scispace - formally typeset
Open AccessJournal ArticleDOI

A general protocol for the generation of Nanobodies for structural biology

TLDR
A general protocol for the generation of Nanobodies to be used as crystallization chaperones for the structural investigation of diverse conformational states of flexible (membrane) proteins and complexes thereof.
Abstract
There is growing interest in using antibodies as auxiliary tools to crystallize proteins. Here we describe a general protocol for the generation of Nanobodies to be used as crystallization chaperones for the structural investigation of diverse conformational states of flexible (membrane) proteins and complexes thereof. Our technology has a competitive advantage over other recombinant crystallization chaperones in that we fully exploit the natural humoral response against native antigens. Accordingly, we provide detailed protocols for the immunization with native proteins and for the selection by phage display of in vivo-matured Nanobodies that bind conformational epitopes of functional proteins. Three representative examples illustrate that the outlined procedures are robust, making it possible to solve by Nanobody-assisted X-ray crystallography in a time span of 6-12 months.

read more

Content maybe subject to copyright    Report

Vrije Universiteit Brussel
A general protocol for the generation of Nanobodies for structural Biology
Pardon, Els; Laeremans, Toon; Triest, Sarah; Rasmussen, Soren G. F.; Ruf, Armin;
Muyldermans, Serge; Hol, Wim G. J.; Kobilka, Brian K; Steyaert, Jan
Published in:
Nature Protocols
DOI:
10.1038/nprot.2014.039
Publication date:
2014
Document Version:
Final published version
Link to publication
Citation for published version (APA):
Pardon, E., Laeremans, T., Triest, S., Rasmussen, S. G. F., Ruf, A., Muyldermans, S., ... Steyaert, J. (2014). A
general protocol for the generation of Nanobodies for structural Biology. Nature Protocols, 9, 674-693.
https://doi.org/10.1038/nprot.2014.039
Copyright
No part of this publication may be reproduced or transmitted in any form, without the prior written permission of the author(s) or other rights
holders to whom publication rights have been transferred, unless permitted by a license attached to the publication (a Creative Commons
license or other), or unless exceptions to copyright law apply.
Take down policy
If you believe that this document infringes your copyright or other rights, please contact openaccess@vub.be, with details of the nature of the
infringement. We will investigate the claim and if justified, we will take the appropriate steps.
Download date: 09. Aug. 2022

© 2014 Nature America, Inc. All rights reserved.
PROTOCOL
674
|
VOL.9 NO.3
|
2014
|
NATURE PROTOCOLS
INTRODUCTION
The production of diffraction-quality crystals remains the major
bottleneck in macromolecular X-ray crystallography. Collective
efforts of several laboratories have demonstrated that Nanobodies
are exquisite chaperones for crystallizing complex biological
systems such as membrane proteins
1–3
, transient multiprotein
assemblies
2,4–6
, transient conformational states
1
and intrinsically
disordered proteins
7,8
. Further, they can be used as structural
probes of protein misfolding and fibril formation
9,10
. Nanobodies
are the small (15 kDa) and stable single-domain fragments
harboring the full antigen-binding capacity of the original
heavy chain–only antibodies that naturally occur in camelids
11,12
.
Nanobodies are encoded by single gene fragments, they are
easily produced in microorganisms and they exhibit a superior
stability compared with derivatives of conventional antibodies
such as Fabs or scFvs. Because of their compact prolate shape,
Nanobodies expose a convex paratope and have access to
cavities or clefts on the surface of proteins
1,13,14
that are often
inaccessible to conventional antibodies. These cryptic epitopes
can be readily recognized by the long CDR3 loop of the Nanobody.
In our experience, Nanobodies raised in vivo by immunization
against, and selected on, properly folded proteins systematically
recognize discontinuous amino acid segments of the native
protein conformation (i.e., conformational epitopes), making
them ideal tools to selectively stabilize specific conformational
states of (membrane) proteins.
For the discovery of Nanobodies as crystallization chaper-
ones, ~1 mg of functional protein is required. The generation of
in vivo–matured Nanobodies can therefore be incorporated in
the crystallization pipeline even before the purification of the
protein has been fully optimized and scaled up. Nanobodies
binding conformational epitopes (conformational Nanobodies)
can subsequently be used for preparing pure, homogeneous and
highly concentrated monodisperse samples that are required for
crystallization
15
. If no native purified protein is available, genetic
and cell-based vaccinations, combined with cell-based selection
approaches, have been successfully applied in our laboratory and
elsewhere to generate Nanobodies against target proteins in their
native conformation
16–18
.
Comparisons with other approaches
Here we present a general protocol for the generation, selection
and purification of recombinant in vivo–matured Nanobodies
for structural biology
1–5,7,9,19–30
that takes 3–4 months. Our
Nanobody discovery platform has the competitive advantage over
other recombinant crystallization chaperones
31–33
that the cloned
Nanobody library represents the full collection of the naturally cir-
culating, humoral antigen-binding repertoire of heavy chain–only
antibodies, in contrast to combinatorial libraries of conventional
antibody fragments. Because Nanobodies are encoded by single
exons, the full antigen-binding capacity of in vivo–matured anti-
bodies can be cloned and efficiently screened for high-affinity
binders, allowing one to fully exploit the humoral response of large
mammals against native antigens. To our knowledge, there are no
indications that in vivomatured Nanobodies induce non-native
conformations. Surely, immature B cells expressing antibodies
that have to pay a substantial energetic penalty for distorting the
antigen structure will have a lower probability of proliferation and
differentiation into mature antibody-secreting B lymphocytes.
Limitations
With nearly 20 years of experience we have learned that conforma-
tional Nanobodies can be identified against any properly folded
protein. In those cases in which we failed in a first attempt, we
successfully performed new immunizations or pannings, paying
special attention to the quality of the antigen, thereby learning
that good protein biochemistry is the key to success. Although
Nanobodies are good at binding conformational epitopes on
folded proteins with high affinity, they perform poorly at binding
A general protocol for the generation of Nanobodies
for structural biology
Els Pardon
1,2
, Toon Laeremans
1,2
, Sarah Triest
1,2
, Søren G F Rasmussen
3
, Alexandre Wohlkönig
1,2
, Armin Ruf
4
,
Serge Muyldermans
2,5
, Wim G J Hol
6
, Brian K Kobilka
7
& Jan Steyaert
1,2
1
Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
2
Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB),
Brussels, Belgium.
3
Department of Neuroscience and Pharmacology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark.
4
Pharma Research
and Early Development (pRED), Small Molecule Research, Discovery Technologies, F. Hoffmann-La Roche, Basel, Switzerland.
5
Cellular and Molecular Immunology,
VUB, Brussels, Belgium.
6
Department of Biochemistry, Biomolecular Structure Center, School of Medicine, University of Washington, Seattle, Washington, USA.
7
Department of Molecular and Cellular Physiology, School of Medicine, Stanford University, Stanford, California, USA. Correspondence should be addressed
to J.S. (jan.steyaert@vib-vub.be).
Published online 27 February 2014; doi:10.1038/nprot.2014.039
There is growing interest in using antibodies as auxiliary tools to crystallize proteins. Here we describe a general protocol
for the generation of Nanobodies to be used as crystallization chaperones for the structural investigation of diverse
conformational states of flexible (membrane) proteins and complexes thereof. Our technology has a competitive advantage
over other recombinant crystallization chaperones in that we fully exploit the natural humoral response against native antigens.
Accordingly, we provide detailed protocols for the immunization with native proteins and for the selection by phage display
of in vivo–matured Nanobodies that bind conformational epitopes of functional proteins. Three representative examples
illustrate that the outlined procedures are robust, making it possible to solve by Nanobody-assisted X-ray crystallography
in a time span of 6–12 months.

© 2014 Nature America, Inc. All rights reserved.
PROTOCOL
NATURE PROTOCOLS
|
VOL.9 NO.3
|
2014
|
675
peptides or intrinsically unfolded parts of proteins. For linear
epitopes, conventional antibodies may be a better alternative.
Applications
The Nanobodies to be used as crystallization chaperones can also
be valuable for other applications in structural biology. For exam-
ple, domain-specific Nanobodies have been used in single-particle
electron microscopy (EM) as a marker to track these domains
in particle projections
34,35
. Because many Nanobodies can be
functionally expressed as intrabodies in eukaryotic cells, these
single-domain antibodies can also be used as biosensors to track
conformational properties of their targets inside a living cell
36–39
.
Ultimately, Nanobodies that constrain protein targets in unique
disease-linked conformations may facilitate the discovery of new
therapeutic molecules
40
.
Experimental design
General considerations. The workflow for generating, isolating
and characterizing the Nanobodies to be used as crystallization
chaperones (Fig. 1) is inherently dependent on the nature of the
antigen and on the purpose of the structural study. Several steps
in the Nanobody discovery process, including the preparation of
the immunogen, the selection strategy, the screening approach
and the functional and biophysical characterization, differ when
the target is a soluble protein, a membrane protein or a multi-
protein complex. To make this protocol broadly applicable for
the structural biology community, several modifications to the
standard protocol are referred to in the text.
Protein production and antigen quality control. Today, research-
ers are faced with a bewildering array of methods to produce and
purify recombinant proteins
41
. Although this is not the focus of
this paper, the supply of properly folded protein is crucial for the
generation of conformational Nanobodies. We discourage immu-
nization of animals with poorly characterized protein samples.
Our standard protocol typically requires 1 mg of the purified
protein: 700 µg for immunization and the remaining portion
for all Nanobody selection, identification and characterization
efforts. Ideally, a single batch of the purified protein is dispensed
into aliquots and stored under conditions that ensure the sta-
bility of the protein over time. Often, flash-freezing in liquid
nitrogen and storage at −80 °C is favored. We insist on confirm-
ing protein quality of one thawed aliquot before the protein is
administered as an immunogen. Biochemical or cellular assays
that quantitatively assess the functionality of the antigen (e.g.,
enzymatic and signaling activity, interaction with (radio)ligands
or binding of certified conventional antibodies against discon-
tinuous epitopes) can be used to test whether your protein is
properly folded. If your protein cannot be tested functionally,
we advise performing a rigorous biophysical characterization to
confirm its folded state (reviewed in ref. 42). Unless it has been
demonstrated not to have any effect on the functionality of the
protein, we strongly discourage multiple freeze-thaw cycles in
order to minimize protein denaturation. If samples cannot be
frozen, consider using freshly prepared protein throughout the
immunization and discovery effort.
Conformational locking of the antigens. Nanobodies have been
shown to stabilize proteins such as kinases and G protein–coupled
receptors (GPCRs) in unique biologically relevant conforma-
tions
33,43,44
. To identify such Nanobodies, we advise immunizing
animals with proteins constrained in the desired conformation
with cofactors, enzyme inhibitors
27
, orthosteric ligands
1
, allosteric
ligands or any molecule that conformationally traps the macro-
molecule in a particular state
45
. We recommend using ligands
that dissociate slowly to maximize the lifetime of the constrained
target conformation in the immunized animal. Purified, detergent-
solubilized membrane proteins may denature after immunization
owing to the dissociation of detergents. To maintain a native con-
formation after immunization, reconstitution of the protein into
a lipid environment, typically phospholipid vesicles
1
, or the use of
a very tight binding detergent
2
may be required. Reconstitution
of membrane proteins in lipid vesicles may also reduce ligand
dissociation. A ligand trapped in the vesicle can rebind the intra-
vesicular binding site of the protein, prolonging the extravesicular
exposure of the desired conformational state to the immune system.
Alternatively, particular mutations may trap the target in a
unique conformational state. Finally, chemical cross-linking
between protein domains or different proteins may stabilize
epitopes that are unique to the complex
2
.
Immunogen preparation, camelid immunization and repertoire
cloning. Antibodies with a homodimeric heavy-chain composition
devoid of light chains are only found in Tylopoda (camels, drom-
edaries and llamas) and sharks
46
. Our protocols can be applied to
llamas, camels, dromaderies and alpacas. All vaccination experi-
ments are executed according to EU animal welfare legislation and
after approval of the local ethics committee. It is known that stress
can cause immunosuppression. Animals should be manipulated
by authorized staff, preferably by an experienced veterinarian.
We allow animals to acclimatize to new housing conditions for
at least 1 week before immunization starts.
Protein production
Antigen quality control
LIama immunization
Functional antigen immobilization
Selection by phage display
Selection by yeast or bacterial display
Screening for antigen binders
Nanobody purification
Co crystallization
Structure refinement
Nanobody engineering for other biophysical applications
Biochemical and biophysical
characterization of binders
Repertoire cloning &
library construction
Reconstitution of antigen
in liposomes
Genetic and cell-based
vaccinations
(Bio)chemical cross-linking of
multiprotein antigens
Conformational locking of
the antigen
Step 1
Step 4
Step 7
Step 46
Step 62
Step 70
Figure 1
|
Workflow for the generation of conformational Nanobodies for
structural biology.

© 2014 Nature America, Inc. All rights reserved.
PROTOCOL
676
|
VOL.9 NO.3
|
2014
|
NATURE PROTOCOLS
In llamas, repetitive immunogen administrations generate a
robust immune response mediated by both conventional and
heavy chain–only antibodies. On the basis of 10 years of expe-
rience, GERBU LQ is a good immunostimulating adjuvant for
conformational Nanobody discovery, and it is well tolerated by
llamas. In general, we inject our animals subcutaneously with
cocktails of 1–5 antigens mixed with adjuvant. Alternatively,
different immunogens can be injected separately at different s.c.
locations. If small-molecule ligands are used for the conforma-
tional locking of an antigen, we always add these compounds in
excess to the antigen. In our hands, we have successfully reused
llamas for different Nanobody discovery projects by respecting a
grace period of at least 6 months.
A blood sample of 100 ml from an immunized llama contains suf-
ficient expressing B cells to clone a diverse set of the affinity-matured
Nanobodies with high specificity for their cognate antigen
47
.
Peripheral blood lymphocytes (PBLs) should be isolated without
delay from the noncoagulated blood for the purification of total
RNA and the synthesis of cDNA. From this cDNA, the Nanobody-
encoding open reading frames can be amplified by PCR and
cloned into an appropriate phage display vector.
Primer design. Over the years, several PCR strategies have been
developed to amplify Nanobody gene fragments from lymphocyte
cDNA. We prefer to use a two-step nested PCR approach. One
pair of primers (CALL001 and CALL002) has been designed for
the first PCR by using the cDNA of B lymphocytes as the tem-
plate
48
. The CALL002 primer anneals in a region of the second
constant heavy-chain domain (CH2) that is conserved among
all IgG isotypes of all camelids, whereas the CALL001 primer
anneals in a well-conserved region of the leader signal sequence
of all V elements of family III (by far the most abundant V fam-
ily in camelids). The primers VHH-Back and VHH-For are
used to amplify the Nanobody repertoire via a second nested
PCR (Supplementary Fig. 1a). From our experience, the prim-
ers described in PROCEDURE Steps 21 and 24 are adequate to
amplify Nanobody-encoding genes of family III from dromedary
(Camelus dromedaries), camel (Camelus bactrianus), llama (Lama
glama), and also alpaca (Vicugna pacos).
Van der Linden et al.
49
developed dedicated primers annealing
to the hinge of each heavy chain–only IgG isotype
49
of all camelid
species (Supplementary Fig. 1b). Maass et al.
50
designed prim-
ers dedicated for alpaca Nanobodies (Supplementary Fig. 1c).
Kastelic et al.
51
used primers that amplify mixtures of the VHH
(variable fragment of the heavy-chain antibody) and the VH
(variable fragment of the classical antibody) domains from llama
(Supplementary Fig. 1d).
Selection by phage display. Many excellent reviews on selection
methodologies to enrich for target-specific antibodies against
native epitopes have been published
52
. Phage display is certainly
the most robust technique, but yeast display
53,54
or bacterial
display
55
can also be used to select Nanobodies from immune
libraries. For conformational binders, it is essential to perform
the in vitro selection (panning) experiments under conditions
(buffer with appropriate detergent, pH, temperature and cofac-
tors) that produce the desired conformation of the protein during
phage incubation. To reduce the background of phage-expressing
nonspecific Nanobodies, the target protein should be highly pure
(>95%) and homogeneous in conformation.
Aspecific adsorption onto the solid surface of an ELISA plate is
still the most common way to immobilize targets for selection by
phage display, but this method can result in (partial) denaturation
of the protein
56
. Although the use of streptavidin-coated magnetic
beads is a valid alternative, we perform most panning experiments
in 96-well plates, which allows multiple parallel selection condi-
tions. Our preferred method is to capture biotinylated or tagged
target protein on a solid phase coated with NeutrAvidin or a tag-
specific antibody, respectively. Alternative strategies for presenting
target proteins during selection are summarized in Table 1. The
structural integrity and homogeneity of the presented target is
the most decisive factor for selecting Nanobodies with the desired
properties. If possible, we try different capturing or immobiliza-
tion methods, vary the antigen concentration, use different deter-
gents or ligands to keep the target in the desired conformation, try
different washing and incubation buffers or use different elution
methods in parallel pannings by using different selection wells on
the same 96-well plate. In many cases, magnetic beads can be used
as a valid alternative in order to perform selections in solution.
Depending on the magnitude of the heavy chain–only
antibody-mediated humoral response in the llama, typically
one or two rounds of panning are sufficient to enrich for target-
specific Nanobodies. Rather than selecting for target specificity
alone, we prefer implementing conditions early on that allow
the identification of Nanobodies with the desired functional or
biophysical properties: high affinity, stabilization of a unique
protein conformation, modulation of receptor function, binding
to a particular target domain or epitope, interference with
TABLE 1
|
Target presentation methods for the identification of
Nanobodies with demonstrated crystallization chaperone activity.
Target selection format Negative control
Biotinylated target protein
captured onto a
NeutrAvidin-coated well
27
NeutrAvidin-coated well
Membrane protein that is
packed into a (biotinylated)
nanodisc and captured onto a
NeutrAvidin- or antibody-coated
well
2
(see Anticipated Results)
Irrelevant membrane
protein reconstituted in
(biotinylated) nanodiscs
and captured onto
a NeutrAvidin- or
antibody-coated well
Protein captured onto an
immobilized monoclonal antibody
that is specific for your protein
or a protein tag (His-tag,
Strep-tag, GST, etc.)
Well that is coated with
the monoclonal antibody
Solid-phase immobilized protein
by aspecific adsorption
6,70
Empty well
Solid-phase immobilized membrane
protein reconstituted in lipid
vesicles
1
(see Anticipated Results)
Well coated with lipid
vesicles harboring an
irrelevant membrane protein
Solid-phase immobilized membrane
protein that is packed into
virus-like particles
71
Well coated with an
irrelevant virus-like particle

© 2014 Nature America, Inc. All rights reserved.
PROTOCOL
NATURE PROTOCOLS
|
VOL.9 NO.3
|
2014
|
677
(orthosteric) ligand binding and stabilization of a protein com-
plex, among others. If no purified protein with demonstrated
functionality is available, Nanobody-panning methodologies that
use target-expressing cells (or derivatives) have been reported and
are implemented in our laboratory
57
.
Screening and functional/biophysical characterization of
conformational Nanobodies. After panning, we routinely
pick 96 individual clones from distinct selection outputs and
express the Nanobodies in the E. coli periplasm in 96-well plates.
In the past 10 years, we have rarely found Nanobodies that
bind linear epitopes. Therefore, we discourage the use of western
blotting or any other technique that tends to unfold the target.
In parallel to the assessment of antigen specificity, assays should
be implemented for the identification of Nanobodies with the
desired functional or biophysical properties. If the target is a
membrane protein, we recommend additionally assessing
Nanobody binding to target-overexpressing cells via fluorescence-
activated flow cytometry.
Our display vectors permit the inducible periplasmic expression
of Nanobodies as soluble C-terminally His
6
-tagged proteins in E. coli
strain WK6. Milligram quantities of >95% pure Nanobody can rou-
tinely be obtained by immobilized metal ion affinity chromatography
(IMAC) from the periplasmic extract of a 1-liter bacterial culture
48
.
As it is not known upfront which Nanobodies will behave as
the best crystallization chaperones, it is crucial to identify a large
panel of sequence-diverse Nanobodies (see Anticipated Results).
Typically, after a protein-based immunization and selection
discovery, between 3 and 30 Nanobody families are identified.
A Nanobody family is defined as a group of Nanobodies with
a high similarity in their CDR3 sequence (identical length and
>80% sequence identity). Nanobodies from the same family
derive from the same B-cell lineage and bind to the same epitope
on the target.
Nanobodies as crystallization chaperones. Electrophoretic
mobility shift assays on native gels provide a quick and easy
strategy for verifying whether purified Nanobodies form a
homogeneous complex with the target protein. Figure 2 shows how
mobility shift assays have been used to characterize Nanobodies
that bind an editosome protein of T. brucei
24
. Cocrystallization
experiments can be successful just by mixing the Nanobody and
target protein in a 1.2:1 molar ratio. Alternatively, the complex
can be further purified by size-exclusion chromatography after
co-incubation. Because Nanobodies are resistant to additives,
extremes of pH and temperature, and proteases
58
, they are
ideally suited for screening a broad range of crystallization
conditions with variations in pH, ionic strength, temperature,
protein concentration, salts, ligands or additives, and type of
precipitant. Conditions that can be screened are limited by the
stability of the target protein rather than by the stability of the
crystallization chaperones. More important is that Nanobodies
are extremely soluble proteins (40 mg ml
−1
), maximizing the
chance that they crystallize in complex with their cocrystallization
target rather than yielding Nanobody-only crystals. Because
they are efficiently produced in the periplasm of E. coli, SeMet-
labeled Nanobodies may ultimately be used for phasing via
single-anomalous dispersion technique without the need for
introducing SeMet into the target protein
28
. Alternatively, phase
information can be obtained from a molecular replacement
solution of the Nanobody in the complex.
Considerations about the laboratory facilities. We advise
dedicating two separate laboratories for Nanobody discovery.
One phage-free laboratory is used for Nanobody repertoire
amplification and cloning. We use dedicated reagents for library
construction. The other laboratory is used for all phage work,
including the amplification of phage and pannings. To reduce
phage contamination, we use filter tips and clean benches, equip-
ment and glassware with 1% (wt/vol) sodium hypochlorite
after each experiment. Use disposables whenever possible, and
discard them in 1% (wt/vol) sodium hypochlorite to inactivate
the remaining phage particles.
MATERIALS
REAGENTS
Llamas ! CAUTION All vaccination experiments should be executed in
accordance with the applicable animal welfare legislation, and they must be
approved by the local ethics committee.
Acetic acid, glacial (Merck, cat. no. 1.00058.2500) ! CAUTION Glacial acetic
acid is corrosive. Avoid inhalation and exposure to skin and eyes.
AEBSF protease inhibitor (Carl Roth, cat. no. 2931.3)
Agarose MP (Roche, cat. no. 11 388 991 001)
+ +
+
+
7
8
8
8
+
Nanobody
Target protein
Target-Nb7 complex
Target-Nb8 complex
Target alone
++
Nb7
Target alone
Nb8
Figure 2
|
Native gel analysis of Nanobodies interacting with the
oligonucleotide-binding fold of the A1 protein of the editosome of the
sleeping sickness parasite Trypanosoma brucei
24
. Nanobodies Nb7 and Nb8
were generated with this protocol. The target protein was incubated with
Nb7 or Nb8 for 30 min at 4 °C in 20 mM Tris (pH 7.5), 1 mM DTT and
300 mM NaCl. Complex formation of target and Nanobody was analyzed on
a 4–15% (wt/vol) native gel at 110 V for 1 h and stained with Coomassie
Blue. The positions of the target protein alone (first lane), the target-Nb7
complex (third lane), the Nanobody Nb8 alone (fourth lane) and the
target-Nb8 complex (fifth lane) are indicated with symbols above the bands.
Owing to its high isoelectric point, Nb7 is too positively charged to run into
the native gel (second lane).
AP-conjugate substrate: 4-nitrophenyl phosphate disodium salt hexahydrate
(DNPP, Sigma-Aldrich, cat. no. 71768)
CaCl
2
·2H
2
O (Merck, cat. no. 2382)
Chloroform (Merck, cat. no. 1.02445.0250)
Citric acid·H
2
O (Sigma-Aldrich, cat. no. C1909)
E. coli strains CRITICAL Use an amber codon suppression (supE)
strain such as TG1 (electrocompetent cells 4 × 10
10
c.f.u. µg
−1
;
Lucigen, cat. no. 60502-1) for cloning and preparation of phage libraries,

Citations
More filters
Journal ArticleDOI

Structural insights into µ-opioid receptor activation

TL;DR: A 2.1 Å X-ray crystal structure of the murine μOR bound to the morphinan agonist BU72 and a G protein mimetic camelid antibody fragment is reported, revealing an extensive polar network between the ligand-binding pocket and the cytoplasmic domains appears to play a similar role in signal propagation for all three G-protein-coupled receptors.
Journal ArticleDOI

The P2X7 Receptor Channel: Recent Developments and the Use of P2X7 Antagonists in Models of Disease

TL;DR: The cloning and characterization of human, rat, mouse, guinea pig, dog, and Rhesus macaque P2X7, as well as recent observations regarding the gating and permeability of P2x7, are discussed.
Journal ArticleDOI

Nanobodies as therapeutics: big opportunities for small antibodies

TL;DR: How Nbs are being explored as therapeutics in many fields of medicine, including oncology, inflammatory, infectious and neurological diseases, and imaging, is discussed and their potential for use in the diagnosis and monitoring of diseases is highlighted.
Journal ArticleDOI

Structure of the Nanobody-Stabilized Active State of the Kappa Opioid Receptor

TL;DR: A crystal structure of human KOP in complex with the potent epoxymorphinan opioid agonist MP1104 and an active-state-stabilizing nanobody is provided and key residues that propagate larger-scale structural rearrangements and transducer binding are illuminated that elucidate the structural determinants of KOP pharmacology, function, and biased signaling.
References
More filters
Journal ArticleDOI

Naturally occurring antibodies devoid of light chains

TL;DR: The presence of considerable amounts of IgG-like material of Mr 100K in the serum of the camel, which is composed of heavy-chain dimers and devoid of light chains, but nevertheless have an extensive antigen-binding repertoire, opens new perspectives in the engineering of antibodies.
Journal ArticleDOI

The single-step method of RNA isolation by acid guanidinium thiocyanate–phenol–chloroform extraction: twenty-something years on

TL;DR: The original protocol, enabling the isolation of RNA from cells and tissues in less than 4 hours, greatly advanced the analysis of gene expression in plant and animal models as well as in pathological samples, as demonstrated by the overwhelming number of citations the paper gained over 20 years.
Journal ArticleDOI

Structure of a nanobody-stabilized active state of the β2 adrenoceptor

TL;DR: A camelid antibody fragment to the human β2 adrenergic receptor is generated, and an agonist-bound, active-state crystal structure of the receptor-nanobody complex is obtained, providing insights into the process of agonist binding and activation.
Journal ArticleDOI

Nanobodies: Natural Single-Domain Antibodies

TL;DR: The facile identification of antigen-specific VHHs and their beneficial biochemical and economic properties have encouraged antibody engineering of these single-domain antibodies for use as a research tool and in biotechnology and medicine.
Related Papers (5)
Frequently Asked Questions (15)
Q1. What are the contributions in this paper?

Nanobodies have been used as chaperones for crystallizing complex biological systems such as membrane proteins, transient multiprotein assemblies, and intrinsically disordered proteins this paper. 

chemical cross-linking between protein domains or different proteins may stabilize epitopes that are unique to the complex2. 

Because Nanobodies are encoded by single exons, the full antigen-binding capacity of in vivo–matured antibodies can be cloned and efficiently screened for high-affinity binders, allowing one to fully exploit the humoral response of large mammals against native antigens. 

Because they are efficiently produced in the periplasm of E. coli, SeMetlabeled Nanobodies may ultimately be used for phasing via single-anomalous dispersion technique without the need for introducing SeMet into the target protein28. 

More important is that Nanobodies are extremely soluble proteins (≥40 mg ml−1), maximizing the chance that they crystallize in complex with their cocrystallization target rather than yielding Nanobody-only crystals. 

The structural integrity and homogeneity of the presented target is the most decisive factor for selecting Nanobodies with the desired properties. 

Collective efforts of several laboratories have demonstrated that Nanobodies are exquisite chaperones for crystallizing complex biological systems such as membrane proteins1–3, transient multiprotein assemblies2,4–6, transient conformational states1 and intrinsically disordered proteins7,8. 

Electrophoretic mobility shift assays on native gels provide a quick and easy strategy for verifying whether purified Nanobodies form ahomogeneous complex with the target protein. 

Depending on the magnitude of the heavy chain–only antibody-mediated humoral response in the llama, typically one or two rounds of panning are sufficient to enrich for targetspecific Nanobodies. 

Two DNA fragments should be amplified: fragments of ~700 bp representing the heavy chain–only antibody repertoire and fragments of 1,000 bp corresponding to the heavy chain of the conventional antibodies (supplementary Fig. 1a). 

After each wash step, remove excess liquid by tapping the emptied plates once on a paper towel, avoiding cross-contamination of the phage to the neighboring wells. 

The generation of in vivo–matured Nanobodies can therefore be incorporated in the crystallization pipeline even before the purification of the protein has been fully optimized and scaled up. 

These customsynthesized primers (Sigma-Aldrich) are stored at −20 °C for years as stock solutions at 100 µM in 10 mM Tris-HCl buffer (pH 8.5) dN6 random primers (5′-NNNNNN-3′) are custom-synthesized and stored at −20° for years as a stock solution in 10 mM Tris-HCl buffer (pH 8.5) at a 2.5 µg µl−1 concentration EQUIPMENT 

Phage display is certainly the most robust technique, but yeast display53,54 or bacterial display55 can also be used to select Nanobodies from immune libraries. 

The workflow for generating, isolating and characterizing the Nanobodies to be used as crystallization chaperones (Fig. 1) is inherently dependent on the nature of the antigen and on the purpose of the structural study.