19 research outputs found

    Impact of Immunization Technology and Assay Application on Antibody Performance – A Systematic Comparative Evaluation

    Get PDF
    Antibodies are quintessential affinity reagents for the investigation and determination of a protein's expression patterns, localization, quantitation, modifications, purification, and functional understanding. Antibodies are typically used in techniques such as Western blot, immunohistochemistry (IHC), and enzyme-linked immunosorbent assays (ELISA), among others. The methods employed to generate antibodies can have a profound impact on their success in any of these applications. We raised antibodies against 10 serum proteins using 3 immunization methods: peptide antigens (3 per protein), DNA prime/protein fragment-boost (“DNA immunization”; 3 per protein), and full length protein. Antibodies thus generated were systematically evaluated using several different assay technologies (ELISA, IHC, and Western blot). Antibodies raised against peptides worked predominantly in applications where the target protein was denatured (57% success in Western blot, 66% success in immunohistochemistry), although 37% of the antibodies thus generated did not work in any of these applications. In contrast, antibodies produced by DNA immunization performed well against both denatured and native targets with a high level of success: 93% success in Western blots, 100% success in immunohistochemistry, and 79% success in ELISA. Importantly, success in one assay method was not predictive of success in another. Immunization with full length protein consistently yielded the best results; however, this method is not typically available for new targets, due to the difficulty of generating full length protein. We conclude that DNA immunization strategies which are not encumbered by the limitations of efficacy (peptides) or requirements for full length proteins can be quite successful, particularly when multiple constructs for each protein are used

    Prion protein-specific antibodies-development, modes of action and therapeutics application

    Get PDF
    Prion diseases or Transmissible Spongiform Encephalopathies (TSEs) are lethal neurodegenerative disorders involving the misfolding of the host encoded cellular prion protein, PrPC. This physiological form of the protein is expressed throughout the body, and it reaches the highest levels in the central nervous system where the pathology occurs. The conversion into the pathogenic isoform denoted as prion or PrPSc is the key event in prion disorders. Prominent candidates for the treatment of prion diseases are antibodies and their derivatives. Anti-PrPC antibodies are able to clear PrPSc from cell culture of infected cells. Furthermore, application of anti-PrPC antibodies suppresses prion replication in experimental animal models. Major drawbacks of immunotherapy are immune tolerance, the risks of neurotoxic side effects, limited ability of compounds to cross the blood-brain barrier and their unfavorable pharmacokinetic. The focus of this review is to recapitulate the current understanding of the molecular mechanisms for antibody mediated anti-prion activity. Although relevant for designing immunotherapeutic tools, the characterization of key antibody parameters shaping the molecular mechanism of the PrPC to PrPSc conversion remains elusive. Moreover, this review illustrates the various attempts towards the development of anti-PrP antibody compounds and discusses therapeutic candidates that modulate PrP expression

    Specific Binding of the Pathogenic Prion Isoform: Development and Characterization of a Humanized Single-Chain Variable Antibody Fragment

    Get PDF
    Murine monoclonal antibody V5B2 which specifically recognizes the pathogenic form of the prion protein represents a potentially valuable tool in diagnostics or therapy of prion diseases. As murine antibodies elicit immune response in human, only modified forms can be used for therapeutic applications. We humanized a single-chain V5B2 antibody using variable domain resurfacing approach guided by computer modelling. Design based on sequence alignments and computer modelling resulted in a humanized version bearing 13 mutations compared to initial murine scFv. The humanized scFv was expressed in a dedicated bacterial system and purified by metal-affinity chromatography. Unaltered binding affinity to the original antigen was demonstrated by ELISA and maintained binding specificity was proved by Western blotting and immunohistochemistry. Since monoclonal antibodies against prion protein can antagonize prion propagation, humanized scFv specific for the pathogenic form of the prion protein might become a potential therapeutic reagent

    APPORTS DE L'IMMUNISATION GENIQUE A L'OBTENTION D'ANTICORPS A VISEE THERAPEUTIQUE : VERS UNE IMMUNOTHERAPIE DES MALADIES A PRIONS

    Get PDF
    Prion diseases are fatal neurodegenerative disorders affecting many animal species, including humans. They are characterized by the accumulation, especially in the central nervous system, of an abnormal isoform of an ubiquist membrane protein, termed cellular prion protein (PrPc). There is to date no really efficient treatment for these diseases. During last years, both passive and active immunotherapy, based on the use of antibodies directed against the native form of PrPc and able to interfer with its transconformation into the pathogenic isoform, has appeared as a promising approach.To obtain antibodies for a possible use in passive immunotherapy in humans, we have developed several genetic immunization protocols, a technique generally recognized to allow the production of antibodies against the native form of the antigen, but which is often decribed as poorly efficient. The intramuscular electrotransfer of DNA coding for the human PrP allowed us, through a considerable increase of the in vivo expression of the protein, to raise in wild-type mice high amounts of polyclonal antibodies to native human PrP. These results open the way to the production of monoclonal antibodies, then to recombinant antibodies, under a format which remains to be defined in view of investigating and optimizing their potential therapeutic properties .In the context of an active immunotherapeutic approach (vaccination), we tried to overcome the immunological tolerance to endogenous PrPc in wild-type mice. The high levels of specific antibodies, raised through genetic immunization and electrotransfer of human PrP DNA, enabled us to demonstrate a clear cross-reactivity of these antibodies with native murine PrP.From a general point of view, our present findings may therefore significantly contribute in defining novel experimental conditions for effective passive and active immunotherapeutic strategies of several pathologies, including other neurodegenerative conformational disorders, e.g. Alzheimer disease, or some cancers characterized by an overexpression of cell membrane proteins.Les maladies à prions sont des maladies neurodégénératives fatales, affectant de nombreuses espèces animales, dont l'homme. Elles se caractérisent par l'accumulation, notamment dans le système nerveux central, d'une isoforme anormale d'une protéine membranaire ubiquiste, la protéine prion cellulaire (PrPc). Il n'existe à l'heure actuelle aucun traitement réellement efficace. Ces dernières années, l'immunothérapie, passive et active, basée sur l'utilisation d'anticorps dirigés contre la conformation native de la PrPc et capables de bloquer sa transconformation en forme pathogène, est apparue comme une méthode prometteuse.En vue d'obtenir des anticorps susceptibles d'être utilisés en immunothérapie passive chez l'homme, nous avons développé différents protocoles d'immunisation génique chez des souris de type sauvage, une technique reconnue pour favoriser la production d'anticorps dirigés contre la conformation native de l'antigène, mais souvent décrite comme peu efficace. L'injection intramusculaire par électrotransfert de l'ADNc codant la PrP humaine nous a permis d'obtenir, en augmentant fortement le niveau d'expression in vivo de la protéine, de fortes quantités d'anticorps polyclonaux contre la PrP humaine native. Ces résultats ouvrent la voie à la production d'anticorps monoclonaux puis recombinants, dans un format d'expression qu'il reste à définir pour tester et optimiser leur potentiel thérapeutique in vivo.Dans le cadre d'une approche par immunothérapie active (vaccination), nous avons essayé de rompre la tolérance immunitaire vis-à-vis de la PrPc endogène chez des souris de type sauvage. La potentialisation de la réponse en anticorps spécifiques, induite par l'électrotransfert in vivo de l'ADNc de la PrP humaine, nous a permis de mettre en évidence une réaction croisée de ces anticorps avec la PrPc murine.D'un point de vue général, nos résultats pourraient contribuer à ouvrir de nouvelles perspectives pour l'immunothérapie, passive et active, de nombreuses pathologies, en particulier d'autres maladies neurodégénératives, comme la maladie d'Alzheimer, ou certains cancers caractérisés par une surexpression de protéines membranaire

    Apports de l'immunisation génique à l'obtention d'anticorps à visée thérapeutique (vers une immunothérapie des maladies à prions)

    No full text
    LE KREMLIN-B.- PARIS 11-BU MĂ©d (940432101) / SudocPARIS-BIUP (751062107) / SudocSudocFranceF

    Curative properties of antibodies against prion protein: A comparative in vitro study of monovalent fragments and divalent antibodies

    No full text
    International audiencePrion diseases, which include Creutzfeldt-Jakob disease (CJD) in humans, are a group of devastating neurodegenerative disorders for which no therapy is yet available. However, passive immunotherapy appears to be a promising therapeutic approach, given that antibodies against the cellular prion protein (PrPc) have been shown in vitro to antagonize deposition of the disease-associated prion protein (PrPSc). Nevertheless, in vivo deleterious side effects of injected anti-PrP antibodies have been reported, mainly due to their Fc fragments and divalence. In this context, we examined here the ability of five Fabs (monovalent fragments devoid of the Fc part), prepared from antibodies already characterized in the laboratory, to inhibit prion replication in infected neuronal cells. We show that all Fabs (which all retain the same apparent affinity for PrPc as their whole antibody counterpart, as measured in EIA experiments) recognize quite well membrane bound-PrP in neuronal cells (as shown by flow cytometry analysis) and inhibit PrPSc formation in infected cells in a dose-dependent manner, most of them (four out of five) exhibiting a similar efficiency as whole antibodies. From a fundamental point of view, this report indicates that the in vitro curative effect of antibodies i) is epitope independent and only related to the efficiency of recognizing the native, membrane-inserted form of neuronal PrP and ii) probably occurs by directly or indirectly masking the PrPc epitopes involved in PrPSc interaction, rather than by cross-linking membrane bound PrPc. From a practical point of view, i.e. in the context of a possible immunotherapy of prion diseases, our data promote the use of monovalent antibodies (either Fabs or engineered recombinant fragments) for further in vivo studies

    Electrotransfer of cDNA Coding for a Heterologous Prion Protein Generates Autoantibodies Against Native Murine Prion Protein in Wild-Type Mice

    No full text
    International audiencePrion diseases (e.g., Creutzfeldt-Jakob disease in humans) are always fatal neurodegenerative disorders characterized by conversion of the ubiquitous cellular prion protein (PrP(c)) into a pathological conformer. Immunological strategies are considered as promising prophylactic or therapeutic approaches but, unfortunately, vaccination attempts until now have been very disappointing in wild-type animals because of immune tolerance to self PrP(c). Encouraging results have come from recent experiments carried out through genetic immunization (i.e., injection in mice of cDNA coding for murine prion protein [PrP]) or heterologous protein immunization (i.e., injection in mice of PrP from another species), albeit the levels of autoantibodies in wild-type animals remained generally low. Here we investigated whether combining the potential benefits of these two last approaches, namely using genetic immunization with the cDNA coding for a heterologous PrP, could more efficiently break immune tolerance. Wild-type mice were thus vaccinated with cDNA coding for human PrP(c), fused or unfused to a stimulatory T-cell epitope, using or not using electrotransfer of DNA. After three DNA injections, mice receiving electrotransferred DNA developed a strong immune response, oriented toward the humoral Th2 type, characterized not only by high IgG1 and IgG2a antibody titers against the heterologous human PrP(c), but also, as expected, by significant amounts of autoantibodies recognizing the native conformation of murine PrP(c) expressed on cell membranes as revealed by flow cytometry and immunofluorescence. These results hence open the way for investigation of the possible protective effects of anti-PrP(c) autoantibodies in infected mouse models. More generally, our results suggest that this original immunization strategy could be of value for circumventing tolerance to poorly immunogenic proteins

    Generating antibodies against the native form of the human prion protein (hPrP) in wild-type animals: A comparison between DNA and protein immunizations

    No full text
    International audienceGeneration of therapeutic antibodies against human proteins is hampered by the difficulty of obtaining large quantities of correctly folded immunogens when following classic immunization procedures. Here we compared several genetic immunization protocols for their potential ability to generate high levels of antibodies against proteins expressed in their native form. We chose as a model the prion protein (PrP) because it has been demonstrated that the recognition of the native conformation of PrP is an absolute prerequisite for anti-PrP antibodies to be used as therapeutic tools for prion diseases, a group of lethal neurodegenerative disorders. We designed two human PrP-DNA vectors, containing or not a stimulatory T cell epitope, which were injected into mice following four different protocols: in the naked form with or without electroporation, or protected by cationic polymers or block copolymers. For comparison, other animals received conventional injections of recombinant human PrP with Freund's adjuvant or alum. We found that genetic immunization, carried out especially through DNA electroporation and, to a lesser extent, through injection of block copolymer-protected DNA, was able to generate high amounts of antibodies recognizing native PrP as expressed on the cell surface. Conversely, protein immunizations led to very high levels of antibodies against PrP immobilized on microtiter plates, but unable to recognize the native cell membrane-bound PrP. This clearly demonstrates the usefulness of genetic immunization, when performed under well defined conditions, in raising antibodies to native proteins. These results are of interest not only in view of passive immunotherapy of prion diseases, but also, more generally, in view of generating antibodies to human membrane proteins for immunotherapeutic or immunodiagnostic purposes

    Immunological Determinants of Clinical Outcome in Peruvian Patients with Tegumentary Leishmaniasis Treated with Pentavalent Antimonials ▿ †

    No full text
    The mechanisms linking the immune response to cutaneous and mucosal leishmaniasis (CL and ML, respectively) lesions and the response to treatment are incompletely understood. Our aims were to prospectively assess, by quantitative reverse transcription-PCR, the levels of mRNA for gamma interferon, tumor necrosis factor alpha, interleukin-10 (IL-10), IL-4, and IL-13, as well as the presence of T cells (CD2) and macrophages (CD68), in CL and ML lesions and to follow their changes in response to treatment with pentavalent antimonials. The leishmanin skin test (LST) was performed on all CL and ML patients before treatment. The patient population included individuals living in areas of Peru where the disease is endemic, i.e., 129 with CL and 43 with ML. Compared to CL patients, the LST induration size was larger, the levels of all cytokine mRNAs but IL-10 were higher, T-cell mRNA was similar, and macrophage mRNA was lower in ML patients. The proportion of CL patients with an LST induration size of >8 mm was higher among responders to treatment. In CL, the pretreatment levels of cytokine mRNAs did not discriminate between responders and nonresponders; however, treatment was more often accompanied by a reduction in the levels of T-cell and cytokine mRNAs in responders than in nonresponders. Furthermore, the production of cytokines per T cell and macrophage decreased with treatment but IL-10 production remained high in nonresponders. Overall, these findings point to complex relationships among New World Leishmania parasites, skin and mucosal immune responses, and treatment outcome. The persistence of high levels of IL-10 in CL is characteristically associated with a poor response to treatment
    corecore