Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1844(11): 1889-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25280770
2.
Biochim Biophys Acta ; 1844(11): 2016-2023, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25038323

ABSTRACT

Antibodies have evolved to function in oxidative, extracellular environments. A pair of cysteines in close proximity will oxidatively react to form a disulfide bond that fixes and stabilizes the tertiary structure of a protein. Immunoglobulin G (IgG) includes several disulfide bonds, and the patterns of inter-chain disulfide bonds characterize different IgG sub-classes. Moreover, the Ig-fold domains are characterized by a buried intra-domain disulfide bond, which is important for its structural stability. However, the intra-domain disulfide bond can be replaced without crucial effects on the structure and function, if the domain structure is intrinsically stable or has been stabilized by protein engineering. In previous studies, disulfide bonds were removed by amino-acid substitution indicating that Val and/or Ala (i.e. Ala-Ala, Ala-Val, Val-Ala, and Val-Ala) pairs were preferred for cysteine replacement in the Ig-fold domain. As such, these mutations may be useful for the intracellular use of antibodies. Recently, additional intra-domain disulfide bonds have been shown to stabilize Ig-fold domains and whole IgGs. In heavy chain variable or light chain variable domains, the introduction of additional disulfide bonds into the framework region did not reduce antigen-binding affinity, suggesting that generating disulfide bonds may be a method for stabilizing IgG and antibody fragments, such as the antigen-binding fragment, and single-chain and single-domain antibodies. This article is part of a Special Issue entitled: Recent advances in molecular engineering of antibody.

3.
PLoS Negl Trop Dis ; 6(11): e1902, 2012.
Article in English | MEDLINE | ID: mdl-23166849

ABSTRACT

BACKGROUND: The discovery of Nanobodies (Nbs) with a direct toxic activity against African trypanosomes is a recent advancement towards a new strategy against these extracellular parasites. The anti-trypanosomal activity relies on perturbing the highly active recycling of the Variant-specific Surface Glycoprotein (VSG) that occurs in the parasite's flagellar pocket. METHODOLOGY/PRINCIPAL FINDINGS: Here we expand the existing panel of Nbs with anti-Trypanosoma brucei potential and identify four categories based on their epitope specificity. We modified the binding properties of previously identified Nanobodies Nb_An05 and Nb_An33 by site-directed mutagenesis in the paratope and found this to strongly affect trypanotoxicity despite retention of antigen-targeting properties. Affinity measurements for all identified anti-trypanosomal Nbs reveal a strong correlation between trypanotoxicity and affinity (K(D)), suggesting that it is a crucial determinant for this activity. Half maximal effective (50%) affinity of 57 nM was calculated from the non-linear dose-response curves. In line with these observations, Nb humanizing mutations only preserved the trypanotoxic activity if the K(D) remained unaffected. CONCLUSIONS/SIGNIFICANCE: This study reveals that the binding properties of Nanobodies need to be compatible with achieving an occupancy of >95% saturation of the parasite surface VSG in order to exert an anti-trypanosomal activity. As such, Nb-based approaches directed against the VSG target would require binding to an accessible, conserved epitope with high affinity.


Subject(s)
Antibodies, Protozoan/immunology , Single-Domain Antibodies/immunology , Trypanosoma brucei brucei/immunology , Animals , Antibodies, Protozoan/pharmacology , Antibody Affinity , Mice , Mice, Inbred C57BL , Microbial Viability/drug effects , Single-Domain Antibodies/pharmacology , Trypanosoma brucei brucei/drug effects
4.
Methods Mol Biol ; 911: 399-416, 2012.
Article in English | MEDLINE | ID: mdl-22886265

ABSTRACT

The successful medical application of single domain antibodies largely depends on their functionality. This feature is partly determined by the intrinsic stability of the single domain. Therefore a lot of research has gone into the elucidation of rules to uniformly increase stability of antibodies. Recently, a novel intra-domain disulfide bond was independently discovered by two research groups, after either rational design or careful investigation of the naturally occurring camelid antibody repertoire. By introducing this particular disulfide bond within a single domain antibody, the conformational stability can be increased in general. In this chapter it is described how to introduce this extra intra-domain disulfide bond and how to estimate the biophysical and biochemical impact of this cystine on the domain.


Subject(s)
Disulfides/chemistry , Single-Domain Antibodies/chemistry , Amino Acid Sequence , Antibody Affinity , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Denaturation , Protein Engineering , Protein Folding , Protein Stability , Protein Structure, Tertiary , Single-Domain Antibodies/genetics , Temperature
5.
Methods Mol Biol ; 911: 491-505, 2012.
Article in English | MEDLINE | ID: mdl-22886272

ABSTRACT

Chromatin immunoprecipitation (ChIP), followed by microarray hybridization (ChIP-chip) or high-throughput sequencing (ChIP-seq), is becoming a widely used powerful method for the analysis of the in vivo DNA-protein interactions at genomic scale.The success of ChIP largely depends on the quality of antibodies. Although polyclonal antibodies have been successfully used for ChIP, their production requires regular immunization and they exhibit high aspecificity and batch to batch variability. These problems can be circumvented by generating monoclonal antibodies (mAbs) via hybridoma technology. However, such mAbs do not often capture DNA-protein complexes and are not amenable to engineering. Nanobodies are recombinant single domain antibody fragments derived from camelid Heavy-Chain antibodies. Nanobodies exhibit high affinity and specificity towards their cognate antigens and often capture their target antigens in solution. Moreover, the Nanobody genes can be easily tailored to streamline ChIP.Here, we describe a Nanobody-based ChIP protocol which we have successfully used for genome-wide identification of the binding sites of the low-abundant transcription factor Ss-LrpB from the hyperthermoacidophilic archaeon Sulfolobus solfataricus.


Subject(s)
Chromatin Immunoprecipitation/methods , Single-Domain Antibodies/metabolism , Sulfolobus solfataricus/drug effects , Sulfolobus solfataricus/genetics , Sulfolobus solfataricus/metabolism
6.
J Biol Chem ; 287(3): 1970-9, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22128183

ABSTRACT

The antigen-binding fragment of functional heavy chain antibodies (HCAbs) in camelids comprises a single domain, named the variable domain of heavy chain of HCAbs (VHH). The VHH harbors remarkable amino acid substitutions in the framework region-2 to generate an antigen-binding domain that functions in the absence of a light chain partner. The substitutions provide a more hydrophilic, hence more soluble, character to the VHH but decrease the intrinsic stability of the domain. Here we investigate the functional role of an additional hallmark of dromedary VHHs, i.e. the extra disulfide bond between the first and third antigen-binding loops. After substituting the cysteines forming this interloop cystine by all 20 amino acids, we selected and characterized several VHHs that retain antigen binding capacity. Although VHH domains can function in the absence of an interloop disulfide bond, we demonstrate that its presence constitutes a net advantage. First, the disulfide bond stabilizes the domain and counteracts the destabilization by the framework region-2 hallmark amino acids. Second, the disulfide bond rigidifies the long third antigen-binding loop, leading to a stronger antigen interaction. This dual beneficial effect explains the in vivo antibody maturation process favoring VHH domains with an interloop disulfide bond.


Subject(s)
Cysteine/chemistry , Disulfides/chemistry , Immunoglobulin Heavy Chains/chemistry , Single-Chain Antibodies/chemistry , Animals , Camelus , Cysteine/genetics , Cysteine/metabolism , Disulfides/metabolism , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Protein Stability , Protein Structure, Tertiary , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
7.
Methods Mol Biol ; 790: 239-59, 2011.
Article in English | MEDLINE | ID: mdl-21948420

ABSTRACT

The immunization of an animal with a whole proteome or the infection of an animal and the screening of the resulting antibody repertoire on either the same or different proteome(s) or the infecting agent(s), omits the laborious steps of recombinant protein expression and purification to obtain multiple antigen binders. This procedure allows the identification of antibodies that are specific to unique or common signatures of different proteomes without prior knowledge of these signatures.Nanobodies are the smallest (15 kDa, 2.2 nm diameter, 4 nm height) in vivo affinity-matured functional antigen-binding entities that are derived from camelid heavy-chain antibodies. Due to their small size, recognition of unique epitopes, high affinity, and easy tailoring, nanobodies are attractive affinity reagents for various applications, including diagnosis and therapy.We detail a protocol to generate, isolate, express, and purify anti-infectome/anti-proteome nanobodies.


Subject(s)
Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/immunology , Nanostructures , Proteome/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Trypanosoma/immunology , Animals , Antibody Specificity , Chromatography, Affinity , DNA Restriction Enzymes/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Escherichia coli/cytology , Escherichia coli/genetics , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/isolation & purification , Leukocytes, Mononuclear/immunology , Peptide Library , Periplasm/metabolism , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
8.
Protein Eng Des Sel ; 24(9): 727-35, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21798998

ABSTRACT

During scorpion envenoming, highly toxic small polypeptides of the venom diffuse rapidly within the victim, causing serious medical problems. Nanobodies (Nbs), the recombinant single-domain antigen-binding fragments of camel-specific heavy-chain only antibodies, offer special advantages in therapy over classic antibody fragments due to their robustness and smaller size, matching the size of the scorpion toxins. Recently, a potent AahII scorpion toxin-neutralizing Nb was identified. However, this NbAahII10 contains a single Cys in its first antigen-binding loop, leading to Nb dimerization upon prolonged storage. In this work, we first investigate the efficacy of NbAahII10 variants in which this Cys was substituted by Ala, Ser or Thr. Second, the NbAahII10 Cys/Ser mutant displaying the best functional properties is subsequently humanized. It is demonstrated that the maximally humanized version of NbAahII10 Cys/Ser maintains its high affinity for the antigen without conceding much on expression yield and stability. More importantly, its neutralizing capacity is preserved as all mice survive injections of seven LD(50) and 50% of mice survived nine LD(50) of the scorpion toxin. Thus, this humanized Nb is the best candidate to develop a therapy in human against the most toxic venom compound of one of the most dangerous scorpions.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neutralizing/immunology , Cysteine/chemistry , Scorpion Venoms/immunology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/metabolism , Antibody Affinity , Camelus , Chromatography, Gel , Cysteine/genetics , Cysteine/metabolism , Electrophoresis, Polyacrylamide Gel , Gene Knockdown Techniques , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/metabolism , Lethal Dose 50 , Mice , Models, Molecular , Molecular Sequence Data , Neutralization Tests , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Scorpion Venoms/metabolism
9.
PLoS Pathog ; 7(6): e1002072, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21698216

ABSTRACT

The African trypanosome Trypanosoma brucei, which persists within the bloodstream of the mammalian host, has evolved potent mechanisms for immune evasion. Specifically, antigenic variation of the variant-specific surface glycoprotein (VSG) and a highly active endocytosis and recycling of the surface coat efficiently delay killing mediated by anti-VSG antibodies. Consequently, conventional VSG-specific intact immunoglobulins are non-trypanocidal in the absence of complement. In sharp contrast, monovalent antigen-binding fragments, including 15 kDa nanobodies (Nb) derived from camelid heavy-chain antibodies (HCAbs) recognizing variant-specific VSG epitopes, efficiently lyse trypanosomes both in vitro and in vivo. This Nb-mediated lysis is preceded by very rapid immobilisation of the parasites, massive enlargement of the flagellar pocket and major blockade of endocytosis. This is accompanied by severe metabolic perturbations reflected by reduced intracellular ATP-levels and loss of mitochondrial membrane potential, culminating in cell death. Modification of anti-VSG Nbs through site-directed mutagenesis and by reconstitution into HCAbs, combined with unveiling of trypanolytic activity from intact immunoglobulins by papain proteolysis, demonstrates that the trypanolytic activity of Nbs and Fabs requires low molecular weight, monovalency and high affinity. We propose that the generation of low molecular weight VSG-specific trypanolytic nanobodies that impede endocytosis offers a new opportunity for developing novel trypanosomiasis therapeutics. In addition, these data suggest that the antigen-binding domain of an anti-microbial antibody harbours biological functionality that is latent in the intact immunoglobulin and is revealed only upon release of the antigen-binding fragment.


Subject(s)
Antibodies, Protozoan/pharmacology , Endocytosis/drug effects , Trypanosoma brucei brucei/immunology , Variant Surface Glycoproteins, Trypanosoma/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Antibodies, Protozoan/therapeutic use , Antibody Affinity , Cells, Cultured , Down-Regulation/drug effects , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Models, Molecular , Molecular Sequence Data , Nanoparticles , Trypanocidal Agents/pharmacology , Trypanocidal Agents/therapeutic use , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei brucei/physiology , Trypanosoma brucei brucei/ultrastructure , Trypanosomiasis, African/immunology , Trypanosomiasis, African/metabolism , Trypanosomiasis, African/therapy
10.
J Virol Methods ; 173(2): 300-5, 2011 May.
Article in English | MEDLINE | ID: mdl-21371506

ABSTRACT

Recently, single-domain recombinant antibody fragments (VHHs or nanobodies) against poliovirus type 1 were isolated. To examine the antigenicity of poliovirus using these recombinant VHHs, an alternative technique mimicking protein A immunoprecipitation had to be developed that was designed specifically for VHHs. The current study validated an affinity capturing assay that is based on the magnetic separation of unbound antigen and antigen-VHH complexes. The technique is simple, fast, reliable, quantitative and inexpensive and was employed to assess the reactivity of 15 VHHs for native infectious poliovirus (N antigen), heat-denatured virus (H antigen) and 14S subviral particles. Three distinct subsets of VHHs were tentatively distinguished based on their specificity for the antigens: one that binds only to 14S precursors, another that binds to the H antigen and a third that binds to the N antigen. Some VHHs of the latter two subsets bound 14S subviral particles with equal affinity but others had at least 100-fold less affinity for the precursors. All neutralizing VHHs were demonstrated to recognize N antigen and all N-specific VHHs were shown to be neutralizing. This study corroborates the findings that VHHs mainly target conformational epitopes and that they target additional epitopes as compared to classical antibodies. The described technique may be useful for epitope mapping and tracking conformational changes of proteins.


Subject(s)
Antibodies, Viral/immunology , Antigen-Antibody Complex/isolation & purification , Antigens, Viral/isolation & purification , Immunomagnetic Separation/methods , Poliovirus/isolation & purification , Epitope Mapping/methods , Epitopes/immunology , Protein Conformation
11.
Expert Rev Mol Diagn ; 10(6): 777-85, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20843201

ABSTRACT

With the advent of new antibody engineering technologies, conventional antibodies have been minimized into smaller antibody formats. Small size is an important advantage for current and future diagnostic development. Nanobodies® (Ablynx) are among the smallest known antigen-binding antibody fragments, and are derived from the heavy-chain only antibodies that occur naturally in the serum of Camelidae. Endowed by natural evolution, these Nanobodies inherently exhibit unique biophysical, biochemical and pharmacological characteristics. In addition to their excellent potential as molecules in drug development, Nanobodies possess very attractive functional properties that aid in their development for diagnostic tools. Here we present several examples of currently available applications of Nanobodies to the field of immunosensor for cancer, immunoaffinity chromatography, in vivo and intracellular imaging.


Subject(s)
Antibodies/immunology , Immunoglobulin Fragments/immunology , Molecular Diagnostic Techniques , Animals , Antibodies/blood , Antibodies/genetics , Antibody Specificity , Antigen-Antibody Reactions , Camelids, New World , Humans , Immunoglobulin Fragments/blood , Immunoglobulin Fragments/genetics , Molecular Probes/metabolism
12.
FASEB J ; 24(9): 3479-89, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20410443

ABSTRACT

Envenoming following scorpion sting is a common emergency in many parts of the world. Our aim was to ameliorate the current 100-kDa horse plasma antivenom serum (PAS)-derived Fab'(2) to more quickly reach the highly diffusible scorpion toxins (7 kDa). We immunized dromedaries with toxins from Androctonus australis hector (Aah) scorpions and cloned the single-domain antibody fragments or nanobodies (15 kDa) from their B cells. Nanobodies against AahI' toxin (with AahII the most toxic compound of the venom) were retrieved from the libraries, and their AahI'-toxin neutralization was monitored in mice. Remarkably, the NbAahI'F12 fully protected mice against 100 LD(50) of AahI' administered intracerebroventricularly. Moreover, where PAS failed completely to neutralize 2 LD(50) of crude venom injected subcutaneously, the designed bispecific NbF12-10 against AahI'/AahII toxins succeeded in neutralizing 5 LD(50). Finally, in a challenge assay in which mice were subcutaneously injected with a lethal dose of scorpion venom, the subsequent intravenous injection of 85 microg of NbF12-10 protected all mice, even if the whole procedure was repeated 3 times. Furthermore, the NbF12-10 remained fully protective when mice with severe signs of envenoming were treated a few minutes before the untreated mice died.


Subject(s)
Immunoglobulin Fragments/immunology , Scorpion Venoms/immunology , Animals , Camelus , Epitope Mapping , Immunoglobulin Fragments/isolation & purification , Male , Mice
13.
World J Biol Chem ; 1(7): 235-8, 2010 Jul 26.
Article in English | MEDLINE | ID: mdl-21537479

ABSTRACT

It is well established that all camelids have unique antibodies circulating in their blood. Unlike antibodies from all other species, these special antibodies are devoid of light chains, and are composed of a heavy chain homodimer. These so-called heavy-chain antibodies (HCAbs) are expressed after a V-D-J rearrangement and require dedicated constant gamma genes. An immune response is raised in these HCAbs following a classical immunization protocol. These HCAbs are easily purified from serum, and their antigen-binding fragment interacts with parts of the target that are less antigenic to conventional antibodies. The antigen binding site of the dromedary HCAb comprises one single domain, referred to as VHH or nanobody (Nb), therefore, a strategy was designed to clone the Nb repertoire of an immunized dromedary and to select the Nb with specificity for our target antigens. The monoclonal Nb is produced well in bacteria, is very stable and highly soluble, and it binds the antigen with high affinity and specificity. Currently, the recombinant Nb has been developed successfully for research purposes, as a probe in biosensors, to diagnose infections, or to treat diseases such as cancer or trypanosomiasis.

14.
Biochem J ; 424(2): 263-72, 2009 Nov 11.
Article in English | MEDLINE | ID: mdl-19732033

ABSTRACT

Scorpion venom, containing highly toxic, small polypeptides that diffuse rapidly within the patient, causes serious medical problems. Nanobodies, single-domain antigen-binding fragments derived from dromedary heavy-chain antibodies, have a size that closely matches that of scorpion toxins. Therefore these nanobodies might be developed into potent immunotherapeutics to treat scorpion envenoming. Multiple nanobodies of sub-nanomolar affinity to AahII, the most toxic polypeptide within the Androctonus australis hector venom, were isolated from a dromedary immunized with AahII. These nanobodies neutralize the lethal effect of AahII to various extents without clear correlation with the kinetic rate constants kon or koff, or the equilibrium dissociation constant, KD. One particular nanobody, referred to as NbAahII10, which targets a unique epitope on AahII, neutralizes 7 LD50 of this toxin in mice, corresponding to a neutralizing capacity of approx. 37000 LD50 of AahII/mg of nanobody. Such high neutralizing potency has never been reached before by any other monoclonal antibody fragment.


Subject(s)
Antibodies/immunology , Camelus/immunology , Neurotoxins/immunology , Peptides/immunology , Scorpion Venoms/immunology , Scorpions/immunology , Amino Acid Sequence , Animals , Antibodies/therapeutic use , Antibody Formation , Antibody Specificity , Epitopes/chemistry , Epitopes/immunology , Female , Mice , Molecular Sequence Data , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/therapy , Neurotoxins/chemistry , Neurotoxins/toxicity , Peptides/chemistry , Peptides/toxicity , Scorpion Venoms/chemistry , Scorpion Venoms/toxicity , Scorpions/chemistry , Sequence Alignment , Sequence Homology, Amino Acid
15.
J Biol Chem ; 284(5): 3273-3284, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19010777

ABSTRACT

Nanobodies, single-domain antigen-binding fragments of camelid-specific heavy-chain only antibodies offer special advantages in therapy over classic antibody fragments because of their smaller size, robustness, and preference to target unique epitopes. A Nanobody differs from a human heavy chain variable domain in about ten amino acids spread all over its surface, four hallmark Nanobody-specific amino acids in the framework-2 region (positions 42, 49, 50, and 52), and a longer third antigen-binding loop (H3) folding over this area. For therapeutic applications the camelid-specific amino acid sequences in the framework have to be mutated to their human heavy chain variable domain equivalent, i.e. humanized. We performed this humanization exercise with Nanobodies of the subfamily that represents close to 80% of all dromedary-derived Nanobodies and investigated the effects on antigen affinity, solubility, expression yield, and stability. It is demonstrated that the humanization of Nanobody-specific residues outside framework-2 are neutral to the Nanobody properties. Surprisingly, the Glu-49 --> Gly and Arg-50 --> Leu humanization of hallmark amino acids generates a single domain that is more stable though probably less soluble. The other framework-2 substitutions, Phe-42 --> Val and Gly/Ala-52 --> Trp, are detrimental for antigen affinity, due to a repositioning of the H3 loop as shown by their crystal structures. These insights were used to identify a soluble, stable, well expressed universal humanized Nanobody scaffold that allows grafts of antigen-binding loops from other Nanobodies with transfer of the antigen specificity and affinity.


Subject(s)
Antibodies/immunology , Camelus/immunology , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/genetics , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Sequence Homology, Amino Acid
16.
Biochemistry ; 47(42): 11041-54, 2008 Oct 21.
Article in English | MEDLINE | ID: mdl-18816062

ABSTRACT

A single-domain fragment, cAb-HuL22, of a camelid heavy-chain antibody specific for the active site of human lysozyme has been generated, and its effects on the properties of the I56T and D67H amyloidogenic variants of human lysozyme, which are associated with a form of systemic amyloidosis, have been investigated by a wide range of biophysical techniques. Pulse-labeling hydrogen-deuterium exchange experiments monitored by mass spectrometry reveal that binding of the antibody fragment strongly inhibits the locally cooperative unfolding of the I56T and D67H variants and restores their global cooperativity to that characteristic of the wild-type protein. The antibody fragment was, however, not stable enough under the conditions used to explore its ability to perturb the aggregation behavior of the lysozyme amyloidogenic variants. We therefore engineered a more stable version of cAb-HuL22 by adding a disulfide bridge between the two beta-sheets in the hydrophobic core of the protein. The binding of this engineered antibody fragment to the amyloidogenic variants of lysozyme inhibited their aggregation into fibrils. These findings support the premise that the reduction in global cooperativity caused by the pathogenic mutations in the lysozyme gene is the determining feature underlying their amyloidogenicity. These observations indicate further that molecular targeting of enzyme active sites, and of protein binding sites in general, is an effective strategy for inhibiting or preventing the aberrant self-assembly process that is often a consequence of protein mutation and the origin of pathogenicity. Moreover, this work further demonstrates the unique properties of camelid single-domain antibody fragments as structural probes for studying the mechanism of aggregation and as potential inhibitors of fibril formation.


Subject(s)
Amyloid/antagonists & inhibitors , Camelus/immunology , Immunoglobulin Fragments/metabolism , Muramidase/immunology , Amino Acid Sequence , Amyloid/chemistry , Amyloid/immunology , Amyloid/metabolism , Animals , Antibody Affinity , Camelus/genetics , Catalytic Domain/immunology , Humans , Immunoglobulin Fragments/genetics , In Vitro Techniques , Molecular Sequence Data , Muramidase/antagonists & inhibitors , Muramidase/chemistry , Muramidase/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Engineering , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
17.
Curr Opin Pharmacol ; 8(5): 600-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18691671

ABSTRACT

Antibodies are large and complex molecules, with two identical parts that bind independently of each other onto the antigen and the third part of the molecule that dictates the effector function(s). To improve the therapeutic value of antibodies, protein-engineering endeavors reduced the size of the antigen-binding moiety to a single-domain unit. Occasionally, it was demonstrated that the single-domain antigen-binding derivatives of antibodies can have--on their own--an agonistic (or antagonistic) effect on their target. The small size and strict monomeric behavior, in combination with other biochemical properties such as high solubility and high specificity and affinity for the cognate antigen, make single-domain antibodies ideal to design novel man-made conjugates harnessed with innovative effector functions outside the reach of classical antibodies.


Subject(s)
Antibodies/chemistry , Antibodies/therapeutic use , Animals , Antibodies/genetics , Antibodies, Blocking/chemistry , Antibodies, Blocking/genetics , Antibodies, Blocking/therapeutic use , Humans , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/therapeutic use
18.
Mol Immunol ; 45(14): 3847-56, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18614235

ABSTRACT

Many efforts aim at solving the serious problems encountered with immunotherapy against scorpion envenoming. The most attractive approach consists in generating single-chain antibody fragments (scFv) as their pharmaco-kinetic properties should match closely those of the scorpion toxins. Although high affinity scFv reagents have been generated in the past, their production level, stability, and toxin neutralizing capacity remain disappointingly poor. In the current study, we identified one Nanobody (Nb), a single-domain antigen-binding fragment of a dromedary Heavy-chain antibody (HCAb) that recognizes specifically the Androctonus australis hector AahI' toxin. This Nb has excellent production, stability and solubility characteristics. With this Nb we further manufactured a tandem linked bivalent construct and assembled a HCAb with improved antigen binding due to avidity effects. All these constructs were shown in mouse models to possess a scorpion toxin neutralization capacity that exceeds by far all previous attempts with scFv-based materials, even when used at lower doses. It is therefore clear that in the near future Nanobodies will be at the core of novel serotherapeutics as they combine multiple benefits over other reagents to treat scorpion envenomed patients.


Subject(s)
Antibodies/immunology , Camelus/immunology , Scorpion Venoms/chemistry , Scorpion Venoms/immunology , Scorpions , Animals , Antibodies/genetics , Humans , Neutralization Tests , Protein Structure, Tertiary/genetics , Recombinant Proteins/immunology , Scorpion Venoms/antagonists & inhibitors , Scorpion Venoms/isolation & purification
19.
Brief Funct Genomic Proteomic ; 7(4): 275-82, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18586755

ABSTRACT

Today's proteomic analyses are generating increasing numbers of biomarkers, making it essential to possess highly specific probes able to recognize those targets. Antibodies are considered to be the first choice as molecular recognition units due to their target specificity and affinity, which make them excellent probes in proteomics. In the post-genomic era and with high-throughput techniques available, the goal is to discriminate between all individual proteins from the proteome including their splice variants and post-translationally modified derivatives. Aided by advances in generation, selection and engineering of antibody-based recognition units, antibody fragments provide tools for detection of high- as well as low-abundant analytes even in complex, non-fractionated proteomes in conjunction with usage of small amounts of samples and reagents. In addition, large consortia aim at generating vast numbers of antibody-based recognition units suitable for future diagnostics and therapeutics.


Subject(s)
Antibodies/immunology , Oligonucleotide Array Sequence Analysis/methods , Protein Array Analysis/methods , Proteome/immunology , Proteomics/methods , Antigen-Antibody Complex , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Proteins/genetics , Proteins/immunology , Proteome/analysis
20.
Acta Trop ; 107(2): 139-44, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18589393

ABSTRACT

African trypanosomiasis is a severe parasitic disease affecting both man and livestock. It is crucial to expand our fundamental knowledge of the intimate interactions between trypanosomes and their vertebrate hosts in order to develop new and efficient control strategies. The mouse model of trypanosomiasis is the most popular for research purposes because of all the logistic advantages of using this species. Studies of any aspect of trypanosomiases in the mouse systematically require the quantification of some phenotypic traits which translate its degree of resistance/susceptibility to the disease, as blood cell counts. The present study presents a methodological approach combining everyday microsampling of tail blood and its analysis by flow cytometry. The technical options and conditions permitting a fast, reliable and reproducible daily quantification of erythrocyte, reticulocyte, leucocyte and trypanosome counts in the inoculated mouse were established. The protocol proposed allows the multiplication of blood samplings without being exposed to the time-consuming constraint of visual countings, without causing iatrogenic blood cell alterations in the mouse and without requiring specific anti-trypanosome antibodies.


Subject(s)
Flow Cytometry/methods , Parasitemia/blood , Parasitemia/parasitology , Trypanosoma/isolation & purification , Trypanosoma/pathogenicity , Trypanosomiasis/blood , Trypanosomiasis/parasitology , Animals , Blood Cell Count , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Specific Pathogen-Free Organisms , Trypanosoma/classification
SELECTION OF CITATIONS
SEARCH DETAIL
...