1
|
Sultana S, Geyer CR. Effect of framework and complementarity determining region H1 charge on the human VH-B1a domain expression, folding, stability, and solubility. Biochem Biophys Res Commun 2024; 739:150956. [PMID: 39541928 DOI: 10.1016/j.bbrc.2024.150956] [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: 08/27/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Many difficulties related to using antibodies in diagnostic and therapeutic applications can sometimes be circumvented by using smaller, less complex single domain antibodies based on variable heavy chain (VH) domain constructs such as camelid VHH domains. However, VH domains have their own limitations, including an increased tendency to aggregate. VH domains often contain hydrophobic residues within their complementarity-determining regions (CDRs) that facilitate binding to target antigens but can also mediate VH domain aggregation, which is a concern for therapeutic applications since this can trigger immune responses. In this study, we engineered the human VH-1Ba domain to improve its stability and solubility by introducing charged amino acids in the VH domain framework region and CDRH1. We followed two strategies to improve the stability and solubility of VH domains. First, we introduced positive and negatively charged amino acids in the framework region of an autonomous human VH domain (VH-B1a) and observed the effect of framework net charge on VH domain refolding, stability, and solubility. Introducing positive charge into the VH-B1a framework increased its thermostability but slightly lowered its refolding ability and solubility. We were not able to obtain correctly folded negatively charged (-VH) VH domains. Second, we introduced a series of positive and negatively charged amino acids in the CDRH1 loop of near-neutral (VH-B1a) and positively charged (+VH) VH domains, and observed their effect on expression, refolding, stability, and solubility. For both the VH-B1a and +VH domains, we observed a decrease in melting temperature (Tm) and room temperature solubility as more negative or positive charged amino acids were added to the CDRH1. The VH-B1a domain had higher room temperature solubility for negative and slightly positive CDRH1 net charges. The + VH domain had higher Tms for all CDRH1 net charges and was better able to tolerate the adverse effects of adding positive charge to CDRH1. We observed a similar response in refolding and solubility of VH-B1a and +VH in response to changes in CDRH1 net charge after temperature-induced denaturation for negative and neutral CDRH1s. We observed a positional effect with a single Lys (31K) and double Lys (31, 32 KK) substitutions in CDRH1, which promoted VH-B1a aggregation and was partially overcome by the +VH domain. In summary, this study provides information for designing VH domains with improved biophysical properties and a +VH domain that will be useful for applications where positive surface charge and CDRH1 are desirable.
Collapse
Affiliation(s)
- Sharmin Sultana
- Department of Biochemistry, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - C Ronald Geyer
- Department of Pathology, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada.
| |
Collapse
|
2
|
Rizk SS, Moustafa DM, ElBanna SA, Nour El-Din HT, Attia AS. Nanobodies in the fight against infectious diseases: repurposing nature's tiny weapons. World J Microbiol Biotechnol 2024; 40:209. [PMID: 38771414 PMCID: PMC11108896 DOI: 10.1007/s11274-024-03990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
Nanobodies are the smallest known antigen-binding molecules to date. Their small size, good tissue penetration, high stability and solubility, ease of expression, refolding ability, and negligible immunogenicity in the human body have granted them excellence over conventional antibodies. Those exceptional attributes of nanobodies make them promising candidates for various applications in biotechnology, medicine, protein engineering, structural biology, food, and agriculture. This review presents an overview of their structure, development methods, advantages, possible challenges, and applications with special emphasis on infectious diseases-related ones. A showcase of how nanobodies can be harnessed for applications including neutralization of viruses and combating antibiotic-resistant bacteria is detailed. Overall, the impact of nanobodies in vaccine design, rapid diagnostics, and targeted therapies, besides exploring their role in deciphering microbial structures and virulence mechanisms are highlighted. Indeed, nanobodies are reshaping the future of infectious disease prevention and treatment.
Collapse
Affiliation(s)
- Soha S Rizk
- Microbiology and Immunology Postgraduate Program, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Dina M Moustafa
- Department of Medical Sciences, Faculty of Dentistry, The British University in Egypt, El Sherouk City, Cairo, 11837, Egypt
| | - Shahira A ElBanna
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Hanzada T Nour El-Din
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed S Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
3
|
Smith CA, Sonneson GJ, Hoey RJ, Hinerman JM, Sheehy K, Walter R, Herr AB, Horn JR. Molecular recognition requires dimerization of a VHH antibody. MAbs 2023; 15:2215363. [PMID: 37243579 PMCID: PMC10228392 DOI: 10.1080/19420862.2023.2215363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/02/2023] [Accepted: 05/15/2023] [Indexed: 05/29/2023] Open
Abstract
Camelid heavy-chain-only antibodies are a unique class of antibody that possesses only a single variable domain (termed VHH) for antigen recognition. Despite their apparent canonical mechanism of target recognition, where a single VHH domain binds a single target, an anti-caffeine VHH has been observed to possess 2:1 stoichiometry. Here, the structure of the anti-caffeine VHH/caffeine complex enabled the generation and biophysical analysis of variants that were used to better understand the role of VHH homodimerization in caffeine recognition. VHH interface mutants and caffeine analogs, which were examined to probe the mechanism of caffeine binding, suggested caffeine recognition is only possible with the VHH dimer species. Correspondingly, in the absence of caffeine, the anti-caffeine VHH was found to form a dimer with a dimerization constant comparable to that observed with VH:VL domains in conventional antibody systems, which was most stable near physiological temperature. While the VHH:VHH dimer structure (at 1.13 Å resolution) is reminiscent of conventional VH:VL heterodimers, the homodimeric VHH possesses a smaller angle of domain interaction, as well as a larger amount of apolar surface area burial. To test the general hypothesis that the short complementarity-determining region-3 (CDR3) may help drive VHH:VHH homodimerization, an anti-picloram VHH domain containing a short CDR3 was generated and characterized, which revealed it also existed as dimer species in solution. These results suggest homodimer-driven recognition may represent a more common method of VHH ligand recognition, opening opportunities for novel VHH homodimer affinity reagents and helping to guide their use in chemically induced dimerization applications.
Collapse
Affiliation(s)
- Christopher A. Smith
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL, USA
| | - Gregory J. Sonneson
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL, USA
| | - Robert J. Hoey
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL, USA
| | - Jennifer M. Hinerman
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Kimberly Sheehy
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL, USA
| | - Richard Walter
- Shamrock Structures, LLC, Woodridge, IL, USA
- Joliet XTALPAC LLC, USA
| | - Andrew B. Herr
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - James R. Horn
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL, USA
| |
Collapse
|
4
|
Liu C, Lin H, Cao L, Wang K, Sui J. Research progress on unique paratope structure, antigen binding modes, and systematic mutagenesis strategies of single-domain antibodies. Front Immunol 2022; 13:1059771. [PMID: 36479130 PMCID: PMC9720397 DOI: 10.3389/fimmu.2022.1059771] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
Single-domain antibodies (sdAbs) showed the incredible advantages of small molecular weight, excellent affinity, specificity, and stability compared with traditional IgG antibodies, so their potential in binding hidden antigen epitopes and hazard detection in food, agricultural and veterinary fields were gradually explored. Moreover, its low immunogenicity, easy-to-carry target drugs, and penetration of the blood-brain barrier have made sdAbs remarkable achievements in medical treatment, toxin neutralization, and medical imaging. With the continuous development and maturity of modern molecular biology, protein analysis software and database with different algorithms, and next-generation sequencing technology, the unique paratope structure and different antigen binding modes of sdAbs compared with traditional IgG antibodies have aroused the broad interests of researchers with the increased related studies. However, the corresponding related summaries are lacking and needed. Different antigens, especially hapten antigens, show distinct binding modes with sdAbs. So, in this paper, the unique paratope structure of sdAbs, different antigen binding cases, and the current maturation strategy of sdAbs were classified and summarized. We hope this review lays a theoretical foundation to elucidate the antigen-binding mechanism of sdAbs and broaden the further application of sdAbs.
Collapse
|
5
|
Macias LA, Wang X, Davies BW, Brodbelt JS. Mapping paratopes of nanobodies using native mass spectrometry and ultraviolet photodissociation. Chem Sci 2022; 13:6610-6618. [PMID: 35756525 PMCID: PMC9172568 DOI: 10.1039/d2sc01536f] [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: 03/16/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022] Open
Abstract
Following immense growth and maturity of the field in the past decade, native mass spectrometry has garnered widespread adoption for the structural characterization of macromolecular complexes. Routine analysis of biotherapeutics by this technique has become commonplace to assist in the development and quality control of immunoglobulin antibodies. Concurrently, 193 nm ultraviolet photodissociation (UVPD) has been developed as a structurally sensitive ion activation technique capable of interrogating protein conformational changes. Here, UVPD was applied to probe the paratopes of nanobodies, a class of single-domain antibodies with an expansive set of applications spanning affinity reagents, molecular imaging, and biotherapeutics. Comparing UVPD sequence fragments for the free nanobodies versus nanobody·antigen complexes empowered assignment of nanobody paratopes and intermolecular salt-bridges, elevating the capabilities of UVPD as a new strategy for characterization of nanobodies. Ultraviolet photodissociation mass spectrometry is used to probe the paratopes of nanobodies, a class of single-domain antibodies, and to determine intersubunit salt-bridges and explore the nanobody·antigen interfaces.![]()
Collapse
Affiliation(s)
- Luis A Macias
- Department of Chemistry, University of Texas at Austin Austin TX 78712 USA
| | - Xun Wang
- Department of Molecular Biosciences, University of Texas at Austin Austin TX 78712 USA
| | - Bryan W Davies
- Department of Molecular Biosciences, University of Texas at Austin Austin TX 78712 USA
| | | |
Collapse
|
6
|
Moliner-Morro A, McInerney GM, Hanke L. Nanobodies in the limelight: Multifunctional tools in the fight against viruses. J Gen Virol 2022; 103. [PMID: 35579613 DOI: 10.1099/jgv.0.001731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antibodies are natural antivirals generated by the vertebrate immune system in response to viral infection or vaccination. Unsurprisingly, they are also key molecules in the virologist's molecular toolbox. With new developments in methods for protein engineering, protein functionalization and application, smaller antibody-derived fragments are moving in focus. Among these, camelid-derived nanobodies play a prominent role. Nanobodies can replace full-sized antibodies in most applications and enable new possible applications for which conventional antibodies are challenging to use. Here we review the versatile nature of nanobodies, discuss their promise as antiviral therapeutics, for diagnostics, and their suitability as research tools to uncover novel aspects of viral infection and disease.
Collapse
Affiliation(s)
- Ainhoa Moliner-Morro
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Gerald M McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
7
|
Camelid Single-Domain Antibodies: Promises and Challenges as Lifesaving Treatments. Int J Mol Sci 2022; 23:ijms23095009. [PMID: 35563400 PMCID: PMC9100996 DOI: 10.3390/ijms23095009] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023] Open
Abstract
Since the discovery of camelid heavy-chain antibodies in 1993, there has been tremendous excitement for these antibody domains (VHHs/sdAbs/nanobodies) as research tools, diagnostics, and therapeutics. Commercially, several patents were granted to pioneering research groups in Belgium and the Netherlands between 1996–2001. Ablynx was established in 2001 with the aim of exploring the therapeutic applications and development of nanobody drugs. Extensive efforts over two decades at Ablynx led to the first approved nanobody drug, caplacizumab (Cablivi) by the EMA and FDA (2018–2019) for the treatment of rare blood clotting disorders in adults with acquired thrombotic thrombocytopenic purpura (TPP). The relatively long development time between camelid sdAb discovery and their entry into the market reflects the novelty of the approach, together with intellectual property restrictions and freedom-to-operate issues. The approval of the first sdAb drug, together with the expiration of key patents, may open a new horizon for the emergence of camelid sdAbs as mainstream biotherapeutics in the years to come. It remains to be seen if nanobody-based drugs will be cheaper than traditional antibodies. In this review, I provide critical perspectives on camelid sdAbs and present the promises and challenges to their widespread adoption as diagnostic and therapeutic agents.
Collapse
|
8
|
Duznovic I, Gräwe A, Weber W, Müller LK, Ali M, Ensinger W, Tietze A, Stein V. Ultrasensitive and Selective Protein Recognition with Nanobody-Functionalized Synthetic Nanopores. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101066. [PMID: 34216425 DOI: 10.1002/smll.202101066] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/17/2021] [Indexed: 06/13/2023]
Abstract
The development of flexible and reconfigurable sensors that can be readily tailored toward different molecular analytes constitutes a key goal and formidable challenge in biosensing. In this regard, synthetic nanopores have emerged as potent physical transducers to convert molecular interactions into electrical signals. Yet, systematic strategies to functionalize their surfaces with receptor proteins for the selective detection of molecular analytes remain scarce. Addressing these limitations, a general strategy is presented to immobilize nanobodies in a directional fashion onto the surface of track-etched nanopores exploiting copper-free click reactions and site-specific protein conjugation systems. The functional immobilization of three different nanobodies is demonstrated in ligand binding experiments with green fluorescent protein, mCherry, and α-amylase (α-Amy) serving as molecular analytes. Ligand binding is resolved using a combination of optical and electrical recordings displaying quantitative dose-response curves. Furthermore, a change in surface charge density is identified as the predominant molecular factor that underlies quantitative dose-responses for the three different protein analytes in nanoconfined geometries. The devised strategy should pave the way for the systematic functionalization of nanopore surfaces with biological receptors and their ability to detect a variety of analytes for diagnostic purposes.
Collapse
Affiliation(s)
- Ivana Duznovic
- Technische Universität Darmstadt, Fachbereich Material- und Geowissenschaften, Fachgebiet Materialanalytik, Alarich-Weiss-Str. 2, D-64287, Darmstadt, Germany
| | - Alexander Gräwe
- Department of Biology, TU Darmstadt, 64287, Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283, Darmstadt, Germany
| | - Wadim Weber
- Department of Biology, TU Darmstadt, 64287, Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283, Darmstadt, Germany
| | - Lena K Müller
- Technische Universität Darmstadt, Clemens-Schöpf Institute of Organic Chemistry and Biochemistry, Alarich-Weiss Str. 4, 64287, Darmstadt, Germany
| | - Mubarak Ali
- Technische Universität Darmstadt, Fachbereich Material- und Geowissenschaften, Fachgebiet Materialanalytik, Alarich-Weiss-Str. 2, D-64287, Darmstadt, Germany
- GSI Helmholtz-Center for Heavy Ion Research, Planckstr. 1, D-64291, Darmstadt, Germany
| | - Wolfgang Ensinger
- Technische Universität Darmstadt, Fachbereich Material- und Geowissenschaften, Fachgebiet Materialanalytik, Alarich-Weiss-Str. 2, D-64287, Darmstadt, Germany
| | - Alesia Tietze
- Technische Universität Darmstadt, Clemens-Schöpf Institute of Organic Chemistry and Biochemistry, Alarich-Weiss Str. 4, 64287, Darmstadt, Germany
- University of Gothenburg, Department of Chemistry and Molecular Biology, Wallenberg Centre for Molecular and Translational Medicine, Kemigården 4, Göteborg, 412 96, Sweden
| | - Viktor Stein
- Department of Biology, TU Darmstadt, 64287, Darmstadt, Germany
- Centre for Synthetic Biology, TU Darmstadt, 64283, Darmstadt, Germany
| |
Collapse
|
9
|
Trinh TTN, Gaubert A, Melani P, Cambillau C, Roussel A, Leone P. Crystal structures of two camelid nanobodies raised against GldL, a component of the type IX secretion system from Flavobacterium johnsoniae. Acta Crystallogr F Struct Biol Commun 2021; 77:171-176. [PMID: 34100775 PMCID: PMC8186413 DOI: 10.1107/s2053230x21005185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/14/2021] [Indexed: 02/09/2023] Open
Abstract
GldL is an inner-membrane protein that is essential for the function of the type IX secretion system (T9SS) in Flavobacterium johnsoniae. The complex that it forms with GldM is supposed to act as a new rotary motor involved in the gliding motility of the bacterium. In the context of structural studies of GldL to gain information on the assembly and function of the T9SS, two camelid nanobodies were selected, produced and purified. Their interaction with the cytoplasmic domain of GldL was characterized and their crystal structures were solved. These nanobodies will be used as crystallization chaperones to help in the crystallization of the cytoplasmic domain of GldL and could also help to solve the structure of the complex using molecular replacement.
Collapse
Affiliation(s)
- Thi Trang Nhung Trinh
- Faculty of Medical Technology, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Vietnam
- PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No. 167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Vietnam
| | - Anaïs Gaubert
- Centre National de la Recherche Scientifique, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
| | - Pauline Melani
- Centre National de la Recherche Scientifique, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
| | - Christian Cambillau
- Centre National de la Recherche Scientifique, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
| | - Alain Roussel
- Centre National de la Recherche Scientifique, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
| | - Philippe Leone
- Centre National de la Recherche Scientifique, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, UMR 7257, Marseille, France
| |
Collapse
|
10
|
Soler MA, Medagli B, Wang J, Oloketuyi S, Bajc G, Huang H, Fortuna S, de Marco A. Effect of Humanizing Mutations on the Stability of the Llama Single-Domain Variable Region. Biomolecules 2021; 11:163. [PMID: 33530572 PMCID: PMC7911018 DOI: 10.3390/biom11020163] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/21/2021] [Accepted: 01/24/2021] [Indexed: 11/24/2022] Open
Abstract
In vivo clinical applications of nanobodies (VHHs) require molecules that induce minimal immunoresponse and therefore possess sequences as similar as possible to the human VH domain. Although the relative sequence variability in llama nanobodies has been used to identify scaffolds with partially humanized signature, the transformation of the Camelidae hallmarks in the framework2 still represents a major problem. We assessed a set of mutants in silico and experimentally to elucidate what is the contribution of single residues to the VHH stability and how their combinations affect the mutant nanobody stability. We described at molecular level how the interaction among residues belonging to different structural elements enabled a model llama nanobody (C8WT, isolated from a naïve library) to be functional and maintain its stability, despite the analysis of its primary sequence would classify it as aggregation-prone. Five chimeras formed by grafting CDRs isolated from different nanobodies into C8WT scaffold were successfully expressed as soluble proteins and both tested clones preserved their antigen binding specificity. We identified a nanobody with human hallmarks that seems suitable for humanizing selected camelid VHHs by grafting heterologous CDRs in its scaffold and could serve for the preparation of a synthetic library of human-like single domains.
Collapse
Affiliation(s)
- Miguel A. Soler
- CONCEPT Lab, Italian Institute of Technology (IIT), 16152 Genova, Italy
| | - Barbara Medagli
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy; (B.M.); (S.F.)
| | - Jiewen Wang
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, China; (J.W.); (H.H.)
| | - Sandra Oloketuyi
- Lab of Environmental and Life Sciences, University of Nova Gorica, 5000 Rožna Dolina-Nova Gorica, Slovenia;
| | - Gregor Bajc
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - He Huang
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, China; (J.W.); (H.H.)
| | - Sara Fortuna
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy; (B.M.); (S.F.)
| | - Ario de Marco
- Lab of Environmental and Life Sciences, University of Nova Gorica, 5000 Rožna Dolina-Nova Gorica, Slovenia;
| |
Collapse
|
11
|
Dhawanjewar AS, Roy AA, Madhusudhan MS. A knowledge-based scoring function to assess quaternary associations of proteins. Bioinformatics 2020; 36:3739-3748. [PMID: 32246820 DOI: 10.1093/bioinformatics/btaa207] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/01/2020] [Accepted: 03/30/2020] [Indexed: 12/21/2022] Open
Abstract
MOTIVATION The elucidation of all inter-protein interactions would significantly enhance our knowledge of cellular processes at a molecular level. Given the enormity of the problem, the expenses and limitations of experimental methods, it is imperative that this problem is tackled computationally. In silico predictions of protein interactions entail sampling different conformations of the purported complex and then scoring these to assess for interaction viability. In this study, we have devised a new scheme for scoring protein-protein interactions. RESULTS Our method, PIZSA (Protein Interaction Z-Score Assessment), is a binary classification scheme for identification of native protein quaternary assemblies (binders/nonbinders) based on statistical potentials. The scoring scheme incorporates residue-residue contact preference on the interface with per residue-pair atomic contributions and accounts for clashes. PIZSA can accurately discriminate between native and non-native structural conformations from protein docking experiments and outperform other contact-based potential scoring functions. The method has been extensively benchmarked and is among the top 6 methods, outperforming 31 other statistical, physics based and machine learning scoring schemes. The PIZSA potentials can also distinguish crystallization artifacts from biological interactions. AVAILABILITY AND IMPLEMENTATION PIZSA is implemented as a web server at http://cospi.iiserpune.ac.in/pizsa and can be downloaded as a standalone package from http://cospi.iiserpune.ac.in/pizsa/Download/Download.html. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Abhilesh S Dhawanjewar
- Indian Institute of Science Education and Research, Pashan, Pune 411008, India.,School of Biological Sciences, University of Nebraska, Lincoln, NE 68588, USA
| | - Ankit A Roy
- Indian Institute of Science Education and Research, Pashan, Pune 411008, India
| | | |
Collapse
|
12
|
Bathula NV, Bommadevara H, Hayes JM. Nanobodies: The Future of Antibody-Based Immune Therapeutics. Cancer Biother Radiopharm 2020; 36:109-122. [PMID: 32936001 DOI: 10.1089/cbr.2020.3941] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeted therapy is a fast evolving treatment strategy to reduce unwanted damage to healthy tissues, while increasing efficacy and specificity. Driven by state-of-the-art technology, this therapeutic approach is especially true of cancer. Antibodies with their remarkable specificity have revolutionized therapeutic strategies for autoimmune conditions and cancer, particularly blood-borne cancers, but have severe limitations in treating solid tumors. This is mainly due to their large molecular size, low stability, tumor-tissue penetration difficulties, and pharmacokinetic properties. The tumor microenvironment, rich in immune-suppressing molecules is also a major barrier in targeting solid tumors by antibody-based drugs. Nanobodies have recently emerged as an alternative therapeutic agent to overcome some of the drawbacks of traditional antibody treatment. Nanobodies are the VHH domains found on the heavy-chain only antibodies of camelids and are the smallest naturally available antibody fragments with excellent antigen-binding specificity and affinity, equivalent to conventional antibodies but with molecular weights as low as 15 kDa. The compact size, high stability, enhanced hydrophilicity, particularly in framework regions, excellent epitope interactions with protruding CDR3 regions, and improved tissue penetration make nanobodies the next-generation therapeutics (Nano-BioDrugs). In this review, the authors discuss the interesting properties of nanobodies and their advantages over their conventional counterparts and provide insight into how nanobodies are being utilized as agonists and antagonists, bispecific constructs, and drug and enzyme-conjugates to combat the tumor microenvironment and treat disease.
Collapse
Affiliation(s)
- Nuthan V Bathula
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Hemashree Bommadevara
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Jerrard M Hayes
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Hirano Y, Tsukamoto K, Ariki S, Naka Y, Ueda M, Tamada T. X-ray crystallographic structural studies of α-amylase I from Eisenia fetida. Acta Crystallogr D Struct Biol 2020; 76:834-844. [DOI: 10.1107/s2059798320010165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/23/2020] [Indexed: 11/11/2022] Open
Abstract
The earthworm Eisenia fetida possesses several cold-active enzymes, including α-amylase, β-glucanase and β-mannanase. E. fetida possesses two isoforms of α-amylase (Ef-Amy I and II) to digest raw starch. Ef-Amy I retains its catalytic activity at temperatures below 10°C. To identify the molecular properties of Ef-Amy I, X-ray crystal structures were determined of the wild type and of the inactive E249Q mutant. Ef-Amy I has structural similarities to mammalian α-amylases, including the porcine pancreatic and human pancreatic α-amylases. Structural comparisons of the overall structures as well as of the Ca2+-binding sites of Ef-Amy I and the mammalian α-amylases indicate that Ef-Amy I has increased structural flexibility and more solvent-exposed acidic residues. These structural features of Ef-Amy I may contribute to its observed catalytic activity at low temperatures, as many cold-adapted enzymes have similar structural properties. The structure of the substrate complex of the inactive mutant of Ef-Amy I shows that a maltohexaose molecule is bound in the active site and a maltotetraose molecule is bound in the cleft between the N- and C-terminal domains. The recognition of substrate molecules by Ef-Amy I exhibits some differences from that observed in structures of human pancreatic α-amylase. This result provides insights into the structural modulation of the recognition of substrates and inhibitors.
Collapse
|
14
|
Rudolph MJ, Czajka TF, Davis SA, Thi Nguyen CM, Li XP, Tumer NE, Vance DJ, Mantis NJ. Intracellular Neutralization of Ricin Toxin by Single-domain Antibodies Targeting the Active Site. J Mol Biol 2020; 432:1109-1125. [PMID: 31931008 PMCID: PMC7066583 DOI: 10.1016/j.jmb.2020.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/16/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
The extreme potency of the plant toxin, ricin, is due to its enzymatic subunit, RTA, which inactivates mammalian ribosomes with near-perfect efficiency. Here we characterized, at the functional and structural levels, seven alpaca single-domain antibodies (VHHs) previously reported to recognize epitopes in proximity to RTA's active site. Three of the VHHs, V2A11, V8E6, and V2G10, were potent inhibitors of RTA in vitro and protected Vero cells from ricin when expressed as intracellular antibodies ("intrabodies"). Crystal structure analysis revealed that the complementarity-determining region 3 (CDR3) elements of V2A11 and V8E6 penetrate RTA's active site and interact with key catalytic residues. V2G10, by contrast, sits atop the enzymatic pocket and occludes substrate accessibility. The other four VHHs also penetrated/occluded RTA's active site, but lacked sufficient binding affinities to outcompete RTA-ribosome interactions. Intracellular delivery of high-affinity, single-domain antibodies may offer a new avenue in the development of countermeasures against ricin toxin.toxin, antibody, structure, intracellular.
Collapse
Affiliation(s)
- Michael J Rudolph
- New York Structural Biology Center, New York, NY 10027, United States.
| | - Timothy F Czajka
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, 12208, United States; Department of Biomedical Sciences, University at Albany, Albany, NY 12201, United States
| | - Simon A Davis
- New York Structural Biology Center, New York, NY 10027, United States
| | - Chi My Thi Nguyen
- New York Structural Biology Center, New York, NY 10027, United States
| | - Xiao-Ping Li
- Department of Plant Biology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
| | - Nilgun E Tumer
- Department of Plant Biology, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
| | - David J Vance
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, 12208, United States
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, 12208, United States; Department of Biomedical Sciences, University at Albany, Albany, NY 12201, United States.
| |
Collapse
|
15
|
Hanlon A, Metjian A. Caplacizumab in adult patients with acquired thrombotic thrombocytopenic purpura. Ther Adv Hematol 2020; 11:2040620720902904. [PMID: 32095224 PMCID: PMC7011322 DOI: 10.1177/2040620720902904] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/29/2019] [Indexed: 12/27/2022] Open
Abstract
Thrombotic thrombocytopenic purpura (TTP) is usually a fatal disease caused by a
deficiency of the metalloproteinase, ADAMTS13, often due to autoimmunity. This
leads to the development of pathogenic multimers of von Willebrand factor (vWF),
causing an inappropriate interaction of platelets and vWF. This results in a
thrombotic microangiopathy, which is treated with therapeutic plasma exchange
and immune suppression. Although this treatment has reduced the mortality of TTP
to only about 20%, there have been no recent significant advances in the
treatment of TTP. Recently, a novel agent has been approved for use in TTP.
Caplacizumab, which binds to the A1 domain of vWF, prevents the adhesion of
platelets to vWF. It is a first in-class ‘nanobody’, that in clinical trials has
shown marked efficacy in treating TTP and its complications. This review will
discuss the development and implications of caplacizumab in the treatment of
TTP.
Collapse
Affiliation(s)
| | - Ara Metjian
- Duke University, Box #3422, 100 Trent Drive, Durham, NC 27708-0187, USA
| |
Collapse
|
16
|
Hoey RJ, Eom H, Horn JR. Structure and development of single domain antibodies as modules for therapeutics and diagnostics. Exp Biol Med (Maywood) 2019; 244:1568-1576. [PMID: 31594404 PMCID: PMC6920669 DOI: 10.1177/1535370219881129] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Since their discovery just over 25 years ago, the single variable domain from heavy-chain-only antibodies plays a role in an increasing number of antibody-based applications. Structural and biophysical studies have revealed that the small, ∼15 kDa, single variable domain found in camelids displays versatility in target recognition. Such insight has served as the foundation to develop and engineer VHH domains with enhanced properties capable of targeting a range of therapeutically relevant protein antigens or low-molecular weight haptens. Furthermore, the modular nature of VHH domains allows them to be introduced into constructs that are simply not possible with conventional antibodies. Here, we review the structural and biophysical properties of VHH domains, highlight recent VHH-based therapeutics and diagnostics, and provide insight into VHH engineering that may pave the way to next-generation single domain antibody applications.
Collapse
Affiliation(s)
- Robert J Hoey
- Department of Chemistry and Biochemistry,
Northern
Illinois University, DeKalb, IL 60115,
USA
| | - Hyeyoung Eom
- Department of Chemistry and Biochemistry,
Northern
Illinois University, DeKalb, IL 60115,
USA
| | - James R Horn
- Department of Chemistry and Biochemistry,
Northern
Illinois University, DeKalb, IL 60115,
USA
| |
Collapse
|
17
|
Sherwood LJ, Taylor AB, Hart PJ, Hayhurst A. Paratope Duality and Gullying are Among the Atypical Recognition Mechanisms Used by a Trio of Nanobodies to Differentiate Ebolavirus Nucleoproteins. J Mol Biol 2019; 431:4848-4867. [PMID: 31626803 PMCID: PMC6990103 DOI: 10.1016/j.jmb.2019.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/28/2019] [Accepted: 10/07/2019] [Indexed: 02/08/2023]
Abstract
We had previously shown that three anti–Marburg virus nanobodies (VHH or single-domain antibody [sdAb]) targeted a cryptotope within an alpha-helical assembly at the nucleoprotein (NP) C-terminus that was conserved through half a century of viral evolution. Here, we wished to determine whether an anti–Ebola virus sdAb, that was cross-reactive within the Ebolavirus genus, recognized a similar structural feature upstream of the ebolavirus NP C-terminus. In addition, we sought to determine whether the specificities of a less cross-reactive anti–Zaire ebolavirus sdAb and a totally specific anti–Sudan ebolavirus sdAb were the result of exclusion from this region. Binding and X-ray crystallographic studies revealed that the primary determinant of cross-reactivity did indeed appear to be a preference for the helical feature. Specificity, in the case of the Zaire ebolavirus–specific sdAb, arose from the footprint shifting away from the helices to engage more variable residues. While both sdAbs used CDRs, they also had atypical side-on approaches, with framework 2 helping to accommodate parts of the epitope in sizeable paratope gullies. The Sudan ebolavirus–specific sdAb was more remarkable and appeared to bind two C-terminal domains simultaneously via nonoverlapping epitopes—“paratope duality.” One mode involved paratope gullying, whereas the other involved only CDRs, with CDR3 restructuring to wedge in between opposing walls of an interdomain crevice. The varied routes used by sdAbs to engage antigens discovered here deepen our appreciation of the small scaffold’s architectural versatility and also reveal lucrative opportunities within the ebolavirus NP C-termini that might be leveraged for diagnostics and novel therapeutic targeting.
Collapse
Affiliation(s)
- Laura Jo Sherwood
- Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Alexander Bryan Taylor
- X-ray Crystallography Core Laboratory, Institutional Research Cores and Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Peter John Hart
- X-ray Crystallography Core Laboratory, Institutional Research Cores and Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA; Department of Veterans Affairs, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
| | - Andrew Hayhurst
- Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA. http://
| |
Collapse
|
18
|
Yu W, Jo S, Lakkaraju SK, Weber DJ, MacKerell AD. Exploring protein-protein interactions using the site-identification by ligand competitive saturation methodology. Proteins 2019; 87:289-301. [PMID: 30582220 PMCID: PMC6408985 DOI: 10.1002/prot.25650] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/06/2018] [Accepted: 12/19/2018] [Indexed: 01/05/2023]
Abstract
Protein docking methods are powerful computational tools to study protein-protein interactions (PPI). While a significant number of docking algorithms have been developed, they are usually based on rigid protein models or with limited considerations of protein flexibility and the desolvation effect is rarely considered in docking energy functions, which may lower the accuracy of the predictions. To address these issues, we introduce a PPI energy function based on the site-identification by ligand competitive saturation (SILCS) framework and utilize the fast Fourier transform (FFT) correlation approach. The free energy content of the SILCS FragMaps represent an alternative to traditional energy grids and they can be efficiently utilized to guide FFT-based protein docking. Application of the approach to eight diverse test cases, including seven from Protein Docking Benchmark 5.0, showed the PPI prediction using SILCS approach (SILCS-PPI) to be competitive with several commonly used protein docking methods indicating that the method has the ability to both qualitatively and quantitatively inform the prediction of PPI. Results show the utility of the SILCS-PPI docking approach for determination of probability distributions of PPI interactions over the surface of both partner proteins, allowing for identification of alternate binding poses. Such binding poses are confirmed by experimental crystal contacts in our test cases. While more computationally demanding than available PPI docking technologies, we anticipate that the SILCS-PPI docking approach will offer an alternative methodology for improved evaluation of PPIs that could be used in a variety of fields from systems biology to excipient design for biologics-based drugs.
Collapse
Affiliation(s)
- Wenbo Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD 20850
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD 21201
| | | | | | - David J. Weber
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD 20850
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Alexander D. MacKerell
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201
- Institute for Bioscience and Biotechnology Research (IBBR), Rockville, MD 20850
- Center for Biomolecular Therapeutics (CBT), School of Medicine, University of Maryland, Baltimore, MD 21201
- SilcsBio LLC, Baltimore, MD 21202
| |
Collapse
|
19
|
Zavrtanik U, Lukan J, Loris R, Lah J, Hadži S. Structural Basis of Epitope Recognition by Heavy-Chain Camelid Antibodies. J Mol Biol 2018; 430:4369-4386. [PMID: 30205092 DOI: 10.1016/j.jmb.2018.09.002] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 10/28/2022]
Abstract
Truncated versions of heavy-chain antibodies (HCAbs) from camelids, also termed nanobodies, comprise only one-tenth the mass of conventional antibodies, yet retain similar, high binding affinities for the antigens. Here we analyze a large data set of nanobody-antigen crystal structures and investigate how nanobody-antigen recognition compares to the one by conventional antibodies. We find that nanobody paratopes are enriched in aromatic residues just like conventional antibodies, but additionally, they also bear a more hydrophobic character. Most striking differences were observed in the characteristics of the antigen's epitope. Unlike conventional antibodies, nanobodies bind to more rigid, concave, conserved and structured epitopes enriched with aromatic residues. Nanobodies establish fewer interactions with the antigens compared to conventional antibodies, and we speculate that high binding affinities are achieved due to less unfavorable conformational and more favorable solvation entropy contributions. We observed that interactions with antigen are mediated not only by three CDR loops but also by numerous residues from the nanobody framework. These residues are not distributed uniformly; rather, they are concentrated into four structurally distinct regions and mediate mostly charged interactions. Our findings suggest that in some respects nanobody-antigen interactions are more similar to the general protein-protein interactions rather than antibody-antigen interactions.
Collapse
Affiliation(s)
- Uroš Zavrtanik
- Department of Physical Chemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Junoš Lukan
- Statistical Office of the Republic of Slovenia, Litostrojska cesta 54, 1000 Ljubljana, Slovenia
| | - Remy Loris
- Structural Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel, B-1050 Brussel, Belgium; VIB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050 Brussel, Belgium
| | - Jurij Lah
- Department of Physical Chemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - San Hadži
- Department of Physical Chemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; Structural Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel, B-1050 Brussel, Belgium; VIB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050 Brussel, Belgium.
| |
Collapse
|
20
|
Abstract
Single-domain antibodies (sdAbs), the autonomous variable domains of heavy chain-only antibodies produced naturally by camelid ungulates and cartilaginous fishes, have evolved to bind antigen using only three complementarity-determining region (CDR) loops rather than the six present in conventional VH:VL antibodies. It has been suggested, based on limited evidence, that sdAbs may adopt paratope structures that predispose them to preferential recognition of recessed protein epitopes, but poor or non-recognition of protuberant epitopes and small molecules. Here, we comprehensively surveyed the evidence in support of this hypothesis. We found some support for a global structural difference in the paratope shapes of sdAbs compared with those of conventional antibodies: sdAb paratopes have smaller molecular surface areas and diameters, more commonly have non-canonical CDR1 and CDR2 structures, and have elongated CDR3 length distributions, but have similar amino acid compositions and are no more extended (interatomic distance measured from CDR base to tip) than conventional antibody paratopes. Comparison of X-ray crystal structures of sdAbs and conventional antibodies in complex with cognate antigens showed that sdAbs and conventional antibodies bury similar solvent-exposed surface areas on proteins and form similar types of non-covalent interactions, although these are more concentrated in the compact sdAb paratope. Thus, sdAbs likely have privileged access to distinct antigenic regions on proteins, but only owing to their small molecular size and not to general differences in molecular recognition mechanism. The evidence surrounding the purported inability of sdAbs to bind small molecules was less clear. The available data provide a structural framework for understanding the evolutionary emergence and function of autonomous heavy chain-only antibodies.
Collapse
Affiliation(s)
- Kevin A Henry
- a Human Health Therapeutics Research Centre , National Research Council Canada , Ottawa , Ontario , Canada
| | - C Roger MacKenzie
- a Human Health Therapeutics Research Centre , National Research Council Canada , Ottawa , Ontario , Canada.,b School of Environmental Sciences , University of Guelph , Guelph , Ontario , Canada
| |
Collapse
|
21
|
Tabares-da Rosa S, Wogulis LA, Wogulis MD, González-Sapienza G, Wilson DK. Structure and specificity of several triclocarban-binding single domain camelid antibody fragments. J Mol Recognit 2018; 32:e2755. [PMID: 30033524 DOI: 10.1002/jmr.2755] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 12/29/2022]
Abstract
The variable VHH domains of camelid single chain antibodies have been useful in numerous biotechnology applications due to their simplicity, biophysical properties, and abilities to bind to their cognate antigens with high affinities and specificity. Their interactions with proteins have been well-studied, but considerably less work has been done to characterize their ability to bind haptens. A high-resolution structural study of three nanobodies (T4, T9, and T10) which have been shown to bind triclocarban (TCC, 3-(4-chlorophenyl)-1-(3,4-dichlorophenyl)urea) with near-nanomolar affinity shows that binding occurs in a tunnel largely formed by CDR1 rather than a surface or lateral binding mode seen in other nanobody-hapten interactions. Additional significant interactions are formed with a non-hypervariable loop, sometimes dubbed "CDR4". A comparison of apo and holo forms of T9 and T10 shows that the binding site undergoes little conformational change upon binding of TCC. Structures of three nanobody-TCC complexes demonstrated there was not a standard binding mode. T4 and T9 have a high degree of sequence identity and bind the hapten in a nearly identical manner, while the more divergent T10 binds TCC in a slightly displaced orientation with the urea moiety rotated approximately 180° along the long axis of the molecule. In addition to methotrexate, this is the second report of haptens binding in a tunnel formed by CDR1, suggesting that compounds with similar hydrophobicity and shape could be recognized by nanobodies in analogous fashion. Structure-guided mutations failed to improve binding affinity for T4 and T9 underscoring the high degree of natural optimization.
Collapse
Affiliation(s)
| | - Linda A Wogulis
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | - Mark D Wogulis
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | | | - David K Wilson
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| |
Collapse
|
22
|
Chaisri U, Chaicumpa W. Evolution of Therapeutic Antibodies, Influenza Virus Biology, Influenza, and Influenza Immunotherapy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9747549. [PMID: 29998138 PMCID: PMC5994580 DOI: 10.1155/2018/9747549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/19/2018] [Accepted: 03/31/2018] [Indexed: 02/07/2023]
Abstract
This narrative review article summarizes past and current technologies for generating antibodies for passive immunization/immunotherapy. Contemporary DNA and protein technologies have facilitated the development of engineered therapeutic monoclonal antibodies in a variety of formats according to the required effector functions. Chimeric, humanized, and human monoclonal antibodies to antigenic/epitopic myriads with less immunogenicity than animal-derived antibodies in human recipients can be produced in vitro. Immunotherapy with ready-to-use antibodies has gained wide acceptance as a powerful treatment against both infectious and noninfectious diseases. Influenza, a highly contagious disease, precipitates annual epidemics and occasional pandemics, resulting in high health and economic burden worldwide. Currently available drugs are becoming less and less effective against this rapidly mutating virus. Alternative treatment strategies are needed, particularly for individuals at high risk for severe morbidity. In a setting where vaccines are not yet protective or available, human antibodies that are broadly effective against various influenza subtypes could be highly efficacious in lowering morbidity and mortality and controlling unprecedented epidemic/pandemic. Prototypes of human single-chain antibodies to several conserved proteins of influenza virus with no Fc portion (hence, no ADE effect in recipients) are available. These antibodies have high potential as a novel, safe, and effective anti-influenza agent.
Collapse
Affiliation(s)
- Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
23
|
Chen F, Liu H, Sun H, Pan P, Li Y, Li D, Hou T. Assessing the performance of the MM/PBSA and MM/GBSA methods. 6. Capability to predict protein-protein binding free energies and re-rank binding poses generated by protein-protein docking. Phys Chem Chem Phys 2018; 18:22129-39. [PMID: 27444142 DOI: 10.1039/c6cp03670h] [Citation(s) in RCA: 355] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Understanding protein-protein interactions (PPIs) is quite important to elucidate crucial biological processes and even design compounds that interfere with PPIs with pharmaceutical significance. Protein-protein docking can afford the atomic structural details of protein-protein complexes, but the accurate prediction of the three-dimensional structures for protein-protein systems is still notoriously difficult due in part to the lack of an ideal scoring function for protein-protein docking. Compared with most scoring functions used in protein-protein docking, the Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) and Molecular Mechanics/Poisson Boltzmann Surface Area (MM/PBSA) methodologies are more theoretically rigorous, but their overall performance for the predictions of binding affinities and binding poses for protein-protein systems has not been systematically evaluated. In this study, we first evaluated the performance of MM/PBSA and MM/GBSA to predict the binding affinities for 46 protein-protein complexes. On the whole, different force fields, solvation models, and interior dielectric constants have obvious impacts on the prediction accuracy of MM/GBSA and MM/PBSA. The MM/GBSA calculations based on the ff02 force field, the GB model developed by Onufriev et al. and a low interior dielectric constant (εin = 1) yield the best correlation between the predicted binding affinities and the experimental data (rp = -0.647), which is better than MM/PBSA (rp = -0.523) and a number of empirical scoring functions used in protein-protein docking (rp = -0.141 to -0.529). Then, we examined the capability of MM/GBSA to identify the possible near-native binding structures from the decoys generated by ZDOCK for 43 protein-protein systems. The results illustrate that the MM/GBSA rescoring has better capability to distinguish the correct binding structures from the decoys than the ZDOCK scoring. Besides, the optimal interior dielectric constant of MM/GBSA for re-ranking docking poses may be determined by analyzing the characteristics of protein-protein binding interfaces. Considering the relatively high prediction accuracy and low computational cost, MM/GBSA may be a good choice for predicting the binding affinities and identifying correct binding structures for protein-protein systems.
Collapse
Affiliation(s)
- Fu Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Hui Liu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Huiyong Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Peichen Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Youyong Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Dan Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China. and State Key Lab of CAD&CG, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| |
Collapse
|
24
|
Arbabi-Ghahroudi M. Camelid Single-Domain Antibodies: Historical Perspective and Future Outlook. Front Immunol 2017; 8:1589. [PMID: 29209322 PMCID: PMC5701970 DOI: 10.3389/fimmu.2017.01589] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/03/2017] [Indexed: 11/13/2022] Open
Abstract
Tremendous effort has been expended over the past two and a half decades to understand many aspects of camelid heavy chain antibodies, from their biology, evolution, and immunogenetics to their potential applications in various fields of research and medicine. In this article, I present a historical perspective on the development of camelid single-domain antibodies (sdAbs or VHHs, also widely known as nanobodies) since their discovery and discuss the advantages and disadvantages of these unique molecules in various areas of research, industry, and medicine. Commercialization of camelid sdAbs exploded in 2001 with a flurry of patents issued to the Vrije Universiteit Brussel (VUB) and later taken on by the Vlaams Interuniversitair Instituut voor Biotechnologie (VIB) and, after 2002, the VIB-founded spin-off company, Ablynx. While entrepreneurial spirit has certainly catalyzed the exploration of nanobodies as marketable products, IP restrictions may be partially responsible for the relatively long time span between the discovery of these biomolecules and their entry into the pharmaceutical market. It is now anticipated that the first VHH-based antibody drug, Caplacizumab, a bivalent anti-vWF antibody for treating rare blood clotting disorders, may be approved and commercialized in 2018 or shortly thereafter. This elusive first approval, along with the expiry of key patents, may substantially alter the scientific and biomedical landscape surrounding camelid sdAbs and pave the way for their emergence as mainstream biotherapeutics.
Collapse
Affiliation(s)
- Mehdi Arbabi-Ghahroudi
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada.,Department of Biology, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
25
|
Enhanced expression and purification of camelid single domain VHH antibodies from classical inclusion bodies. Protein Expr Purif 2017; 136:39-44. [DOI: 10.1016/j.pep.2017.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/10/2017] [Accepted: 02/11/2017] [Indexed: 01/05/2023]
|
26
|
Meera C, Meenakumari S, Thirumavalavan M, Pachaiappan R. Isolation and characterization of α-amylase inhibitor from Leucas aspera (Willd) Link: α-amylase assay combined with FPLC chromatography for expedited identification. JOURNAL OF PLANT BIOCHEMISTRY AND BIOTECHNOLOGY 2017; 26:346-355. [DOI: 10.1007/s13562-017-0397-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
|
27
|
Nguyen VS, Logger L, Spinelli S, Legrand P, Huyen Pham TT, Nhung Trinh TT, Cherrak Y, Zoued A, Desmyter A, Durand E, Roussel A, Kellenberger C, Cascales E, Cambillau C. Type VI secretion TssK baseplate protein exhibits structural similarity with phage receptor-binding proteins and evolved to bind the membrane complex. Nat Microbiol 2017. [DOI: 10.1038/nmicrobiol.2017.103] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
28
|
Maggi M, Scotti C. Data on enhanced expression and purification of camelid single domain antibodies from Escherichia coli classical inclusion bodies. Data Brief 2017; 12:132-137. [PMID: 28413818 PMCID: PMC5384857 DOI: 10.1016/j.dib.2017.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/08/2017] [Accepted: 03/28/2017] [Indexed: 11/04/2022] Open
Abstract
Heterologous expression of high amounts of recombinant proteins is a milestone for research and industrial purposes. Single domain antibodies (sdAbs) are heavy-chain only antibody fragments with applications in the biotechnological, medical and industrial fields. The simple nature and small size of sdAbs allows for efficient expression of the soluble molecule in different hosts. However, in some cases, it results in low functional protein yield. To overcome this limitation, expression of a 6xHistag sdAb was attempted in different conditions in Escherichia coli BL21(DE3) cells. Data showed that high amount of sdAb can be expressed in E. coli classical inclusion bodies, efficiently extracted by urea in a short-time, and properly purified by metal ion affinity chromatography. These data originate from the research article "Enhanced expression and purification of camelid single domain VHH antibodies from classical inclusion bodies" Maggi and Scotti (2017) [1] (DOI: http://dx.doi.org/10.1016/j.pep.2017.02.007).
Collapse
Affiliation(s)
- Maristella Maggi
- Department of Molecular Medicine, Unit of Immunology and General Pathology, University of Pavia, Via Ferrata, 9, 27100 Pavia, Italy
| | - Claudia Scotti
- Department of Molecular Medicine, Unit of Immunology and General Pathology, University of Pavia, Via Ferrata, 9, 27100 Pavia, Italy
| |
Collapse
|
29
|
Rapid Discovery of Potent and Selective Glycosidase-Inhibiting De Novo Peptides. Cell Chem Biol 2017; 24:381-390. [PMID: 28262556 DOI: 10.1016/j.chembiol.2017.02.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 01/14/2017] [Accepted: 01/31/2017] [Indexed: 12/20/2022]
Abstract
Human pancreatic α-amylase (HPA) is responsible for degrading starch to malto-oligosaccharides, thence to glucose, and is therefore an attractive therapeutic target for the treatment of diabetes and obesity. Here we report the discovery of a unique lariat nonapeptide, by means of the RaPID (Random non-standard Peptides Integrated Discovery) system, composed of five amino acids in a head-to-side-chain thioether macrocycle and a further four amino acids in a 310 helical C terminus. This is a potent inhibitor of HPA (Ki = 7 nM) yet exhibits selectivity for the target over other glycosidases tested. Structural studies show that this nonapeptide forms a compact tertiary structure, and illustrate that a general inhibitory motif involving two phenolic groups is often accessed for tight binding of inhibitors to HPA. Furthermore, the work reported here demonstrates the potential of this methodology for the discovery of de novo peptide inhibitors against other glycosidases.
Collapse
|
30
|
Noël F, Malpertuy A, de Brevern AG. Global analysis of VHHs framework regions with a structural alphabet. Biochimie 2016; 131:11-19. [PMID: 27613403 DOI: 10.1016/j.biochi.2016.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/05/2016] [Accepted: 09/05/2016] [Indexed: 02/08/2023]
Abstract
The VHHs are antigen-binding region/domain of camelid heavy chain antibodies (HCAb). They have many interesting biotechnological and biomedical properties due to their small size, high solubility and stability, and high affinity and specificity for their antigens. HCAb and classical IgGs are evolutionary related and share a common fold. VHHs are composed of regions considered as constant, called the frameworks (FRs) connected by Complementarity Determining Regions (CDRs), a highly variable region that provide interaction with the epitope. Actually, no systematic structural analyses had been performed on VHH structures despite a significant number of structures. This work is the first study to analyse the structural diversity of FRs of VHHs. Using a structural alphabet that allows approximating the local conformation, we show that each of the four FRs do not have a unique structure but exhibit many structural variant patterns. Moreover, no direct simple link between the local conformational change and amino acid composition can be detected. These results indicate that long-range interactions affect the local conformation of FRs and impact the building of structural models.
Collapse
Affiliation(s)
- Floriane Noël
- INSERM, U 1134, DSIMB, F-75739 Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1134, F-75739 Paris, France; Institut National de la Transfusion Sanguine (INTS), F-75739 Paris, France; Laboratoire d'Excellence GR-Ex, F-75739 Paris, France
| | | | - Alexandre G de Brevern
- INSERM, U 1134, DSIMB, F-75739 Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1134, F-75739 Paris, France; Institut National de la Transfusion Sanguine (INTS), F-75739 Paris, France; Laboratoire d'Excellence GR-Ex, F-75739 Paris, France.
| |
Collapse
|
31
|
Kumada Y, Kang B, Yamakawa K, Kishimoto M, Horiuchi JI. Efficient preparation and site-directed immobilization of VHH antibodies by genetic fusion of poly(methylmethacrylate)-binding peptide (PMMA-Tag). Biotechnol Prog 2015; 31:1563-70. [DOI: 10.1002/btpr.2169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/07/2015] [Indexed: 01/21/2023]
Affiliation(s)
- Yoichi Kumada
- Dept. of Biomolecular Engineering; Kyoto Institute of Technology; Hashigami-Cho 1, Matsugasaki, Sakyo-Ku Kyoto 606-8585 Japan
| | - Bongmun Kang
- Venture Laboratory; Kyoto Institute of Technology; Hashigami-Cho 1, Matsugasaki, Sakyo-Ku Kyoto 606-8585 Japan
| | - Kagenari Yamakawa
- Dept. of Biomolecular Engineering; Kyoto Institute of Technology; Hashigami-Cho 1, Matsugasaki, Sakyo-Ku Kyoto 606-8585 Japan
| | - Michimasa Kishimoto
- Dept. of Biomolecular Engineering; Kyoto Institute of Technology; Hashigami-Cho 1, Matsugasaki, Sakyo-Ku Kyoto 606-8585 Japan
| | - Jun-Ichi Horiuchi
- Dept. of Biomolecular Engineering; Kyoto Institute of Technology; Hashigami-Cho 1, Matsugasaki, Sakyo-Ku Kyoto 606-8585 Japan
| |
Collapse
|
32
|
Dorresteijn B, Rotman M, Faber D, Schravesande R, Suidgeest E, van der Weerd L, van der Maarel SM, Verrips CT, El Khattabi M. Camelid heavy chain only antibody fragment domain against β-site of amyloid precursor protein cleaving enzyme 1 inhibits β-secretase activityin vitroandin vivo. FEBS J 2015; 282:3618-31. [DOI: 10.1111/febs.13367] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/24/2015] [Accepted: 07/01/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Bram Dorresteijn
- Biomolecular Imaging Group; Division of Cell Biology; Department of Biology; Faculty of Science; Utrecht University; The Netherlands
| | - Maarten Rotman
- Department of Radiology; Leiden University Medical Center; Leiden The Netherlands
- Department of Human Genetics; Leiden University Medical Center; Leiden The Netherlands
| | - Dorien Faber
- Biomolecular Imaging Group; Division of Cell Biology; Department of Biology; Faculty of Science; Utrecht University; The Netherlands
| | - Ruud Schravesande
- Biomolecular Imaging Group; Division of Cell Biology; Department of Biology; Faculty of Science; Utrecht University; The Netherlands
| | - Ernst Suidgeest
- Department of Radiology; Leiden University Medical Center; Leiden The Netherlands
| | - Louise van der Weerd
- Department of Radiology; Leiden University Medical Center; Leiden The Netherlands
- Department of Human Genetics; Leiden University Medical Center; Leiden The Netherlands
| | | | - Cornelis T. Verrips
- Biomolecular Imaging Group; Division of Cell Biology; Department of Biology; Faculty of Science; Utrecht University; The Netherlands
- QVQ Holding BV; Utrecht The Netherlands
| | - Mohamed El Khattabi
- Biomolecular Imaging Group; Division of Cell Biology; Department of Biology; Faculty of Science; Utrecht University; The Netherlands
- QVQ Holding BV; Utrecht The Netherlands
| |
Collapse
|
33
|
Pain C, Dumont J, Dumoulin M. Camelid single-domain antibody fragments: Uses and prospects to investigate protein misfolding and aggregation, and to treat diseases associated with these phenomena. Biochimie 2015; 111:82-106. [DOI: 10.1016/j.biochi.2015.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/23/2015] [Indexed: 12/19/2022]
|
34
|
Scior T, Paiz-Candia B, Islas ÁA, Sánchez-Solano A, Millan-Perez Peña L, Mancilla-Simbro C, Salinas-Stefanon EM. Predicting a double mutant in the twilight zone of low homology modeling for the skeletal muscle voltage-gated sodium channel subunit beta-1 (Nav1.4 β1). Comput Struct Biotechnol J 2015; 13:229-40. [PMID: 25904995 PMCID: PMC4402383 DOI: 10.1016/j.csbj.2015.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/18/2015] [Accepted: 03/21/2015] [Indexed: 12/18/2022] Open
Abstract
The molecular structure modeling of the β1 subunit of the skeletal muscle voltage-gated sodium channel (Nav1.4) was carried out in the twilight zone of very low homology. Structural significance can per se be confounded with random sequence similarities. Hence, we combined (i) not automated computational modeling of weakly homologous 3D templates, some with interfaces to analogous structures to the pore-bearing Nav1.4 α subunit with (ii) site-directed mutagenesis (SDM), as well as (iii) electrophysiological experiments to study the structure and function of the β1 subunit. Despite the distant phylogenic relationships, we found a 3D-template to identify two adjacent amino acids leading to the long-awaited loss of function (inactivation) of Nav1.4 channels. This mutant type (T109A, N110A, herein called TANA) was expressed and tested on cells of hamster ovary (CHO). The present electrophysiological results showed that the double alanine substitution TANA disrupted channel inactivation as if the β1 subunit would not be in complex with the α subunit. Exhaustive and unbiased sampling of “all β proteins” (Ig-like, Ig) resulted in a plethora of 3D templates which were compared to the target secondary structure prediction. The location of TANA was made possible thanks to another “all β protein” structure in complex with an irreversible bound protein as well as a reversible protein–protein interface (our “Rosetta Stone” effect). This finding coincides with our electrophysiological data (disrupted β1-like voltage dependence) and it is safe to utter that the Nav1.4 α/β1 interface is likely to be of reversible nature.
Collapse
Affiliation(s)
- Thomas Scior
- Facultad de Ciencias Químicas, Universidad Autónoma de Puebla, Puebla, Mexico
| | - Bertin Paiz-Candia
- Facultad de Ciencias Químicas, Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ángel A Islas
- Laboratorio de Biofísica, Instituto de Fisiología, Universidad Autónoma de Puebla, Puebla, Mexico
| | - Alfredo Sánchez-Solano
- Laboratorio de Biofísica, Instituto de Fisiología, Universidad Autónoma de Puebla, Puebla, Mexico
| | | | - Claudia Mancilla-Simbro
- Laboratorio de Biofísica, Instituto de Fisiología, Universidad Autónoma de Puebla, Puebla, Mexico
| | | |
Collapse
|
35
|
Segura J, Marín-López MA, Jones PF, Oliva B, Fernandez-Fuentes N. VORFFIP-driven dock: V-D2OCK, a fast and accurate protein docking strategy. PLoS One 2015; 10:e0118107. [PMID: 25763838 PMCID: PMC4357426 DOI: 10.1371/journal.pone.0118107] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/27/2014] [Indexed: 12/24/2022] Open
Abstract
The experimental determination of the structure of protein complexes cannot keep pace with the generation of interactomic data, hence resulting in an ever-expanding gap. As the structural details of protein complexes are central to a full understanding of the function and dynamics of the cell machinery, alternative strategies are needed to circumvent the bottleneck in structure determination. Computational protein docking is a valid and valuable approach to model the structure of protein complexes. In this work, we describe a novel computational strategy to predict the structure of protein complexes based on data-driven docking: VORFFIP-driven dock (V-D2OCK). This new approach makes use of our newly described method to predict functional sites in protein structures, VORFFIP, to define the region to be sampled during docking and structural clustering to reduce the number of models to be examined by users. V-D2OCK has been benchmarked using a validated and diverse set of protein complexes and compared to a state-of-art docking method. The speed and accuracy compared to contemporary tools justifies the potential use of VD2OCK for high-throughput, genome-wide, protein docking. Finally, we have developed a web interface that allows users to browser and visualize V-D2OCK predictions from the convenience of their web-browsers.
Collapse
Affiliation(s)
- Joan Segura
- Leeds Institute of Molecular Medicine, School of Medicine, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Manuel Alejandro Marín-López
- Structural Bioinformatics Lab (GRIB-IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| | - Pamela F. Jones
- Leeds Institute of Molecular Medicine, School of Medicine, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Baldo Oliva
- Structural Bioinformatics Lab (GRIB-IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain
| | - Narcis Fernandez-Fuentes
- Leeds Institute of Molecular Medicine, School of Medicine, University of Leeds, Leeds, LS9 7TF, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Rouet R, Dudgeon K, Christie M, Langley D, Christ D. Fully Human VH Single Domains That Rival the Stability and Cleft Recognition of Camelid Antibodies. J Biol Chem 2015; 290:11905-17. [PMID: 25737448 DOI: 10.1074/jbc.m114.614842] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Indexed: 01/01/2023] Open
Abstract
Human VH single domains represent a promising class of antibody fragments with applications as therapeutic modalities. Unfortunately, isolated human VH domains also generally display poor biophysical properties and a propensity to aggregate. This has encouraged the development of non-human antibody domains as alternative means of antigen recognition and, in particular, camelid (VHH) domains. Naturally devoid of light chain partners, these domains are characterized by favorable biophysical properties and propensity for cleft binding, a highly desirable characteristic, allowing the targeting of cryptic epitopes. In contrast, previously reported structures of human VH single domains had failed to recapitulate this property. Here we report the engineering and characterization of phage display libraries of stable human VH domains and the selection of binders against a diverse set of antigens. Unlike "camelized" human domains, the domains do not rely on potentially immunogenic framework mutations and maintain the structure of the VH/VL interface. Structure determination in complex with hen egg white lysozyme revealed an extended VH binding interface, with complementarity-determining region 3 deeply penetrating into the active site cleft, highly reminiscent of what has been observed for camelid domains. Taken together, our results demonstrate that fully human VH domains can be constructed that are not only stable and well expressed but also rival the cleft binding properties of camelid antibodies.
Collapse
Affiliation(s)
- Romain Rouet
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and
| | - Kip Dudgeon
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and
| | - Mary Christie
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and the Faculty of Medicine, St. Vincent's Clinical School, University of New South Wales, Darlinghurst, Sydney, New South Wales 2010, Australia
| | - David Langley
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and
| | - Daniel Christ
- From the Department of Immunology, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia and the Faculty of Medicine, St. Vincent's Clinical School, University of New South Wales, Darlinghurst, Sydney, New South Wales 2010, Australia
| |
Collapse
|
37
|
Desmyter A, Spinelli S, Roussel A, Cambillau C. Camelid nanobodies: killing two birds with one stone. Curr Opin Struct Biol 2015; 32:1-8. [PMID: 25614146 DOI: 10.1016/j.sbi.2015.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/23/2014] [Accepted: 01/05/2015] [Indexed: 01/09/2023]
Abstract
In recent years, the use of single-domain camelid immunoglobulins, termed vHHs or nanobodies, has seen increasing growth in biotechnology, pharmaceutical applications and structure/function research. The usefulness of nanobodies in structural biology is now firmly established, as they provide access to new epitopes in concave and hinge regions - and stabilize them. These sites are often associated with enzyme inhibition or receptor neutralization, and, at the same time, provide favorable surfaces for crystal packing. Remarkable results have been achieved by using nanobodies with flexible multi-domain proteins, large complexes and, last but not least, membrane proteins. While generating nanobodies is still a rather long and expensive procedure, the advent of naive libraries might be expected to facilitate the whole process.
Collapse
Affiliation(s)
- Aline Desmyter
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Silvia Spinelli
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Alain Roussel
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France
| | - Christian Cambillau
- Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques, France; Centre National de la Recherche Scientifique, AFMB, UMR 7257, case 932, 13288 Marseille Cedex 09, France.
| |
Collapse
|
38
|
Sun Z, Lu W, Liu P, Wang H, Huang Y, Zhao Y, Kong Y, Cui Z. Isolation and characterization of a proteinaceous α-amylase inhibitor AAI-CC5 from Streptomyces sp. CC5, and its gene cloning and expression. Antonie van Leeuwenhoek 2014; 107:345-56. [PMID: 25411086 DOI: 10.1007/s10482-014-0333-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/11/2014] [Indexed: 11/26/2022]
Abstract
An α-amylase inhibitor producing Streptomyces sp. strain CC5 was isolated from soil. A proteinaceous α-amylase inhibitor AAI-CC5 was purified from strain CC5. AAI-CC5 specifically inhibited mammalian α-amylases. The molecular weight of the inhibitor was determined to be 8,212 Da by MALDI-TOF Mass Spectrum. The N-terminal 15 amino acid residues of the purified AAI-CC5 were DTGSPAPECVEYFQS, which is dissimilar to other reported proteinaceous α-amylase inhibitors. AAI-CC5 is a pH insensitive and heat-stable protein, and cannot be hydrolysed by trypsin. AAI-CC5 was cloned and expressed in Escherichia coli BL21 (DE3) with a hexa-histidine tag on the C terminal. AAI-CC5 shared 82 % identity with Parvulustat. The recombinant α-amylase inhibitor was purified to homogeneity by one-step affinity chromatography using Ni(2+)-NTA resin with molecular mass of 9,404 Da. Steady state kinetics studies of α-amylase and the inhibitor revealed an irreversible, non-competitive inhibition mechanism with IC50 and Ki value of 6.43 ×1 10(-11) and 4.45 × 10(-11) M respectively. These results suggest this novel α-amylase inhibitor possessed powerful inhibitory activity for α-amylase, and it may be a candidate in research of diabetes therapy and obesity treatment.
Collapse
Affiliation(s)
- Zhibin Sun
- Key Laboratory of Environmental Microbiology of Ministry of Agriculture, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Single-domain antibodies targeting neuraminidase protect against an H5N1 influenza virus challenge. J Virol 2014; 88:8278-96. [PMID: 24829341 DOI: 10.1128/jvi.03178-13] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Influenza virus neuraminidase (NA) is an interesting target of small-molecule antiviral drugs. We isolated a set of H5N1 NA-specific single-domain antibodies (N1-VHHm) and evaluated their in vitro and in vivo antiviral potential. Two of them inhibited the NA activity and in vitro replication of clade 1 and 2 H5N1 viruses. We then generated bivalent derivatives of N1-VHHm by two methods. First, we made N1-VHHb by genetically joining two N1-VHHm moieties with a flexible linker. Second, bivalent N1-VHH-Fc proteins were obtained by genetic fusion of the N1-VHHm moiety with the crystallizable region of mouse IgG2a (Fc). The in vitro antiviral potency against H5N1 of both bivalent N1-VHHb formats was 30- to 240-fold higher than that of their monovalent counterparts, with 50% inhibitory concentrations in the low nanomolar range. Moreover, single-dose prophylactic treatment with bivalent N1-VHHb or N1-VHH-Fc protected BALB/c mice against a lethal challenge with H5N1 virus, including an oseltamivir-resistant H5N1 variant. Surprisingly, an N1-VHH-Fc fusion without in vitro NA-inhibitory or antiviral activity also protected mice against an H5N1 challenge. Virus escape selection experiments indicated that one amino acid residue close to the catalytic site is required for N1-VHHm binding. We conclude that single-domain antibodies directed against influenza virus NA protect against H5N1 virus infection, and when engineered with a conventional Fc domain, they can do so in the absence of detectable NA-inhibitory activity. IMPORTANCE Highly pathogenic H5N1 viruses are a zoonotic threat. Outbreaks of avian influenza caused by these viruses occur in many parts of the world and are associated with tremendous economic loss, and these viruses can cause very severe disease in humans. In such cases, small-molecule inhibitors of the viral NA are among the few treatment options for patients. However, treatment with such drugs often results in the emergence of resistant viruses. Here we show that single-domain antibody fragments that are specific for NA can bind and inhibit H5N1 viruses in vitro and can protect laboratory mice against a challenge with an H5N1 virus, including an oseltamivir-resistant virus. In addition, plant-produced VHH fused to a conventional Fc domain can protect in vivo even in the absence of NA-inhibitory activity. Thus, NA of influenza virus can be effectively targeted by single-domain antibody fragments, which are amenable to further engineering.
Collapse
|
40
|
Pardon E, Laeremans T, Triest S, Rasmussen SGF, Wohlkönig A, Ruf A, Muyldermans S, Hol WGJ, Kobilka BK, Steyaert J. A general protocol for the generation of Nanobodies for structural biology. Nat Protoc 2014; 9:674-93. [PMID: 24577359 PMCID: PMC4297639 DOI: 10.1038/nprot.2014.039] [Citation(s) in RCA: 564] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
There is growing interest in using antibodies as auxiliary tools to crystallize proteins. Here we describe a general protocol for the generation of Nanobodies to be used as crystallization chaperones for the structural investigation of diverse conformational states of flexible (membrane) proteins and complexes thereof. Our technology has a competitive advantage over other recombinant crystallization chaperones in that we fully exploit the natural humoral response against native antigens. Accordingly, we provide detailed protocols for the immunization with native proteins and for the selection by phage display of in vivo-matured Nanobodies that bind conformational epitopes of functional proteins. Three representative examples illustrate that the outlined procedures are robust, making it possible to solve by Nanobody-assisted X-ray crystallography in a time span of 6-12 months.
Collapse
Affiliation(s)
- Els Pardon
- 1] Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium. [2] Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Brussels, Belgium
| | - Toon Laeremans
- 1] Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium. [2] Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Brussels, Belgium
| | - Sarah Triest
- 1] Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium. [2] Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Brussels, Belgium
| | - Søren G F Rasmussen
- Department of Neuroscience and Pharmacology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Alexandre Wohlkönig
- 1] Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium. [2] Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Brussels, Belgium
| | - Armin Ruf
- Pharma Research and Early Development (pRED), Small Molecule Research, Discovery Technologies, F. Hoffmann-La Roche, Basel, Switzerland
| | - Serge Muyldermans
- 1] Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Brussels, Belgium. [2] Cellular and Molecular Immunology, VUB, Brussels, Belgium
| | - Wim G J Hol
- Department of Biochemistry, Biomolecular Structure Center, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, School of Medicine, Stanford University, Stanford, California, USA
| | - Jan Steyaert
- 1] Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium. [2] Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Brussels, Belgium
| |
Collapse
|
41
|
Kovalenko OV, Olland A, Piché-Nicholas N, Godbole A, King D, Svenson K, Calabro V, Müller MR, Barelle CJ, Somers W, Gill DS, Mosyak L, Tchistiakova L. Atypical antigen recognition mode of a shark immunoglobulin new antigen receptor (IgNAR) variable domain characterized by humanization and structural analysis. J Biol Chem 2013; 288:17408-19. [PMID: 23632026 DOI: 10.1074/jbc.m112.435289] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The immunoglobulin new antigen receptors (IgNARs) are a class of Ig-like molecules of the shark immune system that exist as heavy chain-only homodimers and bind antigens by their single domain variable regions (V-NARs). Following shark immunization and/or in vitro selection, V-NARs can be generated as soluble, stable, and specific high affinity monomeric binding proteins of ∼12 kDa. We have previously isolated a V-NAR from an immunized spiny dogfish shark, named E06, that binds specifically and with high affinity to human, mouse, and rat serum albumins. Humanization of E06 was carried out by converting over 60% of non-complementarity-determining region residues to those of a human germ line Vκ1 sequence, DPK9. The resulting huE06 molecules have largely retained the specificity and affinity of antigen binding of the parental V-NAR. Crystal structures of the shark E06 and its humanized variant (huE06 v1.1) in complex with human serum albumin (HSA) were determined at 3- and 2.3-Å resolution, respectively. The huE06 v1.1 molecule retained all but one amino acid residues involved in the binding site for HSA. Structural analysis of these V-NARs has revealed an unusual variable domain-antigen interaction. E06 interacts with HSA in an atypical mode that utilizes extensive framework contacts in addition to complementarity-determining regions that has not been seen previously in V-NARs. On the basis of the structure, the roles of various elements of the molecule are described with respect to antigen binding and V-NAR stability. This information broadens the general understanding of antigen recognition and provides a framework for further design and humanization of shark IgNARs.
Collapse
Affiliation(s)
- Oleg V Kovalenko
- Global Biotherapeutics Technologies, Pfizer Research and Development, Cambridge, Massachusetts 02140, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
MβL (metallo-β-lactamase) enzymes are usually produced by multi-resistant Gram-negative bacterial strains and have spread worldwide. An approach on the basis of phage display was used to select single-domain antibody fragments (VHHs, also called nanobodies) that would inhibit the clinically relevant VIM (Verona integron-encoded MβL)-4 MβL. Out of more than 50 selected nanobodies, only the NbVIM_38 nanobody inhibited VIM-4. The paratope, inhibition mechanism and epitope of the NbVIM_38 nanobody were then characterized. An alanine scan of the NbVIM_38 paratope showed that its binding was driven by hydrophobic amino acids. The inhibitory concentration was in the micromolar range for all β-lactams tested. In addition, the inhibition was found to follow a mixed hyperbolic profile with a predominantly uncompetitive component. Moreover, substrate inhibition was recorded only after nanobody binding. These kinetic data are indicative of a binding site that is distant from the active site. This finding was confirmed by epitope mapping analysis that was performed using peptides, and which identified two stretches of amino acids in the L6 loop and at the end of the α2 helix. Because this binding site is distant from the active site and alters both the substrate binding and catalytic properties of VIM-4, this nanobody can be considered as an allosteric inhibitor.
Collapse
|
43
|
Viral infection modulation and neutralization by camelid nanobodies. Proc Natl Acad Sci U S A 2013; 110:E1371-9. [PMID: 23530214 DOI: 10.1073/pnas.1301336110] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Lactococcal phages belong to a large family of Siphoviridae and infect Lactococcus lactis, a gram-positive bacterium used in commercial dairy fermentations. These phages are believed to recognize and bind specifically to pellicle polysaccharides covering the entire bacterium. The phage TP901-1 baseplate, located at the tip of the tail, harbors 18 trimeric receptor binding proteins (RBPs) promoting adhesion to a specific lactococcal strain. Phage TP901-1 adhesion does not require major conformational changes or Ca(2+), which contrasts other lactococcal phages. Here, we produced and characterized llama nanobodies raised against the purified baseplate and the Tal protein of phage TP901-1 as tools to dissect the molecular determinants of phage TP901-1 infection. Using a set of complementary techniques, surface plasmon resonance, EM, and X-ray crystallography in a hybrid approach, we identified binders to the three components of the baseplate, analyzed their affinity for their targets, and determined their epitopes as well as their functional impact on TP901-1 phage infectivity. We determined the X-ray structures of three nanobodies in complex with the RBP. Two of them bind to the saccharide binding site of the RBP and are able to fully neutralize TP901-1 phage infectivity, even after 15 passages. These results provide clear evidence for a practical use of nanobodies in circumventing lactococcal phages viral infection in dairy fermentation.
Collapse
|
44
|
Chowdhury R, Rasheed M, Keidel D, Moussalem M, Olson A, Sanner M, Bajaj C. Protein-protein docking with F(2)Dock 2.0 and GB-rerank. PLoS One 2013; 8:e51307. [PMID: 23483883 PMCID: PMC3590208 DOI: 10.1371/journal.pone.0051307] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 10/31/2012] [Indexed: 12/03/2022] Open
Abstract
Motivation Computational simulation of protein-protein docking can expedite the process of molecular modeling and drug discovery. This paper reports on our new F2 Dock protocol which improves the state of the art in initial stage rigid body exhaustive docking search, scoring and ranking by introducing improvements in the shape-complementarity and electrostatics affinity functions, a new knowledge-based interface propensity term with FFT formulation, a set of novel knowledge-based filters and finally a solvation energy (GBSA) based reranking technique. Our algorithms are based on highly efficient data structures including the dynamic packing grids and octrees which significantly speed up the computations and also provide guaranteed bounds on approximation error. Results The improved affinity functions show superior performance compared to their traditional counterparts in finding correct docking poses at higher ranks. We found that the new filters and the GBSA based reranking individually and in combination significantly improve the accuracy of docking predictions with only minor increase in computation time. We compared F2 Dock 2.0 with ZDock 3.0.2 and found improvements over it, specifically among 176 complexes in ZLab Benchmark 4.0, F2 Dock 2.0 finds a near-native solution as the top prediction for 22 complexes; where ZDock 3.0.2 does so for 13 complexes. F2 Dock 2.0 finds a near-native solution within the top 1000 predictions for 106 complexes as opposed to 104 complexes for ZDock 3.0.2. However, there are 17 and 15 complexes where F2 Dock 2.0 finds a solution but ZDock 3.0.2 does not and vice versa; which indicates that the two docking protocols can also complement each other. Availability The docking protocol has been implemented as a server with a graphical client (TexMol) which allows the user to manage multiple docking jobs, and visualize the docked poses and interfaces. Both the server and client are available for download. Server: http://www.cs.utexas.edu/~bajaj/cvc/software/f2dock.shtml. Client: http://www.cs.utexas.edu/~bajaj/cvc/software/f2dockclient.shtml.
Collapse
Affiliation(s)
- Rezaul Chowdhury
- Department of Computer Science, Institute of Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Muhibur Rasheed
- Department of Computer Science, Institute of Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Donald Keidel
- The Scripps Research Institute, La Jolla, California, United States of America
| | - Maysam Moussalem
- Department of Computer Science, Institute of Computational Engineering and Sciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Arthur Olson
- The Scripps Research Institute, La Jolla, California, United States of America
| | - Michel Sanner
- The Scripps Research Institute, La Jolla, California, United States of America
| | - Chandrajit Bajaj
- The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
45
|
Bodelón G, Palomino C, Fernández LÁ. Immunoglobulin domains inEscherichia coliand other enterobacteria: from pathogenesis to applications in antibody technologies. FEMS Microbiol Rev 2013; 37:204-50. [DOI: 10.1111/j.1574-6976.2012.00347.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 06/07/2012] [Accepted: 06/14/2012] [Indexed: 11/28/2022] Open
|
46
|
Analysis of heavy-chain antibody responses and resistance to Parelaphostrongylus tenuis in experimentally infected alpacas. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1019-26. [PMID: 22593238 DOI: 10.1128/cvi.00178-12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The parasitic nematode Parelaphostrongylus tenuis is an important cause of neurologic disease of camelids in central and eastern North America. The aim of this study was to determine whether alpacas develop resistance to disease caused by P. tenuis in response to a previous infection or a combination of controlled infection and immunization. Alpacas were immunized with a homogenate of third-stage larvae (L3) and simultaneously implanted subcutaneously with diffusion chambers containing 20 live L3. Sham-treated animals received adjuvant alone and empty chambers. The protocol was not effective in inducing resistance to oral challenge with 10 L3, and disease developed between 60 and 71 days following infection. Immediately following the onset of neurologic disease, affected animals were treated with a regimen of anthelmintic and anti-inflammatory drugs, and all recovered. One year later, a subset of alpacas from this experiment was challenged with 20 L3 and the results showed that prior infection induced resistance to disease. Primary and secondary infections induced production of conventional and heavy-chain IgGs that reacted with soluble antigens in L3 homogenates but did not consistently recognize a recombinant form of a parasite-derived aspartyl protease inhibitor. Thus, the latter antigen may not be a good candidate for serology-based diagnostic tests. Antibody responses to parasite antigens occurred in the absence of overt disease, demonstrating that P. tenuis infection can be subclinical in a host that has been considered to be highly susceptible to disease. The potential for immunoprophylaxis to be effective in preventing disease caused by P. tenuis was supported by evidence of resistance to reinfection.
Collapse
|
47
|
Li JW, Xia L, Su Y, Liu H, Xia X, Lu Q, Yang C, Reheman K. Molecular imprint of enzyme active site by camel nanobodies: rapid and efficient approach to produce abzymes with alliinase activity. J Biol Chem 2012; 287:13713-21. [PMID: 22374998 DOI: 10.1074/jbc.m111.336370] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Screening of inhibitory Ab1 antibodies is a critical step for producing catalytic antibodies in the anti-idiotypic approach. However, the incompatible surface of the active site of the enzyme and the antigen-binding site of heterotetrameric conventional antibodies become the limiting step. Because camelid-derived nanobodies possess the potential to preferentially bind to the active site of enzymes due to their small size and long CDR3, we have developed a novel approach to produce antibodies with alliinase activities by exploiting the molecular mimicry of camel nanobodies. By screening the camelid-derived variable region of the heavy chain cDNA phage display library with alliinase, we obtained an inhibitory nanobody VHHA4 that recognizes the active site. Further screening with VHHA4 from the same variable domain of the heavy chain of a heavy-chain antibody library led to a higher incidence of anti-idiotypic Ab2 abzymes with alliinase activities. One of the abzymes, VHHC10, showed the highest activity that can be inhibited by Ab1 VHHA4 and alliinase competitive inhibitor penicillamine and significantly suppressed the B16 tumor cell growth in the presence of alliin in vitro. The results highlight the feasibility of producing abzymes via anti-idiotypic nanobody approach.
Collapse
Affiliation(s)
- Jiang-Wei Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Antibodies are invaluable macromolecules effectively utilized as detection reagents and therapeutics. Traditionally, researchers have relied upon the entire immunoglobulin molecule, however advances in protein engineering have ushered the use of antibody fragments as equally important biological tools such that at present, the downstream application generally dictates the antibody format employed. We provide herein robust and proven protocols for the isolation of autonomous human antibody variable heavy domains (VH). The strategy utilizes combinatorial phage-displayed libraries targeting human VH domain positions previously shown to promote autonomous behavior, and selection against a specified antigen. Subsequently, autonomous VH domains are characterized and chosen using standard biophysical methods.
Collapse
Affiliation(s)
- Raffi Tonikian
- Terrence Donnelly Center for Cellular and Biomolecular Research and Banting and Best Department of Medical Research, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
49
|
Monegal A, Olichon A, Bery N, Filleron T, Favre G, de Marco A. Single domain antibodies with VH hallmarks are positively selected during panning of llama (Lama glama) naïve libraries. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2012; 36:150-156. [PMID: 21767565 DOI: 10.1016/j.dci.2011.06.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 06/24/2011] [Accepted: 06/26/2011] [Indexed: 05/31/2023]
Abstract
Independent variable domains with VH hallmarks have been repeatedly identified in immune and pre-immune VHH libraries. In some cases, stable independent VH domains have been also isolated in mouse and human recombinant antibody repertoires. However, we have come to realize that VHs were selected with a higher efficiency than VHHs during biopanning of a pre-immune (VHH) library. The biochemical and biophysical comparison did not indicate a presence of any feature that would favor the VH binders during the selection process. In contrast, selected VHHs seemed to be more stable than the VHs, ruling out the existence of a thermodynamically - favored VH sub-class. Therefore, we reasoned that a certain degree of thermodynamic instability may be beneficial for both displaying and expression of VH(H)s when the Sec-pathway is used for their secretion to avoid the cytoplasmic trapping of fast-folding polypeptides. Indeed, VHHs, but not VHs, were accumulated at higher concentrations when expressed fused to the dsbA leader peptide, a sequence that drives the linked polypeptides to the co-translational SRP secretion machinery. These data suggest that the thermodynamically favored VHHs can be lost during biopanning, as previously observed for DARPins and in contrast to the recombinant antibodies in scFv format.
Collapse
Affiliation(s)
- Ana Monegal
- Cogentech - Protein Chemistry Unit, IFOM-IEO Campus, Via Adamello 16, 20139 Milano, Italy
| | | | | | | | | | | |
Collapse
|
50
|
de Marco A. User-friendly expression plasmids enable the fusion of VHHs to application-specific tags. Methods Mol Biol 2012; 911:507-522. [PMID: 22886273 DOI: 10.1007/978-1-61779-968-6_32] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
One of the advantages of using recombinant instead of conventional antibodies is that these can be easily manipulated by means of standard molecular biology techniques. Therefore this opportunity can be exploited to prepare fusion constructs composed of VHHs and suitable tags. According to the applications in which the antibodies will be applied, molecules such as fluorescent probes, biotin, and PEG can be either covalently or non-covalently linked to the antibodies. Within this chapter, practical tips for the choice and expression of the most appropriate among the available plasmids are listed, keeping in mind the experimental conditions in which usually the fusion antibodies will be applied.
Collapse
Affiliation(s)
- Ario de Marco
- University of Nova Gorica (UNG), Rožna Dolina, Nova Gorica, Slovenia.
| |
Collapse
|