Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Hum Mol Genet ; 32(11): 1772-1785, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-36282524

RESUMEN

Autosomal dominant adult-onset neuronal ceroid lipofuscinosis (ANCL) is a rare neurodegenerative disorder characterized by progressive dementia and premature death. Four ANCL-causing mutations have been identified, all mapping to the DNAJC5 gene that encodes cysteine string protein α (CSPα). Here, using Caenorhabditis elegans, we describe an animal model of ANCL in which disease-causing mutations are introduced into their endogenous chromosomal locus, thereby mirroring the human genetic disorder. This was achieved through CRISPR/Cas9-mediated gene editing of dnj-14, the C. elegans ortholog of DNAJC5. The resultant homozygous ANCL mutant worms exhibited reduced lifespans and severely impaired chemotaxis, similar to isogenic dnj-14 null mutants. Importantly, these phenotypes were also seen in balanced heterozygotes carrying one wild-type and one ANCL mutant dnj-14 allele, mimicking the heterozygosity of ANCL patients. We observed a more severe chemotaxis phenotype in heterozygous ANCL mutant worms compared with haploinsufficient worms lacking one copy of CSP, consistent with a dominant-negative mechanism of action. Additionally, we provide evidence of CSP haploinsufficiency in longevity, as heterozygous null mutants exhibited significantly shorter lifespan than wild-type controls. The chemotaxis phenotype of dnj-14 null mutants was fully rescued by transgenic human CSPα, confirming the translational relevance of the worm model. Finally, a focused compound screen revealed that the anti-epileptic drug ethosuximide could restore chemotaxis in dnj-14 ANCL mutants to wild-type levels. This suggests that ethosuximide may have therapeutic potential for ANCL and demonstrates the utility of this C. elegans model for future larger-scale drug screening.


Asunto(s)
Caenorhabditis elegans , Lipofuscinosis Ceroideas Neuronales , Adulto , Animales , Humanos , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Etosuximida/farmacología , Etosuximida/uso terapéutico , Mutación , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo
2.
Cell Tissue Res ; 396(1): 41-55, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38403745

RESUMEN

Cysteine string protein (CSP) is a member of the DnaJ/Hsp40 family of molecular chaperones. CSP is enriched in neurons, where it mainly localises to synaptic vesicles. Mutations in CSP-encoding genes in flies, worms, mice and humans result in neuronal dysfunction, neurodegeneration and reduced lifespan. Most attention has therefore focused on CSP's neuronal functions, although CSP is also expressed in non-neuronal cells. Here, we used genome editing to fluorescently tag the Caenorhabditis elegans CSP orthologue, dnj-14, to identify which tissues preferentially express CSP and hence may contribute to the observed mutant phenotypes. Replacement of dnj-14 with wrmScarlet caused a strong chemotaxis defect, as seen with other dnj-14 null mutants. In contrast, inserting the reporter in-frame to create a DNJ-14-wrmScarlet fusion protein had no effect on chemotaxis, indicating that C-terminal tagging does not impair DNJ-14 function. WrmScarlet fluorescence appeared most obvious in the intestine, head/pharynx, spermathecae and vulva/uterus in the reporter strains, suggesting that DNJ-14 is preferentially expressed in these tissues. Crossing the DNJ-14-wrmScarlet strain with GFP marker strains confirmed the intestinal and pharyngeal expression, but only a partial overlap with neuronal GFP was observed. DNJ-14-wrmScarlet fluorescence in the intestine was increased in response to starvation, which may be relevant to mammalian CSPα's role in microautophagy. DNJ-14's enrichment in worm reproductive tissues (spermathecae and vulva/uterus) parallels the testis-specific expression of CSPß and CSPγ isoforms in mammals. Furthermore, CSPα messenger RNA is highly expressed in the human proximal digestive tract, suggesting that CSP may have a conserved, but overlooked, function within the gastrointestinal system.


Asunto(s)
Sistemas CRISPR-Cas , Caenorhabditis elegans , Proteínas del Choque Térmico HSP40 , Masculino , Animales , Ratones , Humanos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Distribución Tisular , Sistemas CRISPR-Cas/genética , Proteínas de la Membrana/metabolismo , Mamíferos/metabolismo
3.
Biochem J ; 478(24): 4153-4167, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34661239

RESUMEN

Dietary restriction (DR) has been shown to increase lifespan in organisms ranging from yeast to mammals. This suggests that the underlying mechanisms may be evolutionarily conserved. Indeed, upstream signalling pathways, such as TOR, are strongly linked to DR-induced longevity in various organisms. However, the downstream effector proteins that ultimately mediate lifespan extension are less clear. To shed light on this, we used a proteomic approach on budding yeast. Our reasoning was that analysis of proteome-wide changes in response to DR might enable the identification of proteins that mediate its physiological effects, including replicative lifespan extension. Of over 2500 proteins we identified by liquid chromatography-mass spectrometry, 183 were significantly altered in expression by at least 3-fold in response to DR. Most of these proteins were mitochondrial and/or had clear links to respiration and metabolism. Indeed, direct analysis of oxygen consumption confirmed that mitochondrial respiration was increased several-fold in response to DR. In addition, several key proteins involved in mating, including Ste2 and Ste6, were down-regulated by DR. Consistent with this, shmoo formation in response to α-factor pheromone was reduced by DR, thus confirming the inhibitory effect of DR on yeast mating. Finally, we found that Hsp26, a member of the conserved small heat shock protein (sHSP) family, was up-regulated by DR and that overexpression of Hsp26 extended yeast replicative lifespan. As overexpression of sHSPs in Caenorhabditis elegans and Drosophila has previously been shown to extend lifespan, our data on yeast Hsp26 suggest that sHSPs may be universally conserved effectors of longevity.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteoma
4.
Epilepsia ; 61(4): 810-821, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32112430

RESUMEN

OBJECTIVE: Genetic variants in STXBP1, which encodes the conserved exocytosis protein Munc18-1, are associated with a variety of infantile epilepsy syndromes. We aimed to develop an in vivo Caenorhabditis elegans model that could be used to test the pathogenicity of such variants in a cost-effective manner. METHODS: The CRISPR/Cas9 method was used to introduce a null mutation into the unc-18 gene (the C. elegans orthologue of STXBP1), thereby creating a paralyzed worm strain. We subsequently rescued this strain with transgenes encoding the human STXBP1/Munc18-1 protein (wild-type and eight different epilepsy-associated missense variants). The resulting humanized worm strains were then analyzed via behavioral, electrophysiological, and biochemical approaches. RESULTS: Transgenic expression of wild-type human STXBP1 protein fully rescued locomotion in both solid and liquid media to the same level as the standard wild-type worm strain, Bristol N2. Six variant strains (E59K, V84D, C180Y, R292H, L341P, R551C) exhibited impaired locomotion, whereas two (P335L, R406H) were no different from worms expressing wild-type STXBP1. Electrophysiological recordings revealed that all eight variant strains displayed less frequent and more irregular pharyngeal pumping in comparison to wild-type STXBP1-expressing strains. Four strains (V84D, C180Y, R292H, P335L) exhibited pentylenetetrazol-induced convulsions in an acute assay of seizure-like activity, in contrast to worms expressing wild-type STXBP1. No differences were seen between wild-type and variant STXBP1 strains in terms of mRNA abundance. However, STXBP1 protein levels were reduced to 20%-30% of wild-type in all variants, suggesting that the mutations result in STXBP1 protein instability. SIGNIFICANCE: The approach described here is a cost-effective in vivo method for establishing the pathogenicity of genetic variants in STXBP1 and potentially other conserved neuronal proteins. Furthermore, the humanized strains we created could potentially be used in the future for high-throughput drug screens to identify novel therapeutics.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia/genética , Proteínas Munc18/genética , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Humanos , Mutación , Proteínas de Transporte Vesicular/genética
5.
Neurobiol Dis ; 118: 40-54, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29940336

