1
|
Ncobeni N, de la Torre BG, Albericio F, Kruger HG, Parboosing R. Active targeting of CD4 +T lymphocytes by PEI-capped, peptide-functionalized gold nanoparticles. NANOTECHNOLOGY 2022; 33:405101. [PMID: 35700711 DOI: 10.1088/1361-6528/ac7885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
Active targeting is a promising approach for the treatment of viral infections. In particular, site-specific formulations for the treatment of HIV infection may overcome challenges associated with current ARV regimens. In this study we explored active targeting by synthesizing a gold nanoparticle construct decorated with an anti-CD4 cyclic peptide. The aim was to demonstrate selectivity of the system for the CD4 receptor and to deliver the RNA payload into T-lymphocytes. Colloidal gold nanoparticles functionalized withN-succinimidyl 3-(2-pyridyldithio) propionate (SPDP) were formed by a one-pot synthesis method where thiol modified polyethyleneimine (PEI) was mixed with chloroauric acid. PEI-SPDP AuNPs (gold nanoparticles) were conjugated to an anti-CD4 peptide and loaded with RNA. We measured toxicity and uptake using TZM-bl and HeLa cells. Our findings show that the nanoparticles bind selectively to CD4 + cells. UV-vis characterisation of the nanoparticles revealed a surface plasmon resonance (SPR) peak at 527 nm, corresponding to a 6 nm diameter. HRTEM of the complete nanoparticles visualised circular shaped particles with average diameter of ∼7 nm. The polydispersity index was calculated to be 0.08, indicating monodispersity of complete NPS in solution. Through the pyridine-2-thione assay each nanoparticle was calculated to carry 1.37 × 105SPDP molecules available for peptide binding. Flow cytometry showed that 13.6% of TZM-bl cells, and 0.14% of HeLa cells retained fluorescence after an overnight incubation, an indication of system binding. No internal RNA delivery was demonstrated. Further work is required to improve internalization.
Collapse
Affiliation(s)
- Nomfundo Ncobeni
- Department of Virology-University of KwaZulu-Natal and National Health Laboratory Service, Durban, South Africa
- Catalysis and Peptide Research Labs, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Beatriz G de la Torre
- KwaZulu Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Fernando Albericio
- Peptide Science Laboratory, School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), E-08034 Barcelona, Spain
- CIBER-BBN, Networking Centre on Bioengineering, Biomaterials and Nanomedicine, and Department of Organic Chemistry, University of Barcelona, E-08028 Barcelona, Spain
| | - Hendrik G Kruger
- Catalysis and Peptide Research Labs, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Raveen Parboosing
- Department of Virology-University of KwaZulu-Natal and National Health Laboratory Service, Durban, South Africa
| |
Collapse
|
2
|
Van Holsbeeck K, Martins JC, Ballet S. Downsizing antibodies: Towards complementarity-determining region (CDR)-based peptide mimetics. Bioorg Chem 2021; 119:105563. [PMID: 34942468 DOI: 10.1016/j.bioorg.2021.105563] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/20/2021] [Accepted: 12/12/2021] [Indexed: 12/27/2022]
Abstract
Monoclonal antibodies emerged as an important therapeutic drug class with remarkable specificity and binding affinity. Nonetheless, these heterotetrameric immunoglobulin proteins come with high manufacturing and therapeutic costs which can take extraordinary proportions, besides other limitations such as their limited in cellulo access imposed by their molecular size (ca. 150 kDa). These drawbacks stimulated the development of downsized functional antibody fragments (ca. 15-50 kDa), together with smaller synthetic peptides (ca. 1-3 kDa) derived from the antibodies' crucial complementarity-determining regions (CDR). Despite the general lack of success in the literal translation of CDR loops in peptide mimetics, rational structure-based and computational approaches have shown their potential for obtaining functional CDR-based peptide mimetics. In this review, we describe the efforts made in the development of antibody and nanobody paratope-derived peptide mimetics with particular focus on the used design strategies, in addition to highlighting the challenges associated with their development.
Collapse
Affiliation(s)
- Kevin Van Holsbeeck
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; NMR and Structure Analysis Unit, Ghent University, Krijgslaan 281 S4, 9000 Ghent, Belgium
| | - José C Martins
- NMR and Structure Analysis Unit, Ghent University, Krijgslaan 281 S4, 9000 Ghent, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
3
|
Zhuang X, Stahl SJ, Watts NR, DiMattia MA, Steven AC, Wingfield PT. A cell-penetrating antibody fragment against HIV-1 Rev has high antiviral activity: characterization of the paratope. J Biol Chem 2014; 289:20222-33. [PMID: 24878961 DOI: 10.1074/jbc.m114.581090] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The HIV-1 protein Rev oligomerizes on viral transcripts and directs their nuclear export. Previously, a Fab against Rev generated by phage display was used to crystallize and solve the structure of the Rev oligomerization domain. Here we have investigated the capability of this Fab to block Rev oligomerization and inhibit HIV-1 replication. The Fab itself did not have antiviral activity, but when a Tat-derived cell-penetrating peptide was appended, the resulting molecule (FabRev1-Tat) was strongly inhibitory of three different CCR5-tropic HIV-1 isolates (IC50 = 0.09-0.44 μg/ml), as assessed by suppression of reverse transcriptase activity in infected peripheral blood mononuclear cells, and had low cell toxicity (TC50 > 100 μg/ml). FabRev1-Tat was taken up by both peripheral blood mononuclear and HEK293T cells, appearing in both the cytoplasm and nucleus, as shown by immunofluorescence confocal laser scanning microscopy. Computational alanine scanning was used to identify key residues in the complementarity-determining regions to guide mutagenesis experiments. Residues in the light chain CDR3 (LCDR3) were assessed to be important. Residues in LCDR3 were mutated, and LCDR3-Tyr(92) was found to be critical for binding to Rev, as judged by surface plasmon resonance and electron microscopy. Peptides corresponding to all six CDR regions were synthesized and tested for Rev binding. None of the linear peptides had significant affinity for Rev, but four of the amide-cyclic forms did. Especially cyclic-LCDR3 (LGGYPAASYRTA) had high affinity for Rev and was able to effectively depolymerize Rev filaments, as shown by both surface plasmon resonance and electron microscopy.
Collapse
Affiliation(s)
| | | | | | - Michael A DiMattia
- the Laboratory of Structural Biology Research, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Alasdair C Steven
- the Laboratory of Structural Biology Research, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | | |
Collapse
|
4
|
Timmerman P, Shochat SG, Desmet J, Barderas R, Casal JI, Meloen RH, Altschuh D. Binding of CDR-derived peptides is mechanistically different from that of high-affinity parental antibodies. J Mol Recognit 2010; 23:559-68. [DOI: 10.1002/jmr.1017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
5
|
Boschek CB, Apiyo DO, Soares TA, Engelmann HE, Pefaur NB, Straatsma TP, Baird CL. Engineering an ultra-stable affinity reagent based on Top7. Protein Eng Des Sel 2009; 22:325-32. [DOI: 10.1093/protein/gzp007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
6
|
Kiss C, Fisher H, Pesavento E, Dai M, Valero R, Ovecka M, Nolan R, Phipps ML, Velappan N, Chasteen L, Martinez JS, Waldo GS, Pavlik P, Bradbury AR. Antibody binding loop insertions as diversity elements. Nucleic Acids Res 2006; 34:e132. [PMID: 17023486 PMCID: PMC1635297 DOI: 10.1093/nar/gkl681] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2006] [Accepted: 09/03/2006] [Indexed: 11/13/2022] Open
Abstract
In the use of non-antibody proteins as affinity reagents, diversity has generally been derived from oligonucleotide-encoded random amino acids. Although specific binders of high-affinity have been selected from such libraries, random oligonucleotides often encode stop codons and amino acid combinations that affect protein folding. Recently it has been shown that specific antibody binding loops grafted into heterologous proteins can confer the specific antibody binding activity to the created chimeric protein. In this paper, we examine the use of such antibody binding loops as diversity elements. We first show that we are able to graft a lysozyme-binding antibody loop into green fluorescent protein (GFP), creating a fluorescent protein with lysozyme-binding activity. Subsequently we have developed a PCR method to harvest random binding loops from antibodies and insert them at predefined sites in any protein, using GFP as an example. The majority of such GFP chimeras remain fluorescent, indicating that binding loops do not disrupt folding. This method can be adapted to the creation of other nucleic acid libraries where diversity is flanked by regions of relative sequence conservation, and its availability sets the stage for the use of antibody loop libraries as diversity elements for selection experiments.
Collapse
Affiliation(s)
- Csaba Kiss
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Hugh Fisher
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Emanuele Pesavento
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Minghua Dai
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Rosa Valero
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Milan Ovecka
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Rhiannon Nolan
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - M. Lisa Phipps
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Nileena Velappan
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Leslie Chasteen
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | | | - Geoffrey S. Waldo
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | - Peter Pavlik
- HCDR3s as diversity elements, Los Alamos National LaboratoryLos Alamos, NM, USA
| | | |
Collapse
|
7
|
Bès C, Troadec S, Chentouf M, Breton H, Lajoix AD, Heitz F, Gross R, Plückthun A, Chardès T. PIN-bodies: a new class of antibody-like proteins with CD4 specificity derived from the protein inhibitor of neuronal nitric oxide synthase. Biochem Biophys Res Commun 2006; 343:334-44. [PMID: 16540093 DOI: 10.1016/j.bbrc.2006.02.126] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Accepted: 02/06/2006] [Indexed: 11/24/2022]
Abstract
By inserting the CB1 paratope-derived peptide (PDP) from the anti-CD4 13B8.2 antibody binding pocket into each of the three exposed loops of the protein inhibitor of neuronal nitric oxide synthase (PIN), we have combined the anti-CD4 specificity of the selected PDP with the stability, ease of expression/purification, and the known molecular architecture of the phylogenetically well-conserved PIN scaffold protein. Such "PIN-bodies" were able to bind CD4 with a better affinity and specificity than the soluble PDP; additionally, in competitive ELISA experiments, CD4-specific PIN-bodies were more potent inhibitors of the binding of the parental recombinant antibody 13B8.2 to CD4 than the soluble PDP. The efficiency of CD4-specific CB1-inserted PIN-bodies was confirmed in biological assays where these constructs showed higher potencies to block antigen presentation by inhibition of IL-2 secretion and to inhibit the one-way and two-way mixed lymphocyte reactions, compared with soluble anti-CD4 PDP CB1. Insertion of the PDP into the first exposed loop (position 33/34) of PIN appeared to be the most promising scaffold. Taken together, our findings demonstrate that the PIN molecule is a suitable scaffold to expose new peptide loops and generate small artificial ligand-binding products with defined specificities.
Collapse
Affiliation(s)
- Cédric Bès
- CNRS UMR 5160, Centre de Pharmacologie et Biotechnologie pour la Santé, Faculté de Pharmacie, 15 Avenue Charles Flahault, BP 14491, 34093 Montpellier Cedex 5, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Peter JC, Wallukat G, Tugler J, Maurice D, Roegel JC, Briand JP, Hoebeke J. Modulation of the M2 muscarinic acetylcholine receptor activity with monoclonal anti-M2 receptor antibody fragments. J Biol Chem 2004; 279:55697-706. [PMID: 15485827 DOI: 10.1074/jbc.m407213200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Antibodies directed against the second extracellular loop of G protein-coupled receptors are known to have functional activities. From a partial agonist monoclonal antibody directed against the M2 muscarinic receptor, we constructed and produced a single chain variable fragment with high affinity for its target epitope. The fragment is able to recognize its receptor on Chinese hamster ovary cells transfected with the M2 muscarinic acetylcholine receptor to block the effect of carbachol on this receptor and to exert an inverse agonist activity on the basal activity of the receptor. The antibody fragment is also able to increase the basal rhythm of cultured neonatal rat cardiomyocytes and to inhibit in a non-competitive manner the negative chronotropic effect of carbachol. This antibody fragment is able to exert its inverse agonist activity in vivo on mouse heart activity. The immunological strategy presented here could be useful to develop specific allosteric inverse agonist reagents for G protein-coupled receptors.
Collapse
MESH Headings
- Allosteric Site
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/chemistry
- Base Sequence
- Binding, Competitive
- Blotting, Western
- CHO Cells
- Carbachol/pharmacology
- Cells, Cultured
- Cricetinae
- Dose-Response Relationship, Drug
- Dose-Response Relationship, Immunologic
- Electrophoresis, Polyacrylamide Gel
- Epitopes/chemistry
- Escherichia coli/metabolism
- Immunoglobulin Fab Fragments
- Immunohistochemistry
- Mice
- Models, Molecular
- Molecular Sequence Data
- Myocytes, Cardiac/metabolism
- Nucleotides/chemistry
- Peptides/chemistry
- Protein Structure, Tertiary
- Rats
- Receptor, Muscarinic M2/chemistry
- Receptor, Muscarinic M2/metabolism
- Receptors, Cholinergic/chemistry
- Sequence Homology, Amino Acid
- Surface Plasmon Resonance
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Jean-Christophe Peter
- CNRS, Unité Propre de Recherche 9021, Institut de Biologie Moléculaire et Cellulaire, Laboratory of Therapeutical Chemistry and Immunology, 15 rue René Descartes, F-67084 Strasbourg, France
| | | | | | | | | | | | | |
Collapse
|
9
|
Perosa F, Favoino E, Caragnano MA, Dammacco F. Human CD4 mimicry by anti-idiotypic monoclonal antibody 16D7 is based on a conformational epitope. Immunol Lett 2004; 95:145-53. [PMID: 15388254 DOI: 10.1016/j.imlet.2004.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2004] [Accepted: 06/30/2004] [Indexed: 10/26/2022]
Abstract
The mouse anti-idiotypic mAb (mAb2) 16D7 recognizes the paratope (combining site-associated epitope) of the syngeneic anti-human CD4 mAb HP2/6 (mAb1), a down-modulator of T cell function. 16D7 mimics CD4 in xenogeneic settings in humans and can thus be used to target CD4+ T cells for therapeutic purposes. To define the minimum structural requirement for CD4 mimicry, 16D7 isolated L and H chains were tested for their ability to inhibit mAb1 binding to either CD4 Ag or to mAb2. 16D7 L only specifically inhibited these interactions. Alignment of 16D7 L variable region sequence with that of CD4 defined peptides L2, L3 and L7 with at least 68% of similarity and 30% of identity to exposed regions of CD4. Furthermore, peptides L1, L4, L5, L6 and L8 were selected from a set of 10-mer overlapping peptides (covering the FW and CDR regions of 16D7 L) because of their reactivity with mAb1. Only the synthesized L2 reacted with HP2/6 in slot dot assay and inhibited mAb2-mAb1 interaction. Immunization of BALB/c mice with 16D7, 16D7-F(ab')2, 16D7 L and L2, resulted in the production of CD4-specific Ab3 only in mice immunized with 16D7 and its F(ab')2 fragments. The lack of CD4 reactivity with sera elicited with 16D7 L and L2 was not due to their lack of immunogenicity since both sera reacted with the corresponding immunogen and the former also inhibited the binding of mAb1 to 16D7. The results suggest that CD4 mimicry by 16D7 is based on a conformational epitope and L2 is only one of the HP2/6-specific contact points of 16D7.
Collapse
Affiliation(s)
- Federico Perosa
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Internal Medicine, University of Bari Medical School, Piazza G. Cesare 11, 70124 Bari, Italy.
| | | | | | | |
Collapse
|
10
|
Bès C, Briant-Longuet L, Cerutti M, Heitz F, Troadec S, Pugnière M, Roquet F, Molina F, Casset F, Bresson D, Péraldi-Roux S, Devauchelle G, Devaux C, Granier C, Chardes T. Mapping the paratope of anti-CD4 recombinant Fab 13B8.2 by combining parallel peptide synthesis and site-directed mutagenesis. J Biol Chem 2003; 278:14265-73. [PMID: 12566463 DOI: 10.1074/jbc.m210694200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We analyzed antigen-binding residues from the variable domains of anti-CD4 antibody 13B8.2 using the Spot method of parallel peptide synthesis. Sixteen amino acids, defined as Spot critical residues (SCR), were identified on the basis of a 50% decrease in CD4 binding to alanine analogs of reactive peptides. Recombinant Fab 13B8.2 mutants were constructed with alanine residues in place of each of the 16 SCR, expressed in the baculovirus cell system, and purified. CD measurements indicated that the mutated proteins were conformationally intact, with a beta-sheet secondary structure similar to that of wild-type Fab. Compared with the CD4-binding capacity of wild-type Fab 13B8.2, 11 light (Y32-L, W35-L, Y36-L, H91-L, and Y92-L) and heavy chain (H35-H, R38-H, W52-H, R53-H, F100K-H, and W103-H) Fab single mutants showed a decrease in CD4 recognition as demonstrated by enzyme-linked immunosorbent assay, BIAcore, and flow cytometry analyses. The five remaining Fab mutants showed antigen-binding properties similar to those of wild-type Fab. Recombinant Fab mutants that showed decreased CD4 binding also lost their capacity to inhibit human immunodeficiency virus promoter activation and the antigen-presenting ability that wild-type Fab displays. Molecular modeling of the 13B8.2 antibody paratope indicated that most of these critical residues are appropriately positioned inside the putative CD4-binding pocket, whereas the five SCR that were not confirmed by mutagenesis show an unfavorable positioning. Taken together, these results indicate that most of the residues defined by the Spot method as critical matched with important residues defined by mutagenesis in the whole protein context. The identification of critical residues for CD4 binding in the paratope of anti-CD4 recombinant Fab 13B8.2 provides the opportunity for the generation of improved anti-CD4 molecules with more efficient pharmacological properties.
Collapse
Affiliation(s)
- Cédric Bès
- CNRS UMR 5094, Institut de Biotechnologie et Pharmacologie, Faculté de Pharmacie, 15 Avenue Charles Flahault, 34093 Montpellier Cedex 5, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Laune D, Molina F, Ferrières G, Villard S, Bès C, Rieunier F, Chardès T, Granier C. Application of the Spot method to the identification of peptides and amino acids from the antibody paratope that contribute to antigen binding. J Immunol Methods 2002; 267:53-70. [PMID: 12135800 DOI: 10.1016/s0022-1759(02)00140-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Overlapping peptide scans prepared by Spot synthesis have been used to map interaction sites in several systems. Here we report our experience with this approach to identify peptides from the variable parts of anti-hapten, anti-peptide and anti-protein antibodies that retain their specific antigen-binding capacity in the Spot format. In general, the identification by the Spot method of antigen-reactive peptides was confirmed by using soluble peptides which demonstrated antigen-binding capacity in ELISA or Biacore and, biological activity for some peptides derived from anti-CD4 antibodies. The Spot method was also used to map precisely key residues from the antibody paratope. The identification of critical residues from an anti-troponin I antibody of diagnostic interest is reported as well as the compiled results from the analysis of five other antibodies of various specificities. A critical assessment of our results is provided by comparing results obtained by our approach in the mapping of antibody residues critical for antigen binding with data from the literature concerning the structural analysis of antigen-antibody complexes.
Collapse
Affiliation(s)
- Daniel Laune
- CNRS UMR 5094, Faculté de Pharmacie, Montpellier, France.
| | | | | | | | | | | | | | | |
Collapse
|