Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brief Bioinform ; 25(6)2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39311700

RESUMEN

De novo mutations in the synaptic GTPase activating protein (SynGAP) are associated with neurological disorders like intellectual disability, epilepsy, and autism. SynGAP is also implicated in Alzheimer's disease and cancer. Although pathogenic variants are highly penetrant in neurodevelopmental conditions, a substantial number of them are caused by missense mutations that are difficult to diagnose. Hence, in silico mutagenesis was performed for probing the missense effects within the N-terminal region of SynGAP structure. Through extensive molecular dynamics simulations, encompassing three 150-ns replicates for 211 variants, the impact of missense mutations on the protein fold was assessed. The effect of the mutations on the folding stability was also quantitatively assessed using free energy calculations. The mutations were categorized as potentially pathogenic or benign based on their structural impacts. Finally, the study introduces wild-type-SynGAP in complex with RasGTPase at the inner membrane, while considering the potential effects of mutations on these key interactions. This study provides structural perspective to the clinical assessment of SynGAP missense variants and lays the foundation for future structure-based drug discovery.


Asunto(s)
Simulación de Dinámica Molecular , Mutación Missense , Proteínas Activadoras de ras GTPasa , Humanos , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/química , Proteínas Activadoras de ras GTPasa/metabolismo , Pliegue de Proteína , Relación Estructura-Actividad
2.
Brain Behav Immun ; 121: 351-364, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39089536

RESUMEN

BACKGROUND: Maternal immune activation (MIA) triggers neurobiological changes in offspring, potentially reshaping the molecular synaptic landscape, with the hippocampus being particularly vulnerable. However, critical details regarding developmental timing of these changes and whether they differ between males and females remain unclear. METHODS: We induced MIA in C57BL/6J mice on gestational day nine using the viral mimetic poly(I:C) and performed mass spectrometry-based proteomic analyses on hippocampal synaptoneurosomes of embryonic (E18) and adult (20 ± 1 weeks) MIA offspring. RESULTS: In the embryonic synaptoneurosomes, MIA led to lipid, polysaccharide, and glycoprotein metabolism pathway disruptions. In the adult synaptic proteome, we observed a dynamic shift toward transmembrane trafficking, intracellular signalling cascades, including cell death and growth, and cytoskeletal organisation. In adults, many associated pathways overlapped between males and females. However, we found distinct sex-specific enrichment of dopaminergic and glutamatergic pathways. We identified 50 proteins altered by MIA in both embryonic and adult samples (28 with the same directionality), mainly involved in presynaptic structure and synaptic vesicle function. We probed human phenome-wide association study data in the cognitive and psychiatric domains, and 49 of the 50 genes encoding these proteins were significantly associated with the investigated phenotypes. CONCLUSIONS: Our data emphasise the dynamic effects of viral-like MIA on developing and mature hippocampi and provide novel targets for study following prenatal immune challenges. The 22 proteins that changed directionality from the embryonic to adult hippocampus, suggestive of compensatory over-adaptions, are particularly attractive for future investigations.


Asunto(s)
Hipocampo , Ratones Endogámicos C57BL , Efectos Tardíos de la Exposición Prenatal , Proteoma , Sinapsis , Animales , Hipocampo/metabolismo , Femenino , Proteoma/metabolismo , Embarazo , Masculino , Ratones , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Sinapsis/metabolismo , Poli I-C/farmacología , Proteómica/métodos , Humanos
3.
World J Surg ; 47(6): 1348-1357, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36811667

RESUMEN

BACKGROUND: The ward round is an integral part of everyday surgical practice. It is a complex clinical activity that requires both sound clinical management and communication skills. This study reports the results of a consensus-building exercise on the common aspects of the general surgical ward rounds. METHODS: The consensus-building committee involving a range of stakeholders from 16 United Kingdom (UK) National Health Service trusts took part in this consensus exercise. The members discussed and suggested a series of statements concerning surgical ward round. An agreement of ≥ 70% among members was regarded as a consensus. RESULTS: Thirty-two members voted on 60 statements. There was a consensus on fifty-nine statements after the first round of voting, and one statement was modified before it reached consensus in the second round. The statements covered nine sections: a preparation phase, team allocation, multidisciplinary approach to the ward round, structure of the round, teaching considerations, confidentiality and privacy, documentation, post-round arrangements, and weekend round. There was a consensus on spending time to prepare for the round, a consultant-led round, involvement of the nursing staff, an MDT round at the beginning and end of the week, a minimum of 5 min allocated to each patient, utilisation of a round checklist, afternoon virtual round, and a clear handover and plan for the weekend. CONCLUSION: The consensus committee achieved agreement on several aspects concerning the surgical ward rounds in the UK NHS. This should help improve the care of surgical patients in the UK.


Asunto(s)
Medicina Estatal , Humanos , Consenso , Técnica Delphi , Reino Unido
4.
Bioinformatics ; 34(18): 3196-3204, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29897415

RESUMEN

Motivation: The collective and co-ordinated synaptic activity of large neuronal populations is relevant to neuronal development as well as a range of neurological diseases. Quantification of synaptically-mediated neuronal signalling permits further downstream analysis as well as potential application in target validation and in vitro screening assays. Our aim is to develop a phenotypic quantification for neuronal activity imaging data of large populations of neurons, in particular relating to the spatial component of the activity. Results: We extend the use of Markov random field (MRF) models to achieve this aim. In particular, we consider Bayesian posterior densities of model parameters in Gaussian MRFs to directly model changes in calcium fluorescence intensity rather than using spike trains. The basis of our model is defining neuron 'neighbours' by the relative spatial positions of the neuronal somata as obtained from the image data whereas previously this has been limited to defining an artificial square grid across the field of view and spike binning. We demonstrate that our spatial phenotypic quantification is applicable for both in vitro and in vivo data consisting of thousands of neurons over hundreds of time points. We show how our approach provides insight beyond that attained by conventional spike counting and discuss how it could be used to facilitate screening assays for modifiers of disease-associated defects of communication between cells. Availability and implementation: We supply the MATLAB code and data to obtain all of the results in the paper. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Modelos Neurológicos , Neuronas , Animales , Teorema de Bayes , Gatos , Fenotipo , Ratas
5.
Acta Neuropsychiatr ; 31(6): 305-315, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31364522

RESUMEN

OBJECTIVE: Despite its numerous side effects, clozapine is still the most effective antipsychotics making it an ideal reference substance to validate the efficacy of novel compounds for the treatment of schizophrenia. However, blood-brain barrier permeability for most new molecular entities is unknown, requiring central delivery. Thus, we performed a dose-finding study for chronic intracerebroventricular (icv) delivery of clozapine in mice. METHODS: Specifically, we implanted wild-type C57BL/6J mice with osmotic minipumps (Alzet) delivering clozapine at a rate of 0.15 µl/h at different concentrations (0, 3.5, 7 and 14 mg/ml, i.e. 0, 12.5, 25 and 50 µg/day). Mice were tested weekly in a modified SHIRPA paradigm, for locomotor activity in the open field and for prepulse inhibition (PPI) of the acoustic startle response (ASR) for a period of 3 weeks. RESULTS: None of the clozapine concentrations caused neurological deficits or evident gross behavioural alterations in the SHIRPA paradigm. In male mice, clozapine had no significant effect on locomotor activity or PPI of the ASR. In female mice, the 7 and 14 mg/ml dose of clozapine significantly affected both open field activity and PPI, while 3.5 mg/ml of clozapine increased PPI but had no effects on locomotor activity. CONCLUSION: Our findings indicate that 7 mg/ml may be the optimal dose for chronic icv delivery of clozapine in mice, allowing comparison to screen for novel antipsychotic compounds.


Asunto(s)
Clozapina/administración & dosificación , Clozapina/farmacología , Relación Dosis-Respuesta a Droga , Animales , Femenino , Infusiones Intraventriculares , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Inhibición Prepulso/efectos de los fármacos , Caracteres Sexuales
6.
Mol Pain ; 14: 1744806918801224, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30157705

RESUMEN

Elevated N-methyl-D-aspartate receptor activity contributes to central sensitization. Our laboratories and others recently reported that disrupting protein-protein interactions downstream of N-methyl-D-aspartate receptors suppresses pain. Specifically, disrupting binding between the enzyme neuronal nitric oxide synthase and either its upstream (postsynaptic density 95 kDa, PSD95) or downstream (e.g. nitric oxide synthase 1 adaptor protein, NOS1AP) protein partners suppressed inflammatory and/or neuropathic pain. However, the lack of a small-molecule neuronal nitric oxide synthase-NOS1AP inhibitor has hindered efforts to validate the therapeutic utility of disrupting the neuronal nitric oxide synthase-NOS1AP interface as an analgesic strategy. We, therefore, evaluated the ability of a putative small-molecule neuronal nitric oxide synthase-NOS1AP inhibitor ZLc002 to disrupt binding between neuronal nitric oxide synthase and NOS1AP using ex vivo, in vitro, and purified recombinant systems and asked whether ZLc002 would suppress inflammatory and neuropathic pain in vivo. In vitro, ZLc002 reduced co-immunoprecipitation of full-length NOS1AP and neuronal nitric oxide synthase in cultured neurons and in HEK293T cells co-expressing full-length neuronal nitric oxide synthase and NOS1AP. However, using a cell-free biochemical binding assay, ZLc002 failed to disrupt the in vitro binding between His-neuronal nitric oxide synthase1-299 and glutathione S-transferase-NOS1AP400-506, protein sequences containing the required binding domains for this protein-protein interaction, suggesting an indirect mode of action in intact cells. ZLc002 (4-10 mg/kg i.p.) suppressed formalin-evoked inflammatory pain in rats and reduced Fos protein-like immunoreactivity in the lumbar spinal dorsal horn. ZLc002 also suppressed mechanical and cold allodynia in a mouse model of paclitaxel-induced neuropathic pain. Anti-allodynic efficacy was sustained for at least four days of once daily repeated dosing. ZLc002 also synergized with paclitaxel when administered in combination to reduce breast (4T1) or ovarian (HeyA8) tumor cell line viability but did not alter tumor cell viability without paclitaxel. Our results verify that ZLc002 disrupts neuronal nitric oxide synthase-NOS1AP interaction in intact cells and demonstrate, for the first time, that systemic administration of a putative small-molecule inhibitor of neuronal nitric oxide synthase-NOS1AP suppresses inflammatory and neuropathic pain.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Antineoplásicos/efectos adversos , Inhibidores Enzimáticos/uso terapéutico , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Paclitaxel/efectos adversos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico , Neuronas , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Umbral del Dolor/efectos de los fármacos , Ratas , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
7.
J Neurosci ; 35(19): 7349-64, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25972165

RESUMEN

The protein NOS1AP/CAPON mediates signaling from a protein complex of NMDA receptor, PSD95 and nNOS. The only stroke trial for neuroprotectants that showed benefit to patients targeted this ternary complex. NOS1AP/nNOS interaction regulates small GTPases, iron transport, p38MAPK-linked excitotoxicity, and anxiety. Moreover, the nos1ap gene is linked to disorders from schizophrenia, post-traumatic stress disorder, and autism to cardiovascular disorders and breast cancer. Understanding protein interactions required for NOS1AP function, therefore, has broad implications for numerous diseases. Here we show that the interaction of NOS1AP with nNOS differs radically from the classical PDZ docking assumed to be responsible. The NOS1AP PDZ motif does not bind nNOS as measured by multiple methods. In contrast, full-length NOS1AP forms an unusually stable interaction with nNOS. We mapped the discrepancy between full-length and C-terminal PDZ motif to a novel internal region we call the ExF motif. The C-terminal PDZ motif, although neither sufficient nor necessary for binding, nevertheless promotes the stability of the complex. It therefore potentially affects signal transduction and suggests that functional interaction of nNOS with NOS1AP might be targetable at two distinct sites. We demonstrate that excitotoxic pathways can be regulated, in cortical neuron and organotypic hippocampal slice cultures from rat, either by the previously described PDZ ligand TAT-GESV or by the ExF motif-bearing region of NOS1AP, even when lacking the critical PDZ residues as long as the ExF motif is intact and not mutated. This previously unrecognized heterodivalent interaction of nNOS with NOS1AP may therefore provide distinct opportunities for pharmacological intervention in NOS1AP-dependent signaling and excitotoxicity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Animales Recién Nacidos , Células COS , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Chlorocebus aethiops , Agonistas de Aminoácidos Excitadores/farmacología , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Técnicas In Vitro , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Mutación/genética , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/genética , Técnicas de Cultivo de Órganos , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Neurosci ; 33(19): 8185-201, 2013 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-23658158

RESUMEN

Neuronal nitric oxide synthase (nNOS) and p38MAPK are strongly implicated in excitotoxicity, a mechanism common to many neurodegenerative conditions, but the intermediary mechanism is unclear. NOS1AP is encoded by a gene recently associated with sudden cardiac death, diabetes-associated complications, and schizophrenia (Arking et al., 2006; Becker et al., 2008; Brzustowicz, 2008; Lehtinen et al., 2008). Here we find it interacts with p38MAPK-activating kinase MKK3. Excitotoxic stimulus induces recruitment of NOS1AP to nNOS in rat cortical neuron culture. Excitotoxic activation of p38MAPK and subsequent neuronal death are reduced by competing with the nNOS:NOS1AP interaction and by knockdown with NOS1AP-targeting siRNAs. We designed a cell-permeable peptide that competes for the unique PDZ domain of nNOS that interacts with NOS1AP. This peptide inhibits NMDA-induced recruitment of NOS1AP to nNOS and in vivo in rat, doubles surviving tissue in a severe model of neonatal hypoxia-ischemia, a major cause of neonatal death and pediatric disability. The highly unusual sequence specificity of the nNOS:NOS1AP interaction and involvement in excitotoxic signaling may provide future opportunities for generation of neuroprotectants with high specificity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neuronas/fisiología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Análisis de Varianza , Animales , Animales Recién Nacidos , Encéfalo/citología , Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hipoxia/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , L-Lactato Deshidrogenasa/metabolismo , N-Metilaspartato/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/genética , Péptidos/farmacología , Conformación Proteica , ARN Interferente Pequeño/farmacología , Ratas , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección
9.
J Neurosci ; 32(5): 1847-58, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22302823