RESUMEN

The antiepileptic drug ethosuximide has recently been shown to be neuroprotective in various Caenorhabditis elegans and rodent neurodegeneration models. It is therefore a promising repurposing candidate for the treatment of multiple neurodegenerative diseases. However, high concentrations of the drug are required for its protective effects in animal models, which may impact on its translational potential and impede the identification of its molecular mechanism of action. Therefore, we set out to develop more potent neuroprotective lead compounds based on ethosuximide as a starting scaffold. Chemoinformatic approaches were used to identify compounds with structural similarity to ethosuximide and to prioritise these based on good predicated blood-brain barrier permeability and C. elegans bioaccumulation properties. Selected compounds were initially screened for anti-convulsant activity in a C. elegans pentylenetetrazol-induced seizure assay, as a rapid primary readout of bioactivity; and then assessed for neuroprotective properties in a C. elegans TDP-43 proteinopathy model based on pan-neuronal expression of human A315T mutant TDP-43. The most potent compound screened, α-methyl-α-phenylsuccinimide (MPS), ameliorated the locomotion defects and extended the shortened lifespan of TDP-43 mutant worms. MPS also directly protected against neurodegeneration by reducing the number of neuronal breaks and cell body losses in GFP-labelled GABAergic motor neurons. Importantly, optimal neuroprotection was exhibited by external application of 50 µM MPS, compared to 8 mM for ethosuximide. This greater potency of MPS was not due to bioaccumulation to higher internal levels within the worm, based on 1H-nuclear magnetic resonance analysis. Like ethosuximide, the activity of MPS was abolished by mutation of the evolutionarily conserved FOXO transcription factor, daf-16, suggesting that both compounds act via the same neuroprotective pathway(s). In conclusion, we have revealed a novel neuroprotective activity of MPS that is >100-fold more potent than ethosuximide. This increased potency will facilitate future biochemical studies to identify the direct molecular target(s) of both compounds, as we have shown here that they share a common downstream DAF-16-dependent mechanism of action. Furthermore, MPS is the active metabolite of another approved antiepileptic drug, methsuximide. Therefore, methsuximide may have repurposing potential for treatment of TDP-43 proteinopathies and possibly other human neurodegenerative diseases.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Modelos Animales de Enfermedad , Succinimidas/uso terapéutico , Proteinopatías TDP-43/tratamiento farmacológico , Proteinopatías TDP-43/genética , Animales , Animales Modificados Genéticamente , Anticonvulsivantes/química , Anticonvulsivantes/uso terapéutico , Caenorhabditis elegans , Femenino , Masculino , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Succinimidas/química , Proteinopatías TDP-43/patología
6.
Hum Mol Genet ; 23(22): 5916-27, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24947438

RESUMEN

Adult onset neuronal lipofuscinosis (ANCL) is a human neurodegenerative disorder characterized by progressive neuronal dysfunction and premature death. Recently, the mutations that cause ANCL were mapped to the DNAJC5 gene, which encodes cysteine string protein alpha. We show here that mutating dnj-14, the Caenorhabditis elegans orthologue of DNAJC5, results in shortened lifespan and a small impairment of locomotion and neurotransmission. Mutant dnj-14 worms also exhibited age-dependent neurodegeneration of sensory neurons, which was preceded by severe progressive chemosensory defects. A focussed chemical screen revealed that resveratrol could ameliorate dnj-14 mutant phenotypes, an effect mimicked by the cAMP phosphodiesterase inhibitor, rolipram. In contrast to other worm neurodegeneration models, activation of the Sirtuin, SIR-2.1, was not required, as sir-2.1; dnj-14 double mutants showed full lifespan rescue by resveratrol. The Sirtuin-independent neuroprotective action of resveratrol revealed here suggests potential therapeutic applications for ANCL and possibly other human neurodegenerative diseases.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de la Membrana/metabolismo , Lipofuscinosis Ceroideas Neuronales/metabolismo , Sirtuinas/metabolismo , Estilbenos/farmacología , Adulto , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Evaluación Preclínica de Medicamentos , Proteínas del Choque Térmico HSP40/genética , Humanos , Esperanza de Vida , Proteínas de la Membrana/genética , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Lipofuscinosis Ceroideas Neuronales/genética , Resveratrol , Sirtuinas/genética
7.
Epilepsia Open ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38813985

RESUMEN

OBJECTIVE: GABAA receptor subunit mutations pose a significant risk for genetic generalized epilepsy; however, there are over 150 identified variants, many with unknown or unvalidated pathogenicity. We aimed to develop in vivo models for testing GABAA receptor variants using the model organism, Caenorhabditis elegans. METHODS: CRISPR-Cas9 gene editing was used to create a complete deletion of unc-49, a C. elegans GABAA receptor, and to create homozygous epilepsy-associated mutations in the endogenous unc-49 gene. The unc-49 deletion strain was rescued with transgenes for either the C. elegans unc-49B subunit or the α1, ß3, and γ2 subunits for the human GABAA receptor. All newly created strains were analyzed for phenotype and compared against existing unc-49 mutations. RESULTS: Nematodes with a full genetic deletion of the entire unc-49 locus were compared with existing unc-49 mutations in three separate phenotypic assays-coordinated locomotion, shrinker frequency and seizure-like convulsions. The full unc-49 deletion exhibited reduced locomotion and increased shrinker frequency and PTZ-induced convulsions, but were not found to be phenotypically stronger than existing unc-49 mutations. Rescue with the unc-49B subunit or creation of humanized worms for the GABAA receptor both showed partial phenotypic rescue for all three phenotypes investigated. Finally, two epilepsy-associated variants were analyzed and deemed to be loss of function, thus validating their pathogenicity. SIGNIFICANCE: These findings establish C. elegans as a genetic model to investigate GABAA receptor mutations and delineate a platform for validating associated variants in any epilepsy-associated gene. PLAIN LANGUAGE SUMMARY: Epilepsy is a complex human disease that can be caused by mutations in specific genes. Many possible mutations have been identified, but it is unknown for most of them whether they cause the disease. We tested the role of mutations in one specific gene using a small microscopic worm as an animal model. Our results establish this worm as a model for epilepsy and confirm that the two unknown mutations are likely to cause the disease.

8.
J Neurosci ; 32(20): 7042-51, 2012 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-22593072

RESUMEN

Diacylglycerol (DAG)/protein kinase C (PKC) signaling plays an integral role in the regulation of neuronal function. This is certainly true in Caenorhabditis elegans and in particular for thermosensory signaling and behavior. Downstream molecular targets for transduction of this signaling cascade remain, however, virtually uncharacterized. We investigated whether PKC phosphorylation of Munc18-1, an essential protein in vesicle trafficking and exocytosis, was the downstream effector for DAG regulation of thermosensory behavior. We demonstrate here that the C. elegans ortholog of Munc18-1, UNC-18, was phosphorylated in vitro at Ser322. Transgenic rescue of unc-18-null worms with Ser322 phosphomutants displayed altered thermosensitivity. C. elegans expresses three DAG-regulated PKCs, and blocking UNC-18 Ser322 phosphorylation was phenocopied only by deletion of calcium-activated PKC-2. Expression of nonphosphorylatable UNC-18 S322A, either pan-neuronally or specifically in AFD thermosensory neurons, converted wild-type worms to a pkc-2-null phenotype. These data demonstrate that an individual DAG-dependent thermosensory behavior of an organism is effected specifically by the downstream PKC-2 phosphorylation of UNC-18 on Ser322 in AFD neurons.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Locomoción/fisiología , Fosfoproteínas/fisiología , Proteína Quinasa C/fisiología , Células Receptoras Sensoriales/fisiología , Sensación Térmica/fisiología , Proteínas de Transporte Vesicular/fisiología , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Diglicéridos/metabolismo , Diglicéridos/fisiología , Isoenzimas/genética , Isoenzimas/fisiología , Mutación , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteínas Qa-SNARE/metabolismo , Células Receptoras Sensoriales/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
9.
MicroPubl Biol ; 20232023.
Artículo en Inglés | MEDLINE | ID: mdl-37090152

RESUMEN

Changes in neuronal function that occur with age are an area of increasing importance. A potential significant contributor to age-dependent decline may be alterations to neurotransmitter release. Protein kinases, such as Protein Kinase C and Protein Kinase A, are well characterised modulators of neuronal function and neurotransmission. Protein Kinase D (PRKD) is a serine/threonine kinase whose role in neurons is less well characterised. Here we report that mutations in the C. elegans PRKD homolog, dkf-1 , show an acceleration in age-dependent decline of locomotion rate and an alteration to age-dependent changes in aldicarb sensitivity. These effects could be explained by a pre- or post-synaptic function of the protein kinase as the animal ages.

10.
Sci Rep ; 13(1): 20616, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996473

RESUMEN

Biological pathways between alcohol consumption and alcohol liver disease (ALD) are not fully understood. We selected genes with known effect on (1) alcohol consumption, (2) liver function, and (3) gene expression. Expression of the orthologs of these genes in Caenorhabditis elegans and Drosophila melanogaster was suppressed using mutations and/or RNA interference (RNAi). In humans, association analysis, pathway analysis, and Mendelian randomization analysis were performed to identify metabolic changes due to alcohol consumption. In C. elegans, we found a reduction in locomotion rate after exposure to ethanol for RNAi knockdown of ACTR1B and MAPT. In Drosophila, we observed (1) a change in sedative effect of ethanol for RNAi knockdown of WDPCP, TENM2, GPN1, ARPC1B, and SCN8A, (2) a reduction in ethanol consumption for RNAi knockdown of TENM2, (3) a reduction in triradylglycerols (TAG) levels for RNAi knockdown of WDPCP, TENM2, and GPN1. In human, we observed (1) a link between alcohol consumption and several metabolites including TAG, (2) an enrichment of the candidate (alcohol-associated) metabolites within the linoleic acid (LNA) and alpha-linolenic acid (ALA) metabolism pathways, (3) a causal link between gene expression of WDPCP to liver fibrosis and liver cirrhosis. Our results imply that WDPCP might be involved in ALD.


Asunto(s)
Caenorhabditis elegans , Drosophila melanogaster , Metabolismo de los Lípidos , Hepatopatías Alcohólicas , Animales , Humanos , Consumo de Bebidas Alcohólicas/genética , Caenorhabditis elegans/genética , Drosophila melanogaster/genética , Etanol/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Cirrosis Hepática/patología , Hepatopatías Alcohólicas/metabolismo
11.
J Neurosci ; 31(24): 9055-66, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677188

RESUMEN

The release of hormones and neurotransmitters, mediated by regulated exocytosis, can be modified by regulation of the fusion pore. The fusion pore is considered stable and narrow initially, eventually leading to the complete merger of the vesicle and the plasma membranes. By using the high-resolution patch-clamp capacitance technique, we studied single vesicles and asked whether the Sec1/Munc18 proteins, interacting with the membrane fusion-mediating SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins, affect fusion pore properties. Munc18-1 mutants were transfected into lactotrophs to affect the interaction of Munc18-1 with syntaxin1 (Synt1) (R39C), Rab3A (E466K), and Mints (P242S). Compared with wild-type, Munc18-1 E466K increased the frequency of the fusion event. The latter two mutants increased the fusion pore dwell-time. All the mutants stabilized narrow fusion pores and increased the amplitude of fusion events, likely via preferential fusion of larger vesicles, since overexpression of Munc18-1 R39C did not affect the average size of vesicles, as determined by stimulated emission depletion (STED) microscopy. Single-molecule atomic force microscopy experiments revealed that wild-type Munc18-1, but not Munc18-1 R39C, abrogates the interaction between synaptobrevin2 (Syb2) and Synt1 binary trans-complexes. However, neither form of Munc18-1 affected the interaction of Syb2 with the preformed binary cis-Synt1A-SNAP25B complexes. This indicates that Munc18-1 performs a proofing function by inhibiting tethering of Syb2-containing vesicles solely to Synt1 at the plasmalemma and favoring vesicular tethering to the preformed binary cis-complex of Synt1A-SNAP25B. The association of Munc18-1 with the ternary SNARE complex leads to tuning of fusion pores via multiple and converging mechanisms involving Munc18-1 interactions with Synt1A, Rab3A, and Mints.


Asunto(s)
Vesículas Citoplasmáticas/fisiología , Fusión de Membrana/fisiología , Proteínas Munc18/genética , Mutación/genética , Análisis de Varianza , Animales , Células Cultivadas , Capacidad Eléctrica , Glutamina/genética , Proteínas Fluorescentes Verdes/genética , Lactotrofos/citología , Lisina/genética , Masculino , Fusión de Membrana/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Mentha/genética , Mentha/metabolismo , Microscopía de Fuerza Atómica/métodos , Microscopía Confocal , Modelos Biológicos , Proteínas Munc18/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Wistar , Estadísticas no Paramétricas , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo , Sintaxina 1/genética , Sintaxina 1/metabolismo , Transfección/métodos , Proteína de Unión al GTP rab3A/genética , Proteína de Unión al GTP rab3A/metabolismo
12.
Biochem Soc Trans ; 38(2): 559-63, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20298221

RESUMEN

Neurodegenerative disorders such as Alzheimer's, Parkinson's and Huntington's diseases affect millions of people. These disorders are age-dependent, progressive and, at present, incurable. A practical and relevant model is needed to investigate the molecular determinants of these debilitating diseases. Mammalian models are often prohibitively expensive, time-consuming and very complex. Given the highly conserved neurological pathways between mammals and invertebrates, Caenorhabditis elegans has emerged as a powerful tool for the investigation of the pathophysiology of these disorders. We describe recent findings in this area and show how C. elegans is being used to broaden our knowledge of human neurodegenerative diseases.


Asunto(s)
Caenorhabditis elegans , Modelos Animales , Degeneración Nerviosa/genética , Enfermedades Neurodegenerativas/patología , Animales , Caenorhabditis elegans/genética , Humanos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Enfermedades Neurodegenerativas/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Tauopatías/genética , Tauopatías/patología
13.
Biochem Soc Trans ; 38(Pt 1): 172-6, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20074054

RESUMEN

Acute exposure to ethanol is known to modulate signalling within the nervous system. Physiologically these effects are both presynaptic and postsynaptic in origin; however, considerably more research has focused primarily on postsynaptic targets. Recent research using the model organism Caenorhabditis elegans has determined a role for specific proteins (Munc18-1 and Rab3) and processes (synaptic vesicle recruitment and fusion) in transducing the presynaptic effects of ethanol. In the present paper, we review these results, identifying the proteins and protein interactions involved in ethanol sensitivity and discuss their links with mammalian studies of alcohol abuse.


Asunto(s)
Etanol/farmacología , Terminales Presinápticos/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Humanos , Proteínas Munc18/metabolismo , Terminales Presinápticos/metabolismo , Proteínas SNARE/metabolismo , Transmisión Sináptica/fisiología , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
14.
Biochem J ; 418(1): 73-80, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19032153

RESUMEN

SNAREs (soluble N-ethylmaleimide-sensitive fusion protein-attachment protein receptors) are widely accepted to drive all intracellular membrane fusion events. SM (Sec1/Munc18-like) proteins bind to SNAREs and this interaction may underlie their ubiquitous requirement for efficient membrane fusion. SM proteins bind to SNAREs in at least three modes: (i) to a closed conformation of syntaxin; (ii) to the syntaxin N-terminus; and (iii) to the assembled SNARE complex. Munc18-1 exhibits all three binding modes and recent in vitro reconstitution assays suggest that its interaction with the syntaxin N-terminus is essential for neuronal SNARE complex binding and efficient membrane fusion. To investigate the physiological relevance of these binding modes, we studied the UNC-18/UNC-64 SM/SNARE pair, which is essential for neuronal exocytosis in Caenorhabditis elegans. Mutations in the N-terminus of UNC-64 strongly inhibited binding to UNC-18, as did mutations targeting closed conformation binding. Complementary mutations in UNC-18 designed to selectively impair binding to either closed syntaxin or its N-terminus produced a similarly strong inhibition of UNC-64 binding. Therefore high-affinity UNC18/UNC-64 interaction in vitro involves both binding modes. To determine the physiological relevance of each mode, unc-18-null mutant worms were transformed with wild-type or mutant unc-18 constructs. The UNC-18(R39C) construct, that is defective in closed syntaxin binding, fully rescued the locomotion defects of the unc-18 mutant. In contrast, the UNC-18(F113R) construct, that is defective in binding to the N-terminus of UNC-64, provided no rescue. These results suggest that binding of UNC-18 to closed syntaxin is dispensable for membrane fusion, whereas interaction with the syntaxin N-terminus is essential for neuronal exocytosis in vivo.


Asunto(s)
Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/química , Caenorhabditis elegans/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas Qa-SNARE/química , Proteínas Qa-SNARE/metabolismo , Transmisión Sináptica , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Secuencia Conservada , Humanos , Datos de Secuencia Molecular , Mutación/genética , Fosfoproteínas/genética , Unión Proteica , Proteínas Qa-SNARE/genética , Alineación de Secuencia , Proteínas de Transporte Vesicular/genética
15.
Genetics ; 180(1): 165-78, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18757920

RESUMEN

SNAP receptor (SNARE) and Sec1/Munc18 (SM) proteins are required for all intracellular membrane fusion events. SNAREs are widely believed to drive the fusion process, but the function of SM proteins remains unclear. To shed light on this, we screened for dominant-negative mutants of yeast Sec1 by random mutagenesis of a GAL1-regulated SEC1 plasmid. Mutants were identified on the basis of galactose-inducible growth arrest and inhibition of invertase secretion. This effect of dominant-negative sec1 was suppressed by overexpression of the vesicle (v)-SNAREs, Snc1 and Snc2, but not the target (t)-SNAREs, Sec9 and Sso2. The mutations isolated in Sec1 clustered in a hotspot within domain 3a, with F361 mutated in four different mutants. To test if this region was generally involved in SM protein function, the F361-equivalent residue in mammalian Munc18-1 (Y337) was mutated. Overexpression of the Munc18-1 Y337L mutant in bovine chromaffin cells inhibited the release kinetics of individual exocytosis events. The Y337L mutation impaired binding of Munc18-1 to the neuronal SNARE complex, but did not affect its binary interaction with syntaxin1a. Taken together, these data suggest that domain 3a of SM proteins has a functionally important role in membrane fusion. Furthermore, this approach of screening for dominant-negative mutants in yeast may be useful for other conserved proteins, to identify functionally important domains in their mammalian homologs.


Asunto(s)
Proteínas Munc18/genética , Mutagénesis , Proteínas de Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Animales , Bovinos , Membrana Celular/metabolismo , Células Cromafines/metabolismo , Exocitosis , Regulación de la Expresión Génica , Genes Dominantes , Cinética , Datos de Secuencia Molecular , Proteínas Munc18/química , Mutación , Estructura Terciaria de Proteína , Proteínas de Saccharomyces cerevisiae/química
16.
Biochem J ; 409(2): 407-16, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17919117

RESUMEN

Munc18-1 plays a crucial role in regulated exocytosis in neurons and neuroendocrine cells through modulation of vesicle docking and membrane fusion. The molecular basis for Munc18 function is still unclear, as are the links with Rabs and SNARE [SNAP (soluble N-ethylmaleimide-sensitive factor-attachment protein) receptor] proteins that are also required. Munc18-1 can bind to SNAREs through at least three modes of interaction, including binding to the closed conformation of syntaxin 1. Using a gain-of-function mutant of Munc18-1 (E466K), which is based on a mutation in the related yeast protein Sly1p, we have identified a direct interaction of Munc18-1 with Rab3A, which is increased by the mutation. Expression of Munc18-1 with the E466K mutation increased exocytosis in adrenal chromaffin cells and PC12 cells (pheochromocytoma cells) and was found to increase the density of secretory granules at the periphery of PC12 cells, suggesting a stimulatory effect on granule recruitment through docking or tethering. Both the increase in exocytosis and changes in granule distribution appear to require Munc18-1 E466K binding to the closed form of syntaxin 1, suggesting a role for this interaction in bridging Rab- and SNARE-mediated events in exocytosis.


Asunto(s)
Exocitosis/fisiología , Proteínas Munc18/genética , Proteínas Munc18/metabolismo , Mutación , Vesículas Secretoras/metabolismo , Proteínas de Unión al GTP rab3/metabolismo , Animales , Células Cultivadas , Microscopía Confocal , Células PC12 , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas SNARE/metabolismo
17.
Biochem J ; 413(3): 479-91, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18452404

RESUMEN

Exocytosis is regulated by NO in many cell types, including neurons. In the present study we show that syntaxin 1a is a substrate for S-nitrosylation and that NO disrupts the binding of Munc18-1 to the closed conformation of syntaxin 1a in vitro. In contrast, NO does not inhibit SNARE {SNAP [soluble NSF (N-ethylmaleimide-sensitive fusion protein) attachment protein] receptor} complex formation or binding of Munc18-1 to the SNARE complex. Cys(145) of syntaxin 1a is the target of NO, as a non-nitrosylatable C145S mutant is resistant to NO and novel nitrosomimetic Cys(145) mutants mimic the effect of NO on Munc18-1 binding in vitro. Furthermore, expression of nitrosomimetic syntaxin 1a in living cells affects Munc18-1 localization and alters exocytosis release kinetics and quantal size. Molecular dynamic simulations suggest that NO regulates the syntaxin-Munc18 interaction by local rearrangement of the syntaxin linker and H3c regions. Thus S-nitrosylation of Cys(145) may be a molecular switch to disrupt Munc18-1 binding to the closed conformation of syntaxin 1a, thereby facilitating its engagement with the membrane fusion machinery.


Asunto(s)
Cisteína/metabolismo , Proteínas Munc18/metabolismo , Sintaxina 1/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Línea Celular , Membrana Celular/metabolismo , Simulación por Computador , Cisteína/química , Cisteína/genética , Exocitosis , Células HeLa , Humanos , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas Munc18/química , Proteínas Munc18/genética , Óxido Nítrico/metabolismo , Plásmidos/genética , Unión Proteica , Proteínas SNARE/metabolismo , Homología de Secuencia de Aminoácido , Sintaxina 1/química , Sintaxina 1/genética , Termodinámica
18.
Biophys J ; 94(3): 1084-93, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17921224

RESUMEN

Carbon fiber amperometry is a popular method for measuring single exocytotic events; however, the functional interpretation of the data can prove hazardous. For example, changes to vesicle transmitter levels can appear to cause changes in the timing and rate of the fusion process itself. Use of an analytical technique based on differentiation revealed that an increase in dense-core granule catecholamine content by exogenous application of l-DOPA did not affect initial release rates. Changes to the timing and amplitude of amperometric spikes from l-DOPA-treated cells are, then, likely a reflection of the increased quantal size rather than any direct effect on exocytosis itself. Applying this new analysis to individual fusion events from cells expressing Munc-18-1 with various specific point mutations demonstrated that Munc-18-1 functions at a late stage involved in the determination of the initial rate of fusion. Furthermore, a mutation of the protein that inhibits its biochemical interaction with the t-SNARE syntaxin-1 in a closed conformation caused premature termination of the fusion event. Through these two late-stage functions, Munc-18-1 could act as a key protein involved in the presynaptic control of signaling strength and duration.


Asunto(s)
Células Cromafines/fisiología , Exocitosis/fisiología , Fusión de Membrana/fisiología , Proteínas Munc18/metabolismo , Proteínas Qa-SNARE/metabolismo , Animales , Bovinos , Células Cultivadas
19.
Mol Biol Cell ; 16(2): 470-82, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15563604

RESUMEN

Sec1/Munc18 (SM) proteins are involved in various intracellular membrane trafficking steps. Many SM proteins bind to appropriate syntaxin homologues involved in these steps, suggesting that SM proteins function as syntaxin chaperones. Organisms with mutations in SM genes, however, exhibit defects in either early (docking) or late (fusion) stages of exocytosis, implying that SM proteins may have multiple functions. To gain insight into the role of SM proteins, we introduced mutations modeled on those identified in Caenorhabditis elegans, Drosophila melanogaster, and Saccharomyces cerevisiae into mammalian Munc18-1. As expected, several mutants exhibited reduced binding to syntaxin1A. However, three mutants displayed wild-type syntaxin binding affinities, indicating syntaxin-independent defects. Expression of these mutants in chromaffin cells either increased the rate and extent of exocytosis or altered the kinetics of individual release events. This latter effect was associated with a reduced Mint binding affinity in one mutant, implying a potential mechanism for the observed alteration in release kinetics. Furthermore, this phenotype persisted when the mutation was combined with a second mutation that greatly reduced syntaxin binding affinity. These results clarify the data on the function of SM proteins in mutant organisms and indicate that Munc18-1 controls multiple stages of exocytosis via both syntaxin-dependent and -independent protein interactions.


Asunto(s)
Exocitosis/fisiología , Regulación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas/metabolismo , Proteínas de Transporte Vesicular/genética , Médula Suprarrenal/citología , Alelos , Animales , Bovinos , Células Cultivadas , Células Cromafines/fisiología , Electrofisiología , Exocitosis/genética , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Cinética , Modelos Moleculares , Proteínas Munc18 , Mutación Missense , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Proteínas Qa-SNARE , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Proteínas de Transporte Vesicular/metabolismo
20.
J Neurosci Methods ; 309: 132-142, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30189284

RESUMEN

BACKGROUND: Epilepsy affects around 1% of people, but existing antiepileptic drugs (AEDs) only offer symptomatic relief and are ineffective in approximately 30% of patients. Hence, new AEDs are sorely needed. However, a major bottleneck is the low-throughput nature of early-stage AED screens in conventional rodent models. This process could potentially be expedited by using simpler invertebrate systems, such as the nematode Caenorhabditis elegans. NEW METHOD: Head-bobbing convulsions were previously reported to be inducible by pentylenetetrazol (PTZ) in C. elegans with loss-of-function mutations in unc-49, which encodes a GABAA receptor. Given that epilepsy-linked mutations in human GABAA receptors are well documented, this could represent a clinically-relevant system for early-stage AED screens. However, the original agar plate-based assay is unsuited to large-scale screening and has not been validated for identifying AEDs. Therefore, we established an alternative streamlined, higher-throughput approach whereby mutants were treated with PTZ and AEDs via liquid-based incubation. RESULTS: Convulsions induced within minutes of PTZ exposure in unc-49 mutants were strongly inhibited by the established AED ethosuximide. This protective activity was independent of ethosuximide's suggested target, the T-type calcium channel, as a null mutation in the worm cca-1 ortholog did not affect ethosuximide's anticonvulsant action. COMPARISON WITH EXISTING METHOD: Our streamlined assay is AED-validated, feasible for higher throughput compound screens, and can facilitate insights into AED mechanisms of action. CONCLUSIONS: Based on an epilepsy-associated genetic background, this C. elegans unc-49 model of seizure-like activity presents an ethical, higher throughput alternative to conventional rodent seizure models for initial AED screens.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Convulsiones/prevención & control , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Convulsivantes/administración & dosificación , Etosuximida/administración & dosificación , Pentilenotetrazol/administración & dosificación , Receptores de GABA-A/genética , Convulsiones/inducido químicamente
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda