GET THE APP

ER Intrabodies: Potent Molecules for Specific Knockdown of Protei
Journal of Cell Science & Therapy

Journal of Cell Science & Therapy
Open Access

ISSN: 2157-7013

+44 1300 500008

Short Communication - (2015) Volume 6, Issue 4

ER Intrabodies: Potent Molecules for Specific Knockdown of Proteins Passing the ER

Thomas Boldicke*
Helmholtz Centre for Infection Research, Department Structural and Functional Protein Research, Germany
*Corresponding Author: Phd Thomas Boldicke, Helmholtz Centre for Infection Research, Department Structural and Functional Protein Research, Inhoffenstr. 7, 38124 Braunschweig, Germany, Tel: 0531-6181-5050 Email:

Letter to Editor

Today the function of proteins can be analyzed by nucleotide based functional genomics for example siRNA [1] or protein knockdown approaches. Knockdown approaches based on direct interference of the target protein with the inhibitor results in detailed insights of protein functions not obtainable with genetic methods.

A very promising protein knockdown technique is based on recombinant antibody fragments expressed inside the ER (ER intrabodies). ER intrabodies mediate inhibition of the function of proteins passing the ER very efficiently and specifically. Many ER intrabodies against a large range of attractive targets have been described [2,3]. Targets include oncogenic receptors, virus proteins (to prevent virus assembly), cellular virus receptors (to block virus entry), proteins of the immune system and the nervous system [4-8]. Even ER intrabodies with therapeutic potential have been generated [9-15].

Intrabodies are recombinant antibody fragments, mainly constructed in the scFv or Fab format. They are produced inside target cells expressing the corresponding antigens. Intrabodies can be targeted to the nucleus, ER or mitochondrium by fusion of an appropriate signal sequence to the N-terminus of the antibody coding sequence or expressed without any signal sequence inside the cytoplasm [16].

ER intrabodies retain proteins passing the ER via a sequence encoding the short retention peptide KDEL which is fused to the Cterminus of the intrabody gene [17]. The inhibition of the translocation of the antigen from the ER to the cell compartment where it normally acts leads to inhibition of antigen function. The method is suitable to retain the transport of secretory molecules, cell surface proteins or proteins which are active in the Golgi or endosomal compartment [2,13,18]. Even an intrabody without retention sequence inactivated the ribosomal P proteins of Trypanosoma cruzi [19].

Due to the high specificity of antibodies, this technology is an attractive alternative to RNAi/miRNA based methods and small molecule inhibitor molecules such as receptor tyrosine kinases inhibitors, which often show off target effects [20-23].

An ER intrabody just requires efficient binding to its antigen to retain it inside the ER. In contrast, a cytosolic antibody must inactivate its target, or interfere with the binding of the target protein to its corresponding binding partner. In contrast to ER intrabodies which are correctly folded in the ER, cytosolic intrabodies are often not correctly folded due to the fact that disulfide bridges are not formed in this environment. Consequently, a considerable effort is required to select stable cytosolic intrabodies [24]. Today the most used methods for selection of cytosolic intrabodies are the Intracellular Antibody Capture Technology based on an antigen-dependent two hybrid system [25] and construction of single domain antibodies derived from camels [26] or shark which are very stable expressed in the cytoplasm [27-29].

The antigen specific scFv antibody fragment for ER intrabody generation can be selected from (1) a hybridoma clone [30] or (2) from in vitro display systems such as phage display or yeast display antibody repertoires [31,32] (Figure 1).

cell-science-therapy-establishment

Figure 1: Scheme of ER intrabody construction and establishment of transgenic intrabody mouse.

The advantages of ER intrabodies are

• Very high specificity to the antigen

• In vitro display techniques open the door for constructing ER intrabodies with a single cloning step.

• Splice variants can be targeted

• Post translational modifications can be analyzed

• Isoforms of a protein can be inactivated by one intrabody

• Transgenic intrabody mice can be generated

The number of ER intrabodies will grow much more faster in the future compared to the last twenty years, as (1) thousands of new scFv antibody fragments have been generated by phage display and yeast display and the corresponding V region genes are available for construction of new ER intrabodies, and (2) optimized consensus primer mixes, RACE and adapter-ligated RT-PCR enables reliable isolation of antibody variable domains from hybridoma clones today.

Until now most ER intrabodies have been tested in vitro. In addition some ER intrabodies have been applied in xenograft tumor mouse models [9,33], in an Alzheimer’s disease mouse model [34] and in an human papillomavirus (HPV)-associated tumor mouse model [35]. Recently two transgenic intrabody mice has been generated showing significant knock down of VCAM and gesolin in vivo [36,37]. The VCAM intrabody mouse expressed the intrabody as scFv fragment and the gesolin intrabody mouse as VHH antibody. Interestingly, the transgenic VCAM intrabody mouse was viable in contrast to the lethal knock out counterpart [38]. In addition an anti- EVH1 intrabody has been investigated in vivo in a transgenic intrabody mouse [39]. However this report has been questioned because the intrabody was targeted to the secretory pathway but interacted with a cytosolic protein [40].

The results obtained from the VCAM and gesolin intrabody transgenic intrabody mice pave the way to new fascinating possibilities: for example the possibility to establish transgenic intrabody mice with inducible or tissue specific ER intrabody expression which can further be regulated by different exogenous promoters.

Very interesting would be therapeutic clinical approaches of ER intrabodies [15,16]. Problems not yet solved in gene therapeutic appraches is the need of safety viral or efficient non-viral transfection systems. Construction of retrovirus with low safety risk, of transductional and transcriptional targeting vectors for cell-specific gene transfer and the use of mRNA might be promising tools for successful gene therapy in the future [41-43].

In summary, ER intrabodies are very potent and specific molecules to study the function of proteins in vitro and in vivo. Particularly the availability of variable antibody domains selected from phage display libraries simplifies the generation of new ER intrabodies for a more broad application in functional analysis of proteins.

(A) Shows a scFv fragment, which will be converted into an ER intrabody by cloning it in one step into ER targeting vector (D) for in vitro characterization of ER intrabody or into Embronic Stem Cell vector for generation of transgenic mice (E). The scFv comprises the variable domain of the heavy chain (VH) and light chain (VL) linked together by a synthetic 15 (Gly4Ser)3 amino acid linker. CDR1, CDR2, CDR3, complementarity determing regions.

(B) scFv’s can be selected using universal antibody phage display libraries which comprises the complete genes of a man’s antibody repertoire. Seen is a typically filamentous M13 antibody phage with the antibody gene inside the phage and the corresponding protein fused to P3 on the surface of the phage. In a process called biopanning the recombinant antibody phages are incubated with antigen and after washing, specific phages can be eluted and the antibody genes further amplified in E.coli.

(C) scFv’s can also be constructed from the variable domains of a hybridoma clone. Therefore the variable domains of the heavy and light chain will be amplified from the cDNA by PCR. In general the variable genes are amplified with consensus primer. If it is not possible to amplify less common non-consensus antibody sequences, two other methods are available: Rapid amplification of cDNA ends (RACE, [44] and adapter ligated PCR [45]. Assembly of the variable domains with the synthetic linker by PCR will result in the scFv.

(D) The gene of the scFv will be cloned into ER targeting vector providing a secretion signal at the N-terminus and a peptide affinity tag and the retention sequence KDEL at the C-terminus of the antibody coding sequence.

(E) After cloning the retention of the corresponding cell surface antigen inside the ER can be estimated by FACS. Shown is as an example of the inhibition of Neural Cell Adhesion Molecule (NCAM) cell surface expression by anti-NCAM ER intrabodies. TE671cells (NCAM+, shaded area) and TE671 cells expressing anti-NCAM intrabodies (bold line) incubated with anti-NCAM antibodies and secondary anti-mouse phycoerythrin conjugated antibodies. TE671 cells expressing ER intrabodies against NCAM stained with secondary antibody only (thin lane).

(F) To generate transgenic mice the coding sequence of the antibody will be cloned into an Embryonic stem cell (ES) vector and the vector transfected into ES cells. Then recombinant ES cells will be used for the generation of transgenic intrabody mice. The intrabody gene can be oriented in reverse orientation and flanked by reverse LoxP sites. To express the intrabody gene, the established intrabody mouse will be crossed with a mouse expressing the cre recombinase (Cre mouse [46]). The cre recombinase inverts the intrabody gene at the two inverse DNA recognition sites (LoxP sites), thereby the intrabody gene is activated.

(G) Transgenic intrabody mice can be established with constitutive intrabody expression or inducible or tissues specific intrabody expression [46].

References

  1. Dorsett Y, Tuschl T (2004) siRNAs: applications in functional genomics and potential as therapeutics. Nat Rev Drug Discov 3: 318-329.
  2. Boldicke T (2007) Blocking translocation of cell surface molecules from the ER to the cell surface by intracellular antibodies targeted to the ER. J Cell Mol Med 11: 54-70.
  3. Lo AS, Zhu Q, Marasco WA (2008) Intracellular antibodies (intrabodies) and their therapeutic potential. Handb Exp Pharmacol : 343-373.
  4. Wheeler YY, Kute TE, Willingham MC, Chen SY, Sane DC (2003) Intrabody-based strategies for inhibition of vascular endothelial growth factor receptor-2: effects on apoptosis, cell growth, and angiogenesis. FASEB J 17: 1733-1735.
  5. Liao W, Strube RW, Milne RW, Chen SY, Chan L (2008) Cloning of apoB intrabodies: specific knockdown of apoB in HepG2 cells. Biochem Biophys Res Commun 373: 235-240.
  6. Steinberger P, Andris-Widhopf J, Buhler B, Torbett BE, Barbas CF (2000) 3rd. Functional deletion of the CCR5 receptor by intracellular immunization produces cells that are refractory to CCR5-dependent HIV-1 infection and cell fusion. Proc Natl Acad Sci 97: 805-810.
  7. Intasai N, Tragoolpua K, Pingmuang P, Khunkaewla P, Moonsom S, et al. (2008) Potent inhibition of OKT3-induced T cell proliferation and suppression of CD147 cell surface expression in HeLa cells by scFv-M6-1B9. Immunobiology 214: 410-421.
  8. Zhang C, Helmsing S, Zagrebelsky M, Schirrmann T, Marschall AL, et al. (2012) Suppression of p75 neurotrophin receptor surface expression with intrabodies influences Bcl-xL mRNA expression and neurite outgrowth in PC12 cells. PLoS One 7: e30684.
  9. Popkov M, Jendreyko N, McGavern DB, Rader C, Barbas CF 3rd (2005) Targeting tumor angiogenesis with adenovirus-delivered anti-Tie-2 intrabody. Cancer Res 65: 972-981.
  10. Jannot CB, Beerli RR, Mason S, Gullick WJ, Hynes NE (1996) Intracellular expression of a single-chain antibody directed to the EGFR leads to growth inhibition of tumor cells. Oncogene 13: 275-282.
  11. Boldicke T, Weber H, Mueller PP, Barleon B, Bernal M (2005) Novel highly efficient intrabody mediates complete inhibition of cell surface expression of the human vascular endothelial growth factor receptor- (VEGFR-2/KDR). J Immunol Methods 300: 146-159.
  12. Kirschning CJ, Dreher S, Maass B, Fichte S, Schade J, et al. (2010) Generation of anti-TLR2 intrabody mediating inhibition of macrophage surface TLR2 expression and TLR2-driven cell activation. BMC Biotechnol 10: 31.
  13. Reimer E, Somplatzki S, Zegenhagen D, Hänel S, Fels A, et al. (2013) Molecular cloning and characterization of a novel anti-TLR9 intrabody. Cell Mol Biol Lett 18: 433-446.
  14. Serruys B, Van Houtte F, Farhoudi-Moghadam A, Leroux-Roels G, Vanlandschoot P (2010) Production, characterization and in vitro testing of HBcAg-specific VHH intrabodies. J Gen Virol 91: 643-652.
  15. Cardinale A, Biocca S (2013) Gene-based antibody strategies for prion diseases. Int J Cell Biol 2013: 710406.
  16. Lobato MN, Rabbitts TH (2003) Intracellular antibodies and challenges facing their use as therapeutic agents. Trends Mol Med 9: 390-396.
  17. Munro S, Pelham HR (1987) A C-terminal signal prevents secretion of luminal ER proteins. Cell 48: 899-907.
  18. Zehner M, Marschall AL, Bos E, Schloetel JG, Kreer C, et al. (2015) The translocon protein Sec61 mediates antigen transport from endosomes in the cytosol for cross-presentation to CD8(+) T cells. Immunity 42: 850-863.
  19. Ayub MJ, Nyambega B, Simonetti L, Duffy T, Longhi SA, et al. (2012) Selective blockade of trypanosomatid protein synthesis by a recombinant antibody anti-Trypanosoma cruzi P2β protein. PLoS One 7: e36233.
  20. Davies SP, Reddy H, Caivano M, Cohen P (2000) Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J 351: 95-105.
  21. Jackson AL, Linsley PS (2010) Recognizing and avoiding siRNA off-target effects for target identification and therapeutic application. Nat Rev Drug Discov 9: 57-67.
  22. Snøve O Jr, Holen T (2004) Many commonly used siRNAs risk off-target activity. Biochem Biophys Res Commun 319: 256-263.
  23. Cao T, Heng BC (2005) Intracellular antibodies (intrabodies) versus RNA interference for therapeutic applications. Ann Clin Lab Sci 35: 227-229.
  24. Warn A, Auf der Maur A, Escher D, Honegger A, Barberis A, et al. (2000) Correlation between in vitro stability and in vivo performance of anti-GCN4 intrabodies as cytoplasmic inhibitors. J Biol Chem 275: 2795-2803.
  25. Visintin M, Melchionna T, Cannistraci I, Cattaneo A (2008) In vivo selection of intrabodies specifically targeting protein-protein interactions: a general platform for an "undruggable" class of disease targets. J Biotechnol 135: 1-15.
  26. Muyldermans S (2013) Nanobodies: natural single-domain antibodies. Annu Rev Biochem 82: 775-797.
  27. Staus DP, Wingler LM, Strachan RT, Rasmussen SG, Pardon E, et al. (2014) Regulation of β2-adrenergic receptor function by conformationally selective single-domain intrabodies. Mol Pharmacol 85: 472-481.
  28. Van Impe K, Bethuyne J, Cool S, Impens F, Ruano-Gallego D, et al. (2013) A nanobody targeting the F-actin capping protein CapG restrains breast cancer metastasis. Breast Cancer Res 15: R116.
  29. Liu JL, Zabetakis D, Brown JC, Anderson GP, Goldman ER (2014) Thermal stability and refolding capability of shark derived single domain antibodies. Mol Immunol 59: 194-199.
  30. Böldicke T, Somplatzki S, Sergeev G, Mueller PP (2012) Functional inhibition of transitory proteins by intrabody-mediated retention in the endoplasmatic reticulum. Methods 56: 338-350.
  31. Bradbury AR, Sidhu S, Dübel S, McCafferty J (2011) Beyond natural antibodies: the power of in vitro display technologies. Nat Biotechnol 29: 245-254.
  32. Schofield DJ, Pope AR, Clementel V, Buckell J, Chapple SDj, et al. (2007) Application of phage display to high throughput antibody generation and characterization. Genome Biol 8: R254.
  33. Jendreyko N, Popkov M, Rader C, Barbas CF 3rd (2005) Phenotypic knockout of VEGF-R2 and Tie-2 with an intradiabody reduces tumor growth and angiogenesis in vivo. Proc Natl Acad Sci U S A 102: 8293-8298.
  34. Sudol KL, Mastrangelo MA, Narrow WC, Frazer ME, Levites YR, et al. (2009) Generating differentially targeted amyloid-beta specific intrabodies as a passive vaccination strategy for Alzheimer's disease. Mol Ther 17: 2031-2040.
  35. Accardi L, Paolini F, Mandarino A, Percario Z, Di Bonito P, et al. (2014) In vivo antitumor effect of an intracellular single-chain antibody fragment against the E7 oncoprotein of human papillomavirus 16. Int J Cancer 134: 2742-2747.
  36. Marschall AL, Single FN, Schlarmann K, Bosio A, Strebe N, et al. (2014) Functional knock down of VCAM1 in mice mediated by endoplasmatic reticulum retained intrabodies. MAbs 6: 1394-1401.
  37. Van Overbeke W, Wongsantichon J, Everaert I, Verhelle A, Zwaenepoel O, et al. (2015) An ER-directed gelsolin nanobody targets the first step in amyloid formation in a gelsolin amyloidosis mouse model. Hum Mol Genet 24: 2492-2507.
  38. Kwee L, Baldwin HS, Shen HM, Stewart CL, Buck C, et al. (1995) Defective development of the embryonic and extraembryonic circulatory systems in vascular cell adhesion molecule (VCAM-1) deficient mice. Development 121: 489-503.
  39. Sato M, Iwaya R, Ogihara K, Sawahata R, Kitani H, et al. (2005) Intrabodies against the EVH1 domain of Wiskott-Aldrich syndrome protein inhibit T cell receptor signaling in transgenic mice T cells. FEBS J 272: 6131-6144.
  40. Cardinale A, Biocca S (2009) Can intrabodies targeted to the secretory compartment interact with a cytosolic protein? Exp Mol Pathol. 86: 138.
  41. Papayannakos C, Daniel R (2013) Understanding lentiviral vector chromatin targeting: working to reduce insertional mutagenic potential for gene therapy. Gene Ther 20: 581-588.
  42. Waehler R, Russell SJ, Curiel DT (2007) Engineering targeted viral vectors for gene therapy. Nat Rev Genet 8: 573-587.
  43. Dolgin E (2015) Business: The billion-dollar biotech. Nature 522: 26-28.
  44. Ruberti F, Cattaneo A, Bradbury A (1994) The use of the RACE method to clone hybridoma cDNA when V region primers fail. J Immunol Methods 173: 33-39.
  45. Ladiges W, Osman GE (2000) Molecular characterization of immunoglobulin genes, Basic Methods in Antibody Production and Characterization. CRC Press Ltd, Bocuraton, Florida 169-191
  46. Lewandoski M (2001) Conditional control of gene expression in the mouse. Nat Rev Genet 2: 743-755.
Citation: Boldicke T (2015) ER Intrabodies: Potent Molecules for Specific Knockdown of Proteins Passing the ER. J Cell Sci Ther 6:214.

Copyright: ©2015 Boldicke T, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top