90 results on '"Ruby H. P. Law"'
Search Results
2. The first transmembrane region of complement component-9 acts as a brake on its self-assembly
- Author
-
Bradley A. Spicer, Ruby H. P. Law, Tom T. Caradoc-Davies, Sue M. Ekkel, Charles Bayly-Jones, Siew-Siew Pang, Paul J. Conroy, Georg Ramm, Mazdak Radjainia, Hariprasad Venugopal, James C. Whisstock, and Michelle A. Dunstone
- Subjects
Science - Abstract
The Complement component 9 (C9) is the pore-forming component of the Membrane Attack Complex which targets pathogens. Here authors use structural biology to compare monomeric C9 to C9 within the polymeric assembly and identify the element which inhibits C9 self-assembly in the absence of the target membrane.
- Published
- 2018
- Full Text
- View/download PDF
3. Structure of the poly-C9 component of the complement membrane attack complex
- Author
-
Natalya V. Dudkina, Bradley A. Spicer, Cyril F. Reboul, Paul J. Conroy, Natalya Lukoyanova, Hans Elmlund, Ruby H. P. Law, Susan M. Ekkel, Stephanie C. Kondos, Robert J. A. Goode, Georg Ramm, James C. Whisstock, Helen R. Saibil, and Michelle A. Dunstone
- Subjects
Science - Abstract
The membrane attack complex is a heteromeric assembly of complement proteins where multiple copies of C9 are recruited by the C5b678 complex to form lytic pores in pathogen membranes. Here the authors present the structure of a soluble pore-like form of the C9 component that reveals details of the oligomerization interfaces.
- Published
- 2016
- Full Text
- View/download PDF
4. The role of NINJ1 protein in programmed cellular destruction
- Author
-
James C. Whisstock and Ruby H. P. Law
- Subjects
Multidisciplinary - Published
- 2023
- Full Text
- View/download PDF
5. Front Cover: Synthesis and Structural Characterization of Macrocyclic Plasmin Inhibitors (ChemMedChem 6/2023)
- Author
-
Simon J. A. Wiedemeyer, Guojie Wu, T. L. Phuong Pham, Heike Lang‐Henkel, Benjamin Perez Urzua, James C Whisstock, Ruby H. P. Law, and Torsten Steinmetzer
- Subjects
Pharmacology ,Organic Chemistry ,Drug Discovery ,Molecular Medicine ,General Pharmacology, Toxicology and Pharmaceutics ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
6. Synthesis and Structural Characterization of Macrocyclic Plasmin Inhibitors
- Author
-
Simon J. A. Wiedemeyer, Guojie Wu, T. L. Phuong Pham, Heike Lang‐Henkel, Benjamin Perez Urzua, James C Whisstock, Ruby H. P. Law, and Torsten Steinmetzer
- Subjects
Pharmacology ,Organic Chemistry ,Drug Discovery ,Molecular Medicine ,General Pharmacology, Toxicology and Pharmaceutics ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
7. A mechanism for hereditary angioedema caused by a lysine 311–to–glutamic acid substitution in plasminogen
- Author
-
S. Kent Dickeson, Sunil Kumar, Mao-Fu Sun, Bassem M. Mohammed, Dennis R. Phillips, James C. Whisstock, Adam J. Quek, Edward P. Feener, Ruby H. P. Law, and David Gailani
- Subjects
Mammals ,Kininogens ,Fibrinolysis ,Lysine ,Immunology ,Angioedemas, Hereditary ,Glutamic Acid ,Plasminogen ,Cell Biology ,Hematology ,Factor XIIa ,Bradykinin ,Biochemistry ,Thrombosis and Hemostasis ,Mice ,Tissue Plasminogen Activator ,Animals ,Humans ,Fibrinolysin ,Plasma Kallikrein ,circulatory and respiratory physiology - Abstract
Patients with hereditary angioedema (HAE) experience episodes of bradykinin (BK)-induced swelling of skin and mucosal membranes. The most common cause is reduced plasma activity of C1 inhibitor, the main regulator of the proteases plasma kallikrein (PKa) and factor XIIa (FXIIa). Recently, patients with HAE were described with a Lys311 to glutamic acid substitution in plasminogen (Plg), the zymogen of the protease plasmin (Plm). Adding tissue plasminogen activator to plasma containing Plg-Glu311 vs plasma containing wild-type Plg (Plg-Lys311) results in greater BK generation. Similar results were obtained in plasma lacking prekallikrein or FXII (the zymogens of PKa and FXIIa) and in normal plasma treated with a PKa inhibitor, indicating Plg-Glu311 induces BK generation independently of PKa and FXIIa. Plm-Glu311 cleaves high and low molecular weight kininogens (HK and LK, respectively), releasing BK more efficiently than Plm-Lys311. Based on the plasma concentrations of HK and LK, the latter may be the source of most of the BK generated by Plm-Glu311. The lysine analog ε-aminocaproic acid blocks Plm-catalyzed BK generation. The Glu311 substitution introduces a lysine-binding site into the Plg kringle 3 domain, perhaps altering binding to kininogens. Plg residue 311 is glutamic acid in most mammals. Glu311 in patients with HAE, therefore, represents reversion to the ancestral condition. Substantial BK generation occurs during Plm-Glu311 cleavage of human HK, but not mouse HK. Furthermore, mouse Plm, which has Glu311, did not liberate BK from human kininogens more rapidly than human Plg-Lys311. This indicates Glu311 is pathogenic in the context of human Plm when human kininogens are the substrates.
- Published
- 2022
8. Application and Structural Analysis of Triazole‐Bridged Disulfide Mimetics in Cyclic Peptides
- Author
-
Gordon J. King, David J. Craik, Kuok Yap, Andrew M. White, Peta J. Harvey, Guojie Wu, Ruby H. P. Law, Simon J. de Veer, Joakim E. Swedberg, Conan K. Wang, and Thomas Durek
- Subjects
Peptidomimetic ,Stereochemistry ,Triazole ,Peptide ,Crystallography, X-Ray ,010402 general chemistry ,Peptides, Cyclic ,01 natural sciences ,Ruthenium ,Catalysis ,chemistry.chemical_compound ,medicine ,Molecule ,Amino Acid Sequence ,Disulfides ,Nuclear Magnetic Resonance, Biomolecular ,chemistry.chemical_classification ,010405 organic chemistry ,Molecular Mimicry ,Disulfide bond ,General Medicine ,General Chemistry ,Triazoles ,Trypsin ,Cycloaddition ,Cyclic peptide ,0104 chemical sciences ,chemistry ,Cyclization ,medicine.drug - Abstract
Ruthenium-catalysed azide-alkyne cycloaddition (RuAAC) provides access to 1,5-disubstituted 1,2,3-triazole motifs in peptide engineering applications. However, investigation of this motif as a disulfide mimetic in cyclic peptides has been limited, and the structural consequences remain to be studied. We report synthetic strategies to install various triazole linkages into cyclic peptides through backbone cyclisation and RuAAC cross-linking reactions. These linkages were evaluated in four serine protease inhibitors based on sunflower trypsin inhibitor-1. NMR and X-ray crystallography revealed exceptional consensus of bridging distance and backbone conformations (RMSD
- Published
- 2020
- Full Text
- View/download PDF
9. Effective targeting of intact and proteolysed CDCP1 for imaging and treatment of pancreatic ductal adenocarcinoma
- Author
-
Stephen E. Rose, Chao Li, Paul Thomas, James C. Whisstock, Kamil A. Sokolowski, Simon Puttick, T. Cuda, Elena I. Deryugina, James P. Quigley, Cameron Snell, David Wyld, Ashleigh Parkin, Tashbib Khan, Ruby H. P. Law, Thomas Kryza, Marina Pajic, Andrew D. Riddell, Brian W.C. Tse, Madeline Gough, Yaowu He, John D. Hooper, Nicholas Lyons, Andrew Barbour, and Julia Yin
- Subjects
0301 basic medicine ,theranostics ,endocrine system diseases ,CDCP1 ,pancreatic cancer ,PET-CT ,Medicine (miscellaneous) ,Context (language use) ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Antigens, Neoplasm ,Pancreatic cancer ,Cell Line, Tumor ,Biomarkers, Tumor ,Medicine ,Animals ,Humans ,Precision Medicine ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,business.industry ,Cancer ,medicine.disease ,digestive system diseases ,3. Good health ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,Proteolysis ,Cancer research ,Disease Progression ,monoclonal-antibody ,Signal transduction ,business ,Cell Adhesion Molecules ,Immunostaining ,Research Paper ,Carcinoma, Pancreatic Ductal - Abstract
Background: CUB domain-containing protein 1 (CDCP1) is a cell surface receptor regulating key signalling pathways in malignant cells. CDCP1 has been proposed as a molecular target to abrogate oncogenic signalling pathways and specifically deliver anti-cancer agents to tumors. However, the development of CDCP1-targeting agents has been questioned by its frequent proteolytic processing which was thought to result in shedding of the CDCP1 extracellular domain limiting its targetability. In this study, we investigated the relevance of targeting CDCP1 in the context of pancreatic ductal adenocarcinoma (PDAC) and assess the impact of CDCP1 proteolysis on the effectiveness of CDCP1 targeting agents. Methods: The involvement of CDCP1 in PDAC progression was assessed by association analysis in several PDAC cohorts and the proteolytic processing of CDCP1 was evaluated in PDAC cell lines and patient-derived cells. The consequences of CDCP1 proteolysis on its targetability in PDAC cells was assessed using immunoprecipitation, immunostaining and biochemical assays. The involvement of CDCP1 in PDAC progression was examined by loss-of-function in vitro and in vivo experiments employing PDAC cells expressing intact or cleaved CDCP1. Finally, we generated antibody-based imaging and therapeutic agents targeting CDCP1 to demonstrate the feasibility of targeting this receptor for detection and treatment of PDAC tumors. Results: High CDCP1 expression in PDAC is significantly associated with poorer patient survival. In PDAC cells proteolysis of CDCP1 does not always result in the shedding of CDCP1-extracellular domain which can interact with membrane-bound CDCP1 allowing signal transduction between the different CDCP1-fragments. Targeting CDCP1 impairs PDAC cell functions and PDAC tumor growth independently of CDCP1 cleavage status. A CDCP1-targeting antibody is highly effective at delivering imaging radionuclides and cytotoxins to PDAC cells allowing specific detection of tumors by PET/CT imaging and superior anti-tumor effects compared to gemcitabine in in vivo models. Conclusion: Independent of its cleavage status, CDCP1 exerts oncogenic functions in PDAC and has significant potential to be targeted for improved radiological staging and treatment of this cancer. Its elevated expression by most PDAC tumors and lack of expression by normal pancreas and other major organs, suggest that targeting CDCP1 could benefit a significant proportion of PDAC patients. These data support the further development of CDCP1-targeting agents as personalizable tools for effective imaging and treatment of PDAC.
- Published
- 2020
10. Anti-CDCP1 immuno-conjugates for detection and inhibition of ovarian cancer
- Author
-
Paul J. Conroy, Kamil A. Sokolowski, Simon Puttick, S. John Weroha, Thomas Kryza, James C. Whisstock, Brittney S. Harrington, Samantha J. Stehbens, Paul Haluska, T. Cuda, Rohan Lourie, Sarah Reed, Brian W.C. Tse, Buddhika J. Arachchige, Lewis Perrin, Yaowu He, Tashbib Khan, Katherine K. Robbins, Ruby H. P. Law, Carlos Salomon, Pamela M. Pollock, and John D. Hooper
- Subjects
0301 basic medicine ,Immunoconjugates ,CDCP1 ,Transplantation, Heterologous ,Medicine (miscellaneous) ,Flow cytometry ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Western blot ,Antigens, Neoplasm ,Cell Movement ,In vivo ,antibody ,Cell Line, Tumor ,medicine ,Animals ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Ovarian Neoplasms ,Radioisotopes ,biology ,medicine.diagnostic_test ,Chemistry ,Cell Membrane ,Membrane Proteins ,Cancer ,Cell migration ,Surface Plasmon Resonance ,medicine.disease ,3. Good health ,ovarian cancer ,src-Family Kinases ,030104 developmental biology ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Models, Animal ,biology.protein ,Cancer research ,immuno-conjugate ,Female ,Zirconium ,Antibody ,Ovarian cancer ,Cell Adhesion Molecules ,Research Paper - Abstract
CUB-domain containing protein 1 (CDCP1) is a cancer associated cell surface protein that amplifies pro-tumorigenic signalling by other receptors including EGFR and HER2. Its potential as a cancer target is supported by studies showing that anti-CDCP1 antibodies inhibit cell migration and survival in vitro, and tumor growth and metastasis in vivo. Here we characterize two anti-CDCP1 antibodies, focusing on immuno-conjugates of one of these as a tool to detect and inhibit ovarian cancer. Methods: A panel of ovarian cancer cell lines was examined for cell surface expression of CDCP1 and loss of expression induced by anti-CDCP1 antibodies 10D7 and 41-2 using flow cytometry and Western blot analysis. Surface plasmon resonance analysis and examination of truncation mutants was used to analyse the binding properties of the antibodies for CDCP1. Live-cell spinning-disk confocal microscopy of GFP-tagged CDCP1 was used to track internalization and intracellular trafficking of CDCP1/antibody complexes. In vivo, zirconium 89-labelled 10D7 was detected by positron-emission tomography imaging, of an ovarian cancer patient-derived xenograft grown intraperitoneally in mice. The efficacy of cytotoxin-conjugated 10D7 was examined against ovarian cancer cells in vitro and in vivo. Results: Our data indicate that each antibody binds with high affinity to the extracellular domain of CDCP1 causing rapid internalization of the receptor/antibody complex and degradation of CDCP1 via processes mediated by the kinase Src. Highlighting the potential clinical utility of CDCP1, positron-emission tomography imaging, using zirconium 89-labelled 10D7, was able to detect subcutaneous and intraperitoneal xenograft ovarian cancers in mice, including small (diameter
- Published
- 2020
- Full Text
- View/download PDF
11. Stability of the octameric structure affects plasminogen-binding capacity of streptococcal enolase.
- Author
-
Amanda J Cork, Daniel J Ericsson, Ruby H P Law, Lachlan W Casey, Eugene Valkov, Carlo Bertozzi, Anna Stamp, Blagojce Jovcevski, J Andrew Aquilina, James C Whisstock, Mark J Walker, and Bostjan Kobe
- Subjects
Medicine ,Science - Abstract
Group A Streptococcus (GAS) is a human pathogen that has the potential to cause invasive disease by binding and activating human plasmin(ogen). Streptococcal surface enolase (SEN) is an octameric α-enolase that is localized at the GAS cell surface. In addition to its glycolytic role inside the cell, SEN functions as a receptor for plasmin(ogen) on the bacterial surface, but the understanding of the molecular basis of plasmin(ogen) binding is limited. In this study, we determined the crystal and solution structures of GAS SEN and characterized the increased plasminogen binding by two SEN mutants. The plasminogen binding ability of SENK312A and SENK362A is ~2- and ~3.4-fold greater than for the wild-type protein. A combination of thermal stability assays, native mass spectrometry and X-ray crystallography approaches shows that increased plasminogen binding ability correlates with decreased stability of the octamer. We propose that decreased stability of the octameric structure facilitates the access of plasmin(ogen) to its binding sites, leading to more efficient plasmin(ogen) binding and activation.
- Published
- 2015
- Full Text
- View/download PDF
12. Model for surface-dependent factor XII activation: the roles of factor XII heavy chain domains
- Author
-
Aleksandr Shamanaev, Ivan Ivanov, Mao-Fu Sun, Maxim Litvak, Priyanka Srivastava, Bassem M. Mohammed, Rabia Shaban, Ashoka Maddur, Ingrid M. Verhamme, Owen J. T. McCarty, Ruby H. P. Law, and David Gailani
- Subjects
Lysine ,Aminocaproic Acid ,Factor XII ,Prekallikrein ,Hematology ,Blood Coagulation ,Fibronectins - Abstract
Factor XII (FXII) is the zymogen of a plasma protease (FXIIa) that contributes to bradykinin generation by converting prekallikrein to the protease plasma kallikrein (PKa). FXII conversion to FXIIa by autocatalysis or PKa-mediated cleavage is enhanced when the protein binds to negatively charged surfaces such as polymeric orthophosphate. FXII is composed of noncatalytic (heavy chain) and catalytic (light chain) regions. The heavy chain promotes FXII surface-binding and surface-dependent activation but restricts activation when FXII is not surface bound. From the N terminus, the heavy chain contains fibronectin type 2 (FN2), epidermal growth factor-1 (EGF1), fibronectin type 1 (FN1), EGF2, and kringle (KNG) domains and a proline-rich region. It shares this organization with its homolog, pro–hepatocyte growth factor activator (Pro-HGFA). To study the importance of heavy chain domains in FXII function, we prepared FXII with replacements of each domain with corresponding Pro-HGFA domains and tested them in activation and activity assays. EGF1 is required for surface-dependent FXII autoactivation and surface-dependent prekallikrein activation by FXIIa. KNG and FN2 are important for limiting FXII activation in the absence of a surface by a process that may require interactions between a lysine/arginine binding site on KNG and basic residues elsewhere on FXII. This interaction is disrupted by the lysine analog ε-aminocaproic acid. A model is proposed in which an ε-aminocaproic acid–sensitive interaction between the KNG and FN2 domains maintains FXII in a conformation that restricts activation. Upon binding to a surface through EGF1, the KNG/FN2-dependent mechanism is inactivated, exposing the FXII activation cleavage site.
- Published
- 2021
13. The cryo-EM structure of the acid activatable pore-forming immune effector Macrophage-expressed gene 1
- Author
-
Phillip I. Bird, Bart W. Hoogenboom, Adrian W. Hodel, Georg Ramm, Susan M. Ekkel, Hariprasad Venugopal, Charles Bayly-Jones, Mazdak Radjainia, Ilia Voskoboinik, Ruby H. P. Law, Paul J. Conroy, Yann Gambin, Siew Siew Pang, Michelle A. Dunstone, Emma Sierecki, Bradley A. Spicer, Edward S. Parsons, and James C. Whisstock
- Subjects
Pore Forming Cytotoxic Proteins ,0301 basic medicine ,030103 biophysics ,Cryo-electron microscopy ,Science ,Immunology ,Protein domain ,General Physics and Astronomy ,Microscopy, Atomic Force ,Phagolysosome ,Article ,Protein Structure, Secondary ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Protein structure ,Protein Domains ,Humans ,lcsh:Science ,Liposome ,Multidisciplinary ,Bacteria ,Chemistry ,Macrophages ,Cell Membrane ,Cryoelectron Microscopy ,Membrane Proteins ,General Chemistry ,030104 developmental biology ,Membrane ,Membrane protein ,Phagolysosome membrane ,Liposomes ,Biophysics ,lcsh:Q ,Lysosomes - Abstract
Macrophage-expressed gene 1 (MPEG1/Perforin-2) is a perforin-like protein that functions within the phagolysosome to damage engulfed microbes. MPEG1 is thought to form pores in target membranes, however, its mode of action remains unknown. We use cryo-Electron Microscopy (cryo-EM) to determine the 2.4 Å structure of a hexadecameric assembly of MPEG1 that displays the expected features of a soluble prepore complex. We further discover that MPEG1 prepore-like assemblies can be induced to perforate membranes through acidification, such as would occur within maturing phagolysosomes. We next solve the 3.6 Å cryo-EM structure of MPEG1 in complex with liposomes. These data reveal that a multi-vesicular body of 12 kDa (MVB12)-associated β-prism (MABP) domain binds membranes such that the pore-forming machinery of MPEG1 is oriented away from the bound membrane. This unexpected mechanism of membrane interaction suggests that MPEG1 remains bound to the phagolysosome membrane while simultaneously forming pores in engulfed bacterial targets., Macrophage-expressed gene 1 (MPEG1) functions within the phagolysosome to damage engulfed microbes, presumably via forming pores in target membranes. In order to provide insights into the mechanism of MPEG1 function and membrane binding, the authors present structures of hexadecameric MPEG1 prepores both in solution and in complex with liposomes.
- Published
- 2019
14. Structural studies of plasmin inhibition
- Author
-
James C. Whisstock, Guojie Wu, Ruby H. P. Law, Adam J. Quek, Blake A. Mazzitelli, Tom T. Caradoc-Davies, and Sue M. Ekkel
- Subjects
Plasmin ,medicine.medical_treatment ,Apoptosis ,Inflammation ,030204 cardiovascular system & hematology ,Pharmacology ,Biochemistry ,Plasminogen Activators ,03 medical and health sciences ,0302 clinical medicine ,Zymogen ,Antifibrinolytic agent ,Fibrinolysis ,medicine ,Animals ,Humans ,Protease Inhibitors ,Protease inhibitor (pharmacology) ,030304 developmental biology ,Serine protease ,0303 health sciences ,biology ,Chemistry ,fungi ,Plasminogen ,Antifibrinolytic Agents ,biology.protein ,medicine.symptom ,Wound healing ,Signal Transduction ,medicine.drug - Abstract
Plasminogen (Plg) is the zymogen form of the serine protease plasmin (Plm), and it plays a crucial role in fibrinolysis as well as wound healing, immunity, tissue remodeling and inflammation. Binding to the targets via the lysine-binding sites allows for Plg activation by plasminogen activators (PAs) present on the same target. Cellular uptake of fibrin degradation products leads to apoptosis, which represents one of the pathways for cross-talk between fibrinolysis and tissue remodeling. Therapeutic manipulation of Plm activity plays a vital role in the treatments of a range of diseases, whereas Plm inhibitors are used in trauma and surgeries as antifibrinolytic agents. Plm inhibitors are also used in conditions such as angioedema, menorrhagia and melasma. Here, we review the rationale for the further development of new Plm inhibitors, with a particular focus on the structural studies of the active site inhibitors of Plm. We compare the binding mode of different classes of inhibitors and comment on how it relates to their efficacy, as well as possible future developments.
- Published
- 2019
- Full Text
- View/download PDF
15. Toward Better Understanding on How Group AStreptococcusManipulates Human Fibrinolytic System
- Author
-
James C. Whisstock, Ruby H. P. Law, and Adam J. Quek
- Subjects
Plasmin ,Streptococcus ,Chemistry ,Streptokinase ,medicine ,Hemolytic streptococcus ,medicine.disease_cause ,Group A ,medicine.drug ,Microbiology - Published
- 2020
- Full Text
- View/download PDF
16. Highly Potent and Selective Plasmin Inhibitors Based on the Sunflower Trypsin Inhibitor-1 Scaffold Attenuate Fibrinolysis in Plasma
- Author
-
Joakim E. Swedberg, David J. Craik, Ruby H. P. Law, Thomas Durek, Tunjung Mahatmanto, Tom T. Caradoc-Davies, Guojie Wu, and James C. Whisstock
- Subjects
Proteases ,Serine Proteinase Inhibitors ,Antifibrinolytic ,Plasmin ,medicine.drug_class ,medicine.medical_treatment ,Molecular Dynamics Simulation ,Pharmacology ,Crystallography, X-Ray ,Peptides, Cyclic ,01 natural sciences ,Serine ,03 medical and health sciences ,Drug Discovery ,Fibrinolysis ,medicine ,Humans ,Aprotinin ,Amino Acid Sequence ,Fibrinolysin ,030304 developmental biology ,Serine protease ,0303 health sciences ,Binding Sites ,biology ,Chemistry ,Trypsin ,0104 chemical sciences ,3. Good health ,010404 medicinal & biomolecular chemistry ,Drug Design ,biology.protein ,Molecular Medicine ,Peptides ,medicine.drug - Abstract
Antifibrinolytic drugs provide important pharmacological interventions to reduce morbidity and mortality from excessive bleeding during surgery and after trauma. Current drugs used for inhibiting the dissolution of fibrin, the main structural component of blood clots, are associated with adverse events due to lack of potency, high doses, and nonselective inhibition mechanisms. These drawbacks warrant the development of a new generation of highly potent and selective fibrinolysis inhibitors. Here, we use the 14-amino acid backbone-cyclic sunflower trypsin inhibitor-1 scaffold to design a highly potent (Ki = 0.05 nM) inhibitor of the primary serine protease in fibrinolysis, plasmin. This compound displays a million-fold selectivity over other serine proteases in blood, inhibits fibrinolysis in plasma more effectively than the gold-standard therapeutic inhibitor aprotinin, and is a promising candidate for development of highly specific fibrinolysis inhibitors with reduced side effects.
- Published
- 2018
- Full Text
- View/download PDF
17. A Cyclic Peptide Inhibitor of the iNOS–SPSB Protein–Protein Interaction as a Potential Anti-Infective Agent
- Author
-
Maiada M. Sadek, Nicholas Barlow, Eleanor W. W. Leung, Billy J. Williams-Noonan, Beow Keat Yap, Fairolniza Mohd Shariff, Tom T. Caradoc-Davies, Sandra E. Nicholson, David K. Chalmers, Philip E. Thompson, Ruby H. P. Law, and Raymond S. Norton
- Subjects
0301 basic medicine ,Intracellular Signaling Peptides and Proteins ,Nitric Oxide Synthase Type II ,Suppressor of Cytokine Signaling Proteins ,General Medicine ,Peptides, Cyclic ,Biochemistry ,Mice ,03 medical and health sciences ,RAW 264.7 Cells ,030104 developmental biology ,0302 clinical medicine ,Anti-Infective Agents ,Drug Design ,030220 oncology & carcinogenesis ,Animals ,Humans ,Molecular Medicine ,Oligopeptides ,Protein Binding - Abstract
SPRY domain- and SOCS box-containing proteins SPSB1, SPSB2, and SPSB4 interact with inducible nitric oxide synthase (iNOS), causing the iNOS to be polyubiquitinated and targeted for degradation. Inhibition of this interaction increases iNOS levels, and consequently cellular nitric oxide (NO) concentrations, and has been proposed as a potential strategy for killing intracellular pathogens. We previously described two DINNN-containing cyclic peptides (CP1 and CP2) as potent inhibitors of the murine SPSB-iNOS interaction. In this study, we report the crystal structures of human SPSB4 bound to CP1 and CP2 and human SPSB2 bound to CP2. We then used these structures to design a new inhibitor in which an intramolecular hydrogen bond was replaced with a hydrocarbon linkage to form a smaller macrocycle while maintaining the bound geometry of CP2 observed in the crystal structures. This resulting pentapeptide SPSB-iNOS inhibitor (CP3) has a reduced macrocycle ring size, fewer nonbinding residues, and includes additional conformational constraints. CP3 has a greater affinity for SBSB2 ( K
- Published
- 2018
- Full Text
- View/download PDF
18. Inhibition of a K9/K36 demethylase by an H3.3 point mutation found in paediatric glioblastoma
- Author
-
Wendi Lin, Ruby H. P. Law, Jeffrey R. Mann, Hsiao P.J. Voon, Partha Pratim Das, Linda Hii, Maheshi Udugama, David Steer, and Lee H. Wong
- Subjects
0301 basic medicine ,Science ,Mutant ,Glycine ,General Physics and Astronomy ,Arginine ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Histones ,Mice ,03 medical and health sciences ,medicine ,Animals ,Humans ,Point Mutation ,Biotinylation ,Transgenes ,Epigenetics ,Child ,lcsh:Science ,Embryonic Stem Cells ,Regulation of gene expression ,Mutation ,Multidisciplinary ,biology ,Brain Neoplasms ,Sequence Analysis, RNA ,Point mutation ,General Chemistry ,Chromatin ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Histone ,Cancer research ,biology.protein ,Demethylase ,lcsh:Q ,Glioblastoma ,Protein Binding - Abstract
An array of oncogenic histone point mutations have been identified across a number of different cancer studies. It has been suggested that some of these mutant histones can exert their effects by inhibiting epigenetic writers. Here, we report that the H3.3 G34R (glycine to arginine) substitution mutation, found in paediatric gliomas, causes widespread changes in H3K9me3 and H3K36me3 by interfering with the KDM4 family of K9/K36 demethylases. Expression of a targeted single-copy of H3.3 G34R at endogenous levels induced chromatin alterations that were comparable to a KDM4 A/B/C triple-knockout. We find that H3.3 G34R preferentially binds KDM4 while simultaneously inhibiting its enzymatic activity, demonstrating that histone mutations can act through inhibition of epigenetic erasers. These results suggest that histone point mutations can exert their effects through interactions with a range of epigenetic readers, writers and erasers., Recent studies have identified a number of oncogenic histone point mutations in different cancers. Here the authors provide evidence that H3.3 G34R substitution mutation, which is found in paediatric gliomas, causes changes in H3K9me3 and H3K36me3 by interfering with the KDM4 family of K9/K36 demethylases.
- Published
- 2018
19. The subtilisin-like protease AprV2 is required for virulence and uses a novel disulphide-tethered exosite to bind substrates.
- Author
-
Ruth M Kennan, Wilson Wong, Om P Dhungyel, Xiaoyan Han, David Wong, Dane Parker, Carlos J Rosado, Ruby H P Law, Sheena McGowan, Shane B Reeve, Vita Levina, Glenn A Powers, Robert N Pike, Stephen P Bottomley, A Ian Smith, Ian Marsh, Richard J Whittington, James C Whisstock, Corrine J Porter, and Julian I Rood
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Many bacterial pathogens produce extracellular proteases that degrade the extracellular matrix of the host and therefore are involved in disease pathogenesis. Dichelobacter nodosus is the causative agent of ovine footrot, a highly contagious disease that is characterized by the separation of the hoof from the underlying tissue. D. nodosus secretes three subtilisin-like proteases whose analysis forms the basis of diagnostic tests that differentiate between virulent and benign strains and have been postulated to play a role in virulence. We have constructed protease mutants of D. nodosus; their analysis in a sheep virulence model revealed that one of these enzymes, AprV2, was required for virulence. These studies challenge the previous hypothesis that the elastase activity of AprV2 is important for disease progression, since aprV2 mutants were virulent when complemented with aprB2, which encodes a variant that has impaired elastase activity. We have determined the crystal structures of both AprV2 and AprB2 and characterized the biological activity of these enzymes. These data reveal that an unusual extended disulphide-tethered loop functions as an exosite, mediating effective enzyme-substrate interactions. The disulphide bond and Tyr92, which was located at the exposed end of the loop, were functionally important. Bioinformatic analyses suggested that other pathogenic bacteria may have proteases that utilize a similar mechanism. In conclusion, we have used an integrated multidisciplinary combination of bacterial genetics, whole animal virulence trials in the original host, biochemical studies, and comprehensive analysis of crystal structures to provide the first definitive evidence that the extracellular secreted proteases produced by D. nodosus are required for virulence and to elucidate the molecular mechanism by which these proteases bind to their natural substrates. We postulate that this exosite mechanism may be used by proteases produced by other bacterial pathogens of both humans and animals.
- Published
- 2010
- Full Text
- View/download PDF
20. Characterisation of peptide microarrays for studying antibody-antigen binding using surface plasmon resonance imagery.
- Author
-
Claude Nogues, Hervé Leh, Christopher G Langendorf, Ruby H P Law, Ashley M Buckle, and Malcolm Buckle
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Non-specific binding to biosensor surfaces is a major obstacle to quantitative analysis of selective retention of analytes at immobilized target molecules. Although a range of chemical antifouling monolayers has been developed to address this problem, many macromolecular interactions still remain refractory to analysis due to the prevalent high degree of non-specific binding. We describe how we use the dynamic process of the formation of self assembling monolayers and optimise physical and chemical properties thus reducing considerably non-specific binding and allowing analysis of specific binding of analytes to immobilized target molecules. METHODOLOGY/PRINCIPAL FINDINGS: We illustrate this approach by the production of specific protein arrays for the analysis of interactions between the 65kDa isoform of human glutamate decarboxylase (GAD65) and a human monoclonal antibody. Our data illustrate that we have effectively eliminated non-specific interactions with the surface containing the immobilised GAD65 molecules. The findings have several implications. First, this approach obviates the dubious process of background subtraction and gives access to more accurate kinetic and equilibrium values that are no longer contaminated by multiphase non-specific binding. Second, an enhanced signal to noise ratio increases not only the sensitivity but also confidence in the use of SPR to generate kinetic constants that may then be inserted into van't Hoff type analyses to provide comparative DeltaG, DeltaS and DeltaH values, making this an efficient, rapid and competitive alternative to ITC measurements used in drug and macromolecular-interaction mechanistic studies. Third, the accuracy of the measurements allows the application of more intricate interaction models than simple Langmuir monophasic binding. CONCLUSIONS: The detection and measurement of antibody binding by the type 1 diabetes autoantigen GAD65 represents an example of an antibody-antigen interaction where good structural, mechanistic and immunological data are available. Using SPRi we were able to characterise the kinetics of the interaction in greater detail than ELISA/RIA methods. Furthermore, our data indicate that SPRi is well suited to a multiplexed immunoassay using GAD65 proteins, and may be applicable to other biomarkers.
- Published
- 2010
- Full Text
- View/download PDF
21. Prodepth: predict residue depth by support vector regression approach from protein sequences only.
- Author
-
Jiangning Song, Hao Tan, Khalid Mahmood, Ruby H P Law, Ashley M Buckle, Geoffrey I Webb, Tatsuya Akutsu, and James C Whisstock
- Subjects
Medicine ,Science - Abstract
Residue depth (RD) is a solvent exposure measure that complements the information provided by conventional accessible surface area (ASA) and describes to what extent a residue is buried in the protein structure space. Previous studies have established that RD is correlated with several protein properties, such as protein stability, residue conservation and amino acid types. Accurate prediction of RD has many potentially important applications in the field of structural bioinformatics, for example, facilitating the identification of functionally important residues, or residues in the folding nucleus, or enzyme active sites from sequence information. In this work, we introduce an efficient approach that uses support vector regression to quantify the relationship between RD and protein sequence. We systematically investigated eight different sequence encoding schemes including both local and global sequence characteristics and examined their respective prediction performances. For the objective evaluation of our approach, we used 5-fold cross-validation to assess the prediction accuracies and showed that the overall best performance could be achieved with a correlation coefficient (CC) of 0.71 between the observed and predicted RD values and a root mean square error (RMSE) of 1.74, after incorporating the relevant multiple sequence features. The results suggest that residue depth could be reliably predicted solely from protein primary sequences: local sequence environments are the major determinants, while global sequence features could influence the prediction performance marginally. We highlight two examples as a comparison in order to illustrate the applicability of this approach. We also discuss the potential implications of this new structural parameter in the field of protein structure prediction and homology modeling. This method might prove to be a powerful tool for sequence analysis.
- Published
- 2009
- Full Text
- View/download PDF
22. Perforin—A key (shaped) weapon in the immunological arsenal
- Author
-
Ruby H. P. Law, Ilia Voskoboinik, James C. Whisstock, Bradley A. Spicer, and Paul J. Conroy
- Subjects
Models, Molecular ,0301 basic medicine ,Immunological Synapses ,chemical and pharmacologic phenomena ,Crystallography, X-Ray ,Granzymes ,Pore forming protein ,03 medical and health sciences ,Immune system ,Protein Domains ,Animals ,Humans ,Cytotoxic T cell ,MACPF ,biology ,Perforin ,Models, Immunological ,Cell Biology ,Cell biology ,Granzyme B ,030104 developmental biology ,Granzyme ,biology.protein ,T-Lymphocytes, Cytotoxic ,Developmental Biology - Abstract
Cytotoxic lymphocytes play a key role in immune homeostasis through elimination of virally-infected and transformed target cells. They do this by employing the potent pore-forming protein, perforin, a molecule that permits cytotoxic proteases, such as granzyme B, to enter the target cell cytoplasm. The synergistic activities of perforin and granzymes bring about the destruction of target cells in a process that is now more clearly understood as a result of structural and cellular biology. These data are helping the development of new classes of immunosuppressive molecules for use in treating immune driven disease and in enhancing the success of transplant therapies. This review focuses on structural and biological aspects of perforin function.
- Published
- 2017
- Full Text
- View/download PDF
23. Regulation of perforin activation and pre‐synaptic toxicity through C‐terminal glycosylation
- Author
-
Omer Gilan, Colin M. House, Joseph A. Trapani, Ilia Voskoboinik, Ruby H. P. Law, Imran G House, James C. Whisstock, Mark A. Dawson, and Amelia Jean Brennan
- Subjects
0301 basic medicine ,Glycosylation ,Immunological Synapses ,chemical and pharmacologic phenomena ,Biology ,Cytoplasmic Granules ,Endoplasmic Reticulum ,Biochemistry ,Natural killer cell ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Secretion ,Molecular Biology ,MACPF ,Membrane Glycoproteins ,Perforin ,Endoplasmic reticulum ,Scientific Reports ,Degranulation ,Cell biology ,Killer Cells, Natural ,Granzyme B ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Proteolysis ,biology.protein ,Interleukin-2 ,Protein Processing, Post-Translational - Abstract
Perforin is a highly cytotoxic pore‐forming protein essential for immune surveillance by cytotoxic lymphocytes. Prior to delivery to target cells by exocytosis, perforin is stored in acidic secretory granules where it remains functionally inert. However, how cytotoxic lymphocytes remain protected from their own perforin prior to its export to secretory granules, particularly in the Ca2+‐rich endoplasmic reticulum, remains unknown. Here, we show that N‐linked glycosylation of the perforin C‐terminus at Asn549 within the endoplasmic reticulum inhibits oligomerisation of perforin monomers and thus protects the host cell from premature pore formation. Subsequent removal of this glycan occurs through proteolytic processing of the C‐terminus within secretory granules and is imperative for perforin activation prior to secretion. Despite evolutionary conservation of the C‐terminus, we found that processing is carried out by multiple proteases, which we attribute to the unstructured and exposed nature of the region. In sum, our studies reveal a post‐translational regulatory mechanism essential for maintaining perforin in an inactive state until its secretion from the inhibitory acidic environment of the secretory granule.
- Published
- 2017
- Full Text
- View/download PDF
24. X-ray crystal structure of plasmin with tranexamic acid–derived active site inhibitors
- Author
-
James C. Whisstock, Tom T. Caradoc-Davies, Yuko Tsuda, Koushi Hidaka, Raymond S. Norton, Paul J. Conroy, Eleanor W. W. Leung, Guojie Wu, Nigel Kirby, Adam J. Quek, Ruby H. P. Law, and Devadharshini Jeevarajah
- Subjects
0301 basic medicine ,Protease ,biology ,Plasmin ,Chemistry ,medicine.medical_treatment ,Active site ,Cell migration ,Hematology ,Small molecule ,Hemorrhagic disorder ,Thrombosis and Hemostasis ,03 medical and health sciences ,030104 developmental biology ,Biochemistry ,Zymogen ,medicine ,biology.protein ,Tyrosine ,circulatory and respiratory physiology ,medicine.drug - Abstract
The zymogen protease plasminogen and its active form plasmin perform key roles in blood clot dissolution, tissue remodeling, cell migration, and bacterial pathogenesis. Dysregulation of the plasminogen/plasmin system results in life-threatening hemorrhagic disorders or thrombotic vascular occlusion. Accordingly, inhibitors of this system are clinically important. Currently, tranexamic acid (TXA), a molecule that prevents plasminogen activation through blocking recruitment to target substrates, is the most widely used inhibitor for the plasminogen/plasmin system in therapeutics. However, TXA lacks efficacy on the active form of plasmin. Thus, there is a need to develop specific inhibitors that target the protease active site. Here we report the crystal structures of plasmin in complex with the novel YO (trans-4-aminomethylcyclohexanecarbonyl-l-tyrosine-n-octylamide) class of small molecule inhibitors. We found that these inhibitors form key interactions with the S1 and S3' subsites of the catalytic cleft. Here, the TXA moiety of the YO compounds inserts into the primary (S1) specificity pocket, suggesting that TXA itself may function as a weak plasmin inhibitor, a hypothesis supported by subsequent biochemical and biophysical analyses. Mutational studies reveal that F587 of the S' subsite plays a key role in mediating the inhibitor interaction. Taken together, these data provide a foundation for the future development of small molecule inhibitors to specifically regulate plasmin function in a range of diseases and disorders.
- Published
- 2017
- Full Text
- View/download PDF
25. Structure and Function Characterization of the a1a2 Motifs of Streptococcus pyogenes M Protein in Human Plasminogen Binding
- Author
-
Yue Yuan, Martina L. Sanderson-Smith, Adam J. Quek, Devadharshini Jeevarajah, James C. Whisstock, Ruby H. P. Law, Paul J. Conroy, Yetunde A. Ayinuola, Guojie Wu, Blake A. Mazzitelli, Eleanor W. W. Leung, Gordon J. Lloyd, Francis J. Castellino, and Tom T. Caradoc-Davies
- Subjects
Myeloma protein ,Streptococcus pyogenes ,Lysine ,Cell ,Amino Acid Motifs ,Peptide ,medicine.disease_cause ,Crystallography, X-Ray ,Article ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,Bacterial Proteins ,Protein Domains ,Structural Biology ,Cell surface receptor ,medicine ,Humans ,Amino Acid Sequence ,Molecular Biology ,Pathogen ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Chemistry ,Protein Stability ,Mutagenesis ,Plasminogen ,medicine.anatomical_structure ,Biochemistry ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Plasminogen (Plg)-binding M protein (PAM) is a group A streptococcal cell surface receptor that is crucial for bacterial virulence. Previous studies revealed that, by binding to the kringle 2 (KR2) domain of host Plg, the pathogen attains a proteolytic microenvironment on the cell surface that facilitates its dissemination from the primary infection site. Each of the PAM molecules in their dimeric assembly consists of two Plg binding motifs (called the a1 and a2 repeats). To date, the molecular interactions between the a1 repeat and KR2 have been structurally characterized, whereas the role of the a2 repeat is less well defined. Here, we report the 1.7-A x-ray crystal structure of KR2 in complex with a monomeric PAM peptide that contains both the a1 and a2 motifs. The structure reveals how the PAM peptide forms key interactions simultaneously with two KR2 via the high-affinity lysine isosteres within the a1a2 motifs. Further studies, through combined mutagenesis and functional characterization, show that a2 is a stronger KR2 binder than a1, suggesting that these two motifs may play discrete roles in mediating the final PAM-Plg assembly.
- Published
- 2019
26. The structure of Macrophage Expressed Gene-1, a phagolysosome immune effector that is activated upon acidification
- Author
-
Bart W. Hoogenboom, Siew Siew Pang, Mazdak Radjainia, Hariprasad Venugopal, Georg Ramm, Ruby H. P. Law, Yann Gambin, Emma Sierecki, Ilia Voskoboinik, Bradley A. Spicer, Michelle A. Dunstone, James C. Whisstock, Sue M. Ekkel, Phillip I. Bird, Charles Bayly-Jones, Paul J. Conroy, and Adrian W. Hodel
- Subjects
0303 health sciences ,Liposome ,biology ,Chemistry ,Endosome ,Phagolysosome ,03 medical and health sciences ,0302 clinical medicine ,Membrane ,Perforin ,Phagolysosome membrane ,Biophysics ,biology.protein ,Macrophage ,Complement membrane attack complex ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Macrophage Expressed Gene-1 (MPEG-1; also termed Perforin-2) is an endosomal / phagolysosomal perforin-like protein that is conserved across the metazoan kingdom and that functions within the phagolysosome to damage engulfed microbes. Like the Membrane Attack Complex and perforin, MPEG-1 has been postulated to form pores in target membranes, however, its mode of action remains to be established. We used single particle cryo-Electron Microscopy to determine the 2.4 Å structure of a hexadecameric assembly of MPEG-1 that displays the expected features of a soluble pre-pore complex. We further discovered that the MPEG-1 pre-pore-like assemblies can be induced to perforate membranes through mild acidification, such as would occur within maturing phagolysosomes. We next solved the 3.6 Å cryo-EM structure of MPEG-1 in complex with liposomes. Remarkably these data revealed that a C-terminal Multi-vesicular body of 12 kDa (MVB12)-associatedβ-prism (MABP) domain interacts with target membranes in a mode that positions the pore forming machinery of MPEG-1 to point away from the bound membrane. This unexpected mechanism of membrane interaction raises the intriguing possibility that MPEG-1 may be able to remain bound to the phagolysosome membrane while simultaneously forming pores in engulfed bacterial targets.
- Published
- 2019
- Full Text
- View/download PDF
27. Molecular pathogenesis of plasminogen Hakodate: the second Japanese family case of severe type I plasminogen deficiency manifested late-onset multi-organic chronic pseudomembranous mucositis
- Author
-
Naohiro Izumi, Tsukasa Osaki, Akitada Ichinose, Masayoshi Souri, Ruby H. P. Law, and Youngseok Song
- Subjects
Male ,Mucositis ,0301 basic medicine ,medicine.medical_specialty ,Pathology ,Plasmin ,medicine.medical_treatment ,Mutation, Missense ,030204 cardiovascular system & hematology ,Compound heterozygosity ,Fibrin ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Japan ,Internal medicine ,Ligneous conjunctivitis ,Fibrinolysis ,medicine ,Humans ,Missense mutation ,Enterocolitis, Pseudomembranous ,Aged ,biology ,business.industry ,Skin Diseases, Genetic ,Plasminogen ,Hematology ,Conjunctivitis ,medicine.disease ,030104 developmental biology ,Endocrinology ,Chronic Disease ,biology.protein ,RNA Splice Sites ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
A 64-year-old man first developed ligneous conjunctivitis at the age of 58 years after right pulmonary resection because of suspected cancer; otherwise, he had been healthy. Since then, he began to suffer from various forms of chronic pseudomembranous mucositis. Laboratory tests demonstrated that he had 7.8 % of plasminogen activity and 5.9 % of the normal antigen level. Thus, he was diagnosed as having severe type I plasminogen deficiency, making him the third case in Japan. DNA sequencing and PCR-restriction fragment length polymorphism analyses revealed that this patient was a compound heterozygote of a G-to-A missense mutation (G266E) in exon VIII and a g-to-a mutation at the obligatory splicing acceptor site in intron 12 (IVS12-1g>a). These two mutations were confirmed to be novel. Molecular modeling and splice site strength calculation predicted conformational disorder(s) for the Glu266 mutant and a drastic decrease in splicing efficiency for intron 12, respectively. Western blot analysis demonstrated that the patient contained a small amount of the normal-sized plasminogen protein. Mass spectrometric analysis of the patient's plasminogen revealed a peptide containing the wild-type Gly266 residue and no peptides with mutations at Glu266. However, he had never suffered from thrombosis. Low levels of fibrinogen/fibrin degradation products (FDP), D-dimer, and plasmin-α2-plasmin inhibitor complex clearly indicated a hypo-fibrinolytic condition. However, his plasma concentration of elastase-digested crosslinked FDPs was 4.8 U/mL, suggesting the presence of an on-going plasmin(ogen)-independent "alternative" fibrinolytic system, which may protect the patient from thrombosis. The patient has been free from recurrence of ligneous conjunctivitis for approximately 2.5 years.
- Published
- 2016
- Full Text
- View/download PDF
28. Structure of the poly-C9 component of the complement membrane attack complex
- Author
-
Robert J. A. Goode, Bradley A. Spicer, Michelle A. Dunstone, S.C. Kondos, Georg Ramm, James C. Whisstock, Susan M. Ekkel, Cyril F. Reboul, Natalya Lukoyanova, Natalya V Dudkina, Helen R. Saibil, Paul J. Conroy, Hans Elmlund, and Ruby H. P. Law
- Subjects
0301 basic medicine ,MAC assembly ,Models, Molecular ,Polymers ,Science ,General Physics and Astronomy ,Complement Membrane Attack Complex ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Humans ,Multidisciplinary ,Membrane insertion ,Molecular Structure ,Component (thermodynamics) ,Complement component 9 ,Cryoelectron Microscopy ,General Chemistry ,Protein superfamily ,Complement C9 ,3. Good health ,030104 developmental biology ,Membrane ,Biochemistry ,Perforin ,biology.protein ,Biophysics ,Complement membrane attack complex - Abstract
The membrane attack complex (MAC)/perforin-like protein complement component 9 (C9) is the major component of the MAC, a multi-protein complex that forms pores in the membrane of target pathogens. In contrast to homologous proteins such as perforin and the cholesterol-dependent cytolysins (CDCs), all of which require the membrane for oligomerisation, C9 assembles directly onto the nascent MAC from solution. However, the molecular mechanism of MAC assembly remains to be understood. Here we present the 8 Å cryo-EM structure of a soluble form of the poly-C9 component of the MAC. These data reveal a 22-fold symmetrical arrangement of C9 molecules that yield an 88-strand pore-forming β-barrel. The N-terminal thrombospondin-1 (TSP1) domain forms an unexpectedly extensive part of the oligomerisation interface, thus likely facilitating solution-based assembly. These TSP1 interactions may also explain how additional C9 subunits can be recruited to the growing MAC subsequent to membrane insertion., The membrane attack complex is a heteromeric assembly of complement proteins where multiple copies of C9 are recruited by the C5b678 complex to form lytic pores in pathogen membranes. Here the authors present the structure of a soluble pore-like form of the C9 component that reveals details of the oligomerization interfaces.
- Published
- 2016
29. Inside Cover: Application and Structural Analysis of Triazole‐Bridged Disulfide Mimetics in Cyclic Peptides (Angew. Chem. Int. Ed. 28/2020)
- Author
-
Joakim E. Swedberg, Peta J. Harvey, Andrew M. White, Conan K. Wang, Simon J. de Veer, David J. Craik, Gordon J. King, Kuok Yap, Ruby H. P. Law, Thomas Durek, and Guojie Wu
- Subjects
chemistry.chemical_classification ,chemistry.chemical_compound ,chemistry ,Peptidomimetic ,Stereochemistry ,INT ,Triazole ,Disulfide bond ,Cover (algebra) ,General Chemistry ,Catalysis ,Cyclic peptide - Published
- 2020
- Full Text
- View/download PDF
30. Innentitelbild: Application and Structural Analysis of Triazole‐Bridged Disulfide Mimetics in Cyclic Peptides (Angew. Chem. 28/2020)
- Author
-
Thomas Durek, Gordon J. King, Andrew M. White, David J. Craik, Guojie Wu, Joakim E. Swedberg, Peta J. Harvey, Conan K. Wang, Ruby H. P. Law, Kuok Yap, and Simon J. de Veer
- Subjects
chemistry.chemical_classification ,chemistry.chemical_compound ,Peptidomimetic ,Chemistry ,Stereochemistry ,Disulfide bond ,Triazole ,General Medicine ,Cyclic peptide - Published
- 2020
- Full Text
- View/download PDF
31. Tranexamic acid is an active site inhibitor of urokinase plasminogen activator
- Author
-
Guojie Wu, Tom T. Caradoc-Davies, Jonathan G. Schoenecker, Paul J. Conroy, James C. Whisstock, Kellie L. Tuck, Matthew J. Veldman, Blake A. Mazzitelli, Ruby H. P. Law, Adam J. Quek, and Lisa M Ooms
- Subjects
0301 basic medicine ,Proteases ,Plasmin ,030204 cardiovascular system & hematology ,Pharmacology ,Tissue plasminogen activator ,Fibrin ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Antifibrinolytic agent ,Catalytic Domain ,Cell Line, Tumor ,medicine ,Humans ,Fibrinolysin ,Binding site ,Urokinase ,biology ,Chemistry ,Plasminogen ,Hematology ,Urokinase-Type Plasminogen Activator ,Stimulus Report ,Antifibrinolytic Agents ,030104 developmental biology ,Tranexamic Acid ,biology.protein ,Tranexamic acid ,medicine.drug - Abstract
Lysine binding sites (LBSs) of plasminogen (Plg) are essential for maintaining a closed conformation in circulation and binding to fibrin and cell surface receptors.1,2 Upon binding to the targets, Plg is activated to plasmin (Plm) by the fibrin-bound tissue plasminogen activator (tPA) or the cell receptor–bound urokinase plasminogen activator (uPA). Tranexamic acid (TXA) is a lysine analog that binds to the LBSs of Plg with 1 high-affinity (1.1 μM) and 3 medium-affinity (∼0.75 mM) binding sites.1-3 Accordingly, TXA at submicromolar concentrations significantly attenuates in situ Plm formation4 and is used frequently as an antifibrinolytic agent in trauma, as well as in major surgeries, including cardiac, orthopedic, and hepatic surgeries.5 The CRASH-26 and MATTERs7 clinical studies revealed that, when TXA is administered within 3 hours after injury, mortality is reduced by up to 20%.8 Recent reanalysis of the clinical data further showed that the survival benefit of TXA decreased by 10% for every 15 minutes of delayed administration, with no benefit obtained after 3 hours.9 This lack of efficacy outside of the “3-hour window” has been associated with the upregulation of plasma uPA postinjury.6,10,11 We have previously observed that a very high concentration (∼25 mM) of TXA inhibits Plm activity via binding to the primary catalytic (S1) pocket of the enzyme.12 However, because of the low affinity of TXA for Plm, we do not anticipate that TXA functions as a Plm inhibitor during clinical use.12 However, given these findings, we investigated whether TXA may have an inhibitory effect on other proteases in the Plg-activation system. Unexpectedly, our results revealed that TXA attenuates uPA activity with an inhibitory constant (Ki) of 2 mM. In contrast, similar efficacy is not observed in the presence of e-aminocaproic acid (EACA; an alternative drug to TXA).
- Published
- 2018
32. The first transmembrane region of complement component-9 acts as a brake on its self-assembly
- Author
-
Michelle A. Dunstone, Tom T. Caradoc-Davies, James C. Whisstock, Charles Bayly-Jones, Mazdak Radjainia, Ruby H. P. Law, Hariprasad Venugopal, Paul J. Conroy, Georg Ramm, Bradley A. Spicer, Siew Siew Pang, and Sue M. Ekkel
- Subjects
0301 basic medicine ,Models, Molecular ,Conformational change ,Science ,Protein domain ,General Physics and Astronomy ,Plasma protein binding ,Complement Membrane Attack Complex ,Crystallography, X-Ray ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Mice ,Protein Domains ,Animals ,Humans ,lcsh:Science ,Multidisciplinary ,030102 biochemistry & molecular biology ,Chemistry ,Cryoelectron Microscopy ,Membrane Proteins ,General Chemistry ,Complement System Proteins ,Complement C9 ,Transmembrane protein ,Complement system ,030104 developmental biology ,Membrane protein ,Structural biology ,Biophysics ,lcsh:Q ,Complement membrane attack complex ,Protein Binding - Abstract
Complement component 9 (C9) functions as the pore-forming component of the Membrane Attack Complex (MAC). During MAC assembly, multiple copies of C9 are sequentially recruited to membrane associated C5b8 to form a pore. Here we determined the 2.2 Å crystal structure of monomeric murine C9 and the 3.9 Å resolution cryo EM structure of C9 in a polymeric assembly. Comparison with other MAC proteins reveals that the first transmembrane region (TMH1) in monomeric C9 is uniquely positioned and functions to inhibit its self-assembly in the absence of C5b8. We further show that following C9 recruitment to C5b8, a conformational change in TMH1 permits unidirectional and sequential binding of additional C9 monomers to the growing MAC. This mechanism of pore formation contrasts with related proteins, such as perforin and the cholesterol dependent cytolysins, where it is believed that pre-pore assembly occurs prior to the simultaneous release of the transmembrane regions., The Complement component 9 (C9) is the pore-forming component of the Membrane Attack Complex which targets pathogens. Here authors use structural biology to compare monomeric C9 to C9 within the polymeric assembly and identify the element which inhibits C9 self-assembly in the absence of the target membrane.
- Published
- 2018
33. Characterising the Subsite Specificity of Urokinase-Type Plasminogen Activator and Tissue-Type Plasminogen Activator using a Sequence-Defined Peptide Aldehyde Library
- Author
-
Ruby H. P. Law, James C. Whisstock, David J. Craik, Joakim E. Swedberg, Choi Yi Li, and Simon J. de Veer
- Subjects
Proteases ,medicine.medical_treatment ,Peptide ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Substrate Specificity ,Serine ,Peptide Library ,Catalytic Domain ,Fibrinolysis ,medicine ,Humans ,Enzyme Inhibitors ,Molecular Biology ,Urokinase ,chemistry.chemical_classification ,Aldehydes ,Protease ,Tetrapeptide ,010405 organic chemistry ,Organic Chemistry ,Urokinase-Type Plasminogen Activator ,0104 chemical sciences ,chemistry ,Tissue Plasminogen Activator ,Molecular Medicine ,Peptides ,Plasminogen activator ,medicine.drug - Abstract
Urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA) are two serine proteases that contribute to initiating fibrinolysis by activating plasminogen. uPA is also an important tumour-associated protease due to its role in extracellular matrix remodelling. Overexpression of uPA has been identified in several different cancers and uPA inhibition has been reported as a promising therapeutic strategy. Although several peptide-based uPA inhibitors have been developed, the extent to which uPA tolerates different tetrapeptide sequences that span the P1-P4 positions remains to be thoroughly explored. In this study, we screened a sequence-defined peptide aldehyde library against uPA and tPA. Preferred sequences from the library screen yielded potent inhibitors for uPA, led by Ac-GTAR-H (Ki =18 nm), but not for tPA. Additionally, synthetic peptide substrates corresponding to preferred inhibitor sequences were cleaved with high catalytic efficiency by uPA but not by tPA. These findings provide new insights into the binding specificity of uPA and tPA and the relative activity of tetrapeptide inhibitors and substrates against these enzymes.
- Published
- 2018
34. Perforin proteostasis is regulated through its C2 domain: supra-physiological cell death mediated by T431D-perforin
- Author
-
Annette Ciccone, Natalya Lukoyanova, Joseph A. Trapani, Helen R. Saibil, Sandra Verschoor, Ruby H. P. Law, James C. Whisstock, Tahereh Noori, Paul J. Conroy, Ilia Voskoboinik, Hideo Yagita, and Amelia J. Brennan
- Subjects
0301 basic medicine ,Protein Folding ,chemical and pharmacologic phenomena ,Apoptosis ,Crystallography, X-Ray ,Endoplasmic Reticulum ,Article ,03 medical and health sciences ,Mice ,Protein Domains ,Cell Line, Tumor ,Cytotoxic T cell ,Animals ,Transition Temperature ,Molecular Biology ,C2 domain ,biology ,Chemistry ,Perforin ,Protein Stability ,Perforin Deficiency ,Endoplasmic reticulum ,hemic and immune systems ,Cell Biology ,Recombinant Proteins ,Transport protein ,Cell biology ,Protein Structure, Tertiary ,Rats ,Protein Transport ,030104 developmental biology ,Proteostasis ,Granzyme ,biology.protein ,Mutagenesis, Site-Directed ,Calcium - Abstract
The pore forming, Ca(2+)-dependent protein, perforin, is essential for the function of cytotoxic lymphocytes, which are at the frontline of immune defence against pathogens and cancer. Perforin is a glycoprotein stored in the secretory granules prior to release into the immune synapse. Congenital perforin deficiency causes fatal immune dysregulation, and is associated with various haematological malignancies. At least 50% of pathological missense mutations in perforin result in protein misfolding and retention in the endoplasmic reticulum. However, the regulation of perforin proteostasis remains unexplored. Using a variety of biochemical assays that assess protein stability and acquisition of complex glycosylation, we demonstrated that the binding of Ca(2+) to the C2 domain stabilises perforin and regulates its export from the endoplasmic reticulum to the secretory granules. As perforin is a thermo-labile protein, we hypothesised that by altering its C2 domain it may be possible to improve protein stability. On the basis of the X-ray crystal structure of the perforin C2 domain, we designed a mutation (T431D) in the Ca(2+) binding loop. Mutant perforin displayed markedly enhanced thermal stability and lytic function, despite its trafficking from the endoplasmic reticulum remaining unchanged. Furthermore, by introducing the T431D mutation into A90V perforin, a pathogenic mutation, which results in protein misfolding, we corrected the A90V folding defect and completely restored perforin’s cytotoxic function. These results revealed an unexpected role for the Ca(2+)-dependent C2 domain in maintaining perforin proteostasis and demonstrated the possibility of designing perforin with supra-physiological cytotoxic function through stabilisation of the C2 domain.
- Published
- 2018
35. Structural Basis for Ca2+-mediated Interaction of the Perforin C2 Domain with Lipid Membranes
- Author
-
James C. Whisstock, Paul J. Conroy, Eleanor W. W. Leung, Joseph A. Trapani, Hiromasa Yagi, Raymond S. Norton, Ruby H. P. Law, and Ilia Voskoboinik
- Subjects
Protein Conformation ,viruses ,Phosphorylcholine ,Membrane lipids ,Molecular Sequence Data ,chemical and pharmacologic phenomena ,Crystallography, X-Ray ,Biochemistry ,Membrane Lipids ,Mice ,Calcium-binding protein ,Animals ,Amino Acid Sequence ,Binding site ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,C2 domain ,Sequence Homology, Amino Acid ,biology ,Perforin ,hemic and immune systems ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,Membrane ,Granzyme ,Protein Structure and Folding ,Biophysics ,biology.protein ,bacteria ,Calcium ,Binding domain - Abstract
Natural killer cells and cytotoxic T-lymphocytes deploy perforin and granzymes to kill infected host cells. Perforin, secreted by immune cells, binds target membranes to form pores that deliver pro-apoptotic granzymes into the target cell. A crucial first step in this process is interaction of its C2 domain with target cell membranes, which is a calcium-dependent event. Some aspects of this process are understood, but many molecular details remain unclear. To address this, we investigated the mechanism of Ca(2+) and lipid binding to the C2 domain by NMR spectroscopy and x-ray crystallography. Calcium titrations, together with dodecylphosphocholine micelle experiments, confirmed that multiple Ca(2+) ions bind within the calcium-binding regions, activating perforin with respect to membrane binding. We have also determined the affinities of several of these binding sites and have shown that this interaction causes a significant structural rearrangement in CBR1. Thus, it is proposed that Ca(2+) binding at the weakest affinity site triggers changes in the C2 domain that facilitate its interaction with lipid membranes.
- Published
- 2015
- Full Text
- View/download PDF
36. Preferential Acquisition and Activation of Plasminogen Glycoform II by PAM Positive Group A Streptococcal Isolates
- Author
-
Simon M. Cook, Ruby H. P. Law, Martina L. Sanderson-Smith, James C. Whisstock, David M. P. De Oliveira, Diane Ly, Adam J. Quek, and Jason D. McArthur
- Subjects
Aminocaproates ,Binding Sites ,Glycosylation ,Angiostatin ,Protein Conformation ,Streptococcus pyogenes ,Chemistry ,Lysine ,Plasminogen ,Biochemistry ,Group A ,Molecular biology ,Streptococcal M protein ,Enzyme Activation ,chemistry.chemical_compound ,Bacterial Proteins ,Kringles ,Streptococcal Infections ,Humans ,Surface plasmon resonance ,Binding site ,Protein Binding - Abstract
Plasminogen (Plg) circulates in the host as two predominant glycoforms. Glycoform I Plg (GI-Plg) contains glycosylation sites at Asn289 and Thr346, whereas glycoform II Plg (GII-Plg) is exclusively glycosylated at Thr346. Surface plasmon resonance experiments demonstrated that Plg binding group A streptococcal M protein (PAM) exhibits comparative equal affinity for GI- and GII-Plg in the "closed" conformation (for GII-Plg, KD = 27.4 nM; for GI-Plg, KD = 37.0 nM). When Plg was in the "open" conformation, PAM exhibited an 11-fold increase in affinity for GII-Plg (KD = 2.8 nM) compared with that for GI-Plg (KD = 33.2 nM). The interaction of PAM with Plg is believed to be mediated by lysine binding sites within kringle (KR) 2 of Plg. PAM-GI-Plg interactions were fully inhibited with 100 mM lysine analogue ε-aminocaproic acid (εACA), whereas PAM-GII-Plg interactions were shown to be weakened but not inhibited in the presence of 400 mM εACA. In contrast, binding to the KR1-3 domains of GII-Plg (angiostatin) by PAM was completely inhibited in the presence 5 mM εACA. Along with PAM, emm pattern D GAS isolates express a phenotypically distinct SK variant (type 2b SK) that requires Plg ligands such as PAM to activate Plg. Type 2b SK was able to generate an active site and activate GII-Plg at a rate significantly higher than that of GI-Plg when bound to PAM. Taken together, these data suggest that GAS selectively recruits and activates GII-Plg. Furthermore, we propose that the interaction between PAM and Plg may be partially mediated by a secondary binding site outside of KR2, affected by glycosylation at Asn289.
- Published
- 2015
- Full Text
- View/download PDF
37. Dimerization Is Not a Determining Factor for Functional High Affinity Human Plasminogen Binding by the Group A Streptococcal Virulence Factor PAM and Is Mediated by Specific Residues within the PAM a1a2 Domain
- Author
-
Ruby H. P. Law, Victoria A. Ploplis, Sarbani Bhattacharya, Francis J. Castellino, Zhong Liang, and Adam J. Quek
- Subjects
endocrine system ,Protein Conformation ,Streptococcus pyogenes ,Virulence Factors ,Plasmin ,Molecular Sequence Data ,Protein domain ,Peptide ,Biochemistry ,Protein structure ,Bacterial Proteins ,stomatognathic system ,parasitic diseases ,medicine ,Humans ,Amino Acid Sequence ,Binding site ,Molecular Biology ,Peptide sequence ,chemistry.chemical_classification ,Binding Sites ,Plasminogen ,Cell Biology ,Protein engineering ,chemistry ,Protein Structure and Folding ,embryonic structures ,Dimerization ,Ultracentrifugation ,Binding domain ,medicine.drug - Abstract
A emm53 subclass of Group A Streptococcus pyogenes (GAS) interacts tightly with human plasma plasminogen (hPg) and plasmin (hPm) via the kringle 2 (K2hPg) domain of hPg/hPm and the N-terminal a1a2 regions of a GAS coiled-coil M-like protein (PAM). Previous studies have shown that a monomeric PAM fragment, VEK30 (residues 97-125 + Tyr), interacted specifically with isolated K2hPg. However, the binding strength of VEK30 (KD = 56 nm) was ∼60-fold weaker than that of full-length dimeric PAM (KD = 1 nm). To assess whether this attenuated binding was due to the inability of VEK30 to dimerize, we defined the minimal length of PAM required to dimerize using a series of peptides with additional PAM residues placed at the NH2 and COOH termini of VEK30. VEK64 (PAM residues 83-145 + Tyr) was found to be the smallest peptide that adopted an α-helical dimer, and was bound to K2hPg with nearly the same affinity as PAM (KD = 1-2 nm). However, addition of two PAM residues (Arg(126)-His(127)) to the COOH terminus of VEK30 (VEK32) maintained a monomeric peptidic structure, but exhibited similar K2hPg binding affinity as full-length dimeric PAM. We identified five residues in a1a2 (Arg(113), His(114), Glu(116), Arg(126), His(127)), mutation of which reduced PAM binding affinity for K2hPg by ∼ 1000-fold. Replacement of these critical residues by Ala in the GAS genome resulted in reduced virulence, similar to the effects of inactivating the PAM gene entirely. We conclude that rather than dimerization of PAM, the five key residues in the binding domain of PAM are essential to mediate the high affinity interaction with hPg, leading to increased GAS virulence.
- Published
- 2014
- Full Text
- View/download PDF
38. New insights into the structure and function of the plasminogen/plasmin system
- Author
-
Ruby H. P. Law, Diana Abu-ssaydeh, and James C. Whisstock
- Subjects
Serine protease ,chemistry.chemical_classification ,Conformational change ,biology ,Plasmin ,Plasminogen ,Bacterial Physiological Phenomena ,Kringle domain ,Fibrin ,Extracellular matrix ,Enzyme ,chemistry ,Biochemistry ,Structural Biology ,Zymogen ,medicine ,biology.protein ,Carbohydrate Metabolism ,Humans ,Disease ,Fibrinolysin ,Molecular Biology ,medicine.drug - Abstract
Plasminogen is the zymogen form of plasmin, an enzyme that plays a fundamental role in the dissolution of fibrin clots, the extracellular matrix and other key proteins involved in immunity and tissue repair. Comprising seven distinct domains (an N-terminal Pan-apple domain (PAp), 5 kringle domains (KR) and the serine protease domain (SP)), plasminogen undergoes a complex, incompletely understood conformational change that is key to its activation. Here, we review our current understanding of the structural basis for plasminogen activation with regard to new insights derived from crystallographic and biochemical studies.
- Published
- 2013
- Full Text
- View/download PDF
39. Defining the interaction of perforin with calcium and the phospholipid membrane
- Author
-
Raymond S. Norton, Eleanor W. W. Leung, Annette Ciccone, Matthew A. Perugini, Natalya Lukoyanova, Hideo Yagita, Gordon J. Lloyd, Jamie A. Lopez, Joseph A. Trapani, Daouda A K Traore, James C. Whisstock, Sandra Verschoor, Kylie A. Browne, Con Dogovski, Ruby H. P. Law, Ilia Voskoboinik, and Amelia J. Brennan
- Subjects
Pore Forming Cytotoxic Proteins ,Conformational change ,Biochemistry ,Pore forming protein ,Cell membrane ,Jurkat Cells ,Mice ,medicine ,Animals ,Humans ,Molecular Biology ,Phospholipids ,C2 domain ,Mice, Knockout ,MACPF ,biology ,Cell Membrane ,Cell Biology ,Protein Structure, Tertiary ,Rats ,Cell biology ,medicine.anatomical_structure ,Perforin ,Granzyme ,biology.protein ,Calcium ,K562 Cells ,Complement membrane attack complex - Abstract
Following its secretion from cytotoxic lymphocytes into the immune synapse, perforin binds to target cell membranes through its Ca2+-dependent C2 domain. Membrane-bound perforin then forms pores that allow passage of pro-apoptopic granzymes into the target cell. In the present study, structural and biochemical studies reveal that Ca2+ binding triggers a conformational change in the C2 domain that permits four key hydrophobic residues to interact with the plasma membrane. However, in contrast with previous suggestions, these movements and membrane binding do not trigger irreversible conformational changes in the pore-forming MACPF (membrane attack complex/perforin-like) domain, indicating that subsequent monomer–monomer interactions at the membrane surface are required for perforin pore formation.
- Published
- 2013
- Full Text
- View/download PDF
40. Perforin forms transient pores on the target cell plasma membrane to facilitate rapid access of granzymes during killer cell attack
- Author
-
Angus P. R. Johnston, Ruby H. P. Law, Joseph A. Trapani, James C. Whisstock, Helen R. Saibil, Jamie A. Lopez, Catherina H. Bird, Natalya Lukoyanova, Olivia Susanto, Misty R. Jenkins, Vivien R. Sutton, Ilia Voskoboinik, and Phillip I. Bird
- Subjects
Pore Forming Cytotoxic Proteins ,Time Factors ,Synaptic cleft ,Immunology ,Apoptosis ,Complement Membrane Attack Complex ,Biochemistry ,Exocytosis ,Granzymes ,Immunological synapse ,Cell membrane ,Jurkat Cells ,Mice ,medicine ,Animals ,Humans ,Cytotoxic T cell ,biology ,Perforin ,Cell Membrane ,Cell Biology ,Hematology ,Endocytosis ,Cell biology ,Killer Cells, Natural ,medicine.anatomical_structure ,Granzyme ,biology.protein ,Complement membrane attack complex ,HeLa Cells ,T-Lymphocytes, Cytotoxic - Abstract
Cytotoxic lymphocytes serve a key role in immune homeostasis by eliminating virus-infected and transformed target cells through the perforin-dependent delivery of proapoptotic granzymes. However, the mechanism of granzyme entry into cells remains unresolved. Using biochemical approaches combined with time-lapse microscopy of human primary cytotoxic lymphocytes engaging their respective targets, we defined the time course of perforin pore formation in the context of the physiological immune synapse. We show that, on recognition of targets, calcium influx into the lymphocyte led to perforin exocytosis and target cell permeabilization in as little as 30 seconds. Within the synaptic cleft, target cell permeabilization by perforin resulted in the rapid diffusion of extracellular milieu-derived granzymes. Repair of these pores was initiated within 20 seconds and was completed within 80 seconds, thus limiting granzyme diffusion. Remarkably, even such a short time frame was sufficient for the delivery of lethal amounts of granzymes into the target cell. Rapid initiation of apoptosis was evident from caspase-dependent target cell rounding within 2 minutes of perforin permeabilization. This study defines the final sequence of events controlling cytotoxic lymphocyte immune defense, in which perforin pores assemble on the target cell plasma membrane, ensuring efficient delivery of lethal granzymes.
- Published
- 2013
- Full Text
- View/download PDF
41. The X-ray Crystal Structure of Full-Length Human Plasminogen
- Author
-
Qingwei Zhang, Bernadine G.C. Lu, Nathan Cowieson, Angus Cowan, Tom T. Caradoc-Davies, Robert N. Pike, Adam J. Quek, Joanna Amarante Encarnacao, Anita J Horvath, Ruby H. P. Law, James C. Whisstock, A. Ian Smith, David Steer, and Paul Bernard Coughlin
- Subjects
Models, Molecular ,Conformational change ,Glycosylation ,Plasmin ,medicine.medical_treatment ,Crystallography, X-Ray ,Protein Structure, Secondary ,General Biochemistry, Genetics and Molecular Biology ,Kringle domain ,chemistry.chemical_compound ,Protein structure ,Kringles ,medicine ,Humans ,lcsh:QH301-705.5 ,Urokinase ,Serine protease ,Protease ,biology ,Plasminogen ,Enzyme Activation ,lcsh:Biology (General) ,chemistry ,Biochemistry ,Mutation ,Biophysics ,biology.protein ,Protein Binding ,medicine.drug - Abstract
SummaryPlasminogen is the proenzyme precursor of the primary fibrinolytic protease plasmin. Circulating plasminogen, which comprises a Pan-apple (PAp) domain, five kringle domains (KR1-5), and a serine protease (SP) domain, adopts a closed, activation-resistant conformation. The kringle domains mediate interactions with fibrin clots and cell-surface receptors. These interactions trigger plasminogen to adopt an open form that can be cleaved and converted to plasmin by tissue-type and urokinase-type plasminogen activators. Here, the structure of closed plasminogen reveals that the PAp and SP domains, together with chloride ions, maintain the closed conformation through interactions with the kringle array. Differences in glycosylation alter the position of KR3, although in all structures the loop cleaved by plasminogen activators is inaccessible. The ligand-binding site of KR1 is exposed and likely governs proenzyme recruitment to targets. Furthermore, analysis of our structure suggests that KR5 peeling away from the PAp domain may initiate plasminogen conformational change.
- Published
- 2012
- Full Text
- View/download PDF
42. The structural basis for membrane binding and pore formation by lymphocyte perforin
- Author
-
Michelle A. Dunstone, Katherine Baran, James C. Whisstock, Fasséli Coulibaly, Ilia Voskoboinik, Michael E. D'Angelo, Tom T. Caradoc-Davies, Helen R. Saibil, Phillip I. Bird, Michael J. Kuiper, Natalya Lukoyanova, Ruby H. P. Law, Joseph A. Trapani, Sandra Verschoor, Annette Ciccone, Kylie A. Browne, and Elena V. Orlova
- Subjects
Models, Molecular ,Pore Forming Cytotoxic Proteins ,chemical and pharmacologic phenomena ,Crystallography, X-Ray ,Cholesterol-dependent cytolysin ,Granzymes ,Natural killer cell ,Mice ,medicine ,Animals ,Humans ,Lymphocytes ,MACPF ,Multidisciplinary ,Epidermal Growth Factor ,biology ,Perforin Deficiency ,Cell Membrane ,Cryoelectron Microscopy ,hemic and immune systems ,Protein Structure, Tertiary ,Cell biology ,Granzyme B ,Cholesterol ,medicine.anatomical_structure ,Granzyme ,Perforin ,biology.protein ,Complement membrane attack complex - Abstract
Natural killer cells and cytotoxic T lymphocytes accomplish the critically important function of killing virus-infected and neoplastic cells. They do this by releasing the pore-forming protein perforin and granzyme proteases from cytoplasmic granules into the cleft formed between the abutting killer and target cell membranes. Perforin, a 67-kilodalton multidomain protein, oligomerizes to form pores that deliver the pro-apoptopic granzymes into the cytosol of the target cell. The importance of perforin is highlighted by the fatal consequences of congenital perforin deficiency, with more than 50 different perforin mutations linked to familial haemophagocytic lymphohistiocytosis (type 2 FHL). Here we elucidate the mechanism of perforin pore formation by determining the X-ray crystal structure of monomeric murine perforin, together with a cryo-electron microscopy reconstruction of the entire perforin pore. Perforin is a thin 'key-shaped' molecule, comprising an amino-terminal membrane attack complex perforin-like (MACPF)/cholesterol dependent cytolysin (CDC) domain followed by an epidermal growth factor (EGF) domain that, together with the extreme carboxy-terminal sequence, forms a central shelf-like structure. A C-terminal C2 domain mediates initial, Ca(2+)-dependent membrane binding. Most unexpectedly, however, electron microscopy reveals that the orientation of the perforin MACPF domain in the pore is inside-out relative to the subunit arrangement in CDCs. These data reveal remarkable flexibility in the mechanism of action of the conserved MACPF/CDC fold and provide new insights into how related immune defence molecules such as complement proteins assemble into pores.
- Published
- 2010
- Full Text
- View/download PDF
43. A major cathepsin B protease from the liver fluke Fasciola hepatica has atypical active site features and a potential role in the digestive tract of newly excysted juvenile parasites
- Author
-
Terence W Spithill, Peter M. Smooker, Deanne L.V. Greenwood, Ruby H. P. Law, Carolyn I Phillips, Robert N. Pike, Nirma Samarawickrema, Lakshmi C. Wijeyewickrema, Boris Turk, David Piedrafita, Noelene Sheila Quinsey, Theresa H.T. Coetzer, James A. Irving, Matthew Bogyo, Steven H. L. Verhelst, and Simone A. Beckham
- Subjects
Cysteine Proteinase Inhibitors ,Biochemistry ,Article ,Cathepsin B ,Substrate Specificity ,Enzyme activator ,Catalytic Domain ,Animals ,Humans ,Fasciola hepatica ,Parasites ,Cathepsin ,Life Cycle Stages ,Exopeptidase activity ,Sheep ,biology ,Cell Biology ,Exopeptidase ,biology.organism_classification ,Cathepsins ,Cystatins ,Cysteine protease ,Enzyme Activation ,Gastrointestinal Tract ,Kinetics ,Protein Transport ,Structural Homology, Protein ,Molecular Probes ,biology.protein ,Cystatin - Abstract
The newly excysted juvenile (NEJ) stage of the Fasciola hepatica lifecycle occurs just prior to invasion into the wall of the gut of the host, rendering it an important target for drug development. The cathepsin B enzymes from NEJ flukes have recently been demonstrated to be crucial to invasion and migration by the parasite. Here we characterize one of the cathepsin B enzymes (recombinant FhcatB1) from NEJ flukes. FhcatB1 has biochemical properties distinct from mammalian cathepsin B enzymes, with an atypical preference for Ile over Leu or Arg residues at the P(2) substrate position and an inability to act as an exopeptidase. FhcatB1 was active across a broad pH range (optimal activity at pH 5.5-7.0) and resistant to inhibition by cystatin family inhibitors from sheep and humans, suggesting that this enzyme would be able to function in extracellular environments in its mammalian hosts. It appears, however, that the FhcatB1 protease functions largely as a digestive enzyme in the gut of the parasite, due to the localization of a specific, fluorescently labeled inhibitor with an Ile at the P(2) position. Molecular modelling and dynamics were used to predict the basis for the unusual substrate specificity: a P(2) Ile residue positions the substrate optimally for interaction with catalytic residues of the enzyme, and the enzyme lacks an occluding loop His residue crucial for exopeptidase activity. The unique features of the enzyme, particularly with regard to its specificity and likely importance to a vital stage of the parasite's life cycle, make it an excellent target for therapeutic inhibitors or vaccination.
- Published
- 2009
- Full Text
- View/download PDF
44. Structural Mechanisms of Inactivation in Scabies Mite Serine Protease Paralogues
- Author
-
James C. Whisstock, David J. Kemp, James A. Irving, Charlene Willis, Sundy N.Y. Yang, Simone L. Reynolds, Katja Fischer, Tanya Ann Bashtannyk-Puhalovich, Robert N. Pike, Ashley M. Buckle, Christopher G. Langendorf, Simone A. Beckham, Ruby H. P. Law, and Sheena McGowan
- Subjects
Models, Molecular ,Proteases ,Protein Conformation ,medicine.medical_treatment ,Sarcoptes scabiei ,Crystallography, X-Ray ,Microbiology ,Serine ,Peptide Library ,Structural Biology ,Catalytic Domain ,Catalytic triad ,medicine ,Mite ,Animals ,Molecular Biology ,Phylogeny ,Serine protease ,Protease ,biology ,Serine Endopeptidases ,biology.organism_classification ,Enzyme Activation ,Mutation ,biology.protein ,Antibody - Abstract
The scabies mite (Sarcoptes scabiei) is a parasite responsible for major morbidity in disadvantaged communities and immuno-compromised patients worldwide. In addition to the physical discomfort caused by the disease, scabies infestations facilitate infection by Streptococcal species via skin lesions, resulting in a high prevalence of rheumatic fever/heart disease in affected communities. The scabies mite produces 33 proteins that are closely related to those in the dust mite group 3 allergen and belong to the S1-like protease family (chymotrypsin-like). However, all but one of these molecules contain mutations in the conserved active-site catalytic triad that are predicted to render them catalytically inactive. These molecules are thus termed scabies mite inactivated protease paralogues (SMIPPs). The precise function of SMIPPs is unclear; however, it has been suggested that these proteins might function by binding and protecting target substrates from cleavage by host immune proteases, thus preventing the host from mounting an effective immune challenge. In order to begin to understand the structural basis for SMIPP function, we solved the crystal structures of SMIPP-S-I1 and SMIPP-S-D1 at 1.85 A and 2.0 A resolution, respectively. Both structures adopt the characteristic serine protease fold, albeit with large structural variations over much of the molecule. In both structures, mutations in the catalytic triad together with occlusion of the S1 subsite by a conserved Tyr200 residue is predicted to block substrate ingress. Accordingly, we show that both proteases lack catalytic function. Attempts to restore function (via site-directed mutagenesis of catalytic residues as well as Tyr200) were unsuccessful. Taken together, these data suggest that SMIPPs have lost the ability to bind substrates in a classical "canonical" fashion, and instead have evolved alternative functions in the lifecycle of the scabies mite.
- Published
- 2009
- Full Text
- View/download PDF
45. Stability of the octameric structure affects plasminogen-binding capacity of streptococcal enolase
- Author
-
Mark J. Walker, Daniel J. Ericsson, Anna Stamp, J. Andrew Aquilina, Blagojce Jovcevski, Bostjan Kobe, James C. Whisstock, Ruby H. P. Law, Lachlan W. Casey, Eugene Valkov, Carlo Bertozzi, and Amanda J. Cork
- Subjects
Protein Conformation ,Streptococcus pyogenes ,Plasmin ,Enolase ,lcsh:Medicine ,Plasma protein binding ,Crystallography, X-Ray ,medicine.disease_cause ,Protein structure ,Bacterial Proteins ,medicine ,Humans ,Histone octamer ,Binding site ,lcsh:Science ,Multidisciplinary ,Chemistry ,lcsh:R ,Plasminogen ,Lyase ,Biochemistry ,Phosphopyruvate Hydratase ,lcsh:Q ,Research Article ,Protein Binding ,medicine.drug - Abstract
Group A Streptococcus (GAS) is a human pathogen that has the potential to cause invasive disease by binding and activating human plasmin(ogen). Streptococcal surface enolase (SEN) is an octameric α-enolase that is localized at the GAS cell surface. In addition to its glycolytic role inside the cell, SEN functions as a receptor for plasmin(ogen) on the bacterial surface, but the understanding of the molecular basis of plasmin(ogen) binding is limited. In this study, we determined the crystal and solution structures of GAS SEN and characterized the increased plasminogen binding by two SEN mutants. The plasminogen binding ability of SENK312A and SENK362A is ~2- and ~3.4-fold greater than for the wild-type protein. A combination of thermal stability assays, native mass spectrometry and X-ray crystallography approaches shows that increased plasminogen binding ability correlates with decreased stability of the octamer. We propose that decreased stability of the octameric structure facilitates the access of plasmin(ogen) to its binding sites, leading to more efficient plasmin(ogen) binding and activation.
- Published
- 2015
46. Production and processing of a recombinant Fasciola hepatica cathepsin B-like enzyme (FhcatB1) reveals potential processing mechanisms in the parasite
- Author
-
Noelene Sheila Quinsey, Ruby H-P Law, Simone A. Beckham, Robert N. Pike, James H. McKerrow, Conor R. Caffrey, Terence W Spithill, and Peter M. Smooker
- Subjects
Models, Molecular ,Proteases ,Time Factors ,Protein Conformation ,Clinical Biochemistry ,Biology ,Crystallography, X-Ray ,Biochemistry ,Cathepsin B ,Pichia pastoris ,law.invention ,Structure-Activity Relationship ,law ,Animals ,Fasciola hepatica ,Molecular Biology ,chemistry.chemical_classification ,Cathepsin ,Binding Sites ,Dose-Response Relationship, Drug ,Heparin ,Dextran Sulfate ,Hydrogen-Ion Concentration ,biology.organism_classification ,Recombinant Proteins ,Endopeptidase ,Protein Structure, Tertiary ,Enzyme Activation ,Enzyme ,chemistry ,Recombinant DNA ,Protein Processing, Post-Translational - Abstract
The liver fluke,Fasciola hepatica, apparently uses a number of cysteine proteases during its life cycle, most likely for feeding, immune evasion and invasion of tissues. A cathepsin B-like enzyme (herein referred to as FhcatB1) appears to be a major enzyme secreted by the invasive, newly excysted juvenile flukes of this parasite. To examine the processing mechanisms for this enzyme, a recombinant form was expressed inPichia pastorisand purified to yield a homogenous pool of the enzyme. The purified enzyme could be autoactivated at low pH via a bi-molecular mechanism, a process that was greatly accelerated by the presence of large, negatively charged molecules such as dextran sulfate. The enzyme could also apparently be processed to the correct size by an asparaginyl endopeptidase via cleavage in an unusual insertion N-terminal to the normal cleavage site used to yield the active form of the enzyme. Thus, there appear to be a number of ways in which this enzyme can be processed to its optimally active form prior to secretion byF. hepatica.
- Published
- 2006
- Full Text
- View/download PDF
47. The Murine Orthologue of Human Antichymotrypsin
- Author
-
Stephen P. Bottomley, Noelene Sheila Quinsey, Robert N. Pike, Anita J Horvath, James C. Whisstock, Jamie Rossjohn, Paul Bernard Coughlin, James A. Irving, and Ruby H. P. Law
- Subjects
biology ,Cell Biology ,Cathepsin G ,Serpin ,Biochemistry ,Molecular biology ,Alpha 1-antichymotrypsin ,Protease inhibitor (biology) ,chemistry.chemical_compound ,Protein structure ,chemistry ,biology.protein ,medicine ,Structural motif ,Molecular Biology ,Peptide sequence ,Reactive center ,medicine.drug - Abstract
Antichymotrypsin (SERPINA3) is a widely expressed member of the serpin superfamily, required for the regulation of leukocyte proteases released during an inflammatory response and with a permissive role in the development of amyloid encephalopathy. Despite its biological significance, there is at present no available structure of this serpin in its native, inhibitory state. We present here the first fully refined structure of a murine antichymotrypsin orthologue to 2.1 A, which we propose as a template for other antichymotrypsin-like serpins. A most unexpected feature of the structure of murine serpina3n is that it reveals the reactive center loop (RCL) to be partially inserted into the A beta-sheet, a structural motif associated with ligand-dependent activation in other serpins. The RCL is, in addition, stabilized by salt bridges, and its plane is oriented at 90 degrees to the RCL of antitrypsin. A biochemical and biophysical analysis of this serpin demonstrates that it is a fast and efficient inhibitor of human leukocyte elastase (ka: 4 +/- 0.9 x 10(6) m(-1) s(-)1) and cathepsin G (ka: 7.9 +/- 0.9 x 10(5) m(-1) s(-)1) giving a spectrum of activity intermediate between that of human antichymotrypsin and human antitrypsin. An evolutionary analysis reveals that residues subject to positive selection and that have contributed to the diversity of sequences in this sub-branch (A3) of the serpin superfamily are essentially restricted to the P4-P6' region of the RCL, the distal hinge, and the loop between strands 4B and 5B.
- Published
- 2005
- Full Text
- View/download PDF
48. The High Resolution Crystal Structure of the Human Tumor Suppressor Maspin Reveals a Novel Conformational Switch in the G-helix
- Author
-
Jamie Rossjohn, Marguerite S. Buzza, Tanya Ann Bashtannyk-Puhalovich, Stephen P. Bottomley, Ashley M. Buckle, Ruby H. P. Law, Phillip I. Bird, Margaret D Worrall, James A. Irving, Travis Clarke Beddoe, Katya Ruzyla, Kim-Ngoc Nguyen, and James C. Whisstock
- Subjects
Models, Molecular ,Conformational change ,Protein Conformation ,Cathepsin L ,Xenopus ,Static Electricity ,Plasma protein binding ,Serpin ,Biology ,Crystallography, X-Ray ,Bioinformatics ,Biochemistry ,Protein Structure, Secondary ,law.invention ,Mice ,Protein structure ,law ,Animals ,Humans ,Genes, Tumor Suppressor ,Binding site ,Molecular Biology ,Serpins ,Cofactor binding ,Binding Sites ,Circular Dichroism ,Homozygote ,Temperature ,Maspin ,Cell Biology ,Cathepsins ,Recombinant Proteins ,Extracellular Matrix ,Rats ,Cell biology ,Cysteine Endopeptidases ,Spectrophotometry ,Suppressor ,Chickens ,Plasmids ,Protein Binding - Abstract
Maspin is a serpin that acts as a tumor suppressor in a range of human cancers, including tumors of the breast and lung. Maspin is crucial for development, because homozygous loss of the gene is lethal; however, the precise physiological role of the molecule is unclear. To gain insight into the function of human maspin, we have determined its crystal structure in two similar, but non-isomorphous crystal forms, to 2.1- and 2.8-A resolution, respectively. The structure reveals that maspin adopts the native serpin fold in which the reactive center loop is expelled fully from the A beta-sheet, makes minimal contacts with the core of the molecule, and exhibits a high degree of flexibility. A buried salt bridge unique to maspin orthologues causes an unusual bulge in the region around the D and E alpha-helices, an area of the molecule demonstrated in other serpins to be important for cofactor recognition. Strikingly, the structural data reveal that maspin is able to undergo conformational change in and around the G alpha-helix, switching between an open and a closed form. This change dictates the electrostatic character of a putative cofactor binding surface and highlights this region as a likely determinant of maspin function. The high resolution crystal structure of maspin provides a detailed molecular framework to elucidate the mechanism of function of this important tumor suppressor.
- Published
- 2005
- Full Text
- View/download PDF
49. Conformational changes during pore formation by the perforin-related protein pleurotolysin
- Author
-
James C. Whisstock, Rodney K. Tweten, Michelle A. Dunstone, Oded Kleifeld, Helen R. Saibil, Daouda A K Traore, S.C. Kondos, Bradley A. Spicer, Ruby H. P. Law, Cyril F. Reboul, Joseph A. Trapani, Katherine V. Oliver, Maya Topf, Ilia Voskoboinik, Tamas Zsolt Hatfaludi, Natalya Lukoyanova, Eileen M. Hotze, Irene Farabella, Susan M. Ekkel, and Tom T. Caradoc-Davies
- Subjects
Models, Molecular ,Conformational change ,Protein Folding ,Erythrocytes ,QH301-705.5 ,Recombinant Fusion Proteins ,Gene Expression ,Complement Membrane Attack Complex ,Biology ,Cholesterol-dependent cytolysin ,Q1 ,bcs ,Crystallography, X-Ray ,Pleurotus ,General Biochemistry, Genetics and Molecular Biology ,Protein Structure, Secondary ,Fungal Proteins ,Hemolysin Proteins ,Protein structure ,Escherichia coli ,Animals ,Protein Isoforms ,Biology (General) ,Integral membrane protein ,Fungal protein ,MACPF ,Sheep ,General Immunology and Microbiology ,General Neuroscience ,QH ,Cell Membrane ,Cryoelectron Microscopy ,Transmembrane protein ,Cell biology ,Protein Structure, Tertiary ,Biophysics ,Protein folding ,General Agricultural and Biological Sciences ,Protein Binding ,Research Article - Abstract
Membrane attack complex/perforin-like (MACPF) proteins comprise the largest superfamily of pore-forming proteins, playing crucial roles in immunity and pathogenesis. Soluble monomers assemble into large transmembrane pores via conformational transitions that remain to be structurally and mechanistically characterised. Here we present an 11 Å resolution cryo-electron microscopy (cryo-EM) structure of the two-part, fungal toxin Pleurotolysin (Ply), together with crystal structures of both components (the lipid binding PlyA protein and the pore-forming MACPF component PlyB). These data reveal a 13-fold pore 80 Å in diameter and 100 Å in height, with each subunit comprised of a PlyB molecule atop a membrane bound dimer of PlyA. The resolution of the EM map, together with biophysical and computational experiments, allowed confident assignment of subdomains in a MACPF pore assembly. The major conformational changes in PlyB are a ∼70° opening of the bent and distorted central β-sheet of the MACPF domain, accompanied by extrusion and refolding of two α-helical regions into transmembrane β-hairpins (TMH1 and TMH2). We determined the structures of three different disulphide bond-trapped prepore intermediates. Analysis of these data by molecular modelling and flexible fitting allows us to generate a potential trajectory of β-sheet unbending. The results suggest that MACPF conformational change is triggered through disruption of the interface between a conserved helix-turn-helix motif and the top of TMH2. Following their release we propose that the transmembrane regions assemble into β-hairpins via top down zippering of backbone hydrogen bonds to form the membrane-inserted β-barrel. The intermediate structures of the MACPF domain during refolding into the β-barrel pore establish a structural paradigm for the transition from soluble monomer to pore, which may be conserved across the whole superfamily. The TMH2 region is critical for the release of both TMH clusters, suggesting why this region is targeted by endogenous inhibitors of MACPF function., Author Summary Animals, plants, fungi, and bacteria all use pore-forming proteins of the membrane attack complex-perforin (MACPF) family as lethal, cell-killing weapons. These proteins are able to insert into the plasma membranes of target cells, creating large pores that short circuit the natural separation between the intracellular and extracellular milieu, with catastrophic results. However, the pore-forming proteins must undergo a substantial transformation from soluble precursors to a large barrel-shaped transmembrane complex as they punch their way into cells. Using a combination of X-ray crystallography and cryo electron microscopy, we have visualized, for the first time, the mechanism of action of one of these pore-forming proteins—pleurotolysin, a MACPF protein from the edible oyster mushroom. This enabled us to propose a model of the pleurotolysin pore by fitting the crystallographic structures of the pore proteins into a three-dimensional map of the pore obtained by cryo electron microscopy. We then designed a set of double mutants that allowed us to chemically trap intermediate states along the trajectory of the pore formation process, and to determine their structures too. By combining these data we proposed a detailed molecular mechanism for pore formation. The pleurotolysin first assembles into rings of 13 subunits, each of which then opens up by about 70° during pore formation. This process is accompanied by refolding and extrusion of two compact regions from each subunit into long hairpins that then zipper together to form an 80-Å wide barrel-shaped channel through the membrane., A combination of structural methods reveals the complex process by which the perforin-like fungal toxin Pleurotolysin rearranges its structure to form a pore that punches a hole in target cell membranes.
- Published
- 2014
50. Reconciling the Structural Attributes of Avian Antibodies*
- Author
-
Paul J. Conroy, James C. Whisstock, Richard O'Kennedy, Sarah Gilgunn, Ruby H. P. Law, Stephen Hearty, Tom T. Caradoc-Davies, and Gordon J. Lloyd
- Subjects
Phage display ,Avian immune system ,Molecular Sequence Data ,Computational biology ,Crystallography, X-Ray ,Biochemistry ,Antibodies ,Antigen-Antibody Reactions ,Structure-Activity Relationship ,Immune system ,Antigen ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,biology ,Mechanism (biology) ,Repertoire ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,respiratory system ,Single-Domain Antibodies ,Multiple species ,Virology ,Protein Structure, Tertiary ,Kinetics ,Protein Structure and Folding ,biology.protein ,Antibody ,Crystallization ,human activities ,Chickens - Abstract
Antibodies are high value therapeutic, diagnostic, biotechnological, and research tools. Combinatorial approaches to antibody discovery have facilitated access to unique antibodies by surpassing the diversity limitations of the natural repertoire, exploitation of immune repertoires from multiple species, and tailoring selections to isolate antibodies with desirable biophysical attributes. The V-gene repertoire of the chicken does not utilize highly diverse sequence and structures, which is in stark contrast to the mechanism employed by humans, mice, and primates. Recent exploitation of the avian immune system has generated high quality, high affinity antibodies to a wide range of antigens for a number of therapeutic, diagnostic and biotechnological applications. Furthermore, extensive examination of the amino acid characteristics of the chicken repertoire has provided significant insight into mechanisms employed by the avian immune system. A paucity of avian antibody crystal structures has limited our understanding of the structural consequences of these uniquely chicken features. This paper presents the crystal structure of two chicken single chain fragment variable (scFv) antibodies generated from large libraries by phage display against important human antigen targets, which capture two unique CDRL1 canonical classes in the presence and absence of a non-canonical disulfide constrained CDRH3. These structures cast light on the unique structural features of chicken antibodies and contribute further to our collective understanding of the unique mechanisms of diversity and biochemical attributes that render the chicken repertoire of particular value for antibody generation.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.