Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/319986158

Immuno-detection of cleaved SNAP-25 from differentiated mouse embryonic


stem cells provides a sensitive assay for determination of botulinum A toxin
and antitoxin potency

Article  in  Journal of Immunological Methods · September 2017


DOI: 10.1016/j.jim.2017.09.007

CITATIONS READS

14 311

5 authors, including:

Shalini P Rajagopal Dorothea Sesardic


Medicines and Healthcare products Regulatory Agency (MHRA) National Institute for Biological Standards and Control
9 PUBLICATIONS   169 CITATIONS    179 PUBLICATIONS   5,254 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

EU AntiBOTABE View project

Replacing animals for botulinum testing View project

All content following this page was uploaded by Shalini P Rajagopal on 29 September 2017.

The user has requested enhancement of the downloaded file.


Journal of Immunological Methods xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Journal of Immunological Methods


journal homepage: www.elsevier.com/locate/jim

Research paper

Immuno-detection of cleaved SNAP-25 from differentiated mouse


embryonic stem cells provides a sensitive assay for determination of
botulinum A toxin and antitoxin potency
G. Yadirgi, P. Stickings, S. Rajagopal, Y. Liu, D. Sesardic⁎
Division of Bacteriology, National Institute for Biological Standards and Control, a center of the Medicines and Healthcare Products Regulatory Agency, South Mimms,
Potters Bar, Hertfordshire EN6 3QG, UK

A R T I C L E I N F O A B S T R A C T

Keywords: Botulinum toxin type A is a causative agent of human botulism. Due to high toxicity and ease of production it is
BoNT/A classified by the Centres for Disease Control and Prevention as a category A bioterrorism agent. The same
Mouse embryonic stem cells serotype, BoNT/A, is also the most widely used in pharmaceutical preparations for treatment of a diverse range
Cell-based assay of neuromuscular disorders. Traditionally, animals are used to confirm the presence and activity of toxin and to
SNAP-25
establish neutralizing capabilities of countermeasures in toxin neutralization tests. Cell based assays for BoNT/A
Capture ELISA
have been reported as the most viable alternative to animal models, since they are capable of reflecting all key
In vitro potency
Toxin neutralization steps (binding, translocation, internalization and cleavage of intracellular substrate) involved in toxin activity. In
this paper we report preliminary development of a simple immunochemical method for specifically detecting
BoNT/A cleaved intracellular substrate, SNAP-25, in cell lysates of neurons derived from mouse embryonic stem
cells. The assay offers sensitivity of better than 0.1 LD50/ml (3 fM) which is not matched by other functional
assays, including the mouse bioassay, and provides serotype specificity for quantitative detection of BoNT/A and
anti-BoNT/A antitoxin. Subject to formal validation, the method described here could potentially be used as a
substitute for the mouse bioassay to measure potency and consistency of therapeutic products.

1. Introduction considerable ethical and safety limitations and alternative non-animal


methods have been sought as replacements for a number of years
Botulinum neurotoxins (BoNT/s) are considered the most lethal (Sesardic and Gaines Das, 2008; NIH Publication, 2008; Sesardic, 2012;
toxins (Gill, 1982). They induce prolonged muscle paralysis and block Adler et al., 2010).
muscle function through inhibition of neuronal transmitters from per- To date, seven serologically distinct botulinum toxin serotypes
ipheral cholinergic nerve endings (Rossetto, et al., 2014). Although (A–G) have been identified. They all comprise a 150 kDa single-chain
they are highly toxic and the causative agents of botulism, BoNTs have progenitor toxin which is subsequently activated by protease to gen-
been used for over twenty years in many medical interventions to treat erate a disulphide bond-linked structure containing a 50 kDa light chain
muscle hyperactivity, such as dystonia and spastic conditions, exocrine (LC) and a 100 kDa heavy chain (HC). The HC contains two functional
gland hyperactivity, such as hyperhidrosis, and pain disorders domains that are both required for toxin binding and uptake into the
(Dressler, 2012). A number of licensed products are manufactured nerve cells which involves a dual-receptor mechanism involving gang-
globally and for many years the in vivo mouse bioassay has been ap- liosides, such as GT1b, and a protein receptor. Once the toxin is in-
plied to determine toxin activity per vial, monitor lot to lot variability ternalized, the inter-chain disulphide bond is broken by reducing con-
and to define shelf life. Animals are still used for assessment of toxin ditions within the endocytic vesicles. This activates and releases the
potency by many manufacturers, for clinical diagnosis, and for potency endopeptidase light chain (LC) which specifically cleaves one of three
testing of antitoxins (Sesardic, 2012). The mouse bioassays have proteins involved in neurotransmitter release (Pirazzini et al., 2017).

Abbreviations: BoNT/A, botulinum neurotoxin A; SNAP-25, synaptosomal protein of molecular mass 25 kDa; EB, embryoid bodies; ESDN, embryonic stem cell-derived neurons; mESC,
mouse embryonic stem cells; HC, heavy chain; LC, light chain; LHn/A, recombinant endopeptidase fragment of BoNT/A LC and the N-terminus of the HC; ELISA, enzyme linked
immunosorbent assay; WB, Western blotting; PBS, phosphate buffered saline; CM, culture medium; RA, retinoic acid; LIF, leukemia inhibitory factor; GMEM, Glasgow's Minimal Essential
Medium; Shh, Sonic Hedgehog; FD, final differentiation; NGF-β, nerve growth factor beta; BDNF, brain-derived neurotrophic factor; RT, room temperature

Corresponding author.
E-mail addresses: gokhanyadirgi@yahoo.co.uk (G. Yadirgi), paul.stickings@nibsc.org (P. Stickings), shalini.rajagopal@nibsc.org (S. Rajagopal), Yvonne.Liu@nibsc.org (Y. Liu),
thea.sesardic@nibsc.org (D. Sesardic).

http://dx.doi.org/10.1016/j.jim.2017.09.007
Received 12 July 2017; Received in revised form 19 September 2017; Accepted 20 September 2017
0022-1759/ © 2017 Published by Elsevier B.V.

Please cite this article as: Yadirgi, G., Journal of Immunological Methods (2017), http://dx.doi.org/10.1016/j.jim.2017.09.007
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

The endopeptidase activity of BoNT LC is highly specific so that each (~ 35 LD50/ml), offers a sufficiently sensitive and robust method for
toxin has a unique substrate/target bond combination. Whereas the LCs potency testing of BoNT/A in Botox® (Fernandez-Salas et al., 2012).
of botulinum toxin serotypes A, E and C1 all cleave SNAP-25 (sy- The availability of neurons derived from pluripotent stem cells has
naptosomal associated protein of molecular mass 25 kDa), they target provided a new platform for highly sensitive detection of botulinum
this protein at distinct sites (Q197-R198, R180-I181, and R198-A199, neurotoxins (Kiris et al., 2011; McNutt et al., 2011, Pellet et al., 2011;
respectively) (Schiavo et al., 1993; Vadakkanchery et al., 1999; Whitemarsh et al., 2012; Beske et al., 2015; Jenkinson et al., 2017). In
Rossetto et al., 2014; Pirazzini et al., 2017). addition to the source of cells, development of cell based assays is
Once the molecular mechanism of BoNT/s was unravelled in the highly dependent on the use of a relevant and quantitative endpoint to
early 1990′s, it became possible to develop immunoassays designed to measure the botulinum toxin activity. Measurement of neuro-
detect the endopeptidase activity of the BoNT LCs. This approach re- transmitter release has been applied in assay development studies
quired production of a recombinant or synthetic substrate (SNAP-25 for (Keller and Neale, 2001; Rasetti-Escargueil et al., 2011; Hall et al.,
BoNT/A and BoNT/E or VAMP2 for BoNT/B) and a specific neoepitope 2004) and, while it is an important endpoint and research tool, it is
antibody, recognising only a peptide sequence that becomes exposed considered less useful for routine quality control purposes because it is
upon toxin-mediated cleavage of the substrate. Taking this approach, relatively nonspecific and highly dependent on cell type (Pellet, 2013).
serotype specific assays have been developed for several BoNT/s (Ekong Most recently, blockade of synaptic transmission in a network culture of
et al., 1997; Jones et al., 2008; Jones et al., 2009; Sesardic et al., 2004; neurons derived from human and rodent stem cells has been described
Simon et al., 2015). However, endopeptidase assays can suffer from as a functionally relevant readout for BoNT intoxication (Beske et al.,
unwanted matrix interference from albumin and human serum (Jones 2015, 2016; Jenkinson et al., 2017). However, this approach relies on
et al., 2011; Simon et al., 2015), cannot detect changes to the binding the use of specialized whole cell electrophysiology equipment or multi-
and translocation domains of the toxin, and can detect free LC activity electrode arrays which is not applicable in a routine control laboratory
in the absence of a functional binding domain, thus potentially giving setting and does not offer high throughput capability. Western blotting
false positive results (Sesardic, 2012). Furthermore, these methods are (Pellett et al., 2007, 2015, 2017, Whitemarsh et al., 2012; McNutt et al.,
not considered to be indicators of stability. While these endopeptidase 2011), quantitative immunofluorescence (Dong et al., 2004; Gilmore
assays are suitable for monitoring lot-to-lot consistency following va- et al., 2011; Kiris et al., 2011) or ELISA (Nuss et al. 2010; Pellett et al.,
lidation for a given toxin product, they cannot be considered as a full 2017) are methods that are all based on detection of intracellular
replacement for the mouse LD50 potency test (NIH Publication, 2008, substrates after cleavage by BoNT/s which is considered to be the most
Adler et al., 2010). specific endpoint and can be applied to any neuronal cell type.
To circumvent the limitations of the first generation of in vitro en- In this study, we report preliminary development of a simple cap-
dopeptidase assays, second generation methods were developed that ture ELISA method for detection of BoNT/A activity in neurons derived
require both functional binding to toxin receptor or antibody and cat- from mouse embryonic stem cells. Our results suggest that the assay is
alytic endopeptidase activity for detection of BoNT/A and BoNT/B likely to be suitable for testing the activity of therapeutic and com-
(Evans et al., 2009; Liu et al., 2012; Rosen et al., 2016). However, the mercial grade BoNT/A toxins and could also be used to measure the
concept of biochemical assays which mimic only two steps in the bo- potency of antitoxin to BoNT/A. The method described here is more
tulinum intoxication process is also considered limited as these assays sensitive than the mouse bioassay for detection of BoNT/A and other
do not reflect all the events required for toxin activity in vivo, such as cell based assays that have been developed for this toxin serotype.
the internalization step and, particularly, the persistence of toxin within
the cell, which are known to contribute to toxin potency (Keller and 2. Materials and methods
Neale, 2001).
Cell based bioassays for BoNT/s are superior to other in vitro models 2.1. Toxins
because they are capable of reflecting all major steps of botulinum toxin
action (Pellet, 2013) and they are therefore considered to offer the most Purified type BoNT/A haemagglutinin (HA) free toxin purchased
realistic strategy towards complete replacement of the mouse LD50 test from Metabiologics Inc., (Madison, USA, @ 2.3 × 10 (Dong et al.,
(NIH Publication, 2008; Adler et al., 2010). Primary cells have been 2007) LD50/mg, 1 mg/ml from Hall strain) was used. A working stock
used for BoNT research for many years, and the initial studies for cell was prepared at 20,000 LD50/ml (87 ng/ml) in Gelatine (0.2% w/v)
based methods focused on neurons derived from rat (Keller et al., Phosphate (50 mM di-sodium hydrogen orthophosphate) Buffer (GPB,
2004), mouse (Dong et al., 2007) and chicken embryos (Nuss et al., pH 6.5) as previously described (Jones et al., 2008). All stocks of ali-
2010). However, these studies did not offer sensitivity comparable to quoted toxins were stored at −80 °C prior to use.
the mouse bioassay since only nM rather than low pM concentrations of Two formulations of therapeutic BoNT/A toxin for injection, from
BoNT/A could be detected. The first highly sensitive cell based assay for the same manufacturer, containing different concentrations of BoNT/A
detection of BoNT/A was developed using primary rat embryonic spinal as active component were also used. Recombinant LHn/A fragment,
cord cells. Comparable sensitivity to the mouse bioassays was reported with an LD50 of 0.3 mg/mouse, was a generous gift from Syntaxin Ltd,
using detection of cleaved SNAP-25 quantified by Western blotting and was used as reported previously (Liu et al., 2012). All BoNT/A used
(Pellet et al., 2010). The approach to use primary rat spinal cord cells in this study were sub-type A1. Purified BoNT/B1 and BoNT/E3 were
for detection and quantification of botulinum neurotoxin antibodies purchased from Metabiologics (USA) and working stocks of 20,000 and
was also reported as an opportunity to replace the mouse toxin neu- 12,560 mouse LD50/ml, respectively were prepared in GPB buffer and
tralization test (Hall et al., 2004). Still, it was recognised that primary stored at − 80 °C before use.
cells do not provide an easily standardized and robust model that would
be preferred for routine laboratory use, and they also require animals 2.2. Antibodies
for the supply of cells.
Immortalized cell lines can offer the desired standardization prop- BoNT/A cleavage site-specific polyclonal anti-peptide antibody
erties, but they are typically at least 1000-fold less sensitive for BoNT/A against SNAP-25190–197 was made in rabbits as previously described
compared to the mouse bioassay (Purkiss et al., 2001; Yowler et al., (Ekong et al., 1997; Jones et al., 2008) and affinity purified against the
2002, Pellet, 2013), even after lengthy differentiation protocols and immunizing peptide. The same antibody was also custom made by Bio
addition of GT1b to improve sensitivity (Rasetti-Escargueil, et al., Trend (Chemikalin GmbH, Köln, Germany) and purchased affinity
2011). The notable exception is the cancer derived SiMa cell line purified to capture BoNT/A cleaved SNAP-25 from cell lysates.
(human neuroblastoma cell line) which, with an EC50 of ~1 pM Two separate polyclonal detection antibodies (sheep anti-SNAP-

2
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

251–57 and SNAP25111–157) were raised as previously reported (Ekong Cells were washed three times with PBS followed by application of
et al., 1997) and were used to detect captured SNAP-25 in order to the appropriate Alexa-conjugated secondary antibody (Invitrogen) - all
increase the signal and improve the sensitivity of the ELISA. Optimal used at 1:500 dilution in 1% (v/v) BSA in PBS, and incubated in the
results were obtained by mixing two antibodies together at equivalent dark for 2 h at RT. Application of secondary antibody in the absence of
concentrations. primary antibody was used as a negative control. Cells were counter-
Botulinum antitoxin equine type A (NIBSC product code 59/021), stained with DAPI (1 μg/ml, Sigma) and coverslips were mounted with
with assigned potency of 2000 IU/ampoule, and botulinum antitoxin Fluoroshield (Sigma). Cells were visualised using a fluorescent micro-
equine type E (NIBSC product code 02/318), with assigned potency of scope (Olympus IX73).
197 IU/ampoule, were used in the toxin neutralization assays (Jones
et al., 2006). 2.5. Treatment of cells

2.3. Maintenance and neuronal differentiation of mouse embryonic stem For the ELISA based detection assay, all treatments were performed
(mES) cells on 96-well plates (Nunc), with ESDNs seeded at a density of
4–5 × 104 cells per well and differentiated between 9 and 12 days
The mES cell line used in this study was E14tg2a (provided by Dr. (DIV9 to DIV12). Initial studies included treatment of cells from DIV3.
Jennifer Nichols of Cambridge University, Welcome Trust Centre for Toxin dose finding and time course studies involved addition of a range
Stem Cell Research). These feeder-independent pluripotent cells were of concentrations of BoNT/A (0.08 to 40 LD50/ml) to DIV9 differ-
maintained at 37 °C in a 5% (v/v) CO2 incubator in 0.2% gelatin-coated entiated cells in 200 μl volumes of cell culture medium and incubating
flasks (Nunc) in culture medium (CM) made up of Glasgow's Minimal tissue culture plates for different lengths of time (12, 24, 48 or 72 h) at
Essential Medium (GMEM, Invitrogen) supplemented with 10% ESC- 37 °C.
qualified fetal bovine serum (Invitrogen), 10 ng/ml Leukemia in- In all other studies, ESDNs were used at DIV9 were used unless
hibitory factor (LIF, Invitrogen) and 0.1 mM 2-mercaptoethanol. otherwise indicated. Purified BoNT/A, BoNT/B or BoNT/E, or com-
Passage 18 of mES cells (1 × 10) (Bigalke et al., 1985) were cultured in plexed BoNT/A, from therapeutic preparations diluted in DM, were
a T25 flask (Nunc) for 3 days until 90–95% confluence was reached. To added in 200 μl volumes (range 0.002 to 800 LD50/ml) and cells were
form embryoid bodies (EBs), cells were washed with pre-warmed PBS incubated for 48 h at 37 °C. For specificity studies, inactivated BoNT/A
prior to trypsinization [2 ml trypsin-EDTA/PBS (1:1)] for 60 s. Re- or recombinant LHn/A (at molar excess) were used. Controls included
sulting clumps of 100 to 200 cells (primed to form small and large sized wells with cells but no toxin or treatment.
EBs) were transferred into bacterial grade Petri dishes (Nunc) in 12 ml A BoNT/A toxin neutralization study was performed to determine
of CM without LIF, for 24 h (Day 1). the concentration of botulinum type A reference antitoxin needed to
EBs were treated with 1 μM retinoic acid (RA, Sigma) for two con- prevent intracellular cleavage of SNAP-25 induced by a fixed dose
secutive days in fresh CM without LIF (Day 2 and 3). On Day 4, Sonic (20 LD50/ml, 4 LD50/dose) of BoNT/A. Toxin was pre-incubated for
Hedgehog (Shh) 100 ng/ml, R & D Systems) and purmorphamine (1 μM, 1 h at RT with an equal volume of botulinum type A or E reference
Calbiochem) was added to cultures for 24 h. On Day 5 EBs were antitoxin (ranging between 0.1 IU/ml and 0.03 mIU/ml) in a separate
transferred to new dishes containing CM without LIF, supplemented 96-well tissue culture plate. All dilutions were performed in DM and
with glial cell-derived neurotrophic factor (GDNF, 100 ng/ml, R & D 200 μl of toxin/antitoxin mixture was added to ESDNs at DIV12 and
Systems) and brain-derived neurotrophic factor (BDNF, 100 ng/ml, tissue culture plates incubated for 48 h at 37 °C. Controls in this study
R & D Systems), for 60 h. included cells exposed to antitoxin alone with no toxin treatment.
After 9–12 days, EBs were dissociated into single cell cultures by For Western blotting studies, cells were plated into laminin-coated 6
washing with PBS and then incubating for 5 min with trypsin-EDTA/ well plates at 1 × 106 cells per well and treated with BoNT/A toxin
PBS (1:1) at 37 °C in a 5% (v/v) CO2 incubator. Dissociation into single (diluted in DM, ranging from 0.2–125 LD50/ml) for 48 h.
cells was encouraged using mechanical force by pipetting. For the final
differentiation (FD) into embryonic stem cell derived neurons (ESDNs), 2.6. Preparation of cell lysate
single cells were seeded at a density of 4–5 × 104 cells/well on laminin
coated surfaces (1 μg/well, Invitrogen) in 96-well plates (Nunc). For At the end of the treatment, cells were carefully washed once for
this FD stage, cells were maintained in differentiation medium (DM) 5 min with PBS. Total protein from cells was harvested by lysing with
consisting of: Neurobasal medium (Invitrogen) supplemented with 1× 110 μl per well (for 96 well plates) or 400 μl per well (for 6 well plates)
B27 (Invitrogen), 0.5 mM L-glutamine and nerve growth factor beta of freshly M-PER lysis buffer (Mammalian Protein Extraction Reagent,
(NGF-β, 20 ng/ml, Sigma). Media was changed every 2–3 days for up to Pierce 87786) containing EDTA-free protease inhibitor (Pierce) and
21 days (DIV21). DIV indicates number of days that cells are cultured as 150 mM NaCl. Cell lysates were stored at − 20 °C until use in ELISA or
a monolayer in the final differentiation medium. Western blotting.

2.4. Immunocytochemistry and cell imaging 2.7. Immuno-detection of cleaved SNAP-25 by ELISA

Cells at the FD stage were plated on laminin coated (20 μg/ml) A schematic diagram showing the general principle of the capture
16 mm glass coverslips (Fisher Scientific), and differentiated for 3, 10 ELISA for immuno-detection of BoNT/A cleaved SNAP-25 is shown in
or 21 days in vitro (DIV3, 10 or 21 respectively). ESDNs were fixed for Fig. 1. Polystyrene Maxisorp 96-well plates (Nunc) were coated with
10 min in 4% paraformaldehyde, permeabilized with either Tween-20 50 μl/well of affinity purified capture antibody against SNAP-25190–197
(0.2% v/v) or TritonX-100 (0.2% v/v) in PBS for 10 min and blocked diluted to 5 μg/ml in 50 mM carbonate buffer (pH 9.6). Sealed plates
with 5% BSA for 1 h at room temperature (RT). Primary antibodies were incubated overnight at 4 °C. The coating antibody was decanted
were from Abcam unless otherwise stated, and were diluted in 1% (v/v) and plates were blocked for 1 h at RT with 250 μl per well of 2.5% (w/
BSA in PBS and applied overnight at 4 °C: rabbit anti-Map2 (1:250; v) of skimmed milk powder (Marvel) in PBS, containing 0.05% v/v
ab32454); mouse anti-Tau (1:250; ab80579); mouse anti-VAMP-1/2/3 Tween-20 (M-PBST). Plates were then washed 3 times with PBST,
(1:100; Santa Cruz Biotechnology sc-133,129); rabbit anti-SV2A R-300 which was the same for all subsequent wash steps. Cell lysates (50 μl
(1:1000; Santa Cruz Biotechnology sc-28,955); mouse anti-synapto- per well) were added together with an equal amount of M-PBST and
tagmin I/II (1:500; ab13259); mouse anti-choline acetyltransferase plates were shaken at 400 rpm for 10 min, and then incubated without
(1:250; ab68779) and, mouse anti-noradrenaline (1:250; ab8887). shaking for 90 min at RT. M-PBST (100 μl per well) was added to buffer

3
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

Fig. 1. Schematic overview of the capture ELISA for immuno-detection of BoNT/A cleaved SNAP-25. BoNT/A cleaves SNAP-25 (pink) between amino acids 197 and 198 in embryonic
stem cell derived neurons, and the cleavage product is captured using a specific neoepitope antibody raised against a peptide corresponding to amino acids 190–197 of SNAP-25 (SNAP-
25190–197). This antibody only recognises the BoNT/A-cleaved SNAP-25 and does not recognise intact SNAP-25. The captured cleavage product is then detected using two polyclonal
detection antibodies that bind to two distinct sites, SNAP-251–57 and SNAP-25111–157. (For interpretation of the references to colour in this figure legend, the reader is referred to the web
version of this article.)

control wells. After washing, an equal concentration of polyclonal 165 V for 60 min and then transferred onto 0.2 μm nitrocellulose
sheep antibodies raised against SNAP-25 epitopes (1–57, 111–157), membranes at 10 V for 60 min using Bolt transfer buffer, both using a
diluted 1/1000 from 10 mg/ml stock in M-PBST was added to plates Novex Bolt system (Invitrogen).
(100 μl per well), which were then incubated for 90 min at RT. Plates Presence of total or truncated SNAP-25 in cells before and after
were washed and 100 μl per well of rabbit anti-sheep-HRP conjugate treatment with BoNT/A was performed by incubating membranes with
(Thermo 31480) diluted 1:16,000 in M-PBST was added. Plates were commercially, or in house, sourced antibodies to SNAP-25 (monoclonal
incubated for 90 min at RT followed by a final wash and addition of mouse antibody from Synaptic systems #111 111, rabbit anti-SNAP-25
100 μl/well of substrate solution [50 mM citric acid pH 4.0, 0.05% w/v from abcam #ab109105 or rabbit antibody to SNAP-25190–197) diluted
ABTS (2,2′-Azino-bis(3-thylbenzothiazoline-6-sulfonic acid) diammo- in PBS with 1% skimmed milk powder (Marvel). Specific detection was
nium salt (A9941, Sigma), and 0.05% v/v of 30% Hydrogen Peroxide visualised by addition of either goat anti-mouse IgG (Sigma, A9044) or
Solution (Merck)]. Colour was allowed to develop for 30 min at RT goat anti-rabbit IgG (Sigma, A0545) conjugated to HRP, followed by
prior to reading absorbance at 405 nm using a Multiskan plate reader. detection using ECL2 chemiluminescence reagent (Pierce 80,196) ac-
cording to the manufacturer's instructions. Gels were developed and
capture images recorded using Gel Logic 2200 Pro Imaging Systems.
2.8. Immuno-detection of cleaved SNAP-25 by Western blotting

For Western blotting studies, 20 μg of total protein from cell lysates, 3. Results
diluted in Tris-Tricine sample buffer containing 2% β-mercaptoethanol,
was separated on a 10–12% Tris-Tricine gel (Bio-Rad 456–3113) with 3.1. Differentiation and characterization of mouse ES cells
Tris-Tricine SDS buffer under denaturing conditions. Gels were trans-
ferred onto 0.2 μm nitrocellulose membrane (Bio-Rad 162–0212) using Differentiated cells were fully characterized prior to assay devel-
a wet transfer system, blocked with PBS containing 5% skimmed milk opment by immunocytochemistry as shown in Fig. 2. By day 10 of
powder and exposed to primary antibody. Alternatively, cell lysates differentiation (DIV10), presence of complex axodendritic arbours and
were diluted with 4 × LDS sample buffer, 10 × reducing agent (both networks of cells are observed appearing together with positive im-
from Invitrogen) and ultrapure water, to 7.5 μg total protein, denatured munostaining for specific markers of neuronal differentiation, axonal
at 70 °C for 10 min, then loaded onto 12% Bis-Tris Plus gels marker Tau and dendritic marker MAP-2 (Fig. 2a). At DIV21, im-
(Invitrogen). Molecular weight markers were a 1:1 mixture of Novex munostaining confirmed presence of synaptotagamin I/III and SV2A,
Sharp pre-stained protein standard and MagicMark XP western protein indicating presence of botulinum toxin specific receptors (Fig. 2b). At
standard (Invitrogen). Gels were run with Bolt MES running buffer at DIV21, immunostaining confirmed the presence of choline

4
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

Fig. 2. Characterization of neurons derived from mouse ESDNs by immunostaining and Western Blotting. (a) Cells at DIV10 show positive immunostaining for specific markers of
neuronal differentiation: Tau (red) and MAP-2 (green). (b) Cells at DIV21 cell show positive immunostaining for synaptotagmin I/III (green), SV2A (red) and Tau (grey). Cells at DIV21
have positive immunostaining for neurotransmitters: c) choline acetyltransferase (red) and Tau (green) and d) noradrenaline (red) and Tau (green). Cells at DIV21 also had positive
immunostaining for VAMP 1/2/3 (green), SV2A (red) and Tau (grey). Scale bar is 50 μm, and nuclear counterstaining is shown in blue. (f) Western blotting for presence of SNAP-25 in
differentiated cells at DIV12 (lane 2) DIV13 (lane 3), DIV14 (lane 4), DIV19 (lane 5) and DIV21 (lane 6). Lanes 1 and 7 are molecular weight markers. (For interpretation of the references
to colour in this figure legend, the reader is referred to the web version of this article.)

acetyltransferase and noradrenaline neurotransmitters (Fig. 2c and d SNAP-25 cleavage product in samples exposed to BoNT/A at con-
respectively), and the presence of VAMP-1/2/3 (Fig. 2e). SNAP-25 ex- centrations of 125, 25 and 5 LD50/ml but not in the control sample,
pression in differentiated cells was confirmed by Western blotting using with a very faint band also visible in the lysate from samples exposed to
antibody detecting only intact SNAP-25 (Fig. 2f). 1 LD50/ml (Fig. 3b).

3.2. Western blotting 3.3. Capture ELISA for detection of BoNT/A cleaved SNAP25 from
differentiated mouse ES cells
Western blotting was also used to confirm cleavage of SNAP-25 in
lysates from ESDNs that had been treated with a range of BoNT/A In a preliminary study, ESDNs at DIV3 and DIV9 were treated with
concentrations (125, 25, 5.0, 1.0 and 0.2 LD50/ml). Immunodetection pure BoNT/A (80–0.039 LD50/ml = 16.0–0.0078 LD50/dose) for 48
was performed using two different antibodies: a mouse monoclonal and 72 h. One set of cells from DIV3 were also given the same treatment
antibody recognising both cleaved and un-cleaved SNAP-25 (Fig. 3a) but BoNT/A was incubated with cells for 24 h after which time the
and a rabbit polyclonal antibody specific for BoNT/A cleavage site only toxin was removed and cells incubated for further 48 h in culture
(Fig. 3b). The mouse monoclonal antibody was able to detect intact medium without toxin. Results from this preliminary study (Fig. 4a)
SNAP-25 in all samples and there was a visibly reduced intensity of the show a positive dose response for all 3 treatments, with a dose de-
band corresponding to intact SNAP-25 at the highest concentration of pendent increase in the detection of cleaved SNAP-25. There was a clear
BoNT/A (125 LD50/ml). The reduction of intensity of signal for intact increase in sensitivity when the differentiation time increased from
SNAP-25 was accompanied by the appearance of a band corresponding DIV3 to DIV9 despite the longer incubation time with toxin at DIV3
to the smaller SNAP-25 cleavage product, and this band was clearly (EC50 of 3.25 LD50/ml for DIV3 and 1.08 LD50/ml for DIV9). A shorter
visible at the highest BoNT/A concentrations of 125 and 25 LD50/ml exposure to BoNT/A followed by 2 days of culture in medium without
(Fig. 3a). The rabbit polyclonal (neoepitope) antibody detected the toxin only did not improve sensitivity (EC50 of 7.17 LD50/ml).

5
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

Fig. 3. Detection of cleaved SNAP-25 from ESDNs treated with BoNT/A, by Western blotting. (a) Presence of SNAP-25 and BoNT/A cleaved SNAP-25 in lysed ESDNs at DIV12 treated for
48 h with 125 (lane 3), 25 (lane 4), 5.0 (lane 5), 1.0 (lane 6) and 0.2 (lane 7) LD50/ml of BoNT/A. Lane 2 = control (no toxin). Lane 1 = molecular weight markers. Blots were visualised
with mouse monoclonal antibody, recognising both cleaved and un-cleaved SNAP25. (b) Presence of BoNT/A cleaved SNAP-25 in lysed ESDNs at DIV12 treated for 48 h with 125 (lane 3),
25 (lane 4), 5.0 (lane 5), 1.0 (lane 6) and 0.2 (lane 7) LD50 of BoNT/A. Lane 2 = control (no toxin). Lane 1 = molecular weight markers. Blots were visualised with rabbit BoNT/A
cleavage site specific antibody, detecting only cleaved SNAP-25.

A preliminary time course study using ESDNs at DIV9 was per- Table 1
formed by incubating cells with BoNT/A, 80–0.039 LD50/ml Variation in EC50 values for different preparations of ESDNs and different ELISA plates.
(16–0.0078 LD50/dose) for between 12 and 72 h (Fig. 4b), followed by
Plate No. Cell batch 1 Cell batch 2 Cell batch 3
detection of cleaved SNAP-25 by ELISA. Assay sensitivity increased on
extended exposure to toxin, with EC50 values (LD50/ml) ranging from DIV9 DIV9 DIV12
9.41 at 12 h to 0.74 LD50/ml at 72 h, although there was little differ-
1 1.08 1.70 1.94
ence beyond the 48 h time point.
1.82
The dose response and EC50 for BoNT/A was confirmed for two 2 1.06 1.15 1.10
different preparations of ESDNs at DIV9 and for a third cell preparation 1.26
at DIV12, and in each case cleaved SNAP-25 was measured on at least 3 1.46 1.52 ND
two ELISA plates. The results (shown as EC50 in LD50/ml) are sum- 1.43
1.41
marised in Table 1. Taking all the data together, the minimum level of
1.41
detection was determined as at least 0.1 LD50/ml (3 fM) with an EC50 4 ND 1.42 ND
of 1.39 (1.24–1.55) LD50/ml (6 pg/ml; 45 fM). The EC50 data from all
3 cell batches for all plates were log10 transformed and a random effects Data are the EC50 for the dose response to pure BoNT/A (48 h treatment). Results are
model was used to determine between-plate variance, which was con- shown for 3 different cell batches (DIV9–DIV12), titrated on up to 4 plates with single or
replicate titrations per plate as indicated. EC50 values were calculated from log trans-
verted to geometric coefficient of variation (GCV). The between-plate
formed data normalised as a percentage of the largest value in each data set. ND = not
GCV was estimated as 22.4%. done.

activity even when this component was used at a 4-order of magnitude


3.4. Specificity of the capture ELISA for active BoNT/A
higher concentration (Fig. 5b).
The specificity of the cell based assay with SNAP-25 capture ELISA
was determined using botulinum toxin from serotype B (which cleaves 3.5. Detection of BoNT/A in pharmaceutical products
a different SNARE protein) and E (which cleaves SNAP-25 but at a
different position to toxin A). A dose dependent cleavage of SNAP-25 The cell based assay was used to measure the activity of two
was confirmed for BoNT/A but there was no measureable response for pharmaceutical products containing BoNT/A in the form of complexed
either the B or E toxin and OD values for these samples were compar- toxin with bulking/stabilising excipients. Both products had a very si-
able to those seen for cells incubated without any toxin (Fig. 5a). The milar dose response profile to the commercial BoNT/A (Fig. 6) sug-
positive response for toxin A was also absent in lysates from cells that gesting that this assay might be suitable to assess products of this type.
had been incubated with a heat inactivated BoNT/A preparation Using the commercial BoNT/A as a reference toxin, the precision of the
(Fig. 5a). Specificity of the assay for active A toxin was further con- relative potency estimate for both product 1 and product 2 was calcu-
firmed using the non-toxic fragment of the BoNT/A (BoNT/A LHn) lated as 85–118% which complies with recommended validity criteria
which lacks the receptor binding domain. There was no detectable for a valid potency estimate according to the European Pharmacopeia

a 3.0 DIV9 (48h) b


3.0 DIV9 (12h)
DIV3 (72h)
DIV9 (24h)
2.5 DIV3 (24h + 48h)
2.5 DIV9 (48h)
DIV9 (72h)
2.0 2.0
OD 405 nm

OD 405 nm

1.5 1.5

1.0 1.0

0.5 0.5

0.0 0.0
0.01 0.1 1 10 100 0.01 0.1 1 10 100
BoNT/A LD50/ml BoNT/A LD50/ml

Fig. 4. Detection of cleaved SNAP-25 from ESDNs treated with BoNT/A by capture ELISA. (a) cells were differentiated for 3 (DIV3) or 9 (DIV9) days on 96-well tissue culture plates and
treated with 200 μl purified BoNT/A (80 to 0.04 LD50/ml;16 to 0.008 LD50/well) for 24, 48 and 72 h. (b) cells at DIV9 were exposed to same dose range of pure BoNT/A (80 to
0.04 LD50/ml) for 12, 24, 48 and 72 h. Results presented are from one assay and each data set is from a single titration. In both figures the dotted line shows the absorbance reading for
control cells control without toxin.

6
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

a 1.5 b
BoNT/A
BoNT/B 1.5

BoNT/E
1.0 BoNT/A (Heat Inactivated)
OD 405 nm

BoNT/A LHn
1.0

OD 405 nm
BoNT/A

0.5
0.5

0.0 0.0
0.001 0.01 0.1 1 10 100 1000

-8

-6

-4

-2

4
-1

10

10

10
10

10

10

10
10
BoNT/A LD50/ml BoNT/A [µg/ml]

Fig. 5. Specificity of capture ELISA for detection of biologically active BoNT/A. ESDNs at DIV9 were incubated for 48 h with (a) purified BoNT/A, BoNT/B, BoNT/E (650 to 0.0037 LD50/
ml) and equivalent concentration of heat inactivated BoNT/A. The heat inactivation of Metabiologics pure holotoxin of BoNT/A1 was achieved by boiling at 95 °C for 5 min. b) purified
BoNT/A (10− 2 and 10− 8 μg/ml) and non-toxic fragment of BoNT/A, LHn/A (300 to 0.0017 μg/ml) lacking receptor binding domain. Results are from a single assay and each data set is
from a single titration. Dotted lines in each experiment indicate controls where cells were not exposed to any treatments.

1.5 1.5
BoNT/A Anti-A + BoNT/A
Product 1 Anti-E + BoNT/A
Product 2 Anti-A
1.0 1.0
OD 405 nm

OD 405 nm
0.5 0.5

0.0 0.0
0.0001 0.001 0.01 0.1 1 10 100 1000 0.00001 0.0001 0.001 0.01 0.1 1
BoNT/A LD50/ml Antitoxin IU/ml

Fig. 6. Detection of cleaved SNAP-25 from ESDNs treated with BoNT/A in therapeutic Fig. 7. Inhibition of BoNT/A cleavage of SNAP-25 in ESDNs by antibodies against bo-
formulations. After 48 h incubation cells were lysed and the lysates from each individual tulinum A toxin. Cells at DIV12 were treated for 48 h with a mixture of purified BoNT/A
well were subjected to BoNT/A specific capture ELISA to detect cleaved SNAP-25. Dotted toxin (20 LD50/ml; 4 LD50/well) and reference standard for type A antitoxin (NIBSC
lines are cell controls with no toxin. Results are from a typical experiment performed on product code 59/021), or reference antitoxin for BoNT/E (NIBSC product code 02/318),
two independent occasions and each data set is the mean from triplicate titration ± SD. used in the range between 0.1 IU and 0.05 mIU. Results are from a representative ex-
periment where each data set is a mean from a duplicate titration ± SD. Negative controls
included cells treated with reference antisera against BoNT/A in the absence of BoNT/A
monograph for Botulinum toxin A for injection (01/2012:2113). toxin (n = 1).

3.6. Neutralization of BoNT/A effect on neurons from mouse EC cells


based assay for detection of BoNT/A reported to date.
determined by capture ELISA
During the past decade, much effort has focused on development of
sensitive assays for BoNT/s which could provide sensitivity equivalent
To evaluate the suitability of the cell based assay for measuring
to the mouse bioassay. Primary rodent neuronal cells have been used in
antitoxin potency, neutralization assays were performed with a fixed
BoNT research for many years (Keller et al., 2004; Bigalke et al.,1985;
dose (20 LD50/ml) of pure BoNT/A which corresponds to 4 LD50/dose
Dong et al., 2007), but it was not until rat spinal cord cells were first
(~ 15 pg of pure BoNT/A per dose). This dose of toxin had been shown
applied for quantitative detection of purified BoNT/A that a cell based
to be towards the top of the linear part of the dose response curve. This
assay for potency determination was realistically possible (Pellet et al.,
concentration of toxin was neutralized by anti-A antitoxin in a dose
2010). This assay is dependent on immunodetection of cleaved and
dependent manner in the range between 1 and 100 mIU/ml (Fig.7).
uncleaved SNAP-25, quantified by Western blotting (WB) using a
There was no neutralization of BoNT/A activity in the presence of an
commercial antibody that detects both molecules. The level of sensi-
equine polyclonal antibody to BoNT/E further demonstrating the spe-
tivity (EC50) was reported to be 133 LD50/ml (5.7 pg or 3.8 LD50/
cificity of the assay and confirming that neutralization is serotype
well) which was claimed to be comparable to the mouse bioassay
specific. Furthermore, the anti-A antitoxin alone had no effect on SNAP-
(Pellet et al., 2010). However assays using primary neuronal cells do
25 cleavage (Fig. 7).
not provide an easily standardized and robust model suitable for rou-
tine laboratory use, require relatively lengthy manipulation of cells, and
4. Discussion use of animals as a source of the cells. Assays using these cells were
therefore not used widely as an alternative to the animal potency test,
Cell based assays for detection of BoNT/s are considered to be the despite their high sensitivity in comparison to all other established cell
most suitable approach to replace the mouse bioassay. This is due to the lines (Pellet, 2013; Hubbard et al., 2012).
fact that they are capable of reflecting all key steps required for toxin Availability of neurons derived from pluripotent stem cells (Bibel
activity in vivo and can be standardized and applied for high et al., 2004, 2007) has significantly contributed to the field of cell based
throughput routine testing of therapeutic toxin products (NIH assay development and provided a new platform for highly sensitive
Publication number, 2008; Adler et al., 2010). In this study we report detection of BoNT/s. Improvements in procedures for the culture and
preliminary development of a cell based assay that has a sensitivity differentiation of stem cells, and reduction in cost of critical reagents,
exceeding that of the mouse bioassay and is the most sensitive cell

7
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

has further advanced the field of assay development for detection of BoNT/A for 48 h and effect of toxin on SNAP-25 determined by WB and
BoNT/A (Hubbard et al., 2012; Kiris et al., 2014). capture ELISA (Pellett et al., 2017). In the capture ELISA, the EC50 for
Several studies have used mESC combined with immunoblotting to both HIP and iCell neurons was 0.34 LD50 per dose which is very
monitor dose dependent proteolysis of SNAP-25 by BoNT/A (McNutt comparable to our findings presented in this study using ESDNs. The
et al., 2011; Pellet et al., 2011; Kiris et al., 2011; Hubbard et al., 2012,) capture ELISA was more sensitive compared to WB where EC50s of 0.49
and other BoNT/serotypes (Hubbard et al., 2012). McNutt et al. (2011) and 1.5 LD/50 per dose were reported for iCell and HIPS neurons, re-
determined an EC50 of 0.4 pM for BoNT/A in mESC derived neurons at spectively. Different detection antibodies were used in the capture
DIV12 (based on detection of cleaved SNAP-25 by WB) following 48 h ELISA and WB which may contribute to the differences in sensitivity.
treatment with toxin. Pellet et al. (2011) reported a minimum detection Whereas human stem cells may be the preferred option in some studies,
of 104 fM, with EC50 of 1.7 pM, using neurons from mES cells after more robust differentiation and a higher protein yield in a 96 well
9 days in culture and 24 h incubation with pure BoNT/A. A study by format makes mES cells more attractive for replacement of the mouse
Hubbard et al. (2012) confirmed only 50% cleavage of SNAP-25 with bioassay in the majority of applications currently relying on animal
67 pM of BoNT/A in enriched glutamatergic neurons from mESCs. Kiris models. It is worth noting that the low protein yield in cell lysates from
et al. (2011) used a high-throughput ELISA method combined with human neurons necessitated modification of capture ELISA to improve
immunofluorescence to monitor disappearance of full length SNAP-25 the signal detection (Pellett et al., 2017).
using BoNT/A cleavage sensitive antibodies. In this study they reported Detection of cleavage products is specific and quantitative for
a dynamic dose response range between 25 and 1000 pM for BoNT/A, BoNTs and therefore is a desirable endpoint for potency determination
which is considerably inferior in sensitivity compared to results pre- of pharmaceutical BoNTs. An ELISA based detection method for cleaved
sented in this study. Furthermore, in the previous study of Nuss et al. SNAP-25 has been developed for detection of BoNT/A in Botox®
(2010) using these same antibodies, sensitive to the BoNT/A cleavage pharmaceutical products as a replacement of the mouse LD50 potency
site, in combination with primary neuronal cells (embryonic chicken test (Fernandez-Salas et al., 2012). In this study, differentiated SiMa
spinal motor neurons) detected BoNT/A only in the range between 0.1 cells (human neuroblastoma cell line) were used in an ELISA format
and 10 nM. incorporating a monoclonal antibody detecting cleaved SNAP-25. The
Our method includes a capture ELISA read-out which is comparable detection of BoNT/A in Botox® formulations was reported with an EC50
to studies of Kiris et al. (2011) and Nuss et al. (2010) and offers sig- of 4–6 LD50/ml (1.0–0.4 LD50/well), for BoNT/A complex an EC50 of
nificantly higher throughput capacity compared to WB, with higher 3.3 pM, and for pure BoNT/A an EC50 of 0.12 nM. In our study, EC50s
sensitivity. The assay reported in this study has a minimum detection for formulated HA complexed toxin products were ~1.0 LD50/ml and
level of 0.1 LD50/ml which is equivalent to 0.02 LD50 per dose for pure BoNT/A ~1.5 LD50/ml (48 fM) confirming that ESDNs are
(0.44 pg/ml or 3 fM) and an EC50 of 1.5 LD50/ml (6.5 pg/ml; 48 fM). more sensitive to BoNT/A compared to SiMa cells.
Since neurons derived from mESC were also used in previous studies, it There are limited reports describing cell based assays for toxin
is likely that the differentiation protocol and format of the capture neutralization from which an appropriate level of sensitivity can be
ELISA, including the capture and detection antibodies used contributed determined. Pellett et al. (2007) used rat primary neuronal cells and
to the enhances detection of BoNT/A in comparison to previous studies. confirmed detection of antibodies in patients resistant to BoNT/A
One key difference to the method previously reported (Nuss et al., therapy. The authors did not include an International Standard to de-
2010; Kiris et al., 2011) is that in our approach, the capture antibody termine the level of sensitivity, but applied a theoretical value that one
used in the ELISA specifically and selectively detects only BoNT/A- International Unit of reference type A antitoxin would neutralize 104
cleaved SNAP-25 product so that appearance of product, rather than mouse LD50 units. They used this extrapolation to estimate potencies of
loss of SNAP-25 is detected. The same antibody has been used pre- 6–7.5 mIU/ml for samples derived from patients resistant to toxin
viously in development of highly sensitive biochemical assays for therapy. However, it should be noted that this extrapolation is not
BoNT/A, that have even higher sensitivity compared to the cell based necessarily transferrable to systems other than the mouse bioassay.
assay (Jones et al., 2008; Liu et al., 2012), but suffer from the limitation Furthermore, the mouse toxin neutralization assay has been confirmed
previously noted that they are incapable of reflecting all key steps in- not to be sufficiently sensitive to detect low levels of serum antibodies
volved in toxin activity in vivo. It is worth noting that antibody de- in patients resistant to toxin therapy (Göschel et al., 1997). Hall et al.
tecting only the BoNT/A cleaved SNAP25, and used as a capture anti- (2004) used rat embryonic spinal cord cells combined with a [3H]
body, was at least 25-fold more effective in detecting this fragment glycine release assay and reported a linear dose response in the range of
compared to the commercially sourced antibody detecting both cleaved 0.001 to 0.1 IU/ml. This is comparable to our study where we confirm a
and un-cleaved SNAP-25 as determined by WB studies reported here. similar sensitivity and dose response for the same International Re-
This suggests that the cleavage site specific antibody plays an important ference Standard for type A antitoxin. It should also be noted that the
role in determining sensitivity of the assay. In addition to the capture detection limit for anti-A antitoxin in the cell based assay we describe
antibody, the cell based assay described here includes two separate here is likely to be enhanced when a lower toxin dose is used in the
detection antibodies directed against SNAP-25111–157 and SNAP251–57 neutralization assay.
(regions of SNAP25 protein which are unaffected by toxin-mediated Most recently, the blockade of synaptic transmission in network
proteolysis) that are used to amplify the detection signal. Although the cultures of neurons derived from stem cells has been described as a
study presented here does not represent a formal validation, the results functionally relevant readout for BoNT intoxication (Beske et al., 2015,
obtained suggest that the assay is likely to be suitable for development 2016, Jenkinson et al., 2017). Using stem cell derived neurons on multi
into a robust, high throughput routine assay. Although the dose re- electrodes, Jenkinson et al. (2017) confirmed a significant reduction of
sponse curve and absolute EC50 differs between plates (from the same both spontaneous network bursts and average spike rates with 1.66 pM
cell batch) and between cell batches, it should be noted that in routine of BoNT/A and 33 LD50/ml of Botox®. Beske et al. (2016) measured
use test toxin or antitoxin samples would be titrated alongside a re- spontaneous neurotransmission frequency on neurons derived from
ference (as is the case for the mouse bioassay) which should sig- mouse ESC and reported synaptic activity for BoNT/A with an IC50 of
nificantly reduce the between plate and between cell batch variability 0.05pM for BoNT/A. Exposure of mESC to clinical concentrations of
observed in this preliminary study. Botox® demonstrated that exposure to 1 or 2 LD50/ml resulted in re-
Recent studies on neurons derived from human induced pluripotent duced neurotransmission and production of cleaved SNAP-25 after 20 h
stem cells also confirm highly sensitive detection of BoNTs (Pellett exposure. The sensitivity to BoNT/A reported in these studies is com-
et al., 2015; Whitemarsh et al., 2012). Two commercially sourced parable to the level of detection presented in our study for both com-
human stem cell derived neurons (iCell and HIP™) were exposed to pure mercial BoNT/A and therapeutic toxin preparations. However, despite

8
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

being functionally relevant, the measurement of the blockade of sy- Gill, D.M., 1982. Bacterial toxins: A table of lethal amounts. Microbiol. Rev. 46, 86–94.
Gilmore, M.A., Williams, D., Okawa, Y., Holguin, B., James, N.G., Ross, J.A., Aoki, R.K.,
naptic transmission as an endpoint relies on the use of specialized Jameson, D.M., Steward, L.E., 2011. Depolarization after resonance energy transfer
equipment which is not readily available in most routine control la- (DARET): a sensitive fluorescence-based assay for botulinum neurotoxin protease
boratory settings, and less favourable for high throughput capability. activity. Anal. Biochem. 413, 36–42. http://dx.doi.org/10.1016/j.ab.2011.01.043.
Göschel, H., Wohlfarth, K., Frevert, J., Dengler, R., Bigalke, H., 1997. Botulinum A toxin
In conclusion, our study shows that neurons from mESCs combined therapy: neutralizing and nonneutralizing antibodies-therapeutic consequences. Exp.
with a simple capture ELISA for detection of toxin-cleaved substrate Neurol. 147, 96–102. http://dx.doi.org/10.1006/exnr.1997.6580.
could be used to develop a highly sensitive in vitro assay for measuring Hall, Y.H., Chaddock, J.A., Moulsdale, H.J., Kirby, E.R., Alexander, F.C., Marks, J.D.,
Foster, K.A., 2004. Novel application of an in vitro technique to the detection and
the potency of BoNT/A and anti-BoNT/A antitoxin. Subject to formal quantification of botulinum neurotoxin antibodies. J. Immunol. Methods 288, 55–60.
validation, this assay could serve as a complete replacement for mouse http://dx.doi.org/10.1016/j.jim.2004.02.011.
bioassays currently used to measure activity of BoNT/A toxin and an- Hubbard, K.S., Gut, I.M., Lyman, M.E., Tuznik, K.M., Mesngon, M.T., McNutt, P.M., 2012.
High yield derivation of enriched glutamatergic neurons from suspension-cultured
titoxin.
mouse ESCs for neurotoxicology research. BMC Neurosci. 13, 127. http://dx.doi.org/
10.1186/1471-2202-13-127.
Acknowledgements Jenkinson, S.P., Grandgirard, D., Heidemann, M., Tscherter, A., Avondet, M.-A., Leib,
S.L., 2017. Embryonic stem cell-derived neurons grown on multi-electrode arrays as a
novel in vitro bioassay for the detection of clostridium botulinum neurotoxins. Front.
This study was funded by NC3Rs grant ID 94812 (reference no Pharmacol. 8, 73. http://dx.doi.org/10.3389/fphar.2017.00073.
G1000086) and National Institute for Biological Standards and Control. Jones, R.G.A., Corbel, M.J., Sesardic, D., 2006. A review of WHO international standards
Dr. RGA Jones provided training to GY during initial assay set up. The for botulinum antitoxins. Biologicals 34, 223–226. http://dx.doi.org/10.1016/j.
biologicals.2005.11.009.
authors would also like to thank Rob Tierney for proof reading and Jones, R.G.A., Ochiai, M., Liu, Y., Ekong, T., Sesardic, D., 2008. Development of improved
Peter Rigsby for statistical assistance. SNAP25 endopeptidase immune-assays for botulinum type A and E toxins. J.
Immunol. Methods 329, 92–101. http://dx.doi.org/10.1016/j.jim.2007.09.014.
Jones, R.J., Liu, Y., Sesardic, D., 2009. Development of a novel highly specific en-
Authors contributions dopeptidase assay for botulinum toxin type C1 activity. J. Immunol. Methods 343,
21–27. http://dx.doi.org/10.1016/j.jim.2009.01.001.
DS and PS conceived and designed the experiments. GY, SR, PS and Jones, R.J., Liu, Y., Hallis, C., Thorpe, S.J., Longstaff, C., Matejtschuk, P., Sesardic, D.,
2011. Release of proteolytic activity following reduction in therapeutic human serum
YL performed the experiments and analysed the data. PS and SR pre-
albumin containing products: detection with a new neoepitope endopeptidase im-
pared figures and critically reviewed the paper. DS wrote the paper. munoassays. J. Pharm. Biomed. Anal. 54, 74–80. http://dx.doi.org/10.1016/j.jpba.
2010.08.013.
Keller, J.E., Neale, E.A., 2001. The role of the synaptic protein snap-25 in the potency of
Conflict of interest
botulinum neurotoxin type A. J. Biol. Chem. 276, 1348–13476. http://dx.doi.org/10.
1074/jbc.M010992200.
The authors declare no conflict of interest. Keller, J.E., Cai, F., Neale, E.A., 2004. Uptake of botulinum neurotoxin into cultured
neurons. Biochemistry 43 (2), 526–532. http://dx.doi.org/10.1021/bi0356698.
Kiris, E., Nuss, J.E., Burnett, J.C., Kota, K.P., Koh, D.C., Wanner, L.M., Torres-Melendez,
References E., Gussio, R., Tessarollo, L., Bavari, S., 2011. Embryonic stem cell-derived moto-
neurons provide a highly sensitive cell culture model for botulinum neurotoxin stu-
Adler, S., Bicker, G., Bigalke, H., Bishop, C., Blumel, J., Dressler, D., Fitzgerald, J., Gessler, dies, with implications for high-throughput drug discovery. Stem Cell Res. 6,
F., Heuschen, H., Kegel, B., Luch, A., Milne, C., Pickett, A., Ratsch, H., Ruhdel, I., 195–205. http://dx.doi.org/10.1016/j.scr.2011.01.002.
Sesardic, D., Stephens, M., Stiens, G., Thornton, P.D., Thurmer, R., Vey, M., Kiris, E., Kota, K.P., Burnett, J.C., Soloveva, V., Kane, C.D., Bavari, S., 2014. Recent de-
Spielmann, H., Grune, B., Liebsch, M., 2010. The current scientific and legal status of velopments in cell-based assays and stem cell technologies for Botulinum neurotoxin
alternative methods to the LD50 test for botulinum neurotoxin potency testing. research and drug discovery. Expert. Rev. Mol. Diagn. 14, 153–168. http://dx.doi.
Report and recommendations of the ZEBET Expert meeting. Altern. Lab. Anim 38 (4), org/10.1586/14737159.2014.867808.
315–330. Liu, Y., Rigsby, P., Sesardic, D., Marks, J.D., Jones, R.G., 2012. A functional dual-coated
Beske, P.H., Scheeler, S.M., Adler, A., McNutt, P.M., 2015. Accelerated intoxication of (FDC) microtitre plate method to replace the botulinum toxin LD50 test. Anal.
GABAergic synapses by botulinum neurotoxin A disinhibits stem cell-derived neuron Biochem. 425, 28–35. http://dx.doi.org/10.1016/j.ab.2012.02.038.
networks prior to network silencing. Front. Cell. Neurosci. 9, 159. http://dx.doi.org/ McNutt, P., Celver, J., Hamilton, T., Mesngon, M., 2011. Embryonic stem cell-derived
10.3389/fncel.2015.00159. neurons are a novel, highly sensitive tissue culture platform for botulinum research.
Beske, P.H., Bradford, A.B., Grynovicki, J.O., Glotfelty, E.J., Hoffman, K.M., Hubbard, Biochem. Biophys. Res. Commun. 405, 85–90. http://dx.doi.org/10.1016/j.bbrc.
K.S., Tuznik, K.M., McNutt, P.M., 2016. Botulinum and tetanus neurotoxin-induced 2010.12.132.
blockade of synaptic transmission in networked cultures of human and rodent neu- National Institute of Environment and Health Services, 2008. Report on the ICCVAM-
rons. Toxicol. Sci 149 (2), 503–515. http://dx.doi.org/10.1093/toxsci/kfv254. NICEATM/ECVAM Scientific Workshop on Alternatives Methods to Refine, Reduce or
Bibel, M., Richter, J., Schrenk, K., Schrenk, K., Kerry, Tucker L., Staiger, V., Korte, L., Replace the Mouse LD50 Assay for Botulinum Toxin Testing. (NIH Publication
Goetz, M., Barde, Y.-A., 2004. Differentiation of mouse embryonic stem cells into a number 08-6416).
defined neuronal lineage. Nat. Neurosci. 7, 1003–1009. http://dx.doi.org/10.1038/ Nuss, J.E., Ruthel, G., Tressler, L.E., Wanner, L.M., Torres-Melendez, E., Hale, M.L.,
nn130. Bavari, S., 2010. Development of cell-based assays to measure botulinum neurotoxin
Bibel, M., Richter, J., Lacroix, E., Barde, Y.-A., 2007. Generation of a defined and uniform serotype A activity using cleavage-sensitive antibodies. J. Biomol. Screen. 15 (1),
population of CNS progenitors and neurons from mouse embryonic stem cells. Nat. 42–51. http://dx.doi.org/10.1177/1087057109354779.
Protoc. 2, 1034–1043. http://dx.doi.org/10.1038/nprot.2007.147. Pellet, S., 2013. Progress in cell based assays for botulinum neurotoxin detection. Curr.
Bigalke, H., Dreyer, F., Bergey, G., 1985. Botulinum A neurotoxin inhibits non-cholinergic Top. Microbiol. Immunol. 364, 257–285. http://dx.doi.org/10.1007/978-3-642-
synaptic transmission in mouse spinal cord neurons in culture. Brain Res. 360 (1–2), 33570-9_12.
318–324. Pellet, S., Tepp, W., Toth, S.I., Johnson, E.A., 2010. Comparison of the primary rat spinal
Dong, M., Tepp, W.H., Johnson, E., Chapman, E.R., 2004. Using fluorescent sensors to cord cell (RSC) assay and the mouse bioassay for Botulinum neurotoxin type A po-
detect botulinum neurotoxin activity in vitro and in living cells. Proc. Natl. Acad. Sci. tency determination. J. Pharmacol. Toxicol. Methods 61 (10), 304–310. http://dx.
U. S. A. 101, 14701–14706. http://dx.doi.org/10.1073/pnas.0404107101. doi.org/10.1016/j.vascn.2010.01.003.
Dong, M., Tepp, W.H., Liu, H., Johnson, E.A., Chapman, E.R., 2007. Mechanism of bo- Pellet, S., Du, Z.W., Pier, C.L., Tepp, W.H., Zhang, S.C., Johnson, E.A., 2011. Sensitive and
tulinum neurotoxin B and G entry into hippocampal neurons. J. Cell Biol. 179, quantitative detection of botulinum neurotoxin in neurons derived from mouse em-
1511–1522. http://dx.doi.org/10.1083/jcb.200707184. bryonic stem cells. Biochem. Biophys. Res. Commun. 404, 388–392. http://dx.doi.
Dressler, D., 2012. Clinical applications of botulinum toxin. Curr. Opin. Microbiol. 15, org/10.1016/j.bbrc.2010.11.128.
325–336. http://dx.doi.org/10.1016/j.mib.2012.05.012. Pellett, S., Tepp, W.H., Clancy, C.M., Borodic, G.E., Johnson, E.A., 2007. A neuronal cell-
Ekong, T.A.N., Feavers, I., Sesardic, D., 1997. Recombinant SNAP25 is an effective sub- based neurotoxin assay for highly sensitive and specific detection of neutralizing
strate for clostridium botulinum type A toxin endopeptidase activity in vitro. serum antibodies. FEBS Lett. 581 (25), 4803–4808. http://dx.doi.org/10.1016/j.
Microbiology 143, 3337–3347. http://dx.doi.org/10.1099/00221287-143-10-3337. febslet.2007.08.078.
Evans, E.R., Skipper, P.J., Shone, C.C., 2009. An assay for botulinum toxin types A, B and Pellett, S., Schwartz, M.P., Tepp, W.H., Josephson, R., Scherf, J.M., Pier, C.L., Thomson,
F that requires both functional binding and catalytic activities within the neurotoxin. J.A., Murphy, W.L., Johnson, E.A., 2015. Human induced pluripotent stem cell de-
J. Appl. Microbiol. 107, 1384–1391. http://dx.doi.org/10.1111/j.1365-2672.2009. rived neuronal cells cultured on chemically-defined hydrogels for sensitive in vitro
04325. detection of botulinum neurotoxin. Sci Rep 5, 14566. http://dx.doi.org/10.1038/
Fernandez-Salas, E., Wang, J., Molina, Y., Nelson, J.B., Jacky, B.P.S., Aoki, R.K., 2012. srep14566.
Botulinum neurotoxin serotype A specific cell based potency assay to replace the Pellett, S., Tepp, W.H., Johnson, E.A., Sesardic, D., 2017. Assessment of ELISA as endpoint
mouse bioassay. PLoS One 7 (11), e49516. http://dx.doi.org/10.1371/journal.pone. in neuronal cell-based assay for BoNT detection using hiPSC derived neurons. J.
0049516. Pharmacol. Toxicol. Methods 88, 1–6. http://dx.doi.org/10.1016/j.vascn.2017.04.

9
G. Yadirgi et al. Journal of Immunological Methods xxx (xxxx) xxx–xxx

013. Sesardic, D., Gaines Das, R., 2008. Alternatives to the LD50 for botulinum toxin potency
Pirazzini, M., Rossetto, O., Eleopra, R., Montecucco, C., 2017. Botulinum neurotoxins: testing: strategies and progress towards refinement, reduction and replacement. In:
biology, pharmacology, and toxicology. Pharmacol. Rev. 69, 200–235. http://dx.doi. Proceedings of the 6th World Congress on Alternatives, Tokyo, 2007, pp. 581–585.
org/10.1124/pr.116.012658. Sesardic, D., Jones, R.G., Leung, T., Alsop, T., Tierney, R., 2004. Detection of antibodies
Purkiss, J.R., Friis, L.M., Doward, S., Quinn, C.P., 2001. Clostridium botulinum neuro- against botulinum toxins. Mov. Disord. 19, 85–91. http://dx.doi.org/10.1002/mds.
toxins act with a wide range of potencies on SH-SY5Y human neuroblastoma cells. 20021.
Neurotoxicology 22 (4), 447–453. Simon, S., Fiebig, U., Liu, Y., Tierney, R., Dano, J., Worbs, S., Endermann, T., Nevers, M.-
Rasetti-Escargueil, C., Machado, C.M., Preneta-Blanc, R., Fleck, R.A., Sesardic, D., 2011. C., Volland, H., Sesardic, D., Dorner, M.B., 2015. Recommended immunological
Enhanced sensitivity to botulinum type A neurotoxin of human neuroblastoma SH- strategies to screen for botulinum neurotoxin-containing samples. Toxins 7 (12),
SY5Y cells after differentiation into mature neuronal cells. Botulinum J. 22 (1), 5011–5034. http://dx.doi.org/10.3390/toxins7124860.
30–48. http://dx.doi.org/10.1504/TBJ.2011.041814. Vaidyanathan, V.V., Yoshino, K., Jahnz, M., Dörries, C., Bade, S., Nauenburg, S.,
Rosen, O., Ozeri, E., Barnea, A., David, A.B., Zichel, R., 2016. Development of an in- Niemann, H., Binz, T., 1999. Proteolysis of SNAP-25 isoforms by botulinum neuro-
novative in vitro potency assay for anti-botulinum antitoxins. Toxins 8 (10), 276. toxin types A, C, and E: domains and amino acid residues controlling the formation of
http://dx.doi.org/10.3390/toxins8100276. enzyme-substrate complexes and cleavage. J. Neurochem. 72, 327–337.
Rossetto, O., Pirazzini, M., Montecucco, C., 2014. Botulinum neurotoxins: genetic, Whitemarsh, R.C.M., Strathman, M.J., Chase, L.G., Stankewicz, C., Tepp, W.H., Johnson,
structural and mechanistic insights. Nat. Rev. Microbiol. 12, 535–549. http://dx.doi. E.A., Pellett, S., 2012. Novel application of human neurons derived from induced
org/10.1038/nrmicro3295. pluripotent stem cells for highly sensitive botulinum neurodetection. Toxicol. Sci.
Schiavo, G., Santucci, A., Dasgupta, B.R., Mehta, P.P., Jontes, J., Benfenati, F., Wilson, 126 (2), 426–435. http://dx.doi.org/10.1093/toxsci/kfr354.
M.C., Montecucco, C., 1993. Botulinum neurotoxins serotypes A and E cleave SNAP- Yowler, B.C., Kensinger, R.D., Schengrund, C.L., 2002. Botulinum neurotoxin A activity is
25 at distinct COOH-terminal peptide bonds. FEBS Lett. 335, 99–103. dependent upon the presence of specific gangliosides in neuroblastoma cells ex-
Sesardic, D., 2012. Bioassays for evaluation of medical products derived from bacterial pressing synaptotagmin I. J. Biol. Chem. 277 (36), 32815–32819. http://dx.doi.org/
toxins. Curr. Opin. Microbiol. 15 (3), 310–317. http://dx.doi.org/10.1016/j.mib. 10.1074/jbc.M205258200.
2012.05.008.

10

View publication stats

You might also like