Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(26): eadl0030, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38924398

RESUMEN

How can short-lived molecules selectively maintain the potentiation of activated synapses to sustain long-term memory? Here, we find kidney and brain expressed adaptor protein (KIBRA), a postsynaptic scaffolding protein genetically linked to human memory performance, complexes with protein kinase Mzeta (PKMζ), anchoring the kinase's potentiating action to maintain late-phase long-term potentiation (late-LTP) at activated synapses. Two structurally distinct antagonists of KIBRA-PKMζ dimerization disrupt established late-LTP and long-term spatial memory, yet neither measurably affects basal synaptic transmission. Neither antagonist affects PKMζ-independent LTP or memory that are maintained by compensating PKCs in ζ-knockout mice; thus, both agents require PKMζ for their effect. KIBRA-PKMζ complexes maintain 1-month-old memory despite PKMζ turnover. Therefore, it is not PKMζ alone, nor KIBRA alone, but the continual interaction between the two that maintains late-LTP and long-term memory.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Potenciación a Largo Plazo , Ratones Noqueados , Proteína Quinasa C , Animales , Proteína Quinasa C/metabolismo , Proteína Quinasa C/genética , Ratones , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Memoria/fisiología , Memoria a Largo Plazo/fisiología , Sinapsis/metabolismo , Sinapsis/fisiología , Unión Proteica , Fosfoproteínas
2.
PLoS One ; 19(1): e0295504, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38166102

RESUMEN

BACKGROUND: Stroke is a major cause of death, disability, and public health problems. Its intervention is limited to early treatment with thrombolytics and/or endovascular clot removal with mechanical thrombectomy without any available subacute or chronic neuroprotective treatments. RNS60 has reduced neuroinflammation and increased neuronal survival in several animal models of neurodegeneration and trauma. The aim here was to evaluate whether RNS60 protects the brain and cognitive function in a mouse stroke model. METHODS: Male C57BL/6J mice were subjected to sham or ischemic stroke surgery using 60-minute transient middle cerebral artery occlusion (tMCAo). In each group, mice received blinded daily administrations of RNS60 or control fluids (PNS60 or normal saline [NS]), beginning 2 hours after surgery over 13 days. Multiple neurobehavioral tests were conducted (Neurological Severity Score [mNSS], Novel Object Recognition [NOR], Active Place Avoidance [APA], and the Conflict Variant of APA [APAc]). On day 14, cortical microvascular perfusion (MVP) was measured, then brains were removed and infarct volume, immunofluorescence of amyloid beta (Aß), neuronal density, microglial activation, and white matter damage/myelination were measured. SPSS was used for analysis (e.g., ANOVA for parametric data; Kruskal Wallis for non-parametric data; with post-hoc analysis). RESULTS: Thirteen days of treatment with RNS60 reduced brain infarction, amyloid pathology, neuronal death, microglial activation, white matter damage, and increased MVP. RNS60 reduced brain pathology and resulted in behavioral improvements in stroke compared to sham surgery mice (increased memory-learning in NOR and APA, improved cognitive flexibility in APAc). CONCLUSION: RNS60-treated mice exhibit significant protection of brain tissue and improved neurobehavioral functioning after tMCAo-stroke. Additional work is required to determine mechanisms, time-window of dosing, and multiple dosing volumes durations to support clinical stroke research.


Asunto(s)
Isquemia Encefálica , Ataque Isquémico Transitorio , Fármacos Neuroprotectores , Accidente Cerebrovascular , Ratones , Masculino , Animales , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Péptidos beta-Amiloides , Ratones Endogámicos C57BL , Accidente Cerebrovascular/patología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad
3.
bioRxiv ; 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38260257

