Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Ther ; 31(3): 788-800, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36575794

ABSTRACT

The COVID-19 pandemic and the need for additional safe, effective, and affordable vaccines gave new impetus into development of vaccine genetic platforms. Here we report the findings from the phase 1, first-in-human, dose-escalation study of COVID-eVax, a DNA vaccine encoding the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. Sixty-eight healthy adults received two doses of 0.5, 1, or 2 mg 28 days apart, or a single 2-mg dose, via intramuscular injection followed by electroporation, and they were monitored for 6 months. All participants completed the primary safety and immunogenicity assessments after 8 weeks. COVID-eVax was well tolerated, with mainly mild to moderate solicited adverse events (tenderness, pain, bruising, headache, and malaise/fatigue), less frequent after the second dose, and it induced an immune response (binding antibodies and/or T cells) at all prime-boost doses tested in up to 90% of the volunteers at the highest dose. However, the vaccine did not induce neutralizing antibodies, while particularly relevant was the T cell-mediated immunity, with a robust Th1 response. This T cell-skewed immunological response adds significant information to the DNA vaccine platform and should be assessed in further studies for its protective capacity and potential usefulness also in other therapeutic areas, such as oncology.


Subject(s)
COVID-19 , Vaccines, DNA , Adult , Humans , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Double-Blind Method , Pandemics/prevention & control , SARS-CoV-2 , Vaccines, DNA/adverse effects
2.
RSC Adv ; 8(16): 8638-8656, 2018 Feb 23.
Article in English | MEDLINE | ID: mdl-35539867

ABSTRACT

A small series of Morita-Baylis-Hillman adduct (MBHA) derivatives was synthesized and made to react with imidazole, N-acetylhistidine, and N-acetylhexahistidine as models of poly-histidine derivatives. Intriguingly, the reaction of MBHA derivatives 1a and b with imidazole in acetonitrile-phosphate buffered saline (PBS) gave the imidazolium salt biadducts 3a and b as the main reaction products. These results were confirmed by experiments performed with N-acetylhistidine and 1b and suggested the possible occurrence of these structures in the products of poly-histidine labeling with MBHA derivatives 1a and b. These compounds were then transformed into the corresponding water-soluble derivatives 1c-e by introducing oligo(ethylene glycol) chains and their reactivity was evaluated in preliminary experiments with imidazole and then with N-acetylhexahistidine in PBS. The structure of polymeric materials Ac-His-6-MBHA-1d and Ac-His-6-MBHA-1e obtained using ten-fold excesses of compounds 1d and e was investigated using mass spectrometry, NMR spectroscopy, and photophysical studies, which suggested the presence of biadduct residues in both polymeric materials. These results provide the basis for the preparation of fishbone-like polymer brushes, the characterization of their properties, and the exploration of their potential applications in different fields of science such as in vivo fluorogenic labeling, fluorescence microscopy, protein PEGylation, up to the production of smart materials and biosensors.

3.
ACS Omega ; 2(9): 5453-5459, 2017 Sep 30.
Article in English | MEDLINE | ID: mdl-31457813

ABSTRACT

In order to obtain new fluorophores potentially useful in imidazole labeling and subsequent conjugation, a small series of Morita-Baylis-Hillman acetates (3a-c) was designed, synthesized, and reacted with imidazole. The optical properties of the corresponding imidazole derivatives 4a-c were analyzed both in solution and in the solid state. Although the solutions display a very weak emission, the powders show a blue emission, particularly enhanced in the case of compound 4c possessing two methoxy groups in the cinnamic scaffold. The photophysical study confirmed the hypothesis that the molecular rigidity of the solid state enhances the emission properties of these compounds by triggering the restriction of intramolecular motions, paving the way for their applications in fluorogenic labeling.

4.
Oncotarget ; 7(13): 15539-53, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26788990

ABSTRACT

Single-chain variable fragments (scFvs) expressed as "intracellular antibodies" (intrabodies) can target intracellular antigens to hamper their function efficaciously and specifically. Here we use an intrabody targeting the E6 oncoprotein of Human papillomavirus 16 (HPV16) to address the issue of a non-invasive therapy for HPV cancer patients.A scFv against the HPV16 E6 was selected by Intracellular Antibody Capture Technology and expressed as I7nuc in the nucleus of HPV16-positive SiHa, HPV-negative C33A and 293T cells. Colocalization of I7nuc and recombinant E6 was observed in different cell compartments, obtaining evidence of E6 delocalization ascribable to I7nuc. In SiHa cells, I7nuc expressed by pLNCX retroviral vector was able to partially inhibit degradation of the main E6 target p53, and induced p53 accumulation in nucleus. When analyzing in vitro activity on cell proliferation and survival, I7nuc was able to decrease growth inducing late apoptosis and necrosis of SiHa cells.Finally, I7nuc antitumor activity was demonstrated in two pre-clinical models of HPV tumors. C57BL/6 mice were injected subcutaneously with HPV16-positive TC-1 or C3 tumor cells, infected with pLNCX retroviral vector expressing or non-expressing I7nuc. All the mice injected with I7nuc-expressing cells showed a clear delay in tumor onset; 60% and 40% of mice receiving TC-1 and C3 cells, respectively, remained tumor-free for 17 weeks of follow-up, whereas 100% of the controls were tumor-bearing 20 days post-inoculum. Our data support the therapeutic potential of E6-targeted I7nuc against HPV tumors.


Subject(s)
Immunotherapy/methods , Neoplasms, Experimental/virology , Oncogene Proteins, Viral/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Single-Chain Antibodies/pharmacology , Animals , Cell Line, Tumor , Heterografts , Humans , Mice , Mice, Inbred C57BL , Papillomavirus Infections/complications
5.
Arch Biochem Biophys ; 522(1): 26-36, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22516657

ABSTRACT

proNGF, the precursor of the neurotrophin NGF, is widely expressed in central and peripheral nervous system. Its physiological functions are still largely unknown, although it emerged from studies in the last decade that proNGF has additional and distinct functions with respect to NGF, besides acting chaperone-like for NGF folding during its biogenesis. The regulation of proNGF/NGF ratio represents a crucial process for homeostasis of brain and other tissues, and understanding the molecular aspects of these differences is important. We report the selection and characterization of a recombinant monoclonal anti-proNGF antibody in single chain Fv fragment (scFv) format. The selection exploited the Intracellular Antibody Capture Technology (IACT), starting from a naïve mouse SPLINT (Single Pot Library of INTracellular antibodies) library. This antibody (scFv FPro10) was expressed recombinantly in Escherichia coli, was proven to be highly soluble and stable, and thoroughly characterized from the biochemical-biophysical point of view. scFv FPro10 displays high affinity and specificity for proNGF, showing no cross-reactivity with other pro-neurotrophins. A structural model was obtained by SAXS. scFv FPro10 represents a new tool to be exploited for the selective immunoanalysis of proNGF, both in vitro and in vivo, and might help in understanding the molecular function of proNGF in neurodegeneration.


Subject(s)
Antibodies, Monoclonal/immunology , Nerve Growth Factor/immunology , Protein Precursors/immunology , Single-Chain Antibodies/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibody Affinity , Base Sequence , Circular Dichroism , Cross Reactions , DNA Primers , Enzyme-Linked Immunosorbent Assay , Humans , Isoelectric Focusing , Mice , Recombinant Proteins/metabolism , Scattering, Small Angle , Single-Chain Antibodies/metabolism , Spectrometry, Fluorescence , Surface Plasmon Resonance , X-Ray Diffraction
6.
J Mol Biol ; 387(3): 584-606, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19361429

ABSTRACT

The development of conformation-sensitive antibody domains targeting the misfolding beta amyloid (Abeta) peptide is of great interest for research into Alzheimer's disease (AD). We describe the direct selection, by the Intracellular Antibody Capture Technology (IACT), of a panel of anti-Abeta single chain Fv antibody fragments (scFvs), targeting pathologically relevant conformations of Abeta. A LexA-Abeta1-42 fusion protein was expressed in yeast cells, as the "intracellular antigen". Two different scFv antibody libraries (Single Pot Libraries of Intracellular Antibodies, SPLINT) were used for the intracellular selections: (i) a naïve library, derived from a natural, non-immune, source of mouse antibody variable region (V) genes; and (ii) an immune library constructed from the repertoire of antibody V genes of Abeta-immunized mice. This led to the isolation of 18 different anti-Abeta scFvs, which bind Abeta both in the yeast cell, as well as in vitro, if used as purified recombinant proteins. Surprisingly, all the anti-Abeta scFvs isolated are conformation-sensitive, showing a high degree of specificity towards Abeta oligomers with respect to monomeric Abeta, while also displaying some degree of sequence-specificity, recognizing either the N-terminal or the C-terminal part of Abeta1-42; in particular, the scFvs selected from Abeta-immune SPLINT library show a relevant N-terminal epitope bias. Representative candidates from this panel of the anti-Abeta scFvs were shown to recognize in vivo-produced Abeta "deposits" in histological sections from human AD brains and to display good neutralization properties, significantly inhibiting Abeta oligomer-induced toxicity and synaptic binding of Abeta oligomers in neuronal cultured cells. The properties of these anti-Abeta antibody domains, as well as their direct availability for intra- or extra-cellular "genetic delivery" make them ideally suited for new experimental approaches to study and image the intracellular processing and trafficking of Abeta oligomers.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/chemistry , Antibodies/chemistry , Antibodies/immunology , Protein Conformation , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/immunology , Animals , Antibodies/genetics , Cells, Cultured , Epitopes/chemistry , Humans , Immunoassay/methods , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neurons/cytology , Neurons/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Library , Phylogeny , Protein Binding , Protein Multimerization , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sequence Analysis, DNA , Synapses/metabolism , Two-Hybrid System Techniques
7.
J Biotechnol ; 135(1): 1-15, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18395925

ABSTRACT

Protein-protein interactions represent a major potential drug target for many human diseases, but these are unanimously considered undruggable with small chemical molecules. We have developed 3-SPLINT, a novel technology for the selection of antibodies that are intrinsically endowed with the ability to interfere with a given protein-protein interaction. The selection procedure exploits the recently described yeast SPLINT libraries of intrabodies, adapting them to a reverse-hybrid system, yielding the selection of recombinant antibodies that are able to disrupt a target protein-protein interaction in vivo. This class of antibodies should therefore perturb an individual protein-protein interaction, without perturbing the scaffolding function of the target protein in that complex, or other protein interactions of that same protein. We provide here a proof of concept of the technology, by the de novo selection of antibodies against two distinct interacting protein pairs: the GABARAP, which interact with the gamma2 subunit of GABA(A) receptor, and the p65 protein dimer, involved in the NF-kappaB-mediated signalling transduction pathway. Intrabodies selected against the latter were functionally validated in cells. Such antibodies, by interfering with the dimerization domain of p65, lead to an activation of the NF-kappaB-mediated transcriptional activity, which is normally inhibited by p65 knock-down RNAi. This provides a clear-cut demonstration that interfering with a protein interaction can be functionally very different from physically removing one of the interacting proteins. The 3-SPLINT approach provides a general and finer tool for the functional validation of selected protein interactions in protein networks, and is ideally applied to protein "hubs", displaying multiple distinct interactions. 3-SPLINT will therefore complement RNAi-based approaches, in the toolkit of target validation strategies, and is amenable to the systematic isolation of comprehensive sets of antibodies against most protein-protein interactions of a given protein network.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Drug Delivery Systems/methods , Peptide Library , Protein Engineering/methods , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/immunology , Protein Interaction Mapping , Antibodies, Monoclonal/metabolism
8.
J Mol Neurosci ; 34(2): 141-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18008186

ABSTRACT

The microtubule-binding protein gephyrin is known to play a pivotal role in targeting and clustering postsynaptic inhibitory receptors. Here, the Intracellular Antibodies Capture Technology (IATC) was used to select two single-chain antibody fragments or intrabodies, which, fused to nuclear localization signals (NLS), were able to efficiently and selectively remove gephyrin from glycine receptor (GlyR) clusters. Co-transfection of NLS-tagged individual intrabodies with gephyrin-enhanced green fluorescent protein (EGFP) in HEK 293 cells revealed a partial relocalization of gephyrin aggregates onto the nucleus or in the perinuclear area. When expressed in cultured neurons, these intrabodies caused a significant reduction in the number of immunoreactive GlyR clusters, which was associated with a decrease in the peak amplitude of glycine-evoked whole cell currents as assessed with electrophysiological experiments. Hampering protein function at a posttranslational level may represent an attractive alternative for interfering with gephyrin function in a more spatially localized manner.


Subject(s)
Carrier Proteins/metabolism , Immunoglobulin Fragments/metabolism , Membrane Proteins/metabolism , Receptors, Glycine/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Cells, Cultured , Excitatory Amino Acid Agonists/metabolism , Glycine/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoglobulin Fragments/genetics , Neurons/cytology , Neurons/metabolism , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Patch-Clamp Techniques , Rats , Rats, Wistar , Recombinant Fusion Proteins/genetics , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
9.
J Immunol Methods ; 301(1-2): 31-40, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15907924

ABSTRACT

Intracellular antibodies or intrabodies have great potential in protein knockout strategies for intracellular antigens. We applied the Intracellular Antibody Capture Technology for the direct selection in yeast of a mouse scFv library (V(L)-V(H) format) constructed from animals immunised with recombinant non-structural protein NSP5 of Rotavirus. We selected five different intracellular antibodies (ICAbs), which specifically recognize Delta2, an NSP5 deletion mutant used as bait. The anti-NSP5 ICAbs were well expressed both in yeast and mammalian cells as cytoplasmic or nuclear-tagged forms. By immunofluorescence and co-immunoprecipitation assays we characterised the intracellular interaction of the five anti-NSP5 ICAbs with the co-expressed antigens.


Subject(s)
Antibodies/genetics , Antibodies/immunology , Antibody Specificity , Gene Library , Viral Nonstructural Proteins/immunology , Amino Acid Sequence , Animals , Antibodies/chemistry , Antigens, Viral/immunology , Cell Line , Genetic Vectors/genetics , Mice , Microscopy, Confocal , Molecular Sequence Data , Sequence Alignment , Spleen/immunology , Spleen/metabolism
10.
J Gen Virol ; 85(Pt 11): 3285-3290, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15483242

ABSTRACT

Intracellular antibodies or intrabodies (ICAbs) have great potential in protein knockout strategies for intracellular antigens. In this study, they have been used to investigate the role of the rotavirus non-structural protein NSP5 in the virus replication cycle. Intracellular antibody-capture technology was used to select single-chain Fv format (scFv) ICAbs against an NSP5 mutant. Five different specific ICAbs were selected and expressed in MA104 cells, in the scFv format, as cytoplasmic- and nuclear-tagged forms. By confocal microscopy, it was found that three of these ICAbs recognized the full-length wild-type NSP5 specifically, forming antigen-specific aggresomes in the cytoplasm of cotransfected cells. Expression of the ICAbs in rotavirus-infected cells largely reduced the assembly of viroplasms and cellular cytopathic effect. Replication of dsRNA was partially inhibited, despite there being no reduction in virus titre. These results demonstrate for the first time a key role for NSP5 during the virus replicative cycle.


Subject(s)
Antibodies, Viral/biosynthesis , Intracellular Space/virology , Rotavirus/physiology , Viral Nonstructural Proteins/immunology , Virus Replication/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Cell Line , Immunoglobulin Fragments/biosynthesis , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Intracellular Space/immunology , Microscopy, Confocal , RNA, Viral/antagonists & inhibitors , RNA, Viral/biosynthesis , Recombinant Proteins/biosynthesis , Rotavirus/genetics , Rotavirus/metabolism , Transfection
11.
Methods ; 34(2): 200-14, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15312673

ABSTRACT

Several approaches have been developed over the past decade to study the complex interactions that occur in biological system. The ability to carry out a comprehensive genetic analysis of an organism becomes more limited and difficult as the complexity of the organism increases because complex organisms are likely to have not only more genes than simple organisms but also more elaborate networks of interactions among those genes. The development of technologies to systematically disrupt protein networks at the genomic scale would greatly accelerate the comprehensive understanding of the cell as molecular machinery. Intracellular antibodies (intrabodies) can be targeted to different intracellular compartments to specifically interfere with function of selected intracellular gene products in mammalian cells. This technique should prove important for studies of mammalian cells, where genetic approaches are more difficult. In the context of large-scale protein interaction mapping projects, intracellular antibodies (ICAbs) promise to be an important tool to knocking out protein function inside the cell. In this context, however, the need for speed and high throughput requires the development of simple and robust methods to derive antibodies which function within cells, without the need for optimization of each individual ICAb. The successful inhibition of biological processes by intrabodies has been demonstrated in a number of different cells. The performance of antibodies that are intracellularly expressed is, however, somewhat unpredictable, because the reducing environment of the cell cytoplasm in which they are forced to work prevents some antibodies, but not others, to fold properly. For this reason, we have developed an in vivo selection procedure named Intracellular Antibody Capture Technology (IACT) that allows the isolation of functional intrabodies. The IAC technology has been used for the rapid identification of antigen-antibody pairs in intracellular compartments and for the in vivo identification of epitopes recognized by the selected intracellular antibodies. Several optimizations of the IAC technology for protein knock-out have been developed so far. This system offers a powerful and versatile proteomic tool to dissect diverse functional properties of cellular proteins in different cell lines.


Subject(s)
Antibodies/chemistry , Antibodies/physiology , Drug Delivery Systems/methods , Intracellular Fluid/chemistry , Intracellular Fluid/immunology , Animals , Antibodies/genetics , Genetic Vectors , Humans , Intracellular Fluid/drug effects
12.
J Immunol Methods ; 290(1-2): 135-53, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15261577

ABSTRACT

The intracellular antibody technology has many applications for proteomics studies. The potential of intracellular antibodies for the systematic study of the proteome has been made possible by the development of new experimental strategies that allow the selection of antibodies under conditions of intracellular expression. The Intracellular Antibody Capture Technology (IACT) is an in vivo two-hybrid-based method originally developed for the selection of antibodies readily folded for ectopic expression. IACT has been used for the rapid and effective identification of novel antigen-antibody pairs in intracellular compartments and for the in vivo identification of epitopes recognized by selected intracellular antibodies. IACT opens the way to the use of intracellular antibody technology for large-scale applications in proteomics. In its present format, its use is however somewhat limited by the need of a preselection of the input phage antibody libraries on protein antigens or by the construction of an antibody library from mice immunized against the target protein(s), to provide an enriched input library to compensate for the suboptimal efficiency of transformation of the yeast cells. These enrichment steps require expressing the corresponding proteins, which represents a severe bottleneck for the scaling up of the technology. We describe here the construction of a single pot library of intracellular antibodies (SPLINT), a naïve library of scFv fragments expressed directly in the yeast cytoplasm in a format such that antigen-specific intrabodies can be isolated directly from gene sequences, with no manipulation whatsoever of the corresponding proteins. We describe also the isolation from SPLINT of a panel of intrabodies against a number of different proteins. The application of SPLINT on a genome-wide scale should help the systematic study of the functional organization of cell proteome.


