1
|
Li H, Ai Y, Wu Y, Fan R, Tian Y, Chen S, Wan W, Wu C. Development of a Graphene Oxide-Based Aptamer Nanoarray for Improved Neutralization and Protection Effects Against Ricin. Pharmaceutics 2024; 16:1455. [PMID: 39598578 PMCID: PMC11597295 DOI: 10.3390/pharmaceutics16111455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Ricin's high toxicity and potential as a bioweapon underscore the need for effective antidotes. Monoclonal antibodies, though effective, are limited by complex production. This study aimed to develop a graphene oxide-based aptamer nanoarray (ARMAN) for improved neutralization and protection against ricin. Methods: High-affinity aptamers targeting ricin's RTA and RTB subunits were selected using SELEX technology and conjugated to graphene oxide (GO) via click chemistry. ARMAN's characteristics, including morphology, stability, and biosecurity, were assessed. Its performance was evaluated in terms of affinity for ricin, neutralization capacity, and therapeutic effects in cellular assays and a mouse model of ricin poisoning. Results: ARMAN exhibited a uniform morphology with an average particle size of 217 nm and demonstrated significantly enhanced affinity for ricin compared to free aptamers. ARMAN showed rapid and effective neutralization ability, significantly increasing cell viability in BEAS-2B, GES-1, and HL7702 cell lines exposed to ricin. In vivo, ARMAN treatment led to a notable prolongation of survival in ricin-poisoned mice, highlighting its potential for both pre- and post-exposure treatment. These findings indicate that ARMAN not only neutralizes ricin effectively but also provides a therapeutic window for treatment. Conclusions: ARMAN's superior binding affinity, serum stability, biocompatibility, and broad therapeutic efficacy make it a promising new antidote against ricin poisoning. This study's findings represent significant progress in the development of rapid-response antidotes, with ARMAN offering a potential solution for both military and civilian emergency response scenarios.
Collapse
Affiliation(s)
- Huafei Li
- School of Lifesciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Yanwen Ai
- School of Lifesciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Yanjin Wu
- School of Lifesciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Ruyu Fan
- Clinical Research Unit, The First Affiliated Hospital of Navy Medical University (Changhai Hospital), 168 Changhai Road, Shanghai 200433, China
| | - Yuan Tian
- School of Lifesciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Shuangqun Chen
- School of Lifesciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Wei Wan
- Department of Orthopedic Oncology, Spinal Tumor Center, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Cong Wu
- Clinical Research Unit, The First Affiliated Hospital of Navy Medical University (Changhai Hospital), 168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
2
|
Czajka TF, Vance DJ, Song R, Mantis NJ. A Biparatopic Intrabody Renders Vero Cells Impervious to Ricin Intoxication. Biochemistry 2024; 63:2391-2396. [PMID: 39297955 DOI: 10.1021/acs.biochem.4c00385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Expression of camelid-derived, single-domain antibodies (VHHs) within the cytoplasm of mammalian cells as "intrabodies" has opened up novel avenues for medical countermeasures against fast-acting biothreat agents. In this report, we describe a heterodimeric intrabody that renders Vero cells virtually impervious to ricin toxin (RT), a potent Category B ribosome-inactivating protein. The intrabody consists of two structurally defined VHHs that target distinct epitopes on RT's enzymatic subunit (RTA): V9E1 targets RTA's P-stalk recruitment site, and V2A11 targets RTA's active site. Resistance to RT conferred by the biparatopic VHH construct far exceeded that of either of the VHHs alone and effectively inhibited all measurable RT-induced cytotoxicity in vitro. We propose that the targeted delivery of bispecific intrabodies to lung tissues may represent a novel means to shield the airways from the effects of inhalational RT exposure.
Collapse
Affiliation(s)
- Timothy F Czajka
- Department of Biomedical Sciences, University at Albany, Albany, New York 12201, United States
| | - David J Vance
- Department of Biomedical Sciences, University at Albany, Albany, New York 12201, United States
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States of America
| | - Renji Song
- Division of Research, Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States of America
| | - Nicholas J Mantis
- Department of Biomedical Sciences, University at Albany, Albany, New York 12201, United States
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States of America
| |
Collapse
|
3
|
Lequesne L, Dano J, Rouaix A, Kropp C, Plaisance M, Gelhaye S, Lequesne ML, Piquet P, Avril A, Becher F, Orsini Delgado ML, Simon S. A Monoclonal Antibody with a High Affinity for Ricin Isoforms D and E Provides Strong Protection against Ricin Poisoning. Toxins (Basel) 2024; 16:412. [PMID: 39453188 PMCID: PMC11510859 DOI: 10.3390/toxins16100412] [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: 09/06/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/26/2024] Open
Abstract
Ricin is a highly potent toxin that has been used in various attempts at bioterrorism worldwide. Although a vaccine for preventing ricin poisoning (RiVax™) is in clinical development, there are currently no commercially available prophylaxis or treatments for ricin intoxication. Numerous studies have highlighted the potential of passive immunotherapy using anti-ricin monoclonal antibodies (mAbs) and have shown promising results in preclinical models. In this article, we describe the neutralizing and protective efficacy of a new generation of high-affinity anti-ricin mAbs, which bind and neutralize very efficiently both ricin isoforms D and E in vitro through cytotoxicity cell assays. In vivo, protection assay revealed that one of these mAbs (RicE5) conferred over 90% survival in a murine model challenged intranasally with a 5 LD50 of ricin and treated by intravenous administration of the mAbs 6 h post-intoxication. Notably, a 35% survival rate was observed even when treatment was administered 24 h post-exposure. Moreover, all surviving mice exhibited long-term immunity to high ricin doses. These findings offer promising results for the clinical development of a therapeutic candidate against ricin intoxication and may also pave the way for novel vaccination strategies against ricin or other toxins.
Collapse
Affiliation(s)
- Loïs Lequesne
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Julie Dano
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Audrey Rouaix
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Camille Kropp
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Marc Plaisance
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Stéphanie Gelhaye
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Marie-Lou Lequesne
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Paloma Piquet
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Arnaud Avril
- Microbiology and Infectious Diseases Department, French Armed Forces Biomedical Research Institute, 91220 Brétigny-sur-Orge, France
| | - François Becher
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Maria Lucia Orsini Delgado
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| | - Stéphanie Simon
- Département Médicaments et Technologies pour la Santé (DMTS), SPI, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France; (L.L.); (M.L.O.D.)
| |
Collapse
|
4
|
Czajka TF, Vance DJ, Song R, Mantis NJ. A Biparatopic Intrabody Renders Vero Cells Impervious to Ricin Intoxication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601761. [PMID: 39005371 PMCID: PMC11244990 DOI: 10.1101/2024.07.02.601761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Expression of camelid-derived, single-domain antibodies (VHHs) within the cytoplasm of mammalian cells as "intrabodies" has opened-up novel avenues for medical countermeasures against fast-acting biothreat agents. In this report, we describe a heterodimeric intrabody that renders Vero cells virtually impervious to ricin toxin (RT), a potent Category B ribosome-inactivating protein (RIP). The intrabody consists of two structurally defined VHHs that target distinct epitopes on RT's enzymatic subunit (RTA): V9E1 targets RTA's P-stalk recruitment site, and V2A11 targets RTA's active site. Resistance to RT conferred by the biparatopic VHH construct far exceeded that of either of the VHHs alone and effectively inhibited all measurable RT-induced cytotoxicty in vitro. We propose that targeted delivery of bispecific intrabodies to lung tissues may represent a novel means to shield the airways from the effects of inhalational RT exposure.
Collapse
Affiliation(s)
- Timothy F. Czajka
- Department of Biomedical Sciences, University at Albany, Albany, NY 12201 United States
| | - David J. Vance
- Department of Biomedical Sciences, University at Albany, Albany, NY 12201 United States
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, 12208
| | - Renji Song
- Division of Research, Wadsworth Center, New York State Department of Health, Albany, NY, 12208
| | - Nicholas J. Mantis
- Department of Biomedical Sciences, University at Albany, Albany, NY 12201 United States
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, 12208
| |
Collapse
|
5
|
De Greve H, Fioravanti A. Single domain antibodies from camelids in the treatment of microbial infections. Front Immunol 2024; 15:1334829. [PMID: 38827746 PMCID: PMC11140111 DOI: 10.3389/fimmu.2024.1334829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/29/2024] [Indexed: 06/04/2024] Open
Abstract
Infectious diseases continue to pose significant global health challenges. In addition to the enduring burdens of ailments like malaria and HIV, the emergence of nosocomial outbreaks driven by antibiotic-resistant pathogens underscores the ongoing threats. Furthermore, recent infectious disease crises, exemplified by the Ebola and SARS-CoV-2 outbreaks, have intensified the pursuit of more effective and efficient diagnostic and therapeutic solutions. Among the promising options, antibodies have garnered significant attention due to their favorable structural characteristics and versatile applications. Notably, nanobodies (Nbs), the smallest functional single-domain antibodies of heavy-chain only antibodies produced by camelids, exhibit remarkable capabilities in stable antigen binding. They offer unique advantages such as ease of expression and modification and enhanced stability, as well as improved hydrophilicity compared to conventional antibody fragments (antigen-binding fragments (Fab) or single-chain variable fragments (scFv)) that can aggregate due to their low solubility. Nanobodies directly target antigen epitopes or can be engineered into multivalent Nbs and Nb-fusion proteins, expanding their therapeutic potential. This review is dedicated to charting the progress in Nb research, particularly those derived from camelids, and highlighting their diverse applications in treating infectious diseases, spanning both human and animal contexts.
Collapse
Affiliation(s)
- Henri De Greve
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Antonella Fioravanti
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
- VIB-VUB Center for Structural Biology, Vrije Universiteit Brussel, Brussels, Belgium
- Fondazione ParSeC – Parco delle Scienze e della Cultura, Prato, Italy
| |
Collapse
|
6
|
Liu ML, Liang XM, Jin MY, Huang HW, Luo L, Wang H, Shen X, Xu ZL. Food-Borne Biotoxin Neutralization in Vivo by Nanobodies: Current Status and Prospects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10753-10771. [PMID: 38706131 DOI: 10.1021/acs.jafc.4c02257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Food-borne biotoxins from microbes, plants, or animals contaminate unclean, spoiled, and rotten foods, posing significant health risks. Neutralizing such toxins is vital for human health, especially after food poisoning. Nanobodies (Nbs), a type of single-domain antibodies derived from the genetic cloning of a variable domain of heavy chain antibodies (VHHs) in camels, offer unique advantages in toxin neutralization. Their small size, high stability, and precise binding enable effective neutralization. The use of Nbs in neutralizing food-borne biotoxins offers numerous benefits, and their genetic malleability allows tailored optimization for diverse toxins. As nanotechnology continues to evolve and improve, Nbs are poised to become increasingly efficient and safer tools for toxin neutralization, playing a pivotal role in safeguarding human health and environmental safety. This review not only highlights the efficacy of these agents in neutralizing toxins but also proposes innovative solutions to address their current challenges. It lays a solid foundation for their further development in this crucial field and propels their commercial application, thereby contributing significantly to advancements in this domain.
Collapse
Affiliation(s)
- Min-Ling Liu
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Research Center for Green Development of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Min Liang
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Research Center for Green Development of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Ming-Yu Jin
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Research Center for Green Development of Agriculture, South China Agricultural University, Guangzhou 510642, China
- School of Life and Health Technology, Dongguan, University of Technology, Dongguan 523808, China
| | - Hui-Wei Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Research Center for Green Development of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Lin Luo
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Research Center for Green Development of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Hong Wang
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Research Center for Green Development of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Xing Shen
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Research Center for Green Development of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Zhen-Lin Xu
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Research Center for Green Development of Agriculture, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
7
|
Vance DJ, Basir S, Piazza CL, Willsey GG, Haque HME, Tremblay JM, Rudolph MJ, Muriuki B, Cavacini L, Weis DD, Shoemaker CB, Mantis NJ. Single-domain antibodies reveal unique borrelicidal epitopes on the Lyme disease vaccine antigen, outer surface protein A (OspA). Infect Immun 2024; 92:e0008424. [PMID: 38470113 PMCID: PMC11003225 DOI: 10.1128/iai.00084-24] [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: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/13/2024] Open
Abstract
Camelid-derived, single-domain antibodies (VHHs) have proven to be extremely powerful tools in defining the antigenic landscape of immunologically heterogeneous surface proteins. In this report, we generated a phage-displayed VHH library directed against the candidate Lyme disease vaccine antigen, outer surface protein A (OspA). Two alpacas were immunized with recombinant OspA serotype 1 from Borrelia burgdorferi sensu stricto strain B31, in combination with the canine vaccine RECOMBITEK Lyme containing lipidated OspA. The phage library was subjected to two rounds of affinity enrichment ("panning") against recombinant OspA, yielding 21 unique VHHs within two epitope bins, as determined through competition enzyme linked immunosorbent assays (ELISAs) with a panel of OspA-specific human monoclonal antibodies. Epitope refinement was conducted by hydrogen exchange-mass spectrometry. Six of the monovalent VHHs were expressed as human IgG1-Fc fusion proteins and shown to have functional properties associated with protective human monoclonal antibodies, including B. burgdorferi agglutination, outer membrane damage, and complement-dependent borreliacidal activity. The VHHs displayed unique reactivity profiles with the seven OspA serotypes associated with B. burgdorferi genospecies in the United States and Europe consistent with there being unique epitopes across OspA serotypes that should be considered when designing and evaluating multivalent Lyme disease vaccines.
Collapse
Affiliation(s)
- David J. Vance
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, USA
- Department of Biomedical Sciences, University at Albany, Albany, New York, USA
| | - Saiful Basir
- Department of Biomedical Sciences, University at Albany, Albany, New York, USA
| | - Carol Lyn Piazza
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, USA
| | - Graham G. Willsey
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, USA
| | | | - Jacque M. Tremblay
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | | | - Beatrice Muriuki
- Department of Medicine, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, USA
| | - Lisa Cavacini
- Department of Medicine, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, USA
| | - David D. Weis
- Department of Chemistry, The University of Kansas, Lawrence, Kansas, USA
| | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Nicholas J. Mantis
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, USA
- Department of Biomedical Sciences, University at Albany, Albany, New York, USA
| |
Collapse
|
8
|
Botelho FD, Franca TCC, LaPlante SR. The Search for Antidotes Against Ricin. Mini Rev Med Chem 2024; 24:1148-1161. [PMID: 38350844 DOI: 10.2174/0113895575270509231121060105] [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: 07/04/2023] [Revised: 09/30/2023] [Accepted: 10/18/2023] [Indexed: 02/15/2024]
Abstract
The castor plant (Ricinus communis) is primarily known for its seeds, which contain a unique fatty acid called ricinoleic acid with several industrial and commercial applications. Castor seeds also contain ricin, a toxin considered a chemical and biological warfare agent. Despite years of investigation, there is still no effective antidote or vaccine available. However, some progress has been made, and the development of an effective treatment may be on the horizon. To provide an updated overview of this issue, we have conducted a comprehensive review of the literature on the current state of research in the fight against ricin. This review is based on the reported research and aims to address the challenges faced by researchers, as well as highlight the most successful cases achieved thus far. Our goal is to encourage the scientific community to continue their efforts in this critical search.
Collapse
Affiliation(s)
- Fernanda Diniz Botelho
- Laboratory of Molecular Modeling Applied to the Chemical and Biological Defense (LMCBD), Military Institute of Engineering, Praça General Tibúrcio 80, 22290-270, Rio de Janeiro, RJ, Brazil
| | - Tanos Celmar Costa Franca
- Laboratory of Molecular Modeling Applied to the Chemical and Biological Defense (LMCBD), Military Institute of Engineering, Praça General Tibúrcio 80, 22290-270, Rio de Janeiro, RJ, Brazil
- Université de Québec, INRS - Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec, H7V 1B7, Canada
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Steven R LaPlante
- Université de Québec, INRS - Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec, H7V 1B7, Canada
| |
Collapse
|
9
|
Harmsen MM, Cornelissen JC, van der Wal FJ, Bergervoet JHW, Koene M. Single-Domain Antibody Multimers for Detection of Botulinum Neurotoxin Serotypes C, D, and Their Mosaics in Endopep-MS. Toxins (Basel) 2023; 15:573. [PMID: 37755999 PMCID: PMC10535107 DOI: 10.3390/toxins15090573] [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: 08/21/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are highly toxic proteins that require high-affinity immunocapture reagents for use in endopeptidase-based assays. Here, 30 novel and 2 earlier published llama single-domain antibodies (VHHs) against the veterinary-relevant BoNT serotypes C and D were yeast-produced. These VHHs recognized 10 independent antigenic sites, and many cross-reacted with the BoNT/DC and CD mosaic variants. As VHHs are highly suitable for genetically linking to increase antigen-binding affinity, 52 VHH multimers were produced and their affinity for BoNT/C, D, DC, and CD was determined. A selection of 15 multimers with high affinity (KD < 0.1 nM) was further shown to be resilient to a high salt wash that is used for samples from complex matrices and bound native BoNTs from culture supernatants as shown by Endopep-MS. High-affinity multimers suitable for further development of a highly sensitive Endopep-MS assay include four multimers that bind both BoNT/D and CD with KD of 14-99 pM, one multimer for BoNT/DC (65 pM) that also binds BoNT/C (75 pM), and seven multimers for BoNT/C (<1-19 pM), six of which also bind BoNT/DC with lower affinity (93-508 pM). In addition to application in diagnostic tests, these VHHs could be used for the development of novel therapeutics for animals or humans.
Collapse
Affiliation(s)
- Michiel M. Harmsen
- Wageningen Bioveterinary Research, Wageningen University & Research, 8221 RA Lelystad, The Netherlands (F.J.v.d.W.)
| | - Jan C. Cornelissen
- Wageningen Bioveterinary Research, Wageningen University & Research, 8221 RA Lelystad, The Netherlands (F.J.v.d.W.)
| | - Fimme J. van der Wal
- Wageningen Bioveterinary Research, Wageningen University & Research, 8221 RA Lelystad, The Netherlands (F.J.v.d.W.)
| | - Jan H. W. Bergervoet
- Wageningen Plant Research, Wageningen University & Research, 6708 PB Wageningen, The Netherlands
| | - Miriam Koene
- Wageningen Bioveterinary Research, Wageningen University & Research, 8221 RA Lelystad, The Netherlands (F.J.v.d.W.)
| |
Collapse
|
10
|
Thran M, Pönisch M, Danz H, Horscroft N, Ichtchenko K, Tzipori S, Shoemaker CB. Co-administration of an effector antibody enhances the half-life and therapeutic potential of RNA-encoded nanobodies. Sci Rep 2023; 13:14632. [PMID: 37670025 PMCID: PMC10480410 DOI: 10.1038/s41598-023-41092-7] [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: 05/30/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
The incidence of Clostridioides difficile infection (CDI) and associated mortality have increased rapidly worldwide in recent years. Therefore, it is critical to develop new therapies for CDI. Here we report on the development of mRNA-LNPs encoding camelid-derived VHH-based neutralizing agents (VNAs) targeting toxins A and/or B of C. difficile. In preclinical models, intravenous administration of the mRNA-LNPs provided serum VNA levels sufficient to confer protection of mice against severe disease progression following toxin challenge. Furthermore, we employed an mRNA-LNP encoded effector antibody, a molecular tool designed to specifically bind an epitopic tag linked to the VNAs, to prolong VNA serum half-life. Co-administration of VNA-encoding mRNA-LNPs and an effector antibody, either provided as recombinant protein or encoded by mRNA-LNP, increased serum VNA half-life in mice and in gnotobiotic piglets. Prolonged serum half-life was associated with higher concentrations of serum VNA and enhanced prophylactic protection of mice in challenge models.
Collapse
Affiliation(s)
| | | | - Hillary Danz
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA
| | | | - Konstantin Ichtchenko
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA
| | - Charles B Shoemaker
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, MA, 01536, USA.
| |
Collapse
|
11
|
Zupancic JM, Smith MD, Trzeciakiewicz H, Skinner ME, Ferris SP, Makowski EK, Lucas MJ, McArthur N, Kane RS, Paulson HL, Tessier PM. Quantitative flow cytometric selection of tau conformational nanobodies specific for pathological aggregates. Front Immunol 2023; 14:1164080. [PMID: 37622125 PMCID: PMC10445546 DOI: 10.3389/fimmu.2023.1164080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/15/2023] [Indexed: 08/26/2023] Open
Abstract
Single-domain antibodies, also known as nanobodies, are broadly important for studying the structure and conformational states of several classes of proteins, including membrane proteins, enzymes, and amyloidogenic proteins. Conformational nanobodies specific for aggregated conformations of amyloidogenic proteins are particularly needed to better target and study aggregates associated with a growing class of associated diseases, especially neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. However, there are few reported nanobodies with both conformational and sequence specificity for amyloid aggregates, especially for large and complex proteins such as the tau protein associated with Alzheimer's disease, due to difficulties in selecting nanobodies that bind to complex aggregated proteins. Here, we report the selection of conformational nanobodies that selectively recognize aggregated (fibrillar) tau relative to soluble (monomeric) tau. Notably, we demonstrate that these nanobodies can be directly isolated from immune libraries using quantitative flow cytometric sorting of yeast-displayed libraries against tau aggregates conjugated to quantum dots, and this process eliminates the need for secondary nanobody screening. The isolated nanobodies demonstrate conformational specificity for tau aggregates in brain samples from both a transgenic mouse model and human tauopathies. We expect that our facile approach will be broadly useful for isolating conformational nanobodies against diverse amyloid aggregates and other complex antigens.
Collapse
Affiliation(s)
- Jennifer M. Zupancic
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Matthew D. Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Hanna Trzeciakiewicz
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Mary E. Skinner
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Sean P. Ferris
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Emily K. Makowski
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Michael J. Lucas
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Nikki McArthur
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Ravi S. Kane
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Henry L. Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, MI, United States
- Michigan Alzheimer’s Disease Center, University of Michigan, Ann Arbor, MI, United States
| | - Peter M. Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, MI, United States
- Michigan Alzheimer’s Disease Center, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
12
|
Hussack G, Rossotti MA, van Faassen H, Murase T, Eugenio L, Schrag JD, Ng KKS, Tanha J. Structure-guided design of a potent Clostridiodes difficile toxin A inhibitor. Front Microbiol 2023; 14:1110541. [PMID: 36778856 PMCID: PMC9909335 DOI: 10.3389/fmicb.2023.1110541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Crystal structures of camelid heavy-chain antibody variable domains (VHHs) bound to fragments of the combined repetitive oligopeptides domain of Clostridiodes difficile toxin A (TcdA) reveal that the C-terminus of VHH A20 was located 30 Å away from the N-terminus of VHH A26. Based on this observation, we generated a biparatopic fusion protein with A20 at the N-terminus, followed by a (GS)6 linker and A26 at the C-terminus. This A20-A26 fusion protein shows an improvement in binding affinity and a dramatic increase in TcdA neutralization potency (>330-fold [IC 50]; ≥2,700-fold [IC 99]) when compared to the unfused A20 and A26 VHHs. A20-A26 also shows much higher binding affinity and neutralization potency when compared to a series of control antibody constructs that include fusions of two A20 VHHs, fusions of two A26 VHHs, a biparatopic fusion with A26 at the N-terminus and A20 at the C-terminus (A26-A20), and actoxumab. In particular, A20-A26 displays a 310-fold (IC 50) to 29,000-fold (IC 99) higher neutralization potency than A26-A20. Size-exclusion chromatography-multiangle light scattering (SEC-MALS) analyses further reveal that A20-A26 binds to TcdA with 1:1 stoichiometry and simultaneous engagement of both A20 and A26 epitopes as expected based on the biparatopic design inspired by the crystal structures of TcdA bound to A20 and A26. In contrast, the control constructs show varied and heterogeneous binding modes. These results highlight the importance of molecular geometric constraints in generating highly potent antibody-based reagents capable of exploiting the simultaneous binding of more than one paratope to an antigen.
Collapse
Affiliation(s)
- Greg Hussack
- Life Sciences Division, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Martin A. Rossotti
- Life Sciences Division, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Henk van Faassen
- Life Sciences Division, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Tomohiko Murase
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Luiz Eugenio
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Joseph D. Schrag
- Life Sciences Division, Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, QC, Canada
| | - Kenneth K.-S. Ng
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada,Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON, Canada,*Correspondence: Kenneth K.-S. Ng,
| | - Jamshid Tanha
- Life Sciences Division, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada,Jamshid Tanha,
| |
Collapse
|
13
|
Rasetti-Escargueil C, Avril A. Medical Countermeasures against Ricin Intoxication. Toxins (Basel) 2023; 15:toxins15020100. [PMID: 36828415 PMCID: PMC9966136 DOI: 10.3390/toxins15020100] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/14/2022] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
Ricin toxin is a disulfide-linked glycoprotein (AB toxin) comprising one enzymatic A chain (RTA) and one cell-binding B chain (RTB) contained in the castor bean, a Ricinus species. Ricin inhibits peptide chain elongation via disruption of the binding between elongation factors and ribosomes, resulting in apoptosis, inflammation, oxidative stress, and DNA damage, in addition to the classically known rRNA damage. Ricin has been used in traditional medicine throughout the world since prehistoric times. Because ricin toxin is highly toxic and can be readily extracted from beans, it could be used as a bioweapon (CDC B-list). Due to its extreme lethality and potential use as a biological weapon, ricin toxin remains a global public health concern requiring specific countermeasures. Currently, no specific treatment for ricin intoxication is available. This review focuses on the drugs under development. In particular, some examples are reviewed to demonstrate the proof of concept of antibody-based therapy. Chemical inhibitors, small proteins, and vaccines can serve as alternatives to antibodies or may be used in combination with antibodies.
Collapse
Affiliation(s)
- Christine Rasetti-Escargueil
- Unité des Bactéries Anaérobies et Toxines, Institut Pasteur, 25 Avenue du Docteur Roux, 75015 Paris, France
- Correspondence:
| | - Arnaud Avril
- Unité Immunopathologies, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France
| |
Collapse
|
14
|
Tsukahara N, Murakami A, Motohashi M, Nakayama H, Kondo Y, Ito Y, Azuma T, Kishimoto H. An alpaca single-domain antibody (VHH) phage display library constructed by CDR shuffling provided high-affinity VHHs against desired protein antigens. Int Immunol 2022; 34:421-434. [PMID: 35689594 DOI: 10.1093/intimm/dxac022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Antigen-combining sites of the camelid heavy-chain antibody variable domain (VHH) are constructed by three complementarity-determining regions (CDR1, CDR2 and CDR3). We prepared cDNA using mRNA extracted from peripheral lymphocytes of alpacas that had been non-immunized or immunized with human serum albumin (HSA). The VHH gene fragments encoding the amino-terminal half-containing CDR1 as well as CDR2 and the carboxy-terminal half-containing CDR3 were amplified independently by PCR, and then full-length VHH gene fragments were generated by overlap extension PCR and cloned into the phagemid vector. This protocol, referred to as CDR shuffling, allowed us to construct an alpaca VHH phage display library possessing repertoires different from those naturally occurring in animals. We asked, first, whether this library was able to provide the functional VHH fragments against HSA, an immunized antigen, and obtained 29 anti-HSA VHH clones, 41% possessed KD values of lower than 10-8 M, 5 of which had KD values of 10-10 M. We also obtained VHH clones against non-immunized protein antigens such as cardiac troponin T and I, Ebola virus glycoprotein 1 and human immunoglobulin G by biopanning. We compared the amino acid sequences and affinities and found that 43% of VHHs had KD values of less than 10-8 M, although those having KD values of 10-10 M were unavailable. These results suggested that the CDR-shuffled VHH phage display library could potentially provide VHHs against non-immunized protein antigens with similar levels of affinities to those against immunized antigens.
Collapse
Affiliation(s)
- Narutoshi Tsukahara
- Department of Immunology & Parasitology, Graduate School of Medicine, University of the Ryukyus, Uehara, Nishihara, Nakagami, Okinawa, Japan.,RePHAGEN Co., Ltd., Suzaki, Uruma, Okinawa, Japan
| | - Akikazu Murakami
- RePHAGEN Co., Ltd., Suzaki, Uruma, Okinawa, Japan.,Department of Oral Microbiology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto, Tokushima, Tokushima, Japan
| | - Maiko Motohashi
- Department of Immunology & Parasitology, Graduate School of Medicine, University of the Ryukyus, Uehara, Nishihara, Nakagami, Okinawa, Japan
| | | | | | - Yuji Ito
- Graduate School of Science and Engineering, Kagoshima University, Korimoto, Kagoshima, Kagoshima, Japan
| | - Takachika Azuma
- Antibody Engineering Research Center Co., Ltd., Yamazaki, Noda, Chiba, Japan
| | - Hidehiro Kishimoto
- Department of Immunology & Parasitology, Graduate School of Medicine, University of the Ryukyus, Uehara, Nishihara, Nakagami, Okinawa, Japan
| |
Collapse
|
15
|
Intramuscular delivery of formulated RNA encoding six linked nanobodies is highly protective for exposures to three Botulinum neurotoxin serotypes. Sci Rep 2022; 12:11664. [PMID: 35803998 PMCID: PMC9266081 DOI: 10.1038/s41598-022-15876-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 06/30/2022] [Indexed: 01/07/2023] Open
Abstract
Single domain antibodies (sdAbs), also called nanobodies, have substantial biophysical advantages over conventional antibodies and are increasingly being employed as components of immunotherapeutic agents. One particularly favorable property is the ability to link different sdAbs into heteromultimers. This feature allows production of single molecules capable of simultaneously targeting more than one antigen. In addition, cooperative binding of multiple linked sdAbs to non-overlapping epitopes on the same target can produce synergistic improvements in target affinity, variant specificity, and in vivo potencies. Here we seek to test the option of increased component sdAbs in these heteromultimers by testing different sdAb heterohexamers in which each of the six camelid sdAb components (VHHs) can neutralize one of three different Botulinum neurotoxin (BoNT) serotypes, A, B or E. Each heterohexamer bound all three targeted BoNT serotypes and protected mice from at least 100 MIPLD50 of each serotype. To test the potential of mRNA therapeutics encoding long sdAb heteromultimers, one heterohexamer was encoded as replicating RNA (repRNA), formulated with a cationic nanocarrier, and delivered to mice via intramuscular injection. Heterohexamer antitoxin serum expression levels were easily detected by 8 h post-treatment, peaked at 5–10 nM around two days, and persisted for more than three days. Mice treated with the formulated repRNA one day post-treatment survived challenge with 100 MIPLD50 of each toxin serotype, demonstrating the function of all six component VHHs. Use of long sdAb multimers, administered as proteins or repRNA, offer the potential for substantially improved versatility in the development of antibody-based therapeutics.
Collapse
|
16
|
Ricin toxin and its neutralizing antibodies: A review. Toxicon 2022; 214:47-53. [PMID: 35595086 DOI: 10.1016/j.toxicon.2022.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/22/2022]
Abstract
Ricin toxin (RT) belongs to the ribosome-inactivating protein (RIP) family of toxins and is considered to be a moderate threat by the US Center of Disease Control and Prevention (CDC). RT poses a great potential threat to the public, but there has been a lack of effective treatment options so far. Over the past few decades, researches on the prevention and treatment of RT poisoning have been investigated, among which neutralizing antibodies targeting RT specifically have always been a research hotspot. In this review, we have summarized the mechanism of action of RT, the research results and the design strategies of RT neutralizing antibodies, and discussed the key issues in the development of RT neutralizing antibody researches.
Collapse
|
17
|
Neutralization of typhoid toxin by alpaca-derived, single-domain antibodies targeting the PltB and CdtB subunits. Infect Immun 2021; 90:e0051521. [PMID: 34898253 PMCID: PMC8852740 DOI: 10.1128/iai.00515-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Typhoid toxin is secreted by the typhoid fever-causing bacterial pathogen Salmonella enterica serovar Typhi and has tropism for immune cells and brain endothelial cells. Here, we generated a camelid single-domain antibody (VHH) library from typhoid toxoid-immunized alpacas and identified 41 VHHs selected on the glycan receptor-binding PltB and nuclease CdtB. VHHs exhibiting potent in vitro neutralizing activities from each sequence-based family were epitope binned via competition enzyme-linked immunosorbent assays (ELISAs), leading to 6 distinct VHHs, 2 anti-PltBs (T2E7 and T2G9), and 4 anti-CdtB VHHs (T4C4, T4C12, T4E5, and T4E8), whose in vivo neutralizing activities and associated toxin-neutralizing mechanisms were investigated. We found that T2E7, T2G9, and T4E5 effectively neutralized typhoid toxin in vivo, as demonstrated by 100% survival of mice administered a lethal dose of typhoid toxin and with little to no typhoid toxin-mediated upper motor function defect. Cumulatively, these results highlight the potential of the compact antibodies to neutralize typhoid toxin by targeting the glycan-binding and/or nuclease subunits.
Collapse
|
18
|
Structural Insights into Rational Design of Single-Domain Antibody-Based Antitoxins against Botulinum Neurotoxins. Cell Rep 2021; 30:2526-2539.e6. [PMID: 32101733 PMCID: PMC7138525 DOI: 10.1016/j.celrep.2020.01.107] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/23/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Botulinum neurotoxin (BoNT) is one of the most acutely lethal toxins known to humans, and effective treatment for BoNT intoxication is urgently needed. Single-domain antibodies (VHH) have been examined as a countermeasure for BoNT because of their high stability and ease of production. Here, we investigate the structures and the neutralization mechanisms for six unique VHHs targeting BoNT/A1 or BoNT/B1. These studies reveal diverse neutralizing mechanisms by which VHHs prevent host receptor binding or block transmembrane delivery of the BoNT protease domain. Guided by this knowledge, we design heterodimeric VHHs by connecting two neutralizing VHHs via a flexible spacer so they can bind simultaneously to the toxin. These bifunctional VHHs display much greater potency in a mouse co-intoxication model than similar heterodimers unable to bind simultaneously. Taken together, our studies offer insight into antibody neutralization of BoNTs and advance our ability to design multivalent anti-pathogen VHHs with improved therapeutic properties. Botulinum neurotoxins (BoNTs) are extremely toxic biothreats. Lam et al. report the crystal structures and neutralizing mechanisms of six unique antitoxin VHHs against BoNT/A1 and BoNT/B1, the two major human pathogenic BoNTs. They then develop a platform for structure-based rational design of bifunctional VHH heterodimers with superior antitoxin potencies.
Collapse
|
19
|
Wang W, Yuan J, Jiang C. Applications of nanobodies in plant science and biotechnology. PLANT MOLECULAR BIOLOGY 2021; 105:43-53. [PMID: 33037986 PMCID: PMC7547553 DOI: 10.1007/s11103-020-01082-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/05/2020] [Indexed: 05/15/2023]
Abstract
Present review summarizes the current applications of nanobodies in plant science and biotechnology, including plant expression of nanobodies, plant biotechnological applications, nanobody-based immunodetection, and nanobody-mediated resistance against plant pathogens. Nanobodies (Nbs) are variable domains of heavy chain-only antibodies (HCAbs) isolated from camelids. In spite of their single domain structure, nanobodies display many unique features, such as small size, high stability, and cryptic epitopes accessibility, which make them ideal for sophisticated applications in plants and animals. In this review, we summarize the current applications of nanobodies in plant science and biotechnology, focusing on nanobody expression in plants, plant biotechnological applications, determination of plant toxins and pathogens, and nanobody-mediated resistance against plant pathogens. Prospects and challenges of nanobody applications in plants are also discussed.
Collapse
Affiliation(s)
- Wenyi Wang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China.
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China.
| | - Jumao Yuan
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Changan Jiang
- Precision Medicine R&D Center, Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong Province, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| |
Collapse
|
20
|
Vance DJ, Poon AY, Mantis NJ. Sites of vulnerability on ricin B chain revealed through epitope mapping of toxin-neutralizing monoclonal antibodies. PLoS One 2020; 15:e0236538. [PMID: 33166282 PMCID: PMC7652295 DOI: 10.1371/journal.pone.0236538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022] Open
Abstract
Ricin toxin's B subunit (RTB) is a multifunctional galactose (Gal)-/N-acetylgalactosamine (GalNac)-specific lectin that promotes uptake and intracellular trafficking of ricin's ribosome-inactivating subunit (RTA) into mammalian cells. Structurally, RTB consists of two globular domains (RTB-D1, RTB-D2), each divided into three homologous sub-domains (α, β, γ). The two carbohydrate recognition domains (CRDs) are situated on opposite sides of RTB (sub-domains 1α and 2γ) and function non-cooperatively. Previous studies have revealed two distinct classes of toxin-neutralizing, anti-RTB monoclonal antibodies (mAbs). Type I mAbs, exemplified by SylH3, inhibit (~90%) toxin attachment to cell surfaces, while type II mAbs, epitomized by 24B11, interfere with intracellular toxin transport between the plasma membrane and the trans-Golgi network (TGN). Localizing the epitopes recognized by these two classes of mAbs has proven difficult, in part because of RTB's duplicative structure. To circumvent this problem, RTB-D1 and RTB-D2 were expressed as pIII fusion proteins on the surface of filamentous phage M13 and subsequently used as "bait" in mAb capture assays. We found that SylH3 captured RTB-D1 (but not RTB-D2) in a dose-dependent manner, while 24B11 captured RTB-D2 (but not RTB-D1) in a dose-dependent manner. We confirmed these domain assignments by competition studies with an additional 8 RTB-specific mAbs along with a dozen a single chain antibodies (VHHs). Collectively, these results demonstrate that type I and type II mAbs segregate on the basis of domain specificity and suggest that RTB's two domains may contribute to distinct steps in the intoxication pathway.
Collapse
Affiliation(s)
- David J. Vance
- Division of Infectious Disease, New York State Department of Health,Wadsworth Center, Albany, NY, United States of America
| | - Amanda Y. Poon
- Department of Biomedical Sciences, University at Albany School of Public Health, Albany, NY, United States of America
| | - Nicholas J. Mantis
- Division of Infectious Disease, New York State Department of Health,Wadsworth Center, Albany, NY, United States of America
- Department of Biomedical Sciences, University at Albany School of Public Health, Albany, NY, United States of America
| |
Collapse
|
21
|
Tremblay JM, Vazquez-Cintron E, Lam KH, Mukherjee J, Bedenice D, Ondeck CA, Conroy MT, Bodt SML, Winner BM, Webb RP, Ichtchenko K, Jin R, McNutt PM, Shoemaker CB. Camelid VHH Antibodies that Neutralize Botulinum Neurotoxin Serotype E Intoxication or Protease Function. Toxins (Basel) 2020; 12:toxins12100611. [PMID: 32987745 PMCID: PMC7598594 DOI: 10.3390/toxins12100611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022] Open
Abstract
Botulinum neurotoxin (BoNT) serotype E is one of three serotypes that cause the preponderance of human botulism cases and is a Tier 1 Select Agent. BoNT/E is unusual among BoNT serotypes for its rapid onset and short duration of intoxication. Here we report two large panels of unique, unrelated camelid single-domain antibodies (VHHs) that were selected for their ability to bind to BoNT/E holotoxin and/or to the BoNT/E light chain protease domain (LC/E). The 19 VHHs which bind to BoNT/E were characterized for their subunit specificity and 8 VHHs displayed the ability to neutralize BoNT/E intoxication of neurons. Heterodimer antitoxins consisting of two BoNT/E-neutralizing VHHs, including one heterodimer designed using structural information for simultaneous binding, were shown to protect mice against co-administered toxin challenges of up to 500 MIPLD50. The 22 unique VHHs which bind to LC/E were characterized for their binding properties and 9 displayed the ability to inhibit LC/E protease activity. Surprisingly, VHHs selected on plastic-coated LC/E were virtually unable to recognize soluble or captured LC/E while VHHs selected on captured LC/E were poorly able to recognize LC/E coated to a plastic surface. This panel of anti-LC/E VHHs offer insight into BoNT/E function, and some may have value as components of therapeutic antidotes that reverse paralysis following BoNT/E exposures.
Collapse
Affiliation(s)
- Jacqueline M. Tremblay
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA; (J.M.T.); (J.M.)
| | - Edwin Vazquez-Cintron
- The United States Army Medical Research Institute of Chemical Defense, Fort Detrick, MD 21010, USA; (E.V.-C.); (C.A.O.); (M.T.C.); (S.M.L.B.); (B.M.W.); (P.M.M.)
| | - Kwok-Ho Lam
- Department of Physiology & Biophysics, University of California, Irvine, CA 92697-4560, USA; (K.-H.L.); (R.J.)
| | - Jean Mukherjee
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA; (J.M.T.); (J.M.)
| | - Daniela Bedenice
- Department of Clinical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA;
| | - Celinia A. Ondeck
- The United States Army Medical Research Institute of Chemical Defense, Fort Detrick, MD 21010, USA; (E.V.-C.); (C.A.O.); (M.T.C.); (S.M.L.B.); (B.M.W.); (P.M.M.)
| | - Matthieu T. Conroy
- The United States Army Medical Research Institute of Chemical Defense, Fort Detrick, MD 21010, USA; (E.V.-C.); (C.A.O.); (M.T.C.); (S.M.L.B.); (B.M.W.); (P.M.M.)
| | - Skylar M. L. Bodt
- The United States Army Medical Research Institute of Chemical Defense, Fort Detrick, MD 21010, USA; (E.V.-C.); (C.A.O.); (M.T.C.); (S.M.L.B.); (B.M.W.); (P.M.M.)
| | - Brittany M. Winner
- The United States Army Medical Research Institute of Chemical Defense, Fort Detrick, MD 21010, USA; (E.V.-C.); (C.A.O.); (M.T.C.); (S.M.L.B.); (B.M.W.); (P.M.M.)
| | - Robert P. Webb
- Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD 21702-5011, USA;
| | - Konstantin Ichtchenko
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA;
| | - Rongsheng Jin
- Department of Physiology & Biophysics, University of California, Irvine, CA 92697-4560, USA; (K.-H.L.); (R.J.)
| | - Patrick M. McNutt
- The United States Army Medical Research Institute of Chemical Defense, Fort Detrick, MD 21010, USA; (E.V.-C.); (C.A.O.); (M.T.C.); (S.M.L.B.); (B.M.W.); (P.M.M.)
| | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA; (J.M.T.); (J.M.)
- Correspondence:
| |
Collapse
|
22
|
Falach R, Sapoznikov A, Evgy Y, Aftalion M, Makovitzki A, Agami A, Mimran A, Lerer E, Ben David A, Zichel R, Katalan S, Rosner A, Sabo T, Kronman C, Gal Y. Post-Exposure Anti-Ricin Treatment Protects Swine Against Lethal Systemic and Pulmonary Exposures. Toxins (Basel) 2020; 12:toxins12060354. [PMID: 32481526 PMCID: PMC7354453 DOI: 10.3390/toxins12060354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 12/04/2022] Open
Abstract
Ricin, a plant-derived toxin originating from the seeds of Ricinus communis (castor bean plant), is one of the most lethal toxins known. To date, there is no approved post-exposure therapy for ricin exposures. This work demonstrates for the first time the therapeutic efficacy of equine-derived anti-ricin F(ab’)2 antibodies against lethal pulmonary and systemic ricin exposures in swine. While administration of the antitoxin at 18 h post-exposure protected more than 80% of both intratracheally and intramuscularly ricin-intoxicated swine, treatment at 24 h post-exposure protected 58% of the intramuscular-exposed swine, as opposed to 26% of the intratracheally exposed animals. Quantitation of the anti-ricin neutralizing units in the anti-toxin preparations confirmed that the disparate protection conferred to swine subjected to the two routes of exposure stems from variance between the two models. Furthermore, dose response experiments showed that approximately 3 times lesser amounts of antibody are needed for high-level protection of the intramuscularly compared to the intratracheally intoxicated swine. This study, which demonstrates the high-level post-exposure efficacy of anti-ricin antitoxin at clinically relevant time-points in a large animal model, can serve as the basis for the formulation of post-exposure countermeasures against ricin poisoning in humans.
Collapse
Affiliation(s)
- Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (R.F.); (A.S.); (Y.E.); (M.A.); (T.S.)
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (R.F.); (A.S.); (Y.E.); (M.A.); (T.S.)
| | - Yentl Evgy
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (R.F.); (A.S.); (Y.E.); (M.A.); (T.S.)
| | - Moshe Aftalion
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (R.F.); (A.S.); (Y.E.); (M.A.); (T.S.)
| | - Arik Makovitzki
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (A.M.); (A.A.); (A.M.); (E.L.); (A.B.D.); (R.Z.)
| | - Avi Agami
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (A.M.); (A.A.); (A.M.); (E.L.); (A.B.D.); (R.Z.)
| | - Avishai Mimran
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (A.M.); (A.A.); (A.M.); (E.L.); (A.B.D.); (R.Z.)
| | - Elad Lerer
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (A.M.); (A.A.); (A.M.); (E.L.); (A.B.D.); (R.Z.)
| | - Alon Ben David
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (A.M.); (A.A.); (A.M.); (E.L.); (A.B.D.); (R.Z.)
| | - Ran Zichel
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (A.M.); (A.A.); (A.M.); (E.L.); (A.B.D.); (R.Z.)
| | - Shahaf Katalan
- Department of Pharmacology, Israel Institute for Biological Research, Ness-Ziona 76100, Israel;
| | - Amir Rosner
- Veterinary Center for Preclinical Research, Israel Institute for Biological Research, Ness-Ziona 76100, Israel;
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (R.F.); (A.S.); (Y.E.); (M.A.); (T.S.)
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (R.F.); (A.S.); (Y.E.); (M.A.); (T.S.)
- Correspondence: (C.K.); (Y.G.); Tel.: +972–8–9381522 (C.K.); +972–8–9381479 (Y.G.)
| | - Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel; (R.F.); (A.S.); (Y.E.); (M.A.); (T.S.)
- Correspondence: (C.K.); (Y.G.); Tel.: +972–8–9381522 (C.K.); +972–8–9381479 (Y.G.)
| |
Collapse
|
23
|
Rong Y, Pauly M, Guthals A, Pham H, Ehrbar D, Zeitlin L, Mantis NJ. A Humanized Monoclonal Antibody Cocktail to Prevent Pulmonary Ricin Intoxication. Toxins (Basel) 2020. [PMID: 32235318 DOI: 10.3390/toxins1204215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
PB10 IgG1, a monoclonal antibody (MAb) directed against an immunodominant epitope on the enzymatic subunit (RTA) of ricin toxin (RT), has been shown to passively protect mice and non-human primates from an aerosolized lethal-dose RT challenge. However, it was recently demonstrated that the therapeutic efficacy of PB10 IgG1 is significantly improved when co-administered with a second MAb, SylH3, targeting RT's binding subunit (RTB). Here we report that the PB10/SylH3 cocktail is also superior to PB10 alone when used as a pre-exposure prophylactic (PrEP) in a mouse model of intranasal RT challenge. The benefit of the PB10/SylH3 cocktail prompted us to engineer a humanized IgG1 version of SylH3 (huSylH3). The huPB10/huSylH3 cocktail proved highly efficacious in the mouse model, thereby opening the door to future testing in non-human primates.
Collapse
MESH Headings
- Administration, Inhalation
- Animals
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/pharmacology
- Antidotes/administration & dosage
- Antidotes/pharmacology
- Chlorocebus aethiops
- Disease Models, Animal
- Drug Therapy, Combination
- Female
- Lung Diseases/chemically induced
- Lung Diseases/prevention & control
- Mice, Inbred BALB C
- Pre-Exposure Prophylaxis
- Ricin/antagonists & inhibitors
- Ricin/immunology
- Vero Cells
Collapse
Affiliation(s)
- Yinghui Rong
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| | - Michael Pauly
- Mapp Biopharmaceutical, Inc. 6160 Lusk Blvd, San Diego, CA 92121, USA
| | - Adrian Guthals
- Mapp Biopharmaceutical, Inc. 6160 Lusk Blvd, San Diego, CA 92121, USA
| | - Henry Pham
- Mapp Biopharmaceutical, Inc. 6160 Lusk Blvd, San Diego, CA 92121, USA
| | - Dylan Ehrbar
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc. 6160 Lusk Blvd, San Diego, CA 92121, USA
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| |
Collapse
|
24
|
A Humanized Monoclonal Antibody Cocktail to Prevent Pulmonary Ricin Intoxication. Toxins (Basel) 2020; 12:toxins12040215. [PMID: 32235318 PMCID: PMC7232472 DOI: 10.3390/toxins12040215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
PB10 IgG1, a monoclonal antibody (MAb) directed against an immunodominant epitope on the enzymatic subunit (RTA) of ricin toxin (RT), has been shown to passively protect mice and non-human primates from an aerosolized lethal-dose RT challenge. However, it was recently demonstrated that the therapeutic efficacy of PB10 IgG1 is significantly improved when co-administered with a second MAb, SylH3, targeting RT’s binding subunit (RTB). Here we report that the PB10/SylH3 cocktail is also superior to PB10 alone when used as a pre-exposure prophylactic (PrEP) in a mouse model of intranasal RT challenge. The benefit of the PB10/SylH3 cocktail prompted us to engineer a humanized IgG1 version of SylH3 (huSylH3). The huPB10/huSylH3 cocktail proved highly efficacious in the mouse model, thereby opening the door to future testing in non-human primates.
Collapse
|
25
|
Mooney B, Torres‐Velez FJ, Doering J, Ehrbar DJ, Mantis NJ. Sensitivity of Kupffer cells and liver sinusoidal endothelial cells to ricin toxin and ricin toxin-Ab complexes. J Leukoc Biol 2019; 106:1161-1176. [PMID: 31313388 PMCID: PMC7008010 DOI: 10.1002/jlb.4a0419-123r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/03/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022] Open
Abstract
Ricin toxin is a plant-derived, ribosome-inactivating protein that is rapidly cleared from circulation by Kupffer cells (KCs) and liver sinusoidal endothelial cells (LSECs)-with fatal consequences. Rather than being inactivated, ricin evades normal degradative pathways and kills both KCs and LSECs with remarkable efficiency. Uptake of ricin by these 2 specialized cell types in the liver occurs by 2 parallel routes: a "lactose-sensitive" pathway mediated by ricin's galactose/N-acetylgalactosamine-specific lectin subunit (RTB), and a "mannose-sensitive" pathway mediated by the mannose receptor (MR; CD206) or other C-type lectins capable of recognizing the mannose-side chains displayed on ricin's A (RTA) and B subunits. In this report, we investigated the capacity of a collection of ricin-specific mouse MAb and camelid single-domain (VH H) antibodies to protect KCs and LSECs from ricin-induced killing. In the case of KCs, individual MAbs against RTA or RTB afforded near complete protection against ricin in ex vivo and in vivo challenge studies. In contrast, individual MAbs or VH Hs afforded little (<40%) or even no protection to LSECs against ricin-induced death. Complete protection of LSECs was only achieved with MAb or VH H cocktails, with the most effective mixtures targeting RTA and RTB simultaneously. Although the exact mechanisms of protection of LSECs remain unknown, evidence indicates that the Ab cocktails exert their effects on the mannose-sensitive uptake pathway without the need for Fcγ receptor involvement. In addition to advancing our understanding of how toxins and small immune complexes are processed by KCs and LSECs, our study has important implications for the development of Ab-based therapies designed to prevent or treat ricin exposure should the toxin be weaponized.
Collapse
Affiliation(s)
- Bridget Mooney
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| | - Fernando J. Torres‐Velez
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| | - Jennifer Doering
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| | - Dylan J. Ehrbar
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| | - Nicholas J. Mantis
- Division of Infectious DiseasesWadsworth CenterNew York State Department of HealthAlbanyNew YorkUSA
| |
Collapse
|
26
|
Schlake T, Thran M, Fiedler K, Heidenreich R, Petsch B, Fotin-Mleczek M. mRNA: A Novel Avenue to Antibody Therapy? Mol Ther 2019; 27:773-784. [PMID: 30885573 PMCID: PMC6453519 DOI: 10.1016/j.ymthe.2019.03.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/12/2022] Open
Abstract
First attempts to use exogenous mRNA for protein expression in vivo were made more than 25 years ago. However, widespread appreciation of in vitro transcribed mRNA as a powerful technology for supplying therapeutic proteins to the body has evolved only during the past few years. Various approaches to turning mRNA into a potent therapeutic have been developed. All of them share utilization of specifically designed, rather than endogenous, sequences and thorough purification protocols. Apart from this, there are two fundamental philosophies, one promoting the use of chemically modified nucleotides, the other advocating restriction to unmodified building blocks. Meanwhile, both strategies have received broad support by successful mRNA-based protein treatments in animal models. For such in vivo use, specifically optimized mRNA had to be combined with potent formulations to enable efficient in vivo delivery. The present review analyzes the applicability of mRNA technology to antibody therapy in all main fields: antitoxins, infectious diseases, and oncology.
Collapse
|
27
|
Whole-Cell Multiparameter Assay for Ricin and Abrin Activity-Based Digital Holographic Microscopy. Toxins (Basel) 2019; 11:toxins11030174. [PMID: 30909438 PMCID: PMC6468687 DOI: 10.3390/toxins11030174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/11/2019] [Accepted: 03/15/2019] [Indexed: 01/25/2023] Open
Abstract
Ricin and abrin are ribosome-inactivating proteins leading to inhibition of protein synthesis and cell death. These toxins are considered some of the most potent and lethal toxins against which there is no available antidote. Digital holographic microscopy (DHM) is a time-lapse, label-free, and noninvasive imaging technique that can provide phase information on morphological features of cells. In this study, we employed DHM to evaluate the morphological changes of cell lines during ricin and abrin intoxication. We showed that the effect of these toxins is characterized by a decrease in cell confluence and changes in morphological parameters such as cell area, perimeter, irregularity, and roughness. In addition, changes in optical parameters such as phase-shift, optical thickness, and effective-calculated volume were observed. These effects were completely inhibited by specific neutralizing antibodies. An enhanced intoxication effect was observed for preadherent compared to adherent cells, as was detected in early morphology changes and confirmed by annexin V/propidium iodide (PI) apoptosis assay. Detection of the dynamic changes in cell morphology at initial stages of cell intoxication by DHM emphasizes the highly sensitive and rapid nature of this method, allowing the early detection of active toxins.
Collapse
|
28
|
Abstract
In this report, we used hydrogen exchange-mass spectrometry (HX-MS) to identify the epitopes recognized by 21 single-domain camelid antibodies (VHHs) directed against the ribosome-inactivating subunit (RTA) of ricin toxin, a biothreat agent of concern to military and public health authorities. The VHHs, which derive from 11 different B-cell lineages, were binned together based on competition ELISAs with IB2, a monoclonal antibody that defines a toxin-neutralizing hotspot ("cluster 3") located in close proximity to RTA's active site. HX-MS analysis revealed that the 21 VHHs recognized four distinct epitope subclusters (3.1-3.4). Sixteen of the 21 VHHs grouped within subcluster 3.1 and engage RTA α-helices C and G. Three VHHs grouped within subcluster 3.2, encompassing a-helices C and G, plus α-helix B. The single VHH in subcluster 3.3 engaged RTA α-helices B and G, while the epitope of the sole VHH defining subcluster 3.4 encompassed α-helices C and E, and β-strand h. Modeling these epitopes on the surface of RTA predicts that the 20 VHHs within subclusters 3.1-3.3 physically occlude RTA's active site cleft, while the single antibody in subcluster 3.4 associates on the active site's upper rim.
Collapse
|
29
|
Generation of Highly Efficient Equine-Derived Antibodies for Post-Exposure Treatment of Ricin Intoxications by Vaccination with Monomerized Ricin. Toxins (Basel) 2018; 10:toxins10110466. [PMID: 30424519 PMCID: PMC6267474 DOI: 10.3390/toxins10110466] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/04/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022] Open
Abstract
Ricin, a highly lethal toxin derived from the seeds of Ricinus communis (castor beans) is considered a potential biological threat agent due to its high availability, ease of production, and to the lack of any approved medical countermeasure against ricin exposures. To date, the use of neutralizing antibodies is the most promising post-exposure treatment for ricin intoxication. The aim of this work was to generate anti-ricin antitoxin that confers high level post-exposure protection against ricin challenge. Due to safety issues regarding the usage of ricin holotoxin as an antigen, we generated an inactivated toxin that would reduce health risks for both the immunizer and the immunized animal. To this end, a monomerized ricin antigen was constructed by reducing highly purified ricin to its monomeric constituents. Preliminary immunizing experiments in rabbits indicated that this monomerized antigen is as effective as the native toxin in terms of neutralizing antibody elicitation and protection of mice against lethal ricin challenges. Characterization of the monomerized antigen demonstrated that the irreversibly detached A and B subunits retain catalytic and lectin activity, respectively, implying that the monomerization process did not significantly affect their overall structure. Toxicity studies revealed that the monomerized ricin displayed a 250-fold decreased activity in a cell culture-based functionality test, while clinical signs were undetectable in mice injected with this antigen. Immunization of a horse with the monomerized toxin was highly effective in elicitation of high titers of neutralizing antibodies. Due to the increased potential of IgG-derived adverse events, anti-ricin F(ab')₂ antitoxin was produced. The F(ab')₂-based antitoxin conferred high protection to intranasally ricin-intoxicated mice; ~60% and ~34% survival, when administered 24 and 48 h post exposure to a lethal dose, respectively. In line with the enhanced protection, anti-inflammatory and anti-edematous effects were measured in the antitoxin treated mice, in comparison to mice that were intoxicated but not treated. Accordingly, this anti-ricin preparation is an excellent candidate for post exposure treatment of ricin intoxications.
Collapse
|
30
|
Rudolph MJ, Vance DJ, Kelow S, Angalakurthi SK, Nguyen S, Davis SA, Rong Y, Middaugh CR, Weis DD, Dunbrack R, Karanicolas J, Mantis NJ. Contribution of an unusual CDR2 element of a single domain antibody in ricin toxin binding affinity and neutralizing activity. Protein Eng Des Sel 2018; 31:277-287. [PMID: 30265352 PMCID: PMC6277176 DOI: 10.1093/protein/gzy022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/03/2018] [Indexed: 11/13/2022] Open
Abstract
Ricin toxin's enzymatic subunit (RTA) has been subjected to intensive B cell epitope mapping studies using a combination of competition ELISAs, hydrogen exchange-mass spectrometry and X-ray crystallography. Those studies identified four spatially distinct clusters (I-IV) of toxin-neutralizing epitopes on the surface of RTA. Here we describe A9, a new single domain camelid antibody (VHH) that was proposed to recognize a novel epitope on RTA that straddles clusters I and III. The X-ray crystal structure of A9 bound to RTA (2.6 Å resolution) revealed extensive antibody contact with RTA's β-strand h (732 Å2 buried surface area; BSA), along with limited engagement with α-helix D (90 Å2) and α-helix C (138 Å2). Collectively, these contacts explain the overlap between epitope clusters I and III, as identified by competition ELISA. However, considerable binding affinity, and, consequently, toxin-neutralizing activity of A9 is mediated by an unusual CDR2 containing five consecutive Gly residues that interact with α-helix B (82 Å2), a known neutralizing hotspot on RTA. Removal of a single Gly residue from the penta-glycine stretch in CDR2 reduced A9's binding affinity by 10-fold and eliminated toxin-neutralizing activity. Computational modeling indicates that removal of a Gly from CDR2 does not perturb contact with RTA per se, but results in the loss of an intramolecular hydrogen bond network involved in stabilizing CDR2 in the unbound state. These results reveal a novel configuration of a CDR2 element involved in neutralizing ricin toxin.
Collapse
Affiliation(s)
| | - David J Vance
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Simon Kelow
- Department of Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Siva Krishna Angalakurthi
- Department of Pharmaceutical Chemistry and Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, KS, USA
| | - Sophie Nguyen
- New York Structural Biology Center, New York, NY, USA
| | - Simon A Davis
- New York Structural Biology Center, New York, NY, USA
| | - Yinghui Rong
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry and Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, KS, USA
| | - David D Weis
- Department of Chemistry and Ralph Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Roland Dunbrack
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John Karanicolas
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
31
|
Vance DJ, Tremblay JM, Rong Y, Angalakurthi SK, Volkin DB, Middaugh CR, Weis DD, Shoemaker CB, Mantis NJ. High-Resolution Epitope Positioning of a Large Collection of Neutralizing and Nonneutralizing Single-Domain Antibodies on the Enzymatic and Binding Subunits of Ricin Toxin. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:e00236-17. [PMID: 29021300 PMCID: PMC5717184 DOI: 10.1128/cvi.00236-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/02/2017] [Indexed: 01/05/2023]
Abstract
We previously produced a heavy-chain-only antibody (Ab) VH domain (VHH)-displayed phage library from two alpacas that had been immunized with ricin toxoid and nontoxic mixtures of the enzymatic ricin toxin A subunit (RTA) and binding ricin toxin B subunit (RTB) (D. J. Vance, J. M. Tremblay, N. J. Mantis, and C. B. Shoemaker, J Biol Chem 288:36538-36547, 2013, https://doi.org/10.1074/jbc.M113.519207). Initial and subsequent screens of that library by direct enzyme-linked immunosorbent assay (ELISA) yielded more than two dozen unique RTA- and RTB-specific VHHs, including 10 whose structures were subsequently solved in complex with RTA. To generate a more complete antigenic map of ricin toxin and to define the epitopes associated with toxin-neutralizing activity, we subjected the VHH-displayed phage library to additional "pannings" on both receptor-bound ricin and antibody-captured ricin. We now report the full-length DNA sequences, binding affinities, and neutralizing activities of 68 unique VHHs: 31 against RTA, 33 against RTB, and 4 against ricin holotoxin. Epitope positioning was achieved through cross-competition ELISAs performed with a panel of monoclonal antibodies (MAbs) and verified, in some instances, with hydrogen-deuterium exchange mass spectrometry. The 68 VHHs grouped into more than 20 different competition bins. The RTA-specific VHHs with strong toxin-neutralizing activities were confined to bins that overlapped two previously identified neutralizing hot spots, termed clusters I and II. The four RTB-specific VHHs with potent toxin-neutralizing activity grouped within three adjacent bins situated at the RTA-RTB interface near cluster II. These results provide important insights into epitope interrelationships on the surface of ricin and delineate regions of vulnerability that can be exploited for the purpose of vaccine and therapeutic development.
