Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Biol Chem ; 299(9): 104927, 2023 09.
Article in English | MEDLINE | ID: mdl-37330175

ABSTRACT

Methicillin-resistant Staphylococcus aureus, or MRSA, is one of the major causative agents of hospital-acquired infections worldwide. Novel antimicrobial strategies efficient against antibiotic-resistant strains are necessary and not only against S. aureus. Among those, strategies that aim at blocking or dismantling proteins involved in the acquisition of essential nutrients, helping the bacteria to colonize the host, are intensively studied. A major route for S. aureus to acquire iron from the host organism is the Isd (iron surface determinant) system. In particular, the hemoglobin receptors IsdH and IsdB located on the surface of the bacterium are necessary to acquire the heme moiety containing iron, making them a plausible antibacterial target. Herein, we obtained an antibody of camelid origin that blocked heme acquisition. We determined that the antibody recognized the heme-binding pocket of both IsdH and IsdB with nanomolar order affinity through its second and third complementary-determining regions. The mechanism explaining the inhibition of acquisition of heme in vitro could be described as a competitive process in which the complementary-determining region 3 from the antibody blocked the acquisition of heme by the bacterial receptor. Moreover, this antibody markedly reduced the growth of three different pathogenic strains of MRSA. Collectively, our results highlight a mechanism for inhibiting nutrient uptake as an antibacterial strategy against MRSA.


Subject(s)
Antibodies, Bacterial , Methicillin-Resistant Staphylococcus aureus , Receptors, Cell Surface , Single-Domain Antibodies , Humans , Anti-Bacterial Agents/pharmacology , Heme/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/therapeutic use , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology , Staphylococcal Infections/drug therapy , Antigens, Bacterial/immunology , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Camelids, New World , Animals , Protein Binding/drug effects , Models, Molecular , Molecular Dynamics Simulation
2.
Int J Mol Sci ; 23(6)2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35328351

ABSTRACT

Nanobodies provide important advantages over traditional antibodies, including their smaller size and robust biochemical properties such as high thermal stability, high solubility, and the ability to be bioengineered into novel multivalent, multi-specific, and high-affinity molecules, making them a class of emerging powerful therapies against SARS-CoV-2. Recent research efforts on the design, protein engineering, and structure-functional characterization of nanobodies and their binding with SARS-CoV-2 S proteins reflected a growing realization that nanobody combinations can exploit distinct binding epitopes and leverage the intrinsic plasticity of the conformational landscape for the SARS-CoV-2 S protein to produce efficient neutralizing and mutation resistant characteristics. Structural and computational studies have also been instrumental in quantifying the structure, dynamics, and energetics of the SARS-CoV-2 spike protein binding with nanobodies. In this review, a comprehensive analysis of the current structural, biophysical, and computational biology investigations of SARS-CoV-2 S proteins and their complexes with distinct classes of nanobodies targeting different binding sites is presented. The analysis of computational studies is supplemented by an in-depth examination of mutational scanning simulations and identification of binding energy hotspots for distinct nanobody classes. The review is focused on the analysis of mechanisms underlying synergistic binding of multivalent nanobodies that can be superior to single nanobodies and conventional nanobody cocktails in combating escape mutations by effectively leveraging binding avidity and allosteric cooperativity. We discuss how structural insights and protein engineering approaches together with computational biology tools can aid in the rational design of synergistic combinations that exhibit superior binding and neutralization characteristics owing to avidity-mediated mechanisms.


Subject(s)
Binding Sites , Molecular Docking Simulation , Molecular Dynamics Simulation , Single-Domain Antibodies/chemistry , Spike Glycoprotein, Coronavirus/chemistry , Amino Acids , Antibody Affinity , Epitopes/chemistry , Epitopes/metabolism , Humans , Multiprotein Complexes/chemistry , Mutagenesis , Protein Binding , Protein Engineering , Protein Interaction Domains and Motifs , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism
3.
J Biomol NMR ; 73(6-7): 375-384, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31073665

ABSTRACT

The insertase BamA is an essential protein of the bacterial outer membrane. Its 16-stranded transmembrane ß-barrel contains a lateral gate as a key functional element. This gate is formed by the C-terminal half of the last ß-strand. The BamA barrel was previously found to sample different conformations in aqueous solution, as well as different gate-open, gate-closed, and collapsed conformations in X-ray crystallography and cryo-electron microscopy structures. Here, we report the successful identification of conformation-selective nanobodies that stabilize BamA in specific conformations. While the initial candidate generation and selection protocol was based on established alpaca immunization and phage display selection procedures, the final selection of nanobodies was enhanced by a solution NMR-based screening step to shortlist the targets for crystallization. In this way, three crystal structures of BamA-nanobody complexes were efficiently obtained, showing two types of nanobodies that indeed stabilized BamA in two different conformations, i.e., with open and closed lateral gate, respectively. Then, by correlating the structural data with high resolution NMR spectra, we could for the first time assign the BamA conformational solution ensemble to defined structural states. The new nanobodies will be valuable tools towards understanding the client insertion mechanism of BamA and towards developing improved antibiotics.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Models, Molecular , Protein Conformation , Single-Domain Antibodies/chemistry , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Single-Domain Antibodies/pharmacology , Solutions
4.
Methods Mol Biol ; 1836: 139-158, 2018.
Article in English | MEDLINE | ID: mdl-30151572

ABSTRACT

Our understanding of infection biology is based on experiments in which pathogen or host proteins are perturbed by small compound inhibitors, mutation, or depletion. This approach has been remarkably successful, as, for example, demonstrated by the independent identification of the endosomal membrane protein Niemann-Pick C1 as an essential factor for Ebola virus infection in both small compound and insertional mutagenesis screens (Côté, Nature 477:344-348, 2011; Carette et al., Nature 477:340-343, 2011). However, many aspects of host-pathogen interactions are poorly understood because we cannot target all of the involved molecules with small molecules, or because we cannot deplete essential proteins. Single domain antibody fragments expressed in the cytosol or other organelles constitute a versatile alternative to perturb the function of any given protein by masking protein-protein interaction interfaces, by stabilizing distinct conformations, or by directly interfering with enzymatic activities. The variable domains of heavy chain-only antibodies (VHHs) from camelid species can be cloned from blood samples of animals immunized with the desired target molecules. We can thus exploit the ability of the camelid immune system to generate affinity-matured single domain antibody fragments to obtain highly specific tools. Interesting VHH candidates are typically identified based on their affinity toward immobilized antigens using techniques such as phage display.The phenotypical screening approach described here allows the direct identification of VHHs that prevent infection of cells with influenza A virus (IAV) or other pathogens. The VHH repertoire is cloned into a lentiviral vector, which is used to generate pseudo-typed lentivirus particles. Target cells are transduced with the lentivirus, so that every cell inducibly expresses a different VHH. This cell collection is then challenged with a lethal dose of virus. Only the cells which express a VHH that prevents infection by targeting virus proteins or host cell components essential for infection will survive. We can thus identify critical target molecules including vulnerable epitopes and conformations, render target molecules accessible to informative perturbation studies, and stabilize intermediates of virus entry for detailed analysis.


Subject(s)
Anti-Retroviral Agents/pharmacology , Lentivirus/drug effects , Phenotype , Single-Domain Antibodies/pharmacology , Amino Acid Sequence , Cell Line , Drug Evaluation, Preclinical/methods , Gene Library , Genetic Vectors/genetics , Humans , Influenza A virus/genetics , Lentivirus/genetics , Lentivirus Infections/drug therapy , Lentivirus Infections/virology , Microbial Sensitivity Tests , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics
5.
BioDrugs ; 32(5): 441-464, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30132211

ABSTRACT

Bispecific antibodies have moved from being an academic curiosity with therapeutic promise to reality, with two molecules being currently commercialized (Hemlibra® and Blincyto®) and many more in clinical trials. The success of bispecific antibodies is mainly due to the continuously growing number of mechanisms of actions (MOA) they enable that are not accessible to monoclonal antibodies. One of the earliest MOA of bispecific antibodies and currently the one with the largest number of clinical trials is the redirecting of the cytotoxic activity of T-cells for oncology applications, now extending its use in infective diseases. The use of bispecific antibodies for crossing the blood-brain barrier is another important application because of its potential to advance the therapeutic options for neurological diseases. Another noteworthy application due to its growing trend is enabling a more tissue-specific delivery or activity of antibodies. The different molecular solutions to the initial hurdles that limited the development of bispecific antibodies have led to the current diverse set of bispecific or multispecific antibody formats that can be grouped into three main categories: IgG-like formats, antibody fragment-based formats, or appended IgG formats. The expanded applications of bispecific antibodies come at the price of additional challenges for clinical development. The rising complexity in their structure may increase the risk of immunogenicity and the multiple antigen specificity complicates the selection of relevant species for safety assessment.


Subject(s)
Antibodies, Bispecific/chemistry , Antibodies, Bispecific/pharmacology , Biological Products/pharmacology , Protein Engineering/methods , Animals , Antibodies, Bispecific/pharmacokinetics , Biological Products/immunology , Cell Membrane/metabolism , Drug Evaluation, Preclinical/methods , Humans , Immunoglobulin G/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Single-Chain Antibodies/chemistry , Single-Domain Antibodies/chemistry
6.
Bioorg Med Chem Lett ; 27(21): 4844-4848, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28974337

ABSTRACT

Survivin, an inhibitor of the apoptosis protein family, is a potent tumor marker for diagnosis and prognosis. The enzyme-linked immunosorbent assay (ELISA) is one of the methods that has been used for detection of survivin. However, ELISA has several disadvantages caused by the use of conventional antibodies, and we have therefore been trying to develop a novel ELISA system using camelid single-domain antibodies (VHHs) as advantageous replacements. Here we report a supplemental approach to improve the VHH-polyclonal antibody sandwich ELISA for survivin detection. Iodoacetyl-functionalized pullulan was synthesized, and its thiol reactivity was characterized by a model reaction with l-cysteine. The thiophilic pullulan was applied to an immunoassay asan additive upon coating of standard assay plates with an anti-survivin VHH fusion protein with C-terminal cysteine. The results showed that the mole ratio of the additive to VHH had a significant effect on the consequent response. Mole ratios of 0.07, 0.7, and 7 led to 90% lower, 15% higher, and 69% lower responses, respectively, than the response of a positive control in which no additive was used. The background levels observed in any additive conditions were as low as that of a negative control lacking both VHH and the additive. These results indicate the applicability of the thiol-reactive pullulan as a response enhancer to VHH-based ELISA.


Subject(s)
Antibodies/immunology , Enzyme-Linked Immunosorbent Assay , Glucans/chemistry , Inhibitor of Apoptosis Proteins/analysis , Single-Domain Antibodies/immunology , Animals , Antibodies/chemistry , Cysteine/chemistry , Humans , Iodoacetic Acid/chemistry , Single-Domain Antibodies/chemistry , Survivin
7.
Sci Rep ; 6: 32256, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27558409

ABSTRACT

Candida albicans (C. albicans) is an important human commensal and opportunistic fungal pathogen. Secreted aspartyl proteinases (Saps) are a major virulence trait of C. albicans, and among these proteases Sap2 has the highest expression levels. It is possible that antibodies against Sap2 could provide an antifungal effect. In this study, two phages displaying anti-rSap2 single chain variable fragments (scFvs) were screened from human single fold scFv libraries, and their potential therapeutic roles were evaluated using a murine model infected by C. albicans. The in vivo efficacies were assessed by mortality rates, fungal burden and histological examination. Overall survival rates were significantly increased while the colony counts and infectious foci were significantly decreased after treatment with the scFv-phages relative to the control groups. In order to investigate the immune response provoked by scFv-phages, three kinds of cytokines (Th1, Th2 and Th17 types) were measured and a clear immune response was observed. These findings suggest that anti-rSap2 scFv-phages have potential in the therapy of systemic infection caused by C. albicans.


Subject(s)
Antibodies, Fungal/pharmacology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Candida albicans/immunology , Candidiasis/drug therapy , Fungal Proteins/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Animals , Antibodies, Fungal/chemistry , Antibodies, Fungal/genetics , Antibodies, Fungal/immunology , Aspartic Acid Endopeptidases/immunology , Bacteriophage M13 , Candidiasis/genetics , Candidiasis/immunology , Candidiasis/pathology , Disease Models, Animal , Female , Fungal Proteins/immunology , Humans , Mice, Inbred BALB C , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , omega-Chloroacetophenone
8.
Bioconjug Chem ; 26(6): 1144-55, 2015 Jun 17.
Article in English | MEDLINE | ID: mdl-25938892

ABSTRACT

A novel immunoconjugate (L-DOS47) was developed and characterized as a therapeutic agent for tumors expressing CEACAM6. The single domain antibody AFAIKL2, which targets CEACAM6, was expressed in the Escherichia coli BL21 (DE3) pT7-7 system. High purity urease (HPU) was extracted and purified from Jack bean meal. AFAIKL2 was activated using N-succinimidyl [4-iodoacetyl] aminobenzoate (SIAB) as the cross-linker and then conjugated to urease. The activation and conjugation reactions were controlled by altering pH. Under these conditions, the material ratio achieved conjugation ratios of 8-11 antibodies per urease molecule, the residual free urease content was practically negligible (<2%), and high purity (>95%) L-DOS47 conjugate was produced using only ultradiafiltration to remove unreacted antibody and hydrolyzed cross-linker. L-DOS47 was characterized by a panel of analytical techniques including SEC, IEC, Western blot, ELISA, and LC-MS(E) peptide mapping. As the antibody-urease conjugate ratio increased, a higher binding signal was observed. The specificity and cytotoxicity of L-DOS47 was confirmed by screening in four cell lines (BxPC-3, A549, MCF7, and CEACAM6-transfected H23). BxPC-3, a CEACAM6-expressing cell line was found to be most susceptible to L-DOS47. L-DOS47 is being investigated as a potential therapeutic agent in human phase I clinical studies for nonsmall cell lung cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Camelids, New World , Canavalia/enzymology , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Single-Domain Antibodies/therapeutic use , Urease/therapeutic use , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Enzyme Therapy , Humans , Immunoconjugates/chemistry , Male , Mice, Nude , Molecular Sequence Data , Single-Domain Antibodies/chemistry , Urease/chemistry
10.
Ther Deliv ; 4(10): 1321-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24116915

ABSTRACT

Nanobodies (Nbs) are small antibody fragments derived from camelid heavy chain antibodies through recombinant gene technology. Their exceptional physicochemical properties, possibility of humanization and unique antigen recognition properties make them excellent candidates for targeted delivery of biologically active components. Several different therapeutic approaches based on the novel camelid Nbs have been developed to treat a wide range of diseases ranging from immune, bone, blood and neurological disorders; infectious diseases and cancer. This review provides a comprehensive overview of the current state of the use of camelid-derived Nbs as novel therapeutic agents against multiple diseases.


Subject(s)
Camelids, New World/immunology , Camelus/immunology , Immunotherapy/methods , Immunotoxins/therapeutic use , Nanomedicine/methods , Single-Domain Antibodies/therapeutic use , Animals , Antibody Specificity , Chemistry, Pharmaceutical , Drug Carriers , Humans , Immunotoxins/chemistry , Immunotoxins/immunology , Protein Conformation , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Structure-Activity Relationship
11.
FEBS J ; 280(15): 3543-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23627412

ABSTRACT

In 1901, the first Nobel Prize in Physiology or Medicine was awarded to Emil von Behring for his ground-breaking discovery of serum therapy: serum from horses vaccinated with toxin-containing culture medium of Corynebacterium diphtheriae contained life-saving 'antitoxins'. The molecular nature of the ADP-ribosylating toxin and the neutralizing antibodies were unraveled only 50 years later. Today, von Behring's antibody therapy is being refined with a new generation of recombinant antibodies and antibody fragments. Nanobodies, which are single-domain antibodies derived from the peculiar heavy-chain antibodies of llamas and other camelids, are emerging as a promising new class of highly specific enzyme inhibitors. In this review, we illustrate the potential of nanobodies as tools to block extracellular and intracellular ADP-ribosyltransferases (ARTs), using the toxin-related membrane-bound mammalian ecto-enzyme ARTC2 and the actin-ADP-ribosylating Salmonella virulence plasmid factor B toxin of Salmonella enterica as examples.


Subject(s)
ADP Ribose Transferases/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , ADP Ribose Transferases/metabolism , Amino Acid Sequence , Animals , Bacterial Infections/drug therapy , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , Camelids, New World , Drug Delivery Systems , Humans , Molecular Sequence Data , Single-Domain Antibodies/chemistry , Virulence Factors/antagonists & inhibitors , Virulence Factors/metabolism
12.
Postepy Hig Med Dosw (Online) ; 66: 348-58, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22706121

ABSTRACT

 Serum of animals belonging to the Camelidae family (camels and llamas) contains fully active antibodies that are naturally devoid of light chains. Variable domains derived from heavy chain antibodies (hcAb) called VHHs or nanobodies™ can bind antigens as effectively as full-length antibodies and are easy to clone and express. Because of their potential, VHHs are being intensively studied as potential therapeutic, diagnostic and imaging tools. The paper reviews the molecular background of heavy chain antibodies and describes methods of obtaining recombinant fragments of heavy chain antibodies as well as their therapeutic, diagnostic and other applications.


Subject(s)
Immunoglobulin Fragments/therapeutic use , Immunoglobulin Heavy Chains/therapeutic use , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Autoimmune Diseases/drug therapy , Camelids, New World , Chromatography, Affinity/methods , Communicable Diseases/drug therapy , Fishes , Hematologic Diseases/drug therapy , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/isolation & purification , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/isolation & purification , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/isolation & purification , Single-Domain Antibodies/therapeutic use , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL