Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
1.
ALTEX ; 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39072354

ABSTRACT

Botulinum neurotoxin type A (BoNT/A) is a widely used biopharmaceutic for the treatment of neurological diseases and aesthetic medicine, allowing months-long paralysis of target muscles and glands. Large numbers of mice are used for multiple botulinum applications including batch release potency testing, antitoxin testing, countermeasure development and basic research. The mouse bioassay (MBA) has historically been the industry gold-standard in the botulinum field and is still heavily used for commercial product testing. BoNT/A intoxication causes severe suffering and application-specific, non-animal alternatives are urgently needed. It is widely accepted, that a cell-based assay (CBA) is the only way to faithfully replicate all the physiological steps of botulinum intoxication; comprising neuronal binding, internalization, endosomal escape, and cleavage of synaptosomal-associated protein of 25 kDa (SNAP25). However, it has not been straightforward to develop these assays and there are only a limited number of CBA currently in use. This is in part, due to the fact that very few cell lines have the appropriate levels of sensitivity to BoNT/A. In this study we have identified that LAN5 cells, a human neuroblastoma derived cell line, are sensitive to BoNT/A and can be engineered to express a recombinant NanoLuc luciferase tagged SNAP25 reporter molecule. On intoxication, the reporter molecule is cleaved and releases a NanoLuc-SNAP25 fragment which can be specifically captured on a 96-well plate for quantitative luminometry. Importantly, we demonstrate this new cell-based assay exhibits sensitivity comparable to the MBA.


Botulinum neurotoxin type A (BoNT/A) is extensively used in the treatment of neurological disorders and aesthetics. When the toxin enters cells, it targets a protein called SNAP25 and inhibits neurotransmitter release. Traditionally, the potency and safety of BoNT/A has been tested using the mouse bioassay, which causes significant distress to the animals being used. Our study introduces a new method for detecting BoNT/A activity based on LAN5 cells, which are a self-replicating, neuroblastoma-derived human cell line. We have engineered the cells to express a version of SNAP25 that allows the potency of BoNT/A to be measured using a luminescence assay. This new cell-based assay is as sensitive as the mouse bioassay and can be used for commercial product testing. This development could lead to fewer animals being used in research and commercial testing of BoNT/A, benefiting both scientific progress and animal welfare.

2.
Toxins (Basel) ; 16(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38668600

ABSTRACT

This review discusses the expanding application of botulinum neurotoxin in treating neurological conditions. The article specifically explores novel approaches to using non-paralytic botulinum molecules. These new molecules, such as BiTox or el-iBoNT, offer an alternative for patients who face limitations in using paralytic forms of botulinum neurotoxin due to concerns about muscle function loss. We highlight the research findings that confirm not only the effectiveness of these molecules but also their reduced paralytic effect. We also discuss a potential cause for the diminished paralytic action of these molecules, specifically changes in the spatial parameters of the new botulinum molecules. In summary, this article reviews the current research that enhances our understanding of the application of new botulinum neurotoxins in the context of common conditions and suggests new avenues for developing more efficient molecules.


Subject(s)
Botulinum Toxins , Humans , Botulinum Toxins/therapeutic use , Animals , Protein Engineering , Nervous System Diseases/drug therapy
3.
J Pain ; 25(6): 104466, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38218509

ABSTRACT

Chronic pain presents an enormous personal and economic burden and there is an urgent need for effective treatments. In a mouse model of chronic neuropathic pain, selective silencing of key neurons in spinal pain signalling networks with botulinum constructs resulted in a reduction of pain behaviours associated with the peripheral nerve. However, to establish clinical relevance it was important to know how long this silencing period lasted. Now, we show that neuronal silencing and the concomitant reduction of neuropathic mechanical and thermal hypersensitivity lasts for up to 120d following a single injection of botulinum construct. Crucially, we show that silencing and analgesia can then be reinstated with a second injection of the botulinum conjugate. Here we demonstrate that single doses of botulinum-toxin conjugates are a powerful new way of providing long-term neuronal silencing and pain relief. PERSPECTIVE: This research demonstrates that botulinum-toxin conjugates are a powerful new way of providing long-term neuronal silencing without toxicity following a single injection of the conjugate and have the potential for repeated dosing when silencing reverses.


Subject(s)
Disease Models, Animal , Neuralgia , Animals , Mice , Neuralgia/drug therapy , Male , Mice, Inbred C57BL , Chronic Pain/drug therapy , Botulinum Toxins, Type A/pharmacology , Botulinum Toxins, Type A/administration & dosage , Hyperalgesia/drug therapy , Botulinum Toxins/administration & dosage , Botulinum Toxins/pharmacology
4.
Life Sci Alliance ; 6(6)2023 06.
Article in English | MEDLINE | ID: mdl-37041008

ABSTRACT

Chronic pain affects one in five people across human societies, with few therapeutic options available. Botulinum neurotoxin (BoNT) can provide long-lasting pain relief by inhibiting local release of neuropeptides and neurotransmitters, but its highly paralytic nature has limited its analgesic potential. Recent advances in protein engineering have raised the possibility of synthesising non-paralysing botulinum molecules for translation to pain sufferers. However, the synthesis of these molecules, via several synthetic steps, has been challenging. Here, we describe a simple platform for safe production of botulinum molecules for treating nerve injury-induced pain. We produced two versions of isopeptide-bonded BoNT from separate botulinum parts using an isopeptide bonding system. Although both molecules cleaved their natural substrate, SNAP25, in sensory neurons, the structurally elongated iBoNT did not cause motor deficit in rats. We show that the non-paralytic elongated iBoNT targets specific cutaneous nerve fibres and provides sustained pain relief in a rat nerve injury model. Our results demonstrate that novel botulinum molecules can be produced in a simple and safe manner and be useful for treating neuropathic pain.


Subject(s)
Botulinum Toxins, Type A , Chronic Pain , Neuralgia , Rats , Humans , Animals , Chronic Pain/drug therapy , Botulinum Toxins, Type A/metabolism , Botulinum Toxins, Type A/pharmacology , Botulinum Toxins, Type A/therapeutic use , Analgesics/pharmacology , Analgesics/therapeutic use , Sensory Receptor Cells/metabolism
5.
Int J Mol Sci ; 23(3)2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35163009

ABSTRACT

The fusion of membranes is a central part of the physiological processes involving the intracellular transport and maturation of vesicles and the final release of their contents, such as neurotransmitters and hormones, by exocytosis. Traditionally, in this process, proteins, such SNAREs have been considered the essential components of the fusion molecular machinery, while lipids have been seen as merely structural elements. Nevertheless, sphingosine, an intracellular signalling lipid, greatly increases the release of neurotransmitters in neuronal and neuroendocrine cells, affecting the exocytotic fusion mode through the direct interaction with SNAREs. Moreover, recent studies suggest that FTY-720 (Fingolimod), a sphingosine structural analogue used in the treatment of multiple sclerosis, simulates sphingosine in the promotion of exocytosis. Furthermore, this drug also induces the intracellular fusion of organelles such as dense vesicles and mitochondria causing cell death in neuroendocrine cells. Therefore, the effect of sphingosine and synthetic derivatives on the heterologous and homologous fusion of organelles can be considered as a new mechanism of action of sphingolipids influencing important physiological processes, which could underlie therapeutic uses of sphingosine derived lipids in the treatment of neurodegenerative disorders and cancers of neuronal origin such neuroblastoma.


Subject(s)
Exocytosis/drug effects , Neuroendocrine Cells/metabolism , Sphingosine/metabolism , Animals , Biological Transport , Humans , Membrane Fusion , SNARE Proteins/metabolism , Sphingosine/pharmacology
6.
Biologicals ; 71: 31-41, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33910767

ABSTRACT

Batch release testing for human and veterinary tetanus vaccines still relies heavily on methods that involve animals, particularly for potency testing. The quantity and quality of tetanus antigen present in these products is of utmost importance for product safety and clinical effect. Immunochemical methods that measure consistency of antigen content and quality, potentially as an indicator of potency, could be a better choice and negate the need for an in vivo potency test. These immunochemical methods require at least one well characterised monoclonal antibody (mAb) that is specific for the target antigen. In this paper we report the results of the comprehensive characterisation of a panel of mAbs against tetanus with a view to select antibodies that can be used for development of an in vitro potency immunoassay. We have assessed binding of the antibodies to native antigen (toxin), detoxified antigen (toxoid), adsorbed antigen and heat-altered antigen. Antibody function was determined using an in-house cell-based neutralisation assay to support prior in vivo potency data that was available for some, but not all, of the antibodies. In addition, antibody affinity was measured, and epitope competition analysis was performed to identify pairs of antibodies that could be deployed in a sandwich immunoassay format. Not all characterisation tests provided evidence of "superiority" of one mAb over another, but together the results from all characterisation studies allowed for selection of an antibody pair to be taken forward to assay development.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoassay , Tetanus Toxoid/immunology , Vaccine Potency , Animal Testing Alternatives , Animals , Tetanus/prevention & control
7.
Neurotherapeutics ; 18(1): 556-568, 2021 01.
Article in English | MEDLINE | ID: mdl-33205382

ABSTRACT

With a prevalence of 15%, migraine is the most common neurological disorder and among the most disabling diseases, taking into account years lived with disability. Current oral medications for migraine show variable effects and are frequently associated with intolerable side effects, leading to the dissatisfaction of both patients and doctors. Injectable therapeutics, which include calcitonin gene-related peptide-targeting monoclonal antibodies and botulinum neurotoxin A (BoNT/A), provide a new paradigm for treatment of chronic migraine but are effective only in approximately 50% of subjects. Here, we investigated a novel engineered botulinum molecule with markedly reduced muscle paralyzing properties which could be beneficial for the treatment of migraine. This stapled botulinum molecule with duplicated binding domain-binary toxin-AA (BiTox/AA)-cleaves synaptosomal-associated protein 25 with a similar efficacy to BoNT/A in neurons; however, the paralyzing effect of BiTox/AA was 100 times less when compared to native BoNT/A following muscle injection. The performance of BiTox/AA was evaluated in cellular and animal models of migraine. BiTox/AA inhibited electrical nerve fiber activity in rat meningeal preparations while, in the trigeminovascular model, BiTox/AA raised electrical and mechanical stimulation thresholds in Aδ- and C-fiber nociceptors. In the rat glyceryl trinitrate (GTN) model, BiTox/AA proved effective in inhibiting GTN-induced hyperalgesia in the orofacial formalin test. We conclude that the engineered botulinum molecule provides a useful prototype for designing advanced future therapeutics for an improved efficacy in the treatment of migraine.


Subject(s)
Analgesics/pharmacology , Botulinum Toxins/pharmacology , Migraine Disorders/drug therapy , Analgesics/administration & dosage , Animals , Botulinum Toxins/administration & dosage , Cell Line, Tumor/drug effects , Disease Models, Animal , Electromyography , Humans , Male , Muscle, Skeletal/drug effects , Nitroglycerin/pharmacology , Rats , Rats, Sprague-Dawley , Trigeminal Ganglion/drug effects
8.
Toxicon X ; 5: 100019, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32140681

ABSTRACT

Neurological diseases constitute a quarter of global disease burden and are expected to rise worldwide with the ageing of human populations. There is an increasing need to develop new molecular systems which can deliver drugs specifically into neurons, non-dividing cells meant to last a human lifetime. Neuronal drug delivery must rely on agents which can recognise neurons with high specificity and affinity. Here we used a recently introduced 'stapling' system to prepare macromolecules carrying duplicated binding domains from the clostridial family of neurotoxins. We engineered individual parts of clostridial neurotoxins separately and combined them using a strong alpha-helical bundle. We show that combining two identical binding domains of tetanus and botulinum type D neurotoxins, in a sterically defined way by protein stapling, allows enhanced intracellular delivery of molecules into neurons. We also engineered a botulinum neurotoxin type C variant with a duplicated binding domain which increased enzymatic delivery compared to the native type C toxin. We conclude that duplication of the binding parts of tetanus or botulinum neurotoxins will allow production of high avidity agents which could deliver imaging reagents and large therapeutic enzymes into neurons with superior efficiency.

9.
Sci Rep ; 9(1): 18471, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804600

ABSTRACT

FTY-720 (Fingolimod) was one of the first compounds authorized for the treatment of multiple sclerosis. Among its other activities, this sphingosine analogue enhances exocytosis in neuroendocrine chromaffin cells, altering the quantal release of catecholamines. Surprisingly, the size of chromaffin granules is reduced within few minutes of treatment, a process that is paralleled by the homotypic fusion of granules and their heterotypic fusion with mitochondria, as witnessed by dynamic confocal and TIRF microscopy. Electron microscopy studies support these observations, revealing the fusion of several vesicles with individual mitochondria to form large, round mixed organelles. This cross-fusion is SNARE-dependent, being partially prevented by the expression of an inactive form of SNAP-25. Fused mitochondria exhibit an altered redox potential, which dramatically enhances cell death. Therefore, the cross-fusion of intracellular organelles appears to be a new mechanism to be borne in mind when considering the effect of FTY-720 on the survival of neuroendocrine cells.


Subject(s)
Chromaffin Granules/drug effects , Fingolimod Hydrochloride/toxicity , Multiple Sclerosis/drug therapy , Neuroendocrine Cells/drug effects , Animals , Cattle , Cells, Cultured , Chromaffin Granules/metabolism , Chromaffin Granules/pathology , Humans , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Primary Cell Culture , Synaptosomal-Associated Protein 25/metabolism , Toxicity Tests
10.
Toxins (Basel) ; 10(8)2018 08 01.
Article in English | MEDLINE | ID: mdl-30071628

ABSTRACT

In the recent past, about 40 botulinum neurotoxin (BoNT) subtypes belonging to serotypes A, B, E, and F pathogenic to humans were identified among hundreds of independent isolates. BoNTs are the etiological factors of botulism and represent potential bioweapons; however, they are also recognized pharmaceuticals for the efficient counteraction of hyperactive nerve terminals in a variety of human diseases. The detailed biochemical characterization of subtypes as the basis for development of suitable countermeasures and possible novel therapeutic applications is lagging behind the increase in new subtypes. Here, we report the primary structure of a ninth subtype of BoNT/F. Its amino-acid sequence diverges by at least 8.4% at the holotoxin and 13.4% at the enzymatic domain level from all other known BoNT/F subtypes. We found that BoNT/F9 shares the scissile Q58/K59 bond in its substrate vesicle associated membrane protein 2 with the prototype BoNT/F1. Comparative biochemical analyses of four BoNT/F enzymatic domains showed that the catalytic efficiencies decrease in the order F1 > F7 > F9 > F6, and vary by up to a factor of eight. KM values increase in the order F1 > F9 > F6 ≈ F7, whereas kcat decreases in the order F7 > F1 > F9 > F6. Comparative substrate scanning mutagenesis studies revealed a unique pattern of crucial substrate residues for each subtype. Based upon structural coordinates of F1 bound to an inhibitor polypeptide, the mutational analyses suggest different substrate interactions in the substrate binding channel of each subtype.


Subject(s)
Botulinum Toxins/chemistry , Peptides/chemistry , Vesicle-Associated Membrane Protein 2/chemistry , Catalysis , Substrate Specificity
11.
FEBS Lett ; 592(21): 3493-3503, 2018 11.
Article in English | MEDLINE | ID: mdl-29962039

ABSTRACT

Membrane fusion is a key event in exocytosis of neurotransmitters and hormones stored in intracellular vesicles. In this process, soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) proteins are essential components of the exocytotic molecular machinery, while lipids have been seen traditionally as structural elements. However, the so-called signalling lipids, such as sphingosine and arachidonic acid, interact with SNAREs and directly modulate the frequency and mode of fusion events. Interestingly, recent work has proved that the sphingosine analogue FTY-720, used in the treatment of multiple sclerosis, mimics the effects of signalling lipids. In the present Review, we discuss recent investigations suggesting that endogenous signalling lipids and synthetic analogues can modulate important physiological aspects of secretion, such as quantal release, vesicle recruitment into active sites, vesicle transport and even organelle fusion in the cytosol. Therefore, these compounds are far from being merely structural components of cellular membranes.


Subject(s)
Arachidonic Acid/metabolism , Exocytosis/physiology , Signal Transduction , Sphingosine/metabolism , Animals , Humans , Membrane Fusion , Protein Binding , SNARE Proteins/metabolism
12.
Sci Transl Med ; 10(450)2018 07 18.
Article in English | MEDLINE | ID: mdl-30021888

ABSTRACT

Chronic pain is a widespread debilitating condition affecting millions of people worldwide. Although several pharmacological treatments for relieving chronic pain have been developed, they require frequent chronic administration and are often associated with severe adverse events, including overdose and addiction. Persistent increased sensitization of neuronal subpopulations of the peripheral and central nervous system has been recognized as a central mechanism mediating chronic pain, suggesting that inhibition of specific neuronal subpopulations might produce antinociceptive effects. We leveraged the neurotoxic properties of the botulinum toxin to specifically silence key pain-processing neurons in the spinal cords of mice. We show that a single intrathecal injection of botulinum toxin conjugates produced long-lasting pain relief in mouse models of inflammatory and neuropathic pain without toxic side effects. Our results suggest that this strategy might be a safe and effective approach for relieving chronic pain while avoiding the adverse events associated with repeated chronic drug administration.


Subject(s)
Botulinum Toxins/toxicity , Chronic Pain/prevention & control , Neurons/metabolism , Analgesics/pharmacology , Animals , Botulinum Toxins/administration & dosage , Cell Death/drug effects , Chronic Pain/pathology , Endocytosis/drug effects , Inflammation/pathology , Inflammation/prevention & control , Male , Mice, Inbred C57BL , Morphine/pharmacology , Neurons/drug effects , Receptors, Neurokinin-1/metabolism , Receptors, Opioid, mu/metabolism
13.
Toxins (Basel) ; 10(7)2018 06 27.
Article in English | MEDLINE | ID: mdl-29954071

ABSTRACT

Immunotoxins are being investigated as anti-cancer therapies and consist of a cytotoxic enzyme fused to a cancer targeting antibody. All currently used toxins function via the inhibition of protein synthesis, making them highly potent in both healthy and transformed cells. This non-specific cell killing mechanism causes dose-limiting side effects that can severely limit the potential of immunotoxin therapy. In this study, the recently characterised bacterial toxin Burkholderia lethal factor 1 (BLF1) is investigated as a possible alternative payload for targeted toxin therapy in the treatment of neuroblastoma. BLF1 inhibits translation initiation by inactivation of eukaryotic initiation translation factor 4A (eIF4A), a putative anti-cancer target that has been shown to regulate a number of oncogenic proteins at the translational level. We show that cellular delivery of BLF1 selectively induces apoptosis in neuroblastoma cells that display MYCN amplification but has little effect on non-transformed cells. Future immunotoxins based on this enzyme may therefore have higher specificity towards MYCN-amplified cancer cells than more conventional ribosome-inactivating proteins, leading to an increased therapeutic window and decreased side effects.


Subject(s)
Antineoplastic Agents/pharmacology , Bacterial Toxins/pharmacology , Burkholderia , Animals , Cell Line, Tumor , Cell Survival/drug effects , Eukaryotic Initiation Factor-4F/metabolism , Fibroblasts/drug effects , Humans , Mice, Inbred C57BL , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/metabolism
14.
Protein Pept Lett ; 25(4): 368-378, 2018.
Article in English | MEDLINE | ID: mdl-29577851

ABSTRACT

BACKGROUND: Alpha-synuclein is a protein involved in the pathogenesis of Parkinson's disease. In vitro observations have shown that specific brain-enriched polyunsaturated fatty acids, such as arachidonic acid, can give rise to a conformational change in alpha-synuclein and ultimately induce its fibrillation. Arachidonic acid is released by phospholipase A2 activity and clinical observations have shown a link between mutations in PLA2G6, the gene responsible for the production of phospholipase A2, and early-onset types of parkinsonism. It is unknown how phospholipase A2-driven release of arachidonic acid can affect the conformation of alphasynuclein. OBJECTIVE: The main objective of this study was to investigate if phospholipase A2-induced release of arachidonic acid can induce changes in conformation and aggregation state of alpha-synuclein. METHODS: Recombinant human alpha-synuclein was expressed and isolated and incubated in the presence of phosphatidylcholine and phosphatidylserine (PC/PS) containing liposomes. The release of free fatty acids from PC/PS liposomes by bee venom phospholipase A2 was measured with the fluorescent probe acrylodated intestinal fatty acid-binding protein (ADIFAB) and radioactive labelling by preparing liposomes in the presence of L- 3-phosphatidylcholine, 1-stearyl-2[1-14C] arachidonoyl. The effect of free fatty acid release on the conformation of alpha-synuclein was assayed by far-UV circular dichroism and resistance against V8 protease-induced limited proteolysis. Aggregation of alpha-synuclein upon exposure to phospholipase A2-induced action on PC/PS liposomes was measured using thioflavin T fluorescence, SDS-PAGE, gel filtration chromatography, and transmission electron microscopy. RAW264.7 cells were transiently transfected with human alpha-synuclein and release of arachidonic acid was quantified using radiolabeling and liquid scintillation counting. RESULTS: Phospholipase A2 is capable of releasing arachidonic acid from biomimetic phospholipid membranes. Exposure of alpha-synuclein to phospholipase A2-induced release of arachidonic acid from PC/PS liposomes induces a conformational transition of the protein and leads to partial resistance against proteolytic cleavage by V8 protease. Prolonged incubation of alpha-synuclein with arachidonic acid, derived from PC/PS liposomes by phospholipase A2 leads to aggregate formation. In line with this, transiently transfected RAW264.7 cells with alpha-synuclein showed arachidonic acid release and punctate alpha-synuclein staining upon phospholipase A2 activation. The ability of arachidonic acid to drive alpha-synuclein to aggregate was independent of its oxidation state. CONCLUSION: We present data that suggest a biological context for the previously reported clinical observation that linked mutations in PLA2G6, the gene responsible for the production of phospholipase A2, and early-onset types of parkinsonism. Release of arachidonic acid, independent of its oxidation state, through activation of phospholipase A2-driven hydrolysis of phospholipid membranes, leads to the structural transition and aggregation of alpha-synuclein.


Subject(s)
Arachidonic Acid/metabolism , Phospholipases A2/metabolism , Protein Aggregates , alpha-Synuclein/metabolism , Animals , Arachidonic Acid/chemistry , Fatty Acid-Binding Proteins/chemistry , Fluorescent Dyes/chemistry , Humans , Liposomes , Mice , Oxidation-Reduction , Parkinson Disease/metabolism , Phosphatidylcholines/chemistry , Phospholipases A2/chemistry , Protein Conformation , RAW 264.7 Cells , Recombinant Proteins/chemistry , alpha-Synuclein/chemistry
15.
Front Pharmacol ; 8: 796, 2017.
Article in English | MEDLINE | ID: mdl-29170639

ABSTRACT

Botulinum neurotoxins (BoNTs) type A and type B are commonly used as biopharmaceutics for neurological diseases, uniquely allowing months-long paralysis of target muscles. Their exquisite neuronal specificity is conferred by a multistep process of binding, internalization, cytosolic escape and cleavage of the neuron-specific proteins, SNAP-25 and vesicle-associated membrane proteins (VAMPs), ultimately to inhibit secretion of neurotransmitters. Currently the mouse lethality bioassay is the only available method for quality control testing of VAMP-cleaving botulinum products. Refined assays for botulinum product testing are urgently needed. Specifically, in vitro replacement assays which can account for all steps of BoNT intoxication are in high demand. Here, we describe a novel SiMa cell-based approach where re-engineering of the VAMP molecule allows detection of all BoNT/B intoxication steps using a luminescent enzymatic reaction with sensitivity comparable to mouse LD50 bioassay. The presented one-step enzyme-linked immunosorbent assay meets 3Rs (replacement, reduction, and refinement of the use of animals) objectives, is user-friendly and will accelerate development of new botulinum drugs. The sensitive enzymatic reporter cell line could also be adapted for the detection of toxin activity during the manufacture of botulinum and tetanus vaccines.

16.
Toxins (Basel) ; 9(11)2017 10 27.
Article in English | MEDLINE | ID: mdl-29076988

ABSTRACT

Ribosome inactivating proteins (RIPs) form a class of toxins that was identified over a century ago. They continue to fascinate scientists and the public due to their very high activity and long-term stability which might find useful applications in the therapeutic killing of unwanted cells but can also be used in acts of terror. We will focus our review on the canonical plant-derived RIPs which display ribosomal RNA N-glycosidase activity and irreversibly inhibit protein synthesis by cleaving the 28S ribosomal RNA of the large 60S subunit of eukaryotic ribosomes. We will place particular emphasis on therapeutic applications and the generation of immunotoxins by coupling antibodies to RIPs in an attempt to target specific cells. Several generations of immunotoxins have been developed and we will review their optimisation as well as their use and limitations in pre-clinical and clinical trials. Finally, we endeavour to provide a perspective on potential future developments for the therapeutic use of immunotoxins.


Subject(s)
Immunotoxins , Plant Proteins , Ribosome Inactivating Proteins , Animals , Humans , Plants/metabolism , RNA, Ribosomal
17.
Sci Rep ; 7(1): 5958, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28729700

ABSTRACT

Neurotransmission and secretion of hormones involve a sequence of protein/lipid interactions with lipid turnover impacting on vesicle trafficking and ultimately fusion of secretory vesicles with the plasma membrane. We previously demonstrated that sphingosine, a sphingolipid metabolite, promotes formation of the SNARE complex required for membrane fusion and also increases the rate of exocytosis in isolated nerve terminals, neuromuscular junctions, neuroendocrine cells and in hippocampal neurons. Recently a fungi-derived sphingosine homologue, FTY720, has been approved for treatment of multiple sclerosis. In its non-phosphorylated form FTY720 accumulates in the central nervous system, reaching high levels which could affect neuronal function. Considering close structural similarity of sphingosine and FTY720 we investigated whether FTY720 has an effect on regulated exocytosis. Our data demonstrate that FTY720 can activate vesicular synaptobrevin for SNARE complex formation and enhance exocytosis in neuroendocrine cells and neurons.


Subject(s)
Fingolimod Hydrochloride/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Neurosecretory Systems/metabolism , R-SNARE Proteins/metabolism , Sphingosine/analogs & derivatives , Synaptic Vesicles/metabolism , Animals , Cytosol/drug effects , Cytosol/metabolism , Excitatory Postsynaptic Potentials/drug effects , Exocytosis/drug effects , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/pharmacology , Glutamic Acid/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Male , Multiple Sclerosis/physiopathology , Neurons/drug effects , Neurons/metabolism , Neurosecretory Systems/drug effects , Neurosecretory Systems/pathology , Neurosecretory Systems/physiopathology , Rats, Wistar , SNARE Proteins/metabolism , Synaptic Vesicles/drug effects , Synaptosomes/drug effects , Synaptosomes/metabolism
18.
Neuroscience ; 331: 148-57, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27339730

ABSTRACT

Sigma-1 receptor (S1R) is a unique pluripotent modulator of living systems and has been reported to be associated with a number of neurological diseases including pathological pain. Intrathecal administration of S1R antagonists attenuates the pain behavior of rodents in both inflammatory and neuropathic pain models. However, the S1R localization in the spinal cord shows a selective ventral horn motor neuron distribution, suggesting the high likelihood of S1R in the dorsal root ganglion (DRG) mediating the pain relief by intrathecally administered drugs. Since primary afferents are the major component in the pain pathway, we examined the mouse and rat DRGs for the presence of the S1R. At both mRNA and protein levels, quantitative RT-PCR (qRT-PCR) and Western confirmed that the DRG contains greater S1R expression in comparison to spinal cord, cortex, or lung but less than liver. Using a custom-made highly specific antibody, we demonstrated the presence of a strong S1R immuno-fluorescence in all rat and mouse DRG neurons co-localizing with the Neuron-Specific Enolase (NSE) marker, but not in neural processes or GFAP-positive glial satellite cells. In addition, S1R was absent in afferent terminals in the skin and in the dorsal horn of the spinal cord. Using immuno-electron microscopy, we showed that S1R is detected in the nuclear envelope and endoplasmic reticulum (ER) of DRG cells. In contrast to other cells, S1R is also located directly at the plasma membrane of the DRG neurons. The presence of S1R in the nuclear envelope of all DRG neurons suggests an exciting potential role of S1R as a regulator of neuronal nuclear activities and/or gene expression, which may provide insight toward new molecular targets for modulating nociception at the level of primary afferent neurons.


Subject(s)
Ganglia, Spinal/metabolism , Ganglia, Spinal/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Receptors, sigma/metabolism , Animals , Antibodies , Blotting, Western , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Immunohistochemistry , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Immunoelectron , Nuclear Envelope/metabolism , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, sigma/genetics , Receptors, sigma/immunology , Sigma-1 Receptor
19.
Oncotarget ; 7(22): 33220-8, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27121208

ABSTRACT

Neuroblastomas constitute a major cause of cancer-related deaths in young children. In recent years, a number of translation-inhibiting enzymes have been evaluated for killing neuroblastoma cells. Here we investigated the potential vulnerability of human neuroblastoma cells to protease activity derived from botulinum neurotoxin type C. We show that following retinoic acid treatment, human neuroblastoma cells, SiMa and SH-SY5Y, acquire a neuronal phenotype evidenced by axonal growth and expression of neuronal markers. Botulinum neurotoxin type C which cleaves neuron-specific SNAP25 and syntaxin1 caused apoptotic death only in differentiated neuroblastoma cells. Direct comparison of translation-inhibiting enzymes and the type C botulinum protease revealed one order higher cytotoxic potency of the latter suggesting a novel neuroblastoma-targeting pathway. Our mechanistic insights revealed that loss of ubiquitous SNAP23 due to differentiation coupled to SNAP25 cleavage due to botulinum activity may underlie the apoptotic death of human neuroblastoma cells.


Subject(s)
Apoptosis , Botulinum Toxins/biosynthesis , Cell Differentiation , Genetic Therapy/methods , Neuroblastoma/therapy , Botulinum Toxins/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Humans , Neuroblastoma/enzymology , Neuroblastoma/genetics , Neuroblastoma/pathology , Phenotype , Protein Synthesis Inhibitors/metabolism , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Signal Transduction , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Transduction, Genetic , Tretinoin/pharmacology
20.
Arch Biochem Biophys ; 593: 38-49, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26850364

ABSTRACT

Angiotensin II (Ang II) is an important mammalian neurohormone involved in reninangiotensin system. Ang II is produced both constitutively and locally by RAS systems, including white fat adipocytes. The influence of Ang II on adipocytes is complex, affecting different systems of signal transduction from early Са(2+) responses to cell proliferation and differentiation, triglyceride accumulation, expression of adipokine-encoding genes and adipokine secretion. It is known that white fat adipocytes express all RAS components and Ang II receptors (АТ1 and АТ2). The current work was carried out with the primary white adipocytes culture, and Са(2+) signaling pathways activated by Ang II were investigated using fluorescent microscopy. Са(2+)-oscillations and transient responses of differentiated adipocytes to Ang II were registered in cells with both small and multiple lipid inclusions. Using inhibitory analysis and selective antagonists, we now show that Ang II initiates periodic Са(2+)-oscillations and transient responses by activating АТ1 and АТ2 receptors and involving branched signaling cascades: 1) Ang II → Gq → PLC → IP3 → IP3Rs → Ca(2+) 2) Gßγ → PI3Kγ → PKB 3) PKB → eNOS → NO → PKG 4) CD38 → cADPR → RyRs → Ca(2+) In these cascades, AT1 receptors play the leading role. The results of the present work open a perspective of using Ang II for correction of signal resistance of adipocytes often observed during obesity and type 2 diabetes.


Subject(s)
Adipocytes, White/metabolism , Angiotensin II/metabolism , Calcium Signaling , Adipocytes, White/cytology , Adipocytes, White/drug effects , Angiotensin II/pharmacology , Animals , Calcium/metabolism , Cell Differentiation , Lipid Droplets/ultrastructure , Mice , Primary Cell Culture , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...