Elsevier

Methods

Volume 34, Issue 2, October 2004, Pages 171-178
Methods

Intracellular targeting and functional analysis of single-chain Fv fragments in mammalian cells

https://doi.org/10.1016/j.ymeth.2004.04.006Get rights and content

Abstract

In the past decade, intracellular antibodies have proven to be a useful tool in obtaining the phenotypic knock-out of selected gene function in different animal and plant systems. This strategy is based on the ectopic expression of recombinant forms of antibodies targeted towards different intracellular compartments, exploiting specific targeting signals to confer the new intracellular location. The functional basis of this technology is closely linked to the ability of intracellular antibodies to interact with their target antigens in vivo. This interaction allows either a direct neutralising effect or the dislodgement of the target protein from its normal intracellular location and, by this mechanism, the inactivation of its function. By using this approach, the function of several antigens has been inhibited in the cytoplasm, the nucleus, and the secretory compartments. In this article, we shall describe all the steps required for expressing single-chain Fv fragments in different subcellular compartments of mammalian cells and their subsequent use in knock-out experiments, starting from a cloned single-chain Fv fragment. This will include the analysis of the solubility properties of the new scFv fragment in transfected mammalian cells, the intracellular distribution of the antigen–antibody complex, and the resulting phenotype.

Introduction

Intracellular antibodies are a relatively new technology based on the concept that antibody chains or domains, if equipped with suitable localisation signals, can be targeted towards new ectopic intracellular sites to interfere with endogenous antigens [1], [2], [3], [4]. This can be obtained by exploiting the property of dominant and autonomous targeting sequences that can be grafted onto other reporter proteins such as antibodies to confer them a new intracellular localisation. Recombinant antibody domains, in particular single-chain Fv (scFv)1 fragments, have been successfully expressed inside cells to inhibit the function of several antigens in the cytoplasm [5], [6], the nucleus [7], [8], and the secretory pathway of mammalian cells [9]. This type of strategy has a great potential for a variety of applications, including gene therapy [10], plant biotechnology [11] and, more recently, functional genomics [4], [12], [26].

Crucial to the efficacy of this approach is the capacity of the intracellular antibody to interact with the endogenous antigen within any compartment of a mammalian cell. This interaction may result in a direct inhibition of the target antigen [6], [13], [14], may restore a mutant deficient activity [15] or may interfere with the protein folding of pathological mutant proteins [16]. In a different mode of action, an intracellular antibody can also act by diverting the intracellular traffic of the antigen. This is the case, for example, of membrane receptors, whose appearance at the surface can be inhibited by the interaction with intracellular antibodies retained in the endoplasmic reticulum by retention signals (intracellular anchors) [17], [18].

As it was clear from the very beginning of work on this technology, the folding properties of the antibodies and antibody domains vary according to individual scFv fragments and to the intracellular compartment where they are located. A systematic comparison of scFvs targeted to the same compartment showed that antibody fragments expressed from identical expression vectors have very distinct properties of solubility and stability [19], [20]. Although some scFvs are soluble in the cell cytoplasm, overexpression of these molecules may lead to the formation of intracellular aggregates. For this reason, there is great interest in engineering frameworks suitable for intracellular expression and onto which other specificities could be grafted. Different approaches have been developed to solve this problem, including modification of the sequence of VH and VL domains utilising random mutations to stabilise scFvs with intrinsic stability [21] or, alternatively, genetic selection approaches to derive functional scFvs, which can tolerate the reducing cellular environments of the cell cytoplasm [22], [23], [24].

Notwithstanding the phenomenon of aggregation, cytosolic scFv fragments maintain the capacity to bind the antigen and, by sequestering it in intracellular aggregates, to divert it from its intracellular location and block its function. A number of examples indicate that the antibody-mediated co-aggregation of the antigen represents a mode of action for intracellular antibodies targeted to the cytoplasm and to the nucleus [25], [26], [27], [28].

The purpose of this paper was to discuss certain aspects of the intracellular antibody technology that are worth considering in order to start a project with this powerful approach. In addition, we shall describe methods for the analysis of: (i) the expressed scFv fragments, (ii) in vivo interaction with the endogenous antigen, and (iii) the assessment of the resulting biological activity. Since, during the past decade, we have focussed our studies on the expression of scFv fragments in the cytoplasm and in the nucleus of mammalian cells, we shall provide examples and protocols for the analysis of antibody domains in these compartments. Reagents and methodological considerations related to each issue are described below in the context of our work on p21Ras and heterochromatin proteins 1 (HP1).

Section snippets

Intracellular targeting of scFvs

Starting from a cloned scFv fragment, a set of general vectors have been described for the expression of these fragments in different cellular compartments of mammalian cells [29]. All the plasmids contain an N- or C-terminal localisation signal that allows the correct retargeting of the scFv fragment. Table 1 shows the amino-acid sequences of some of the sorting peptides that have been successfully used so far for targeting antibodies or antibody domains (in particular scFv fragments) to

References (43)

  • S. Biocca et al.

    Trends Cell Biol.

    (1995)
  • J.H. Richardson et al.

    Trends Biotechnol.

    (1995)
  • S. Biocca et al.

    Biochem. Biophys. Res. Commun.

    (1993)
  • A. Cattaneo et al.

    Trends Biotechnol.

    (1999)
  • S. Jung et al.

    J. Mol. Biol.

    (1999)
  • M. Visintin et al.

    J. Mol. Biol.

    (2002)
  • E. Tse et al.

    J. Mol. Biol.

    (2002)
  • A. Cardinale et al.

    FEBS Lett.

    (1998)
  • L. Persic et al.

    Gene

    (1997)
  • J.D. Chesnut et al.

    J. Immunol. Methods

    (1996)
  • M.A. Hink et al.

    J. Biol. Chem.

    (2000)
  • G. Fenteany et al.

    J. Biol. Chem.

    (1998)
  • S. Biocca et al.

    EMBO J.

    (1990)
  • A. Cattaneo et al.

    Intracellular antibodies: development and application

    (1997)
  • S. Biocca et al.

    Bio/Technology

    (1994)
  • L. Duan et al.

    Proc. Natl. Acad. Sci. USA

    (1994)
  • A.M. Mhashilkar et al.

    EMBO J.

    (1995)
  • W.A. Marasco et al.

    Proc. Natl. Acad. Sci. USA

    (1993)
  • I.J. Rondon et al.

    Annu. Rev. Microbiol.

    (1997)
  • P. Tavladoraki et al.

    Nature

    (1993)
  • M. Visintin et al.

    Proc. Natl. Acad. Sci. USA

    (1999)
  • Cited by (21)

    • TRIM21 and the Function of Antibodies inside Cells

      2017, Trends in Immunology
      Citation Excerpt :

      Here, recombinant antibodies used to block or modulate the function of targeted proteins are expressed inside cells using DNA expression systems. Intracellular antibodies expressed as intrabodies commonly take the form of single chain Fv (scFv) comprising linked variable region heavy and light chains (VH and VL) but other formats are also used, including Fab fragments, diabodies, or single-chain VH fragments (also termed nanobodies) [26–28] (Figure 1). Intrabody technology has been used to identify a large number of antibodies capable of binding to cognate antigen inside cells, including therapeutically relevant targets in cancer, infectious disease, and neurological disorders [27,29–31].

    • Tracking protein-protein interaction and localization in living cells using a high-affinity molecular binder

      2016, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      A confocal microscopy image showed a high-level of expression and an even distribution of EGFP-repebody-B1 across the cytosol and nucleus in mammalian cells (Fig. 1D). It is worth noting that a negligible aggregation of EGFP-repebody-B1 was detected, whereas signs of aggregation of the scFv fragments were found [13]. A Western blot analysis of the total cell extract also supported the negligible aggregation of EGFP-repebody-B1 (Supplementary Fig. S5).

    • An intrabody specific for the nucleophosmin carboxy-terminal mutant and fused to a nuclear localization sequence binds its antigen but fails to relocate it in the nucleus

      2014, Biotechnology Reports
      Citation Excerpt :

      Consequently, any therapeutic perspective should re-establish this equilibrium rather than inactivate NPMc+ by means of neutralizing drugs. Intrabodies have been successfully used in the past to knock-out their targets or sequester their antigen in specific sub-cellular compartments [19–21]. Similarly, we isolated a scFv antibody specific for the de novo exclusive NES motif present in the mutated NPMc+, confirmed its correct folding when it was expressed as an intrabody, and fused it to a sequence corresponding to a repeat of nuclear localization signals (NLS).

    • The use of fluorescent intrabodies to detect endogenous gankyrin in living cancer cells

      2013, Experimental Cell Research
      Citation Excerpt :

      Fluorescent antibodies that bind specifically to the target may offer a means of studying endogenous proteins in living cells; however a technique to deliver these molecules to cells efficiently is still being developed [5]. The advances in recombinant antibodies technologies have allowed cloned antibody variable domains to be stably expressed in mammalian cells either as single-domain antibodies, such as camelid variable heavy chain domains (VHH;) [6,7], or as single chain Fv antibody fragments (scFvs) constituted by the assembly of the variable heavy (VH) and light (VL) chain connected by a flexible linker [8–13]. These so-called intrabodies can be easily fused to FPs to allow the recognized antigen to be imaged, for instance, by fluorescence microscopy [14–17].

    • The potential of intracellular antibodies for therapeutic targeting of protein-misfolding diseases

      2008, Trends in Molecular Medicine
      Citation Excerpt :

      This has been demonstrated not only for nuclear and cytoplasmic intrabodies but also for secretory intrabodies. In the first case, they tend to aggregate and coalesce into aggresome structures, whereas in the second case they are retrotranslocated from the ER, ubiquitinated and, finally, proteasome-degraded [22–24]. For clinical applications of intrabodies, the generation of human-derived antibody domains offers obvious potential advantages.

    View all citing articles on Scopus
    View full text