Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Mol Neurobiol (2013) 48:120–127

DOI 10.1007/s12035-013-8423-9

Botulinum Neurotoxin Type A is Internalized and Translocated


from Small Synaptic Vesicles at the Neuromuscular Junction
Cesare Colasante & Ornella Rossetto & Laura Morbiato &
Marco Pirazzini & Jordi Molgó & Cesare Montecucco

Received: 30 November 2012 / Accepted: 5 February 2013 / Published online: 8 March 2013
# Springer Science+Business Media New York 2013

Abstract Botulinum neurotoxin type A (BoNT/A) is the Introduction


most frequent cause of human botulism and, at the same time,
is largely used in human therapy. Some evidence indicates that The seven known different serotypes of botulinum neuro-
it enters inside nerve terminals via endocytosis of synaptic toxins (BoNT/A to BoNT/G) are produced by bacteria of the
vesicles, though this has not been directly proven. The metal- genus Clostridium and are the sole cause of the flaccid
loprotease L chain of the neurotoxin then reaches the cytosol paralysis of botulism [1]. These neurotoxins are the most
in a process driven by low pH, but the acidic compartment poisonous substances known so far, and their potential use
wherefrom it translocates has not been identified. Using in bioterrorism [2] is a matter of great concern. The BoNTs
immunoelectron microscope, we show that BoNT/A does consist of three domains of 50 kDa each, which are termed L
indeed enter inside synaptic vesicles and that each vesicle chain (the N-terminal one), HN (the intermediate one), and
contains either one or two toxin molecules. This finding HC (the C-terminal domain) [3–5]. These structures are
indicates that it is the BoNT/A protein receptor synaptic closely related to their mechanism of poisoning the nerve
vesicle protein 2, and not its polysialoganglioside receptor terminals, which consists of four steps: (a) binding to nerve
that determines the number of toxin molecules taken up by a terminals mediated by HC, (b) endocytosis, (c) a low pH-
single vesicle. In addition, by rapid quenching the vesicle driven membrane translocation of the L chain mediated by
trans-membrane pH gradient, we show that the neurotoxin HN, and (d) L chain-mediated cleavage and inactivation of
translocation into the cytosol is a fast process. Taken together, the soluble NSF attachment protein receptor (SNARE) pro-
these results strongly indicate that translocation of BoNT/A teins with ensuing inhibition of neuroexocytosis [6–8]. The
takes place from synaptic vesicles, and not from endosomal BoNTs are peculiar with respect to their pre-synaptic mem-
compartments, and that the translocation machinery is operat- brane binding which is mediated by a polysialoganglioside
ed by no more than two neurotoxin molecules. and by a protein receptor consisting of the luminal portion
of a synaptic vesicle (SV) membrane protein [8, 9].
Keywords Botulinum neurotoxin . Electron microscopy . BoNT/A, BoNT/E, and BoNT/F bind to SV2 [10–14], while
Endocytosis . Membrane translocation . Neuromuscular BoNT/B and BoNT/G bind to synaptotagmin [15–18]; the
junction . Synaptic vesicles protein receptor of the other BoNT serotypes has not been
conclusively determined. While the fourth step of intoxica-
tion has been clarified long ago [19, 20], endocytosis and
membrane translocation remain largely unknown. The fact
C. Colasante : J. Molgó (*) that tetanus neurotoxin is taken up by small synaptic vesicle
Laboratoire de Neurobiologie et Développement, Institut de in neurons of the central nervous tissue [21], but not at the
Neurobiologie Alfred Fessard, CNRS-UPR 3294, 91198
neuromuscular junction (NMJ) where it is endocytosed in-
Gif-sur-Yvette Cedex, France
e-mail: Jordi.Molgo@inaf.cnrs-gif.fr side coated vesicles [22], and the fact that the known BoNT
protein receptors are in the lumenal domain of proteins of
O. Rossetto : L. Morbiato : M. Pirazzini : C. Montecucco (*) synaptic vesicles strongly suggest that BoNTs are endocy-
Dipartimento di Scienze Biomediche and Istituto CNR di
tosed inside synaptic vesicles at peripheral nerve terminals
Neuroscienze, Università di Padova, Viale G. Colombo 3,
Padova 35131, Italy [6, 23]. However, their actual presence inside the vesicles
e-mail: cesare.montecucco@gmail.com and the type of synaptic vesicles have not been determined.
Mol Neurobiol (2013) 48:120–127 121

Even less is known about the mode of membrane transloca- then lysed by two passages of pre-cooled French Press. The
tion, but biophysical studies have indicated that, at low pH, lysate was centrifuged (17,000 × g, 20 min), and the
the HN of BoNT forms trans-membrane channel conducing supernatant was loaded on a HiTrap Chelating HP on a
the L chain into the cytosol and involving one to four ÄKTAprime system (GE Healthcare) and eluted with a linear
neurotoxin molecules [8, 24–29]. gradient from 0 to 500 mM imidazole. Pooled fractions were
Here, we have studied the entry of BoNT/A into the dialyzed against 20 mM tris, 200 mM NaCl, 0.1 mM DTT, 5 %
nerve terminal of the mouse NMJ, because this serotype is trehalose, 10 % glycerol, and pH7.4. Protein purity was
the one responsible for the majority of cases of human assessed by staining with SimplyBlue Safestain of a 12 %
botulism [1] and because BoNT/A is almost invariably used sodium dodecyl sulfate polyacrylamide gel. Protein identity
in the therapy of human diseases caused by hyperfunction of was confirmed by Western blotting using an anti-Tag antibody.
peripheral cholinergic nerve terminals [30–32]. We have
generated a chimera consisting of a green fluorescent pro- In Vivo Mouse Intra-muscular Injection of Fusion Proteins
tein linked to the HC domain of BoNT/A (EGFP–BoNT/A-
HC) in order to visualize its distribution at the NMJ using All experiments were performed following French and
fluorescence microscopy and immunoelectron microscopy Italian guidelines for laboratory animal handling and were
with gold-labeled anti-GFP antibodies. This protocol avoids approved by the Animal Committee of the CNRS and of the
artifacts of binding deriving from the cross-linking of a University of Padova, and in accordance with the European
number of neurotoxin molecules to the same gold particle. Community Council Directive of November 24, 1986
We found that the injection of EGFP–BoNT/A-HC into a (86/609/EEC). Young Swiss female mice were obtained from
mouse skeletal muscle leads to its specific binding to the Charles River Breeding Laboratories. EGFP–BoNT/A-HC
pre-synaptic membrane at the active zones of the NMJ, and (25 μg) dissolved in phosphate-buffered saline (PBS) was
that one/two gold particles are present inside small clear injected into the sternocleidomastoid muscle of mice
synaptic vesicles. We also showed that the translocation of anesthetized with sodium pentobarbital (90 mg/Kg, i.p.).
the L chain into the cytosol is a very rapid process, and we The animals were killed through the intra-cardiac perfusion
discuss the general implications of these findings. of freshly prepared 4 % paraformaldeyde (Electron
Microscopy Sciences, Hatfield, PA, USA) 5–10 min after
the injection of EGFP–BoNT/A-HC.
Material and Methods
Mouse Sternocleidomastoid Muscles
Preparation of the BoNT/A-HC Chimera
The muscles were dissected; their fibers were teased apart
Polymerase chain reaction was performed using as a tem- and stained for 30 min at 4 °C with AlexaFluor-594- or
plate the DNA of BoNT/A-HC cloned in pGEX4T1-GST- AlexaFluor680-conjugated α-bungarotoxin (2 μg/ml,
HA (kind gift of Dr. G. Schiavo, Cancer Research, UK) and A594-α-BTx, or A680-α-BTx; Molecular Probes Europe
the DNA of EGFP cloned in the N1 plasmid. The following BV, Leiden, The Netherlands) in PBS and mounted with
primers were used: for the HC of BoNT/A, residues 845– Vectashield anti-fade mounting medium (Vector Laborato-
1295, AAACTCGAGAGTACAGATATACCTTTTCA ries, Inc, Burlingame, CA, USA). Neuromuscular junctions
G C T T a n d A A AT T TA A G C T T T T A C A G T G were analyzed using a Leica upright SP2 DM RXA2 con-
GCCTTTCTCCCCA; for EGFP, AAAGGATCCATGG focal microscope through an oil-immersion objective ×
TGAGCAAGGGCGAG and AAACTCGAGCTTGTA 63/1.32 NA, controlled by Leica Confocal Software version
CAGCTCGTCCATGCC. The amplified sequence of HC 2.61 Build 1537 (argon laser 488 nm for EGFP, HeNe laser
was digested with XhoI/HindIII and then cloned in the 543 nm for A594, and HeNe laser 633 nm for A680).
pRSETa vector to obtain pRSETa–BoNT/A-HC; then, the Intensity profile analyses were performed on series of
EGFP sequence-digested BamHI/XhoI was ligated into the “look-through” projections of mean intensity.
previous vector at the N-terminal domain in order to obtain
finally pRSETa–EGFP–BoNT/A-HC. DNA inserts were se- Immunogold Electron Microscopy
quenced by CRIBI (Padova).
BL21 (DE3) pLysS Escherichia coli strains transformed After 4 % paraformaldehyde perfusion, mouse sternocleido-
with pRSETa–EGFP–BoNT/A-H C were grown in LB mastoid muscles injected with EGFP–BoNT/A-HC were
medium until the optical density at 600 nm reached 0.6. dissected out and post-fixed for 1 h with a mixture of 4 %
Cultures were induced with 1 mM isopropyl β- D -1- paraformaldehyde, 0.1 % glutaraldehyde (Electron Micros-
thiogalactopyranoside for 4 h at 30 °C. Cells were pelleted, copy Sciences, Hatfield, PA, USA), 0.2 % picric acid (Sig-
frozen in liquid nitrogen, and kept at −80 °C. Cells were ma–Aldrich), and 3 % sucrose in PBS solution. Muscle
122 Mol Neurobiol (2013) 48:120–127

regions containing NMJs were treated with 0.5 % osmium Neutralization of the Acidic Compartment on BoNT/A
tetroxide (Electron Microscopy Sciences) for 30 min in Entry into Cultured Neurons
PBS, followed by dehydration in a graded ethanol series
and embedded in LR white resin (Electron Microscopy MNs or CGNs were treated with BoNT/A (10 nM) in high-
Sciences). Silver-gray thin sections (65 nm) were treated potassium buffer (85 mM NaCl, 60 mM KCl, 2 mM CaCl2,
on grids with 0.1 % sodium borohydride in PBS for 2 mM MgCl2, 5.5 mM glucose, 10 mM 4-(2-hydroxyethyl)-
15 min. After washing with PBS containing 0.1 % acetylat- 1-piperazineethanesulfonic acid (HEPES), and pH7.4) for
ed bovine serum albumin (BSA) solution (solution A), non- 5 min. Unbound neurotoxin was washed out using low-K+
specific labeling was prevented with a 30-min incubation buffer (140 mM NaCl, 5 mM KCl, 2 mM CaCl2, 2 mM
with 5 % BSA in 0.1 % gelatin (Aurion, Wageningen, The MgCl2, 5.5 mM glucose, 10 mM HEPES, and pH7.4), and
Netherlands) and 5 % normal goat serum (Sigma). Sections normal culture medium was restored. Before, together or at
were incubated for 2 h with polyclonal rabbit anti-GFP the indicated time points after neurotoxin addition, bafilo-
antibody (diluted 1:100 in solution A, MBL, Watertown, mycin A1 (baf A1, 100 nM, Sigma–Aldrich) alone, or in
USA) followed by 1-h incubation with goat anti-rabbit IgG combination with monensin (Sigma–Aldrich, 100 nM) or
conjugated with 10-nm diameter colloidal gold (Aurion, NH4Cl (50 mM), was added to the culture media. After
ImmunoGold Reagents, The Netherlands) at 1:25 dilution 15 min, the medium was removed and replaced with one
in solution A. After washing, the sections were fixed with containing only baf A1, and the neurons were further incu-
2.5 % glutaraldehyde in PBS. Finally, the grids were coun- bated for 12 h; the cells were lysed directly in Laemmli
terstained with 2.5 % uranyl acetate and 1 % lead citrate buffer. BoNT/A entry was evaluated by determining the
solutions, and examined using a Jeol 1400 transmission extent of synaptosomal-associated protein 25 (SNAP-25)
electron microscope. cleavage by immunoblot analysis using a polyclonal anti-
body which specifically recognizes the C-terminal part of
Cell Cultures SNAP-25 [35]. A monoclonal antibody against vesicle-
associated membrane protein 2 (VAMP-2; Synaptic Sys-
Rat embryonic spinal cord motor neurons (MNs) were tems, Göttingen Germany) was used as an internal loading
prepared essentially as described [33]. Briefly, spinal control. Cleavage was reported as the percentage ratio of the
cords were collected from fetal rats at gestation day 14, whole SNAP-25 with respect to VAMP-2, considering data
which were first mechanically disrupted; then, the cells collected from three independent experiments made in trip-
were dissociated by mild trypsinization in the presence of licate. Values are expressed as the mean ± SD.
DNAse I. Finally, isolated cells were plated on 24-well
plates, pre-treated with poly-ornithine (1.5 μg/ml over-
night) and then with laminin (10 μg/ml for 2 h). Cultures Results
were maintained at 37 °C and 5 % of CO2 before
starting the assay at DIV 15. The culture medium con- A characteristic of BoNT/A, which is essential for its clin-
sisted of Neurobasal (Gibco) supplemented with 2 % ical use, is its rapid and specific binding to the pre-synaptic
heat-inactivated horse serum, 0.5 mM L -glutamine, membrane of the vertebrate NMJ, with limited diffusion
25 mM 2-mercaptoethanol, 25 mM L-glutamic acid, B27 near the site of injection [36]. Figure 1a shows that this
supplement, 10 ng/ml ciliary neurotrophic factor, property is conserved by the EGFP–BoNT/A-HC chimera,
100 pg/ml glial cell-derived neurotrophic factor, and since it labels only the motor nerve terminals within the
5 mg/ml penicillin/streptomycin. boundaries defined by the post-synaptic nicotinic acetylcho-
Rat cerebellar granular neurons (CGNs) were prepared line receptor visualized here by the very specific binding of
as previously described [34]. Rat pups were killed 6 days fluorescent α-BTx. The neuromuscular branches' side view
after birth; cerebella were isolated and cells were dissoci- presented in row B of Fig. 1 clearly shows that the chimera
ated by trypsinization in the presence of DNAse I. Col- binding is restricted to the pre-synaptic membrane. Howev-
lected cells were plated in 24-well plates, pre-treated with er, the level of resolution of this analysis does not allow one
poly-L-lysine (50 μg/mL overnight), at cell density of 3× to distinguish toxin bound to the nerve terminal surface
105 cells per well. Cultures were maintained at 37 °C, from internalized neurotoxin.
5 % CO2, in BME supplemented with 10 % fetal bovine To visualize the distribution of the binding domain of
serum, 25 mM KCl, 2 mM glutamine, and 50 μg/mL BoNT/A at higher resolution, we performed immunoelec-
gentamycin. To arrest growth of non-neuronal cells, cyto- tron microscopy using a GFP-specific antibody as a read
sine arabinoside (10 μM) was added to the medium 18– out, which was then revealed with a gold-labeled secondary
24 h after plating. Experiments were performed on neu- antibody. This procedure is somewhat more laborious than
rons that were 6 DIV. the direct coupling of BoNT/A-HC to gold particles, but was
Mol Neurobiol (2013) 48:120–127 123

Fig. 1 The botulinum neurotoxin type A HC-binding domain (BoNT/ distributed along the motor nerve terminal and restricted to the NMJ
A-HC) binds to axon terminals of the mouse sternocleidomastoid NMJ defined by the α-BTx staining (Merge); scale bar, 20 μm. b Side view
shown by confocal fluorescent microscopy. a Top view of two NMJs of a NMJ branches clearly shows that EGFP–BoNT/A-HC label is
revealed by the staining of the post-synaptic acetylcholine receptors mainly found at the synaptic side of the nerve terminal (around the
with Alexa 594-labeled α-bungarotoxin (α-BTx), showing that the pre-synaptic membrane) and does not co-localize with the facing Alexa
EGFP fluorescence of BoNT/A-HC toxin (EGFP–BoNT/A-HC) is 680-labeled α-BTx (Merge); scale bar, 10 μm

chosen to avoid possible artifacts owing to possible multi- In addition, the simultaneous binding of one gold-labeled
receptor binding of the several BoNT/A-HC molecules secondary antibody to more than one anti-GFP antibodies
cross-linked to the same gold particle. On the other hand, cannot be excluded. Remarkably, the average figure of 1.5
the present procedure can introduce some unspecific gold correlates well with the number of BoNT/A protein–recep-
deposition due to increased possibility of antibody cross- tor–SV2 molecules present in a single SV, estimated to be
reaction. Figure 2 shows that the majority of the neurotoxin between 1 and 2 [38].
molecules are inside small clear SV in muscle samples fixed To provide a functional estimation of the time course of
5 or 10 min after injection (arrowheads), while few are still the translocation process of the BoNT/A-L chain, we used
bound to the pre-synaptic membrane, mainly at active zones different agents able to quench the trans-membrane pH
(long arrows); only occasionally gold particles are found in gradient existing across the SV membrane at defined time
the cytosol, possibly owing to antibody cross-reaction. This points: bafA1 which inhibits the SV ATPase proton pump,
clearly indicates that binding and endocytosis in vivo is monensin which is a very rapid exchanger of H+ for mono-
rapid and that the toxin ends up mainly inside the SV. The valent cations, and ammonium which buffers the lumen pH.
present study was not extended to the motor axon because The effect of their addition at different time points before
the study of the retro-axonal transport of the BoNT/A [37] and after a bacterial toxin, the entry of which into the
was not among the aims of the present study. cytosol occurs via an acidic compartment, provides a reli-
Many motor nerve terminals were analyzed, and Table 1 able estimation of the time course of the entry of the toxin
reports the statistics of the gold particle distribution. This active chain entry into the cytosol [39, 40]. When a rapid
distribution is rather similar in samples analyzed 5 or 10 min quenching is needed, these agents can be combined [39].
after neurotoxin injection, as expected from the fact that Two different primary neuronal cultures were used to obtain
only the HC pre-synaptic membrane binding domain of information of more general relevance: cerebellar granular
BoNT/A is present here; therefore, translocation in the cy- neurons and spinal cord motor neurons. Figure 3 shows that
tosol cannot take place. The average number of gold par- whatever pH gradient quenching method was used (bafA1,
ticles, and therefore of BoNT/A-HC molecules, present bafA1 plus monensin, or bafA1 plus ammonium), the neu-
inside the SV is around 1.5 molecules per vesicle (see tralization of the lumenal pH of SV prevented the low pH-
Table 1). This method may overestimate the number of toxin driven membrane translocation of the L chain of BoNT/A
molecules present in the lumen of a single SV, owing to the only for a few minutes after BoNT/A addition, indicating
use of a secondary antibody, to possible antibody cross- that the entry of its L chain in the cytosol takes place early
reactivity, and to accidental deposition of a gold particle. after endocytosis. These data compare well with previous
124 Mol Neurobiol (2013) 48:120–127

Fig. 2 Immunogold electron


micrographs showing the ultra-
structural localization of the
binding domain of BoNT/A in
the mouse sternocleidomastoid
NMJ. A fluorescent derivative
of the binding domain of
BoNT/A (EGFP–BoNT/A-HC)
was intradermally injected into
the sternocleidomastoid muscle
and immunocytochemically
processed with an anti-EGFP
following a post-embedding
procedure using a secondary
10-nm gold-labeled antibody.
The electron micrographs show
that the toxin is rapidly
internalized and is localized
mainly within the lumen of the
small synaptic vesicles
(arrowheads), with some
molecules still bound to the pre-
synaptic membrane mainly at
the active zones (long arrows)
either after 5 (a and b) or
10 min (c) from toxin injection.
Scale bars, 150 nm

findings obtained with concanamycin A which is another Discussion


inhibitor of the V-ATPase [41, 42] and indicate that the
quenching of the trans-membrane pH gradient of SV is The most important information delivered by the present work
sufficiently rapid also with this slower inhibitor [39]. are the following: (1) BoNT/A is internalized into the lumen
of small synaptic vesicles, (2) it then translocates its L chain
Table 1 Estimates of the number of SVs labeled with EGFP–BoNT/ into the cytosol very rapidly, and (3) the L chain translocating
A-Hc, revealed with a 10 nm colloidal gold labeled anti-EGFP specific
channel is made of one, or at the most, two HN domains. These
antibody, in motor nerve terminals 5 and 10 min after a subcutaneous
toxin injection in the mouse sternocleidomastoid muscle conclusions are derived from a quantitative visualization of
the toxin molecule within the lumen of small synaptic vesicles
Category Time and from a time-resolved quenching of the effector of the
Time after BoNT/A-Hc–EGFP 5 mina 10 minb translocation, which is the lumenal low pH. This latter exper-
injection iment was performed with two different types of neurons and
Average number of SVs counted 57.64±0.76c 54.03±0.59 three different trans-membrane pH gradient quenching meth-
per NMJ-section ods in order to obtain results of more general value. The
Percentage of SVs labeled 37.52±0.12 40.34±0.15
present data fit very well with the estimation that a population
Number of grains/SVs counted 1.46±0.18 1.51±0.25
of small synaptic vesicles contain an average of one to two
SVs synaptic vesicles SV2 molecules per small synaptic vesicle [38]. At the same
a
Data obtained from 26 different motor nerve terminals examined time, this finding indicates that it is the SV2 protein receptor
b
Data obtained from 24 different motor nerve terminals [10, 11] and not the polysialoganglioside receptor [43] that
c
Data represent the mean ± SEM determines the number of toxin molecules loaded within one
Mol Neurobiol (2013) 48:120–127 125

Fig. 3 Time course of the entry of the L chain of BoNT/A in a content of SNARE proteins was estimated by immunoblotting with
cerebellar granular neurons and in b spinal cord motor neurons. Cells specific antibodies. Values are reported as the ratio between the stain-
were incubated with BoNT/A (10 nM) at 37 °C, and at the times ing with the antibody specific for SNAP-25 and the staining with the
indicated, bafilomycin A1 (100 nM) alone (black bars), in combination antibody specific for VAMP-2 expressed as the control value taken as
with monensin (100 nM, striped bars) or with NH4Cl (50 mM, empty 100 %. SD values refer to three different experiments performed in
bars) was added. The incubation was prolonged for 12 h, and the triplicate

SV. This leaves the question of how BoNT/A is bound inside The conclusion that the protein-conducing channel made
the SV: only to SV2 or to both SV2 and polysialogangliosides by the HN domain in the SV membrane involves no more
open. than two molecules has to be confronted with previous data
These considerations are not limited to BoNT/A, but obtained with model systems that the low pH-driven
can be extended with large confidence to BoNT/E which membrane-inserted BoNT/A is a tetramer [24, 25]. Howev-
differs structurally from BoNT/A [3, 5], but enters the er, these data were obtained using very different conditions
cytosol of synaptic terminals within few minutes [41, 42] that may explain the different figures obtained. In one case,
as BoNT/A does. The similar time course and the fact segment 659–681 of BoNT/A was used in a planar lipid
that BoNT/E shares with BoNT/A the SV2 protein re- bilayer [24], while the other one used an artificial liposomal
ceptor [13] speaks strongly in favor of the possibility that system [25]. More recently, using atomic force microscopy
also the L chain of BoNT/E enters the cytosol shortly of BoNT/B interacting with polysialoganglioside-containing
after neurotoxin endocytosis in the SV lumen by crossing lipid bilayers at pH4.4, a significant amount of trimers of
the SV membrane. Together with the rapid kinetics of the toxin was detected, and it was suggested that the L
entry in the cytosol of BoNT/A and BoNT/E, this indi- chain-conducing channel of this neurotoxin is composed
cates that their L chain translocations take place mainly of three HN domains [29].
at the level of SVs. The present study was limited to the In conclusion, the present work visualizes for the first
NMJ and did not include the study of the retro-axonal time BoNT/A inside small clear synaptic vesicles present
transport of BoNT/A, which has been recently well docu- within the main target of this neurotoxin, i.e., the motor
mented [37, 44, 45]. In this respect, the use of the entire nerve terminal of the NMJ. The present data were obtained
BoNT/A should be included in comparison to that of the with a method that avoids the possibility of receptor cross-
binding domain, because one cannot exclude the possi- linking and provide a reliable estimate of the number of
bility that the other domains of the toxin may be in- neurotoxin molecules present within a vesicle. The number
volved in the entry of BoNT/A inside vesicles of retro- of molecules may be one or two, suggesting that membrane
axonal transport, though recent work has shown that the translocation occurs at the level of the membrane of small
binding domain of BoNT/A and BoNT/E is transported clear synaptic vesicles via a protomer consisting of no more
retro-axonally [45]. For what concerns the present paper than two neurotoxin molecules.
aimed at studying the initial trafficking events of
BoNT/A after binding to the NMJ in vivo, all the avail- Acknowledgments The work carried out in the authors' laboratories
able evidence indicate that the use of the binding domain is supported by grants from the Ministero dell'Università e della
Ricerca (Progetto PRIN) and from the University of Padova to OR,
alone is fully appropriate [7, 45]. If anything, the use of
from the Progetto Strategico "An in Vivo Approach to the Physiopa-
a chimera of smaller dimensions with respect to BoNT/A thology of Signal Transduction" of the University of Padova and from
should increase the number of gold particles found inside Fondazione CARIPARO "Synaptic Functions and Role of Glial Cells
the SV with respect to the entire molecule; this is an in Brain and Muscle Diseases" to CM, and by a EU VIIth Frame
Program grant 241835 to JM.
additional indication that the figures provided here may
be overestimated, bringing the real figure closer to one Conflict of interest The authors declare that they have no conflict of
than to two toxin molecules per SV. interest.
126 Mol Neurobiol (2013) 48:120–127

References 21. Matteoli M, Verderio C, Rossetto O, Iezzi N, Coco S, Schiavo G,


Montecucco C (1996) Synaptic vesicle endocytosis mediates the
entry of tetanus neurotoxin into hippocampal neurons. Proc Natl
1. Johnson EA, Montecucco C (2008) Botulism. Handb Clin Neurol Acad Sci USA 93:13310–13315
91:333–368 22. Roux S, Colasante C, Saint Cloment C, Barbier J, Curie T,
2. Arnon SS et al (2001) Botulinum toxin as a biological weap- Girard E, Molgó J, Brulet P (2005) Internalization of a GFP-
on: medical and public health management. JAMA 285:1059– tetanus toxin C-terminal fragment fusion protein at mature
1070 mouse neuromuscular junctions. Mol Cell Neurosci 30:79–89
3. Lacy DB, Stevens RC (1999) Sequence homology and structural 23. Verderio C, Grumelli C, Raiteri L, Coco S, Paluzzi S, Caccin
analysis of the clostridial neurotoxins. J Mol Biol 291:1091–1104 P, Rossetto O, Bonanno G, Montecucco C, Matteoli M (2007)
4. Swaminathan S, Eswaramoorthy S (2000) Structural analysis of Traffic of botulinum toxins A and E in excitatory and inhib-
the catalytic and binding sites of Clostridium botulinum neurotoxin itory neurons. Traffic 8:142–153
B. Nat Struct Biol 7:693–699 24. Montal MS, Blewitt R, Tomich JM, Montal M (1992) Identification
5. Kumaran D, Eswaramoorthy S, Furey W, Navaza J, Sax M, of an ion channel-forming motif in the primary structure of tetanus
Swaminathan S (2009) Domain organization in Clostridium botu- and botulinum neurotoxins. FEBS Lett 313:12–18
linum neurotoxin type E is unique: its implication in faster trans- 25. Schmid MF, Robinson JP, DasGupta BR (1993) Direct visualiza-
location. J Mol Biol 386:233–245 tion of botulinum neurotoxin-induced channels in phospholipid
6. Schiavo G, Matteoli M, Montecucco C (2000) Neurotoxins affect- vesicles. Nature 364:827–830
ing neuroexocytosis. Physiol Rev 80:717–766 26. Montecucco C, Schiavo G (1995) Structure and function of tetanus
7. Binz T, Rummel A (2009) Cell entry strategy of clostridial neuro- and botulinum neurotoxins. Q Rev Biophys 28:423–472
toxins. J Neurochem 109:1584–1595 27. Koriazova LK, Montal M (2003) Translocation of botulinum neu-
8. Montal M (2010) Botulinum neurotoxin: a marvel of protein rotoxin light chain protease through the heavy chain channel. Nat
design. Annu Rev Biochem 79:591–617 Struct Biol 10:13–18
9. Montecucco C, Rossetto O, Schiavo G (2004) Presynaptic receptor 28. Montal M (2009) Translocation of botulinum neurotoxin light
arrays for clostridial neurotoxins. Trends Microbiol 12:442–446 chain protease by the heavy chain protein-conducting channel.
10. Dong M, Yeh F, Tepp WH, Dean C, Johnson EA, Janz R, Toxicon 54:565–569
Chapman ER (2006) SV2 is the protein receptor for botulinum 29. Sun S, Suresh S, Liu H, Tepp WH, Johnson EA, Edwardson JM,
neurotoxin A. Science 312:592–596 Chapman ER (2011) Receptor binding enables botulinum neuro-
11. Mahrhold S, Rummel A, Bigalke H, Davletov B, Binz T toxin B to sense low pH for translocation channel assembly. Cell
(2006) The synaptic vesicle protein 2C mediates the uptake Host Microbe 10:237–247
of botulinum neurotoxin A into phrenic nerves. FEBS Lett 30. Davletov B, Bajohrs M, Binz T (2005) Beyond BOTOX: advan-
580:2011–2014 tages and limitations of individual botulinum neurotoxins. Trends
12. Rummel A et al (2009) Botulinum neurotoxins C, E and F Neurosci 28:446–452
bind gangliosides via a conserved binding site prior to 31. Montecucco C, Molgó J (2005) Botulinal neurotoxins: revival of
stimulation-dependent uptake with botulinum neurotoxin F uti- an old killer. Curr Opin Pharmacol 5:274–279
lising the three isoforms of SV2 as second receptor. J 32. Dressler D (2012) Clinical applications of botulinum toxin. Curr
Neurochem 110:1942–1954 Opin Microbiol 15:325–336
13. Dong M, Liu H, Tepp WH, Johnson EA, Janz R, Chapman ER 33. Ransom BR, Christian CN, Bullock PN, Nelson PG (1977) Mouse
(2008) Glycosylated SV2A and SV2B mediate the entry of spinal cord in cell culture. II. Synaptic activity and circuit behavior.
botulinum neurotoxin E into neurons. Mol Biol Cell 19:5226– J Neurophysiol 40:1151–1162
5237 34. Rigoni M et al (2004) Snake presynaptic neurotoxins with
14. Fu Z, Chen C, Barbieri JT, Kim JJ, Baldwin MR (2009) phospholipase A2 activity induce punctate swellings of neu-
Glycosylated SV2 and gangliosides as dual receptors for botuli- rites and exocytosis of synaptic vesicles. J Cell Sci 117:3561–
num neurotoxin serotype F. Biochemistry 48:5631–5641 3570
15. Nishiki T, Kamata Y, Nemoto Y, Omori A, Ito T, Takahashi M, 35. Pirazzini M, Rossetto O, Bolognese P et al (2011) Double
Kozaki S (1994) Identification of protein receptor for Clostridium anchorage to the membrane and intact inter-chain disulfide bond
botulinum type B neurotoxin in rat brain synaptosomes. J Biol are required for the low pH induced entry of tetanus and
Chem 269:10498–10503 botulinum neurotoxins into neurons. Cell Microbiol 13:1731–
16. Nishiki T et al (1996) The high-affinity binding of 1743
Clostridium botulinum type B neurotoxin to synaptotagmin 36. Carli L, Montecucco C, Rossetto O (2009) Assay of diffusion of
II associated with gangliosides GT1b/GD1a. FEBS Lett different botulinum neurotoxin type a formulations injected in the
378:253–257 mouse leg. Muscle Nerve 40:374–380
17. Dong M, Tepp WH, Liu H, Johnson EA, Chapman ER (2007) 37. Caleo M, Schiavo G (2009) Central effects of tetanus and botuli-
Mechanism of botulinum neurotoxin B and G entry into hippo- num neurotoxins. Toxicon 54:593–599
campal neurons. J Cell Biol 179:1511–1522 38. Takamori S et al (2006) Molecular anatomy of a trafficking organ-
18. Rummel A et al (2007) Identification of the protein receptor elle. Cell 127:831–846
binding site of botulinum neurotoxins B and G proves the 39. Papini E, Rappuoli R, Murgia M, Montecucco C (1993) Cell
double-receptor concept. Proc Natl Acad Sci USA 104:359– penetration of diphtheria toxin. Reduction of the interchain disul-
364 fide bridge is the rate-limiting step of translocation in the cytosol. J
19. Schiavo G, Rossetto O, Benfenati F, Poulain B, Montecucco C Biol Chem 268:567–574
(1994) Tetanus and botulinum neurotoxins are zinc proteases spe- 40. Menard A, Altendorf K, Breves D, Mock M, Montecucco C
cific for components of the neuroexocytosis apparatus. Ann N Y (1996) The vacuolar ATPase proton pump is required for the
Acad Sci 710:65–75 cytotoxicity of Bacillus anthracis lethal toxin. FEBS Lett
20. Niemann H, Blasi J, Jahn R (1994) Clostridial neurotoxins: 386:161–164
new tools for dissecting exocytosis. Trends Cell Biol 4:179– 41. Keller JE, Cai F, Neale EA (2004) Uptake of botulinum neurotoxin
185 into cultured neurons. Biochemistry 43:526–532
Mol Neurobiol (2013) 48:120–127 127

42. Sun S, Tepp WH, Johnson EA, Chapman ER (2012) Botulinum 44. Antonucci F, Rossi C, Gianfranceschi L, Rossetto O, Caleo M
neurotoxins B and E translocate at different rates and exhibit diver- (2008) Long distance retrograde effects of botulinum neurotoxin
gent responses to GT1b and low pH. Biochemistry 51:5655–5662 A. J Neurosci 28:3689–3696
43. Rummel A, Mahrhold S, Bigalke H, Binz T (2004) The HCC- 45. Restani L, Giribaldi F, Bercsenyi K, Manich M, Menedez G,
domain of botulinum neurotoxins A and B exhibits a singular Rossetto O, Caleo M, Schiavo G (2012) Botulinum neurotoxins
ganglioside binding site displaying serotype specific carbohydrate A and E undergo retrograde axonal transport in primary motor
interaction. Mol Microbiol 51:631–643 neurons. PLoS Pathog 8:e1003087

You might also like