Research paper
Engineered cell surface expression of membrane immunoglobulin as a means to identify monoclonal antibody-secreting hybridomas

https://doi.org/10.1016/j.jim.2009.01.005Get rights and content

Abstract

Monoclonal antibodies (mAbs) have proven to be effective biological reagents in the form of therapeutic drugs and diagnostics for many pathologies, as well as valuable research tools. Existing methods for isolating mAb-producing hybridomas are tedious and time consuming. Herein we describe a novel system in which mAb-secreting hybridoma cells were induced to co-express significant amounts of the membrane form of the secreted immunoglobulin (Ig) on their surfaces and are efficiently recovered by fluorescent activated cell sorting (FACS). Fusion of a novel myeloma parent, SP2ab, expressing transgenic Igα and Igβ of the B-cell receptor complex (BCR) with spleen cells resulted in hybridomas demonstrating order of magnitude increases in BCR surface expression. Surface Ig levels correlated with transgenic Igα expression, and these cells also secreted normal levels of mAb. Hundreds of hybridoma lines producing mAbs specific for a variety of antigens were rapidly isolated as single cell-derived clones after FACS. Significant improvements using the Direct Selection of Hybridomas (DiSH) by FACS include reduced time and labor, improved capability of isolating positive hybridomas, and the ease of manipulating cloned cell lines relative to previously existing approaches that require Limiting Dilution Subcloning (LDS).

Introduction

Monoclonal antibodies (mAbs) are essential biotechnology reagents widely used in every phase of the biomedical field from discovery research and diagnosis to therapeutics. The power of mAb based therapeutics lies primarily in their specificity and strong affinity for a target antigen as well as their ability to mediate immune effector functions such as Antibody Dependent Cell Mediated Cytotoxicity and the Complement Cascade. The expansion of downstream technologies, most notably the humanization of mouse mAbs, and resultant low immunogenicity, have greatly expanded the therapeutic applications for mAbs (Milstein, 1999, Glennie and van de Winkel, 2003, Jakobovits et al., 2007). Consequently, there are 21 therapeutic mAb-based reagents currently approved for clinical use, five with annual sales in excess of $3B, and many others in various stages of clinical trials for a variety of diseases (Maggon, 2007) (www.fda.gov/cder/biologics/default.html, www.pipelinereview.com/index.php?option=com_performs&formid=7). In addition, mAbs account for a major market share in clinical and outpatient products and even over-the-counter diagnostics. However, current hybridoma-based mAb development technologies are cumbersome and confounding to the demands of high-throughput facilities. Further, alternative methodologies that avoid hybridomas (e.g. XenoMax and Phage Display), while offering certain advantages, carry burdens of expense and proprietary issues and have their own limitations (Marks et al., 1991, Babcook et al., 1996). Hence, we set out to develop new scientific and technical tools for rapid hybridoma detection and isolation.

Kohler and Milstein's (1975) seminal publication describes the generation and selection of hybridoma cells, heterokaryons resulting from the fusion of mouse B-lymphocytes and immortal myeloma cells, for the production of mAbs. The relevant hybridoma cells producing the mAb of choice are separated into individual clones using Limiting Dilution Subcloning (LDS). Recovering the hybridomas using cell cloning by LDS is perhaps the most problematic, time consuming, and labor-intensive step in generating mAbs (Antczak, 1982, O'Reilly et al., 1998). The fused hybridoma cells are deposited into a few thousand microtiter plate wells containing media supplemented with HAT (hypoxanthine, aminopterin, thymidine). HAT selects for hybridoma cells by killing unfused myeloma cells. The desired hybridomas are identified by screening for the reactivity of mAb secreted into the media using well-known methods such as an Enzyme-Linked Immunosorbent Assay (ELISA). Each HAT resistant cell population testing positive for secreted target mAb must be processed by reiterative cycles of LDS until the progeny of a positive cell is mathematically identified as clonal (Staszewski, 1984). Proposed solutions to this limitation including soft agar culture techniques (Draber et al., 1980), robotics to conduct the repeated cycles of LDS (Wewetzer and Seilheimer, 1995) and micro-encapsulation technologies that trap and assay the secreted Ab in the media around cells (Prokop et al., 2004, Hanania et al., 2005) have attempted to address the various weaknesses of LDS, but are expensive or offer little improvement in the efficiency.

The LDS process could be eliminated if all the desired hybridoma cells expressed the membrane Ig form of the secreted mAb. Cells could then be purified using Fluorescence Activated Cell Sorting (FACS). A few early attempts to use FACS for hybridoma cell cloning (Parks et al., 1979, Meilhoc et al., 1989), while promising, lacked efficiency because most hybridomas poorly express surface Ig (Matsuuchi et al., 1992, Seegmiller et al., 2007). Thus, the immediate objective of our research was to generate hybridomas that would consistently express membrane Ig on the cell surface and thereby facilitate efficient clonal selection by FACS.

We saw two potential obstacles to developing DiSH technology. The first of these was expression of the B-cell receptor subunit proteins Igα (CD79a, NP_031681) and Igβ (CD79b, NP_032365) necessary for assembly and trafficking of a functional BCR complex to the cell surface. Expression of membrane immunoglobulin on the surface of myeloma cells was obtained by transfecting lymphoid cells with cDNAs encoding the membrane isovariant of the antibody heavy chain (HCm) and the Igα and Igβ receptor proteins (Hombach et al., 1990). A diagram of the proposed natural arrangement of these proteins on the cell surface as they are positioned in the B-cell antigen receptor (BCR) complex is shown in Fig. 1A This experimental observation of engineered BCR complex presentation on the cell surface was extended to non-lymphoid cells using a pituitary cell line that is active in secretory functions, but normally would not synthesize the BCR nor express it on its cell surface (Matsuuchi et al., 1992). Once again, transgenic expression of the associated Igα and Igβ proteins, and light chain (LC) and HCm was sufficient to restore cell surface expression of the complete BCR. Thus, the first obstacle to developing hybridomas that efficiently express membrane immunoglobulin on their surface might be the limiting expression of Igα and Igβ proteins, and perhaps other accessory factors necessary for assembly and trafficking of a functional BCR complex.

The second potential obstacle to developing hybridomas that efficiently express membrane Ig might be a deficiency in the expression of the membrane isovariant of the Ig heavy chain (HCm). It is unclear if hybridoma cells express the longer HCm isovariant that is normally part of the BCR complex, along with the usual high levels of HCs, the secreted form. HCm contains the C-terminal peptide partly responsible for efficient insertion of the Ig molecule into the membrane. As B-cells develop into plasma cells an alteration in the post-transcriptional processing of the HC nuclear transcript (Fig. 1B) is reported to result in a switch from HCm to HCs expression. This switch plus an increase in the rate of transcription results in robust antibody (Ab) secretion (Milcarek and Hall, 1985, Genovese et al., 1991, Lassman et al., 1992). HCs protein lacking the membrane-spanning domains is thought to be unable to bind the associated receptor proteins, Igα and Igβ, and therefore is secreted. Thus, we explored HCm, Igα, and Igβ protein expression as possible barriers to obtaining high level BCR expression in hybridomas.

We found that Igα expression was the major limitation to the efficient expression of membrane-associated Ig for typical mAb secreting hybridoma cells. Furthermore, hybridomas derived from a myeloma parent engineered to over-express Igα displayed significant amounts of the BCR complex on the cell surface. Using this engineered myeloma, hundreds of hybridomas making Abs to four diverse antigens were rapidly and efficiently identified and recovered by FACS. DiSH technology greatly simplifies and accelerates mAb development and should help in developing targeted therapeutics and meet the ever-increasing demand for these valuable reagents.

Section snippets

Cell lines and growth conditions

The myeloma cell line SP2/0 (SP2/0-ag14, American Type Culture Collection #CRL-1581) or its transgenic derivative SP2ab was used to generate the hybridomas discussed herein. We also examined two well-characterized hybridoma cell lines, HGS1 (line 12G7) and HECS1 (line 16D8) (Li et al., 2001). SP2/0, SP2ab, and hybridoma cell lines were grown on Iscove's Modified Dulbecco's medium (IMDM, MediaTech, Herndon, VA) supplemented with 20% fetal bovine serum (FBS) (Atlanta Biologicals, Inc.,

Detecting the membrane isovariant of immunoglobulin heavy chain

Quantitative comparisons of mRNAs and mRNAm levels show that hybridomas still express reasonable mRNAm levels relative to immature and mature B-cells for at least one isotype, IgG2a (Matis et al., 1996, Milcarek et al., 1996, Edwalds-Gilbert et al., 1997). Therefore, we considered the possibility that there was sufficient HCm protein already produced by hybridomas for the potential expression of a BCR complex on the cell surface. The addition of the m1 and m2 domains to the heavy chain protein (

Discussion

The scientific goal of this research was to identify a molecular mechanism for producing hybridomas that expressed high levels of the membrane form of the secreted Ab in the BCR complex. The technical goal was to define a method for efficiently isolating hybridomas by capitalizing on the cell sorting and cloning power of FACS. From reconstruction experiments with an existing IgG1-producing hybridoma and from new hybridoma fusions made with the engineered SP2ab myeloma line, it was clear that

Conclusion

More and higher-affinity mAbs are needed for clinical research and newer, improved, faster and more efficient technologies are needed to keep pace with the ever increasing demand for mAbs for use as therapeutic, diagnostic, and research agents. This paper describes the development of one such technology for the rapid selection of hybridomas. We have shown that Igα receptor expression is all that limits strong surface expression of the BCR complex in hybridomas. The transgenic Igα and Igβ

Acknowledgements

We wish to thank Yolanda Lay, Gay Gragson, Lorraine Aron, Ruth Davis, Anthony Truong, Crystal Jackson, Huizhong Chen, Julie Nelson, and Aissa Villareal-Singh for their advice and assistance. Dennis Bagarrozzi generously supplied the samples of influenza virus and Colm Condon supplied an anti-Ig-alpha polyclonal antibody. This work was supported by funds from the University of Georgia's Research Foundation; the Georgia Research Alliance; Abeome Corporation (Athens, GA); and grants to RBM

References (52)

  • WewetzerK. et al.

    Establishment of a single-step hybridoma cloning protocol using an automated cell transfer system: comparison with limiting dilution

    J. Immunol. Methods

    (1995)
  • AntczakD.F.

    Monoclonal antibodies: technology and potential use

    J. Am. Vet. Med. Assoc.

    (1982)
  • BabcookJ.S. et al.

    A novel strategy for generating monoclonal antibodies from single, isolated lymphocytes producing antibodies of defined specificities

    Proc. Natl. Acad. Sci. U. S. A.

    (1996)
  • CairncrossJ.G. et al.

    Cell surface antigens of human astrocytoma defined by mouse monoclonal antibodies: identification of astrocytoma subsets

    Proc. Natl. Acad. Sci. U. S. A.

    (1982)
  • CondonC. et al.

    Aberrant trafficking of the B cell receptor Ig-alpha beta subunit in a B lymphoma cell line

    J. Immunol.

    (2000)
  • DraberP. et al.

    Establishment and characterization of permanent murine hybridomas secreting monoclonal anti-thy-1 antibodies

    J. Immunogenet.

    (1980)
  • Edwalds-GilbertG. et al.

    Alternative poly(A) site selection in complex transcription units: means to an end?

    Nucleic Acids Res.

    (1997)
  • FrankelA.E. et al.

    Human prostate specific and shared differentiation antigens defined by monoclonal antibodies

    Proc. Natl. Acad. Sci. U. S. A.

    (1982)
  • Fuentes-PananaE.M. et al.

    Ig alpha/Ig beta complexes generate signals for B cell development independent of selective plasma membrane compartmentalization

    J. Immunol.

    (2005)
  • GenoveseC. et al.

    Differential mRNA stabilities affect mRNA levels in mutant mouse myeloma cells

    Somat. Cell Mol. Genet.

    (1991)
  • HananiaE.G. et al.

    Automated in situ measurement of cell-specific antibody secretion and laser-mediated purification for rapid cloning of highly-secreting producers

    Biotechnol. Bioeng.

    (2005)
  • HancockR.E. et al.

    Monoclonal antibodies against Pseudomonas aeruginosa outer membrane antigens: isolation and characterization

    Infect. Immun.

    (1982)
  • HombachJ. et al.

    Identification of the genes encoding the IgM-alpha and Ig-beta components of the IgM antigen receptor complex by amino-terminal sequencing

    Eur. J. Immunol.

    (1990)
  • JahnS. et al.

    Cell biology of human IgM-producing hybridomas derived from a fusion of human spleen lymphocytes with mouse myeloma cells

    Hybridoma

    (1987)
  • JakobovitsA. et al.

    From XenoMouse technology to panitumumab, the first fully human antibody product from transgenic mice

    Nat. Biotechnol.

    (2007)
  • Cited by (29)

    • High-throughput single-cell antibody secretion quantification and enrichment using droplet microfluidics-based FRET assay

      2022, iScience
      Citation Excerpt :

      Nonetheless, our results are consistent with previous reports showing that mouse hybridoma cells simultaneously express both the membrane-bound and secreted forms of immunoglobulins. The ratio between the two fractions may depend on alternative RNA splicing (Helman et al., 2014; Liu et al., 2015; Pogson et al., 2016), physiological cell state (Charlet et al., 1995; Corrêa et al., 2016; Listek et al., 2020), transcriptional variability (Price et al., 2009; Shi et al., 2015), and cell cycle (Charlet et al., 1995; Kromenaker and Srienc, 1991). Having confirmed that individual hybridoma cells isolated in 40 pl volume droplets produce sufficient quantities of antibodies for detection by FRET, we next sought to enrich the highest mAb producers from a heterogeneous mixture using a microfluidic FRET-activated droplet sorter (Figures 3, S1B, and Data S3).

    • Rapid and reliable hybridoma screening method that is suitable for production of functional structure-recognizing monoclonal antibody

      2021, Journal of Bioscience and Bioengineering
      Citation Excerpt :

      A method of screening clones that produce antigen-specific antibodies after cell fusion is an FCM-based methodology that enables the selection of a single cell from a bulk mixture of fused hybridoma cells. The method does not require special reagents, is easy to set up, and can be applied in many different types of laboratories (8,11–14). By advancing both methods (SST and MIHS), it will be possible to obtain a variety of monoclonal antibodies suitable for specific applications.

    • Combination of phage and Gram-positive bacterial display of human antibody repertoires enables isolation of functional high affinity binders

      2018, New Biotechnology
      Citation Excerpt :

      Phage display [7,8] is currently the most widely used selection platform, its success mainly owing to its ability to conveniently harbour large libraries together with ease of handling. Other display formats include ribosome display [9,10] and cell based display platforms such as E. coli [11], yeast [12] and mammalian display [13]. The use of cell-based platforms offers several advantages compared to the capture and elution procedure of phage and ribosome display.

    • Flow cytometry-based method for rapid and high-throughput screening of hybridoma cells secreting monoclonal antibody

      2018, Journal of Bioscience and Bioengineering
      Citation Excerpt :

      However, the fluorescence intensity that they used was not strong enough for the separation of labeled and fused cells by FCM of those days, thus returning only modest efficiency to get the desired hybridoma. In 2006, Price et al. (8) improved and patent-protected the method to achieve high cloning efficiency using genetically engineered myeloma to obtain abundant mIg expression. The overexpressed mIg gave stronger fluorescence intensities which made the selection of labeled cells using FCM easier.

    • Comparative characterization of mAb producing hapten-specific hybridoma cells by flow cytometric analysis and ELISA

      2014, Journal of Immunological Methods
      Citation Excerpt :

      The antigen-specific labeling of hybridoma cells has been rarely described, with the exception of Parks et al. (1979), who used antigen-specific microspheres. In another context genetically modified hybridoma cells were sorted antigen-specifically via a streptavidin/biotin system (Price et al., 2009). However, the antigen-specific detection of hapten-specific antibodies on the surface of B cells was reported before (McHeyzer-Williams et al., 1991; Lalor et al., 1992; McHeyzer-Williams et al., 2000).

    View all citing articles on Scopus
    View full text