Collapse
Affiliation(s)
- David J Vance
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Jacqueline M Tremblay
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, USA
| | - Yinghui Rong
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Siva Krishna Angalakurthi
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas, USA
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas, USA
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas, USA
| | - David D Weis
- Department of Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Charles B Shoemaker
- Department of Infectious Disease and Global Health, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, USA
| | - Nicholas J Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Biomedical Sciences, University at Albany, SUNY, Albany, New York, USA
| |
Collapse
|
32
|
A Supercluster of Neutralizing Epitopes at the Interface of Ricin's Enzymatic (RTA) and Binding (RTB) Subunits. Toxins (Basel) 2017; 9:toxins9120378. [PMID: 29168727 PMCID: PMC5744098 DOI: 10.3390/toxins9120378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/10/2017] [Accepted: 11/18/2017] [Indexed: 12/29/2022] Open
Abstract
As part of an effort to engineer ricin antitoxins and immunotherapies, we previously produced and characterized a collection of phage-displayed, heavy chain-only antibodies (VHHs) from alpacas that had been immunized with ricin antigens. In our initial screens, we identified nine VHHs directed against ricin toxin’s binding subunit (RTB), but only one, JIZ-B7, had toxin-neutralizing activity. Linking JIZ-B7 to different VHHs against ricin’s enzymatic subunit (RTA) resulted in several bispecific antibodies with potent toxin-neutralizing activity in vitro and in vivo. JIZ-B7 may therefore be an integral component of a future VHH-based neutralizing agent (VNA) for ricin toxin. In this study, we now localize, using competitive ELISA, JIZ-B7’s epitope to a region of RTB’s domain 2 sandwiched between the high-affinity galactose/N-acetylgalactosamine (Gal/GalNAc)-binding site and the boundary of a neutralizing hotspot on RTA known as cluster II. Analysis of additional RTB (n = 8)- and holotoxin (n = 4)-specific VHHs from a recent series of screens identified a “supercluster” of neutralizing epitopes at the RTA-RTB interface. Among the VHHs tested, toxin-neutralizing activity was most closely associated with epitope proximity to RTA, and not interference with RTB’s ability to engage Gal/GalNAc receptors. We conclude that JIZ-B7 is representative of a larger group of potent toxin-neutralizing antibodies, possibly including many described in the literature dating back several decades, that recognize tertiary and possibly quaternary epitopes located at the RTA-RTB interface and that target a region of vulnerability on ricin toxin.
Collapse
|
33
|
Rosenfeld R, Alcalay R, Mechaly A, Lapidoth G, Epstein E, Kronman C, J Fleishman S, Mazor O. Improved antibody-based ricin neutralization by affinity maturation is correlated with slower off-rate values. Protein Eng Des Sel 2017; 30:611-617. [PMID: 28472478 DOI: 10.1093/protein/gzx028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
While potent monoclonal antibodies against ricin were introduced over the years, the question whether increasing antibody affinity enables better toxin neutralization was not fully addressed yet. The aim of this study was to characterize the contribution of antibody affinity to the ricin neutralization potential of the antibody. cHD23 monoclonal antibody that targets the toxin B-subunit and interferes with its binding to membranal receptors, was isolated. In order to create antibody clones with improved affinity toward ricin, a scFv-phage display library containing mutated versions of the variable regions of cHD23 was constructed and clones with improved binding of ricin were isolated. Structural modeling of these mutants suggests that the inserted mutations may increase the antibody conformational flexibility thus improving its ability to bind ricin. While it was found that the selected clones exhibited improved neutralization of ricin, the correlation between the KD values and potency was only minor (r = 0.55). However, a positive correlation (r = 0.84) exist between the off-rate values (koff) of the affinity matured clones and their ability to neutralize ricin. As cell membranes display inordinately large amounts of potential surface binding sites for ricin, it is suggested that antibodies with improved off-rate values block the ability of the toxin to bind to target receptors, in a highly efficient manner. Currently, antibody-based therapy is the most effective treatment for ricin intoxication and it is anticipated that the findings of this study will provide useful information and a possible strategy to design an improved antibody-based therapy for the toxin.
Collapse
Affiliation(s)
- Ronit Rosenfeld
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Lerrer St., Ness-Ziona 74100, Israel
| | - Ron Alcalay
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Lerrer St., Ness-Ziona 74100, Israel
| | - Adva Mechaly
- Department of Infectious Diseases, Israel Institute for Biological Research, Lerrer St., Ness-Ziona 74100, Israel
| | - Gideon Lapidoth
- Department of Biomolecular Sciences, Weizmann Institute of Science, 234 Herzel St., Rehovot 7610001, Israel
| | - Eyal Epstein
- Department of Biotechnology, Israel Institute for Biological Research, Lerrer St., Ness-Ziona 74100, Israel
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Lerrer St., Ness-Ziona 74100, Israel
| | - Sarel J Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, 234 Herzel St., Rehovot 7610001, Israel
| | - Ohad Mazor
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Lerrer St., Ness-Ziona 74100, Israel
| |
Collapse
|
34
|
Leow CH, Fischer K, Leow CY, Cheng Q, Chuah C, McCarthy J. Single Domain Antibodies as New Biomarker Detectors. Diagnostics (Basel) 2017; 7:diagnostics7040052. [PMID: 29039819 PMCID: PMC5745390 DOI: 10.3390/diagnostics7040052] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 01/02/2023] Open
Abstract
Biomarkers are defined as indicators of biological processes, pathogenic processes, or pharmacological responses to a therapeutic intervention. Biomarkers have been widely used for early detection, prediction of response after treatment, and for monitoring the progression of diseases. Antibodies represent promising tools for recognition of biomarkers, and are widely deployed as analytical tools in clinical settings. For immunodiagnostics, antibodies are now exploited as binders for antigens of interest across a range of platforms. More recently, the discovery of antibody surface display and combinatorial chemistry techniques has allowed the exploration of new binders from a range of animals, for instance variable domains of new antigen receptors (VNAR) from shark and variable heavy chain domains (VHH) or nanobodies from camelids. These single domain antibodies (sdAbs) have some advantages over conventional murine immunoglobulin owing to the lack of a light chain, making them the smallest natural biomarker binders thus far identified. In this review, we will discuss several biomarkers used as a means to validate diseases progress. The potential functionality of modern singe domain antigen binders derived from phylogenetically early animals as new biomarker detectors for current diagnostic and research platforms development will be described.
Collapse
Affiliation(s)
- Chiuan Herng Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang 11800, Malaysia.
| | - Katja Fischer
- Bacterial Pathogenesis and Scabies Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia.
| | - Chiuan Yee Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kelantan 16150, Malaysia.
| | - Qin Cheng
- Department of Drug Resistance and Diagnostics, Australian Army Malaria Institute, Brisbane 4051, Australia.
| | - Candy Chuah
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan 16150, Malaysia.
| | - James McCarthy
- Clinical Tropical Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia.
| |
Collapse
|
35
|
Barta ML, Shearer JP, Arizmendi O, Tremblay JM, Mehzabeen N, Zheng Q, Battaile KP, Lovell S, Tzipori S, Picking WD, Shoemaker CB, Picking WL. Single-domain antibodies pinpoint potential targets within Shigella invasion plasmid antigen D of the needle tip complex for inhibition of type III secretion. J Biol Chem 2017; 292:16677-16687. [PMID: 28842484 DOI: 10.1074/jbc.m117.802231] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/14/2017] [Indexed: 12/18/2022] Open
Abstract
Numerous Gram-negative pathogens infect eukaryotes and use the type III secretion system (T3SS) to deliver effector proteins into host cells. One important T3SS feature is an extracellular needle with an associated tip complex responsible for assembly of a pore-forming translocon in the host cell membrane. Shigella spp. cause shigellosis, also called bacillary dysentery, and invade colonic epithelial cells via the T3SS. The tip complex of Shigella flexneri contains invasion plasmid antigen D (IpaD), which initially regulates secretion and provides a physical platform for the translocon pore. The tip complex represents a promising therapeutic target for many important T3SS-containing pathogens. Here, in an effort to further elucidate its function, we created a panel of single-VH domain antibodies (VHHs) that recognize distinct epitopes within IpaD. These VHHs recognized the in situ tip complex and modulated the infectious properties of Shigella Moreover, structural elucidation of several IpaD-VHH complexes provided critical insights into tip complex formation and function. Of note, one VHH heterodimer could reduce Shigella hemolytic activity by >80%. Our observations along with previous findings support the hypothesis that the hydrophobic translocator (IpaB in Shigella) likely binds to a region within the tip protein that is structurally conserved across all T3SS-possessing pathogens, suggesting potential therapeutic avenues for managing infections by these pathogens.
Collapse
Affiliation(s)
- Michael L Barta
- From the Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047
| | - Jonathan P Shearer
- Department of Infectious Diseases and Global Health, Tufts Clinical and Translational Science Institute, North Grafton, Massachusetts 02111
| | - Olivia Arizmendi
- From the Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047
| | - Jacqueline M Tremblay
- Department of Infectious Diseases and Global Health, Tufts Clinical and Translational Science Institute, North Grafton, Massachusetts 02111
| | - Nurjahan Mehzabeen
- Protein Structure Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, and
| | - Qi Zheng
- From the Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047
| | - Kevin P Battaile
- IMCA-CAT, Hauptman-Woodward Medical Research Institute, Argonne, Illinois 60439
| | - Scott Lovell
- Protein Structure Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, and
| | - Saul Tzipori
- Department of Infectious Diseases and Global Health, Tufts Clinical and Translational Science Institute, North Grafton, Massachusetts 02111
| | - William D Picking
- From the Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047
| | - Charles B Shoemaker
- Department of Infectious Diseases and Global Health, Tufts Clinical and Translational Science Institute, North Grafton, Massachusetts 02111
| | - Wendy L Picking
- From the Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas 66047,
| |
Collapse
|
36
|
Bazzoli A, Vance DJ, Rudolph MJ, Rong Y, Angalakurthi SK, Toth RT, Middaugh CR, Volkin DB, Weis DD, Karanicolas J, Mantis NJ. Using homology modeling to interrogate binding affinity in neutralization of ricin toxin by a family of single domain antibodies. Proteins 2017; 85:1994-2008. [PMID: 28718923 DOI: 10.1002/prot.25353] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/12/2022]
Abstract
In this report we investigated, within a group of closely related single domain camelid antibodies (VH Hs), the relationship between binding affinity and neutralizing activity as it pertains to ricin, a fast-acting toxin and biothreat agent. The V1C7-like VH Hs (V1C7, V2B9, V2E8, and V5C1) are similar in amino acid sequence, but differ in their binding affinities and toxin-neutralizing activities. Using the X-ray crystal structure of V1C7 in complex with ricin's enzymatic subunit (RTA) as a template, Rosetta-based homology modeling coupled with energetic decomposition led us to predict that a single pairwise interaction between Arg29 on V5C1 and Glu67 on RTA was responsible for the difference in ricin toxin binding affinity between V1C7, a weak neutralizer, and V5C1, a moderate neutralizer. This prediction was borne out experimentally: substitution of Arg for Gly at position 29 enhanced V1C7's binding affinity for ricin, whereas the reverse (ie, Gly for Arg at position 29) diminished V5C1's binding affinity by >10 fold. As expected, the V5C1R29G mutant was largely devoid of toxin-neutralizing activity (TNA). However, the TNA of the V1C7G29R mutant was not correspondingly improved, indicating that in the V1C7 family binding affinity alone does not account for differences in antibody function. V1C7 and V5C1, as well as their respective point mutants, recognized indistinguishable epitopes on RTA, at least at the level of sensitivity afforded by hydrogen-deuterium mass spectrometry. The results of this study have implications for engineering therapeutic antibodies because they demonstrate that even subtle differences in epitope specificity can account for important differences in antibody function.
Collapse
Affiliation(s)
- Andrea Bazzoli
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, 66045.,Computational Chemical Biology Core, University of Kansas, Lawrence, Kansas, 66047
| | - David J Vance
- New York State Department of Health, Division of Infectious Diseases, Wadsworth Center, Albany, New York, 12208
| | | | - Yinghui Rong
- New York State Department of Health, Division of Infectious Diseases, Wadsworth Center, Albany, New York, 12208
| | - Siva Krishna Angalakurthi
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas, 66045
| | - Ronald T Toth
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas, 66045
| | - C Russell Middaugh
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas, 66045
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas, 66045
| | - David D Weis
- Department of Chemistry, University of Kansas, Lawrence, Kansas, 66045
| | - John Karanicolas
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, 66045.,Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, 66045.,Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111
| | - Nicholas J Mantis
- New York State Department of Health, Division of Infectious Diseases, Wadsworth Center, Albany, New York, 12208
| |
Collapse
|
37
|
Beghein E, Gettemans J. Nanobody Technology: A Versatile Toolkit for Microscopic Imaging, Protein-Protein Interaction Analysis, and Protein Function Exploration. Front Immunol 2017; 8:771. [PMID: 28725224 PMCID: PMC5495861 DOI: 10.3389/fimmu.2017.00771] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/16/2017] [Indexed: 01/05/2023] Open
Abstract
Over the last two decades, nanobodies or single-domain antibodies have found their way in research, diagnostics, and therapy. These antigen-binding fragments, derived from Camelid heavy chain only antibodies, possess remarkable characteristics that favor their use over conventional antibodies or fragments thereof, in selected areas of research. In this review, we assess the current status of nanobodies as research tools in diverse aspects of fundamental research. We discuss the use of nanobodies as detection reagents in fluorescence microscopy and focus on recent advances in super-resolution microscopy. Second, application of nanobody technology in investigating protein–protein interactions is reviewed, with emphasis on possible uses in mass spectrometry. Finally, we discuss the potential value of nanobodies in studying protein function, and we focus on their recently reported application in targeted protein degradation. Throughout the review, we highlight state-of-the-art engineering strategies that could expand nanobody versatility and we suggest future applications of the technology in the selected areas of fundamental research.
Collapse
Affiliation(s)
- Els Beghein
- Nanobody Laboratory, Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Nanobody Laboratory, Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
38
|
Shriver-Lake LC, Goldman ER, Zabetakis D, Anderson GP. Improved production of single domain antibodies with two disulfide bonds by co-expression of chaperone proteins in the Escherichia coli periplasm. J Immunol Methods 2017; 443:64-67. [DOI: 10.1016/j.jim.2017.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 10/20/2022]
|
39
|
Pairing Alpaca and Llama-Derived Single Domain Antibodies to Enhance Immunoassays for Ricin. Antibodies (Basel) 2017; 6:antib6010003. [PMID: 31548519 PMCID: PMC6698814 DOI: 10.3390/antib6010003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/17/2017] [Indexed: 11/16/2022] Open
Abstract
Previously, our group isolated and evaluated anti-ricin single domain antibodies (sdAbs) derived from llamas, engineered them to further increase their thermal stability, and utilized them for the development of sensitive immunoassays. In work focused on the development of therapeutics, Vance et al. 2013 described anti-ricin sdAbs derived from alpacas. Herein, we evaluated the utility of selected alpaca-derived anti-ricin sdAbs for detection applications, and engineered an alpaca-derived sdAb to increase its melting temperature, providing a highly thermal stable reagent for use in ricin detection. Four of the alpaca-derived anti-ricin A-chain sdAbs were produced and characterized. All four bound to epitopes that overlapped with our previously described llama sdAbs. One alpaca sdAb, F6, was found to possess both a high melting temperature (73 °C) and to work optimally with a thermally stable llama anti-ricin sdAb in sandwich assays for ricin detection. We employed a combination of consensus sequence mutagenesis and the addition of a non-canonical disulfide bond to further enhance the thermal stability of F6 to 85 °C. It is advantageous to have a choice of recognition reagents when developing assays. This work resulted in defining an additional pair of highly thermal stable sdAbs for the sensitive detection of ricin.
Collapse
|
40
|
Rudolph MJ, Vance DJ, Cassidy MS, Rong Y, Mantis NJ. Structural Analysis of Single Domain Antibodies Bound to a Second Neutralizing Hot Spot on Ricin Toxin's Enzymatic Subunit. J Biol Chem 2017; 292:872-883. [PMID: 27903650 PMCID: PMC5247660 DOI: 10.1074/jbc.m116.758102] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/10/2016] [Indexed: 12/15/2022] Open
Abstract
Ricin toxin is a heterodimer consisting of RTA, a ribosome-inactivating protein, and RTB, a lectin that facilitates receptor-mediated uptake into mammalian cells. In previous studies, we demonstrated that toxin-neutralizing antibodies target four spatially distinct hot spots on RTA, which we refer to as epitope clusters I-IV. In this report, we identified and characterized three single domain camelid antibodies (VHH) against cluster II. One of these VHHs, V5E1, ranks as one of the most potent ricin-neutralizing antibodies described to date. We solved the X-ray crystal structures of each of the three VHHs (E1, V1C7, and V5E1) in complex with RTA. V5E1 buries a total of 1,133 Å2 of surface area on RTA and makes primary contacts with α-helix A (residues 18-32), α-helix F (182-194), as well as the F-G loop. V5E1, by virtue of complementarity determining region 3 (CDR3), may also engage with RTB and potentially interfere with the high affinity galactose-recognition element that plays a critical role in toxin attachment to cell surfaces and intracellular trafficking. The two other VHHs, E1 and V1C7, bind epitopes adjacent to V5E1 but display only weak toxin neutralizing activity, thereby providing structural insights into specific residues within cluster II that may be critical contact points for toxin inactivation.
Collapse
Affiliation(s)
- Michael J Rudolph
- From the New York Structural Biology Center, New York, New York 10027,
| | - David J Vance
- the Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York 12208, and
| | - Michael S Cassidy
- From the New York Structural Biology Center, New York, New York 10027
| | - Yinghui Rong
- the Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York 12208, and
| | - Nicholas J Mantis
- the Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York 12208, and
- the Department of Biomedical Sciences, University at Albany, Albany, New York 12201
| |
Collapse
|
41
|
Wagner EK, Wang X, Bui A, Maynard JA. Synergistic Neutralization of Pertussis Toxin by a Bispecific Antibody In Vitro and In Vivo. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:851-862. [PMID: 27581436 PMCID: PMC5098018 DOI: 10.1128/cvi.00371-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/25/2016] [Indexed: 02/08/2023]
Abstract
Bispecific antibodies are a rapidly growing class of therapeutic molecules, originally developed for the treatment of cancer but recently explored for the treatment of autoimmune and infectious diseases. Bordetella pertussis is a reemerging pathogen, and several of the key symptoms of infection are caused by the pertussis toxin (PTx). Two humanized antibodies, hu1B7 and hu11E6, bind distinct epitopes on PTx and, when coadministered, mitigate disease severity in murine and baboon models of infection. Here we describe the generation of a bispecific human IgG1 molecule combining the hu1B7 and hu11E6 binding sites via a knobs-in-holes design. The bispecific antibody showed binding activity equivalent to that of the antibody mixture in a competition enzyme-linked immunosorbent assay (ELISA). A CHO cell neutralization assay provided preliminary evidence for synergy between the two antibodies, while a murine model of PTx-induced leukocytosis definitively showed synergistic neutralization. Notably, the bispecific antibody retained the synergy observed for the antibody mixture, supporting the conclusion that synergy is due to simultaneous blockade of both the catalytic and receptor binding activities of pertussis toxin. These data suggest that a hu1B7/hu11E6 bispecific antibody is a viable alternative to an antibody mixture for pertussis treatment.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/metabolism
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/metabolism
- Antibodies, Monoclonal, Humanized/therapeutic use
- Binding Sites, Antibody
- Bordetella pertussis/immunology
- CHO Cells
- Cricetinae
- Cricetulus
- Drug Synergism
- Enzyme-Linked Immunosorbent Assay
- Epitopes/chemistry
- Epitopes/immunology
- Humans
- Immunoglobulin G/chemistry
- Immunoglobulin G/immunology
- Immunoglobulin G/isolation & purification
- Immunoglobulin G/metabolism
- Mice
- Neutralization Tests
- Pertussis Toxin/immunology
- Whooping Cough/immunology
- Whooping Cough/prevention & control
- Whooping Cough/therapy
Collapse
Affiliation(s)
- Ellen K Wagner
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA
| | - Xianzhe Wang
- Department of Biochemistry, Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Andre Bui
- Proteomics Facility, The University of Texas at Austin, Austin, Texas, USA
| | - Jennifer A Maynard
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas, USA
- Department of Biochemistry, Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
42
|
Legler PM, Compton JR, Hale ML, Anderson GP, Olson MA, Millard CB, Goldman ER. Stability of isolated antibody-antigen complexes as a predictive tool for selecting toxin neutralizing antibodies. MAbs 2016; 9:43-57. [PMID: 27660893 PMCID: PMC5240650 DOI: 10.1080/19420862.2016.1236882] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ricin is an A-B ribosome inactivating protein (RIP) toxin composed of an A-chain subunit (RTA) that contains a catalytic N-glycosidase and a B-chain (RTB) lectin domain that binds cell surface glycans. Ricin exploits retrograde transport to enter into the Golgi and the endoplasmic reticulum, and then dislocates into the cytoplasm where it can reach its substrate, the rRNA. A subset of isolated antibodies (Abs) raised against the RTA subunit protect against ricin intoxication, and RTA-based vaccine immunogens have been shown to provide long-lasting protective immunity against the holotoxin. Anti-RTA Abs are unlikely to cross a membrane and reach the cytoplasm to inhibit the enzymatic activity of the A-chain. Moreover, there is not a strict correlation between the apparent binding affinity (Ka) of anti-RTA Abs and their ability to successfully neutralize ricin toxicity. Some anti-RTA antibodies are toxin-neutralizing, whereas others are not. We hypothesize that neutralizing anti-RTA Abs may interfere selectively with conformational change(s) or partial unfolding required for toxin internalization. To test this hypothesis, we measured the melting temperatures (Tm) of neutralizing single-domain Ab (sdAb)-antigen (Ag) complexes relative to the Tm of the free antigen (Tm-shift = Tmcomplex – TmAg), and observed increases in the Tmcomplex of 9–20 degrees. In contrast, non-neutralizing sdAb-Ag complexes shifted the TmComplex by only 6–7 degrees. A strong linear correlation (r2 = 0.992) was observed between the magnitude of the Tm-shift and the viability of living cells treated with the sdAb and ricin holotoxin. The Tm-shift of the sdAb-Ag complex provided a quantitative biophysical parameter that could be used to predict and rank-order the toxin-neutralizing activities of Abs. We determined the first structure of an sdAb-RTA1-33/44-198 complex, and examined other sdAb-RTA complexes. We found that neutralizing sdAb bound to regions involved in the early stages of unfolding. These Abs likely interfere with steps preceding or following endocytosis that require conformational changes. This method may have utility for the characterization or rapid screening of other Ab that act to prevent conformational changes or unfolding as part of their mechanism of action.
Collapse
Affiliation(s)
| | | | - Martha L Hale
- c US Army Medical Research Institute of Infectious Diseases , Frederick , MD , USA
| | | | - Mark A Olson
- c US Army Medical Research Institute of Infectious Diseases , Frederick , MD , USA
| | - Charles B Millard
- c US Army Medical Research Institute of Infectious Diseases , Frederick , MD , USA
| | | |
Collapse
|
43
|
A Tetraspecific VHH-Based Neutralizing Antibody Modifies Disease Outcome in Three Animal Models of Clostridium difficile Infection. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:774-84. [PMID: 27413067 PMCID: PMC5014919 DOI: 10.1128/cvi.00730-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 07/01/2016] [Indexed: 12/17/2022]
Abstract
Clostridium difficile infection (CDI), a leading cause of nosocomial infection, is a serious disease in North America, Europe, and Asia. CDI varies greatly from asymptomatic carriage to life-threatening diarrhea, toxic megacolon, and toxemia. The incidence of community-acquired infection has increased due to the emergence of hypervirulent antibiotic-resistant strains. These new strains contribute to the frequent occurrence of disease relapse, complicating treatment, increasing hospital stays, and increasing morbidity and mortality among patients. Therefore, it is critical to develop new therapeutic approaches that bypass the development of antimicrobial resistance and avoid disruption of gut microflora. Here, we describe the construction of a single heteromultimeric VHH-based neutralizing agent (VNA) that targets the two primary virulence factors of Clostridium difficile, toxins A (TcdA) and B (TcdB). Designated VNA2-Tcd, this agent has subnanomolar toxin neutralization potencies for both C. difficile toxins in cell assays. When given systemically by parenteral administration, VNA2-Tcd protected against CDI in gnotobiotic piglets and mice and to a lesser extent in hamsters. Protection from CDI was also observed in gnotobiotic piglets treated by gene therapy with an adenovirus that promoted the expression of VNA2-Tcd.
Collapse
|
44
|
Vance DJ, Mantis NJ. Progress and challenges associated with the development of ricin toxin subunit vaccines. Expert Rev Vaccines 2016; 15:1213-22. [PMID: 26998662 PMCID: PMC5193006 DOI: 10.1586/14760584.2016.1168701] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The past several years have seen major advances in the development of a safe and efficacious ricin toxin vaccine, including the completion of two Phase I clinical trials with two different recombinant A subunit (RTA)-based vaccines: RiVax™ and RVEc™ adsorbed to aluminum salt adjuvant, as well as a non-human primate study demonstrating that parenteral immunization with RiVax elicits a serum antibody response that was sufficient to protect against a lethal dose aerosolized ricin exposure. One of the major obstacles moving forward is assessing vaccine efficacy in humans, when neither ricin-specific serum IgG endpoint titers nor toxin-neutralizing antibody levels are accepted as definitive predictors of protective immunity. In this review we summarize ongoing efforts to leverage recent advances in our understanding of RTA-antibody interactions at the structural level to develop novel assays to predict vaccine efficacy in humans.
Collapse
Affiliation(s)
- David J. Vance
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Nicholas J. Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Biomedical Sciences, University at Albany, Albany, New York, USA
| |
Collapse
|
45
|
Vrentas CE, Moayeri M, Keefer AB, Greaney AJ, Tremblay J, O'Mard D, Leppla SH, Shoemaker CB. A Diverse Set of Single-domain Antibodies (VHHs) against the Anthrax Toxin Lethal and Edema Factors Provides a Basis for Construction of a Bispecific Agent That Protects against Anthrax Infection. J Biol Chem 2016; 291:21596-21606. [PMID: 27539858 DOI: 10.1074/jbc.m116.749184] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/08/2016] [Indexed: 01/08/2023] Open
Abstract
Infection with Bacillus anthracis, the causative agent of anthrax, can lead to persistence of lethal secreted toxins in the bloodstream, even after antibiotic treatment. VHH single-domain antibodies have been demonstrated to neutralize diverse bacterial toxins both in vitro and in vivo, with protein properties such as small size and high stability that make them attractive therapeutic candidates. Recently, we reported on VHHs with in vivo activity against the protective antigen component of the anthrax toxins. Here, we characterized a new set of 15 VHHs against the anthrax toxins that act by binding to the edema factor (EF) and/or lethal factor (LF) components. Six of these VHHs are cross-reactive against both EF and LF and recognize the N-terminal domain (LFN, EFN) of their target(s) with subnanomolar affinity. The cross-reactive VHHs block binding of EF/LF to the protective antigen C-terminal binding interface, preventing toxin entry into the cell. Another VHH appears to recognize the LF C-terminal domain and exhibits a kinetic effect on substrate cleavage by LF. A subset of the VHHs neutralized against EF and/or LF in murine macrophage assays, and the neutralizing VHHs that were tested improved survival of mice in a spore model of anthrax infection. Finally, a bispecific VNA (VHH-based neutralizing agent) consisting of two linked toxin-neutralizing VHHs, JMN-D10 and JMO-G1, was fully protective against lethal anthrax spore infection in mice as a single dose. This set of VHHs should facilitate development of new therapeutic VNAs and/or diagnostic agents for anthrax.
Collapse
Affiliation(s)
- Catherine E Vrentas
- From the Department of Biology, Frostburg State University, Frostburg, Maryland 50010.,Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Mahtab Moayeri
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Andrea B Keefer
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Allison J Greaney
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Jacqueline Tremblay
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Maryland 01536
| | - Danielle O'Mard
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Stephen H Leppla
- Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Charles B Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Maryland 01536
| |
Collapse
|
46
|
Steeland S, Vandenbroucke RE, Libert C. Nanobodies as therapeutics: big opportunities for small antibodies. Drug Discov Today 2016; 21:1076-113. [DOI: 10.1016/j.drudis.2016.04.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/26/2016] [Accepted: 04/04/2016] [Indexed: 12/28/2022]
|
47
|
Rudolph MJ, Vance DJ, Cassidy MS, Rong Y, Shoemaker CB, Mantis NJ. Structural analysis of nested neutralizing and non-neutralizing B cell epitopes on ricin toxin's enzymatic subunit. Proteins 2016; 84:1162-72. [PMID: 27159829 DOI: 10.1002/prot.25062] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/31/2016] [Accepted: 04/28/2016] [Indexed: 11/10/2022]
Abstract
In this report, we describe the X-ray crystal structures of two single domain camelid antibodies (VH H), F5 and F8, each in complex with ricin toxin's enzymatic subunit (RTA). F5 has potent toxin-neutralizing activity, while F8 has weak neutralizing activity. F5 buried a total of 1760 Å(2) in complex with RTA and made contact with three prominent secondary structural elements: α-helix B (Residues 98-106), β-strand h (Residues 113-117), and the C-terminus of α-helix D (Residues 154-156). F8 buried 1103 Å(2) in complex with RTA that was centered primarily on β-strand h. As such, the structural epitope of F8 is essentially nested within that of F5. All three of the F5 complementarity determining regions CDRs were involved in RTA contact, whereas F8 interactions were almost entirely mediated by CDR3, which essentially formed a seventh β-strand within RTA's centrally located β-sheet. A comparison of the two structures reported here to several previously reported (RTA-VH H) structures identifies putative contact sites on RTA, particularly α-helix B, associated with potent toxin-neutralizing activity. This information has implications for rational design of RTA-based subunit vaccines for biodefense. Proteins 2016; 84:1162-1172. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - David J Vance
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, 12208
| | | | - Yinghui Rong
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, 12208
| | - Charles B Shoemaker
- Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, 01536
| | - Nicholas J Mantis
- Division of Infectious Diseases, New York State Department of Health, Wadsworth Center, Albany, New York, 12208.,Department of Biomedical Sciences, University at Albany, Albany, New York, 12201
| |
Collapse
|
48
|
Herrera C, Klokk TI, Cole R, Sandvig K, Mantis NJ. A Bispecific Antibody Promotes Aggregation of Ricin Toxin on Cell Surfaces and Alters Dynamics of Toxin Internalization and Trafficking. PLoS One 2016; 11:e0156893. [PMID: 27300140 PMCID: PMC4907443 DOI: 10.1371/journal.pone.0156893] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 05/21/2016] [Indexed: 11/19/2022] Open
Abstract
JJX12 is an engineered bispecific antibody against ricin, a member of the medically important A-B family of toxins that exploits retrograde transport as means to gain entry into the cytosol of target cells. JJX12 consists of RTA-D10, a camelid single variable domain (VHH) antibody directed against an epitope on ricin's enzymatic subunit (RTA), linked via a 15-mer peptide to RTB-B7, a VHH against ricin's bivalent galactose binding subunit (RTB). We previously reported that JJX12, but not an equimolar mixture of RTA-D10 and RTB-B7 monomers, was able to passively protect mice against a lethal dose ricin challenge, demonstrating that physically linking RTB-B7 and RTA-D10 is critical for toxin-neutralizing activity in vivo. We also reported that JJX12 promotes aggregation of ricin in solution, presumably through the formation of intermolecular crosslinking. In the current study, we now present evidence that JJX12 affects the dynamics of ricin uptake and trafficking in human epithelial cells. Confocal microscopy, as well as live cell imaging coupled with endocytosis pathway-specific inhibitors, revealed that JJX12-toxin complexes are formed on the surfaces of mammalian cells and internalized via a pathway sensitive to amiloride, a known inhibitor of macropinocytosis. Moreover, in the presence of JJX12, retrograde transport of ricin to the trans-Golgi network was significantly reduced, while accumulation of the toxin in late endosomes was significantly enhanced. In summary, we propose that JJX12, by virtue of its ability to crosslink ricin toxin, alters the route of toxin uptake and trafficking within cells.
Collapse
Affiliation(s)
- Cristina Herrera
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York, United States of America
| | - Tove Irene Klokk
- Department of Molecular Cell Biology and Centre for Cancer Biomedicine, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Richard Cole
- Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York, United States of America
- Division of Translational Medicine, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - Kirsten Sandvig
- Department of Molecular Cell Biology and Centre for Cancer Biomedicine, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Nicholas J. Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biomedical Sciences, University at Albany School of Public Health, Albany, New York, United States of America
| |
Collapse
|
49
|
Mejías MP, Hiriart Y, Lauché C, Fernández-Brando RJ, Pardo R, Bruballa A, Ramos MV, Goldbaum FA, Palermo MS, Zylberman V. Development of camelid single chain antibodies against Shiga toxin type 2 (Stx2) with therapeutic potential against Hemolytic Uremic Syndrome (HUS). Sci Rep 2016; 6:24913. [PMID: 27118524 PMCID: PMC4847011 DOI: 10.1038/srep24913] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/07/2016] [Indexed: 12/12/2022] Open
Abstract
Shiga toxin (Stx)-producing Escherichia coli (STEC) infections are implicated in the development of the life-threatening Hemolytic Uremic Syndrome (HUS). Despite the magnitude of the social and economic problems caused by STEC infections, no licensed vaccine or effective therapy is presently available for human use. Single chain antibodies (VHH) produced by camelids exhibit several advantages in comparison with conventional antibodies, making them promising tools for diagnosis and therapy. In the present work, the properties of a recently developed immunogen, which induces high affinity and protective antibodies against Stx type 2 (Stx2), were exploited to develop VHHs with therapeutic potential against HUS. We identified a family of VHHs against the B subunit of Stx2 (Stx2B) that neutralize Stx2 in vitro at subnanomolar concentrations. One VHH was selected and was engineered into a trivalent molecule (two copies of anti-Stx2B VHH and one anti-seroalbumin VHH). The resulting molecule presented extended in vivo half-life and high therapeutic activity, as demonstrated in three different mouse models of Stx2-toxicity: a single i.v. lethal dose of Stx2, several i.v. incremental doses of Stx2 and intragastrical STEC infection. This simple antitoxin agent should offer new therapeutic options for treating STEC infections to prevent or ameliorate HUS outcome.
Collapse
Affiliation(s)
- Maria P Mejías
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), P. De Melo 3081, Ciudad de Buenos Aires, (C1425AUM), Argentina
| | - Yanina Hiriart
- INMUNOVA S.A., Av. Patricias Argentinas 435 - Ciudad de Buenos Aires, (C1405BWE), Argentina.,Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires-CONICET, Av. Patricias Argentinas 435 - Ciudad de Buenos Aires. (C1405BWE), Argentina
| | - Constanza Lauché
- INMUNOVA S.A., Av. Patricias Argentinas 435 - Ciudad de Buenos Aires, (C1405BWE), Argentina.,Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires-CONICET, Av. Patricias Argentinas 435 - Ciudad de Buenos Aires. (C1405BWE), Argentina
| | - Romina J Fernández-Brando
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), P. De Melo 3081, Ciudad de Buenos Aires, (C1425AUM), Argentina
| | - Romina Pardo
- INMUNOVA S.A., Av. Patricias Argentinas 435 - Ciudad de Buenos Aires, (C1405BWE), Argentina.,Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires-CONICET, Av. Patricias Argentinas 435 - Ciudad de Buenos Aires. (C1405BWE), Argentina
| | - Andrea Bruballa
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), P. De Melo 3081, Ciudad de Buenos Aires, (C1425AUM), Argentina
| | - María V Ramos
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), P. De Melo 3081, Ciudad de Buenos Aires, (C1425AUM), Argentina
| | - Fernando A Goldbaum
- INMUNOVA S.A., Av. Patricias Argentinas 435 - Ciudad de Buenos Aires, (C1405BWE), Argentina.,Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires-CONICET, Av. Patricias Argentinas 435 - Ciudad de Buenos Aires. (C1405BWE), Argentina
| | - Marina S Palermo
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, (IMEX), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), P. De Melo 3081, Ciudad de Buenos Aires, (C1425AUM), Argentina
| | - Vanesa Zylberman
- INMUNOVA S.A., Av. Patricias Argentinas 435 - Ciudad de Buenos Aires, (C1405BWE), Argentina.,Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires-CONICET, Av. Patricias Argentinas 435 - Ciudad de Buenos Aires. (C1405BWE), Argentina
| |
Collapse
|
50
|
Adenoviral Expression of a Bispecific VHH-Based Neutralizing Agent That Targets Protective Antigen Provides Prophylactic Protection from Anthrax in Mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:213-8. [PMID: 26740390 DOI: 10.1128/cvi.00611-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 12/27/2015] [Indexed: 01/01/2023]
Abstract
Bacillus anthracis, the causative agent of anthrax, secretes three polypeptides, which form the bipartite lethal and edema toxins (LT and ET, respectively). The common component in these toxins, protective antigen (PA), is responsible for binding to cellular receptors and translocating the lethal factor (LF) and edema factor (EF) enzymatic moieties to the cytosol. Antibodies against PA protect against anthrax. We previously isolated toxin-neutralizing variable domains of camelid heavy-chain-only antibodies (VHHs) and demonstrated their in vivo efficacy. In this work, gene therapy with an adenoviral (Ad) vector (Ad/VNA2-PA) (VNA, VHH-based neutralizing agents) promoting the expression of a bispecific VHH-based neutralizing agent (VNA2-PA), consisting of two linked VHHs targeting different PA-neutralizing epitopes, was tested in two inbred mouse strains, BALB/cJ and C57BL/6J, and found to protect mice against anthrax toxin challenge and anthrax spore infection. Two weeks after a single treatment with Ad/VNA2-PA, serum VNA2-PA levels remained above 1 μg/ml, with some as high as 10 mg/ml. The levels were 10- to 100-fold higher and persisted longer in C57BL/6J than in BALB/cJ mice. Mice were challenged with a lethal dose of LT or spores at various times after Ad/VNA2-PA administration. The majority of BALB/cJ mice having serum VNA2-PA levels of >0.1 μg/ml survived LT challenge, and 9 of 10 C57BL/6J mice with serum levels of >1 μg/ml survived spore challenge. Our findings demonstrate the potential for genetic delivery of VNAs as an effective method for providing prophylactic protection from anthrax. We also extend prior findings of mouse strain-based differences in transgene expression and persistence by adenoviral vectors.
Collapse
|