Subject(s)
Immunoglobulin Variable Region/immunology , Peptide Library , Proteomics/methods , Genetic Vectors , Genomics/methods , Plasmids
13.
J Immunol ; 169(5): 2274-83, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12193692

ABSTRACT

Intracellular expression of Ab fragments has been efficiently used to inactivate therapeutic targets, oncogene products, and to induce viral resistance in plants. Ab fragments expressed in the appropriate cell compartment may also help to elucidate the functions of a protein of interest. We report in this study the successful targeting of the protein tyrosine kinase Syk in the RBL-2H3 rat basophilic leukemia cell line. We isolated from a phage display library human single-chain variable fragments (scFv) directed against the portion of Syk containing the Src homology 2 domains and the linker region that separates them. Among them, two scFv named G4G11 and G4E4 exhibited the best binding to Syk in vivo in a yeast two-hybrid selection system. Stable transfectants of RBL-2H3 cells expressing cytosolic G4G11 and G4E4 were established. Immunoprecipitation experiments showed that intracellular G4G11 and G4E4 bind to Syk, but do not inhibit the activation of Syk following FcepsilonRI aggregation, suggesting that the scFv do not affect the recruitment of Syk to the receptor. Nevertheless, FcepsilonRI-mediated calcium mobilization and the release of inflammatory mediators are inhibited, and are consistent with a defect in Bruton's tyrosine kinase and phospholipase C-gamma2 tyrosine phosphorylation and activation. Interestingly, FcepsilonRI-induced mitogen-activated protein kinase phosphorylation is not altered, suggesting that intracellular G4G11 and G4E4 do not prevent the coupling of Syk to the Ras pathway, but they selectively inhibit the pathway involving phospholipase C-gamma2 activation.


Subject(s)
Enzyme Precursors/immunology , Immunoglobulin Variable Region/metabolism , Intracellular Fluid/immunology , Protein-Tyrosine Kinases/immunology , Receptors, IgE/antagonists & inhibitors , Receptors, IgE/physiology , Signal Transduction/immunology , Tumor Cells, Cultured/immunology , src Homology Domains/immunology , Agammaglobulinaemia Tyrosine Kinase , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Binding Sites, Antibody , Calcium/antagonists & inhibitors , Calcium/metabolism , Enzyme Activation/genetics , Enzyme Activation/immunology , Enzyme Precursors/metabolism , Humans , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/pharmacology , Intracellular Fluid/metabolism , Intracellular Signaling Peptides and Proteins , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Molecular Sequence Data , Phospholipase C gamma , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rats , Signal Transduction/genetics , Syk Kinase , Transfection , Tumor Cells, Cultured/metabolism , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Tyrosine/antagonists & inhibitors , Tyrosine/metabolism , src-Family Kinases/metabolism
14.
J Mol Biol ; 317(1): 73-83, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11916379

ABSTRACT

We describe the application of an intracellular antibody capture technology (IACT) as a generic in vivo selection procedure for isolating intracellular antibodies or ICAbs. IACT was applied to the de novo selection of functional ICAbs against the microtubule-associated protein TAU, found in neurofibrillary lesions of Alzheimer's disease brains. A panel of 17 different ICAbs was created which bind TAU inside cells and the epitopes recognized by the selected ICAbs have been determined by an in vivo epitope mapping procedure. Finally, sequence analysis showed that the IACT-derived ICAbs are characterized by a common signature of conserved amino acid residues, suggesting that the IACT naturally selects a sort of "captured consensus sequence" for intracellular antibodies. The development of IACT, together with the possibility of scaling up in a high throughput and automated format, makes IACT a new enabling tool for target validation in functional genomics and global proteomics.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Antibody Specificity/genetics , Antibody Specificity/immunology , Cloning, Molecular/methods , Consensus Sequence , Intracellular Fluid/immunology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Amino Acid Sequence , Animals , Antibodies/genetics , Automation , CHO Cells , COS Cells , Chromatography, Gel , Consensus Sequence/genetics , Cricetinae , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Fibroblasts , Fluorescent Antibody Technique , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Peptide Library , Reproducibility of Results , Sequence Alignment , tau Proteins/genetics , tau Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...