1
|
Huang LD, Gou XY, Yang MJ, Li MJ, Chen SN, Yan J, Liu XX, Sun AH. Peptidoglycan biosynthesis-associated enzymatic kinetic characteristics and β-lactam antibiotic inhibitory effects of different Streptococcus pneumoniae penicillin-binding proteins. Int J Biol Macromol 2024; 254:127784. [PMID: 37949278 DOI: 10.1016/j.ijbiomac.2023.127784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/15/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023]
Abstract
Penicillin-binding proteins (PBPs) include transpeptidases, carboxypeptidases, and endopeptidases for biosynthesis of peptidoglycans in the cell wall to maintain bacterial morphology and survival in the environment. Streptococcus pneumoniae expresses six PBPs, but their enzymatic kinetic characteristics and inhibitory effects on different β-lactam antibiotics remain poorly understood. In this study, all the six recombinant PBPs of S. pneumoniae displayed transpeptidase activity with different substrate affinities (Km = 1.56-9.11 mM) in a concentration-dependent manner, and rPBP3 showed a greater catalytic efficiency (Kcat = 2.38 s-1) than the other rPBPs (Kcat = 3.20-7.49 × 10-2 s-1). However, only rPBP3 was identified as a carboxypeptidase (Km = 8.57 mM and Kcat = 2.57 s-1). None of the rPBPs exhibited endopeptidase activity. Penicillin and cefotaxime inhibited the transpeptidase and carboxypeptidase activity of all the rPBPs but imipenem did not inhibited the enzymatic activities of rPBP3. Except for the lack of binding of imipenem to rPBP3, penicillin, cefotaxime, and imipenem bound to all the other rPBPs (KD = 3.71-9.35 × 10-4 M). Sublethal concentrations of penicillin, cefotaxime, and imipenem induced a decrease of pneumococcal pbps-mRNA levels (p < 0.05). These results indicated that all six PBPs of S. pneumoniae are transpeptidases, while only PBP3 is a carboxypeptidase. Imipenem has no inhibitory effect on pneumococcal PBP3. The pneumococcal genes for encoding endopeptidases remain to be determined.
Collapse
Affiliation(s)
- Li-Dan Huang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China; Yiwu Central Blood Station, Yiwu, Zhejiang 322000, PR China
| | - Xiao-Yu Gou
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China
| | - Mei-Juan Yang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China; The First Hospital of Putian City, Putian, Fujian 351100, PR China
| | - Meng-Jie Li
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China
| | - Sui-Ning Chen
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China
| | - Jie Yan
- Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China
| | - Xiao-Xiang Liu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China.
| | - Ai-Hua Sun
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang 310053, PR China.
| |
Collapse
|
2
|
Onyido EK, James D, Garcia-Parra J, Sinfield J, Moberg A, Coombes Z, Worthington J, Williams N, Francis LW, Conlan RS, Gonzalez D. Elucidating Novel Targets for Ovarian Cancer Antibody-Drug Conjugate Development: Integrating In Silico Prediction and Surface Plasmon Resonance to Identify Targets with Enhanced Antibody Internalization Capacity. Antibodies (Basel) 2023; 12:65. [PMID: 37873862 PMCID: PMC10594448 DOI: 10.3390/antib12040065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/29/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Antibody-drug conjugates (ADCs) constitute a rapidly expanding category of biopharmaceuticals that are reshaping the landscape of targeted chemotherapy. The meticulous process of selecting therapeutic targets, aided by specific monoclonal antibodies' high specificity for binding to designated antigenic epitopes, is pivotal in ADC research and development. Despite ADCs' intrinsic ability to differentiate between healthy and cancerous cells, developmental challenges persist. In this study, we present a rationalized pipeline encompassing the initial phases of the ADC development, including target identification and validation. Leveraging an in-house, computationally constructed ADC target database, termed ADC Target Vault, we identified a set of novel ovarian cancer targets. We effectively demonstrate the efficacy of Surface Plasmon Resonance (SPR) technology and in vitro models as predictive tools, expediting the selection and validation of targets as ADC candidates for ovarian cancer therapy. Our analysis reveals three novel robust antibody/target pairs with strong binding and favourable antibody internalization rates in both wild-type and cisplatin-resistant ovarian cancer cell lines. This approach enhances ADC development and offers a comprehensive method for assessing target/antibody combinations and pre-payload conjugation biological activity. Additionally, the strategy establishes a robust platform for high-throughput screening of potential ovarian cancer ADC targets, an approach that is equally applicable to other cancer types.
Collapse
Affiliation(s)
- Emenike Kenechi Onyido
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK (D.J.); (J.G.-P.); (Z.C.); (L.W.F.); (R.S.C.)
| | - David James
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK (D.J.); (J.G.-P.); (Z.C.); (L.W.F.); (R.S.C.)
| | - Jezabel Garcia-Parra
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK (D.J.); (J.G.-P.); (Z.C.); (L.W.F.); (R.S.C.)
| | - John Sinfield
- Cytiva, Björkgatan 30, 751 84 Uppsala, Sweden; (J.S.); (A.M.)
| | - Anna Moberg
- Cytiva, Björkgatan 30, 751 84 Uppsala, Sweden; (J.S.); (A.M.)
| | - Zoe Coombes
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK (D.J.); (J.G.-P.); (Z.C.); (L.W.F.); (R.S.C.)
| | - Jenny Worthington
- Axis Bioservices Ltd., 189 Castleroe Rd, Coleraine BT51 3RP, UK; (J.W.); (N.W.)
| | - Nicole Williams
- Axis Bioservices Ltd., 189 Castleroe Rd, Coleraine BT51 3RP, UK; (J.W.); (N.W.)
| | - Lewis Webb Francis
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK (D.J.); (J.G.-P.); (Z.C.); (L.W.F.); (R.S.C.)
| | - Robert Steven Conlan
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK (D.J.); (J.G.-P.); (Z.C.); (L.W.F.); (R.S.C.)
| | - Deyarina Gonzalez
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Swansea University, Singleton Park, Swansea SA2 8PP, UK (D.J.); (J.G.-P.); (Z.C.); (L.W.F.); (R.S.C.)
| |
Collapse
|
3
|
Zhang C, Miao Y, Feng Y, Wang J, Tian Z, Dong J, Gao B, Zhang L. Umami polypeptide detection system targeting the human T1R1 receptor and its taste-presenting mechanism. Biomaterials 2022; 287:121660. [PMID: 35792387 DOI: 10.1016/j.biomaterials.2022.121660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/24/2022] [Indexed: 01/30/2023]
Abstract
Umami is one of five basic tastes, the elucidation of its mechanism by the study of the interaction between umami polypeptides and hT1R1 umami receptors is of great significance. However, research on umami peptides targeting human T1R1 receptors is lacking, and the molecular mechanism remains elusive. Here, we successfully established a system to detect umami peptides targeting human T1R1 receptors by fluorescence spectroscopy, Surface Plasmon Resonance (SPR) and computational simulation. The sensory evaluation, calculated Kd value, and experimental affinity results between the four selected umami peptides (GRVSNCAA, KGDEESLA, KGGGGP, and TGDPEK) and glutamate were tested using this system, and all matched well. The maximum Ka value of GRVSNCAA was 479.55 M-1, and the minimum affinity of TGDPEK was 2.67 M-1. Computational simulations showed that the different peptide binding sites in the hT1R1 binding pocket occupied due to conformational changes are important factors for different taste thresholds, and that peptide hydrophobicity plays an important role in regulating affinity. Thus, our study enables rapid screening of high-intensity umami peptides and the development of T1R1 receptor-based umami detection sensors.
Collapse
Affiliation(s)
- Chuanxi Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China; School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China; Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai , 200240, China
| | - Yulu Miao
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Yinghui Feng
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Jiawei Wang
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China; School of Health Science and Engineering, University of Shanghai for Science and Tecchnology, Shanghai, 200093, China
| | - Zhuoli Tian
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Juan Dong
- School of Food Science and Technology, Shihezi University, Shihezi, 832000, China
| | - Bei Gao
- School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China.
| | - Lujia Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China; NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai, 200062, China.
| |
Collapse
|
4
|
Drozd M, Karoń S, Malinowska E. Recent Advancements in Receptor Layer Engineering for Applications in SPR-Based Immunodiagnostics. SENSORS (BASEL, SWITZERLAND) 2021; 21:3781. [PMID: 34072572 PMCID: PMC8198293 DOI: 10.3390/s21113781] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022]
Abstract
The rapid progress in the development of surface plasmon resonance-based immunosensing platforms offers wide application possibilities in medical diagnostics as a label-free alternative to enzyme immunoassays. The early diagnosis of diseases or metabolic changes through the detection of biomarkers in body fluids requires methods characterized by a very good sensitivity and selectivity. In the case of the SPR technique, as well as other surface-sensitive detection strategies, the quality of the transducer-immunoreceptor interphase is crucial for maintaining the analytical reliability of an assay. In this work, an overview of general approaches to the design of functional SPR-immunoassays is presented. It covers both immunosensors, the design of which utilizes well-known and often commercially available substrates, as well as the latest solutions developed in-house. Various approaches employing chemical and passive binding, affinity-based antibody immobilization, and the introduction of nanomaterial-based surfaces are discussed. The essence of their influence on the improvement of the main analytical parameters of a given immunosensor is explained. Particular attention is paid to solutions compatible with the latest trends in the development of label-free immunosensors, such as platforms dedicated to real-time monitoring in a quasi-continuous mode, the use of in situ-generated receptor layers (elimination of the regeneration step), and biosensors using recombinant and labelled protein receptors.
Collapse
Affiliation(s)
- Marcin Drozd
- Faculty of Chemistry, The Chair of Medical Biotechnology, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Sylwia Karoń
- Faculty of Chemistry, The Chair of Medical Biotechnology, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Elżbieta Malinowska
- Faculty of Chemistry, The Chair of Medical Biotechnology, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| |
Collapse
|
5
|
TcpC inhibits toll-like receptor signaling pathway by serving as an E3 ubiquitin ligase that promotes degradation of myeloid differentiation factor 88. PLoS Pathog 2021; 17:e1009481. [PMID: 33788895 PMCID: PMC8041205 DOI: 10.1371/journal.ppat.1009481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 04/12/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
TcpC is a virulence factor of uropathogenic E. coli (UPEC). It was found that TIR domain of TcpC impedes TLR signaling by direct association with MyD88. It has been a long-standing question whether bacterial pathogens have evolved a mechanism to manipulate MyD88 degradation by ubiquitin-proteasome pathway. Here, we show that TcpC is a MyD88-targeted E3 ubiquitin ligase. Kidney macrophages from mice with pyelonephritis induced by TcpC-secreting UPEC showed significantly decreased MyD88 protein levels. Recombinant TcpC (rTcpC) dose-dependently inhibited protein but not mRNA levels of MyD88 in macrophages. Moreover, rTcpC significantly promoted MyD88 ubiquitination and accumulation in proteasomes in macrophages. Cys12 and Trp106 in TcpC are crucial amino acids in maintaining its E3 activity. Therefore, TcpC blocks TLR signaling pathway by degradation of MyD88 through ubiquitin-proteasome system. Our findings provide not only a novel biochemical mechanism underlying TcpC-medicated immune evasion, but also the first example that bacterial pathogens inhibit MyD88-mediated signaling pathway by virulence factors that function as E3 ubiquitin ligase. Toll/interleukin-1 receptor domain-containing protein encoded by E. coli (TcpC) is an important virulence factor in many strains of uropathogenic E. coli (UPEC). TcpC-mediated evasion of innate immunity plays an important role in the pathogenesis of UPEC caused urinary tract infection (UTI) including pyelonephritis. In the present study, we show TcpC is an E3 ubiquitin ligase that promotes ubiquitination and degradation of MyD88, hereby blocking the TLR signaling pathway. Our findings not only illuminate the novel biochemical mechanisms underlying TcpC-mediated evasion of innate immunity, but also provide the first example that bacterial pathogens can subvert TLR signaling pathway through virulence factors that function as MyD88-targeted E3 ubiquitin ligase.
Collapse
|
6
|
Strong interactions between Salp15 homologues from the tick I. ricinus and distinct types of the outer surface OspC protein from Borrelia. Ticks Tick Borne Dis 2020; 12:101630. [PMID: 33401196 DOI: 10.1016/j.ttbdis.2020.101630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 11/24/2022]
Abstract
Ticks belonging to the genus Ixodes are parasites feeding on vertebrate blood and vectors for many pathogenic microbes, including Borrelia burgdorferi sensu lato spirochetes, the causative agent of Lyme borreliosis. The tick saliva contains a mixture of bioactive molecules showing a wide range of properties for efficient engorgement. One of the most extensively studied components of tick saliva is a 15-kDa salivary gland protein (Salp15) from Ixodes scapularis. This multifunctional protein suppresses the immune response of hosts through pleiotropic action on a few crucial defense pathways. Salp15 and its homologue from I. ricinus Iric1 have been also shown to bind to Borrelia burgdorferi sensu stricto outer surface protein C (OspC) permitting the spirochetes to evade antibody-mediated killing in the human host. Further studies revealed that Salp15 and Iric1 protected B. burgdorferi s. s. and B. garinii expressing OspC against the complement system. OspC is the most variable protein on the outer surface of Borrelia, which in addition to Salp15 can also bind other ligands, such as plasminogen, fibrinogen, fibronectin or complement factor 4. So far several OspC variants produced by B. burgdorferi s. l. spirochetes were shown to be capable of binding Salp15 or its homologue, but the protection against borreliacidal antibodies has only been proven in the case of B. burgdorferi s. s. The question of Salp15 contribution to Borrelia survival during the infection has been comprehensively studied during the last decades. In contrast, the organization of the OspC-Salp15 complex has been poorly explored. This report describes the binding between three Salp15 homologues from the tick Ixodes ricinus (Iric1, Iric2 and Iric3) and OspC from four B. burgdorferi sensu lato strains in terms of the binding parameters, analyzed with two independent biophysical methods - Microscale thermophoresis (MST) and Biolayer interferometry (BLI). The results of both experiments show a binding constant at the nanomolar level, which indicates very strong interactions. While the Iric1-OspC binding has been reported before, we show in this study that also Iric2 and Iric3 are capable of OspC binding with high affinity. This observation suggests that these two Salp15 homologues might be used by B. burgdorferi s. l. in a way analogous to Iric1. A comparison of the results from the two methods let us propose that N-terminal immobilization of OspC significantly increases the affinity between the two proteins. Finally, our results indicate that the Iric binding site is located in close proximity of the OspC epitopes recognized by human antibodies, which may have important biological and medical implications.
Collapse
|
7
|
Normal-incidence type solution immersed silicon (SIS) biosensor for ultra-sensitive, label-free detection of cardiac troponin I. Biosens Bioelectron 2020; 168:112525. [PMID: 32858415 DOI: 10.1016/j.bios.2020.112525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 01/25/2023]
Abstract
Early diagnosis of acute myocardial infarction (AMI) significantly reduce the mortality rate and can be achieved via high-sensitive detection of AMI specific cardiac troponin I (cTnI) biomarker. Here, we present normal-incident type solution-immersed silicon (NI-SIS) ellipsometric biosensor, designed for ultra-high sensitive, high-throughput, label-free detection of the target protein. The NI-SIS sensors are equipped with a specially designed prism that maintains the angle of incidence close to the Brewster angle during operation, which significantly reduces SIS noise signals induced by the refractive index fluctuations of the surrounding medium, improves the signal-to-noise ratio, in-results lowers the detection limit. We applied NI-SIS biosensor for ultra-sensitive detection of cTnI biomarkers in human serum. The optimized sensor chip fabrication and detection operation procedures are proposed. The wide linear concentration ranges of fg/mL to ng/mL is achieved with the detection limit of 22.0 fg/mL of cTnI. The analytical correlation was assessed by linear regression analysis with the results of the Pathfast reference system. These impressive biosensing capabilities of NI-SIS technology have huge potentials for accurate detection of target species in different application areas, such as diagnosis, drug discovery, and food contaminations.
Collapse
|
8
|
Development of a Novel SPR Assay to Study CXCR4-Ligand Interactions. BIOSENSORS-BASEL 2020; 10:bios10100150. [PMID: 33096938 PMCID: PMC7589327 DOI: 10.3390/bios10100150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 01/16/2023]
Abstract
G protein-coupled receptors (GPCRs) are involved in a plethora of different diseases. Consequently, these proteins are considered as an important class of drug targets. Measuring detailed kinetic information on these types of proteins has been challenging. Surface plasmon resonance (SPR) can provide this information, however, the use of SPR on GPCRs remains a complex issue. Here, we report an SPR assay to investigate the interactions between the full-length chemokine receptor CXCR4 and nanobody-Fc (Nb-Fc) ligands. Nb-Fcs consist of two monovalent VHH domains fused with an Fc domain of a human IgG molecule. The CXCR4 protein used in this assay was produced with a C-terminal 10x-histidine tag and was immobilized on a nitrilotriacetic acid chip. In order to verify the sensitivity and effectiveness of this assay, the results were compared to data obtained from cellular assays as well as from another SPR assay using CXCR4 virus-like particles (VLPs). CXCR4 remained intact and stable for at least 12 h, and the kinetic results correlated well with both the cellular assays and the VLP SPR assay results. Apart from determining the binding kinetics of Nb-Fc with CXCR4, our results contributed to understanding CXCR4 interaction dynamics. In conclusion, this assay provides a viable experimental platform that has high potential to be expanded for studying other molecules as well as other histidine-tagged GPCRs.
Collapse
|
9
|
Chu R, Gerstein J, Wu H, Huang H, Lou Y, Palmer R. Development of Rabbit Monoclonal Antibodies for Quantitation of Therapeutic Human Antibodies in Preclinical Non-Human Primate Studies. Monoclon Antib Immunodiagn Immunother 2020; 39:175-183. [PMID: 32996830 DOI: 10.1089/mab.2020.0024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
During preclinical studies, there is a great need to develop monoclonal antibodies (mAbs) that are specific to human immunoglobulin (IgG), without binding to monkey IgG, to detect therapeutic human mAb in non-human primates. We took advantage of the latest rabbit B cell cloning technology to develop six unique rabbit anti-human IgG mAb clones for this purpose. These clones are capable of binding to both human IgG and Fab with high affinity without nonspecific binding to cynomolgus monkey IgG. These clones have been evaluated as a generic capture reagent for the detection of human IgG and Fab, in the presence of cynomolgus monkey serum, by Gyrolab™ immunoassay. They may be used in singlet or as pairs for the detection of human IgG, in any host animal, to meet the need for therapeutic mAb development in preclinical studies.
Collapse
Affiliation(s)
- Ruiyin Chu
- Biomarker and Clinical Bioanalysis, Sanofi North America, Framingham, Massachusetts, USA
| | - Jacob Gerstein
- Biomarker and Clinical Bioanalysis, Sanofi North America, Framingham, Massachusetts, USA
| | - Hai Wu
- Yurogen Biosystem LLC, Worcester, Massachusetts, USA
| | - Haibin Huang
- Yurogen Biosystem LLC, Worcester, Massachusetts, USA
| | - Yang Lou
- Yurogen Biosystem LLC, Worcester, Massachusetts, USA
| | - Rachel Palmer
- Biomarker and Clinical Bioanalysis, Sanofi North America, Framingham, Massachusetts, USA
| |
Collapse
|
10
|
Wang SH, Wu TJ, Lee CW, Yu J. Dissecting the conformation of glycans and their interactions with proteins. J Biomed Sci 2020; 27:93. [PMID: 32900381 PMCID: PMC7487937 DOI: 10.1186/s12929-020-00684-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/26/2020] [Indexed: 12/20/2022] Open
Abstract
The use of in silico strategies to develop the structural basis for a rational optimization of glycan-protein interactions remains a great challenge. This problem derives, in part, from the lack of technologies to quantitatively and qualitatively assess the complex assembling between a glycan and the targeted protein molecule. Since there is an unmet need for developing new sugar-targeted therapeutics, many investigators are searching for technology platforms to elucidate various types of molecular interactions within glycan-protein complexes and aid in the development of glycan-targeted therapies. Here we discuss three important technology platforms commonly used in the assessment of the complex assembly of glycosylated biomolecules, such as glycoproteins or glycosphingolipids: Biacore analysis, molecular docking, and molecular dynamics simulations. We will also discuss the structural investigation of glycosylated biomolecules, including conformational changes of glycans and their impact on molecular interactions within the glycan-protein complex. For glycoproteins, secreted protein acidic and rich in cysteine (SPARC), which is associated with various lung disorders, such as chronic obstructive pulmonary disease (COPD) and lung cancer, will be taken as an example showing that the core fucosylation of N-glycan in SPARC regulates protein-binding affinity with extracellular matrix collagen. For glycosphingolipids (GSLs), Globo H ceramide, an important tumor-associated GSL which is being actively investigated as a target for new cancer immunotherapies, will be used to demonstrate how glycan structure plays a significant role in enhancing angiogenesis in tumor microenvironments.
Collapse
Affiliation(s)
- Sheng-Hung Wang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan, 333, Taiwan
| | - Tsai-Jung Wu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan, 333, Taiwan
| | - Chien-Wei Lee
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan, 333, Taiwan
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, and Chang Gung University, Taoyuan, 333, Taiwan. .,Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
11
|
Plácido A, Ferreira-da-Silva F, Leite JRSA, de-los-Santos-Álvarez N, Delerue-Matos C. A convenient renewable surface plasmon resonance chip for relative quantification of genetically modified soybean in food and feed. PLoS One 2020; 15:e0229659. [PMID: 32101588 PMCID: PMC7043770 DOI: 10.1371/journal.pone.0229659] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/12/2020] [Indexed: 01/09/2023] Open
Abstract
The cultivation of genetically modified organisms (GMO) continues to expand worldwide. Still, many consumers express concerns about the use of GMO in food or feed, and many countries have legislated on labelling systems to indicate the presence of GMO in commercial products. To deal with the increased number of GMO events and to address related regulations, alternative detection methods for GMO inspection are required. In this work, a genosensor based on Surface Plasmon Resonance under continuous flow was developed for the detection and quantification of a genetically modified soybean (event GTS 40-3-2). In a single chip, the simultaneous detection of the event-specific and the taxon-specific samples were achieved, whose detection limits were 20 pM and 16 pM, respectively. The reproducibility was 1.4%, which supports the use of the chip as a reliable and cost-effective alternative to other DNA-based techniques. The results indicate that the proposed method is a versatile tool for GMO quantification in food and feed samples.
Collapse
Affiliation(s)
- Alexandra Plácido
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto, Portugal
| | - Frederico Ferreira-da-Silva
- Instituto de Investigação e Inovação em Saúde, i3S, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - José Roberto S. A. Leite
- Área Morfologia, Faculdade de Medicina, Campus Darcy Ribeiro, Universidade de Brasília, Brasília, Federal District, Brazil
| | | | - Cristina Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto, Portugal
| |
Collapse
|
12
|
Capelli D, Parravicini C, Pochetti G, Montanari R, Temporini C, Rabuffetti M, Trincavelli ML, Daniele S, Fumagalli M, Saporiti S, Bonfanti E, Abbracchio MP, Eberini I, Ceruti S, Calleri E, Capaldi S. Surface Plasmon Resonance as a Tool for Ligand Binding Investigation of Engineered GPR17 Receptor, a G Protein Coupled Receptor Involved in Myelination. Front Chem 2020; 7:910. [PMID: 31998697 PMCID: PMC6966494 DOI: 10.3389/fchem.2019.00910] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/16/2019] [Indexed: 01/11/2023] Open
Abstract
The aim of this study was to investigate the potential of surface plasmon resonance (SPR) spectroscopy for the measurement of real-time ligand-binding affinities and kinetic parameters for GPR17, a G protein-coupled receptor (GPCR) of major interest in medicinal chemistry as potential target in demyelinating diseases. The receptor was directly captured, in a single-step, from solubilized membrane extracts on the sensor chip through a covalently bound anti-6x-His-antibody and retained its ligand binding activity for over 24 h. Furthermore, our experimental setup made possible, after a mild regeneration step, to remove the bound receptor without damaging the antibody, and thus to reuse many times the same chip. Two engineered variants of GPR17, designed for crystallographic studies, were expressed in insect cells, extracted from crude membranes and analyzed for their binding with two high affinity ligands: the antagonist Cangrelor and the agonist Asinex 1. The calculated kinetic parameters and binding constants of ligands were in good agreement with those reported from activity assays and highlighted a possible functional role of the N-terminal residues of the receptor in ligand recognition and binding. Validation of SPR results was obtained by docking and molecular dynamics of GPR17-ligands interactions and by functional in vitro studies. The latter allowed us to confirm that Asinex 1 behaves as GPR17 receptor agonist, inhibits forskolin-stimulated adenylyl cyclase pathway and promotes oligodendrocyte precursor cell maturation and myelinating ability.
Collapse
Affiliation(s)
- Davide Capelli
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Chiara Parravicini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giorgio Pochetti
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Roberta Montanari
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Rome, Italy
| | | | - Marco Rabuffetti
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Milan, Italy
| | | | | | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Simona Saporiti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elisabetta Bonfanti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria P Abbracchio
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Ivano Eberini
- Department of Pharmacological and Biomolecular Sciences, Data Science Research Center, Università degli Studi di Milano, Milan, Italy
| | - Stefania Ceruti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Enrica Calleri
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Stefano Capaldi
- Department of Biotechnology, University of Verona, Verona, Italy
| |
Collapse
|
13
|
Sabapathy T, Helmerhorst E, Bottomley S, Babaeff S, Munyard K, Newsholme P, Mamotte CD. Use of virus-like particles as a native membrane model to study the interaction of insulin with the insulin receptor. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1204-1212. [PMID: 30951702 DOI: 10.1016/j.bbamem.2019.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/15/2019] [Accepted: 03/28/2019] [Indexed: 12/15/2022]
Abstract
There is emerging evidence of the utility of virus-like particles (VLPs) as a novel model for the study of receptor-ligand interactions in a native plasma membrane environment. VLPs consist of a viral core protein encapsulated by portions of the cell membrane with membrane proteins and receptors expressed in their native conformation. VLPs can be generated in mammalian cells by transfection with the retroviral core protein (gag). In this study, we used Chinese hamster ovary (CHO T10) cells stably overexpressing the insulin receptor (IR) to generate IR bearing VLPs. The diameter and size uniformity of VLPs were estimated by dynamic light scattering and morphological features examined by scanning electron microscopy. The presence of high affinity IR on VLPs was demonstrated by competitive binding assays (KD: 2.3 ± 0.4 nM, n = 3), which was similar to that on the parental CHO T10 cells (KD: 2.1 ± 0.4 nM, n = 3). We also report that increases or decreases in membrane cholesterol content by treatment with methyl-β-cyclodextrin (MBCD) or cholesterol pre-loaded methyl-β-cyclodextrin (cMBCD), respectively, substantially decreased insulin binding (> 30%) to both VLPs and cells, and we speculate this is due to a change in receptor disposition. We suggest that this novel finding of decreases in insulin binding in response to changes in membrane cholesterol content may largely account for the unexplained decreases in insulin signalling events previously reported elsewhere. Finally, we propose VLPs as a viable membrane model for the study of insulin-IR interactions in a native membrane environment.
Collapse
Affiliation(s)
- Thiru Sabapathy
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| | - Erik Helmerhorst
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| | | | | | - Kylie Munyard
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| | - Cyril D Mamotte
- School of Pharmacy and Biomedical Sciences, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley campus, Perth, Western Australia 6102, Australia.
| |
Collapse
|
14
|
G-Protein Coupled Receptor Protein Synthesis on a Lipid Bilayer Using a Reconstituted Cell-Free Protein Synthesis System. Life (Basel) 2018; 8:life8040054. [PMID: 30400226 PMCID: PMC6316570 DOI: 10.3390/life8040054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/23/2018] [Accepted: 10/30/2018] [Indexed: 12/31/2022] Open
Abstract
Membrane proteins are important drug targets which play a pivotal role in various cellular activities. However, unlike cytosolic proteins, most of them are difficult-to-express proteins. In this study, to synthesize and produce sufficient quantities of membrane proteins for functional and structural analysis, we used a bottom-up approach in a reconstituted cell-free synthesis system, the PURE system, supplemented with artificial lipid mimetics or micelles. Membrane proteins were synthesized by the cell-free system and integrated into lipid bilayers co-translationally. Membrane proteins such as the G-protein coupled receptors were expressed in the PURE system and a productivity ranging from 0.04 to 0.1 mg per mL of reaction was achieved with a correct secondary structure as predicted by circular dichroism spectrum. In addition, a ligand binding constant of 27.8 nM in lipid nanodisc and 39.4 nM in micelle was obtained by surface plasmon resonance and the membrane protein localization was confirmed by confocal microscopy in giant unilamellar vesicles. We found that our method is a promising approach to study the different classes of membrane proteins in their native-like artificial lipid bilayer environment for functional and structural studies.
Collapse
|
15
|
vWA proteins of Leptospira interrogans induce hemorrhage in leptospirosis by competitive inhibition of vWF/GPIb-mediated platelet aggregation. EBioMedicine 2018; 37:428-441. [PMID: 30337247 PMCID: PMC6284457 DOI: 10.1016/j.ebiom.2018.10.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUD Leptospira interrogans is the major causative agent of leptospirosis, a worldwide zoonotic disease. Hemorrhage is a typical pathological feature of leptospirosis. Binding of von Willebrand factor (vWF) to platelet glycoprotein-Ibα (GPIbα) is a crucial step in initiation of platelet aggregation. The products of L. interrogans vwa-I and vwa-II genes contain vWF-A domains, but their ability to induce hemorrhage has not been determined. METHODS Human (Hu)-platelet- and Hu-GPIbα-binding abilities of the recombinant proteins expressed by L. interrogans strain Lai vwa-I and vwa-II genes (rLep-vWA-I and rLep-vWA-II) were detected by flowcytometry, surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC). Hu-platelet aggregation and its signaling kinases and active components were detected by lumiaggregometry, Western analysis, spectrophotometry and confocal microscopy. Hu-GPIbα-binding sites in rLep-vWA-I and rLep-vWA-II were identified by SPR/ITC measurements. FINDINGS Both rLep-vWA-I and rLep-vWA-II were able to bind to Hu-platelets and inhibit rHu-vWF/ristocetin-induced Hu-platelet aggregation, but Hu-GPIbα-IgG, rLep-vWA-I-IgG and rLep-vWA-II-IgG blocked this binding or inhibition. SPR and ITC revealed a tight interaction between Hu-GPIbα and rLep-vWA-I/rLep-vWA-II with KD values of 3.87 × 10-7-8.65 × 10-8 M. Hu-GPIbα-binding of rL-vWA-I/rL-vWA-II neither activated the PI3K/AKT-ERK and PLC/PKC kinases nor affected the NO, cGMP, ADP, Ca2+ and TXA2 levels in Hu-platelets. G13/R36/G47 in Lep-vWA-I and G76/Q126 in Lep-vWA-II were confirmed as the Hu-GPIbα-binding sites. Injection of rLep-vWA-I or rLep-vWA-II in mice resulted in diffuse pulmonary and focal renal hemorrhage but this hemorrhage was blocked by rLep-vWA-I-IgG or rLep-vWA-II-IgG. INTERPRETATION The products of L. interrogans vwa-I and vwa-II genes induce hemorrhage by competitive inhibition of vWF-mediated Hu-platelet aggregation.
Collapse
|
16
|
A novel Fas-binding outer membrane protein and lipopolysaccharide of Leptospira interrogans induce macrophage apoptosis through the Fas/FasL-caspase-8/-3 pathway. Emerg Microbes Infect 2018; 7:135. [PMID: 30061622 PMCID: PMC6066479 DOI: 10.1038/s41426-018-0135-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/18/2018] [Accepted: 06/23/2018] [Indexed: 02/08/2023]
Abstract
Leptospira interrogans is the major causative agent of leptospirosis, an emerging, globally spreading zoonotic infectious disease. The pathogen induces macrophage apoptosis, but the molecular basis and mechanism remain unknown. In the present study, we found that L. interrogans caused apoptosis of phagocytosis-inhibited macrophages, and the product of the L. interrogans LB047 gene (Lep-OMP047) was the unique protein captured by mouse and human Fas proteins. The recombinant expressed Lep-OMP047 (rLep-OMP047) strongly bound mouse and human Fas proteins with equilibrium association constant (KD) values of 5.20 × 10−6 to 2.84 × 10−9 M according to surface plasmon resonance measurement and isothermal titration calorimetry. Flow-cytometric examination showed that 5 μg rLep-OMP047 or 1 μg lipopolysaccharide of L. interrogans (Lep-LPS) caused 43.70% or 21.90% early apoptosis in mouse J774A.1 macrophages and 28.41% or 15.80% for PMA-differentiated human THP-1 macrophages, respectively, but the apoptosis was blocked by Fas-antagonizing IgGs, Fas siRNAs, and caspase-8/-3 inhibitors. Moreover, Lep-OMP047 was significantly upregulated during infection of macrophages. Lep-LPS promoted the expression and cytomembrane translocation of Fas and FasL in macrophages. The JNK and p38 MAPK but not ERK signaling pathways, as well as the transcription factors c-Jun and ATF2 but not CHOP, mediated Lep-LPS-induced Fas/FasL expression and translocation. TLR2 but not TLR4 mediated Lep-LPS-induced JNK/p38 MAPK activation. Therefore, we demonstrated that a novel Fas-binding OMP and LPS of L. interrogans induce macrophage apoptosis through the Fas/FasL-caspase-8/-3 pathway.
Collapse
|
17
|
Chao YC, Fleischer J, Yang RB. Guanylyl cyclase-G is an alarm pheromone receptor in mice. EMBO J 2017; 37:39-49. [PMID: 28963397 DOI: 10.15252/embj.201797155] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 08/27/2017] [Accepted: 08/30/2017] [Indexed: 01/07/2023] Open
Abstract
Many animals respond to threats by releasing alarm pheromones (APs) that warn conspecifics. In mice, detection of the AP 2-sec-butyl-4,5-dihydrothiazole (SBT) is mediated by chemosensory neurons residing in the Grueneberg ganglion (GG) of the anterior nasal region. Although the molecular mechanisms underlying activation of GG neurons by SBT and other substances are still unclear, recent studies have reported an involvement of the transmembrane guanylyl cyclase (GC) subtype GC-G in chemosensory signaling in the GG Here, we show that SBT directly binds with high affinity to the extracellular domain of GC-G and elicits an enhanced enzymatic activity of this protein. In line with this finding, heterologous expression of GC-G renders cells responsive to SBT while activation by SBT was strongly attenuated in GG neurons from GC-G-deficient mice. Consistently, SBT-induced fear-associated behaviors, SBT-evoked elevated blood pressure, and increased serum levels of the stress hormone corticosterone were clearly reduced in GC-G-knockout animals compared to wild-type mice. These observations suggest that GC-G serves as an unusual receptor in GG neurons mediating the detection of the volatile AP substance SBT.
Collapse
Affiliation(s)
- Ying-Chi Chao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Joerg Fleischer
- Department of Animal Physiology, Institute of Biology/Zoology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan .,Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
18
|
Wear MA, Nowicki MW, Blackburn EA, McNae IW, Walkinshaw MD. Thermo-kinetic analysis space expansion for cyclophilin-ligand interactions - identification of a new nonpeptide inhibitor using Biacore™ T200. FEBS Open Bio 2017; 7:533-549. [PMID: 28396838 PMCID: PMC5377415 DOI: 10.1002/2211-5463.12201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 12/31/2022] Open
Abstract
We have established a refined methodology for generating surface plasmon resonance sensor surfaces of recombinant his‐tagged human cyclophilin‐A. Our orientation‐specific stabilisation approach captures his‐tagged protein under ‘physiological conditions’ (150 mm NaCl, pH 7.5) and covalently stabilises it on Ni2+‐nitrilotriacetic acid surfaces, very briefly activated for primary amine‐coupling reactions, producing very stable and active surfaces (≥ 95% specific activity) of cyclophilin‐A. Variation in protein concentration with the same contact time allows straightforward generation of variable density surfaces, with essentially no loss of activity, making the protocol easily adaptable for studying numerous interactions; from very small fragments, ~ 100 Da, to large protein ligands. This new method results in an increased stability and activity of the immobilised protein and allowed us to expand the thermo‐kinetic analysis space, and to determine accurate and robust thermodynamic parameters for the cyclophilin‐A–cyclosporin‐A interaction. Furthermore, the increased sensitivity of the surface allowed identification of a new nonpeptide inhibitor of cyclophilin‐A, from a screen of a fragment library. This fragment, 2,3‐diaminopyridine, bound specifically with a mean affinity of 248 ± 60 μm. The X‐ray structure of this 109‐Da fragment bound in the active site of cyclophilin‐A was solved to a resolution of 1.25 Å (PDB: 5LUD), providing new insight into the molecular details for a potential new series of nonpeptide cyclophilin‐A inhibitors.
Collapse
Affiliation(s)
- Martin A Wear
- The Edinburgh Protein Production Facility (EPPF) Wellcome Trust Centre for Cell Biology (WTCCB) University of Edinburgh UK
| | - Matthew W Nowicki
- The Edinburgh Protein Production Facility (EPPF) Wellcome Trust Centre for Cell Biology (WTCCB) University of Edinburgh UK
| | - Elizabeth A Blackburn
- The Edinburgh Protein Production Facility (EPPF) Wellcome Trust Centre for Cell Biology (WTCCB) University of Edinburgh UK
| | - Iain W McNae
- The Edinburgh Protein Production Facility (EPPF) Wellcome Trust Centre for Cell Biology (WTCCB) University of Edinburgh UK
| | - Malcolm D Walkinshaw
- The Edinburgh Protein Production Facility (EPPF) Wellcome Trust Centre for Cell Biology (WTCCB) University of Edinburgh UK
| |
Collapse
|
19
|
Renaud JP, Chung CW, Danielson UH, Egner U, Hennig M, Hubbard RE, Nar H. Biophysics in drug discovery: impact, challenges and opportunities. Nat Rev Drug Discov 2016; 15:679-98. [PMID: 27516170 DOI: 10.1038/nrd.2016.123] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Over the past 25 years, biophysical technologies such as X-ray crystallography, nuclear magnetic resonance spectroscopy, surface plasmon resonance spectroscopy and isothermal titration calorimetry have become key components of drug discovery platforms in many pharmaceutical companies and academic laboratories. There have been great improvements in the speed, sensitivity and range of possible measurements, providing high-resolution mechanistic, kinetic, thermodynamic and structural information on compound-target interactions. This Review provides a framework to understand this evolution by describing the key biophysical methods, the information they can provide and the ways in which they can be applied at different stages of the drug discovery process. We also discuss the challenges for current technologies and future opportunities to use biophysical methods to solve drug discovery problems.
Collapse
Affiliation(s)
- Jean-Paul Renaud
- NovAliX, Boulevard Sébastien Brant, 67405 Illkirch Cedex, France.,Institut de Génétique et Biologie Moléculaire et Cellulaire, CNRS UMR7104/INSERM U964/Université de Strasbourg, 1 rue Laurent Fries - BP10142, 67404 Illkirch Cedex, France.,RiboStruct, 15 rue Neuve, 67540 Ostwald, France
| | - Chun-Wa Chung
- GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - U Helena Danielson
- Department of Chemistry - BMC and Science for Life Laboratory, Drug Discovery &Development Platform, Uppsala University, SE-751 05 Uppsala, Sweden.,Beactica AB, Uppsala Business Park, 754 50 Uppsala, Sweden
| | - Ursula Egner
- Bayer Pharma AG, Müllerstrasse 178, 13353 Berlin, Germany
| | - Michael Hennig
- Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland.,leadXpro AG, PARK INNOVAARE, CH-5234 Villigen, Switzerland
| | - Roderick E Hubbard
- University of York, Heslington, York, YO10 5DD, UK.,Vernalis (R&D), Granta Park, Cambridge, CB21 6GB, UK
| | - Herbert Nar
- Boehringer Ingelheim GmbH &Co. KG, Birkendorfer Strasse 65, 88400 Biberach, Germany
| |
Collapse
|
20
|
Clausen TM, Pereira MA, Oo HZ, Resende M, Gustavson T, Mao Y, Sugiura N, Liew J, Fazli L, Theander TG, Daugaard M, Salanti A. Real-time and label free determination of ligand binding-kinetics to primary cancer tissue specimens; a novel tool for the assessment of biomarker targeting. SENSING AND BIO-SENSING RESEARCH 2016; 9:23-30. [PMID: 27441183 PMCID: PMC4942562 DOI: 10.1016/j.sbsr.2016.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/29/2016] [Accepted: 05/04/2016] [Indexed: 11/30/2022] Open
Abstract
In clinical oncology, diagnosis and evaluation of optimal treatment strategies are mostly based on histopathological examination combined with immunohistochemical (IHC) expression analysis of cancer-associated antigens in formalin fixed paraffin-embedded (FFPE) tissue biopsies. However, informative IHC analysis depends on both the specificity and affinity of the binding reagent, which are inherently difficult to quantify in situ. Here we describe a label-free method that allows for the direct and real-time assessment of molecular binding kinetics in situ on FFPE tissue specimens using quartz crystal microbalance (QCM) enabled biosensor technology. We analysed the interaction between the rVAR2 protein and its placental-like chondroitin sulfate (pl-CS) receptor in primary human placenta tissue and in breast and prostate tumour specimens in situ. rVAR2 interacted with FFPE human placenta and cancer tissue with an affinity in the nanomolar range, and showed no detectable interaction with pl-CS negative normal tissue. We further validated the method by including analysis with the androgen receptor N-20 antibody (anti-AR). As the KD value produced by this method is independent of the number of epitopes available, this readout offers a quantitative and unbiased readout for in situ binding-avidity and amount of binding epitopes. In summary, this method adds a new and important dimension to classical IHC-based molecular pathology by adding information about the binding characteristics in biologically relevant conditions. This can potentially be used to select optimal biologics for diagnostic and for therapeutic applications as well as guide the development of novel high affinity binding drugs.
Collapse
Affiliation(s)
- Thomas Mandel Clausen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Correspondence to: T.M. Clausen, Centre for Medical Parasitology, Bartholinsgade 2, 1356 Copenhagen, Denmark.Centre for Medical ParasitologyBartholinsgade 2Copenhagen1356Denmark
| | - Marina Ayres Pereira
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Molecular Pathology and Cell Imaging Laboratory, Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Mafalda Resende
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Tobias Gustavson
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Yang Mao
- Copenhagen Center for Glycomics and Department of Cellular and Molecular Medicine, University of Copenhagen, Denmark
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Nobuo Sugiura
- Institute for Molecular Science of Medicine, Aichi Medical University, Japan
| | - Janet Liew
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Molecular Pathology and Cell Imaging Laboratory, Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Molecular Pathology and Cell Imaging Laboratory, Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Thor G. Theander
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mads Daugaard
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- Molecular Pathology and Cell Imaging Laboratory, Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Correspondence to: M. Daugaard, Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada.Vancouver Prostate CentreVancouverBCV6H 3Z6Canada
| | - Ali Salanti
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
21
|
Patra A, Ding T, Engudar G, Wang Y, Dykas MM, Liedberg B, Kah JCY, Venkatesan T, Drum CL. Component-Specific Analysis of Plasma Protein Corona Formation on Gold Nanoparticles Using Multiplexed Surface Plasmon Resonance. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:1174-82. [PMID: 26455731 DOI: 10.1002/smll.201501603] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 07/06/2015] [Indexed: 05/20/2023]
Abstract
At the nano-bio interface, human plasma differentially interacts with engineered nanomaterials through the creation of protein coronas, which in turn become primary determinants of both the pharmacokinetics and pharmacodynamics of circulating nanoparticles. Here, for the first time, the specific binding kinetics of the four major corona forming proteins (human serum albumin, fibrinogen, ApoA1, and polyclonal IgG) are determined for gold nanoparticles (AuNPs). Using a multiplexed surface plasmonic assay, highly reproducible measurements of on rate (k(on)), off rate (k(off)), and disassociation constant (K(D)), in addition to relative amounts of protein binding, are obtained. Dramatic differences in k(on) for individual components are shown as primary determinants of protein affinities, with k(on) ranging over nearly two orders of magnitude for the proteins studied, while k(off) remains within a factor of two for the set. The effect of polyethylene glycol (PEG) modification on plasma component binding is also studied and the effect of PEG length on human serum interaction is characterized through systematic screening of PEG molecular weight (2-30k). The effect of nanoparticle modification on particle targeting is also characterized through study of a hybrid AuNP system.
Collapse
Affiliation(s)
- Abhijeet Patra
- NUSNNI-NanoCore, National University of Singapore, 5A Engineering Drive 1, 11411, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, 117456, Singapore
| | - Tao Ding
- Cardiovascular Research Institute, Department of Cardiology, 1E Kent Ridge Road, 119228, Singapore
| | - Gokce Engudar
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, 117575, Singapore
| | - Yi Wang
- Center for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Michal Marcin Dykas
- NUSNNI-NanoCore, National University of Singapore, 5A Engineering Drive 1, 11411, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, 117456, Singapore
| | - Bo Liedberg
- Center for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - James Chen Yong Kah
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, 117575, Singapore
| | - Thirumalai Venkatesan
- NUSNNI-NanoCore, National University of Singapore, 5A Engineering Drive 1, 11411, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, 117456, Singapore
- Department of Electrical Engineering, Department of Materials Science and Engineering, Department of Physics, National University of Singapore, 117581, Singapore
| | - Chester Lee Drum
- Cardiovascular Research Institute, Department of Cardiology, 1E Kent Ridge Road, 119228, Singapore
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5, 138648, Singapore
| |
Collapse
|
22
|
Real-time and label-free analysis of binding thermodynamics of carbohydrate-protein interactions on unfixed cancer cell surfaces using a QCM biosensor. Sci Rep 2015; 5:14066. [PMID: 26369583 PMCID: PMC4570189 DOI: 10.1038/srep14066] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/17/2015] [Indexed: 11/08/2022] Open
Abstract
A novel approach to the study of binding thermodynamics and kinetics of carbohydrate-protein interactions on unfixed cancer cell surfaces using a quartz crystal microbalance (QCM) biosensor was developed, in which binding events take place at the cell surface, more closely mimicking a biologically relevant environment. In this study, colon adenocarcinoma cells (KM-12) and ovary adenocarcinoma cells (SKOV-3) grew on the optimized polystyrene-coated biosensor chip without fixation. The association and dissociation between the cell surface carbohydrates and a range of lectins, including WGA, Con A, UEA-I, GS-II, PNA and SBA, were monitored in real time and without label for evaluation of cell surface glycosylation. Furthermore, the thermodynamic and kinetic parameters of the interaction between lectins and cell surface glycan were studied, providing detailed information about the interactions, such as the association rate constant, dissociation rate constant, affinity constant, as well as the changes of entropy, enthalpy and Gibbs free energy. This application provides an insight into the cell surface glycosylation and the complex molecular recognition on the intact cell surface, which may have impacts on disease diagnosis and drug discovery.
Collapse
|