Next Article in Journal
Special Issue “H. pylori Virulence Factors in the Induction of Gastric Cancer”
Next Article in Special Issue
The Expanding Therapeutic Utility of Botulinum Neurotoxins
Previous Article in Journal / Special Issue
Botulinum Neurotoxin Injection for the Treatment of Recurrent Temporomandibular Joint Dislocation with and without Neurogenic Muscular Hyperactivity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploiting Botulinum Neurotoxins for the Study of Brain Physiology and Pathology

CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy
*
Author to whom correspondence should be addressed.
Toxins 2018, 10(5), 175; https://doi.org/10.3390/toxins10050175
Submission received: 31 March 2018 / Revised: 21 April 2018 / Accepted: 23 April 2018 / Published: 25 April 2018

Abstract

:
Botulinum neurotoxins are metalloproteases that specifically cleave N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins in synaptic terminals, resulting in a potent inhibition of vesicle fusion and transmitter release. The family comprises different serotypes (BoNT/A to BoNT/G). The natural target of these toxins is represented by the neuromuscular junction, where BoNTs block acetylcholine release. In this review, we describe the actions of botulinum toxins after direct delivery to the central nervous system (CNS), where BoNTs block exocytosis of several transmitters, with near-complete silencing of neural networks. The use of clostridial neurotoxins in the CNS has allowed us to investigate specifically the role of synaptic activity in different physiological and pathological processes. The silencing properties of BoNTs can be exploited for therapeutic purposes, for example to counteract pathological hyperactivity and seizures in epileptogenic brain foci, or to investigate the role of activity in degenerative diseases like prion disease. Altogether, clostridial neurotoxins and their derivatives hold promise as powerful tools for both the basic understanding of brain function and the dissection and treatment of activity-dependent pathogenic pathways.
Key Contribution: This review describes the experimental use of botulinum neurotoxins as tools to block synaptic function in specific brain modules and dissect activity-dependent pathways in CNS pathologies.

1. Introduction

Botulinum neurotoxins (BoNTs) are the pathogenic agents responsible for the manifestation of botulism. The typical flaccid paralysis of botulism induced by BoNTs is due to blockade of cholinergic neurotransmission at the neuromuscular junction and autonomic terminals [1,2,3].
These toxins are produce by anaerobic bacteria of the genus Clostridium and are among the most potent naturally-occurring substances. The family of BoNTs comprises seven antigenically distinct botulinum neurotoxins (BoNT/A–BoNT/G). For serotypes A, B, E, and F, several subtypes have been described based on differences in amino-acid sequences. For BoNT/A, at least eight subtypes (named A1 to A8) are currently known with different enzymatic activity and toxicological properties [4,5,6].
BoNTs share a common molecular structure and are composed of a disulphide-linked, ~100-kDa heavy chain and ~50-kDa light chain. They are metalloproteases that bind to presynaptic terminals, enter the cytosol and block neurotransmitter release by specific cleavage of proteins of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex. The SNARE complex is necessary for synaptic vesicles fusion, thus the net effect is blockade of neurotransmitter release [3,7,8]. The target protein differs according to BoNTs serotype. BoNT/A and E cleave synaptosomal associated protein of 25 kDa (SNAP-25); BoNT/C acts on both SNAP-25 and syntaxin; BoNT/B, D, F and G cleave vesicle-associated membrane proteins (VAMPs, also known as synaptobrevins).
Despite their toxicity, they produce a prolonged but reversible action at the synapses. Thus, it has been speculated, already decades ago, that small amount of BoNTs could be used therapeutically to treat disorders characterized by hyperexcitability. Historically, the first to make therapeutic use of BoNT/s was Alan B. Scott in the 1970s, for the treatment of strabismus [9]. Subsequently, the Food and Drug Administration has continuously increased the approved uses for botulinum neurotoxin A1 (BoNT/A1). BoNT/A1 is indeed the most used serotype in clinical practice, because the protease has a persistent activity and this allows long lasting duration of the therapeutic effects (months).
To date, approved indications include focal dystonias, spasticity, cosmetic treatments and migraine, and several other applications are emerging. In all of these cases, minute amounts of BoNT are administered in peripheral muscles to locally inhibit transmitter release.
However, BoNTs are also effective in blocking transmitter release at central synapses when directly delivered into the brain [10].
Here we will review literature data reporting BoNTs effects following direct injection into the central nervous system. Specifically, we will describe how these potent and selective synaptic blockers may be exploited to gain insight into mechanisms of brain physiology and dysfunction.

2. Action of BoNTs on Central Synaptic Terminals

BoNTs enter central neurons mainly via activity-dependent synaptic endocytosis, indeed depolarization increases toxins uptake [11,12,13,14]. At least for BoNT/A, neuronal entry also occurs via an alternative pathway independent of synaptic vesicle endocytosis [15,16], which may direct the toxin to the retroaxonal transport pathway [17,18].
Analyses on brain synaptosomes have demonstrated that BoNTs (mainly studies on BoNT/A) interfere with neurotransmitter release of acetylcholine, glutamate, noradrenaline, serotonin and dopamine from central synases ([10]). It is interesting to note that GABAergic terminals are more resistant to BoNT/A intoxication compared to excitatory (glutamatergic) terminals [19,20]. One reason could be that SNAP-25, the synaptic target of BoNT/A, is less expressed in inhibitory than in glutamatergic terminals [20,21]. For example, SNAP-25 is almost absent in perisomatic inhibitory terminals impinging onto principal neurons in the pyramidal layer of hippocampal CA1 [22]. However, recent electrophysiological recordings in embryonic stem cell-derived neurons (ESNs), showed that miniature Inhibitory Post Synaptic Currents (mIPSC) frequencies were already reduced more than 70% 30 min after BoNT/A intoxication, while decrease in miniature Excitatory Post Synaptic Currents (mEPSC) frequencies was detectable only after 70 min [23]. This finding supports the initial increase in frequency of mPSCs in the first hour after BoNT/A treatment, followed by basically a complete silencing of activity around 15 h [23].
Silencing of spontaneous and evoked excitatory postsynaptic potentials was already demonstrated in hippocampal neurons [24,25]. Accordingly, in vivo delivery of BoNT/A or BoNT/E in rodent hippocampus prevents neuronal spiking activity in hippocampal CA1 [26,27].
It is worth noting that BoNT/A produces an efficient blockade of neurotransmitter release by cleaving a small percentage (about 10%) of the SNAP-25. This seems to be due to the dominant negative effect of BoNT/A-truncated SNAP-25 [28]. However, it possible to rescue BoNT/A-induced blockade of neurotransmission by increasing extracellular calcium concentration. Although BoNT/A and BoNT/E share the same synaptic target (SNAP-25), this rescue with calcium is not possible with BoNT/E, probably because serotype E cleaves a larger fragment at the C-terminus of SNAP-25 [24,29].
At the ultrastructural level, our group investigated the morphological changes induced by local delivery of BoNT/A into the hippocampus [30,31]. Hippocampal samples were analyzed at different times following BoNT/A injection (2, 4, 8 weeks). Observation of electron microscope images, focused on the CA1 stratum radiatum, revealed that BoNT/A induced an accumulation of synaptic vesicles. This accumulation triggered an enlargement of presynaptic terminals which was maximal at 4 weeks [30]. It is noteworthy that these changes were detectable basically only in asymmetric, excitatory synapses, and not in symmetric, GABAergic synapses, confirming a preferential effect of BoNT/A on excitatory terminals [20,21,22,30]. Axonal enlargements were also observed within the striatum injected with BoNT/A. These enlargements result positive for choline acetyltransferase (ChAT) and tyrosine hydroxylase (TH) in rats, but positive only for ChAT in mice [32,33].

3. BoNTs for the Study of Brain Physiology

A typical feature of BoNTs is that their action is prolonged but reversible. These characteristics make BoNTs, in particular BoNT/E which produces a short-lived blockade, ideal tools to study brain physiology. BoNTs allow a transient “silencing” of specific brain regions after a single administration, which is experimentally more convenient compared to other drugs that need to be continuously infused (e.g., tetrodotoxin or muscimol) [34].
Luvisetto, Pavone and collaborators were among the first to test the impact of direct brain injections of BoNTs in mice. They performed intracerebroventricular (icv) injections of sub-lethal doses of BoNT/A or BoNT/B and assessed various behavioral responses [35], such as active avoidance and object recognition. They also analyzed BoNTs effects on pharmacologically induced locomotor activity. The results indicated no effect on active avoidance acquisition, while there were impairments in the novel object recognition task, and amplified effects of drugs which induce locomotor activity [35]. The same group also tested the effects of central administration of BoNT/A on pain mechanisms [36]. They used a mouse model of formalin-induced pain (injection of formalin into the hindpaw) and the licking response as an index of pain. The data showed that intracerebral BoNT/A affected the licking response in the second phase of formalin test, similar to the effects obtained with peripheral administration [36,37]. Anti-nociceptive effects of central administrations of BoNT/A were later confirmed by other groups in various models of pain [38,39].
Our group has exploited BoNT/E to obtain a sustained but reversible blockade of neurotransmission for about 2 weeks in specific brain regions [27,40]. In particular, to investigate the role of cortical activity in the maturation of visual function, we unilaterally injected BoNT/E into the visual cortex (V1) in rat pups, at the time of eye opening [40]. BoNT/E injection produced a unilateral silencing of V1 for about 2 weeks, completely abolishing visual responses during the so called “critical period” for development of cortical function [41]. We performed electrophysiological recordings 3 weeks following BoNT/E injection (when cleaved SNAP-25 was no longer detectable), in order to assess visual system development when electrical activity was recovered, i.e., at the completion of the normal critical period. We found that BoNT/E-induced silencing of cortical activity did not allow normal maturation of visual function, keeping visual acuity low and extending the duration of the critical period [40]. We also evaluated if these deficits were persistent, or if they reflected only a delay in visual function maturation. Thus we performed behavioral and electrophysiological analyses at a longer time point (more than 2 months following toxin injection), and we confirmed a persistent impairment in visual performance. In conclusion, exploiting BoNT/E delivery to induce a transient silencing of cortical activity during the critical period allowed us to demonstrate that intrinsic cortical activity is necessary for a correct development of visual function [40].
Long-lasting serotypes such as BoNT/A and BoNT/B could be useful to create animal models of pathologies (e.g., dementia, [42]) or to treat hyperexcitability [26] (see below). However, these models could also offer basic knowledge about the role of specific brain regions in behavioral performance. For example, BoNT/B injection into the entorhinal cortex in adult rats produce learning and memory impairments as assessed by maze tests [42].
Similarly, BoNT/E hippocampal injection in adult rats induces deficits in spatial learning during the Morris water maze task, but since BoNT/E action is short-lived, the impairments are completely reversible and confirm a key role of hippocampus in spatial learning [26].
Mapping of the spread of BoNT/E via immunostaining for intact and cleaved SNAP-25 [40,43] demonstrates that toxin action remains confined to the cortical areas close to the injection site, thus allowing regional specificity of the synaptic blockade. Toxin diffusion can be further limited via the use of convection-enhanced delivery (CED), which provides a more homogeneous distribution than conventional bolus injection and does not damage the surrounding tissue [44,45,46,47].

4. Exploiting BoNTs in Pathological Brain Conditions

We have already reported examples of how BoNTs could be exploited to study the role of electrical activity in physiological, developmental brain processes [40]. In addition, BoNTs delivery could be useful to address the impact of electrical activity in neurodegenerative pathologies. Indeed, while it is known that synaptic degeneration precedes cell loss (e.g., [48]), little is known about mechanisms that tag synapses for degeneration. In this context, our group hypothesized that in a hippocampal mouse model of prion disease (a neurodegenerative disease associated with aggregates of misfolded proteins), synaptic degeneration was activity-dependent. To verify this hypothesis, we injected BoNT/A into the hippocampus of mice with prion disease and we analyzed synaptic degeneration at the ultrastructural level by electron microscopy [30]. Contrary to our expectations, we failed to find differences in the density of degenerating synapses between BoNT/A- and vehicle-injected prion mice. The morphology of the degenerating synapses was also indistinguishable between the two groups. These experiments challenge the idea that dysfunctions in synaptic vesicle release trigger the elimination of synaptic boutons, at least in prion-induced neurodegeneration [30,31].
Recently, Spalletti et al. (2017) used BoNT/E to produce a transient silencing of the contralesional hemisphere in a mouse model of focal stroke in the motor cortex. One of the main hypothesis in the stroke field is the “inter-hemispheric competition model”, which posits an enhanced transcallosal inhibition from the healthy to the lesioned side. To reduce this interhemispheric inhibition, the authors applied BoNT/E to block activity in the contralesional motor cortex immediately after the stroke. They found a significant recovery of motor function in the treated animals. Importantly, functional recovery was further enhanced when the silencing of the healthy side was coupled with physical rehabilitation of the affected arm [43].
Since BoNTs block neurotransmitter release, it is not unexpected that they have been exploited to treat, similarly to the peripheral nervous system, pathologies characterized by hyperexcitability. The most frequent category of central pathologies associated with hyperexcitability is epilepsy. About six millions of persons in Europe develop epilepsy, and around 30% of these are pharmaco-resistant [49]. This means that there are no drugs available to suppress or decrease their seizures. In the worst cases, the only clinical solution is a surgical intervention which physically removes the main epileptic focus. Consequently, efforts for discovering new therapeutic treatments are warranted. Our group and others have investigated whether central, local BoNTs delivery could suppress seizures in animal models of epilepsy [4,26,27,45,50]. The first serotype used was BoNT/E, tested in animal models of acute seizures, triggered by hippocampal administration of pro-convulsant agent kainic acid (KA) [26]. To measure BoNT/E effects, authors performed behavioral and electrographic analyses, demonstrating that BoNT/E delivery is effective in decreasing number and duration of seizures triggered by KA. BoNT/E effects were not limited to the electrophysiological level, but the toxin had an impact also on hippocampal histopathological changes, such as neuronal loss. This neuroprotection likely depends on blockade of excitoxicity phenomena occurring during prolonged electrical activity [26]. The neuroprotective action elicited by BoNT/E has been demonstrated also in a model of focal ischemia [51]. The potent vaso-constricting peptide endothelin-1 (ET-1) was delivered intrahippocampally in adult rats, followed 20 min later by BoNT/E injection in CA1. To evaluate BoNT/E action on excitoxicity, that is, on glutamate release, the authors performed in vivo microdialysis. Data showed that BoNT/E-injected rats had a decreased glutamate release. This synaptic effect was matched with a decrease in CA1 neuronal loss, as measured by immunohistochemistry [51]. Thus, the neuroprotective action by BoNT/E depends on the inhibition of the release of glutamate and occurs via downregulation of proapoptotic proteins, such as caspase-3 [52].
Based on these initial, encouraging data on acute seizures, BoNT/E was tested also in a mouse model of chronic seizures that resembles mesial temporal lobe epilepsy (MTLE), one of the most common pharmacoresistant forms of epilepsy in humans, obtained by intrahippocampal injection of KA [27,50]. The authors initially tested the impact of BoNT/E delivery on epileptogenesis (i.e., the development of spontaneous ictal events) following an episode of status epilepticus triggered by KA. The findings indicated that BoNT/E-mediated synaptic blockade during epileptogenesis was not effective in blocking the occurrence of spontaneous seizures. However, BoNT/E treatment was associated with histopatological protection; there was less neuronal loss in CA1 and the dispersion of granule cells in the dentate gyrus was potently prevented [27]. In a second work, the authors investigated if BoNT/E delivery was sufficient to reduce seizures during the chronic phase of epilepsy [50]. Mice injected with KA were implanted with bipolar electrodes, and after a period of baseline recording sessions, BoNT/E was infused directly into the epileptic hippocampus. Subsequent electrophysiological recordings clearly proved that BoNT/E delivery produces a reduction in total seizure duration and frequency [50].
One may argue that to be practically useful in the treatment of epilepsy, focal treatments require a long duration of action. Other serotypes of BoNTs with a prolonged proteolytic activity, like BoNT/A or BoNT/B, are ideal tools. Indeed, a couple of studies have used these serotypes to block seizures for longer periods in the amygdala kindling model, an experimental paradigm that allows to follow seizures for weeks to months. Gasior and colleagues (2013) directly infused BoNT/A or BoNT/B into the amygdala, via convection-enhanced delivery (CED) [45]. Therapeutic effects of both toxins were assessed by measuring after-discharge threshold and other parameters of the amygdala-kindled seizures at different times (3, 7, 10, 15, 21, 35, 50, and 64 days) after the administration. Results pointed to the anti-convulsant effects of both toxins, as assessed with EEG measures (i.e., elevation in after-discharge threshold of stimulation and seizures duration). The anti-convulsant action persisted until day 50. It interesting to note that, whilst BoNT/B was also effective in reduction of behavioral seizures, BoNT/A did not reach significance values in this parameter [45].
Another manuscript exploited infusion of BoNT/A (specifically serotype A2) to reduce seizures in kindled mice [53]. In half of the animals, BoNT/A2 was able to completely block the appearance of seizures. In addition, the toxin decreases the level of seizures, at least until 18 days following injection.
Taken together, these results suggest that BoNTs are quite effective in amelioration of epileptic activity, and they could be potentially used as focal antiepileptic treatments.
One might envision another possible “diagnostic” use of BoNTs in epilepsy, especially for BoNT/E, which has the shorter duration of action. In patients eligible for resection surgery, it is fundamental to precisely map brain epileptic foci, to remove all the hyperexcitable areas and render the patient seizure-free after surgery. The mapping is usually performed by non-invasive imaging techniques (such as magnetoencephalography (MEG) and functional MRI (fMRI)), or by EEG with chronically implanted electrodes [54], however the results are not always satisfactory, and patient could suffer of residual seizures also after surgery. In this context, local delivery of botulinum toxins could represent a strategy to functionally map the epileptogenic areas, and check whether the silencing of the presumptive focus is effective in abolishing seizures.
Another promising application of local delivery of BoNT/A is the therapeutic treatment of movement disorders and neurotransmission dysfunction typical of Parkinson’s disease (PD). PD is characterized by an imbalanced cholinergic hyperactivity in the striatum, due to the loss of dopaminergic neurons of the substantia nigra. Since BoNT/A blocks neurotransmitter release, including acetylcholine (ACh), the toxin was injected directly into the striatum, in animal models of PD [32,33,55,56,57]. In particular, the rodent model of 6-hydroxydopamine (6-OHDA) produces a hemi-parkinsonism. Wree and colleagues (2011) tested effects of BoNT/A injected 6 weeks following lesion with 6-OHDA. BoNT/A action was evaluated using the apomorphine-induced contralateral rotation test. Apomorphine is a dopamine (DA) receptor agonist and stimulates the supersensitive dopamine receptor D2 (DRD2) in the lesioned hemisphere, causing a net rotation away from the side of the lesion, that is, anti-clockwise [58]. Infusion of BoNT/A into the ipsilateral, lesioned striatum is able to reverse this rotation movement until 3 months [32]. Authors observed also enlarged axonal varicosities in BoNT/A (BiVs) injected-animals (possibly due to synaptic vesicles accumulation as seen in hippocampus by Caleo and co-authors [30]). Immunohistochemical analysis revealed that these axonal varicosities were cholinergic, but some of the BiVs were found to be positive for tyrosine hydroxylase (TH) [32,55]. In a subsequent work, these cholinergic varicosities induced by BoNT/A were investigated in detail [55]. They evaluated the number of ChAT-positive interneurons as well as the density and the volumetric size of the BiVs. In the ipsilateral side of BoNT/A-injected rats, with 6-OHDA lesion, the numeric density of BiVs reached a maximum 3 months after BoNT/A, while their volume increased during the whole time course of the experiment. However, no differences were detectable in the number of ChAT-positive neurons, up to 1 year following BoNT/A injection. This last result is important because it speaks in favor of a lack of cytotoxic effects of BoNT/A [55].
A similar study has been performed in mice, to extend possible therapeutic BoNT/A applications to genetics mouse models of PD [33]. Authors injected increasing doses of BoNT/A, finding no differences in the number of ChAT-positive interneurons. Increasing BoNT/A doses (from 25 pg to 200 pg), led to an increased BiV volume, and a decreased number of small BiVs. It is noteworthy that, in contrast to rats, TH-immunoreactive BiVs were not found in BoNT/A-infused mice [33].
Intrastriatally injected BoNT/A appears also to induce changes in receptor expression, likely due to activity silencing. For example, BoNT/A reduced density of dopamine receptor D2/D3, whereas other key receptors (such as dopamine 1 (D1), noradrenergic (a1 and a2) and serotonergic (5HT2A) receptors) remained basically unaltered in rats [57]. Since authors found few weeks after unilateral 6-hydroxydopamine (6-OHDA) lesion a significant increase of D2/D3 receptor ratio, the therapeutic effects of BoNT/A probably resides in reducing the interhemispheric imbalance in D2/D3 receptor density in lesioned rats.
Altogether, these results indicate how intracerebrally injected BoNTs could induce synaptic silencing and long-lasting changes in neurotransmitter-related proteins, that ultimately produce therapeutic benefits (see Table 1 for a summary).

5. Intracerebral BoNTs: Future Directions

BoNT clinical indications are continuously increasing, thanks to advantages such as very long duration, high potency, and complete reversibility of action [3].
There is currently considerable interest in developing novel forms of BoNTs with optimized therapeutic properties and neuronal selectivity (i.e., neuromuscular junction vs. sensory endings), which could offer new treatment opportunities. On one hand, the natural repertoire of BoNTs offers a wide variety of molecules with specific actions in neuronal cells and in vivo mouse models [59]. Second, an engineering approach has been taken to modify the pharmacological properties of native toxins by specific mutations. For example, a mutated BoNT/A1 has been created with faster onset and a shorter duration of action than BoNT/A1 wild type [60], opening the way to design BoNT variants with novel and useful properties.
The group of Bazbek Davletov has quite recently developed a new technology, named “protein-stapling”, by which it is possible to re-assemble chimeric clostridial neurotoxins starting from two separate modules, that is, the light chain/translocation domain and the receptor-binding domain [61,62]. This technology is not only useful to safely produce active toxins, but also allows engineering of toxins. The first engineered toxin was an analogue of the botulinum neurotoxin type A, called BiTox. The structural evaluation of BiTox suggests that the re-assembled BoNT/A could be substantially longer than the native molecule. However, BiTox demonstrated similar efficiency to that of native BoNT/A in proteolytic cleavage of SNAP-25 in vitro and in vivo, and thus in neurotransmitter silencing [61]. Interestingly, and clinically relevant, potency of BiTox at the neuromuscular junction is reduced, probably because of the bigger size of the molecule. Thus, systemic toxicity is reduced in BiTox injected subjects, and this represents a considerable advantage for clinical applications [61].
Engineered neurotoxins could also be exploited to enhance the selectivity for selected neuronal populations, combining the receptor-binding domain with different catalytic chains. For example, the same group has combined BoNT/A protease with the TeNT binding domain, allowing intoxication of different neuron populations compared to the native BoNT/A [62]. This chimera has a nociceptive action at central level, but has no action on motoneurons (as it caused neither flaccid nor spastic paralysis), resulting safer and potentially relevant for medical applications. On the other side, engineered toxins are interesting also for basic neuroscience research. Indeed, this chimera, following direct delivery into the rat visual cortex, was able to modulate sensory function [62].

Author Contributions

M.C. and L.R. wrote and discussed the manuscript.

Acknowledgments

We acknowledge financial support from AIRC (Italian Association for Cancer Research) grant #IG18925, Regione Toscana (RONDA Project, “Programma Attuativo Regionale” financed by FAS—now FSC), CNR InterOmics project, and CNR NanoMax project.

Conflicts of Interest

The authors declare no conflict of interest. The founding sponsors had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish the results.

References

  1. Van der Kloot, W.; Molgó, J. Quantal acetylcholine release at the vertebrate neuromuscular junction. Physiol. Rev. 1994, 74, 899–991. [Google Scholar] [CrossRef] [PubMed]
  2. Rossetto, O.; Pirazzini, M.; Montecucco, C. Botulinum neurotoxins: Genetic, structural and mechanistic insights. Nat. Rev. Microbiol. 2014, 12, 535–549. [Google Scholar] [CrossRef] [PubMed]
  3. Pirazzini, M.; Rossetto, O.; Eleopra, R.; Montecucco, C. Botulinum Neurotoxins: Biology, Pharmacology, and Toxicology. Pharmacol. Rev. 2017, 69, 200–235. [Google Scholar] [CrossRef] [PubMed]
  4. Akaike, N.; Shin, M.-C.; Wakita, M.; Torii, Y.; Harakawa, T.; Ginnaga, A.; Kato, K.; Kaji, R.; Kozaki, S. Transsynaptic inhibition of spinal transmission by A2 botulinum toxin. J. Physiol. 2013, 591, 1031–1043. [Google Scholar] [CrossRef] [PubMed]
  5. Whitemarsh, R.C.M.; Tepp, W.H.; Bradshaw, M.; Lin, G.; Pier, C.L.; Scherf, J.M.; Johnson, E.A.; Pellett, S. Characterization of botulinum neurotoxin A subtypes 1 through 5 by investigation of activities in mice, in neuronal cell cultures, and in vitro. Infect. Immun. 2013, 81, 3894–3902. [Google Scholar] [CrossRef] [PubMed]
  6. Peck, M.W.; Smith, T.J.; Anniballi, F.; Austin, J.W.; Bano, L.; Bradshaw, M.; Cuervo, P.; Cheng, L.W.; Derman, Y.; Dorner, B.G.; et al. Historical Perspectives and Guidelines for Botulinum Neurotoxin Subtype Nomenclature. Toxins 2017, 9. [Google Scholar] [CrossRef] [PubMed]
  7. Schiavo, G.; Matteoli, M.; Montecucco, C. Neurotoxins affecting neuroexocytosis. Physiol. Rev. 2000, 80, 717–766. [Google Scholar] [CrossRef] [PubMed]
  8. Montal, M. Botulinum neurotoxin: A marvel of protein design. Annu. Rev. Biochem. 2010, 79, 591–617. [Google Scholar] [CrossRef] [PubMed]
  9. Scott, A.B.; Rosenbaum, A.; Collins, C.C. Pharmacologic weakening of extraocular muscles. Investig. Ophthalmol. 1973, 12, 924–927. [Google Scholar]
  10. Bozzi, Y.; Costantin, L.; Antonucci, F.; Caleo, M. Action of botulinum neurotoxins in the central nervous system: Antiepileptic effects. Neurotox. Res. 2006, 9, 197–203. [Google Scholar] [CrossRef] [PubMed]
  11. Dong, M.; Yeh, F.; Tepp, W.H.; Dean, C.; Johnson, E.A.; Janz, R.; Chapman, E.R. SV2 is the protein receptor for botulinum neurotoxin A. Science 2006, 312, 592–596. [Google Scholar] [CrossRef] [PubMed]
  12. Verderio, C.; Rossetto, O.; Grumelli, C.; Frassoni, C.; Montecucco, C.; Matteoli, M. Entering neurons: Botulinum toxins and synaptic vesicle recycling. EMBO Rep. 2006, 7, 995–999. [Google Scholar] [CrossRef] [PubMed]
  13. Harper, C.B.; Papadopulos, A.; Martin, S.; Matthews, D.R.; Morgan, G.P.; Nguyen, T.H.; Wang, T.; Nair, D.; Choquet, D.; Meunier, F.A. Botulinum neurotoxin type-A enters a non-recycling pool of synaptic vesicles. Sci. Rep. 2016, 6, 19654. [Google Scholar] [CrossRef] [PubMed]
  14. Kroken, A.R.; Blum, F.C.; Zuverink, M.; Barbieri, J.T. Entry of Botulinum Neurotoxin Subtypes A1 and A2 into Neurons. Infect. Immun. 2017, 85. [Google Scholar] [CrossRef] [PubMed]
  15. Restani, L.; Giribaldi, F.; Manich, M.; Bercsenyi, K.; Menendez, G.; Rossetto, O.; Caleo, M.; Schiavo, G. Botulinum Neurotoxins A and E Undergo Retrograde Axonal Transport in Primary Motor Neurons. PLoS Pathog. 2012, 12. [Google Scholar] [CrossRef] [PubMed]
  16. Bomba-Warczak, E.; Vevea, J.D.; Brittain, J.M.; Figueroa-Bernier, A.; Tepp, W.H.; Johnson, E.A.; Yeh, F.L.; Chapman, E.R. Interneuronal Transfer and Distal Action of Tetanus Toxin and Botulinum Neurotoxins A and D in Central Neurons. Cell Rep. 2016, 16, 1974–1987. [Google Scholar] [CrossRef] [PubMed]
  17. Antonucci, F.; Rossi, C.; Gianfranceschi, L.; Rossetto, O.; Caleo, M. Long-distance retrograde effects of botulinum neurotoxin A. J. Neurosci. 2008, 28, 3689–3696. [Google Scholar] [CrossRef] [PubMed]
  18. Restani, L.; Novelli, E.; Bottari, D.; Leone, P.; Barone, I.; Galli-Resta, L.; Strettoi, E.; Caleo, M. Botulinum neurotoxin A impairs neurotransmission following retrograde transynaptic transport. Traffic 2012, 13, 1083–1089. [Google Scholar] [CrossRef] [PubMed]
  19. Ashton, A.C.; Dolly, J.O. Characterization of the inhibitory action of botulinum neurotoxin type A on the release of several transmitters from rat cerebrocortical synaptosomes. J. Neurochem. 1988, 50, 1808–1816. [Google Scholar] [CrossRef] [PubMed]
  20. Verderio, C.; Grumelli, C.; Raiteri, L.; Coco, S.; Paluzzi, S.; Caccin, P.; Rossetto, O.; Bonanno, G.; Montecucco, C.; Matteoli, M. Traffic of botulinum toxins A and E in excitatory and inhibitory neurons. Traffic 2007, 8, 142–153. [Google Scholar] [CrossRef] [PubMed]
  21. Garbelli, R.; Inverardi, F.; Medici, V.; Amadeo, A.; Verderio, C.; Matteoli, M.; Frassoni, C. Heterogeneous expression of SNAP-25 in rat and human brain. J. Comp. Neurol. 2008, 506, 373–386. [Google Scholar] [CrossRef] [PubMed]
  22. Verderio, C.; Pozzi, D.; Pravettoni, E.; Inverardi, F.; Schenk, U.; Coco, S.; Proux-Gillardeaux, V.; Galli, T.; Rossetto, O.; Frassoni, C.; et al. SNAP-25 Modulation of Calcium Dynamics Underlies Differences in GABAergic and Glutamatergic Responsiveness to Depolarization. Neuron 2004, 41, 599–610. [Google Scholar] [CrossRef]
  23. Beske, P.H.; Scheeler, S.M.; Adler, M.; McNutt, P.M. Accelerated intoxication of GABAergic synapses by botulinum neurotoxin A disinhibits stem cell-derived neuron networks prior to network silencing. Front. Cell. Neurosci. 2015, 9, 159. [Google Scholar] [CrossRef] [PubMed]
  24. Capogna, M.; McKinney, R.A.; O’Connor, V.; Gähwiler, B.H.; Thompson, S.M. Ca2+ or Sr2+ partially rescues synaptic transmission in hippocampal cultures treated with botulinum toxin A and C, but not tetanus toxin. J. Neurosci. 1997, 17, 7190–7202. [Google Scholar] [CrossRef] [PubMed]
  25. Sutton, M.A.; Wall, N.R.; Aakalu, G.N.; Schuman, E.M. Regulation of dendritic protein synthesis by miniature synaptic events. Science 2004, 304, 1979–1983. [Google Scholar] [CrossRef] [PubMed]
  26. Costantin, L.; Bozzi, Y.; Richichi, C.; Viegi, A.; Antonucci, F.; Funicello, M.; Gobbi, M.; Mennini, T.; Rossetto, O.; Montecucco, C.; et al. Antiepileptic effects of botulinum neurotoxin E. J. Neurosci. 2005, 25, 1943–1951. [Google Scholar] [CrossRef] [PubMed]
  27. Antonucci, F.; Di Garbo, A.; Novelli, E.; Manno, I.; Sartucci, F.; Bozzi, Y.; Caleo, M. Botulinum neurotoxin E (BoNT/E) reduces CA1 neuron loss and granule cell dispersion, with no effects on chronic seizures, in a mouse model of temporal lobe epilepsy. Exp. Neurol. 2008, 210, 388–401. [Google Scholar] [CrossRef] [PubMed]
  28. Montecucco, C.; Schiavo, G.; Pantano, S. SNARE complexes and neuroexocytosis: How many, how close? Trends Biochem. Sci. 2005, 30, 367–372. [Google Scholar] [CrossRef] [PubMed]
  29. Keller, J.E.; Neale, E.A. The role of the synaptic protein snap-25 in the potency of botulinum neurotoxin type A. J. Biol. Chem. 2001, 276, 13476–13482. [Google Scholar] [CrossRef] [PubMed]
  30. Caleo, M.; Restani, L.; Vannini, E.; Siskova, Z.; Al-Malki, H.; Morgan, R.; O’Connor, V.; Perry, V.H. The role of activity in Synaptic degeneration in a protein misfolding disease, prion disease. PLoS ONE 2012, 7. [Google Scholar] [CrossRef] [PubMed]
  31. Caleo, M.; Restani, L.; Perry, V.H. Silencing synapses: A route to understanding synapse degeneration in chronic neurodegenerative disease. Prion 2013, 7, 147–150. [Google Scholar] [CrossRef] [PubMed]
  32. Wree, A.; Mix, E.; Hawlitschka, A.; Antipova, V.; Witt, M.; Schmitt, O.; Benecke, R. Intrastriatal botulinum toxin abolishes pathologic rotational behaviour and induces axonal varicosities in the 6-OHDA rat model of Parkinson’s disease. Neurobiol. Dis. 2011, 41, 291–298. [Google Scholar] [CrossRef] [PubMed]
  33. Hawlitschka, A.; Holzmann, C.; Witt, S.; Spiewok, J.; Neumann, A.-M.; Schmitt, O.; Wree, A.; Antipova, V. Intrastriatally injected botulinum neurotoxin-A differently effects cholinergic and dopaminergic fibers in C57BL/6 mice. Brain Res. 2017, 1676, 46–56. [Google Scholar] [CrossRef] [PubMed]
  34. Davletov, B.; Bajohrs, M.; Binz, T. Beyond BOTOX: Advantages and limitations of individual botulinum neurotoxins. Trends Neurosci. 2005, 28, 446–452. [Google Scholar] [CrossRef] [PubMed]
  35. Luvisetto, S.; Marinelli, S.; Rossetto, O.; Montecucco, C.; Pavone, F. Central injection of botulinum neurotoxins: Behavioural effects in mice. Behav. Pharmacol. 2004, 15, 233–240. [Google Scholar] [CrossRef] [PubMed]
  36. Luvisetto, S.; Marinelli, S.; Lucchetti, F.; Marchi, F.; Cobianchi, S.; Rossetto, O.; Montecucco, C.; Pavone, F. Botulinum neurotoxins and formalin-induced pain: Central vs. peripheral effects in mice. Brain Res. 2006, 1082, 124–131. [Google Scholar] [CrossRef] [PubMed]
  37. Cui, M.; Khanijou, S.; Rubino, J.; Aoki, K.R. Subcutaneous administration of botulinum toxin A reduces formalin-induced pain. Pain 2004, 107, 125–133. [Google Scholar] [CrossRef] [PubMed]
  38. Bach-Rojecky, L.; Lacković, Z. Central origin of the antinociceptive action of botulinum toxin type A. Pharmacol. Biochem. Behav. 2009, 94, 234–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Matak, I.; Lacković, Z. Botulinum toxin A, brain and pain. Prog. Neurobiol. 2014, 119–120, 39–59. [Google Scholar] [CrossRef] [PubMed]
  40. Caleo, M.; Restani, L.; Gianfranceschi, L.; Costantin, L.; Rossi, C.; Rossetto, O.; Montecucco, C.; Maffei, L. Transient synaptic silencing of developing striate cortex has persistent effects on visual function and plasticity. J. Neurosci. 2007, 27, 4530–4540. [Google Scholar] [CrossRef] [PubMed]
  41. Takesian, A.E.; Hensch, T.K. Balancing plasticity/stability across brain development. Prog. Brain Res. 2013, 207, 3–34. [Google Scholar] [PubMed]
  42. Ando, S.; Kobayashi, S.; Waki, H.; Kon, K.; Fukui, F.; Tadenuma, T.; Iwamoto, M.; Takeda, Y.; Izumiyama, N.; Watanabe, K.; et al. Animal model of dementia induced by entorhinal synaptic damage and partial restoration of cognitive deficits by BDNF and carnitine. J. Neurosci. Res. 2002, 70, 519–527. [Google Scholar] [CrossRef] [PubMed]
  43. Spalletti, C.; Alia, C.; Lai, S.; Panarese, A.; Conti, S.; Micera, S.; Caleo, M. Combining robotic training and inactivation of the healthy hemisphere restores pre-stroke motor patterns in mice. Elife 2017, 6. [Google Scholar] [CrossRef] [PubMed]
  44. Rogawski, M.A. Convection-enhanced delivery in the treatment of epilepsy. Neurotherapeutics 2009, 6, 344–351. [Google Scholar] [CrossRef] [PubMed]
  45. Gasior, M.; Tang, R.; Rogawski, M.A. Long-lasting attenuation of amygdala-kindled seizures after convection-enhanced delivery of botulinum neurotoxins A and B into the amygdala in rats. J. Pharmacol. Exp. Ther. 2013, 346, 528–534. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, C.; Mastorakos, P.; Sobral, M.; Berry, S.; Song, E.; Nance, E.; Eberhart, C.G.; Hanes, J.; Suk, J.S. Strategies to enhance the distribution of nanotherapeutics in the brain. J. Control. Release 2017, 267, 232–239. [Google Scholar] [CrossRef] [PubMed]
  47. Barua, N.U.; Gill, S.S.; Love, S. Convection-enhanced drug delivery to the brain: Therapeutic potential and neuropathological considerations. Brain Pathol. 2014, 24, 117–127. [Google Scholar] [CrossRef] [PubMed]
  48. Cunningham, C.; Deacon, R.; Wells, H.; Boche, D.; Waters, S.; Diniz, C.P.; Scott, H.; Rawlins, J.N.P.; Perry, V.H. Synaptic changes characterize early behavioural signs in the ME7 model of murine prion disease. Eur. J. Neurosci. 2003, 17, 2147–2155. [Google Scholar] [CrossRef] [PubMed]
  49. Van Vliet, E.A.; Aronica, E.; Gorter, J.A. Role of blood-brain barrier in temporal lobe epilepsy and pharmacoresistance. Neuroscience 2014, 277, 455–473. [Google Scholar] [CrossRef] [PubMed]
  50. Antonucci, F.; Bozzi, Y.; Caleo, M. Intrahippocampal infusion of botulinum neurotoxin E (BoNT/E) reduces spontaneous recurrent seizures in a mouse model of mesial temporal lobe epilepsy. Epilepsia 2009, 50, 963–966. [Google Scholar] [CrossRef] [PubMed]
  51. Antonucci, F.; Cerri, C.; Maya Vetencourt, J.F.; Caleo, M. Acute neuroprotection by the synaptic blocker botulinum neurotoxin E in a rat model of focal cerebral ischaemia. Neuroscience 2010, 169, 395–401. [Google Scholar] [CrossRef] [PubMed]
  52. Manno, I.; Antonucci, F.; Caleo, M.; Bozzi, Y. BoNT/E prevents seizure-induced activation of caspase 3 in the rat hippocampus. Neuroreport 2007, 18, 373–376. [Google Scholar] [CrossRef] [PubMed]
  53. Kato, K.; Akaike, N.; Kohda, T.; Torii, Y.; Goto, Y.; Harakawa, T.; Ginnaga, A.; Kaji, R.; Kozaki, S. Botulinum neurotoxin A2 reduces incidence of seizures in mouse models of temporal lobe epilepsy. Toxicon 2013, 74, 109–115. [Google Scholar] [CrossRef] [PubMed]
  54. Dorfer, C.; Widjaja, E.; Ochi, A.; Carter Snead, O., III; Rutka, J.T. Epilepsy surgery: Recent advances in brain mapping, neuroimaging and surgical procedures. J. Neurosurg. Sci. 2015, 59, 141–155. [Google Scholar] [PubMed]
  55. Mehlan, J.; Brosig, H.; Schmitt, O.; Mix, E.; Wree, A.; Hawlitschka, A. Intrastriatal injection of botulinum neurotoxin-A is not cytotoxic in rat brain—A histological and stereological analysis. Brain Res. 2016, 1630, 18–24. [Google Scholar] [CrossRef] [PubMed]
  56. Wedekind, F.; Oskamp, A.; Lang, M.; Hawlitschka, A.; Zilles, K.; Wree, A.; Bauer, A. Intrastriatal administration of botulinum neurotoxin A normalizes striatal D2R binding and reduces striatal D1R binding in male hemiparkinsonian rats. J. Neurosci. Res. 2018, 96, 75–86. [Google Scholar] [CrossRef] [PubMed]
  57. Mann, T.; Zilles, K.; Dikow, H.; Hellfritsch, A.; Cremer, M.; Piel, M.; Rösch, F.; Hawlitschka, A.; Schmitt, O.; Wree, A. Dopamine, Noradrenaline and Serotonin Receptor Densities in the Striatum of Hemiparkinsonian Rats following Botulinum Neurotoxin-A Injection. Neuroscience 2018, 374, 187–204. [Google Scholar] [CrossRef] [PubMed]
  58. Ungerstedt, U.; Butcher, L.L.; Butcher, S.G.; Andén, N.E.; Fuxe, K. Direct chemical stimulation of dopaminergic mechanisms in the neostriatum of the rat. Brain Res. 1969, 14, 461–471. [Google Scholar] [CrossRef]
  59. Pellett, S.; Bradshaw, M.; Tepp, W.H.; Pier, C.L.; Whitemarsh, R.C.M.; Chen, C.; Barbieri, J.T.; Johnson, E.A. The Light Chain Defines the Duration of Action of Botulinum Toxin Serotype A Subtypes. MBio 2018, 9. [Google Scholar] [CrossRef] [PubMed]
  60. Scheps, D.; López de la Paz, M.; Jurk, M.; Hofmann, F.; Frevert, J. Design of modified botulinum neurotoxin A1 variants with a shorter persistence of paralysis and duration of action. Toxicon 2017, 139, 101–108. [Google Scholar] [CrossRef] [PubMed]
  61. Ferrari, E.; Maywood, E.S.; Restani, L.; Caleo, M.; Pirazzini, M.; Rossetto, O.; Hastings, M.H.; Niranjan, D.; Schiavo, G.; Davletov, B. Re-assembled botulinum neurotoxin inhibits CNS functions without systemic toxicity. Toxins 2011, 3, 345–355. [Google Scholar] [CrossRef] [PubMed]
  62. Ferrari, E.; Gu, C.; Niranjan, D.; Restani, L.; Rasetti-Escargueil, C.; Obara, I.; Geranton, S.M.; Arsenault, J.; Goetze, T.A.; Harper, C.B.; et al. Synthetic self-assembling clostridial chimera for modulation of sensory functions. Bioconjug. Chem. 2013, 24, 1750–1759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Exploiting botulinum neurotoxins (BoNTs) in pathological brain conditions. The table summarizes the main studies that have exploited central delivery of botulinum neurotoxins to treat pathological brain conditions.
Table 1. Exploiting botulinum neurotoxins (BoNTs) in pathological brain conditions. The table summarizes the main studies that have exploited central delivery of botulinum neurotoxins to treat pathological brain conditions.
DiseaseAnimal ModelSpeciesBoNT SerotypeReported EffectsReference
Epilepsyintrahippocampal KAratBoNT/Edecreased number and duration of seizures triggered by KA; decreased neuronal lossCostantin et al, 2005 [26]
intrahippocampal KAratBoNT/Edownregulation of caspase 3Manno et al, 2007 [52]
intrahippocampal KAmouseBoNT/Edecreased neuronal loss and dispersion of granule cells (BoNT/E tested during epileptogenesis)Antonucci et al, 2008 [27]
intrahippocampal KAmouseBoNT/Ereduction of total seizure duration and frequency (BoNT/E tested during chronic phase)Antonucci et al, 2009 [50]
amygdala kindling modelratBoNT/A BoNT/Banti-convulsant effects of both toxins (BoNT/B also at behavioral level)Gasior et al, 2013 [45]
amygdala kindling modelmouseBoNT/A2decreased seizures (in 50% of animals)Kato et al, 2013 [53]
Ischemiaendothelin 1ratBoNT/Eneuroprotective effect (decrease of glutamate release)Antonucci et al, 2010 [48]
phototrombotic strokemouseBoNT/Esynaptic silencing of contralateral hemisphere improved motor recoverySpalletti et al, 2017 [43]
Parkinson’s disease6-OHDA modelratBoNT/Aabolished pathologic rotational behavior; induced ChAT and TH axonal varicositiesWree et al, 2011 [32]
6-OHDA modelratBoNT/Ainduced ChAT and TH axonal varicosities; no changes in ChAT-positive neuronsMehlan et al, 2016 [55]
6-OHDA modelmouseBoNT/Ainduced ChAT axonal varicosities;Hawlitschka et al, 2017 [33]
6-OHDA modelratBoNT/Achanges in receptor expression (rebalance of D2/D3 receptor density)Mann et al, 2018 [57]
Prion diseaseME7 prion diseasemouseBoNT/Aelectrical activity does not impact on synaptic degeneration Caleo et al, 2012 [30]
Painformalin-induced painmouseBoNT/Adecreased licking response in the second phase of formalin testLuvisetto et al, 2006 [36]

Share and Cite

MDPI and ACS Style

Caleo, M.; Restani, L. Exploiting Botulinum Neurotoxins for the Study of Brain Physiology and Pathology. Toxins 2018, 10, 175. https://doi.org/10.3390/toxins10050175

AMA Style

Caleo M, Restani L. Exploiting Botulinum Neurotoxins for the Study of Brain Physiology and Pathology. Toxins. 2018; 10(5):175. https://doi.org/10.3390/toxins10050175

Chicago/Turabian Style

Caleo, Matteo, and Laura Restani. 2018. "Exploiting Botulinum Neurotoxins for the Study of Brain Physiology and Pathology" Toxins 10, no. 5: 175. https://doi.org/10.3390/toxins10050175

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop