Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters








Publication year range
1.
Structure ; 32(9): 1404-1418.e7, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39146931

ABSTRACT

Immunoglobulin G (IgG) antibodies that bind their cognate antigen in a pH-dependent manner (acid-switched antibodies) can release their bound antigen for degradation in the acidic environment of endosomes, while the IgGs are rescued by the neonatal Fc receptor (FcRn). Thus, such IgGs can neutralize multiple antigens over time and therefore be used at lower doses than their non-pH-responsive counterparts. Here, we show that light-chain shuffling combined with phage display technology can be used to discover IgG1 antibodies with increased pH-dependent antigen binding properties, using the snake venom toxins, myotoxin II and α-cobratoxin, as examples. We reveal differences in how the selected IgG1s engage their antigens and human FcRn and show how these differences translate into distinct cellular handling properties related to their pH-dependent antigen binding phenotypes and Fc-engineering for improved FcRn binding. Our study showcases the complexity of engineering pH-dependent antigen binding IgG1s and demonstrates the effects on cellular antibody-antigen recycling.


Subject(s)
Histocompatibility Antigens Class I , Immunoglobulin G , Receptors, Fc , Hydrogen-Ion Concentration , Immunoglobulin G/metabolism , Immunoglobulin G/chemistry , Humans , Receptors, Fc/metabolism , Receptors, Fc/chemistry , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Protein Engineering/methods , Protein Binding , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light Chains/genetics , Antigens/metabolism , Antigens/chemistry , Animals , Models, Molecular
2.
Bioinform Adv ; 4(1): vbae020, 2024.
Article in English | MEDLINE | ID: mdl-38425781

ABSTRACT

Summary: High-throughput sequencing (HTS) offers a modern, fast, and explorative solution to unveil the full potential of display techniques, like antibody phage display, in molecular biology. However, a significant challenge lies in the processing and analysis of such data. Furthermore, there is a notable absence of open-access user-friendly software tools that can be utilized by scientists lacking programming expertise. Here, we present ExpoSeq as an easy-to-use tool to explore, process, and visualize HTS data from antibody discovery campaigns like an expert while only requiring a beginner's knowledge. Availability and implementation: The pipeline is distributed via GitHub and PyPI, and it can either be installed as a package with pip or the user can choose to clone the repository.

3.
Nat Commun ; 15(1): 173, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38228619

ABSTRACT

Improved therapies are needed against snakebite envenoming, which kills and permanently disables thousands of people each year. Recently developed neutralizing monoclonal antibodies against several snake toxins have shown promise in preclinical rodent models. Here, we use phage display technology to discover a human monoclonal antibody and show that this antibody causes antibody-dependent enhancement of toxicity (ADET) of myotoxin II from the venomous pit viper, Bothrops asper, in a mouse model of envenoming that mimics a snakebite. While clinical ADET related to snake venom has not yet been reported in humans, this report of ADET of a toxin from the animal kingdom highlights the necessity of assessing even well-known antibody formats in representative preclinical models to evaluate their therapeutic utility against toxins or venoms. This is essential to avoid potential deleterious effects as exemplified in the present study.


Subject(s)
Bothrops , Neurotoxins , Mice , Animals , Humans , Neurotoxins/toxicity , Bothrops asper , Antibody-Dependent Enhancement , Antibodies, Monoclonal/toxicity
4.
Toxicon ; 238: 107559, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38113945

ABSTRACT

Protein structure determination is a critical aspect of biological research, enabling us to understand protein function and potential applications. Recent advances in deep learning and artificial intelligence have led to the development of several protein structure prediction tools, such as AlphaFold2 and ColabFold. However, their performance has primarily been evaluated on well-characterised proteins and their ability to predict sturtctures of proteins lacking experimental structures, such as many snake venom toxins, has been less scrutinised. In this study, we evaluated three modelling tools on their prediction of over 1000 snake venom toxin structures for which no experimental structures exist. Our findings show that AlphaFold2 (AF2) performed the best across all assessed parameters. We also observed that ColabFold (CF) only scored slightly worse than AF2, while being computationally less intensive. All tools struggled with regions of intrinsic disorder, such as loops and propeptide regions, and performed well in predicting the structure of functional domains. Overall, our study highlights the importance of exercising caution when working with proteins with no experimental structures available, particularly those that are large and contain flexible regions. Nonetheless, leveraging computational structure prediction tools can provide valuable insights into the modelling of protein interactions with different targets and reveal potential binding sites, active sites, and conformational changes, as well as into the design of potential molecular binders for reagent, diagnostic, or therapeutic purposes.


Subject(s)
Artificial Intelligence , Snake Venoms , Binding Sites , Furylfuramide , Proteins/chemistry , Snake Venoms/chemistry
5.
Toxicon ; 234: 107307, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37783315

ABSTRACT

Despite the considerable global impact of snakebite envenoming, available treatments remain suboptimal. Here, we report the discovery of a broadly-neutralizing human monoclonal antibody, using a phage display-based cross-panning strategy, capable of reducing the cytotoxic effects of venom phospholipase A2s from three different snake genera from different continents. This highlights the potential of utilizing monoclonal antibodies to develop more effective, safer, and globally accessible polyvalent antivenoms that can be widely used to treat snakebite envenoming.


Subject(s)
Snake Bites , Animals , Humans , Venoms , Antibodies, Monoclonal , Antivenins/pharmacology , Snakes , Phospholipases A2 , Snake Venoms
6.
Sci Rep ; 13(1): 10181, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37349546

ABSTRACT

Antibodies with cross-reactive binding and broad toxin-neutralizing capabilities are advantageous for treating indications such as infectious diseases and animal envenomings. Such antibodies have been successfully selected against closely related antigens using phage display technology. However, the mechanisms driving antibody cross-reactivity typically remain to be elucidated. Therefore, we sought to explore how a previously reported phage display-based cross-panning strategy drives the selection of cross-reactive antibodies using seven different snake toxins belonging to three protein (sub-)families: phospholipases A2, long-chain α-neurotoxins, and short-chain α-neurotoxins. We showcase how cross-panning can increase the chances of discovering cross-reactive single-chain variable fragments (scFvs) from phage display campaigns. Further, we find that the feasibility of discovering cross-reactive antibodies using cross-panning cannot easily be predicted by analyzing the sequence, structural, or surface similarity of the antigens alone. However, when antigens share the (exact) same functions, this seems to increase the chances of selecting cross-reactive antibodies, which may possibly be due to the existence of structurally similar motifs on the antigens.


Subject(s)
Bacteriophages , Single-Chain Antibodies , Animals , Peptide Library , Neurotoxins , Antigens , Bacteriophages/genetics , Snake Venoms
7.
Drug Discov Today ; 27(8): 2151-2169, 2022 08.
Article in English | MEDLINE | ID: mdl-35550436

ABSTRACT

Phage display technology can be used for the discovery of antibodies for research, diagnostic, and therapeutic purposes. In this review, we present and discuss key parameters that can be optimized when performing phage display selection campaigns, including the use of different antibody formats and advanced strategies for antigen presentation, such as immobilization, liposomes, nanodiscs, virus-like particles, and whole cells. Furthermore, we provide insights into selection strategies that can be used for the discovery of antibodies with complex binding requirements, such as targeting a specific epitope, cross-reactivity, or pH-dependent binding. Lastly, we provide a description of specialized phage display libraries for the discovery of bispecific antibodies and pH-sensitive antibodies. Together, these methods can be used to improve antibody discovery campaigns against all types of antigens.


Subject(s)
Bacteriophages , Peptide Library , Antibodies , Bacteriophages/genetics , Epitopes , Technology
8.
Front Bioeng Biotechnol ; 9: 811905, 2021.
Article in English | MEDLINE | ID: mdl-35127675

ABSTRACT

Animal venoms are complex mixtures containing peptides and proteins known as toxins, which are responsible for the deleterious effect of envenomations. Across the animal Kingdom, toxin diversity is enormous, and the ability to understand the biochemical mechanisms governing toxicity is not only relevant for the development of better envenomation therapies, but also for exploiting toxin bioactivities for therapeutic or biotechnological purposes. Most of toxinology research has relied on obtaining the toxins from crude venoms; however, some toxins are difficult to obtain because the venomous animal is endangered, does not thrive in captivity, produces only a small amount of venom, is difficult to milk, or only produces low amounts of the toxin of interest. Heterologous expression of toxins enables the production of sufficient amounts to unlock the biotechnological potential of these bioactive proteins. Moreover, heterologous expression ensures homogeneity, avoids cross-contamination with other venom components, and circumvents the use of crude venom. Heterologous expression is also not only restricted to natural toxins, but allows for the design of toxins with special properties or can take advantage of the increasing amount of transcriptomics and genomics data, enabling the expression of dormant toxin genes. The main challenge when producing toxins is obtaining properly folded proteins with a correct disulfide pattern that ensures the activity of the toxin of interest. This review presents the strategies that can be used to express toxins in bacteria, yeast, insect cells, or mammalian cells, as well as synthetic approaches that do not involve cells, such as cell-free biosynthesis and peptide synthesis. This is accompanied by an overview of the main advantages and drawbacks of these different systems for producing toxins, as well as a discussion of the biosafety considerations that need to be made when working with highly bioactive proteins.

9.
Front Immunol ; 11: 587825, 2020.
Article in English | MEDLINE | ID: mdl-33262768

ABSTRACT

Widow spiders are among the few spider species worldwide that can cause serious envenoming in humans. The clinical syndrome resulting from Latrodectus spp. envenoming is called latrodectism and characterized by pain (local or regional) associated with diaphoresis and nonspecific systemic effects. The syndrome is caused by α-latrotoxin, a ~130 kDa neurotoxin that induces massive neurotransmitter release. Due to this function, α-latrotoxin has played a fundamental role as a tool in the study of neuroexocytosis. Nevertheless, some questions concerning its mode of action remain unresolved today. The diagnosis of latrodectism is purely clinical, combined with the patient's history of spider bite, as no analytical assays exist to detect widow spider venom. By utilizing antibody phage display technology, we here report the discovery of the first recombinant human monoclonal immunoglobulin G antibody (TPL0020_02_G9) that binds α-latrotoxin from the Mediterranean black widow spider (Latrodectus tredecimguttatus) and show neutralization efficacy ex vivo. Such antibody can be used as an affinity reagent for research and diagnostic purposes, providing researchers with a novel tool for more sophisticated experimentation and analysis. Moreover, it may also find therapeutic application in future.


Subject(s)
Antibodies, Monoclonal , Black Widow Spider/immunology , Immunoglobulin G , Spider Venoms , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Male , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats, Wistar , Spider Venoms/immunology , Spider Venoms/toxicity
10.
Sci Rep ; 10(1): 10765, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612183

ABSTRACT

Broadly-neutralizing monoclonal antibodies are of high therapeutic utility against infectious diseases caused by bacteria and viruses, as well as different types of intoxications. Snakebite envenoming is one such debilitating pathology, which is currently treated with polyclonal antibodies derived from immunized animals. For the development of novel envenoming therapies based on monoclonal antibodies with improved therapeutic benefits, new discovery approaches for broadly-neutralizing antibodies are needed. Here, we present a methodology based on phage display technology and a cross-panning strategy that enables the selection of cross-reactive monoclonal antibodies that can broadly neutralize toxins from different snake species. This simple in vitro methodology is immediately useful for the development of broadly-neutralizing (polyvalent) recombinant antivenoms with broad species coverage, but may also find application in the development of broadly-neutralizing antibodies against bacterial, viral, and parasitic agents that are known for evading therapy via resistance mechanisms and antigen variation.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antivenins/chemistry , Neutralization Tests , Animals , Antigens/chemistry , Biotechnology , Cross Reactions/immunology , Drug Design , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fab Fragments/immunology , In Vitro Techniques , Inhibitory Concentration 50 , Keratinocytes/immunology , Mass Spectrometry , Peptide Library , Proteomics , Recombinant Proteins/chemistry , Snakes , Venoms
11.
Front Pharmacol ; 11: 611, 2020.
Article in English | MEDLINE | ID: mdl-32457615

ABSTRACT

Toxin synergism is a complex biochemical phenomenon, where different animal venom proteins interact either directly or indirectly to potentiate toxicity to a level that is above the sum of the toxicities of the individual toxins. This provides the animals possessing venoms with synergistically enhanced toxicity with a metabolic advantage, since less venom is needed to inflict potent toxic effects in prey and predators. Among the toxins that are known for interacting synergistically are cytotoxins from snake venoms, phospholipases A2 from snake and bee venoms, and melittin from bee venom. These toxins may derive a synergistically enhanced toxicity via formation of toxin complexes by hetero-oligomerization. Using a human keratinocyte assay mimicking human epidermis in vitro, we demonstrate and quantify the level of synergistically enhanced toxicity for 12 cytotoxin/melittin-PLA2 combinations using toxins from elapids, vipers, and bees. Moreover, by utilizing an interaction-based assay and by including a wealth of information obtained via a thorough literature review, we speculate and propose a mechanistic model for how toxin synergism in relation to cytotoxicity may be mediated by cytotoxin/melittin and PLA2 complex formation.

12.
Toxins (Basel) ; 12(4)2020 04 09.
Article in English | MEDLINE | ID: mdl-32283690

ABSTRACT

Antibiotics are often administered with antivenom following snakebite envenomings in order to avoid secondary bacterial infections. However, to this date, no studies have evaluated whether antibiotics may have undesirable potentiating effects on snake venom. Herein, we demonstrate that four commonly used antibiotics affect the enzymatic activities of proteolytic snake venom toxins in two different in vitro assays. Similar findings in vivo could have clinical implications for snakebite management and require further examination.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fibrinogen/metabolism , Fibrinolysis/drug effects , Serine Proteases/metabolism , Snake Venoms/enzymology , Ampicillin/pharmacology , Cloxacillin/pharmacology , Kanamycin/pharmacology
13.
Front Immunol ; 10: 2090, 2019.
Article in English | MEDLINE | ID: mdl-31552038

ABSTRACT

Honey bees can be found all around the world and fulfill key pollination roles within their natural ecosystems, as well as in agriculture. Most species are typically docile, and most interactions between humans and bees are unproblematic, despite their ability to inject a complex venom into their victims as a defensive mechanism. Nevertheless, incidences of bee stings have been on the rise since the accidental release of Africanized bees to Brazil in 1956 and their subsequent spread across the Americas. These bee hybrids are more aggressive and are prone to attack, presenting a significant healthcare burden to the countries they have colonized. To date, treatment of such stings typically focuses on controlling potential allergic reactions, as no specific antivenoms against bee venom currently exist. Researchers have investigated the possibility of developing bee antivenoms, but this has been complicated by the very low immunogenicity of the key bee toxins, which fail to induce a strong antibody response in the immunized animals. However, with current cutting-edge technologies, such as phage display, alongside the rise of monoclonal antibody therapeutics, the development of a recombinant bee antivenom is achievable, and promising results towards this goal have been reported in recent years. Here, current knowledge on the venom biology of Africanized bees and current treatment options against bee envenoming are reviewed. Additionally, recent developments within next-generation bee antivenoms are presented and discussed.


Subject(s)
Bee Venoms , Bees , Insect Bites and Stings/immunology , Insect Bites and Stings/therapy , Americas/epidemiology , Animals , Bees/classification , Bees/physiology , Behavior, Animal , Disease Management , History, 20th Century , History, 21st Century , Hypersensitivity/epidemiology , Hypersensitivity/immunology , Hypersensitivity/therapy , Insect Bites and Stings/epidemiology , Insect Bites and Stings/history , Public Health Surveillance
SELECTION OF CITATIONS
SEARCH DETAIL