Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.873
Filtrar
Más filtros

Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(40): e2319177121, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39298472

RESUMEN

In 2015, the largest recorded harmful algal bloom (HAB) occurred in the Northeast Pacific, causing nearly 100 million dollars in damages to fisheries and killing many protected marine mammals. Dominated by the toxic diatom Pseudo-nitzschia australis, this bloom produced high levels of the neurotoxin domoic acid (DA). Through molecular and transcriptional characterization of 52 near-weekly phytoplankton net-tow samples collected at a bloom hotspot in Monterey Bay, California, we identified active transcription of known DA biosynthesis (dab) genes from the three identified toxigenic species, including P. australis as the primary origin of toxicity. Elevated expression of silicon transporters (sit1) during the bloom supports the previously hypothesized role of dissolved silica (Si) exhaustion in contributing to bloom physiology and toxicity. We find that coexpression of the dabA and sit1 genes serves as a robust predictor of DA one week in advance, potentially enabling the forecasting of DA-producing HABs. We additionally present evidence that low levels of iron could have colimited the diatom population along with low Si. Iron limitation represents an overlooked driver of both toxin production and ecological success of the low-iron-adapted Pseudo-nitzschia genus during the 2015 bloom, and increasing pervasiveness of iron limitation may fuel the escalating magnitude and frequency of toxic Pseudo-nitzschia blooms globally. Our results advance understanding of bloom physiology underlying toxin production, bloom prediction, and the impact of global change on toxic blooms.


Asunto(s)
Diatomeas , Floraciones de Algas Nocivas , Ácido Kaínico , Fitoplancton , Ácido Kaínico/análogos & derivados , Ácido Kaínico/metabolismo , Diatomeas/genética , Diatomeas/metabolismo , Diatomeas/crecimiento & desarrollo , Fitoplancton/genética , Fitoplancton/metabolismo , California , Toxinas Marinas/biosíntesis , Toxinas Marinas/genética , Toxinas Marinas/metabolismo , Neurotoxinas/genética , Neurotoxinas/toxicidad , Neurotoxinas/metabolismo , Hierro/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(44): e2208183119, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36279440

RESUMEN

The origin of methylmercury in pelagic fish remains unclear, with many unanswered questions regarding the production and degradation of this neurotoxin in the water column. We used mercury (Hg) stable isotope ratios of marine particles and biota to elucidate the cycling of methylmercury prior to incorporation into the marine food web. The Hg isotopic composition of particles, zooplankton, and fish reveals preferential methylation of Hg within small (< 53 µm) marine particles in the upper 400 m of the North Pacific Ocean. Mass-dependent Hg isotope ratios (δ202Hg) recorded in small particles overlap with previously estimated δ202Hg values for methylmercury sources to Pacific and Atlantic Ocean food webs. Particulate compound specific isotope analysis of amino acids (CSIA-AA) yield δ15N values that indicate more-significant microbial decomposition in small particles compared to larger particles. CSIA-AA and Hg isotope data also suggest that large particles (> 53 µm) collected in the equatorial ocean are distinct from small particles and resemble fecal pellets. Additional evidence for Hg methylation within small particles is provided by a statistical mixing model of even mass-independent (Δ200Hg and Δ204Hg) isotope values, which demonstrates that Hg within near-surface marine organisms (0-150 m) originates from a combination of rainfall and marine particles. In contrast, in meso- and upper bathypelagic organisms (200-1,400 m), the majority of Hg originates from marine particles with little input from wet deposition. The occurrence of methylation within marine particles is supported further by a correlation between Δ200Hg and Δ199Hg values, demonstrating greater overlap in the Hg isotopic composition of marine organisms with marine particles than with total gaseous Hg or wet deposition.


Asunto(s)
Mercurio , Compuestos de Metilmercurio , Contaminantes Químicos del Agua , Animales , Isótopos de Mercurio/análisis , Mercurio/análisis , Organismos Acuáticos/metabolismo , Neurotoxinas/metabolismo , Monitoreo del Ambiente , Contaminantes Químicos del Agua/análisis , Peces/metabolismo , Isótopos/metabolismo , Agua/metabolismo , Aminoácidos/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35110408

RESUMEN

Domoic acid (DA), the causative agent of amnesic shellfish poisoning, is produced by select organisms within two distantly related algal clades: planktonic diatoms and red macroalgae. The biosynthetic pathway to isodomoic acid A was recently solved in the harmful algal bloom-forming diatom Pseudonitzschia multiseries, establishing the genetic basis for the global production of this potent neurotoxin. Herein, we sequenced the 507-Mb genome of Chondria armata, the red macroalgal seaweed from which DA was first isolated in the 1950s, identifying several copies of the red algal DA (rad) biosynthetic gene cluster. The rad genes are organized similarly to the diatom DA biosynthesis cluster in terms of gene synteny, including a cytochrome P450 (CYP450) enzyme critical to DA production that is notably absent in red algae that produce the simpler kainoid neurochemical, kainic acid. The biochemical characterization of the N-prenyltransferase (RadA) and kainoid synthase (RadC) enzymes support a slightly altered DA biosynthetic model in C. armata via the congener isodomoic acid B, with RadC behaving more like the homologous diatom enzyme despite higher amino acid similarity to red algal kainic acid synthesis enzymes. A phylogenetic analysis of the rad genes suggests unique origins for the red macroalgal and diatom genes in their respective hosts, with native eukaryotic CYP450 neofunctionalization combining with the horizontal gene transfer of N-prenyltransferases and kainoid synthases to establish DA production within the algal lineages.


Asunto(s)
Dimetilaliltranstransferasa/genética , Dimetilaliltranstransferasa/metabolismo , Ácido Kaínico/análogos & derivados , Neurotoxinas/metabolismo , Rhodophyta/metabolismo , Evolución Biológica , Vías Biosintéticas/genética , Diatomeas/genética , Diatomeas/metabolismo , Floraciones de Algas Nocivas/fisiología , Ácido Kaínico/metabolismo , Familia de Multigenes/genética , Neurotoxinas/genética , Filogenia , Intoxicación por Mariscos/metabolismo
4.
Exp Cell Res ; 425(1): 113536, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36858342

RESUMEN

The neurotoxin MPP+ triggers cell death of dopamine neurons and induces Parkinson's disease symptoms in mice and men, but the immediate transcriptional response to this neurotoxin has not been studied. We therefore treated human SH-SY5Y cells with a low dose (0.1 mM) of MPP+ and measured the effect on nascent transcription by precision run-on sequencing (PRO-seq). We found that transcription of the mitochondrial genome was significantly reduced already after 30 min, whereas nuclear gene transcription was unaffected. Inhibition of respiratory complex I by MPP+ led to reduced ATP production, that may explain the diminished activity of mitochondrial RNA polymerase. Our results show that MPP+ has a direct effect on mitochondrial function and transcription, and that other gene expression or epigenetic changes induced by this neurotoxin are secondary effects that reflect a cellular adaptation program.


Asunto(s)
Neuroblastoma , Neurotoxinas , Humanos , Neurotoxinas/toxicidad , Neurotoxinas/metabolismo , 1-Metil-4-fenilpiridinio/toxicidad , Neuronas/metabolismo , Neuroblastoma/metabolismo , Transcripción Genética , Línea Celular Tumoral , Apoptosis
5.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34607950

RESUMEN

Among the organisms that spread into and flourish in Arctic waters with rising temperatures and sea ice loss are toxic algae, a group of harmful algal bloom species that produce potent biotoxins. Alexandrium catenella, a cyst-forming dinoflagellate that causes paralytic shellfish poisoning worldwide, has been a significant threat to human health in southeastern Alaska for centuries. It is known to be transported into Arctic regions in waters transiting northward through the Bering Strait, yet there is little recognition of this organism as a human health concern north of the Strait. Here, we describe an exceptionally large A. catenella benthic cyst bed and hydrographic conditions across the Chukchi Sea that support germination and development of recurrent, locally originating and self-seeding blooms. Two prominent cyst accumulation zones result from deposition promoted by weak circulation. Cyst concentrations are among the highest reported globally for this species, and the cyst bed is at least 6× larger in area than any other. These extraordinary accumulations are attributed to repeated inputs from advected southern blooms and to localized cyst formation and deposition. Over the past two decades, warming has likely increased the magnitude of the germination flux twofold and advanced the timing of cell inoculation into the euphotic zone by 20 d. Conditions are also now favorable for bloom development in surface waters. The region is poised to support annually recurrent A. catenella blooms that are massive in scale, posing a significant and worrisome threat to public and ecosystem health in Alaskan Arctic communities where economies are subsistence based.


Asunto(s)
Dinoflagelados/crecimiento & desarrollo , Dinoflagelados/metabolismo , Floraciones de Algas Nocivas/fisiología , Neurotoxinas/metabolismo , Intoxicación por Mariscos , Alaska , Regiones Árticas , Cambio Climático , Ecosistema , Sedimentos Geológicos/parasitología , Calor , Humanos , Cubierta de Hielo , Salud Pública
6.
Luminescence ; 39(5): e4761, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38807512

RESUMEN

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.


Asunto(s)
Ésteres , Naftoquinonas , Ésteres/química , Naftoquinonas/química , Diabetes Mellitus/metabolismo , Neurotoxinas/química , Neurotoxinas/metabolismo , Agua/química , Estructura Molecular , Hojas de la Planta/química
7.
Eur Biophys J ; 52(4-5): 445-457, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37209172

RESUMEN

We report the solution behavior, oligomerization state, and structural details of myotoxin-II purified from the venom of Bothrops asper in the presence and absence of sodium dodecyl sulfate (SDS) and multiple lipids, as examined by analytical ultracentrifugation and nuclear magnetic resonance. Molecular functional and structural details of the myotoxic mechanism of group II Lys-49 phospholipase A2 homologues have been only partially elucidated so far, and conflicting observations have been reported in the literature regarding the monomeric vs. oligomeric state of these toxins in solution. We observed the formation of a stable and discrete, hexameric form of myotoxin-II, but only in the presence of small amounts of SDS. In SDS-free medium, myotoxin-II was insensitive to mass action and remained monomeric at all concentrations examined (up to 3 mg/ml, 218.2 µM). At SDS concentrations above the critical micelle concentration, only dimers and trimers were observed, and at intermediate SDS concentrations, aggregates larger than hexamers were observed. We found that the amount of SDS required to form a stable hexamer varies with protein concentration, suggesting the need for a precise stoichiometry of free SDS molecules. The discovery of a stable hexameric species in the presence of a phospholipid mimetic suggests a possible physiological role for this oligomeric form, and may shed light on the poorly understood membrane-disrupting mechanism of this myotoxic protein class.


Asunto(s)
Bothrops , Neurotoxinas , Animales , Neurotoxinas/química , Neurotoxinas/metabolismo , Neurotoxinas/toxicidad , Bothrops/metabolismo , Fosfolipasas A2 , Espectroscopía de Resonancia Magnética , Bothrops asper
8.
Proc Natl Acad Sci U S A ; 117(38): 23815-23822, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32900920

RESUMEN

Prions are infectious agents which cause rapidly lethal neurodegenerative diseases in humans and animals following long, clinically silent incubation periods. They are composed of multichain assemblies of misfolded cellular prion protein. While it has long been assumed that prions are themselves neurotoxic, recent development of methods to obtain exceptionally pure prions from mouse brain with maintained strain characteristics, and in which defined structures-paired rod-like double helical fibers-can be definitively correlated with infectivity, allowed a direct test of this assertion. Here we report that while brain homogenates from symptomatic prion-infected mice are highly toxic to cultured neurons, exceptionally pure intact high-titer infectious prions are not directly neurotoxic. We further show that treatment of brain homogenates from prion-infected mice with sodium lauroylsarcosine destroys toxicity without diminishing infectivity. This is consistent with models in which prion propagation and toxicity can be mechanistically uncoupled.


Asunto(s)
Neurotoxinas , Enfermedades por Prión , Priones , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Química Encefálica , Modelos Animales de Enfermedad , Ratones , Neuronas/efectos de los fármacos , Neurotoxinas/aislamiento & purificación , Neurotoxinas/metabolismo , Neurotoxinas/toxicidad , Enfermedades por Prión/metabolismo , Enfermedades por Prión/fisiopatología , Priones/aislamiento & purificación , Priones/metabolismo , Priones/patogenicidad
9.
Proc Natl Acad Sci U S A ; 117(40): 24920-24928, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958636

RESUMEN

Australian funnel-web spiders are infamous for causing human fatalities, which are induced by venom peptides known as δ-hexatoxins (δ-HXTXs). Humans and other primates did not feature in the prey or predator spectrum during evolution of these spiders, and consequently the primate lethality of δ-HXTXs remains enigmatic. Funnel-web envenomations are mostly inflicted by male spiders that wander from their burrow in search of females during the mating season, which suggests a role for δ-HXTXs in self-defense since male spiders rarely feed during this period. Although 35 species of Australian funnel-web spiders have been described, only nine δ-HXTXs from four species have been characterized, resulting in a lack of understanding of the ecological roles and molecular evolution of δ-HXTXs. Here, by profiling venom-gland transcriptomes of 10 funnel-web species, we report 22 δ-HXTXs. Phylogenetic and evolutionary assessments reveal a remarkable sequence conservation of δ-HXTXs despite their deep evolutionary origin within funnel-web spiders, consistent with a defensive role. We demonstrate that δ-HXTX-Ar1a, the lethal toxin from the Sydney funnel-web spider Atrax robustus, induces pain in mice by inhibiting inactivation of voltage-gated sodium (NaV) channels involved in nociceptive signaling. δ-HXTX-Ar1a also inhibited inactivation of cockroach NaV channels and was insecticidal to sheep blowflies. Considering their algogenic effects in mice, potent insecticidal effects, and high levels of sequence conservation, we propose that the δ-HXTXs were repurposed from an initial insecticidal predatory function to a role in defending against nonhuman vertebrate predators by male spiders, with their lethal effects on humans being an unfortunate evolutionary coincidence.


Asunto(s)
Evolución Molecular , Neurotoxinas/genética , Poliaminas/química , Arañas/genética , Secuencia de Aminoácidos/genética , Animales , Australia , Secuencia Conservada/genética , Femenino , Humanos , Masculino , Ratones , Neurotoxinas/química , Neurotoxinas/metabolismo , Péptidos/genética , Filogenia , Poliaminas/metabolismo , Conducta Sexual Animal/fisiología , Venenos de Araña/genética , Arañas/patogenicidad , Transcriptoma/genética , Vertebrados/genética , Vertebrados/fisiología
10.
Bioconjug Chem ; 33(8): 1494-1504, 2022 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-35875886

RESUMEN

Recombinantly produced biotherapeutics hold promise for improving the current standard of care for snakebite envenoming over conventional serotherapy. Nanobodies have performed well in the clinic, and in the context of antivenom, they have shown the ability to neutralize long α-neurotoxins in vivo. Here, we showcase a protein engineering approach to increase the valence and hydrodynamic size of neutralizing nanobodies raised against a long α-neurotoxin (α-cobratoxin) from the venom of the monocled cobraNaja kaouthia. Based on the p53 tetramerization domain, a panel of anti-α-cobratoxin nanobody-p53 fusion proteins, termed Quads, were produced with different valences, inclusion or exclusion of Fc regions for endosomal recycling purposes, hydrodynamic sizes, and spatial arrangements, comprising up to 16 binding sites. Measurements of binding affinity and stoichiometry showed that the nanobody binding affinity was retained when incorporated into the Quad scaffold, and all nanobody domains were accessible for toxin binding, subsequently displaying increased blocking potency in vitro compared to the monomeric format. Moreover, functional assessment using automated patch-clamp assays demonstrated that the nanobody and Quads displayed neutralizing effects against long α-neurotoxins from both N. kaouthia and the forest cobra N. melanoleuca. This engineering approach offers a means of altering the valence, endosomal recyclability, and hydrodynamic size of existing nanobody-based therapeutics in a simple plug-and-play fashion and can thus serve as a technology for researchers tailoring therapeutic properties for improved neutralization of soluble targets such as snake toxins.


Asunto(s)
Elapidae , Anticuerpos de Dominio Único , Animales , Venenos Elapídicos/química , Venenos Elapídicos/metabolismo , Elapidae/metabolismo , Neurotoxinas/química , Neurotoxinas/metabolismo , Anticuerpos de Dominio Único/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
11.
Ecotoxicol Environ Saf ; 244: 114058, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36108432

RESUMEN

Ammonia pollutants were usually found in aquatic environments is due to urban sewage, industrial wastewater discharge, and agricultural runoff and concentrations as high as 180 mg/L (NH4+) have been reported in rivers. High ammonia levels are known to impair multiple tissue and cell functions and cause fish death. Although ammonia is a potent neurotoxin, how sublethal concentrations of ammonia influence the central nervous system (CNS) and the complex behaviors of fish is still unclear. In the present study, we demonstrated that acute sublethal ammonia exposure can change social behavior of adult zebrafish. The exposure to 90 mg /L of (NH4+) for 4 h induced a strong fear response and lower shoaling cohesion; exposure to 180 mg /L of (NH4+) for 4 h reduced the aggressiveness, and social recognition, while the anxiety, social preference, learning, and short-term memory were not affected. Messenger RNA expressions of glutaminase and glutamate dehydrogenase in the brain were induced, suggesting that ammonia exposure altered glutamate neurotransmitters in the CNS. Our findings in zebrafish provided delicate information of ammonia neurotoxicity in complex higher-order social behaviors, which has not been revealed previously. In conclusion, sublethal and acute ammonia exposure can change specific behaviors of fish, which might lead to reductions in individual and population fitness levels.


Asunto(s)
Contaminantes Químicos del Agua , Pez Cebra , Amoníaco/metabolismo , Amoníaco/toxicidad , Animales , Ansiedad/inducido químicamente , Cognición , Glutamato Deshidrogenasa/metabolismo , Glutamatos/metabolismo , Glutaminasa/metabolismo , Neurotoxinas/metabolismo , ARN Mensajero/metabolismo , Aguas del Alcantarillado , Conducta Social , Aguas Residuales , Contaminantes Químicos del Agua/metabolismo , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/metabolismo
12.
Int J Mol Sci ; 23(8)2022 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-35457172

RESUMEN

Tetanus and Botulinum type B neurotoxins are bacterial metalloproteases that specifically cleave the vesicle-associated membrane protein VAMP at an identical peptide bond, resulting in inhibition of neuroexocytosis. The minute amounts of these neurotoxins commonly used in experimental animals are not detectable, nor is detection of their VAMP substrate sensitive enough. The immune detection of the cleaved substrate is much more sensitive, as we have previously shown for botulinum neurotoxin type A. Here, we describe the production in rabbit of a polyclonal antibody raised versus a peptide encompassing the 13 residues C-terminal with respect to the neurotoxin cleavage site. The antibody was affinity purified and found to recognize, with high specificity and selectivity, the novel N-terminus of VAMP that becomes exposed after cleavage by tetanus toxin and botulinum toxin type B. This antibody recognizes the neoepitope not only in native and denatured VAMP but also in cultured neurons and in neurons in vivo in neurotoxin-treated mice or rats, suggesting the great potential of this novel tool to elucidate tetanus and botulinum B toxin activity in vivo.


Asunto(s)
Toxinas Botulínicas Tipo A , Tétanos , Animales , Anticuerpos/metabolismo , Ratones , Neurotoxinas/metabolismo , Péptidos/metabolismo , Proteolisis , Proteínas R-SNARE/química , Proteínas R-SNARE/metabolismo , Conejos , Ratas , Toxina Tetánica/química , Toxina Tetánica/metabolismo
13.
Molecules ; 27(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36500334

RESUMEN

Microtubule targeting agents (MTA) are anti-cancer molecules that bind tubulin and interfere with the microtubule functions, eventually leading to cell death. In the present study, we used an in vitro microtubule polymerization assay to screen several venom families for the presence of anti-microtubule activity. We isolated myotoxin-3, a peptide of the crotamine family, and three isoforms from the venom of the Northern Pacific rattlesnake Crotalus oreganus oreganus, which was able to increase tubulin polymerization. Myotoxin-3 turned out to be a cell-penetrating peptide that slightly diminished the viability of U87 glioblastoma and MCF7 breast carcinoma cells. Myotoxin 3 also induced remodeling of the U87 microtubule network and decreased MCF-7 microtubule dynamic instability. These effects are likely due to direct interaction with tubulin. Indeed, we showed that myotoxin-3 binds to tubulin heterodimer with a Kd of 5.3 µM and stoichiometry of two molecules of peptide per tubulin dimer. Our results demonstrate that exogenous peptides are good candidates for developing new MTA and highlight the richness of venoms as a source of pharmacologically active molecules.


Asunto(s)
Venenos de Crotálidos , Neurotoxinas , Animales , Humanos , Neurotoxinas/metabolismo , Tubulina (Proteína)/metabolismo , Crotalus/metabolismo , Venenos de Crotálidos/farmacología , Venenos de Crotálidos/metabolismo , Péptidos/farmacología , Péptidos/metabolismo
14.
Molecules ; 27(15)2022 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-35956907

RESUMEN

Glutamate excitotoxicity may contribute to retinal ganglion cell (RGC) degeneration in glaucoma and other optic neuropathies, leading to irreversible blindness. Growing evidence has linked impaired mitochondrial quality control with RGCs degeneration, while parkin, an E3 ubiquitin ligase, has proved to be protective and promotes mitophagy in RGCs against excitotoxicity. The purpose of this study was to explore whether a small molecule S3 could modulate parkin-mediated mitophagy and has therapeutic potential for RGCs. The results showed that as an inhibitor of deubiquitinase USP30, S3 protected cultured RGCs and improved mitochondrial health against NMDA-induced excitotoxicity. Administration of S3 promoted the parkin expression and its downstream mitophagy-related proteins in RGCs. An upregulated ubiquitination level of Mfn2 and protein level of OPA1 were also observed in S3-treated RGCs, while parkin knockdown resulted in a major loss of the protective effect of S3 on RGCs under excitotoxicity. These findings demonstrated that S3 promoted RGC survival mainly through enhancing parkin-mediated mitophagy against excitotoxicity. The neuroprotective value of S3 in glaucoma and other optic neuropathies deserves further investigation.


Asunto(s)
Mitofagia , Fármacos Neuroprotectores , Células Ganglionares de la Retina , Ubiquitina-Proteína Ligasas , Glaucoma/tratamiento farmacológico , Glaucoma/metabolismo , Ácido Glutámico/metabolismo , Humanos , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Mitofagia/efectos de los fármacos , Mitofagia/fisiología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/metabolismo , Enfermedades del Nervio Óptico/tratamiento farmacológico , Enfermedades del Nervio Óptico/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Tioléster Hidrolasas/antagonistas & inhibidores , Tioléster Hidrolasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
J Neurochem ; 158(6): 1244-1253, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33629408

RESUMEN

Tetanus is a deadly but preventable disease caused by a protein neurotoxin produced by Clostridium tetani. Spores of C. tetani may contaminate a necrotic wound and germinate into a vegetative bacterium that releases a toxin, termed tetanus neurotoxin (TeNT). TeNT enters the general circulation, binds to peripheral motor neurons and sensory neurons, and is transported retroaxonally to the spinal cord. It then enters inhibitory interneurons and blocks the release of glycine or GABA causing a spastic paralysis. This review attempts to correlate the metalloprotease activity of TeNT and its trafficking and localization into the vertebrate body to the nature and sequence of appearance of the symptoms of tetanus.


Asunto(s)
Encéfalo/metabolismo , Nervios Periféricos/metabolismo , Médula Espinal/metabolismo , Toxina Tetánica/metabolismo , Tétanos/metabolismo , Animales , Encéfalo/microbiología , Humanos , Neurotoxinas/antagonistas & inhibidores , Neurotoxinas/metabolismo , Nervios Periféricos/microbiología , Médula Espinal/microbiología , Tétanos/prevención & control , Toxina Tetánica/antagonistas & inhibidores , Toxoide Tetánico/administración & dosificación , Toxoide Tetánico/metabolismo
16.
Stem Cells ; 38(6): 727-740, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32083763

RESUMEN

Recent studies have demonstrated the generation of midbrain-like organoids (MOs) from human pluripotent stem cells. However, the low efficiency of MO generation and the relatively immature and heterogeneous structures of the MOs hinder the translation of these organoids from the bench to the clinic. Here we describe the robust generation of MOs with homogeneous distribution of midbrain dopaminergic (mDA) neurons. Our MOs contain not only mDA neurons but also other neuronal subtypes as well as functional glial cells, including astrocytes and oligodendrocytes. Furthermore, our MOs exhibit mDA neuron-specific cell death upon treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, indicating that MOs could be a proper human model system for studying the in vivo pathology of Parkinson's disease (PD). Our optimized conditions for producing homogeneous and mature MOs might provide an advanced patient-specific platform for in vitro disease modeling as well as for drug screening for PD.


Asunto(s)
Células-Madre Neurales/metabolismo , Neurotoxinas/metabolismo , Organoides/metabolismo , Enfermedad de Parkinson/genética , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Humanos , Enfermedad de Parkinson/patología
17.
Amino Acids ; 53(9): 1405-1413, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34245370

RESUMEN

Gene-encoded peptides with distinct potent bioactivities enable several animals to take advantage of fierce interspecific interaction, as seen in the skin secretion of amphibians. Unlike, most amphibian species that frequently switches terrestrial-aquatic habitats and hides easily from terrestrial predators, tree frogs of small body size are considered as the vulnerable prey in the arboreal habitat. Here, we show the structural and functional diversity of peptide families based on the skin transcriptome of Hyla japonica, which has evolved to be wrapped as an efficient chemical toolkit for defensive use in arboreal habitat. Generally, the presence of antimicrobial peptide and proteinase inhibitor families reveals the functional consistency of Hyla japonica skin compared to other amphibian species. Furthermore, we found that Anntoxin-like neurotoxins with high expression levels are species-specific in tree frogs. Interestingly, derivatives in the Anntoxin-like family exhibit multiple evolutionary traits in modifying the copy number, folding type, and three-dimensional architecture, which are considered essential for targeting the ion channels of terrestrial predators. Together, our study not only reveals the peptide diversity in the skin secretion of H. japonica, but also draws insights into the predator-deterring strategy for coping with arboreal habitat.


Asunto(s)
Proteínas Anfibias/metabolismo , Péptidos Antimicrobianos/metabolismo , Anuros/fisiología , Neurotoxinas/metabolismo , Conducta Predatoria , Piel/metabolismo , Transcriptoma , Secuencia de Aminoácidos , Proteínas Anfibias/genética , Animales , Péptidos Antimicrobianos/genética , Anuros/clasificación , Secuencia de Bases , Filogenia , Homología de Secuencia , Especificidad de la Especie
18.
Mol Biol Rep ; 48(2): 1475-1483, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33492574

RESUMEN

Rutin is an important flavonoid consumed in the daily diet. It is also known as vitamin P and has been extensively investigated due to its pharmacological properties. On the other hand, neuronal death induced by glutamate excitotoxicity is present in several diseases including neurodegenerative diseases. The neuroprotective properties of rutin have been under investigation, although its mechanism of action is still poorly understood. We hypothesized that the mechanisms of neuroprotection of rutin are associated with the increase in glutamate metabolism in astrocytes. This study aimed to evaluate the protective effects of rutin with a focus on the modulation of glutamate detoxification. We used brain organotypic cultures from post-natal Wistar rats (P7-P9) treated with rutin to evaluate neural cell protection and levels of proteins involved in the glutamate metabolism. Moreover, we used cerebral cortex slices from adult Wistar rats to evaluate glutamate uptake. We showed that rutin inhibited the cell death and loss of glutamine synthetase (GS) induced by glutamate that was associated with an increase in glutamate-aspartate transporter (GLAST) in brain organotypic cultures from post-natal Wistar rats. Additionally, it was observed that rutin increased the glutamate uptake in cerebral cortex slices from adult Wistar rats. We conclude that rutin is a neuroprotective agent that prevents glutamate excitotoxicity and thereof suggest that this effect involves the regulation of astrocytic metabolism.


Asunto(s)
Muerte Celular/efectos de los fármacos , Ácido Glutámico/metabolismo , Neuronas/efectos de los fármacos , Rutina/farmacología , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Transportador 1 de Aminoácidos Excitadores , Glutamato-Amoníaco Ligasa/genética , Ácido Glutámico/toxicidad , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Neurotoxinas/metabolismo , Neurotoxinas/toxicidad , Ratas , Ratas Wistar
19.
Exp Cell Res ; 396(2): 112322, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33068559

RESUMEN

Meningothelial cells (MECs) are the cellular component of the meninges that provide physical protection to the central nervous system (CNS). Their main function is the formation of a barrier enclosing the brain including the cerebrospinal fluid (CSF). Further, MECs are involved in maintaining CSF homeostasis by clearing CSF from bacteria and apoptotic cells. Furthermore, secretion of pro- and anti-inflammatory cytokines and chemokines involves MECs in immunological processes in the CNS. We demonstrated that meningothelial Ben-Men-1 cells ingest neurotoxic peptides amyloid-ß (Aß1-40) and protein α-synuclein up to about 10-fold more efficiently compared to neuronal-like SH-SY5Y cells. Aß1-40 and α-synuclein are mainly taken up via macropinocytosis. Caveolar endocytosis in addition contributes to α-synuclein ingestion. Upon uptake, both are trafficked towards lysosomal degradation. While production of reactive oxygen species (ROS) following exposure to Aß25-35 and α-synuclein was similar between Ben-Men-1 and SH-SY5Y cells, mitochondrial function in Ben-Men-1 was significantly more robust to Aß25-35 treatment compared to neuronal-like SHSY5Y cells. Similarly, Ben-Men-1 were significantly less susceptible to Aß25-35-induced cell death than neuronal-like cells. Furthermore, co-culture with Ben-Men-1 offered significant protection to neuronal-like cells against Aß25-35-induced apoptosis. This study reveals for the first time the function of MECs as scavengers of neurotoxic Aß and α-synuclein, thereby connecting these cells to neuroprotective processes and suggesting a new mechanism and pathway for clearing neurotoxic substances from the CSF.


Asunto(s)
Células Epiteliales/metabolismo , Meninges/citología , Neurotoxinas/metabolismo , Péptidos/metabolismo , Proteínas/metabolismo , Péptidos beta-Amiloides/metabolismo , Línea Celular Tumoral , Endocitosis , Humanos , Mitocondrias/metabolismo , Neuroprotección , Fracciones Subcelulares/metabolismo , alfa-Sinucleína/metabolismo
20.
Proc Natl Acad Sci U S A ; 115(11): E2634-E2643, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29487216

RESUMEN

Exposure of cultured primary neurons to preformed α-synuclein fibrils (PFFs) leads to the recruitment of endogenous α-synuclein and its templated conversion into fibrillar phosphorylated α-synuclein (pα-synF) aggregates resembling those involved in Parkinson's disease (PD) pathogenesis. Pα-synF was described previously as inclusions morphologically similar to Lewy bodies and Lewy neurites in PD patients. We discovered the existence of a conformationally distinct, nonfibrillar, phosphorylated α-syn species that we named "pα-syn*." We uniquely describe the existence of pα-syn* in PFF-seeded primary neurons, mice brains, and PD patients' brains. Through immunofluorescence and pharmacological manipulation we showed that pα-syn* results from incomplete autophagic degradation of pα-synF. Pα-synF was decorated with autophagic markers, but pα-syn* was not. Western blots revealed that pα-syn* was N- and C-terminally trimmed, resulting in a 12.5-kDa fragment and a SDS-resistant dimer. After lysosomal release, pα-syn* aggregates associated with mitochondria, inducing mitochondrial membrane depolarization, cytochrome C release, and mitochondrial fragmentation visualized by confocal and stimulated emission depletion nanoscopy. Pα-syn* recruited phosphorylated acetyl-CoA carboxylase 1 (ACC1) with which it remarkably colocalized. ACC1 phosphorylation indicates low ATP levels, AMPK activation, and oxidative stress and induces mitochondrial fragmentation via reduced lipoylation. Pα-syn* also colocalized with BiP, a master regulator of the unfolded protein response and a resident protein of mitochondria-associated endoplasmic reticulum membranes that are sites of mitochondrial fission and mitophagy. Pα-syn* aggregates were found in Parkin-positive mitophagic vacuoles and imaged by electron microscopy. Collectively, we showed that pα-syn* induces mitochondrial toxicity and fission, energetic stress, and mitophagy, implicating pα-syn* as a key neurotoxic α-syn species and a therapeutic target.


Asunto(s)
Autofagia/efectos de los fármacos , Mitofagia/efectos de los fármacos , Neurotoxinas , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína , Acetil-CoA Carboxilasa/química , Acetil-CoA Carboxilasa/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Química Encefálica , Técnicas de Cultivo de Célula , Células Cultivadas , Humanos , Lisosomas/metabolismo , Ratones , Mitocondrias , Neurotoxinas/química , Neurotoxinas/metabolismo , Neurotoxinas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Fosforilación , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , alfa-Sinucleína/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA