Antibody Structure and Antibody Fragments
Mary Johnson (han at labome dot com)
Synatom Research, Princeton, New Jersey, United States
DOI
//dx.doi.org/10.13070/mm.en.3.160
Date
last modified : 2022-11-05; original version : 2013-12-21
Cite as
MATER METHODS 2013;3:160
Abstract

Here we present an in-depth description of antibody structure and various antibody fragments. We discuss major applications of antibodies as well as advantages and disadvantages of using full-size antibodies versus fragments. We then provide an overview of studies using antibody fragments and a discussion of antibody receptors called Fc receptors.

Conventional Antibodies

Antibodies are immunoglobulin (Ig) glycoproteins produced by plasma cells (B cells) in response to foreign antigenic molecules (immunogens). The primary function of antibodies is to bind specifically to these foreign antigens to disable them and/or mark them for destruction by the immune system, thereby protecting the host from infection. There are several classes of antibodies. The first portion of this summary focuses on conventional full-size antibodies, the IgG and IgM class antibodies, which are heavily utilized in a multitude of research, diagnostic and therapeutic biomedical applications.

Conventional antibody structure

The basic unit of a conventional antibody is a four polypeptide unit consisting of two identical heavy chains and two identical light chains held together by disulfide bonds. The light chains are shorter, with lower molecular weights than the heavy chains. The general shape of an antibody is a Y, with a flexible hinge (interdomain) region at the center of the Y. The flexibility of the interdomain hinge region is important for the bivalent binding of an antibody [2], allowing the two binding pockets to interact with antigenic sites at variable distances. Each polypeptide chain has a constant region, which does not vary significantly among antibodies, and a variable region, which is specific to each particular antibody. The common notation for the light chain variable region is VL and for the light chain constant region is CL (Figure 1). The notation is similar for the heavy chain variable (VH) and constant regions (CH) with CH1, CH2, and CH3 denoting the different constant region domains of the heavy chain. Carbohydrates are normally attached to the CH2 domains of the heavy chains. The fragment crystallizable (Fc) region contains only constant regions from the heavy chains (CH), but the fragment antigen-binding region (Fab) includes both a constant domain and the variable domains of both the heavy and light chains (VH and CL). The fragment variable region FV region contains only the two variable domains (Figure 1). See mouse antibody for discussion on immunoglobulin isotypes, subclasses, and the number of immunoglobulin domains.

Antibody Structure and Antibody Fragments figure 1
Figure 1. The basic structure of a conventional full size antibody (left panel) and of common antibody fragments (right panel). From [1]

Each complete antibody has two antigen-binding pockets, located in the FV regions, and can bind to two antigens (bivalent binding). However, if the two antigens are too close (≤3 nm), or too far apart (≥29nm), the antibody can only bind to one antigen (monovalent binding) [2]. There is a significant affinity change between monovalent and bivalent bindings with a 1,500-fold change in Kd values [2].

The structure of specific antibodies is available from the Structural Antibody Database (SAbDab), a curated database of publicly available antibody structures, as well as structural modelling tools, which is updated weekly from the Protein Data Bank (PDB) [3].

Conventional antibody applications

Conventional full-size antibodies have been utilized in research for protein detection via Western blot analyses [4], immunohistochemistry [5], and enzyme-linked immunosorbent assays (ELISA) [6] for decades. Full-size antibodies have also been developed for diagnostic applications such as pregnancy tests and detection of bacteria and viruses in blood, such as an ELISA that detects HIV. Additionally, conventional full-size antibodies are used commonly in disease therapeutics. For example, Infliximab is one antibody of many available that recognize tumor necrosis factor alpha (TNFα) and it is used in the treatment of Crohn's disease and rheumatoid arthritis [7, 8]. Trastuzumab, or Herceptin, is an antibody that binds to epidermal growth factor receptor 2 and is used in the treatment of metastatic breast cancer [9]. There are also several antibody-based therapies, including Muromonab [10], given to transplant recipients to prevent allograft rejection.

Though conventional full-size antibodies may both be used for therapeutic applications, there are advantages and disadvantages to using the complete antibody. An important advantage of conventional antibodies is the fact the Fc region engages the body's immune response, and can target bound antigens for destruction. This Fc region can also be a disadvantage in some clinical applications because the immune response that it typically elicits may be detrimental to the patient's health. Additionally, full-size antibodies cannot penetrate well into certain tissues due to their relatively large size [11]. In some cases, when using full-size antibodies for diagnostic applications the Fc domain can cause significant nonspecific binding, which may impair detection applications.

Antibody Fragments

For many applications antibody fragments are preferable. Antibody fragments can be produced through chemical or genetic mechanisms. Chemical fragmentation utilizes reducing agents to break the disulfide bonds within the hinge region and digestion of the antibody with proteases including pepsin, papain, and ficin. Genetic construction of fragments offers the ability to create a multitude of fragment-containing molecules, each with unique binding and functional characteristics.

Fab, Fab', (Fab')2, and FV

Chemical and protease digestion of full-size IgG or IgM antibodies yield antigen-binding fragments (Fab; Figure 1), and Fc fragments, comprised only of the heavy chain CH2, and CH3 domains. Biochemical methods of generating antibody fragments produce useful tools for diagnostic and therapeutic applications, but it is quite laborious and requires a large quantity of antibody starting material.

The antigen-binding fragments produced by biochemical digestion include Fab, (Fab')2, Fab', and FV, all of which lack the Fc region. Monovalent F(ab) fragments have one antigen-binding site, whereas divalent (Fab')2 fragments have two antigen-binding regions that are linked by disulfide bonds. Two individual F(ab) fragments are produced when a full-size antibody is digested with papain enzyme. A F(ab')2 fragment, which retains a portion of the hinge region, is produced by pepsin digestion of IgG or IgM antibodies. Reduction of F(ab')2 fragments produces 2 monovalent Fab' fragments, which have a free sulfhydryl group that is useful for conjugation to other molecules. FV fragments are the smallest fragment made from enzymatic cleavage of IgG and IgM class antibodies (Figure 1). FV fragments have the antigen-binding site made of the VH and VL regions, but they lack the constant regions of Fab (CH1 and CL) regions (Figure 1, right panel). The VH and VL are held together in FV fragments by non-covalent interactions. The fragments can be generated through commercially available kits, for example, F(ab')2 Fragmentation Kit from G-Biosciences [12].

scFv, diabody, triabody, tetrabody, Bis-scFv, minibody, Fab2, Fab3

Genetic engineering methods allow the production of single chain variable fragments (scFv), which are FV type fragments that consist of the VH and VL domains linked by an engineered flexible linker peptide (Figure 1) [13]. Manipulation of the orientation of the V-domains and the linker length creates different forms of FV molecules [14]. When the linker is at least 12 residues long, the scFv fragments are primarily monomeric (as shown in Figure 1) [14]. Linkers that are 3-11 residues long yield scFv molecules that are unable to fold into a functional FV domain. These molecules associate with a second scFv molecule, which creates a bivalent diabody [15]. If the linker length is less than three residues, scFv molecules associate into triabodies or tetrabodies [14]. For example, Tao Y et al generated a VH-VL diabody with a short GGGGS linker and scFv with a long GTTAASGSSGGSSSGA linker [16]. Multivalent scFvs possess greater functional binding affinity to their target antigens as a result of having two more target antigen binding sites, which reduces the off-rate of the antibody fragment [17]. Minibodies are scFv-CH3 fusion proteins that assemble into bivalent dimers [18]. Small scFv fragments with two different variable domains can be generated to produce bispecific bis-scFv fragments capable of binding two different epitopes concurrently [19, 20]. Genetic methods are also used to create bispecific Fab dimers (Fab2) and trispecific Fab trimers (Fab3) [20]. These antibody fragments are capable of simultaneously binding 2 or 3 different antigens, respectively. Researchers often use scFv fragments to stabilize protein complexes in protein structure studies [21].

Camelid/shark antibodies and nanobodies

In addition to conventional antibodies, camelid and shark (squalidae) species contain a subset of peculiar Heavy Chain Antibodies (hcAb) exclusively composed by heavy chain homodimers lacking light chains [22, 23]. The Fab portions of these antibodies, called VHH in camelids and VNAR in sharks, are the smallest antigen-binding regions naturally found [24]. Nanobodies are VHH-derived recombinant domains able to bind antigens, often cloned from VHH phage libraries such as those against betacoronarivus S proteins [25] or against SARS-CoV-2 spike protein RBD domain [26]. Their binding thermodynamics and structures have been studied ( [27] and reference therein). Nanobodies are very stable and can be easily produced in huge quantity by using common simple protein expression systems such as bacteria (functional conventional full-size antibodies are difficult to express properly in a bacterial system), thus representing a promising tool for research and therapeutic purposes, especially in the areas of super-resolution microscopy, mass spectrometry, and targeted protein degradation [28]. Nanobodies can also be delivered inside living cells through conjugated with peptides [29, 30], or in vivo [31], or expressed directly in vivo and recognize its targets in vivo, for example, VHH Z70 against tau nucleation [32]. Nanobodies against RFP or GFP, when conjugated with far-red Atto dyes, attained 118-fold magnification of fluorescent signals over GFP or RFP, and were used to generate whole-body mouse neuronal connectivity [33]. They have also been used to stabilize the active state of proteins in structural studies [34]. An expression system with multiple concatenated nanobodies against different influenza strains is being examined as a means to generate a universal flu vaccine [35]. Recombinant anti-IgG secondary nanobodies have great potential to replace widely used polyclonal secondary antibodies produced using animals [36].

Nanobodies have the unique ability to cross the blood-brain barrier [37, 38] ; however, nanobodies tend to be processed and cleared very quickly from the body [39]. Nanobodies can be used for methods like immunoprecipitation, for example, RFP-Trap MA from Chromotek [40], or coupled to fluorescent proteins to track intracellular targets in live cells in real-time [41].

Cow ultralong CDR3H

About 10% of bovine immunoglobulins contain an unusually long third heavy-chain complementarity-determining region (CDR 3H) with a large number of cysteine residues [42-44]. These cysteines pair to form disulfide bonds which leads to a stalk-and-knob like structure in the antigen binding domain [44]. This exceptionally long CDR3H domain with the long-stalk contributes to the diversity of bovine antibody specificity.

Intrabodies

Intrabodies, or intracellular antibodies, refer to antibodies or their fragments (usually of scFv design) expressed directly inside cells or in animals in vivo through an expression vector. For example, Dong JX et al developed several nanobodies against neuronal proteins for intracellular expression as intrabodies [45]. One important consideration/caveat is the reducing intracellular environment which diminishes the affinity of an antibody or antibody fragment whose binding with the antigen is dependent on intradomain disulfide bonds. Another consideration is that intrabodies tend to aggregate. Kabayama H et al designed intrabodies with a net engative charge even at the lowest cytoplasmic pH 6.6 to generate ultra-stable cytoplasmic antibodies [46]. Intrabodies are used as alternatives to pharmacological inhibitors to target specific endocytic participants. For instances, expression of a single-chain variable fragment (scFv) derived from the 3B12A monoclonal antibody against the TDP-43 nuclear export signal in HEK293A cells or after in utero electroporation of its expression vector promoted the proteolysis of TDP-43 aggregates in cultured cells and embryonic mouse brain [47]. Virus-mediated delivery into the nervous system of an scFv antibody against the RNA recognition motif 1 of TDP-43 reduced microgliosis in a mouse model of acute neuroinflammation and mitigated cognitive impairment, motor defects, TDP-43 proteinopathy, and neuroinflammation in transgenic mice expressing TDP-43 mutations linked to amyotrophic lateral sclerosis [48]. The potential of intrabodies as a therapeutical modality remains to emerge.

An antibody fragment can also be linked with a cell-import tag, such as an IPTD tag [49], to facilitate its entry into cells.

Antibody fragment applications

Antibody fragments offer certain advantages over a full-size antibody for some applications. This topic was reviewed by Nelson [50]. One advantage of fragments over full-size antibodies is that antibody fragments are smaller than conventional antibodies and generally lack glycosylation, allowing their production in prokaryotic expression systems, which provide time and cost savings. Additionally, fragments are small enough to infiltrate into some tissues that full-size antibodies are unable to penetrate, which aids in many therapeutic and immunohistochemical procedures [11]. Furthermore, the lack of Fc domain is a substantial advantage for primary antibodies used in immunohistochemistry and other detection applications because they have greatly reduced non-specific binding to the Fc receptor. One scFv that is commonly used in diagnostics is the NC10 antibody against influenza neuraminidase. The MOC-31 antibody against epithelial cell adhesion molecule Ep-CAM is an scFv commonly used in as a cancer therapy. Diabodies, triabodies and tetrabodies have potential uses in applications such as radioimmunotherapy and diagnostic in vivo imaging [51]. However, fragments that lack the Fc domain are degraded in the body much more rapidly than conventional antibodies [52], and are unable to elicit Fc-mediated cytotoxic processes unless they are conjugated to an effector moiety [53], which requires further optimization for antibody fragment-based therapeutics.

Although various antibody fragments offer certain advantages, they are not commonly utilized in experiments. In the more than 60,000 articles manually curated by Labome only a few articles cited applications of antibody Fab fragments. The Roche anti-digoxigenin Fab antibody fragments ( 11093274910) and anti-fluorescein Fab antibody fragments ( 11426338910) are generated in sheep and produced through digestion with papain. Anti-digoxigenin Fab fragments were used for in situ hybridizations since the RNA probes are labeled with digoxigenin [54-56]. They were also used to perform protein folding analysis to study the process of single calmodulin molecule folding through single-molecule force spectroscopy [57]. F(ab) fragments of anti-mouse secondary antibodies are often used to block endogenous mouse Ig during immunostaining, for example, AffiniPure Fab fragment Donkey antimouse IgG from Biozol (JIM-715-007-003) [58].

B Shen et al performed whole mount staining of mouse half bones with Alexa Fluor 647-AffiniPure F(ab')2 fragment donkey anti-chicken IgY at 1:250, Alexa Fluor 488-AffiniPure F(ab')2 fragment donkey anti-rabbit IgG at 1:250, Alexa Fluor 488-AffiniPure F(ab')2 fragment donkey anti-rabbit IgG (at 1:250 (all from Jackson ImmunoResearch) [59]. Invitrogen Alexa Fluor 546-conjugated goat anti-rabbit IgG F(ab')2 fragment was used to perform immunohistochemistry to investigate the role of sFLT-1 in the maintenance of the avascular photoreceptor layer in mouse models [60] and Invitrogen Alexa Fluor 488 F(ab')2 fragment of rabbit anti-goat IgG (H+L) was used in immunohistochemistry to investigate the mechanism of tip link regeneration in auditory hair cells [61]. F(ab')2 secondary antibodies from Jackson ImmunoResearch are commonly used [62].

Fc Receptors

Fc receptors (FcRs) are molecules expressed primarily on/in innate immune cells, that recognize and bind the Fc domain of antibodies and thereby initiate a cell-based immune response. The diverse functions of FcRs reflect the wide range of protective or modulating roles of antibodies, including mediating the neutralization and clearance of targeted substrates, as well as the programming of adaptive immunity [63]. The biological functions of FcRs are regulated by immunoreceptor tyrosine-based activation motifs (ITAMs) and immunoreceptor tyrosine-based inhibitory motifs (ITIMs), that act as the receptor’s interface with activating and inhibitory signaling pathways, respectively. Thus, signaling by ITAMs can elicit cell activation, phagocytosis and endocytosis, whereas signaling by ITIMs has an inhibitory effect on cell activation [64]. FcRs have been described for all classes of immunoglobulins and some of them are discussed below.

IgG receptors

This family includes FcγRI, FcγRII, FcγRIII and their isoforms. They are responsible for antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cell-mediated phagocytosis (ACDP) [63].

Another receptor that binds IgG is neonatal Fc receptor (FcRn), which is involved in the transfer of passive humoral immunity from a mother to her fetus. FcRn also protects IgG from degradation in vivo, explaining their long half-life in the serum [65]. This phenomenon has led to the development of better therapeutic antibodies by introducing alterations in the Fc region to promote the Fc-FcRn interaction.

IgE receptors

They include the high-affinity FcεRI, which is capable of binding monomeric IgE, and the low-affinity C-type lectin FcεRII, which interacts preferentially with complex IgE. FcεRI mediates the immediate hypersensitivity response of many allergic reactions by stimulating degranulation and the release of a range of inflammatory mediators on mast cells and basophils [66]. FcεRII exists both in a membrane-bound form that delivers a downregulating signal for IgE synthesis [66], as well as soluble fragments that generate an opposing upregulation on IgE synthesis [67]. Its role in transcytosis of IgE-allergen complexes in human airway and the intestinal epithelium is actively being investigated as a potential target for allergic airway inflammation as a result of food allergies [68, 69].

IgA receptors

The sole member of the IgA receptor group, FcαRI, is expressed only in cells of the myeloid lineage. It plays a role in both pro- and anti-inflammatory responses depending on the state of the IgA bound. While binding of secretory IgA (SIgA) present at mucosal sites has anti-inflammatory effects including prevention of pathogen invasion, binding of serum IgA leads to inflammatory responses. FcαRI also regulates neutrophil viability depending on the inflammatory microenvironment [70].

TRIM21

TRIM21 can be distinguished from other FcRs as it shows a broad antibody specificity. It can bind IgG, IgM and IgA [71-73]. It is also expressed by cells of most histogenic lineages [74]. TRIM21 participates in antibody-mediated interference of viral replication by targeting cytosolic virus-antibody complexes for proteasomal degradation.

Mitigating Fc Binding

Binding of Fc domains to FcRs can have undesired effects in monoclonal antibody-based analytical methods such as immunohistochemistry (IHC), fluorescence activated cell sorting (FACS) and chromatin immunoprecipitation (ChIP). Non-specific binding to FcRs may introduce background noise that can lead to detection of false positives. Solutions to deal with this problem include the use of (i) isotype controls for gating, (ii) serum to broadly compete for the receptors involved in non‐specific binding, or (iii) purified IgG to block Fc receptors specifically [75]. Innovex Fc Receptor blocker #NB309 can be used to block paraffin or frozen sections during IHC or IC experiments [76] or BD Biosciences #553142 [76, 77], Miltenyi Biotec Fc receptor block [78, 79] for flow cytometry. For example, Chopra S et al blocked FcγR binding with 5 μg/ml TruStain fcX (anti-mouse CD16/32, clone 93) from BioLegend during flow cytometry and cell sorting [80].

Note

Dr. Macarena Fritz Kelly from São José dos Campos, SP, Brazil, contributed the section about Fc receptors in September, 2018.

References
  1. Joosten V, Lokman C, van den Hondel C, Punt P. The production of antibody fragments and antibody fusion proteins by yeasts and filamentous fungi. Microb Cell Fact. 2003;2:1 pubmed
  2. Shaw A, Hoffecker I, Smyrlaki I, Rosa J, Grevys A, Bratlie D, et al. Binding to nanopatterned antigens is dominated by the spatial tolerance of antibodies. Nat Nanotechnol. 2019;14:184-190 pubmed publisher
  3. Dunbar J, Krawczyk K, Leem J, Baker T, Fuchs A, Georges G, et al. SAbDab: the structural antibody database. Nucleic Acids Res. 2014;42:D1140-6 pubmed publisher
  4. Renart J, Reiser J, Stark G. Transfer of proteins from gels to diazobenzyloxymethyl-paper and detection with antisera: a method for studying antibody specificity and antigen structure. Proc Natl Acad Sci U S A. 1979;76:3116-20 pubmed
  5. Matthews J. Immunocytochemical methods: a technical overview. J Oral Pathol. 1987;16:189-95 pubmed
  6. Lequin R. Enzyme immunoassay (EIA)/enzyme-linked immunosorbent assay (ELISA). Clin Chem. 2005;51:2415-8 pubmed
  7. Keyashian K, Annunziata M, Sakuraba A, Hanauer S. Management of inflammatory bowel disease: past, present and future. Expert Rev Clin Immunol. 2012;8:303-5 pubmed publisher
  8. Miller A, Ranatunga S. Immunotherapies in rheumatologic disorders. Med Clin North Am. 2012;96:475-96, ix-x pubmed publisher
  9. Shepard H, Jin P, Slamon D, Pirot Z, Maneval D. Herceptin. Handb Exp Pharmacol. 2008;:183-219 pubmed
  10. Goldstein G. Overview of the development of Orthoclone OKT3: monoclonal antibody for therapeutic use in transplantation. Transplant Proc. 1987;19:1-6 pubmed
  11. Jain R. Physiological barriers to delivery of monoclonal antibodies and other macromolecules in tumors. Cancer Res. 1990;50:814s-819s pubmed
  12. Chew H, De Lima P, Gonzalez Cruz J, Banushi B, Echejoh G, Hu L, et al. Endocytosis Inhibition in Humans to Improve Responses to ADCC-Mediating Antibodies. Cell. 2020;180:895-914.e27 pubmed publisher
  13. Bird R, Hardman K, Jacobson J, Johnson S, Kaufman B, Lee S, et al. Single-chain antigen-binding proteins. Science. 1988;242:423-6 pubmed
  14. Hudson P, Kortt A. High avidity scFv multimers; diabodies and triabodies. J Immunol Methods. 1999;231:177-89 pubmed
  15. Holliger P, Prospero T, Winter G. "Diabodies": small bivalent and bispecific antibody fragments. Proc Natl Acad Sci U S A. 1993;90:6444-8 pubmed
  16. Tao Y, Mis M, Blazer L, Ustav M, Steinhart Z, Chidiac R, et al. Tailored tetravalent antibodies potently and specifically activate Wnt/Frizzled pathways in cells, organoids and mice. elife. 2019;8: pubmed publisher
  17. Kortt A, Lah M, Oddie G, Gruen C, Burns J, Pearce L, et al. Single-chain Fv fragments of anti-neuraminidase antibody NC10 containing five- and ten-residue linkers form dimers and with zero-residue linker a trimer. Protein Eng. 1997;10:423-33 pubmed
  18. Hu S, Shively L, Raubitschek A, Sherman M, Williams L, Wong J, et al. Minibody: A novel engineered anti-carcinoembryonic antigen antibody fragment (single-chain Fv-CH3) which exhibits rapid, high-level targeting of xenografts. Cancer Res. 1996;56:3055-61 pubmed
  19. Schneider D, Xiong Y, Wu D, Hu P, Alabanza L, Steimle B, et al. Trispecific CD19-CD20-CD22-targeting duoCAR-T cells eliminate antigen-heterogeneous B cell tumors in preclinical models. Sci Transl Med. 2021;13: pubmed publisher
  20. Holliger P, Hudson P. Engineered antibody fragments and the rise of single domains. Nat Biotechnol. 2005;23:1126-36 pubmed
  21. Israeli H, Degtjarik O, Fierro F, Chunilal V, Gill A, Roth N, et al. Structure reveals the activation mechanism of the MC4 receptor to initiate satiation signaling. Science. 2021;372:808-814 pubmed publisher
  22. Muyldermans S. Nanobodies: natural single-domain antibodies. Annu Rev Biochem. 2013;82:775-97 pubmed publisher
  23. Wesolowski J, Alzogaray V, Reyelt J, Unger M, Juarez K, Urrutia M, et al. Single domain antibodies: promising experimental and therapeutic tools in infection and immunity. Med Microbiol Immunol. 2009;198:157-74 pubmed publisher
  24. Fridy P, Li Y, Keegan S, Thompson M, Nudelman I, Scheid J, et al. A robust pipeline for rapid production of versatile nanobody repertoires. Nat Methods. 2014;11:1253-60 pubmed publisher
  25. Wrapp D, De Vlieger D, Corbett K, Torres G, Wang N, Van Breedam W, et al. Structural Basis for Potent Neutralization of Betacoronaviruses by Single-Domain Camelid Antibodies. Cell. 2020;181:1004-1015.e15 pubmed publisher
  26. Koenig P, Das H, Liu H, Kummerer B, Gohr F, Jenster L, et al. Structure-guided multivalent nanobodies block SARS-CoV-2 infection and suppress mutational escape. Science. 2021;: pubmed publisher
  27. Akiba H, Tamura H, Kiyoshi M, Yanaka S, Sugase K, Caaveiro J, et al. Structural and thermodynamic basis for the recognition of the substrate-binding cleft on hen egg lysozyme by a single-domain antibody. Sci Rep. 2019;9:15481 pubmed publisher
  28. Beghein E, Gettemans J. Nanobody Technology: A Versatile Toolkit for Microscopic Imaging, Protein-Protein Interaction Analysis, and Protein Function Exploration. Front Immunol. 2017;8:771 pubmed publisher
  29. Akishiba M, Takeuchi T, Kawaguchi Y, Sakamoto K, Yu H, Nakase I, et al. Cytosolic antibody delivery by lipid-sensitive endosomolytic peptide. Nat Chem. 2017;9:751-761 pubmed publisher
  30. Herce H, Schumacher D, Schneider A, Ludwig A, Mann F, Fillies M, et al. Cell-permeable nanobodies for targeted immunolabelling and antigen manipulation in living cells. Nat Chem. 2017;9:762-771 pubmed publisher
  31. Crowley S, Bruck P, Bhuiyan M, Mitchell Gears A, Walsh M, Zhangxu K, et al. Neoleukin-2 enhances anti-tumour immunity downstream of peptide vaccination targeted by an anti-MHC class II VHH. Open Biol. 2020;10:190235 pubmed publisher
  32. Danis C, Dupré E, Zejneli O, Caillierez R, Arrial A, Bégard S, et al. Inhibition of Tau seeding by targeting Tau nucleation core within neurons with a single domain antibody fragment. Mol Ther. 2022;30:1484-1499 pubmed publisher
  33. Stanley S, Kelly L, Latcha K, Schmidt S, Yu X, Nectow A, et al. Bidirectional electromagnetic control of the hypothalamus regulates feeding and metabolism. Nature. 2016;531:647-50 pubmed publisher
  34. Che T, Majumdar S, Zaidi S, Ondachi P, McCorvy J, Wang S, et al. Structure of the Nanobody-Stabilized Active State of the Kappa Opioid Receptor. Cell. 2018;172:55-67.e15 pubmed publisher
  35. Laursen N, Friesen R, Zhu X, Jongeneelen M, Blokland S, Vermond J, et al. Universal protection against influenza infection by a multidomain antibody to influenza hemagglutinin. Science. 2018;362:598-602 pubmed publisher
  36. Pleiner T, Bates M, Gorlich D. A toolbox of anti-mouse and anti-rabbit IgG secondary nanobodies. J Cell Biol. 2018;217:1143-1154 pubmed publisher
  37. Abulrob A, Sprong H, Van Bergen en Henegouwen P, Stanimirovic D. The blood-brain barrier transmigrating single domain antibody: mechanisms of transport and antigenic epitopes in human brain endothelial cells. J Neurochem. 2005;95:1201-14 pubmed
  38. Li T, Bourgeois J, Celli S, Glacial F, Le Sourd A, Mécheri S, et al. Cell-penetrating anti-GFAP VHH and corresponding fluorescent fusion protein VHH-GFP spontaneously cross the blood-brain barrier and specifically recognize astrocytes: application to brain imaging. FASEB J. 2012;26:3969-79 pubmed publisher
  39. Cortez Retamozo V, Lauwereys M, Hassanzadeh Gh G, Gobert M, Conrath K, Muyldermans S, et al. Efficient tumor targeting by single-domain antibody fragments of camels. Int J Cancer. 2002;98:456-62 pubmed
  40. Meertens L, Hafirassou M, Couderc T, Bonnet Madin L, Kril V, Kummerer B, et al. FHL1 is a major host factor for chikungunya virus infection. Nature. 2019;: pubmed publisher
  41. Maier J, Traenkle B, Rothbauer U. Real-time analysis of epithelial-mesenchymal transition using fluorescent single-domain antibodies. Sci Rep. 2015;5:13402 pubmed publisher
  42. Saini S, Allore B, Jacobs R, Kaushik A. Exceptionally long CDR3H region with multiple cysteine residues in functional bovine IgM antibodies. Eur J Immunol. 1999;29:2420-6 pubmed
  43. Shojaei F, Saini S, Kaushik A. Unusually long germline DH genes contribute to large sized CDR3H in bovine antibodies. Mol Immunol. 2003;40:61-7 pubmed
  44. Stanfield R, Wilson I, Smider V. Conservation and diversity in the ultralong third heavy-chain complementarity-determining region of bovine antibodies. Sci Immunol. 2016;1:aaf7962 pubmed publisher
  45. Dong J, Lee Y, Kirmiz M, Palacio S, Dumitras C, Moreno C, et al. A toolbox of nanobodies developed and validated for use as intrabodies and nanoscale immunolabels in brain neurons. elife. 2019;8: pubmed publisher
  46. Kabayama H, Takeuchi M, Tokushige N, Muramatsu S, Kabayama M, Fukuda M, et al. An ultra-stable cytoplasmic antibody engineered for in vivo applications. Nat Commun. 2020;11:336 pubmed publisher
  47. Tamaki Y, Shodai A, Morimura T, Hikiami R, Minamiyama S, Ayaki T, et al. Elimination of TDP-43 inclusions linked to amyotrophic lateral sclerosis by a misfolding-specific intrabody with dual proteolytic signals. Sci Rep. 2018;8:6030 pubmed publisher
  48. Pozzi S, Thammisetty S, Codron P, Rahimian R, Plourde K, Soucy G, et al. Virus-mediated delivery of antibody targeting TAR DNA-binding protein-43 mitigates associated neuropathology. J Clin Invest. 2019;129:1581-1595 pubmed publisher
  49. Pastushok L, Fu Y, Lin L, Luo Y, DeCoteau J, Lee K, et al. A Novel Cell-Penetrating Antibody Fragment Inhibits the DNA Repair Protein RAD51. Sci Rep. 2019;9:11227 pubmed publisher
  50. Nelson A. Antibody fragments: hope and hype. MAbs. 2010;2:77-83 pubmed
  51. Robinson M, Shaller C, Garmestani K, Plascjak P, Hodge K, Yuan Q, et al. Effective treatment of established human breast tumor xenografts in immunodeficient mice with a single dose of the alpha-emitting radioisotope astatine-211 conjugated to anti-HER2/neu diabodies. Clin Cancer Res. 2008;14:875-82 pubmed publisher
  52. Cumber A, Ward E, Winter G, Parnell G, Wawrzynczak E. Comparative stabilities in vitro and in vivo of a recombinant mouse antibody FvCys fragment and a bisFvCys conjugate. J Immunol. 1992;149:120-6 pubmed
  53. Sanz L, Cuesta A, Compte M, Alvarez Vallina L. Antibody engineering: facing new challenges in cancer therapy. Acta Pharmacol Sin. 2005;26:641-8 pubmed
  54. Coolen M, Labusch M, Mannioui A, Bally Cuif L. Mosaic Heterochrony in Neural Progenitors Sustains Accelerated Brain Growth and Neurogenesis in the Juvenile Killifish N. furzeri. Curr Biol. 2020;30:736-745.e4 pubmed publisher
  55. Yasuda S, Tsuchiya H, Kaiho A, Guo Q, Ikeuchi K, Endo A, et al. Stress- and ubiquitylation-dependent phase separation of the proteasome. Nature. 2020;578:296-300 pubmed publisher
  56. Maya Ramos L, Mikawa T. Programmed cell death along the midline axis patterns ipsilaterality in gastrulation. Science. 2020;367:197-200 pubmed publisher
  57. Stigler J, Ziegler F, Gieseke A, Gebhardt J, Rief M. The complex folding network of single calmodulin molecules. Science. 2011;334:512-6 pubmed publisher
  58. Cantuti Castelvetri L, Ojha R, Pedro L, Djannatian M, Franz J, Kuivanen S, et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science. 2020;: pubmed publisher
  59. Shen B, Tasdogan A, Ubellacker J, Zhang J, Nosyreva E, Du L, et al. A mechanosensitive peri-arteriolar niche for osteogenesis and lymphopoiesis. Nature. 2021;591:438-444 pubmed publisher
  60. Luo L, Uehara H, Zhang X, Das S, Olsen T, Holt D, et al. Photoreceptor avascular privilege is shielded by soluble VEGF receptor-1. elife. 2013;2:e00324 pubmed publisher
  61. Indzhykulian A, Stepanyan R, Nelina A, Spinelli K, Ahmed Z, Belyantseva I, et al. Molecular remodeling of tip links underlies mechanosensory regeneration in auditory hair cells. PLoS Biol. 2013;11:e1001583 pubmed publisher
  62. Silva L, Doyle A, Greenwell Wild T, Dutzan N, Tran C, Abusleme L, et al. Fibrin is a critical regulator of neutrophil effector function at the oral mucosal barrier. Science. 2021;374:eabl5450 pubmed publisher
  63. Lee C, Romain G, Yan W, Watanabe M, Charab W, Todorova B, et al. IgG Fc domains that bind C1q but not effector Fc? receptors delineate the importance of complement-mediated effector functions. Nat Immunol. 2017;18:889-898 pubmed publisher
  64. Daeron M. Fc receptor biology. Annu Rev Immunol. 1997;15:203-34 pubmed
  65. Booth B, Ramakrishnan B, Narayan K, Wollacott A, Babcock G, Shriver Z, et al. Extending human IgG half-life using structure-guided design. MAbs. 2018;10:1098-1110 pubmed publisher
  66. Sutton B, Davies A. Structure and dynamics of IgE-receptor interactions: Fc?RI and CD23/Fc?RII. Immunol Rev. 2015;268:222-35 pubmed publisher
  67. Conrad D, Ford J, Sturgill J, Gibb D. CD23: an overlooked regulator of allergic disease. Curr Allergy Asthma Rep. 2007;7:331-7 pubmed
  68. Cooper A, Hobson P, Jutton M, Kao M, Drung B, Schmidt B, et al. Soluble CD23 controls IgE synthesis and homeostasis in human B cells. J Immunol. 2012;188:3199-207 pubmed publisher
  69. Tu Y, Salim S, Bourgeois J, Di Leo V, Irvine E, Marshall J, et al. CD23-mediated IgE transport across human intestinal epithelium: inhibition by blocking sites of translation or binding. Gastroenterology. 2005;129:928-40 pubmed
  70. Aleyd E, Heineke M, van Egmond M. The era of the immunoglobulin A Fc receptor Fc?RI; its function and potential as target in disease. Immunol Rev. 2015;268:123-38 pubmed publisher
  71. Mallery D, McEwan W, Bidgood S, Towers G, Johnson C, James L. Antibodies mediate intracellular immunity through tripartite motif-containing 21 (TRIM21). Proc Natl Acad Sci U S A. 2010;107:19985-90 pubmed publisher
  72. Bidgood S, Tam J, McEwan W, Mallery D, James L. Translocalized IgA mediates neutralization and stimulates innate immunity inside infected cells. Proc Natl Acad Sci U S A. 2014;111:13463-8 pubmed publisher
  73. Morishita R, Sugiyama S, Denda M, Tokunaga S, Kido K, Shioya R, et al. CF-PA2Vtech: a cell-free human protein array technology for antibody validation against human proteins. Sci Rep. 2019;9:19349 pubmed publisher
  74. Reymond A, Meroni G, Fantozzi A, Merla G, Cairo S, Luzi L, et al. The tripartite motif family identifies cell compartments. EMBO J. 2001;20:2140-51 pubmed
  75. Smith P, Chattopadhyay P. Re-visiting Fc-receptor blocking maneuvers in man. Cytometry A. 2016;89:975-977 pubmed publisher
  76. Albrengues J, Shields M, Ng D, Park C, Ambrico A, Poindexter M, et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science. 2018;361: pubmed publisher
  77. Yousefzadeh M, Flores R, Zhu Y, Schmiechen Z, Brooks R, Trussoni C, et al. An aged immune system drives senescence and ageing of solid organs. Nature. 2021;594:100-105 pubmed publisher
  78. Schulte Schrepping J, Reusch N, Paclik D, Baßler K, Schlickeiser S, Zhang B, et al. Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment. Cell. 2020;: pubmed publisher
  79. Rigau M, Ostrouska S, Fulford T, Johnson D, Woods K, Ruan Z, et al. Butyrophilin 2A1 is essential for phosphoantigen reactivity by γδ T cells. Science. 2020;367: pubmed publisher
  80. Chopra S, Giovanelli P, Alvarado Vazquez P, Alonso S, Song M, Sandoval T, et al. IRE1α-XBP1 signaling in leukocytes controls prostaglandin biosynthesis and pain. Science. 2019;365: pubmed publisher
ISSN : 2329-5139