RESUMEN

Excitotoxicity resulting from excessive Ca(2+) influx through glutamate receptors contributes to neuronal injury after stroke, trauma, and seizures. Increased cytosolic Ca(2+) levels activate a family of calcium-dependent proteases with papain-like activity, the calpains. Here we investigated the role of calpain activation during NMDA-induced excitotoxic injury in embryonic (E16-E18) murine cortical neurons that (1) underwent excitotoxic necrosis, characterized by immediate deregulation of Ca(2+) homeostasis, a persistent depolarization of mitochondrial membrane potential (Δψ(m)), and insensitivity to bax-gene deletion, (2) underwent excitotoxic apoptosis, characterized by recovery of NMDA-induced cytosolic Ca(2+) increases, sensitivity to bax gene deletion, and delayed Δψ(m) depolarization and Ca(2+) deregulation, or (3) that were tolerant to excitotoxic injury. Interestingly, treatment with the calpain inhibitor calpeptin, overexpression of the endogenous calpain inhibitor calpastatin, or gene silencing of calpain protected neurons against excitotoxic apoptosis but did not influence excitotoxic necrosis. Calpeptin failed to exert a protective effect in bax-deficient neurons but protected bid-deficient neurons similarly to wild-type cells. To identify when calpains became activated during excitotoxic apoptosis, we monitored calpain activation dynamics by time-lapse fluorescence microscopy using a calpain-sensitive Förster resonance energy transfer probe. We observed a delayed calpain activation that occurred downstream of mitochondrial engagement and directly preceded neuronal death. In contrast, we could not detect significant calpain activity during excitotoxic necrosis or in neurons that were tolerant to excitotoxic injury. Oxygen/glucose deprivation-induced injury in organotypic hippocampal slice cultures confirmed that calpains were specifically activated during bax-dependent apoptosis and in this setting function as downstream cell-death executioners.


Asunto(s)
Apoptosis/fisiología , Calpaína/fisiología , Hipocampo/metabolismo , Proteína X Asociada a bcl-2/fisiología , Animales , Calpaína/antagonistas & inhibidores , Línea Celular Tumoral , Células Cultivadas , Dipéptidos/fisiología , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Hipocampo/efectos de los fármacos , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , N-Metilaspartato/farmacología , Técnicas de Cultivo de Órganos , Embarazo , Proteína X Asociada a bcl-2/agonistas
10.
Res Sq ; 2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37461513

RESUMEN

Maternal infections during pregnancy pose an increased risk for neurodevelopmental psychiatric disorders (NPDs) in the offspring. Here, we examined age- and sex-dependent dynamic changes of the hippocampal synaptic proteome after maternal immune activation (MIA) in embryonic and adult mice. Adult male and female MIA offspring exhibited social deficits and sex-specific depression-like behaviours, among others, validating the model. Furthermore, we observed dose-, age-, and sex-dependent synaptic proteome differences. Analysis of the embryonic synaptic proteome implicates sphingolipid and ketoacid metabolism pathway disruptions during neurodevelopment for NPD-pertinent sequelae. In the embryonic hippocampus, prenatal immune activation also led to changes in neuronal guidance, glycosphingolipid metabolism important for signalling and myelination, and post-translational modification of proteins that regulate intercellular interaction and developmental timing. In adulthood, the observed changes in synaptoneurosomes revealed a dynamic shift toward transmembrane trafficking, intracellular signalling cascades, and hormone-mediated metabolism. Importantly, 68 of the proteins with differential abundance in the embryonic brains of MIA offspring were also altered in adulthood, 75% of which retained their directionality. These proteins are involved in synaptic organisation, neurotransmitter receptor regulation, and the vesicle cycle. A cluster of persistently upregulated proteins, including AKT3, PAK1/3, PPP3CA, formed a functional network enriched in the embryonic brain that is involved in cellular responses to environmental stimuli. To infer a link between the overlapping protein alterations and cognitive and psychiatric traits, we probed human phenome-wise association study data for cognitive and psychiatric phenotypes and all, but PORCN were significantly associated with the investigated domains. Our data provide insights into the dynamic effects of an early prenatal immune activation on developing and mature hippocampi and highlights targets for early intervention in individuals exposed to such immune challenges.

11.
Eur Neuropsychopharmacol ; 67: 66-79, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36513018

RESUMEN

The neuronal isoform of nitric oxide synthase (nNOS) and its interacting protein NOS1AP have been linked to several mental disorders including schizophrenia and depression. An increase in the interaction between nNOS and NOS1AP in the frontal cortex has been suggested to contribute to the emergence of these disorders. Here we aimed to uncover whether disruption of their interactions in the frontal cortex leads to mental disorder endophenotypes. Targeting the medial prefrontal cortex (mPFC), we stereotaxically injected wild-type C57BL/6J mice with recombinant adeno-associated virus (rAAV) expressing either full-length NOS1AP, the nNOS binding region of NOS1AP (i.e. NOS1AP396-503), or the nNOS amino-terminus (i.e. nNOS1-133), which was shown to disrupt the interaction of endogenous nNOS with PSD-95. We tested these mice in a comprehensive behavioural battery, assessing different endophenotypes related to mental disorders. We found no differences in anxiety-related and exploratory behaviours. Likewise, social interaction was comparable in all groups. However, social recognition was impaired in NOS1AP and NOS1AP396-503 mice. These mice, as well as mice overexpressing nNOS1-133 also displayed impaired spatial working memory (SWM) capacity, while spatial reference memory (SRM) remained intact. Finally, mice overexpressing NOS1AP and nNOS1-133, but not NOS1AP396-503, failed to habituate to the startling pulses in an acoustic startle response (ASR) paradigm, though we found no difference in overall startle intensity or prepulse inhibition (PPI) of the ASR. Our findings indicate a distinct role of NOS1AP/nNOS/PSD-95 interactions in the mPFC to contribute to specific endophenotypic changes observed in different mental disorders.


Asunto(s)
Memoria a Corto Plazo , Reflejo de Sobresalto , Ratones , Animales , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo I/genética , Reconocimiento en Psicología , Trastornos de la Memoria , Homólogo 4 de la Proteína Discs Large/metabolismo , Corteza Prefrontal/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética
12.
Eur J Neurosci ; 33(9): 1647-55, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21453290

RESUMEN

Synaptic plasticity is regarded as the major candidate mechanism for synaptic information storage and memory formation in the hippocampus. Mitogen-activated protein kinases have recently emerged as an important regulatory factor in many forms of synaptic plasticity and memory. As one of the subfamilies of mitogen-activated protein kinases, extracellular-regulated kinase is involved in the in vitro induction of long-term potentiation (LTP), whereas p38 mediates metabotropic glutamate receptor-dependent long-term depression (LTD) in vitro. Although c-Jun N-terminal kinase (JNK) has also been implicated in synaptic plasticity, the in vivo relevance of JNK activity to different forms of synaptic plasticity remains to be further explored. We investigated the effect of inhibition of JNK on different forms of synaptic plasticity in the dentate gyrus of freely behaving adult rats. Intracereboventricular application of c-Jun N-terminal protein kinase-inhibiting peptide (D-JNKI) (96 ng), a highly selective JNK inhibitor peptide, did not affect basal synaptic transmission but reduced neuronal excitability with a higher dose (192 ng). Application of D-JNKI, at a concentration that did not affect basal synaptic transmission, resulted in reduced specific phosphorylation of the JNK substrates postsynaptic density 95kD protein (PSD 95) and c-Jun, a significant enhancement of LTD and a facilitation of short-term depression into LTD. Both LTP and short-term potentiation were unaffected. An inhibition of depotentiation (recovery of LTP) occurred. These data suggest that suppression of JNK-dependent signalling may serve to enhance synaptic depression, and indirectly promote LTP through impairment of depotentiation.


Asunto(s)
Conducta Animal/efectos de los fármacos , Hipocampo/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Potenciación a Largo Plazo/fisiología , Depresión Sináptica a Largo Plazo/fisiología , Péptidos/farmacología , Animales , Electrofisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Transmisión Sináptica/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
J Cell Biol ; 173(2): 265-77, 2006 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-16618812

RESUMEN

c-Jun NH(2)-terminal kinases (JNKs) are essential during brain development, when they regulate morphogenic changes involving cell movement and migration. In the adult, JNK determines neuronal cytoarchitecture. To help uncover the molecular effectors for JNKs in these events, we affinity purified JNK-interacting proteins from brain. This revealed that the stathmin family microtubule-destabilizing proteins SCG10, SCLIP, RB3, and RB3' interact tightly with JNK. Furthermore, SCG10 is also phosphorylated by JNK in vivo on sites that regulate its microtubule depolymerizing activity, serines 62 and 73. SCG10-S73 phosphorylation is significantly decreased in JNK1-/- cortex, indicating that JNK1 phosphorylates SCG10 in developing forebrain. JNK phosphorylation of SCG10 determines axodendritic length in cerebrocortical cultures, and JNK site-phosphorylated SCG10 colocalizes with active JNK in embryonic brain regions undergoing neurite elongation and migration. We demonstrate that inhibition of cytoplasmic JNK and expression of SCG10-62A/73A both inhibited fluorescent tubulin recovery after photobleaching. These data suggest that JNK1 is responsible for regulation of SCG10 depolymerizing activity and neurite elongation during brain development.


Asunto(s)
Axones/fisiología , Dendritas/fisiología , Microtúbulos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Proteínas de Unión al Calcio , Proteínas Portadoras , Línea Celular , Células Cultivadas , Embrión de Mamíferos/citología , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Proteínas de Microtúbulos , Proteína Quinasa 8 Activada por Mitógenos/análisis , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Fosforilación , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estatmina
14.
Nat Neurosci ; 10(4): 436-43, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17369826

RESUMEN

Excitotoxic neuronal death contributes to many neurological disorders, and involves calcium influx and stress-activated protein kinases (SAPKs) such as p38alpha. There is indirect evidence that the small Rho-family GTPases Rac and cdc42 are involved in neuronal death subsequent to the withdrawal of nerve growth factor (NGF), whereas Rho is involved in the inhibition of neurite regeneration and the release of the amyloidogenic Abeta(42) peptide. Here we show that Rho is activated in rat neurons by conditions that elevate intracellular calcium and in the mouse cerebral cortex during ischemia. Rho is required for the rapid glutamate-induced activation of p38alpha and ensuing neuronal death. The ability of RhoA to activate p38alpha was not expected, and it was specific to primary neuronal cultures. The expression of active RhoA alone not only activated p38alpha but also induced neuronal death that was sensitive to the anti-apoptotic protein Bcl-2, showing that RhoA was sufficient to induce the excitotoxic pathway. Therefore, Rho is an essential component of the excitotoxic cell death pathway.


Asunto(s)
Calcio/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Neuronas/fisiología , Proteína de Unión al GTP rhoA/fisiología , ADP Ribosa Transferasas/metabolismo , ADP Ribosa Transferasas/farmacología , Animales , Animales Recién Nacidos , Toxinas Botulínicas/metabolismo , Toxinas Botulínicas/farmacología , Encéfalo/citología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Lateralidad Funcional , Ácido Glutámico/farmacología , Isquemia/metabolismo , Isquemia/patología , Ratones , Modelos Moleculares , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Transfección/métodos , Proteína de Unión al GTP cdc42/metabolismo
15.
EBioMedicine ; 71: 103565, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34455393

RESUMEN

BACKGROUND: Nitric oxide synthase 1 adaptor protein (NOS1AP; previously named CAPON) is linked to the glutamatergic postsynaptic density through interaction with neuronal nitric oxide synthase (nNOS). NOS1AP and its interaction with nNOS have been associated with several mental disorders. Despite the high levels of NOS1AP expression in the hippocampus and the relevance of this brain region in glutamatergic signalling as well as mental disorders, a potential role of hippocampal NOS1AP in the pathophysiology of these disorders has not been investigated yet. METHODS: To uncover the function of NOS1AP in hippocampus, we made use of recombinant adeno-associated viruses to overexpress murine full-length NOS1AP or the NOS1AP carboxyterminus in the hippocampus of mice. We investigated these mice for changes in gene expression, neuronal morphology, and relevant behavioural phenotypes. FINDINGS: We found that hippocampal overexpression of NOS1AP markedly increased the interaction of nNOS with PSD-95, reduced dendritic spine density, and changed dendritic spine morphology at CA1 synapses. At the behavioural level, we observed an impairment in social memory and decreased spatial working memory capacity. INTERPRETATION: Our data provide a mechanistic explanation for a highly selective and specific contribution of hippocampal NOS1AP and its interaction with the glutamatergic postsynaptic density to cross-disorder pathophysiology. Our findings allude to therapeutic relevance due to the druggability of this molecule. FUNDING: This study was funded in part by the DFG, the BMBF, the Academy of Finland, the NIH, the Japanese Society of Clinical Neuropsychopharmacology, the Ministry of Education of the Russian Federation, and the European Community.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Endofenotipos , Expresión Génica , Hipocampo/metabolismo , Trastornos Mentales/etiología , Trastornos Mentales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Homólogo 4 de la Proteína Discs Large/metabolismo , Regulación de la Expresión Génica , Hipocampo/fisiopatología , Masculino , Trastornos Mentales/diagnóstico , Ratones , Óxido Nítrico Sintasa de Tipo I/metabolismo , Unión Proteica , Transducción de Señal
16.
J Cell Biol ; 168(1): 117-26, 2005 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-15631993

RESUMEN

The stress-activated protein kinase p38 and nitric oxide (NO) are proposed downstream effectors of excitotoxic cell death. Although the postsynaptic density protein PSD95 can recruit the calcium-dependent neuronal NO synthase (nNOS) to the mouth of the calcium-permeable NMDA receptor, and depletion of PSD95 inhibits excitotoxicity, the possibility that selective uncoupling of nNOS from PSD95 might be neuroprotective is unexplored. The relationship between excitotoxic stress-generated NO and activation of p38, and the significance of the PSD95-nNOS interaction to p38 activation also remain unclear. We find that NOS inhibitors reduce both glutamate-induced p38 activation and the resulting neuronal death, whereas NO donor has effects consistent with NO as an upstream regulator of p38 in glutamate-induced cell death. Experiments using a panel of decoy constructs targeting the PSD95-nNOS interaction suggest that this interaction and subsequent NO production are critical for glutamate-induced p38 activation and the ensuing cell death, and demonstrate that the PSD95-nNOS interface provides a genuine possibility for design of neuroprotective drugs with increased selectivity.


Asunto(s)
Muerte Celular/fisiología , Activación Enzimática , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico Sintasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Cerebelo/citología , Electrofisiología , Transferencia Resonante de Energía de Fluorescencia , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I , Estructura Terciaria de Proteína , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología
17.
Nat Commun ; 11(1): 5107, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-33037199

RESUMEN

Engineered light-dependent switches provide uniquely powerful opportunities to investigate and control cell regulatory mechanisms. Existing tools offer high spatiotemporal resolution, reversibility and repeatability. Cellular optogenetics applications remain limited with diffusible targets as the response of the actuator is difficult to independently validate. Blue light levels commonly needed for actuation can be cytotoxic, precluding long-term experiments. We describe a simple approach overcoming these obstacles. Resonance energy transfer can be used to constitutively or dynamically modulate actuation sensitivity. This simultaneously offers on-line monitoring of light-dependent switching and precise quantification of activation-relaxation properties in intact living cells. Applying this approach to different LOV2-based switches reveals that flanking sequences can lead to relaxation times up to 11-fold faster than anticipated. In situ-measured parameter values guide the design of target-inhibiting actuation trains with minimal blue-light exposure, and context-based optimisation can increase sensitivity and experimental throughput a further 10-fold without loss of temporal precision.


Asunto(s)
Imagen Molecular/métodos , Optogenética/métodos , Fototropinas/metabolismo , Animales , Transferencia de Energía , Femenino , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Hipocampo/citología , Humanos , Luz , Sistema de Señalización de MAP Quinasas , Masculino , Neuronas , Fototropinas/análisis , Fototropinas/genética , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
18.
Pain ; 159(5): 849-863, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29319606

RESUMEN

Elevated N-methyl-D-aspartate receptor (NMDAR) activity is linked to central sensitization and chronic pain. However, NMDAR antagonists display limited therapeutic potential because of their adverse side effects. Novel approaches targeting the NR2B-PSD95-nNOS complex to disrupt signaling pathways downstream of NMDARs show efficacy in preclinical pain models. Here, we evaluated the involvement of interactions between neuronal nitric oxide synthase (nNOS) and the nitric oxide synthase 1 adaptor protein (NOS1AP) in pronociceptive signaling and neuropathic pain. TAT-GESV, a peptide inhibitor of the nNOS-NOS1AP complex, disrupted the in vitro binding between nNOS and its downstream protein partner NOS1AP but not its upstream protein partner postsynaptic density 95 kDa (PSD95). Putative inactive peptides (TAT-cp4GESV and TAT-GESVΔ1) failed to do so. Only the active peptide protected primary cortical neurons from glutamate/glycine-induced excitotoxicity. TAT-GESV, administered intrathecally (i.t.), suppressed mechanical and cold allodynia induced by either the chemotherapeutic agent paclitaxel or a traumatic nerve injury induced by partial sciatic nerve ligation. TAT-GESV also blocked the paclitaxel-induced phosphorylation at Ser15 of p53, a substrate of p38 MAPK. Finally, TAT-GESV (i.t.) did not induce NMDAR-mediated motor ataxia in the rotarod test and did not alter basal nociceptive thresholds in the radiant heat tail-flick test. These observations support the hypothesis that antiallodynic efficacy of an nNOS-NOS1AP disruptor may result, at least in part, from blockade of p38 MAPK-mediated downstream effects. Our studies demonstrate, for the first time, that disrupting nNOS-NOS1AP protein-protein interactions attenuates mechanistically distinct forms of neuropathic pain without unwanted motor ataxic effects of NMDAR antagonists.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Neuralgia/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Animales , Masculino , Ratones , Neuralgia/etiología , Neuralgia/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Dimensión del Dolor , Traumatismos de los Nervios Periféricos/complicaciones , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
19.
Obes Surg ; 28(4): 963-969, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29101716

RESUMEN

BACKGROUND: Bariatric surgery offers excellent weight loss results and improvement in obesity-associated comorbidities. Many patients undergoing surgery are of working age, and so an understanding of any relationship between occupational outcomes and surgery is essential. The aim of this study was to ascertain the occupational outcomes of patients undergoing bariatric surgery at a high-volume centre. METHODS: A retrospective search was performed of a prospectively maintained consecutive electronic database. We collected data on patient demographics and employment status before and after bariatric surgery. All patients with a documented employment status within 30 months of surgery were included. Patients were divided into three groups: within 6 months post-operatively, 7-18 months post-operatively, and 19-30 months post-operatively. RESULTS: A total of 1011 patients were included. Median age was 47 years (range 18-78). Pre-operatively, 59.5% (444/746) were employed compared to 69.9% (707/1011) post-operatively (p < 0.05). The number of unemployed fell from 36.6% (273/746) pre-operatively to 21% (212/1011) post-operatively. The improvement in employment status was seen at all durations of follow-up. For those in employment pre-operatively, approximately 90% were still in employment at each subsequent follow-up. For those patients who were unemployed pre-operatively, approximately 40% were in employment at each subsequent follow-up. A significant improvement in the percentage employed was seen in all working age groups (p < 0.05). CONCLUSION: This is the largest study worldwide looking at employment outcomes following bariatric surgery. It demonstrates a significant increase in number of employed patients following bariatric surgery. Interestingly, it also showed that some patients employed pre-operatively become unemployed afterwards.


Asunto(s)
Cirugía Bariátrica/rehabilitación , Empleo , Obesidad Mórbida/cirugía , Ocupaciones , Adolescente , Adulto , Anciano , Bases de Datos Factuales , Empleo/estadística & datos numéricos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Obesidad Mórbida/epidemiología , Ocupaciones/estadística & datos numéricos , Periodo Posoperatorio , Calidad de Vida , Estudios Retrospectivos , Reinserción al Trabajo/estadística & datos numéricos , Resultado del Tratamiento , Desempleo/estadística & datos numéricos , Pérdida de Peso/fisiología , Adulto Joven
20.
Mol Cell Biol ; 23(17): 6027-36, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12917327

RESUMEN

Lithium has been used as an effective mood-stabilizing drug for the treatment of manic episodes and depression for 50 years. More recently, lithium has been found to protect neurons from death induced by a wide array of neurotoxic insults. However, the molecular basis for the prophylactic effects of lithium have remained obscure. A target of lithium, glycogen synthase kinase 3 (GSK-3), is implicated in neuronal death after trophic deprivation. The mechanism whereby GSK-3 exerts its neurotoxic effects is also unknown. Here we show that lithium blocks the canonical c-Jun apoptotic pathway in cerebellar granule neurons deprived of trophic support. This effect is mimicked by the structurally independent inhibitors of GSK-3, FRAT1, and indirubin. Like lithium, these prevent the stress induced c-Jun protein increase and subsequent apoptosis. These events are downstream of c-Jun transactivation, since GSK-3 inhibitors block neuronal death induced by constitutively active c-Jun (Ser/Thr-->Asp) and FRAT1 expression inhibits AP1 reporter activity. Consistent with this, AP1-dependent expression of proapoptotic Bim requires GSK-3-like activity. These data suggest that a GSK-3-like kinase acts in tandem with c-Jun N-terminal kinase to coordinate the full execution of the c-Jun stress response and neuronal death in response to trophic deprivation.


Asunto(s)
Glucógeno Sintasa Quinasa 3/efectos de los fármacos , Litio/farmacología , Proteínas de la Membrana , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas de Neoplasias , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Reguladoras de la Apoptosis , Proteína 11 Similar a Bcl2 , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Cerebelo/citología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Genes Reporteros , Glucógeno Sintasa Quinasa 3/metabolismo , Indoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Ratones Mutantes , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuronas/metabolismo , Oximas/farmacología , Monoéster Fosfórico Hidrolasas/efectos de los fármacos , Monoéster Fosfórico Hidrolasas/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Estrés Fisiológico , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA