Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.304
Filter
2.
Cell Chem Biol ; 31(7): 1233-1235, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39029451

ABSTRACT

In this issue of Cell Chemical Biology, Elleman et al.1 introduce a transformative chemical approach to control neuronal activity with high spatial and temporal resolution. The authors present STX-bpc, a potent neurotoxin that naturally inhibits voltage-gated sodium channels (NaVs), complementing available optogenetic methods for manipulating neuronal activity, cellular communication, and behavior.


Subject(s)
Neurons , Neurons/drug effects , Neurons/metabolism , Neurons/cytology , Animals , Humans , Optogenetics , Voltage-Gated Sodium Channels/metabolism , Voltage-Gated Sodium Channels/chemistry , Neurotoxins/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/chemistry
3.
Expert Rev Neurother ; 24(8): 787-797, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39049547

ABSTRACT

INTRODUCTION: Cervical dystonia (CD) causes involuntary movements and postures of the head, neck, and shoulders, as well as nonmotor symptoms including pain, mood, and sleep dysfunction, and impacts quality of life. The first-line treatment for CD is botulinum neurotoxin (BoNT) injections. AREAS COVERED: The clinical presentation and diagnosis of CD, as well as where BoNT resides in the treatment landscape, is reviewed first. Next, the mechanism of action and the pharmacological differences in the available preparations of BoNT products are explained. The evidence base for motor and nonmotor efficacy and safety of the available BoNT formulations is reviewed, with attention to duration of benefit as a driver of patient satisfaction. Practical determinants of BoNT efficacy are reviewed including muscle selection, accurate muscle injection, factors related to poor or deteriorating response, and immunogenicity. EXPERT OPINION: BoNT represents a significant advancement in the treatment of CD. More accurate diagnosis, muscle selection and targeting, and dosing can improve outcomes with existing BoNT formulations. Further refinement of BoNT potency, duration of action, safety, and immunogenicity will help reduce unmet needs in the magnitude and duration of benefit. Additional validation of DBS and MRI-guided focused ultrasound may expand options for patients with toxin nonresponse.


Subject(s)
Botulinum Toxins , Neurotoxins , Torticollis , Humans , Torticollis/drug therapy , Botulinum Toxins/therapeutic use , Botulinum Toxins/administration & dosage , Neurotoxins/therapeutic use
4.
Ecotoxicol Environ Saf ; 281: 116678, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964067

ABSTRACT

The non-protein amino acid ß-N-methylamino-L-alanine (BMAA), produced by cyanobacteria, has been recognized as a neurotoxin. L-serine as an antagonist of BMAA can effectively alleviate BMAA-induced neurotoxicity. Although BMAA has long been emphasized as a neurotoxin, with the emergence of BMAA detected in a variety of algae in freshwater around the world and its clear biological enrichment effect, it is particularly important to study the non-neurotoxic adverse effects of BMAA. However, there is only limited evidence to support the ability of BMAA to cause oxidative damage in the liver. The exact molecular mechanism of BMAA-induced liver injury is still unclear. The formation of neutrophil extracellular traps (NETs) is a 'double-edged sword' for the organism, excessive formation of NETs is associated with inflammatory diseases of the liver. Our results innovatively confirmed that BMAA was able to cause the formation of NETs in the liver during the liver injury. The possible mechanism may associated with the regulation of ERK/p38 and cGAS/STING signaling pathways. The massive formation of NETs was able to exacerbate the BMAA-induced oxidative stress and release of inflammatory factors in the mice liver. And the removal of NETs could alleviate this injury. This article will bring a new laboratory evidence for BMAA-induced non-neurotoxicity and immunotoxicity.


Subject(s)
Amino Acids, Diamino , Chemical and Drug Induced Liver Injury , Cyanobacteria Toxins , Extracellular Traps , Oxidative Stress , Animals , Amino Acids, Diamino/toxicity , Extracellular Traps/drug effects , Mice , Oxidative Stress/drug effects , Male , Neutrophils/drug effects , Liver/drug effects , Neurotoxins/toxicity , Signal Transduction/drug effects
5.
Genome Biol Evol ; 16(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39018436

ABSTRACT

Sea anemones are venomous animals that rely on their venom for prey capture, defense against predators, and intraspecific competition. Currently, comprehensive molecular and evolutionary analyses of the toxin repertoire for sea anemones are limited by a lack of proteomic data for most species. In this study, proteo-transcriptomic analysis was used to expand our knowledge of the proteinaceous components of sea anemone venom by determining the secreted venom proteome of Calliactis polypus. Electromechanical stimulation was used to obtain the secreted venom of C. polypus. We identified a low complexity proteome that was dominated by toxins with similarity to known neurotoxins, as well as six novel toxin candidates. The novel putative toxin candidates were found to be taxonomically restricted to species from the superfamily Metridioidea. Furthermore, the secreted venom of C. polypus had only three putative toxins in common with the venom of acontia from the same species and little similarity with the secreted venom of closely related species. Overall, this demonstrates that regionalized and lineage-specific variability in toxin abundance is common among sea anemone species. Moreover, the limited complexity of the toxin repertoire found in C. polypus supports the idea that peptide neurotoxins make up the dominant toxin arsenal found in the venom of sea anemones.


Subject(s)
Proteome , Sea Anemones , Animals , Sea Anemones/genetics , Cnidarian Venoms/genetics , Cnidarian Venoms/chemistry , Cnidarian Venoms/metabolism , Neurotoxins/genetics , Phylogeny , Proteomics
7.
Nature ; 631(8019): 142-149, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926573

ABSTRACT

Interindividual genetic variation affects the susceptibility to and progression of many diseases1,2. However, efforts to study how individual human brains differ in normal development and disease phenotypes are limited by the paucity of faithful cellular human models, and the difficulty of scaling current systems to represent multiple people. Here we present human brain Chimeroids, a highly reproducible, multidonor human brain cortical organoid model generated by the co-development of cells from a panel of individual donors in a single organoid. By reaggregating cells from multiple single-donor organoids at the neural stem cell or neural progenitor cell stage, we generate Chimeroids in which each donor produces all cell lineages of the cerebral cortex, even when using pluripotent stem cell lines with notable growth biases. We used Chimeroids to investigate interindividual variation in the susceptibility to neurotoxic triggers that exhibit high clinical phenotypic variability: ethanol and the antiepileptic drug valproic acid. Individual donors varied in both the penetrance of the effect on target cell types, and the molecular phenotype within each affected cell type. Our results suggest that human genetic background may be an important mediator of neurotoxin susceptibility and introduce Chimeroids as a scalable system for high-throughput investigation of interindividual variation in processes of brain development and disease.


Subject(s)
Cerebral Cortex , Chimera , Genetic Predisposition to Disease , Neurotoxins , Organoids , Female , Humans , Male , Cell Lineage/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Chimera/genetics , Ethanol/adverse effects , Ethanol/toxicity , Genetic Variation , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurotoxins/toxicity , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Tissue Donors , Valproic Acid/adverse effects , Valproic Acid/toxicity , Genetic Predisposition to Disease/genetics
8.
Toxicon ; 247: 107833, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38942241

ABSTRACT

Presynaptic- or ß-neurotoxicity of secreted phospholipases A2 (sPLA2) is a complex process. For full expression of ß-neurotoxicity, the enzymatic activity of the toxin is essential. However, it has been shown that not all toxic effects of a ß-neurotoxin depend on its enzymatic activity, for example, the inhibition of mitochondrial cytochrome c oxidase. The main objective of this study was to verify whether it is possible to observe and study the phospholipase-independent actions of ß-neurotoxins by a standard ex vivo twitch-tension experimental approach. To this end, we compared the effects of a potent snake venom ß-neurotoxin, ammodytoxin A (AtxA), and its enzymatically inactive mutant AtxA(D49S) on muscle contraction of the mouse phrenic nerve-hemidiaphragm preparation. While AtxA significantly affected the amplitude of the indirectly evoked isometric muscle contraction, the resting tension of the neuromuscular (NM) preparation, the amplitude of the end-plate potential (EPP), the EPP half decay time and the resting membrane potential, AtxA(D49S) without enzymatic activity did not. From this, we can conclude that the effects of AtxA independent of enzymatic activity cannot be studied with classical electrophysiological measurements on the isolated NM preparation. Our results also suggest that the inhibition of cytochrome c oxidase activity by AtxA is not involved in the rapid NM blockade by this ß-neurotoxin, but that its pathological consequences are rather long-term. Interestingly, in our experimental setup, AtxA upon direct stimulation reduced the amplitude of muscle contraction and induced contracture of the hemidiaphragm, effects that could be interpreted as myotoxic.


Subject(s)
Viper Venoms , Animals , Mice , Viper Venoms/toxicity , Neurotoxins/toxicity , Muscle Contraction/drug effects , Diaphragm/drug effects , Phrenic Nerve/drug effects , Neuromuscular Junction/drug effects , Male , Electrophysiological Phenomena
9.
Front Cell Infect Microbiol ; 14: 1394713, 2024.
Article in English | MEDLINE | ID: mdl-38836054

ABSTRACT

The rabies virus enters the nervous system by interacting with several molecular targets on host cells to modify behavior and trigger receptor-mediated endocytosis of the virion by poorly understood mechanisms. The rabies virus glycoprotein (RVG) interacts with the muscle acetylcholine receptor and the neuronal α4ß2 subtype of the nicotinic acetylcholine receptor (nAChR) family by the putative neurotoxin-like motif. Given that the neurotoxin-like motif is highly homologous to the α7 nAChR subtype selective snake toxin α-bungarotoxin (αBTX), other nAChR subtypes are likely involved. The purpose of this study is to determine the activity of the RVG neurotoxin-like motif on nAChR subtypes that are expressed in brain regions involved in rabid animal behavior. nAChRs were expressed in Xenopus laevis oocytes, and two-electrode voltage clamp electrophysiology was used to collect concentration-response data to measure the functional effects. The RVG peptide preferentially and completely inhibits α7 nAChR ACh-induced currents by a competitive antagonist mechanism. Tested heteromeric nAChRs are also inhibited, but to a lesser extent than the α7 subtype. Residues of the RVG peptide with high sequence homology to αBTX and other neurotoxins were substituted with alanine. Altered RVG neurotoxin-like peptides showed that residues phenylalanine 192, arginine 196, and arginine 199 are important determinants of RVG peptide apparent potency on α7 nAChRs, while serine 195 is not. The evaluation of the rabies ectodomain reaffirmed the observations made with the RVG peptide, illustrating a significant inhibitory impact on α7 nAChR with potency in the nanomolar range. In a mammalian cell culture model of neurons, we confirm that the RVG peptide binds preferentially to cells expressing the α7 nAChR. Defining the activity of the RVG peptide on nAChRs expands our understanding of basic mechanisms in host-pathogen interactions that result in neurological disorders.


Subject(s)
Glycoproteins , Rabies virus , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Rabies virus/physiology , Rabies virus/metabolism , Humans , Glycoproteins/metabolism , Glycoproteins/genetics , Oocytes/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Host-Pathogen Interactions , Protein Binding , Rabies/metabolism , Rabies/virology , Acetylcholine/metabolism , Acetylcholine/pharmacology , Neurotoxins/metabolism , Neurotoxins/pharmacology
10.
Int J Biol Macromol ; 275(Pt 1): 133461, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38945343

ABSTRACT

Small single-chain variable fragments (scFv) are promising biomolecules to inhibit and neutralize toxins and to act as antivenoms. In this work, we aimed to produce a functional scFv-6009FV in the yeast Pichia pastoris, which inhibits the pure Cn2 neurotoxin and the whole venom of Centruroides noxius. We were able to achieve yields of up to 31.6 ± 2 mg/L in flasks. Furthermore, the protein showed a structure of 6.1 % α-helix, 49.1 % ß-sheet, and 44.8 % of random coil by CD. Mass spectrometry confirmed the amino acid sequence and showed no glycosylation profile for this molecule. Purified scFv-6009FV allowed us to develop anti-scFvs in rabbits, which were then used in affinity columns to purify other scFvs. Determination of its half-maximal inhibitory concentration value (IC50) was 40 % better than the scFvs produced by E. coli as a control. Finally, we found that scFv-6009FV was able to inhibit ex vivo the pure Cn2 toxin and the whole venom from C. noxius in murine rescue experiments. These results demonstrated that under the conditions assayed here, P. pastoris is suited to produce scFv-6009FV that, compared to scFvs produced by E. coli, maintains the characteristics of an antibody and neutralizes the Cn2 toxin more effectively.


Subject(s)
Single-Chain Antibodies , Animals , Mice , Rabbits , Amino Acid Sequence , Animals, Poisonous , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/pharmacology , Gene Expression , Neurotoxins/antagonists & inhibitors , Neurotoxins/chemistry , Neurotoxins/genetics , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/genetics , Saccharomycetales/genetics , Saccharomycetales/metabolism , Scorpion Venoms/antagonists & inhibitors , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Scorpions , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/isolation & purification , Single-Chain Antibodies/pharmacology
11.
Toxins (Basel) ; 16(6)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38922160

ABSTRACT

The growing use of botulinum neurotoxins (BoNTs) for medical and aesthetic purposes has led to the development and marketing of an increasing number of BoNT products. Given that BoNTs are biological medications, their characteristics are heavily influenced by their manufacturing methods, leading to unique products with distinct clinical characteristics. The manufacturing and formulation processes for each BoNT are proprietary, including the potency determination of reference standards and other features of the assays used to measure unit potency. As a result of these differences, units of BoNT products are not interchangeable or convertible using dose ratios. The intrinsic, product-level differences among BoNTs are compounded by differences in the injected tissues, which are innervated by different nerve fiber types (e.g., motor, sensory, and/or autonomic nerves) and require unique dosing and injection sites that are particularly evident when treating complex therapeutic and aesthetic conditions. It is also difficult to compare across studies due to inherent differences in patient populations and trial methods, necessitating attention to study details underlying each outcome reported. Ultimately, each BoNT possesses a unique clinical profile for which unit doses and injection paradigms must be determined individually for each indication. This practice will help minimize unexpected adverse events and maximize efficacy, duration, and patient satisfaction. With this approach, BoNT is poised to continue as a unique tool for achieving individual goals for an increasing number of medical and aesthetic indications.


Subject(s)
Botulinum Toxins , Humans , Botulinum Toxins/therapeutic use , Botulinum Toxins/administration & dosage , Animals , Neurotoxins
12.
Toxins (Basel) ; 16(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38922155

ABSTRACT

Botulinum neurotoxins (BoNTs) have been used for almost half a century in the treatment of excessive muscle contractility. BoNTs are routinely used to treat movement disorders such as cervical dystonia, spastic conditions, blepharospasm, and hyperhidrosis, as well as for cosmetic purposes. In addition to the conventional indications, the use of BoNTs to reduce pain has gained increased recognition, giving rise to an increasing number of indications in disorders associated with chronic pain. Furthermore, BoNT-derived formulations are benefiting a much wider range of patients suffering from overactive bladder, erectile dysfunction, arthropathy, neuropathic pain, and cancer. BoNTs are categorised into seven toxinotypes, two of which are in clinical use, and each toxinotype is divided into multiple subtypes. With the development of bioinformatic tools, new BoNT-like toxins have been identified in non-Clostridial organisms. In addition to the expanding indications of existing formulations, the rich variety of toxinotypes or subtypes in the wild-type BoNTs associated with new BoNT-like toxins expand the BoNT superfamily, forming the basis on which to develop new BoNT-based therapeutics as well as research tools. An overview of the diversity of the BoNT family along with their conventional therapeutic uses is presented in this review followed by the engineering and formulation opportunities opening avenues in therapy.


Subject(s)
Botulinum Toxins , Humans , Botulinum Toxins/therapeutic use , Animals , Neurotoxins/therapeutic use , Neurotoxins/chemistry
13.
Toxins (Basel) ; 16(6)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38922166

ABSTRACT

The larvae of some lampyrid beetles are highly specialized predators of snails. They have been observed to climb on the shells of their prey and use this exposed position to bite and inject secretions potentially originating from the midgut. Besides serving the purpose of extra-oral digestion (EOD), injected compounds also seem to have a paralyzing effect. Up to now, the toxins causing this paralyzing activity have not been identified. In the current study, we provide a first compositional analysis of the midgut secretion from lampyrid larvae, with a focus on identifying putative neurotoxins causing the observed paralyzing effect. For this purpose, we utilized a combined proteo-transcriptomic approach to characterize the compounds present in the midgut secretion of larval stages of Lampyris noctiluca. In terms of the absolute numbers of identified compounds, the midgut secretion is dominated by hydrolyzing enzymes comprising peptidases, carboxylesterases, and glycosidases. However, when considering expression levels, a few rather short cysteine-rich peptides exceed all other compounds. Some of these compounds show moderate similarity to putative neurotoxins identified in the venom of other arthropods and could be responsible for paralyzing effects. In addition to these potential toxins, we provide a list of peptides typical of the midgut secretion of L. noctiluca, supplemented by the corresponding precursor sequences.


Subject(s)
Coleoptera , Larva , Neurotoxins , Snails , Animals , Larva/metabolism , Neurotoxins/toxicity , Neurotoxins/metabolism , Coleoptera/metabolism , Snails/metabolism , Predatory Behavior
14.
Environ Pollut ; 353: 124166, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38754694

ABSTRACT

Potentially toxic cyanobacterial blooms (cyanoHABs) have become a problem in public water supply reservoirs. Temperature rise caused by climate change can increase the frequency and intensity of blooms, which may influence the cyanotoxins concentration in the environment. This study aimed to evaluate the effect of the temperature on the responses of a Neotropical catfish exposed to a neurotoxin-rich cyanobacterial crude extract (Raphidiopsis raciborskii T3). Juveniles of Rhamdia quelen were exposed to four treatments, based on study data: control at 25 °C (C25), control at 30 °C (C30), crude extract equivalent to 105 cells.mL-l of R. raciborskii at 25 °C (CE25) and 30 °C (CE30). After 96 h of exposure, the fish were anesthetized and blood was taken. After euthanasia, the gill, posterior kidney, brain, muscle, liver and gonad were sampled for hematological, biochemical, genotoxic and histopathological biomarker analysis. Liver was sampled for proteomic analysis for identification of proteins related to energy production. Water samples were collected at the beginning and the end of the experiment for neurotoxins quantification. Different parameters in both males and females were altered at CE25, evidencing the effects of neurotoxins in freshwater fish. At CE30, a water warming scenario, more effects were observed in females than at 25 °C, such as activation of saxitoxin metabolism pathway and genotoxicity. More damage to macromolecules was observed in females at the higher temperature, demonstrating that the increase in temperature can aggravate the toxicity of neurotoxins produced by R. raciborskii T3.


Subject(s)
Catfishes , Cyanobacteria , Animals , Catfishes/physiology , Temperature , Microcystins/toxicity , Female , Male , Cyanobacteria Toxins , Climate Change , Neurotoxins/toxicity , Bacterial Toxins/toxicity , Marine Toxins/toxicity
15.
Luminescence ; 39(5): e4761, 2024 May.
Article in English | MEDLINE | ID: mdl-38807512

ABSTRACT

The fear of an increase in blood sugar can be very traumatic. Being diabetic either type I or type II leads to a disorder called diabetes distress having traits of stress, depression, and anxiety. Among risk factors of diabetes mellitus heavy and trace metal toxicity emerges as new risk factors reported in many studies. In this study we target toxic metals, viz., Ni2+, Zn2+, and Cu2+, involved in the pathogenesis of diabetes and diabetic stress with naphthazarin esters. The compounds C1-C3 isolated from the leaves and roots of Arnebia guttata were tested for their metal-binding ability in an aqueous medium in UV-Visible and nuclear magnetic resonance (NMR) studies. These probes are well-known naphthoquinones present in the Arnebia species. In the UV-Visible titrations of compounds C1-C3 with Na2+, K2+, Zn2+, Ca2+, Cu2+, Mg2+, Co2+, and Ni2+ ions, significant binding was observed with Ni2+, Cu2+, and Zn2+ ions in MeOH/H2O. There occurs a beautiful formation of red-shifted bands between the 520 to 620 nm range with a synergistic increase in absorbance. Also, the disappearance of proton peaks in the 1H NMR spectrum on addition of metal ions confirmed binding. Compounds C1-C3 isolated from A. guttata came out as potent Ni2+, Zn2+, and Cu2+ sensors that are reportedly involved in islet function and induction of diabetes.


Subject(s)
Esters , Naphthoquinones , Esters/chemistry , Naphthoquinones/chemistry , Diabetes Mellitus/metabolism , Neurotoxins/chemistry , Neurotoxins/metabolism , Water/chemistry , Molecular Structure , Plant Leaves/chemistry
16.
J Ethnopharmacol ; 332: 118338, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38759762

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The Mesobuthus martensii scorpions, called as "Quanxie", are known Chinese medicinal material base on the "Combat poison with poison" strategy for more than one thousand years, and still widely used to treat various diseases according to the Pharmacopoeia of the People's Republic of China nowadays. AIM OF STUDY: The study aims to investigate the similarity of scorpion neurotoxins at the protein level between the juvenile and adult Mesobuthus martensii scorpions as Chinese medicine materials. MATERIALS AND METHODS: The second-, third- and fourth-instar, and adult Mesobuthus martensii scorpions were collected for the characterization of neurotoxin expression through multiple strategic proteomics, including undigested scorpion venom, endopeptidase-digested, and undigested scorpion telson extract for the sample analysis. RESULTS: Based on the known 107 scorpion neurotoxins from the genomic and transcriptomic analysis of adult Mesobuthus martensii scorpions, the multiple strategic proteomics first revealed that neurotoxins exhibited more stability in telson extract than secreted venom. In the reported transcripts of scorpion neurotoxins, approximately 53%, 56%, 66% and 78% of neurotoxins were detected through undigested scorpion venom, the endopeptidase Arg-C-, Lys-C-digested telson extract, and undigested telson extract strategies, respectively. Nearly 79% of scorpion neurotoxins detected in third-instar Mesobuthus martensii scorpions represent the largest number of scorpion neurotoxins from proteomic analysis to date. Moreover, a total of 84% of scorpion neurotoxins were successfully identified at the protein level, and similar neurotoxin expression profiles in second-, third- and fourth-instar, and adult Mesobuthus martensii scorpions were first revealed by the multiple strategic proteomics. CONCLUSION: These findings for the first time demonstrate the similar neurotoxin expression profiles between the juvenile and adult Mesobuthus martensii scorpions as Chinese medicinal material, which would serve as a paradigm for further toxin analysis from different venomous animals.


Subject(s)
Medicine, Chinese Traditional , Neurotoxins , Proteomics , Scorpion Venoms , Scorpions , Animals , Proteomics/methods , Animals, Poisonous
17.
Microb Biotechnol ; 17(4): e14468, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38635158

ABSTRACT

The sweet potato whitefly Bemisia tabaci (Hemiptera: Aleyrodidae) is responsible for significant crop losses and presents one of the greatest challenges for global agricultural pest management. Management of whitefly populations and associated plant viral diseases is hindered by widespread whitefly resistance to chemical insecticides. An alternative control approach involves the use of insect-specific neurotoxins, but these require delivery from the whitefly gut into the haemocoel. Here we demonstrate that the coat protein (CP) of a begomovirus, Tomato yellow leaf curl virus, is sufficient for delivery of fused proteins into the whitefly haemocoel without virion assembly. Following feeding on the recombinant CP-P-mCherry fusion (where -P- is a proline-rich linker), mCherry fluorescence was detected in the dorsal aorta and pericardial cells of the whitefly, but not in those of whitefly fed on negative control treatments, indicating effective CP-mediated delivery of mCherry into the whitefly haemocoel. Significant mortality was observed in whiteflies fed on a fusion of CP-P to the insect-specific neurotoxin Hv1a, but not in whiteflies fed on CP-P fused to a disarmed Hv1a mutant. Begomovirus coat protein - insect neurotoxin fusions hold considerable potential for transgenic resistance to whitefly providing valuable tools for whitefly management.


Subject(s)
Hemiptera , Plant Viruses , Animals , Neurotoxins , Agriculture , Fluorescence
18.
Sci Rep ; 14(1): 8017, 2024 04 05.
Article in English | MEDLINE | ID: mdl-38580836

ABSTRACT

Cyanobacteria produce neurotoxic non-protein amino acids (NPAAs) that accumulate in ecosystems and food webs. American lobsters (Homarus americanus H. Milne-Edwards) are one of the most valuable seafood industries in Canada with exports valued at > $2 billion. Two previous studies have assessed the occurrence of ß-N-methylamino-L-alanine (BMAA) in a small number of lobster tissues but a complete study has not previously been undertaken. We measured NPAAs in eyeballs, brain, legs, claws, tails, and eggs of 4 lobsters per year for the 2021 and 2022 harvests. Our study included 4 male and 4 female lobsters. We detected BMAA and its isomers, N-(2-aminoethyl)glycine (AEG), 2,4-diaminobutyric acid (DAB) and ß-aminomethyl-L-alanine (BAMA) by a fully validated reverse phase chromatography-tandem mass spectrometry method. We quantified BMAA, DAB, AEG and BAMA in all of the lobster tissues. Our quantification data varied by individual lobster, sex and collection year. Significantly more BMAA was quantified in lobsters harvested in 2021 than 2022. Interestingly, more BAMA was quantified in lobsters harvested in 2022 than 2021. Monitoring of lobster harvests for cyanobacterial neurotoxins when harmful algal bloom events occur could mitigate risks to human health.


Subject(s)
Amino Acids, Diamino , Decapoda , Neurotoxicity Syndromes , Animals , Male , Female , Humans , Nephropidae/metabolism , Ecosystem , Neurotoxins/toxicity , Amino Acids, Diamino/metabolism , Seafood/analysis , Decapoda/metabolism , beta-Alanine
19.
Pestic Biochem Physiol ; 201: 105853, 2024 May.
Article in English | MEDLINE | ID: mdl-38685212

ABSTRACT

Ion channels on cell membrane are molecular targets of more than half peptide neurotoxins from spiders. From Pardosa pseudoannulata, a predatory spider on a range of insect pests, we characterized a peptide neurotoxin PPTX-04 with an insecticidal activity. PPTX-04 showed high toxicity to Nilaparvata lugens, a main prey of P. pseudoannulata, and the toxicity was not affected by the resistance to etofenprox (IUPAC chemical name:1-ethoxy-4-[2-methyl-1-[(3-phenoxyphenyl)methoxy]propan-2-yl]benzene, purity: 99%). On N. lugens voltage-gated sodium channel NlNav1 expressed in Xenopus oocytes, PPTX-04 prolonged the channel opening and induced tail currents, which is similar to pyrethroid insecticides. However, PPTX-04 potency on NlNav1 was not affected by mutations conferring pyrethroid resistance in insects, which revealed that PPTX-04 and pyrethroids should act on different receptors in NlNav1. In contrast, two mutations at the extracellular site 4 significantly reduced PPTX-04 potency, which indicated that PPTX-04 would act on a potential receptor containing the site 4 in NlNav1. The result from the molecular docking supported the conclusion that the binding pocket of PPTX-04 in NlNav1 should contain the site 4. In summary, PPTX-04 had high insecticidal activity through acting on a distinct receptor site in insect Nav, and was a potential resource to control insect pests and manage resistance to pyrethroids.


Subject(s)
Insecticides , Neurotoxins , Spider Venoms , Spiders , Voltage-Gated Sodium Channels , Animals , Insecticides/pharmacology , Insecticides/chemistry , Spider Venoms/chemistry , Spider Venoms/pharmacology , Spider Venoms/genetics , Voltage-Gated Sodium Channels/metabolism , Voltage-Gated Sodium Channels/genetics , Neurotoxins/pharmacology , Neurotoxins/toxicity , Pyrethrins/pharmacology , Hemiptera/drug effects , Oocytes/drug effects , Xenopus laevis , Insect Proteins/genetics , Insect Proteins/metabolism , Insect Proteins/chemistry
20.
Mar Pollut Bull ; 202: 116299, 2024 May.
Article in English | MEDLINE | ID: mdl-38581736

ABSTRACT

The neurotoxin ß-N-methylamino-L-alanine (BMAA) has emerged as an environmental factor related to neurodegenerative diseases. BMAA is produced by various microorganisms including cyanobacteria and diatoms, in diverse ecosystems. In the diatom Phaeodactylum tricornutum, BMAA is known to inhibit growth. The present study investigated the impact of BMAA on the diatom Thalassiosira pseudonana by exposing it to different concentrations of exogenous BMAA. Metabolomics was predominantly employed to investigate the effect of BMAA on T. pseudonana, and MetaboAnalyst (https://www.metabo-analyst.ca/) was used to identify BMAA-associated metabolisms/pathways in T. pseudonana. Furthermore, to explore the unique response, specific metabolites were compared between treatments. When the growth was obstructed by BMAA, 17 metabolisms/pathways including nitrogen and glutathione (i.e. oxidative stress) metabolisms, were influenced in T. pseudonana. This study has further determined that 11 out of 17 metabolisms/pathways could be essentially affected by BMAA, leading to the inhibition of diatom growth.


Subject(s)
Amino Acids, Diamino , Cyanobacteria Toxins , Diatoms , Metabolomics , Neurotoxins , Diatoms/drug effects , Neurotoxins/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL