The ensembles revealed that, while most proteins display a well-ordered core, some proteins exhibit a ‘molten core’ likely supporting functionally important dynamics in ligand binding, enzyme activity and protomer assembly. Order–disorder changes in HIV protease indicate a mechanism of entropy compensation for ordering the catalytic residues upon ligand binding by disordering specific core residues. Thus, ensemble refinement extracts dynamical details from the X-ray data that allow a more comprehensive understanding of structure–dynamics–function relationships.
- Published
- 2012
- Full Text
- View/download PDF
14. Inhibition of cleavage of human complement component C5 and the R885H C5 variant by two distinct high affinity anti-C5 nanobodies
- Author
-
Eva M. Struijf, Karla I De la O Becerra, Maartje Ruyken, Fleur van Oosterom, Danique Y. Siere, Dani A. C. Heesterbeek, Edward Dolk, Raimond Heukers, Bart W. Bardoel, Piet Gros, and Suzan H.M. Rooijakkers
- Abstract
The human complement system plays a crucial role in immune defense. However, its erroneous activation contributes to many serious inflammatory diseases. Since most unwanted complement effector functions result from C5 cleavage, development of C5 inhibitors, such as clinically approved monoclonal antibody Eculizumab, are of great interest. In this study, we developed and characterized two anti-C5 nanobodies, UNbC5-1 and UNbC5-2. Using surface plasmon resonance (SPR), we determined a binding affinity of 120 pM for UNbC5-1 and 8 pM for UNbC5-2. Competition experiments determined that the two nanobodies recognize distinct epitopes on C5. Both nanobodies efficiently interfered with C5 cleavage in a human serum environment, as they prevented red blood cell lysis via membrane attack complexes (C5b-9) and the formation of chemoattractant C5a. The cryo-EM structure of UNbC5-1 and UNbC5-2 in complex with C5 revealed that the binding interfaces of UNbC5-1 and UNbC5-2 overlap with known complement inhibitors Eculizumab and RaCI3, respectively. UNbC5-1 binds to the MG7 domain of C5, facilitated by a hydrophobic core and polar interactions, and UNbC5-2 interacts with the C5d domain mostly by salt bridges and hydrogen bonds. Interestingly, UNbC5-1 potently binds and inhibits C5 R885H, a genetic variant of C5, that is not recognized by Eculizumab. Altogether, we identified and characterized two different, high affinity nanobodies against human C5. Both nanobodies could serve as diagnostic and/or research tools to detect C5 or inhibit C5 cleavage. Furthermore, the residues targeted by UNbC5-1 hold important information for therapeutic inhibition of different polymorphic variants of C5.
- Published
- 2023
15. Releasing Nonperipheral Subunits from Protein Complexes in the Gas Phase
- Author
-
Guanbo Wang, Xinyang Shao, Deniz Ugurlar, Albert J. R. Heck, Lingxiao Chaihu, Rongrong Dai, Rob N. de Jong, Meng Tian, and Piet Gros
- Subjects
Protein function ,Surface Properties ,Chemistry ,010401 analytical chemistry ,Proteins ,010402 general chemistry ,Mass spectrometry ,Tandem mass spectrometry ,01 natural sciences ,Dissociation (chemistry) ,0104 chemical sciences ,Analytical Chemistry ,Gas phase ,Protein Subunits ,Tandem Mass Spectrometry ,Biophysics ,Protein quaternary structure ,Gases ,Particle Size ,Stoichiometry - Abstract
The quaternary structure is an important feature regulating protein function. Native mass spectrometry contributes to untangling quaternary structures by preserving the integrity of protein complexes in the gas phase. Tandem mass spectrometry by collision-induced dissociation (CID) can then be used to release subunits from these intact complexes, thereby providing structural information on the stoichiometry and topology. Cumulatively, such studies have revealed the preferred release of peripheral subunits during CID. In contrast, here we describe and focus on dissociation pathways that release nonperipheral subunits from hetero-complexes in CID at high collision energies. We find that nonperipheral subunits are ejected with a high propensity, as a consequence of sequential dissociation events, upon initial removal of peripheral subunits. Alternatively, nonperipheral subunits can be released directly from a charge-reduced or an elongated intact complex. As demonstrated here for a range of protein assemblies, releasing nonperipheral subunits under controlled conditions may provide unique structural information on the stoichiometry and topology of protein complexes.
- Published
- 2020
16. An Elegant Four-Helical Fold in NOX and STEAP Enzymes Facilitates Electron Transport across Biomembranes—Similar Vehicle, Different Destination
- Author
-
Andrea Mattevi, Wout Oosterheert, Piet Gros, and Joana Reis
- Subjects
inorganic chemicals ,Heme ,Cyanobacteria ,Electron source ,Electron Transport ,03 medical and health sciences ,0302 clinical medicine ,Extracellular ,Humans ,Amino Acid Sequence ,Enzyme Inhibitors ,NOx ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Binding Sites ,Chemistry ,Helix-Loop-Helix Motifs ,NADPH Oxidases ,SUPERFAMILY ,General Medicine ,General Chemistry ,Acceptor ,Electron transport chain ,Protein Structure, Tertiary ,Enzyme ,030220 oncology & carcinogenesis ,Biophysics ,Oxidoreductases ,Reactive Oxygen Species ,Oxidation-Reduction ,Sequence Alignment ,NADP ,Intracellular - Abstract
The ferric reductase superfamily comprises several oxidoreductases that use an intracellular electron source to reduce an extracellular acceptor substrate. NADPH oxidases (NOXs) and six-transmembrane epithelial antigen of the prostate enzymes (STEAPs) are iconic members of the superfamily. NOXs produce extracellular reactive oxygen species that exert potent bactericidal activities and trigger redox-signaling cascades that regulate cell division and differentiation. STEAPs catalyze the reduction of extracellular iron and copper which is necessary for the bioavailability of these essential elements. Both NOXs and STEAPs are present as multiple isozymes with distinct regulatory properties and physiological roles. Despite the important roles of NOXs and STEAPs in human physiology and despite their wide involvement in diseases like cancer, their mode of action at the molecular level remained incompletely understood for a long time, in part due to the absence of high-resolution models of the complete enzymes. Our two laboratories have elucidated the three-dimensional structures of NOXs and STEAPs, providing key insight into their mechanisms and evolution. The enzymes share a conserved transmembrane helical domain with an eye-catching hourglass shape. On the extracellular side, a heme prosthetic group is at the bottom of a pocket where the substrate (O
- Published
- 2020
17. Anti-LRP5/6 VHHs promote differentiation of Wnt-hypersensitive intestinal stem cells
- Author
-
D. Proverbio, T. Kralj, Madelon M. Maurice, Piet Gros, Sarah Stryeck, M. Goldflam, C. Ullman, G. Hermans, T. Aastrup, R.C. van Scherpenzeel, T.Z. Bass, Ingrid Jordens, and Nicola Fenderico
- Subjects
Models, Molecular ,0301 basic medicine ,Transcription, Genetic ,Protein Conformation ,Receptor expression ,Cellular differentiation ,General Physics and Astronomy ,02 engineering and technology ,Crystallography, X-Ray ,Mice ,Models ,Intestine, Small ,Stem Cells/cytology ,Low Density Lipoprotein Receptor-Related Protein-5/antagonists & inhibitors ,lcsh:Science ,Non-U.S. Gov't ,beta Catenin ,Tissue homeostasis ,Tumor ,Crystallography ,Multidisciplinary ,Chemistry ,Research Support, Non-U.S. Gov't ,Stem Cells ,Wnt signaling pathway ,LRP6 ,Cell Differentiation ,LRP5 ,Wnt3A Protein/genetics ,021001 nanoscience & nanotechnology ,Intestine ,3. Good health ,Cell biology ,Organoids ,Low Density Lipoprotein Receptor-Related Protein-5 ,Low Density Lipoprotein Receptor-Related Protein-6 ,Organoids/cytology ,Stem cell ,0210 nano-technology ,Transcription ,Protein Binding ,Single-Domain Antibodies/chemistry ,Intestine, Small/cytology ,beta Catenin/genetics ,Ubiquitin-Protein Ligases ,Science ,Research Support ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Low Density Lipoprotein Receptor-Related Protein-6/antagonists & inhibitors ,Genetic ,Cell Line, Tumor ,Wnt3A Protein ,Small/cytology ,Journal Article ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Ubiquitin-Protein Ligases/genetics ,Cell Proliferation ,Binding Sites ,HEK 293 cells ,Molecular ,General Chemistry ,Fibroblasts ,Single-Domain Antibodies ,HEK293 Cells ,030104 developmental biology ,Fibroblasts/cytology ,Gene Expression Regulation ,X-Ray ,lcsh:Q ,Protein Conformation, beta-Strand ,beta-Strand - Abstract
Wnt-induced β-catenin-mediated transcription is a driving force for stem cell self-renewal during adult tissue homeostasis. Enhanced Wnt receptor expression due to mutational inactivation of the ubiquitin ligases RNF43/ZNRF3 recently emerged as a leading cause for cancer development. Consequently, targeting canonical Wnt receptors such as LRP5/6 holds great promise for treatment of such cancer subsets. Here, we employ CIS display technology to identify single-domain antibody fragments (VHH) that bind the LRP6 P3E3P4E4 region with nanomolar affinity and strongly inhibit Wnt3/3a-induced β-catenin-mediated transcription in cells, while leaving Wnt1 responses unaffected. Structural analysis reveal that individual VHHs variably employ divergent antigen-binding regions to bind a similar surface in the third β-propeller of LRP5/6, sterically interfering with Wnt3/3a binding. Importantly, anti-LRP5/6 VHHs block the growth of Wnt-hypersensitive Rnf43/Znrf3-mutant intestinal organoids through stem cell exhaustion and collective terminal differentiation. Thus, VHH-mediated targeting of LRP5/6 provides a promising differentiation-inducing strategy for treatment of Wnt-hypersensitive tumors., Enhanced Wnt receptor activity is a major cause of cancer development. Here the authors identify camelid single-domain antibody fragments (VHHs) that bind to the Wnt receptor LRP5/6 ectodomain, determine the crystal structures and show that these VHHs selectively inhibit Wnt3- mediated cellular responses and block the growth of mutant Wnt-hypersensitive intestinal tumor organoids.
- Published
- 2019
18. Insight into mode-of-action and structural determinants of the compstatin family of clinical complement inhibitors
- Author
-
Christina, Lamers, Xiaoguang, Xue, Martin, Smieško, Henri, van Son, Bea, Wagner, Nadja, Berger, Georgia, Sfyroera, Piet, Gros, John D, Lambris, and Daniel, Ricklin
- Subjects
Complement Inactivating Agents ,Water ,Complement C3 ,Peptides, Cyclic - Abstract
With the addition of the compstatin-based complement C3 inhibitor pegcetacoplan, another class of complement targeted therapeutics have recently been approved. Moreover, compstatin derivatives with enhanced pharmacodynamic and pharmacokinetic profiles are in clinical development (e.g., Cp40/AMY-101). Despite this progress, the target binding and inhibitory modes of the compstatin family remain incompletely described. Here, we present the crystal structure of Cp40 complexed with its target C3b at 2.0-Å resolution. Structure-activity-relationship studies rationalize the picomolar affinity and long target residence achieved by lead optimization, and reveal a role for structural water in inhibitor binding. We provide explanations for the narrow species specificity of this drug class and demonstrate distinct target selection modes between clinical compstatin derivatives. Functional studies provide further insight into physiological complement activation and corroborate the mechanism of its compstatin-mediated inhibition. Our study may thereby guide the application of existing and development of next-generation compstatin analogs.
- Published
- 2021
19. C1q binding to surface-bound IgG is stabilized by C1r2s2 proteases
- Author
-
Paul W. H. I. Parren, Frank J. Beurskens, Jos A. G. van Strijp, Annemarie Kuipers, Jürgen Strasser, Gestur Vidarsson, Carla J. C. de Haas, Daniel Ricklin, Maartje Ruyken, Johannes Preiner, Piet Gros, Kok P. M. van Kessel, Piet C. Aerts, Deniz Ugurlar, Kevin Widmer, Suzan H.M. Rooijakkers, Janine Schuurman, and Seline A. Zwarthoff
- Subjects
Proteases ,biology ,Chemistry ,Effector ,Phagocytosis ,Igg subclasses ,medicine.disease_cause ,Heterotetramer ,Cell biology ,Lytic cycle ,Staphylococcus aureus ,biology.protein ,medicine ,Antibody - Abstract
Complement is an important effector mechanism for antibody-mediated clearance of infections and tumor cells. Upon binding to target cells, the antibody’s constant (Fc) domain recruits complement component C1 to initiate a proteolytic cascade that generates lytic pores and stimulates phagocytosis. The C1 complex (C1qr2s2) consists of the large recognition protein C1q and a heterotetramer of proteases C1r and C1s (C1r2s2). While interactions between C1 and IgG-Fc’s are believed to be mediated by the globular heads of C1q, we here find that C1r2s2 proteases affect the capacity of C1q to form an avid complex with surface-bound IgG molecules (on various DNP-coated surfaces and pathogenic Staphylococcus aureus). The extent to which C1r2s2 contribute to C1q-IgG stability strongly differs between human IgG subclasses. Using antibody engineering of monoclonal IgG we reveal that hexamer-enhancing mutations improve C1q-IgG stability, both in absence and presence of C1r2s2. In addition, hexamer-enhanced IgGs targeting S. aureus mediate improved complement-dependent phagocytosis by human neutrophils. Altogether, these molecular insights into complement binding to surface-bound IgGs could be important for optimal design of antibody therapies.
- Published
- 2021
20. Author Reply to Peer Reviews of Implications for tetraspanin-enriched microdomain assembly based on structures of CD9 with EWI-F
- Author
-
Piet Gros, Paul M. P. van Bergen en Henegouwen, Martin Lutz, Loes M. J. Kroon-Batenburg, Nicholas M. Pearce, Jip Manshande, Sofia Doulkeridou, Wouter Pos, Viviana Neviani, Katerina T. Xenaki, and Wout Oosterheert
- Published
- 2020
21. Refactoring the B factor: intuitively extracting structural dynamics from macromolecular disorder
- Author
-
Nicholas M. Pearce and Piet Gros
- Subjects
Inorganic Chemistry ,Structural Biology ,General Materials Science ,Physical and Theoretical Chemistry ,Condensed Matter Physics ,Biochemistry - Published
- 2021
22. Preclinical Development of AT1412, a Patient Derived CD9 Antibody That Does Not Induce Thrombosis for Treatment of B ALL
- Author
-
Anikó Szabó, Sjeng Horbach, Remko Schotte, Susan E. van Hal-van Veen, Madalina Cercel, Hans van Eenennaam, Anita W. Rijneveld, Dorien van de Berg, Mette D. Hazenberg, Wouter Pos, Yvonne Claassen, Etsuko Yasuda, Hergen Spits, Piet Gros, Christien Fatmawati, Julien Villaudy, Esmay Frankin, Greta de Jong, Sophie E. Levie, Daniel M Go, Martijn Kedde, and Viviana Neviani
- Subjects
biology ,business.industry ,Immunology ,biology.protein ,medicine ,Cell Biology ,Hematology ,Antibody ,medicine.disease ,business ,Biochemistry ,Thrombosis - Abstract
Despite recent advances in treatment of B-acute lymphoblastic leukemia (B-ALL) there is still a need for novel targeted therapies. The tetraspanin CD9 is expressed in 60-80% of B-ALL and correlates with adverse prognosis. Recently, the mouse CD9 antibody ALB6 was shown to induce leukemia rejection in NOD/SCID mice. However, clinical development of ALB6 and other CD9-targeting antibodies was hampered by their CD9 mediated induction of platelet aggregation. It is known that CD9 is still expressed on tumor cells after treatment with chemotherapy or blinatumomab (Leung, 2019; Linder, 2016). AT1412 is a fully human antibody isolated from B cells of a patient that was cured from stage IV metastatic melanoma (Verdegaal, 2011). AT1412 targets CD9, without inducing platelet aggregation in vitro or thrombosis in cynomolgus monkeys after intravenous administration at therapeutic dose levels. By crystallography AT1412 was shown to bind a unique epitope preventing homodimerization of CD9, distinctly different from other CD9 antibodies. AT1412 binds a majority of patient B-ALL samples, but not T-ALL and induces ADCC and ADCP of CD9 positive B-ALL primary cells and the level of cytotoxicity significantly correlated with that of AT1412 binding. In both NSG and immunodeficient mice harboring a human immune system (HIS mice) AT1412 demonstrated a strong, dose-dependent tumor rejection of B-ALL, most pronounced in the extramedullary sites. In HIS mice AT1412 treatment led to an accumulation of T cells and CD14+ myeloid cells at the tumor sites. To support clinical development, pre-clinical safety of AT1412 was evaluated in cynomolgus monkeys. AT1412 demonstrated a half-life of 8.5 days, supporting 2-3 weekly administration in humans. Besides transient thrombocytopenia no other pathological deviations were observed. No effect on coagulation parameters, bruising or bleeding were observed macro- or microscopically. The thrombocytopenia is reversible, and its recovery accelerated in those animals developing anti-drug antibodies. Taken together, we demonstrate that CD9 on B-ALL cells can be successfully targeted by AT1412. AT1412 targets a unique epitope and does not induce thrombosis. Pre-clinical safety assessment is supporting that AT1412 can be safely administered. A First in Human clinical study is scheduled to start early 2021 in human solid tumors to determine safety and efficacy. AT1412 efficacy will be evaluated in B-ALL in expansion cohorts. Leung, K.T., Zhang, C., Chan, K.Y.Y., Li, K., Cheung, J.T.K., Ng, M.H.L., Zhang, X.-B., Sit, T., Lee, W.Y.W., Kang, W., et al. (2019). CD9 blockade suppresses disease progression of high-risk pediatric B-cell precursor acute lymphoblastic leukemia and enhances chemosensitivity. Leukemia. Linder, K., Gandhiraj, D., Hanmantgad, M., Seiter, K., and Liu, D. (2016). Complete remission after single agent blinatumomab in a patient with pre-B acute lymphoid leukemia relapsed and refractory to three prior regimens: HyperCVAD, high dose cytarabine mitoxantrone and CLAG. Exp. Hematol. Oncol. 5, 5-8. Verdegaal, E.M.E., Visser, M., Ramwadhdoebé, T.H., van der Minne, C.E., van Steijn, J. a Q.M.J., Kapiteijn, E., Haanen, J.B. a G., van der Burg, S.H., Nortier, J.W.R., and Osanto, S. (2011). Successful treatment of metastatic melanoma by adoptive transfer of blood-derived polyclonal tumor-specific CD4+ and CD8+ T cells in combination with low-dose interferon-alpha. Cancer Immunol. Immunother. 60, 953-963. Disclosures Schotte: AIMM Therapeutics: Current Employment, Current equity holder in private company. Villaudy:AIMM Therapeutics: Current Employment, Current equity holder in private company. Levie:AIMM Therapeutics: Current Employment, Current equity holder in private company. Go:AIMM Therapeutics: Current Employment, Current equity holder in private company. Yasuda:AIMM Therapeutics: Current Employment, Current equity holder in private company. Frankin:AIMM Therapeutics: Current Employment, Current equity holder in private company. Cercel:AIMM Therapeutics: Current Employment, Current equity holder in private company. van Hal-van Veen:AIMM Therapeutics: Current Employment, Current equity holder in private company. van de Berg:AIMM Therapeutics: Current Employment, Current equity holder in private company. Fatmawati:AIMM Therapeutics: Current Employment, Current equity holder in private company. Kedde:AIMM Therapeutics: Current Employment, Current equity holder in private company. Claassen:AIMM Therapeutics: Current Employment, Current equity holder in private company. Rijneveld:Amgen: Research Funding; Servier: Research Funding. Spits:AIMM Therapeutics: Current equity holder in private company, Ended employment in the past 24 months, Membership on an entity's Board of Directors or advisory committees. van Eenennaam:AIMM Therapeutics: Current Employment, Current equity holder in private company.
- Published
- 2020
23. ‘Stealth’ corporate innovation: an emerging threat for therapeutic drug development
- Author
-
John P. A. Ioannidis, Antonio M. Risitano, Brian V. Geisbrecht, Bo Nilsson, Daniel Ricklin, Alberto Mantovani, B. Paul Morgan, Tom Eirik Mollnes, Ronald P. Taylor, John D. Lambris, Mohamed R. Daha, Taroh Kinoshita, E. Sander Connolly, Dimitrios C. Mastellos, Robert A. Montgomery, Ruben Pio, V. Michael Holers, Piet Gros, Berhane Ghebrehiwet, George Hajishengallis, Anna M. Blom, Markus Huber-Lang, Mastellos, Dimitrios C, Blom, Anna M, Connolly, E Sander, Daha, Mohamed R, Geisbrecht, Brian V, Ghebrehiwet, Berhane, Gros, Piet, Hajishengallis, George, Holers, V Michael, Huber-Lang, Marku, Kinoshita, Taroh, Mollnes, Tom E, Montgomery, Robert A, Morgan, B Paul, Nilsson, Bo, Pio, Ruben, Ricklin, Daniel, Risitano, Antonio M, Taylor, Ronald P, Mantovani, Alberto, Ioannidis, John P A, and Lambris, John D
- Subjects
0301 basic medicine ,Immunology ,Corporate innovation ,Article ,ComputingMilieux_GENERAL ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Drug development ,Data transparency ,Accountability ,Immunology and Allergy ,Engineering ethics ,Business ,030215 immunology - Abstract
Therapeutic innovations of potentially great clinical impact should embrace the overarching values of research accountability, data transparency and validation through the scientific peer-review process.
- Published
- 2019
24. Insights into IgM-mediated complement activation based on in situ structures of IgM-C1-C4b
- Author
-
Aimee L. Boyle, Piet Gros, Abraham J. Koster, Alexander Kros, Christoph A. Diebolder, and Thomas H. Sharp
- Subjects
0301 basic medicine ,In situ ,Models, Molecular ,Proteases ,IgM ,chemical and pharmacologic phenomena ,medicine.disease_cause ,Antibodies ,Autoimmunity ,03 medical and health sciences ,0302 clinical medicine ,Immunology and Inflammation ,C1 ,Antigen ,Complement C1 ,medicine ,Humans ,subtomogram averaging ,complement ,Antigens ,Complement Activation ,Multidisciplinary ,Binding Sites ,Chemistry ,Hexagonal crystal system ,Complement C4 ,Biological Sciences ,cryoelectron tomography ,Antigen binding ,3. Good health ,Complement system ,030104 developmental biology ,Membrane ,Immunoglobulin M ,030220 oncology & carcinogenesis ,Biophysics ,Technology Platforms - Abstract
Significance IgM antibodies protect mammals against humoral microbial infection and mediate clearance of cellular debris. IgM activates the immune complement system only after binding to cell-surface antigens. Here we report the in situ 3D structures of surface-antigen–bound IgM antibodies in complex with both C1 and C4b complement components. The data indicate the structural arrangement of pentameric and hexameric IgM upon antigen binding, exposing the C1q-binding sites with both adopting hexagonal symmetry. The structures reveal the entire C1qr2s2 complex and elucidate several protein–protein interactions with C4b and IgM. Based on the structural data, we hypothesize a C1q-transmitted surface trigger that activates C1, leading to C4 cleavage and C4b deposition on membranes., Antigen binding by serum Ig-M (IgM) protects against microbial infections and helps to prevent autoimmunity, but causes life-threatening diseases when mistargeted. How antigen-bound IgM activates complement-immune responses remains unclear. We present cryoelectron tomography structures of IgM, C1, and C4b complexes formed on antigen-bearing lipid membranes by normal human serum at 4 °C. The IgM-C1-C4b complexes revealed C4b product release as the temperature-limiting step in complement activation. Both IgM hexamers and pentamers adopted hexagonal, dome-shaped structures with Fab pairs, dimerized by hinge domains, bound to surface antigens that support a platform of Fc regions. C1 binds IgM through widely spread C1q-collagen helices, with C1r proteases pointing outward and C1s bending downward and interacting with surface-attached C4b, which further interacts with the adjacent IgM-Fab2 and globular C1q-recognition unit. Based on these data, we present mechanistic models for antibody-mediated, C1q-transmitted activation of C1 and for C4b deposition, while further conformational rearrangements are required to form C3 convertases.
- Published
- 2019
25. 582P Preclinical development of AT1412, a patient derived CD9 antibody that does not induce thrombosis for treatment of precursor B ALL
- Author
-
Remko Schotte, Anita W. Rijneveld, H. van Eenennaam, Anikó Szabó, Sophie E. Levie, Etsuko Yasuda, Julien Villaudy, Hergen Spits, Martijn Kedde, S.H. van der Burg, Madalina Cercel, Viviana Neviani, Sjeng Horbach, Christien Fatmawati, G. de Jong, Wouter Pos, Piet Gros, P. van Helden, Mette D. Hazenberg, and Els M. E. Verdegaal
- Subjects
Oncology ,biology ,B lymphoblastic leukemia ,business.industry ,Cancer research ,biology.protein ,Medicine ,Hematology ,Antibody ,business ,medicine.disease ,Thrombosis - Published
- 2020
26. Abstract 532: A patient-derived anti-CD9 antibody induces tumor rejection and synergistically enhances anti-PD1 activity
- Author
-
Els M. E. Verdegaal, Piet Gros, Susan van Hal, Hans van Eenennaam, Hergen Spits, Julien Villaudy, Yvonne Claasen, Christien Fatmawati, Pauline M. van Helden, Viviana Neviani, Koen Wagner, Etsuko Yasuda, Martijn Kedde, Wouter Pos, Sjoerd H. van der Burg, Remko Schotte, and Daniel Go
- Subjects
Cancer Research ,Oncology ,biology ,business.industry ,Tumor rejection ,Cancer research ,biology.protein ,Medicine ,Antibody ,business ,Anti pd1 - Abstract
Background Adaptive immunity to cancer cells has been shown to form a crucial part of cancer immunotherapy. Recently, the importance of tumor B-cell signatures were shown to correlate with melanoma survival. We investigated whether anti-tumor antibodies could be isolated from a patient with metastatic melanoma that was tumor-free for 6 years following adoptive transfer of ex vivo expanded autologous T cells (Verdegaal 2011). Methods Peripheral blood memory B cells were immortalized using AIMM's immmortalisation technology (ectopic Bcl-6 and Bcl-xL; Kwakkenbos 2010) expression and analyzed for the presence of tumor-reactive B cells. Results AT1412 antibody was identified by virtue of its differential binding to melanoma cells as compared to healthy melanocytes. AT1412 was found to bind the tetraspanin CD9, a broadly expressed protein involved in multiple cellular activities and disease progression. The crystal structure of the CD9 large extracellular loop in complex with an AT1412 Fab fragment revealed that AT1412 binds the CD9 epitope in an extended and unfolded conformation. In addition to melanoma, AT1412 binds other tumor types including gastric, colon- and pancreatic cancer. AT1412 was shown to induce antibody dependent cellular cytotoxicity (ADCC) and antibody dependent cellular phagocytosis (ADCP) of cancer cells. In addition, AT1412 demonstrated activation of monocytes, most likely via activation of CD9 on platelets-bound monocytes. In mice carrying a human immune system (HIS-mice; van Lent 2010) AT1412 strongly enhanced tumor rejection of A375 and SKMEL-5 tumor cells. AT1412 treatment was shown to enhance tumor infiltration of CD8 T cells and macrophages. AT1412 efficacy was further synergistically enhanced when combined with an anti-PD1 antibody (nivolumab). Other groups have shown that anti-CD9 antibodies can induce tumor rejection in mice. However, these antibodies could not be advanced due to induction of platelet aggregation. We show that AT1412 does not induce aggregation of both human and cynomolgus platelets. Also affinity matured versions of AT1412 do not induce platelet aggregation, supporting that AT1412 binds a unique epitope on CD9. In addition, we studied the safety of AT1412 during a one month exposure in a weekly administration in cynomolgus monkeys. Besides a transient thrombocytopenia AT1412 was well tolerated up to 10 mg/kg antibody (highest dose tested) and did not lead to other adverse events nor were any coagulation factors affected Conclusions Taken together, applying AIMM's proprietary B-cell immortalization technology we isolated antibody AT1412 that targets a unique epitope on CD9. AT1412 was shown to induce tumor rejection as a single agent and enhances the activity of anti-PD-1 antibodies, while not inducing platelet aggregation. One month exposure of AT1412 in monkeys indicated that AT1412 can be safely administered. Preclinical development of AT1412 is ongoing to initiate clinical evaluation in 2020. Funding Dutch Cancer Society, grant UVA 2010-4822 Citation Format: Remko Schotte, Julien Villaudy, Martijn Kedde, Wouter Pos, Koen Wagner, Viviana Neviani, Daniel Go, Etsuko Yasuda, Christien Fatmawati, Els Verdegaal, Susan van Hal, Yvonne Claasen, Hans van Eenennaam, Pauline van Helden, Piet Gros, Sjoerd van der Burg, Hergen Spits. A patient-derived anti-CD9 antibody induces tumor rejection and synergistically enhances anti-PD1 activity [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 532.
- Published
- 2020
27. Regulator-dependent mechanisms of C3b processing by factor I allow differentiation of immune responses
- Author
-
Patrizia Di Crescenzio, Thomas H. Sharp, John D. Lambris, Federico Forneris, Piet Gros, Jin Wu, Daniel Ricklin, Joke C.M. Granneman, Xiaoguang Xue, and Christoph Q. Schmidt
- Subjects
0301 basic medicine ,Models, Molecular ,Protein Conformation ,Immunology ,Regulator ,chemical and pharmacologic phenomena ,Complement factor I ,Biology ,Crystallography, X-Ray ,Article ,03 medical and health sciences ,Scissile bond ,0302 clinical medicine ,Protein structure ,Structural Biology ,Humans ,Molecular Biology ,X-ray crystallography ,Proteases ,CUB domain ,Complement system ,Cell biology ,030104 developmental biology ,Complement Factor I ,Factor H ,Complement Factor H ,Complement C3b ,Proteolysis ,iC3b ,030215 immunology ,Protein Binding - Abstract
The complement system labels microbes and host debris for clearance. Degradation of surface-bound C3b is pivotal to direct immune responses and protect host cells. How the serine protease factor I (FI), assisted by regulators, cleaves either two or three distant peptide bonds in the CUB domain of C3b remains unclear. We present a crystal structure of C3b in complex with FI and regulator factor H (FH; domains 1-4 with 19-20). FI binds C3b-FH between FH domains 2 and 3 and a reoriented C3b C-terminal domain and docks onto the first scissile bond, while stabilizing its catalytic domain for proteolytic activity. One cleavage in C3b does not affect its overall structure, whereas two cleavages unfold CUB and dislodge the thioester-containing domain (TED), affecting binding of regulators and thereby determining the number of cleavages. These data explain how FI generates late-stage opsonins iC3b or C3dg in a context-dependent manner, to react to foreign, danger or healthy self signals.
- Published
- 2017
28. A new approach for generating bispecific antibodies based on a common light chain format and the stable architecture of human immunoglobulin G
- Author
-
Camilla, De Nardis, Linda J A, Hendriks, Emilie, Poirier, Tudor, Arvinte, Piet, Gros, Alexander B H, Bakker, and John, de Kruif
- Subjects
Models, Molecular ,Mice, Inbred BALB C ,Receptor, ErbB-3 ,Protein Conformation ,Protein Stability ,Receptor, ErbB-2 ,Drug Storage ,Antibody Affinity ,Computational Biology ,Antineoplastic Agents ,CHO Cells ,Antibodies, Monoclonal, Humanized ,Protein Engineering ,Recombinant Proteins ,Molecular Docking Simulation ,Bioreactors ,Cricetulus ,Drug Stability ,Immunoglobulin G ,Antibodies, Bispecific ,Editors' Picks Highlights ,Animals ,Humans ,Female ,Half-Life - Abstract
Bispecific antibodies show great promise as intrinsic combination therapies, but often suffer from poor physiochemical properties, many times related to poor heterodimerization. De Nardis et al. identify specific electrostatic interactions that facilitate efficient heterodimerization, resulting in bispecific antibodies with physiochemical properties very similar to those of naturally occurring antibodies. This provides a new platform for the treatment of an array of diseases from cancer and autoimmune diseases to infectious diseases.
- Published
- 2017
29. Author Correction: ‘Stealth’ corporate innovation: an emerging threat for therapeutic drug development
- Author
-
Dimitrios C. Mastellos, Anna M. Blom, Mohamed R. Daha, John D. Lambris, Antonio M. Risitano, Brian V. Geisbrecht, Daniel Ricklin, Berhane Ghebrehiwet, John P. A. Ioannidis, Ronald P. Taylor, Alberto Mantovani, Tom Eirik Mollnes, George Hajishengallis, B. Paul Morgan, Taroh Kinoshita, E. Sander Connolly, Piet Gros, Robert A. Montgomery, Bo Nilsson, Ruben Pio, V. Michael Holers, and Markus Huber-Lang
- Subjects
Drug development ,business.industry ,Immunology ,Immunology and Allergy ,Business ,Public relations ,Corporate innovation - Published
- 2019
30. Regulators of complement activity mediate inhibitory mechanisms through a common C3b-binding mode
- Author
-
Daniel Ricklin, Georgia Sfyroera, John P. Atkinson, Richard E. Hauhart, M. Kathryn Liszewski, Elena B. Volokhina, Joke C.M. Granneman, Federico Forneris, Zhuoer Lin, John D. Lambris, Piet Gros, Apostolia Tzekou, Xiaoguang Xue, Jin Wu, and Paula Bertram
- Subjects
0301 basic medicine ,Immunology ,chemical and pharmacologic phenomena ,Inhibitory postsynaptic potential ,General Biochemistry, Genetics and Molecular Biology ,Cofactor ,Article ,regulators of complement activity ,Complement activity ,Membrane Cofactor Protein ,Viral Matrix Proteins ,03 medical and health sciences ,decay‐accelerating activity ,0302 clinical medicine ,Immune system ,Protein Domains ,Structural Biology ,Homologous chromosome ,Humans ,complement ,Smallpox virus ,cofactor activity ,Molecular Biology ,Complement Activation ,immune evasion ,Binding Sites ,General Immunology and Microbiology ,biology ,CD55 Antigens ,General Neuroscience ,Articles ,3. Good health ,Complement (complexity) ,Complement system ,Cell biology ,Renal disorders Radboud Institute for Molecular Life Sciences [Radboudumc 11] ,030104 developmental biology ,Complement C3b ,biology.protein ,Receptors, Complement 3b ,030215 immunology - Abstract
Regulators of complement activation (RCA) inhibit complement‐induced immune responses on healthy host tissues. We present crystal structures of human RCA (MCP, DAF, and CR1) and a smallpox virus homolog (SPICE) bound to complement component C3b. Our structural data reveal that up to four consecutive homologous CCP domains (i–iv), responsible for inhibition, bind in the same orientation and extended arrangement at a shared binding platform on C3b. Large sequence variations in CCP domains explain the diverse C3b‐binding patterns, with limited or no contribution of some individual domains, while all regulators show extensive contacts with C3b for the domains at the third site. A variation of ~100° rotation around the longitudinal axis is observed for domains binding at the fourth site on C3b, without affecting the overall binding mode. The data suggest a common evolutionary origin for both inhibitory mechanisms, called decay acceleration and cofactor activity, with variable C3b binding through domains at sites ii, iii, and iv, and provide a framework for understanding RCA disease‐related mutations and immune evasion.
- Published
- 2016
31. Molecular Basis of Assembly and Activation of Complement Component C1 in Complex with Immunoglobulin G1 and Antigen
- Author
-
Piet Gros, Rob N. de Jong, Janine Schuurman, Paul W. H. I. Parren, Ewald T. J. van den Bremer, Albert J. R. Heck, Deniz Ugurlar, Aran F. Labrijn, Frank J. Beurskens, and Guanbo Wang
- Subjects
0301 basic medicine ,Glycosylation ,Complement factor I ,Antigen-Antibody Reactions ,03 medical and health sciences ,Classical complement pathway ,Immunoglobulin Fab Fragments ,0302 clinical medicine ,Tandem Mass Spectrometry ,Cell Line, Tumor ,Humans ,Antigens ,Molecular Biology ,Complement Activation ,Complement component 2 ,biology ,CD46 ,Protein Stability ,Complement C1q ,Cell Biology ,Molecular biology ,Complement system ,Cell biology ,030104 developmental biology ,Factor H ,Immunoglobulin G ,Mutation ,biology.protein ,Chromatography, Gel ,Binding Sites, Antibody ,Complement membrane attack complex ,030215 immunology ,Complement control protein ,Protein Binding - Abstract
The classical complement pathway contributes to the natural immune defense against pathogens and tumors. IgG antibodies can assemble at the cell surface into hexamers via Fc:Fc interactions, which recruit complement component C1q and induce complement activation. Biophysical characterization of the C1:IgG complex has remained elusive primarily due to the low affinity of IgG-C1q binding. Using IgG variants that dynamically form hexamers efficient in C1q binding and complement activation, we could assess C1q binding in solution by native mass spectrometry and size-exclusion chromatography. Fc-domain deglycosylation, described to abrogate complement activation, affected IgG hexamerization and C1q binding. Strikingly, antigen binding by IgG hexamers or deletion of the Fab arms substantially potentiated complement initiation, suggesting that Fab-mediated effects impact downstream Fc-mediated events. Finally, we characterized a reconstituted 2,045.3 ± 0.4-kDa complex of intact C1 bound to antigen-saturated IgG hexamer by native mass spectrometry, providing a clear visualization of a complete complement initiation complex.
- Published
- 2015
32. Cryo-electron tomography reveals early steps of complement activation by IgM
- Author
-
Aimee L. Boyle, Piet Gros, Christoph A. Diebolder, Abraham J. Koster, Thomas H. Sharp, and Alexander Kros
- Subjects
Materials science ,Immunology ,Biophysics ,Cryo-electron tomography ,Molecular Biology ,Complement system - Published
- 2018
33. Structure of complement fragment C3b–factor H and implications for host protection by complement regulators
- Author
-
You-Qiang Wu, Daniel Ricklin, Bert J. C. Janssen, Jin Wu, John D. Lambris, and Piet Gros
- Subjects
Models, Molecular ,Complement system ,Macromolecular Substances ,Complement component C3b ,Immunology ,chemical and pharmacologic phenomena ,Complement C3-C5 Convertases ,Complement factor I ,Biology ,Crystallography, X-Ray ,Complement factor B ,Article ,Humans ,Immunology and Allergy ,Innate immunity ,Binding Sites ,Complement component 2 ,Factor H ,Complement component 6 ,Protein Structure, Tertiary ,Regulator of complement activation ,Cell biology ,Biochemistry ,Complement Factor H ,Complement C3b ,Mutation ,CFHR5 ,Protein Binding - Abstract
Factor H (FH) is an abundant regulator of complement activation and protects host cells from self-attack by complement. Here we provide insight into the regulatory activity of FH by solving the crystal structure of the first four domains of FH in complex with its target, complement fragment C3b. FH interacted with multiple domains of C3b, covering a large, extended surface area. The structure indicated that FH destabilizes the C3 convertase by competition and electrostatic repulsion and that FH enables proteolytic degradation of C3b by providing a binding platform for protease factor I while stabilizing the overall domain arrangement of C3b. Our results offer general models for complement regulation and provide structural explanations for disease-related mutations in the genes encoding both FH and C3b.
- Published
- 2009
34. Solid-State Structural Characterization of Cutinase-ECE-Pincer-Metal Hybrids
- Author
-
Lucy Rutten, Gerard van Koten, Cornelis A. Kruithof, Martin Lutz, Piet Gros, Maarten R. Egmond, Birgit Wieczorek, Jean‐Paul B. A. Mannie, Robertus J. M. Klein Gebbink, and Harm P. Dijkstra
- Subjects
Cutinase ,Magnetic Resonance Spectroscopy ,Protein Conformation ,Dimer ,chemistry.chemical_element ,Crystal structure ,Crystallography, X-Ray ,Catalysis ,Coordination complex ,Metal ,chemistry.chemical_compound ,Organometallic Compounds ,Platinum ,chemistry.chemical_classification ,Molecular Structure ,biology ,Organic Chemistry ,Active site ,General Chemistry ,Crystallography ,Models, Chemical ,chemistry ,visual_art ,biology.protein ,visual_art.visual_art_medium ,Carboxylic Ester Hydrolases ,Palladium - Abstract
The first crystal structures of lipases that have been covalently modified through site-selective inhibition by different organometallic phosphonatepincer– metal complexes are described. Two ECE-pincer-type d8-metal complexes, that is, platinum (1) or palladium (2) with phosphonate esters (ECE=[(EtO)-(O=)PACHTUNGTRENUNG(-O-C6H4-ACHTUNGTRENUNG(NO2)- 4)ACHTUNGTRENUNG(-C3H6-4-ACHTUNGTRENUNG(C6H2-ACHTUNGTRENUNG(CH2E)2)] ; E= NMe2 or SMe) were introduced prior to crystallization and have been shown to bind selectively to the Ser120 residue in the active site of the lipase cutinase to give cut-1 (platinum) or cut-2 (palladium) hybrids. For all five presented crystal structures, the ECE-pincer–platplatinum or –palladium head group sticks out of the cutinase molecule and is exposed to the solvent. Depending on the nature of the ECE-pincer–metal head group, the ECE-pincer–platinum and – palladium guests occupy different pockets in the active site of cutinase, with concomitant different stereochemistries on the phosphorous atom for the cut-1 (SP) and cut-2 (RP) structures. When cut-1 was crystallized under halide-poor conditions, a novel metalinduced dimeric structure was formed between two cutinase-bound pincer– platinum head groups, which are interconnected through a single m-Cl bridge. This halide-bridged metal dimer shows that coordination chemistry is possible with protein-modified pincer–metal complexes. Furthermore, we could use NCN-pincer–platinum complex 1 as site-selective tool for the phasing of raw protein diffraction data, which shows the potential use of pincer–platinum complex 1 as a heavy-atom derivative in protein crystallography.
- Published
- 2009
35. Simple Geometry for Diffusion Limited Protein Crystal Growth: Harnessing Gravity to Suppress Convection
- Author
-
M.C.R. Heijna, Piet Gros, W.J.P. van Enckevort, P.W.G. Poodt, Arie Schouten, Elias Vlieg, Crystal and Structural Chemistry, Rontgen participation programme, Sub Crystal and Structural Chemistry, and Dep Scheikunde
- Subjects
Diffraction ,Convection ,Gravity (chemistry) ,Buoyancy ,Chemistry ,Crystal growth ,Geometry ,Solid State Chemistry ,General Chemistry ,engineering.material ,Condensed Matter Physics ,engineering ,General Materials Science ,Growth rate ,Diffusion (business) ,Protein crystallization - Abstract
A new upside-down geometry is proposed to achieve the beneficial effects of microgravity crystal growth by making use of buoyant forces instead of compensating for them. We show by growth experiments on sodium chlorate and lysozyme that crystal growth in an upside-down geometry leads to the formation of a buoyancy assisted depletion zone where convection is suppressed. The effects on growth rate and morphology that are observed are all indicative of diffusion limited growth, just as would happen in the absence of gravity. The optical quality of the lysozyme crystals clearly improved, but no effects of the growth method on X-ray diffraction quality could be observed. The simplicity of the growth geometry offers the possibility to perform large scale protein crystal growth experiments under microgravity-like conditions, without the requirement of compensating for gravity.
- Published
- 2008
36. Structure of Compstatin in Complex with Complement Component C3c Reveals a New Mechanism of Complement Inhibition
- Author
-
Piet Gros, Bert J. C. Janssen, John D. Lambris, and Els F. Halff
- Subjects
Models, Molecular ,Stereochemistry ,Molecular Conformation ,Plasma protein binding ,Biology ,Peptides, Cyclic ,Biochemistry ,Complement C3c ,Substrate Specificity ,Plasma ,03 medical and health sciences ,0302 clinical medicine ,Complement Inactivator Proteins ,Protein structure ,Macroglobulins ,Humans ,Binding site ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Binding Sites ,Complement component 3 ,Cell Biology ,Protein Structure, Tertiary ,3. Good health ,Complement system ,Biophysics ,Peptides ,Immune complex disease ,Protein Binding ,030215 immunology - Abstract
Undesired complement activation is a major cause of tissue injury in various pathological conditions and contributes to several immune complex diseases. Compstatin, a 13-residue peptide, is an effective inhibitor of the activation of complement component C3 and thus blocks a central and crucial step in the complement cascade. The precise binding site on C3, the structure in the bound form, and the exact mode of action of compstatin are unknown. Here we present the crystal structure of compstatin in complex with C3c, a major proteolytic fragment of C3. The structure reveals that the compstatin-binding site is formed by the macroglobulin (MG) domains 4 and 5. This binding site is part of the structurally stable MG-ring formed by domains MG 1-6 and is far away from any other known binding site on C3. Compstatin does not alter the conformation of C3c, whereas compstatin itself undergoes a large conformational change upon binding. We propose a model in which compstatin sterically hinders the access of the substrate C3 to the convertase complexes, thus blocking complement activation and amplification. These insights are instrumental for further development of compstatin as a potential therapeutic.
- Published
- 2007
37. Structural insights into the Slit-Robo complex
- Author
-
Andrew A. McCarthy, Nicole M. Thielens, Stephen Cusack, Cécile Morlot, Piet Gros, Roland A. Romijn, Raimond B. G. Ravelli, and Wieger Hemrika
- Subjects
Models, Molecular ,Heart morphogenesis ,Molecular Sequence Data ,Nerve Tissue Proteins ,Biology ,Crystallography, X-Ray ,Models, Biological ,Structure-Activity Relationship ,ROBO1 ,SLIT2 ,Humans ,Amino Acid Sequence ,Receptors, Immunologic ,Binding site ,Peptide sequence ,Binding Sites ,Multidisciplinary ,Heparin ,Surface Plasmon Resonance ,Biological Sciences ,Slit ,Recombinant Proteins ,Slit-Robo ,Protein Structure, Tertiary ,Cell biology ,Biochemistry ,Mutagenesis ,Intercellular Signaling Peptides and Proteins ,Signal transduction ,Signal Transduction - Abstract
Slits are large multidomain leucine-rich repeat (LRR)-containing proteins that provide crucial guidance cues in neuronal and vascular development. More recently, Slits have been implicated in heart morphogenesis, angiogenesis, and tumor metastasis. Slits are ligands for the Robo (Roundabout) receptors, which belong to the Ig superfamily of transmembrane signaling molecules. The Slit-Robo interaction is mediated by the second LRR domain of Slit and the two N-terminal Ig domains of Robo, but the molecular details of this interaction and how it induces signaling remain unclear. Here we describe the crystal structures of the second LRR domain of human Slit2 (Slit2 D2), the first two Ig domains of its receptor Robo1 (Ig1–2), and the minimal complex between these proteins (Slit2 D2-Robo1 Ig1). Slit2 D2 binds with its concave surface to the side of Ig1 with electrostatic and hydrophobic contact regions mediated by residues that are conserved in other family members. Surface plasmon resonance experiments and a mutational analysis of the interface confirm that Ig1 is the primary domain for binding Slit2. These structures provide molecular insight into Slit-Robo complex formation and will be important for the development of novel cancer therapeutics.
- Published
- 2007
38. Deciphering complement mechanisms: The contributions of structural biology
- Author
-
Sthanam V.L. Narayana, Paul N. Barlow, Piet Gros, Gérard J. Arlaud, and Christine Gaboriaud
- Subjects
Computer science ,Serine Endopeptidases ,Immunology ,Rational design ,Complement System Proteins ,computer.file_format ,Computational biology ,Crystallography, X-Ray ,Bioinformatics ,Protein Data Bank ,Complement (complexity) ,Structure-Activity Relationship ,Structural biology ,Catalytic Domain ,Component (UML) ,Animals ,Humans ,DECIPHER ,Molecular Biology ,computer - Abstract
Since the resolution of the first three-dimensional structure of a complement component in 1980, considerable efforts have been put into the investigation of this system through structural biology techniques, resulting in about a hundred structures deposited in the Protein Data Bank by the beginning of 2007. By revealing its mechanisms at the atomic level, these approaches significantly improve our understanding of complement, opening the way to the rational design of specific inhibitors. This review is co-authored by some of the researchers currently involved in the structural biology of complement and its purpose is to illustrate, through representative examples, how X-ray crystallography and NMR techniques help us decipher the many sophisticated mechanisms that underlie complement functions.
- Published
- 2007
39. Factor B structure provides insights into activation of the central protease of the complement system
- Author
-
Lucio Gomes, Wieger Hemrika, Arie Schouten, Fin J. Milder, Bert J. C. Janssen, Eric G. Huizinga, Anja Roos, Roland A. Romijn, Mohamed R. Daha, and Piet Gros
- Subjects
Models, Molecular ,Stereochemistry ,Chemistry ,Allosteric regulation ,Complement C3-C5 Convertases ,Complement System Proteins ,Regulatory Sequences, Nucleic Acid ,Crystallography, X-Ray ,Ligand (biochemistry) ,Complement factor B ,Protein Structure, Secondary ,Protein Structure, Tertiary ,Complement system ,Enzyme Activation ,Structure-Activity Relationship ,Scissile bond ,Structural Biology ,Catalytic Domain ,von Willebrand Factor ,Helix ,Humans ,Molecular Biology ,Alpha helix ,Complement Factor B - Abstract
Factor B is the central protease of the complement system of immune defense. Here, we present the crystal structure of human factor B at 2.3-A resolution, which reveals how the five-domain proenzyme is kept securely inactive. The canonical activation helix of the Von Willebrand factor A (VWA) domain is displaced by a helix from the preceding domain linker. The two helices conformationally link the scissile-activation peptide and the metal ion-dependent adhesion site required for binding of the ligand C3b. The data suggest that C3b binding displaces the three N-terminal control domains and reshuffles the two central helices. Reshuffling of the helices releases the scissile bond for final proteolytic activation and generates a new interface between the VWA domain and the serine protease domain. This allosteric mechanism is crucial for tight regulation of the complement-amplification step in the immune response.
- Published
- 2007
40. Molecular insights into the surface-specific arrangement of complement C5 convertase enzymes
- Author
-
Jasper A. Soppe, Hatice Orhan, Carla J. C. de Haas, Piet C. Aerts, Maartje Ruyken, Piet Gros, Evelien T.M. Berends, Ronald D. Gorham, and Suzan H.M. Rooijakkers
- Subjects
Physiology ,Complement ,chemical and pharmacologic phenomena ,Plant Science ,Complement receptor ,Biology ,Inflammatory diseases ,General Biochemistry, Genetics and Molecular Biology ,Catalysis ,C5-convertase ,Complement C5 Convertase, Alternative Pathway ,Structural Biology ,Humans ,Ecology, Evolution, Behavior and Systematics ,Innate immunity ,Convertase enzymes ,Agricultural and Biological Sciences(all) ,Complement component 2 ,Biochemistry, Genetics and Molecular Biology(all) ,Methodology Article ,Cell Biology ,C3-convertase ,Microspheres ,Kinetics ,Multi-molecular proteases ,Biochemistry ,Models, Chemical ,Factor H ,Biotinylation ,Complement C3b ,Alternative complement pathway ,Biophysics ,Streptavidin ,General Agricultural and Biological Sciences ,Complement membrane attack complex ,Developmental Biology ,Biotechnology - Abstract
Background Complement is a large protein network in plasma that is crucial for human immune defenses and a major cause of aberrant inflammatory reactions. The C5 convertase is a multi-molecular protease complex that catalyses the cleavage of native C5 into its biologically important products. So far, it has been difficult to study the exact molecular arrangement of C5 convertases, because their non-catalytic subunits (C3b) are covalently linked to biological surfaces through a reactive thioester. Through development of a highly purified model system for C5 convertases, we here aim to provide insights into the surface-specific nature of these important protease complexes. Results Alternative pathway (AP) C5 convertases were generated on small streptavidin beads that were coated with purified C3b molecules. Site-specific biotinylation of C3b via the thioester allowed binding of C3b in the natural orientation on the surface. In the presence of factor B and factor D, these C3b beads could effectively convert C5. Conversion rates of surface-bound C3b were more than 100-fold higher than fluid-phase C3b, confirming the requirement of a surface. We determine that high surface densities of C3b, and its attachment via the thioester, are essential for C5 convertase formation. Combining our results with molecular modeling explains how high C3b densities may facilitate intermolecular interactions that only occur on target surfaces. Finally, we define two interfaces on C5 important for its recognition by surface-bound C5 convertases. Conclusions We establish a highly purified model that mimics the natural arrangement of C5 convertases on a surface. The developed model and molecular insights are essential to understand the molecular basis of deregulated complement activity in human disease and will facilitate future design of therapeutic interventions against these critical enzymes in inflammation. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0203-8) contains supplementary material, which is available to authorized users.
- Published
- 2015
41. Structure of C3b reveals conformational changes that underlie complement activity
- Author
-
Andrew A. McCarthy, Piet Gros, Bert J. C. Janssen, Agni Christodoulidou, and John D. Lambris
- Subjects
Multidisciplinary ,Protein structure ,Complement component 2 ,biology ,Chemistry ,Complement component 7 ,biology.protein ,Anaphylatoxin ,Complement C3-C5 Convertases ,Complement factor B ,Complement system ,Cell biology ,Complement control protein - Abstract
Resistance to infection and clearance of cell debris in mammals depend on the activation of the complement system, which is an important component of innate and adaptive immunity. Central to the complement system is the activated form of C3, called C3b, which attaches covalently to target surfaces to amplify complement response, label cells for phagocytosis and stimulate the adaptive immune response. C3b consists of 1,560 amino-acid residues and has 12 domains. It binds various proteins and receptors to effect its functions. However, it is not known how C3 changes its conformation into C3b and thereby exposes its many binding sites. Here we present the crystal structure at 4-A resolution of the activated complement protein C3b and describe the conformational rearrangements of the 12 domains that take place upon proteolytic activation. In the activated form the thioester is fully exposed for covalent attachment to target surfaces and is more than 85 A away from the buried site in native C3 (ref. 5). Marked domain rearrangements in the alpha-chain present an altered molecular surface, exposing hidden and cryptic sites that are consistent with known putative binding sites of factor B and several complement regulators. The structural data indicate that the large conformational changes in the proteolytic activation and regulation of C3 take place mainly in the first conversion step, from C3 to C3b. These insights are important for the development of strategies to treat immune disorders that involve complement-mediated inflammation.
- Published
- 2006
42. Phosphatidylinositol transfer proteins: From closed for transport to open for exchange
- Author
-
Arie Schouten, Piet Gros, Karel W. A. Wirtz, Kristal- en structuurchemie, Lipide chemie op enkel-cel-niveau, Sub Membrane Enzymology begr. 01-06-12, and Dep Scheikunde
- Subjects
chemistry.chemical_classification ,Gene isoform ,Cancer Research ,Phospholipid ,Biological Transport, Active ,Metabolism ,Phosphatidylinositols ,Amino acid ,chemistry.chemical_compound ,Enzyme ,Biochemistry ,chemistry ,Genetics ,Animals ,Humans ,Molecular Medicine ,Phosphatidylinositol ,Phospholipid Transfer Proteins ,Sphingomyelin ,Molecular Biology ,Phosphatidylinositol transfer protein - Abstract
Since their purification from bovine brain phosphatidylinositol transfer proteins (PI-TP) have been subject to extensive studies mainly prompted by the hypothesis that these proteins play a key role in intracellular phosphatidylinositol (PI) transport and as a consequence, in PI-dependent signalling pathways (Helmkamp et al., 1974; Wirtz, 1997). To date, two isoforms of PI-TP have been identified: PI-TPa (270 amino acids) and PI-TPb (271 amino acids). Either isoform (Mr 32 kDa) is highly conserved (498% sequence identity) among mammalian species. Moreover, both proteins are highly homologous with a sequence identity of 77% (similarity of 94%) yet appear to have distinctly different cellular functions. In agreement with this the gene of human PI-TPa is localized to chromosome 17q13 and of PI-TPb to chromosome 22q12 (Ocaka et al., 2005). Both isoforms act as carriers of PI and phosphatidylcholine (PC) between membranes in vitro, yet PI-TPb can also transfer sphingomyelin (SPM) (Van Paridon et al., 1987b; De Vries et al., 1995). At this point it is still questionable whether these proteins actually transfer these phospholipids between membranes within cells or whether they interact directly with enzymes like PI 4-kinases and PI-specific phospholipase A2 thereby providing these enzymes with substrates (Cunningham et al., 1995; Snoek et al., 1999). In analogy with observations in yeast it has also been proposed that by binding either PI, PC or SPM, PI-TPs act as regulators of phospholipid metabolism at distinct sites in the cell thereby fulfilling the roles of biosensors (McGee
- Published
- 2006
43. Structures of complement component C3 provide insights into the function and evolution of immunity
- Author
-
Anja Roos, H.C.A. Raaijmakers, Kristina Nilsson-Ekdahl, Bert J. C. Janssen, Bo Nilsson, Piet Gros, Mohamed R. Daha, Eric G. Huizinga, Kristal- en structuurchemie, Crystal and Structural Chemistry 1, and Dep Scheikunde
- Subjects
Models, Molecular ,animal diseases ,chemical and pharmacologic phenomena ,Complement C3-C5 Convertases ,Computational biology ,Biology ,Crystallography, X-Ray ,Models, Biological ,Complement C3c ,Evolution, Molecular ,Immune system ,Immunity ,Component (UML) ,Humans ,Complement Activation ,Multidisciplinary ,Complement C3 ,biochemical phenomena, metabolism, and nutrition ,Protein Structure, Tertiary ,Complement (complexity) ,Complement system ,Biochemistry ,International ,bacteria ,Function (biology) - Abstract
The mammalian complement system is a phylogenetically ancient cascade system that has a major role in innate and adaptive immunity. Activation of component C3 (1,641 residues) is central to the three complement pathways and results in inflammation and elimination of self and non-self targets. Here we present crystal structures of native C3 and its final major proteolytic fragment C3c. The structures reveal thirteen domains, nine of which were unpredicted, and suggest that the proteins of the alpha2-macroglobulin family evolved from a core of eight homologous domains. A double mechanism prevents hydrolysis of the thioester group, essential for covalent attachment of activated C3 to target surfaces. Marked conformational changes in the alpha-chain, including movement of a critical interaction site through a ring formed by the domains of the beta-chain, indicate an unprecedented, conformation-dependent mechanism of activation, regulation and biological function of C3.
- Published
- 2005
44. Crystal structure of an Anti-meningococcal subtype P1.4 PorA antibody provides basis for peptide-vaccine design
- Author
-
Loek van Alphen, Peter Hoogerhout, Gestur Vidarsson, Clasien J. Oomen, Betsy Kuipers, Piet Gros, and Landsteiner Laboratory
- Subjects
Models, Molecular ,Protein Conformation ,Molecular Sequence Data ,Molecular Conformation ,Porins ,Enzyme-Linked Immunosorbent Assay ,Meningococcal Vaccines ,Peptide ,Neisseria meningitidis ,Crystallography, X-Ray ,Epitope ,Turn (biochemistry) ,Epitopes ,Mice ,Protein structure ,X-Ray Diffraction ,Antigen ,Structural Biology ,Animals ,Humans ,Cloning, Molecular ,Immunoglobulin Fragments ,Molecular Biology ,chemistry.chemical_classification ,Mice, Inbred BALB C ,Vaccines ,biology ,Sequence Analysis, DNA ,Molecular biology ,Cyclic peptide ,Meningococcal Infections ,chemistry ,biology.protein ,Peptide vaccine ,Antibody ,Peptides ,Software ,Protein Binding - Abstract
In various western countries, subtype P1.4 of Neisseria meningitidis serogroup B causes the greatest incidence of meningococcal disease. To investigate the molecular recognition of this subtype, we crystallised a peptide (P1HVVVNNKVATH(P11)), corresponding to the subtype P1.4 epitope sequence of outer membrane protein PorA, in complex with a Fab fragment of the bactericidal antibody MN20B9.34 directed against this epitope. Structure determination at 1.95 A resolution revealed a unique complex of one P1.4 antigen peptide bound to two identical Fab fragments. One Fab recognises the putative epitope residues in a 2:2 type I beta-turn at residues P5NNKV(P8), whereas the other Fab binds the C-terminal residues of the peptide that we consider a crystallisation artefact. Interestingly, recognition of the P1.4 epitope peptide is mediated almost exclusively through the complementarity-determining regions of the heavy chain. We exploited the observed turn conformation for designing conformationally restricted cyclic peptides for use as a peptide vaccine. The conformational stability of the two peptide designs was assessed by molecular dynamics simulations. Unlike the linear peptide, both cyclic peptides, conjugated to tetanus toxoid as a carrier protein, elicited antibody responses in mice that recognised meningococci of subtype P1.7-2,4. Serum bactericidal assays showed that some, but not all, of the sera induced with the cyclic peptide conjugates could activate the complement system with titres that were very high compared to the titres induced by complete PorA protein in its native conformation administered in outer membrane vesicles.
- Published
- 2005
45. The potentials of conditional optimization in phasing and model building of protein crystal structures
- Author
-
Sjors H.W. Scheres and Piet Gros
- Subjects
Models, Molecular ,Diffraction ,Protein Conformation ,Computer science ,Ab initio ,Proteins ,General Medicine ,Crystallography, X-Ray ,Phaser ,Force field (chemistry) ,Crystallography ,Structural Biology ,Bundle ,von Willebrand Factor ,Atomic model ,Discrete modelling ,Model building ,Algorithm ,Bacterial Outer Membrane Proteins - Abstract
Model building is a pivotal step in protein-structure determination, because with an atomic model available the vast amount of geometrical prior knowledge may be applied to the structure-determination process. Here, conditional optimization, a method that does not require interpretation of the electron-density map, is described. Instead, this method refines loose atoms for which all chemical interpretations are considered simultaneously using an N-particle formalism. This method bears the potential of introducing the geometrical data much earlier in the structure-determination process, i.e. well before an interpretable electron-density map has been obtained. Here, results from two tests are presented: automated model building of three proteins with diffraction data extending to 2.4-3.0 A resolution and ab initio phasing of a small four-helical bundle with diffraction data to 2.0 A resolution. Models built automatically by the widely used programs ARP/wARP and RESOLVE and those from conditional optimization per se, without discrete modelling steps, had comparable phase quality and completeness, except in loop regions, which are poorly modelled by the current force field in conditional optimization. Optimization of multiple random starting models by conditional optimization yielded models revealing the four helices of the four-helical bundle.
- Published
- 2004
46. Novel proteases: common themes and surprising features
- Author
-
Piet Gros, Lucy Vandeputte-Rutten, Kristal- en structuurchemie, Universiteit Utrecht, and Dep Scheikunde
- Subjects
Models, Molecular ,Proteases ,Molecular Structure ,Protein Conformation ,Intracellular protein ,Protozoan Proteins ,Biology ,Pathogenicity ,Protein Structure, Tertiary ,C5-convertase ,Deubiquitinating enzyme ,Cell biology ,Fungal Proteins ,Bacterial Proteins ,Proteasome ,Structural biology ,Structural Biology ,Endopeptidases ,biology.protein ,Animals ,Peptide degradation ,Molecular Biology - Abstract
Proteases perform a wide variety of functions, inside and outside cells, regulating many biological processes. Recent years have witnessed a number of significant advances in the structural biology of proteases, including aspects of intracellular protein and peptide degradation by self-compartmentalizing proteases, activation of proteases in proteolytic cascades of regulatory pathways, and mechanisms of microbial proteases in pathogenicity.
- Published
- 2002
47. Evidence for the formation of disulfide radicals in protein crystals upon X-ray irradiation
- Author
-
Maria L. Raves, Piet Gros, Sean McSweeney, Jacqueline Bergès, Chantal Houée-Levin, Israel Silman, Joel L. Sussman, Martin Weik, and Raimond B. G. Ravelli
- Subjects
Models, Molecular ,Nuclear and High Energy Physics ,Radiation ,Free Radicals ,Absorption spectroscopy ,Protein Conformation ,Chemistry ,Radical ,Proteins ,Dose-Response Relationship, Radiation ,Electrons ,law.invention ,Crystal ,Crystallography ,Protein structure ,X-Ray Diffraction ,law ,Humans ,Disulfides ,Irradiation ,Crystallization ,Protein crystallization ,Instrumentation ,Single crystal - Abstract
Irradiation of proteins with intense X-ray radiation produced by third-generation synchrotron sources generates specific structural and chemical alterations, including breakage of disulfide bonds and decarboxylation. In this paper, disulfide bond lengths in irradiated crystals of the enzyme Torpedo californica acetylcholinesterase are examined based on quantum simulations and on experimental data published previously. The experimental data suggest that one disulfide bond elongates by approximately 0.7 A upon X-ray irradiation as seen in a series of nine data sets collected on a single crystal. Simulation of the same bond suggests elongation by a similar value if a disulfide-radical anion is formed by trapping an electron. The absorption spectrum of a crystal irradiated under similar conditions shows a peak at approximately 400 nm, which in aqueous solution has been attributed to disulfide radicals. The results suggest that the formation of disulfide radicals in protein crystals owing to X-ray irradiation can be observed experimentally, both by structural means and by absorption spectroscopy.
- Published
- 2002
48. Lipopolysaccharide regions involved in the activation of Escherichia coli outer membrane protease OmpT
- Author
-
Klaus Brandenburg, Niek Dekker, R.Arjen Kramer, Marjolein Werkhoven, Piet Gros, Maarten R. Egmond, and Lucy Vandeputte-Rutten
- Subjects
Conformational change ,Protease ,biology ,Chemistry ,medicine.medical_treatment ,Omptin ,Active site ,medicine.disease_cause ,Biochemistry ,OmpT ,Lipid A ,medicine ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Bacterial outer membrane ,Escherichia coli - Abstract
OmpT is an integral outer membrane protease of Escherichia coli. Overexpression of OmpT in E. coli and subsequent in vitro folding of the produced inclusion bodies yielded protein with a native-like structure. However, enzymatically active protease was only obtained after addition of the outer membrane lipid lipopolysaccharide (LPS). OmpT is the first example of an enzyme that requires LPS for activity. In this study, we investigated the nature of this activation. Circular dichroism analysis showed that binding of LPS did not lead to large structural changes. Titration of OmpT with LPS and determining the resulting OmpT activity with a fluorimetric assay yielded a dissociation constant of 10-4 m for E. coli K-12 LPS. Determining the dissociation constants for different LPS chemotypes revealed that a fully acylated lipid A part is minimally required for activation of OmpT. The heptose-bound phosphates in the inner core region were also important for activation. The affinity for LPS was not dependent on the concentration of substrate, neither was affinity for the substrate influenced by the concentration of LPS. This indicated that LPS most likely does not act at the level of substrate binding. We hypothesize that LPS induces a subtle conformational change in the protein that is required for obtaining a native active site geometry.
- Published
- 2002
49. Functional characterization of alternative pathway C3 and C5 convertase inhibitors
- Author
-
Maartje Ruyken, Mihály Józsi, Ronald D. Gorham, Suzan H.M. Rooijakkers, Seline A. Zwarthoff, Piet Gros, and Xiaoguang Xue
- Subjects
Chemistry ,Immunology ,Alternative complement pathway ,Molecular Biology ,C5-convertase ,Cell biology - Published
- 2017
50. Abstract 33: The binding mode of the bispecific anti-HER2xHER3 antibody MCLA-128 is responsible for its potent inhibition of HRG-driven tumorigenesis
- Author
-
Ton Logtenberg, Camilla De Nardis, Mark Throsby, Lex Bakker, Cecile Geuijen, Eric Rovers, John de Kruif, Robert Doornbos, Piet Gros, Carina Bartelink-Clements, David Andre Baptiste Maussang-Detaille, Linda Johanna Aleida Hendriks, and Tristan Gallenne
- Subjects
Cancer Research ,Biodistribution ,biology ,medicine.drug_class ,Chemistry ,Alanine scanning ,Monoclonal antibody ,medicine.disease_cause ,Molecular biology ,Epitope ,In vitro ,Oncology ,In vivo ,medicine ,biology.protein ,Antibody ,skin and connective tissue diseases ,Carcinogenesis - Abstract
Introduction: MCLA-128 is as an ADCC-enhanced IgG1 bispecific antibody that targets the HER2:HER3 dimer and is currently being tested in Phase I/II clinical trials. MCLA-128 demonstrates an in vitro potency superior to other anti-HER2 and anti-HER3 antibodies in cells stimulated with high concentrations of heregulin (HRG) thereby overcoming one of the resistance mechanisms of current HER2 therapies. This study investigates the binding mode of MCLA-128 and proof of concept studies in HRG-driven tumor models. Methods: Alanine scanning shotgun mutagenesis was used to map the epitopes of MCLA-128 to HER2 and HER3. Fab fragments of MCLA-128 were crystallized with the soluble extracellular domains of HER2 and HER3. SAXS analysis on the HER2-HER3-MCLA-128 complex was performed to investigate the binding mode of the bispecific antibody in solution. Ligand-induced dimer specificity was investigated with PathHunter® heterodimerization assays. Bispecific anti-HER2xHER3 antibody and its parental anti-HER3 monoclonal antibody were labelled with 64Cu to compare their biodistribution profiles. The efficacy of MCLA-128 in HRG-driven systems was shown in vitro in MDA-MB-175 cells and in vivo in an orthotopic intracranial patient-derived xenograft (PDX) model originating from a breast cancer brain metastasis Results: The shotgun mutagenesis study identified that the bispecific antibody MCLA-128 binds amino acids T144, R166, R181 in HER2 domain I and R426 in HER3 domain III. Crystallographic studies confirmed the involvement of these critical residues and suggested that MCLA-128 locks the HER3 receptor in its ligand-unbound inactive confirmation. SAXS analysis suggests that the bispecific antibody MCLA-128 forms inter-dimer rather than intra-dimer interactions. In vitro, MCLA-128 specifically blocked HRG-induced signaling of HER2:HER3 but not HER2:HER4 heterodimers. Biodistribution of MCLA-128 in a xenograft model of breast cancer showed that the penetration of MCLA-128 in JIMT-1 HER2-amplified tumors is HER2-dependent despite the high affinity of the HER3 Fab arm for its receptor. MCLA-128 efficiently blocked tumor growth of the HRG-driven HER2 (1+) breast cancer cell line MDA-MB-175 in 3D in vitro. Treatment of orthotopically transplanted HER2-amplified breast cancer brain tumors in mice led to 100% survival with MCLA-128, in contrast to 38% and 0% survival in T-DM1 and vehicle treated mice respectively. Conclusion: MCLA-128 targets HER2-positive tumors via its HER2 arm and locks HER3 in an inactive confirmation. The potent anti-proliferative activity of MCLA-128 in vitro and in vivo supports the clinical development of this bispecific HER2xHER3 antibody in HRG-driven tumors. Citation Format: David Maussang-Detaille, Camilla de Nardis, Linda Hendriks, Carina Bartelink-Clements, Eric Rovers, Tristan Gallenne, Robert Doornbos, Lex Bakker, John de Kruif, Ton Logtenberg, Piet Gros, Cecile Geuijen, Mark Throsby. The binding mode of the bispecific anti-HER2xHER3 antibody MCLA-128 is responsible for its potent inhibition of HRG-driven tumorigenesis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 33. doi:10.1158/1538-7445.AM2017-33
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.