RESUMEN

The rodent hippocampus is a spatially organized neuronal network that supports the formation of spatial and episodic memories. We conducted bulk RNA sequencing and spatial transcriptomics experiments to measure gene expression changes in the dorsal hippocampus following the recall of active place avoidance (APA) memory. Through bulk RNA sequencing, we examined the gene expression changes following memory recall across the functionally distinct subregions of the dorsal hippocampus. We found that recall induced differentially expressed genes (DEGs) in the CA1 and CA3 hippocampal subregions were enriched with genes involved in synaptic transmission and synaptic plasticity, while DEGs in the dentate gyrus (DG) were enriched with genes involved in energy balance and ribosomal function. Through spatial transcriptomics, we examined gene expression changes following memory recall across an array of spots encompassing putative memory-associated neuronal ensembles marked by the expression of the IEGs Arc, Egr1, and c-Jun. Within samples from both trained and untrained mice, the subpopulations of spatial transcriptomic spots marked by these IEGs were transcriptomically and spatially distinct from one another. DEGs detected between Arc+ and Arc- spots exclusively in the trained mouse were enriched in several memory-related gene ontology terms, including "regulation of synaptic plasticity" and "memory." Our results suggest that APA memory recall is supported by regionalized transcriptomic profiles separating the CA1 and CA3 from the DG, transcriptionally and spatially distinct IEG expressing spatial transcriptomic spots, and biological processes related to synaptic plasticity as a defining the difference between Arc+ and Arc- spatial transcriptomic spots.

4.
Commun Biol ; 5(1): 1074, 2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-36209301

RESUMEN

Dysregulation of sphingomyelin and ceramide metabolism have been implicated in Alzheimer's disease. Genome-wide and transcriptome-wide association studies have identified various genes and genetic variants in lipid metabolism that are associated with Alzheimer's disease. However, the molecular mechanisms of sphingomyelin and ceramide disruption remain to be determined. We focus on the sphingolipid pathway and carry out multi-omics analyses to identify central and peripheral metabolic changes in Alzheimer's patients, correlating them to imaging features. Our multi-omics approach is based on (a) 2114 human post-mortem brain transcriptomics to identify differentially expressed genes; (b) in silico metabolic flux analysis on context-specific metabolic networks identified differential reaction fluxes; (c) multimodal neuroimaging analysis on 1576 participants to associate genetic variants in sphingomyelin pathway with Alzheimer's disease pathogenesis; (d) plasma metabolomic and lipidomic analysis to identify associations of lipid species with dysregulation in Alzheimer's; and (e) metabolite genome-wide association studies to define receptors within the pathway as a potential drug target. We validate our hypothesis in amyloidogenic APP/PS1 mice and show prolonged exposure to fingolimod alleviated synaptic plasticity and cognitive impairment in mice. Our integrative multi-omics approach identifies potential targets in the sphingomyelin pathway and suggests modulators of S1P metabolism as possible candidates for Alzheimer's disease treatment.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Ceramidas , Clorhidrato de Fingolimod , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Esfingolípidos/metabolismo , Esfingolípidos/uso terapéutico , Esfingomielinas/uso terapéutico
5.
Mol Cell Neurosci ; 120: 103716, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35276347

RESUMEN

Pelizaeus-Merzbacher-like disease type 1 (PMLD1) is a hypomyelinating disorder arising in patients with mutations in GJC2, encoding Connexin47 (Cx47). PMLD1 causes nystagmus, cerebellar ataxia, spasticity and changes in CNS white matter detected by MRI. At least one mutation (p.I33M) yields a much milder phenotype, spastic paraplegia type 44 (SPG44). Cx47 contributes to gap junction communication channels between oligodendrocytes (OLs), the myelinating cells in the central nervous system (CNS), and between OLs and astrocytes. Prior studies in cell lines have shown that PMLD1 mutants such as p.P87S display defective protein trafficking, intracellular retention in the ER and loss-of-function. Here we show that when expressed in primary OLs, three PMLD1 associated mutants (p.P87S, p.Y269D and p.M283T) show ER retention of Cx47 and evidence of activation of the cellular stress (unfolded protein response, UPR) and apoptotic pathways. On the other hand, the milder SPG44 associated mutation p.I33M shows a wild-type-like subcellular distribution and no activation of the UPR or apoptotic pathways. These studies provide new insight into a potential element of toxic gain of function underlying the mechanism of PMLD1 that should help guide future therapeutic approaches.


Asunto(s)
Enfermedades Desmielinizantes , Enfermedades por Almacenamiento Lisosomal , Enfermedades Neurodegenerativas , Enfermedad de Pelizaeus-Merzbacher , Conexinas/genética , Conexinas/metabolismo , Enfermedades Desmielinizantes/metabolismo , Uniones Comunicantes/genética , Uniones Comunicantes/metabolismo , Humanos , Enfermedades por Almacenamiento Lisosomal/metabolismo , Mutación , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Pelizaeus-Merzbacher/genética , Enfermedad de Pelizaeus-Merzbacher/metabolismo , Respuesta de Proteína Desplegada/genética
6.
Eur J Neurosci ; 54(8): 6795-6814, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33540466

RESUMEN

PKMζ is an autonomously active PKC isoform crucial for the maintenance of synaptic long-term potentiation (LTP) and long-term memory. Unlike other kinases that are transiently stimulated by second messengers, PKMζ is persistently activated through sustained increases in protein expression of the kinase. Therefore, visualizing increases in PKMζ expression during long-term memory storage might reveal the sites of its persistent action and thus the location of memory-associated LTP maintenance in the brain. Using quantitative immunohistochemistry validated by the lack of staining in PKMζ-null mice, we examined the amount and distribution of PKMζ in subregions of the hippocampal formation of wild-type mice during LTP maintenance and spatial long-term memory storage. During LTP maintenance in hippocampal slices, PKMζ increases in the pyramidal cell body and stimulated dendritic layers of CA1 for at least 2 hr. During spatial memory storage, PKMζ increases in CA1 pyramidal cells for at least 1 month, paralleling the persistence of the memory. During the initial expression of the memory, we tagged principal cells with immediate-early gene Arc promoter-driven transcription of fluorescent proteins. The subset of memory-tagged CA1 cells selectively increases expression of PKMζ during memory storage, and the increase persists in dendritic compartments within stratum radiatum for 1 month, indicating long-term storage of information in the CA3-to-CA1 pathway. We conclude that persistent increases in PKMζ trace the molecular mechanism of LTP maintenance and thus the sites of information storage within brain circuitry during long-term memory.


Asunto(s)
Potenciación a Largo Plazo , Proteína Quinasa C , Animales , Hipocampo/metabolismo , Memoria a Largo Plazo , Ratones , Neuronas/metabolismo , Proteína Quinasa C/metabolismo , Memoria Espacial
7.
Brain Commun ; 2(2): fcaa132, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33215083

RESUMEN

Small vessel cerebrovascular disease, visualized as white matter hyperintensities on T2-weighted magnetic resonance imaging, contributes to the clinical presentation of Alzheimer's disease. However, the extent to which cerebrovascular disease represents an independent pathognomonic feature of Alzheimer's disease or directly promotes Alzheimer's pathology is unclear. The purpose of this study was to examine the association between white matter hyperintensities and plasma levels of tau and to determine if white matter hyperintensities and tau levels interact to predict Alzheimer's disease diagnosis. To confirm that cerebrovascular disease promotes tau pathology, we examined tau fluid biomarker concentrations and pathology in a mouse model of ischaemic injury. Three hundred ninety-one participants from the Alzheimer's Disease Neuroimaging Initiative (74.5 ± 7.1 years of age) were included in this cross-sectional analysis. Participants had measurements of plasma total-tau, cerebrospinal fluid beta-amyloid, and white matter hyperintensities, and were diagnosed clinically as Alzheimer's disease (n = 97), mild cognitive impairment (n = 186) or cognitively normal control (n = 108). We tested the relationship between plasma tau concentration and white matter hyperintensity volume across diagnostic groups. We also examined the extent to which white matter hyperintensity volume, plasma tau, amyloid positivity status and the interaction between white matter hyperintensities and plasma tau correctly classifies diagnostic category. Increased white matter hyperintensity volume was associated with higher plasma tau concentration, particularly among those diagnosed clinically with Alzheimer's disease. Presence of brain amyloid and the interaction between plasma tau and white matter hyperintensity volume distinguished Alzheimer's disease and mild cognitive impairment participants from controls with 77.6% and 63.3% accuracy, respectively. In 63 Alzheimer's Disease Neuroimaging Initiative participants who came to autopsy (82.33 ± 7.18 age at death), we found that higher degrees of arteriosclerosis were associated with higher Braak staging, indicating a positive relationship between cerebrovascular disease and neurofibrillary pathology. In a transient middle cerebral artery occlusion mouse model, aged mice that received transient middle cerebral artery occlusion, but not sham surgery, had increased plasma and cerebrospinal fluid tau concentrations, induced myelin loss, and hyperphosphorylated tau pathology in the ipsilateral hippocampus and cerebral hemisphere. These findings demonstrate a relationship between cerebrovascular disease, operationalized as white matter hyperintensities, and tau levels, indexed in the plasma, suggesting that hypoperfusive injury promotes tau pathology. This potential causal association is supported by the demonstration that transient cerebral artery occlusion induces white matter damage, increases biofluidic markers of tau, and promotes cerebral tau hyperphosphorylation in older-adult mice.

8.
Neural Plast ; 2019: 4383258, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31827497

RESUMEN

Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear protein that regulates gene expression through poly(ADP)-ribosylation, resulting in the loosening of chromatin structure. PARP-1 enzymatic activity has been shown to be necessary for the expression of several genes required for memory formation and consolidation. Previously, we showed that nucleolar PARP-1 is significantly decreased in hippocampal pyramidal cells in Alzheimer's disease (AD). We proposed that the displacement of PARP-1 from the nucleolus results in downregulation of new rRNA expression and ribosome biogenesis, leading to cognitive impairment. To further investigate the relationship between nucleolar PARP-1 and memory impairment, we examined PARP-1 expression in the hippocampi of individuals with mild cognitive impairment (MCI) compared to control and AD cases. We used immunohistochemical techniques to examine the nucleolar distribution of PARP-1 in the Cornu Ammonis (CA region) of the hippocampus. PARP-1 positive cells were then scored for the presence or absence of PARP-1 in the nucleolus. We found a significant decrease of PARP-1 staining in the nucleolar compartment of hippocampal pyramidal cells in MCI compared with Control and AD. When the four CA (CA1-4) regions were considered separately, only the CA1 region showed significant differences in nucleolar PARP-1 with Control > AD > MCI cases. Categorization of nucleolar PARP-1 into "distinct" and "diffuse" groups suggest that most of the changes occur within the distinct group. In addition, measurements of the nucleolar diameter of nucleolar PARP-1 positive cells in CA2 and CA4 showed Control > MCI. Thus, MCI cases had a lower percentage of PARP-1 nucleolar positive cells in CA1 and smaller nucleolar diameters in CA2 and CA4, compared to Control. Our data suggest that disruption of nucleolar form and function is an early and important step in the progression of cognitive impairment.


Asunto(s)
Disfunción Cognitiva/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/metabolismo , Cognición/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo
9.
PLoS One ; 13(10): e0203374, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30281601

RESUMEN

The transition from short-term to long-term forms of synaptic plasticity requires protein synthesis and new gene expression. Most efforts to understand experience-induced changes in neuronal gene expression have focused on the transcription products of RNA polymerase II-primarily mRNAs and the proteins they encode. We recently showed that nucleolar integrity and activity-dependent ribosomal RNA (rRNA) synthesis are essential for the maintenance of hippocampal long-term potentiation (LTP). Consequently, the synaptic plasticity and memory hypothesis predicts that nucleolar integrity and activity dependent rRNA synthesis would be required for Long-term memory (LTM). We tested this prediction using the hippocampus-dependent, Active Place Avoidance (APA) spatial memory task and found that training induces de novo rRNA synthesis in mouse dorsal hippocampus. This learning-induced increase in nucleolar activity and rRNA synthesis persists at least 24 h after training. In addition, intra-hippocampal injection of the Pol I specific inhibitor, CX-5461 prior to training, revealed that de novo rRNA synthesis is required for 24 h memory, but not for learning. Using qPCR to assess activity-dependent changes in gene expression, we found that of seven known rRNA expression variants (v-rRNAs), only one, v-rRNA IV, is significantly upregulated right after training. These data indicate that learning induced v-rRNAs are crucial for LTM, and constitute the first evidence that differential rRNA gene expression plays a role in memory.


Asunto(s)
Regulación de la Expresión Génica/genética , Aprendizaje/fisiología , Memoria/fisiología , ARN Ribosómico/genética , Animales , Hipocampo/metabolismo , Consolidación de la Memoria/fisiología , Pruebas de Memoria y Aprendizaje , Memoria a Largo Plazo , Ratones , Plasticidad Neuronal/genética , Sinapsis/genética , Sinapsis/fisiología
10.
Neurobiol Learn Mem ; 138: 135-144, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27417578

RESUMEN

PKMζ is an autonomously active PKC isoform that is thought to maintain both LTP and long-term memory. Whereas persistent increases in PKMζ protein sustain the kinase's action in LTP, the molecular mechanism for the persistent action of PKMζ during long-term memory has not been characterized. PKMζ inhibitors disrupt spatial memory when introduced into the dorsal hippocampus from 1day to 1month after training. Therefore, if the mechanisms of PKMζ's persistent action in LTP maintenance and long-term memory were similar, persistent increases in PKMζ would last for the duration of the memory, far longer than most other learning-induced gene products. Here we find that spatial conditioning by aversive active place avoidance or appetitive radial arm maze induces PKMζ increases in dorsal hippocampus that persist from 1day to 1month, coinciding with the strength and duration of memory retention. Suppressing the increase by intrahippocampal injections of PKMζ-antisense oligodeoxynucleotides prevents the formation of long-term memory. Thus, similar to LTP maintenance, the persistent increase in the amount of autonomously active PKMζ sustains the kinase's action during long-term and remote spatial memory maintenance.


Asunto(s)
Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Memoria a Largo Plazo/fisiología , Proteína Quinasa C/metabolismo , Memoria Espacial/fisiología , Animales , Reacción de Prevención/fisiología , Condicionamiento Operante/fisiología , Potenciales Postsinápticos Excitadores , Masculino , Ratas , Ratas Long-Evans , Retención en Psicología/fisiología
11.
Neural Plast ; 2016: 8987928, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27034851

RESUMEN

Synaptic dysfunction is thought to play a major role in memory impairment in Alzheimer's disease (AD). PARP-1 has been identified as an epigenetic regulator of plasticity and memory. Thus, we hypothesize that PARP-1 may be altered in postmortem hippocampus of individuals with AD compared to age-matched controls without neurologic disease. We found a reduced level of PARP-1 nucleolar immunohistochemical staining in hippocampal pyramidal cells in AD. Nucleolar PARP-1 staining ranged from dispersed and less intense to entirely absent in AD compared to the distinct nucleolar localization in hippocampal pyramidal neurons in controls. In cases of AD, the percentage of hippocampal pyramidal cells with nucleoli that were positive for both PARP-1 and the nucleolar marker fibrillarin was significantly lower than in controls. PARP-1 nucleolar expression emerges as a sensitive marker of functional changes in AD and suggests a novel role for PARP-1 dysregulation in AD pathology.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Nucléolo Celular/metabolismo , Cognición/fisiología , Epigénesis Genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Ribosómico/metabolismo , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Células Piramidales/metabolismo
12.
Commun Integr Biol ; 8(2): e1017163, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26479998

RESUMEN

Widely thought to be a housekeeping process, the regulation and synthesis of rRNA emerges as a potentially central mechanism for the maintenance of synaptic plasticity and memory. We have recently shown that an essential component of late-phase synaptic plasticity is rRNA biosynthesis - the rate-limiting step in the production of new ribosomes. We hypothesize that a particular population of ribosomes is generated upon learning-associated neural activity to alter the rate of synthesis of plasticity factors at tagged synapses that will support the maintenance of synaptic plasticity and memory.

13.
PLoS One ; 9(8): e104364, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25089620

RESUMEN

Long-term memory (LTM) formation requires new protein synthesis and new gene expression. Based on our work in Aplysia, we hypothesized that the rRNA genes, stimulation-dependent targets of the enzyme Poly(ADP-ribose) polymerase-1 (PARP-1), are primary effectors of the activity-dependent changes in synaptic function that maintain synaptic plasticity and memory. Using electrophysiology, immunohistochemistry, pharmacology and molecular biology techniques, we show here, for the first time, that the maintenance of forskolin-induced late-phase long-term potentiation (L-LTP) in mouse hippocampal slices requires nucleolar integrity and the expression of new rRNAs. The activity-dependent upregulation of rRNA, as well as L-LTP expression, are poly(ADP-ribosyl)ation (PAR) dependent and accompanied by an increase in nuclear PARP-1 and Poly(ADP) ribose molecules (pADPr) after forskolin stimulation. The upregulation of PARP-1 and pADPr is regulated by Protein kinase A (PKA) and extracellular signal-regulated kinase (ERK)--two kinases strongly associated with long-term plasticity and learning and memory. Selective inhibition of RNA Polymerase I (Pol I), responsible for the synthesis of precursor rRNA, results in the segmentation of nucleoli, the exclusion of PARP-1 from functional nucleolar compartments and disrupted L-LTP maintenance. Taken as a whole, these results suggest that new rRNAs (28S, 18S, and 5.8S ribosomal components)--hence, new ribosomes and nucleoli integrity--are required for the maintenance of long-term synaptic plasticity. This provides a mechanistic link between stimulation-dependent gene expression and the new protein synthesis known to be required for memory consolidation.


Asunto(s)
Potenciación a Largo Plazo/genética , Memoria a Largo Plazo/fisiología , Plasticidad Neuronal/genética , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Sinapsis/genética , Animales , Colforsina/administración & dosificación , Proteínas Quinasas Dependientes de AMP Cíclico/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Plasticidad Neuronal/fisiología , Poli(ADP-Ribosa) Polimerasa-1 , Poli Adenosina Difosfato Ribosa/biosíntesis , Poli Adenosina Difosfato Ribosa/genética , Poli(ADP-Ribosa) Polimerasas/genética , ARN Ribosómico 28S/biosíntesis , ARN Ribosómico 28S/genética , Sinapsis/fisiología
14.
Philos Trans R Soc Lond B Biol Sci ; 369(1633): 20130140, 2014 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-24298142

RESUMEN

In contrast to protein kinases that participate in long-term potentiation (LTP) induction and memory consolidation, the autonomously active atypical protein kinase C isoform, protein kinase Mzeta (PKMζ), functions in the core molecular mechanism of LTP maintenance and long-term memory storage. Here, using multiple complementary techniques for light and electron microscopic immunolocalization, we present the first detailed characterization of the cellular and subcellular distribution of PKMζ in rat hippocampus and neocortex. We find that PKMζ is widely expressed in forebrain with prominent immunostaining in hippocampal and neocortical grey matter, and weak label in white matter. In hippocampal and cortical pyramidal cells, PKMζ expression is predominantly somatodendritic, and electron microscopy highlights the kinase at postsynaptic densities and in clusters within spines. In addition, nuclear label and striking punctate immunopositive structures in a paranuclear and dendritic distribution are seen by confocal microscopy, occasionally at dendritic bifurcations. PKMζ immunoreactive granules are observed by electron microscopy in cell bodies and dendrites, including endoplasmic reticulum. The widespread distribution of PKMζ in nuclei, nucleoli and endoplasmic reticulum suggests potential roles of this kinase in cell-wide mechanisms involving gene expression, biogenesis of ribosomes and new protein synthesis. The localization of PKMζ within postsynaptic densities and spines suggests sites where the kinase stores information during LTP maintenance and long-term memory.


Asunto(s)
Encéfalo/metabolismo , Densidad Postsináptica/metabolismo , Proteína Quinasa C/metabolismo , Células Piramidales/metabolismo , Animales , Encéfalo/citología , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Microscopía Inmunoelectrónica , Ratas , Ratas Sprague-Dawley
15.
J Neurosci ; 29(30): 9553-62, 2009 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-19641118

RESUMEN

Activity-dependent long-term synaptic plasticity requires gene expression and protein synthesis. Identifying essential genes and studying their transcriptional and translational regulation are key steps to understanding how synaptic changes become long lasting. Recently, the enzyme poly-(ADP-ribose) polymerase 1 (PARP-1) was shown to be necessary for long-term memory (LTM) in Aplysia. Since PARP-1 decondenses chromatin, we hypothesize that this enzyme regulates the expression of specific genes essential for long-term synaptic plasticity that underlies LTM. We cloned Aplysia PARP-1 (ApPARP-1) and determined that its expression in sensory neurons is necessary for serotonin (5-HT)-mediated long-term facilitation (LTF) of sensorimotor neuron synapses. PARP enzymatic activity is also required, since transient application of PARP inhibitors blocked LTF. Differential display and RNA analysis of ganglia dissected from intact animals exposed to 5-HT identified the ribosomal RNA genes as PARP-dependent effector genes. The increase in the expression of rRNAs is long lasting and dynamic. Pulse-labeling RNA studies showed a PARP-dependent increase in rRNAs but not in the total RNA 24 h after 5-HT treatment. Moreover, the expression of both the AprpL27a (Aplysia ribosomal protein L27a) and the ApE2N (Aplysia ubiquitin-conjugating enzyme E2N) mRNAs also increased after 5-HT. Thus, our results suggest that 5-HT, in part by regulating PARP-1 activity, alters the expression of transcripts required for the synthesis of new ribosomes necessary for LTF.


Asunto(s)
Potenciación a Largo Plazo/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Células Receptoras Sensoriales/fisiología , Serotonina/metabolismo , Animales , Aplysia , Secuencia de Bases , Benzamidas/administración & dosificación , Células Cultivadas , Inhibidores Enzimáticos/administración & dosificación , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Datos de Secuencia Molecular , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/enzimología , Neuronas Motoras/fisiología , Fenantrenos/administración & dosificación , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , ARN Mensajero/metabolismo , ARN Ribosómico/metabolismo , Proteínas Ribosómicas/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/enzimología , Sinapsis/efectos de los fármacos , Sinapsis/enzimología , Sinapsis/fisiología , Factores de Tiempo , Enzimas Ubiquitina-Conjugadoras/metabolismo
16.
Brain Res ; 1251: 53-64, 2009 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19056362

RESUMEN

Expression, localization and regulation of different cAMP-dependent protein kinase A (PKA) subunits account for specificity in the intracellular cAMP/PKA signaling pathway. In Aplysia neurons, two classes of PKA (I and II) differing in their regulatory (R) subunits have been characterized. Type I is mostly soluble in the cell body, and type II enriched at the synaptic endings. Although both types are necessary for long-term changes in synaptic plasticity, their differences in cellular localization and expression suggest that they mediate distinct functions. By photoaffinity labeling studies, we previously observed a cAMP-binding 105 kDa band in extracts from Aplysia neurons as a putative third class of R subunit of PKA. Here, we have determined that the 105 kDa band is a high molecular weight complex (HMWC) containing alpha/beta-tubulin and PKA RI, but not RII. This hetero-complex is conserved in vertebrates since mouse brain extracts also contain it. The enrichment of the endogenous HMWC by subcellular fractionation and its synthesis in vitro indicate that it is mainly produced in the cytosol, and then transported to the synapses. The HMWC is functional as a cAMP-sensitive regulatory subunit of PKA since it binds catalytic subunit in the absence of cAMP. Furthermore, serotonin (5-HT) treatment, which produces long-term facilitation in neurons, induced its degradation. In mouse brain RI co-localized with tubulin in neuropils and in COS-7 cells discretely at the cell membrane. These observations suggest that the alpha/beta-tubulin anchoring type I PKA may have an important role in the formation of long-term synaptic plasticity.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Aplysia/enzimología , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/metabolismo , Ganglios de Invertebrados/enzimología , Neuronas/enzimología , Tubulina (Proteína)/metabolismo , Animales , Aplysia/citología , Células COS , Sistema Nervioso Central/citología , Sistema Nervioso Central/enzimología , Chlorocebus aethiops , Subunidad RIIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Ganglios de Invertebrados/citología , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Ratones , Peso Molecular , Plasticidad Neuronal/fisiología , Roedores/metabolismo , Especificidad de la Especie , Fracciones Subcelulares
17.
Biochem Biophys Res Commun ; 359(3): 563-7, 2007 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-17548057

RESUMEN

PKA type I and type II are activated in Aplysia neurons by stimulation with serotonin (5-HT), which causes long-term facilitation (LTF). The proteolysis of the regulatory subunit (R) is thought important for the persistent activation of PKA, which is necessary to produce LTF. In this study, we report that the type I regulatory subunit (RI) and type II regulatory subunit (RII) are differentially regulated by proteolytic cleavage. RI, but not RII, was selectively cleaved after 5-HT treatment for 2h in Aplysia neurons. Interestingly, the proteasome inhibitor MG132 inhibited the cleavage of RI caused by 5-HT treatment in Aplysia neuron. Besides extracts from Aplysia ganglia treated with 5-HT cleaved (35)S-labeled RI synthesized in vitro, but not (35)S-labeled RII. This suggests that 5-HT induces the activation state of RI-specific proteolytic cleavage.


Asunto(s)
Aplysia/efectos de los fármacos , Aplysia/enzimología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neuronas/efectos de los fármacos , Neuronas/enzimología , Serotonina/farmacología , Animales , Extractos Celulares , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína Quinasa Tipo II Dependiente de AMP Cíclico , Fosforilación , Subunidades de Proteína/metabolismo , Tubulina (Proteína)/metabolismo
18.
J Neuropathol Exp Neurol ; 65(4): 319-26, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16691113

RESUMEN

Protein kinase Mzeta (PKMzeta), an atypical protein kinase C (PKC) isoform, plays a key role in the maintenance of long-term potentiation (LTP), a persistent enhancement of AMPA receptor-mediated synaptic transmission, as well as in the persistence of memory in Drosophila. Because memory impairment in Alzheimer disease (AD) has been attributed to disruption of synaptic plasticity, we investigated the expression and distribution of PKMzeta in this disorder. We found that PKMzeta accumulated in neurofibrillary tangles (NFTs), whereas conventional and novel PKC isoforms did not. Unlike tau, which is present in all NFTs regardless of location, PKMzeta was found in a subset of NFTs restricted to limbic or medial temporal lobe structures (i.e. hippocampal formation, entorhinal cortex, and amygdala), areas implicated in memory loss in AD. Interestingly, PKMzeta was not identified in any NFTs in control brains derived from 6 elderly individuals without known cognitive impairment. In medial temporal lobe structures in AD, PKMzeta also occurred within abnormal neurites expressing MAP2, GluR1 and GluR2 as well as in perisomatic granules expressing GluR1 and GluR2, suggesting that aggregation of PKMzeta disrupts glutamatergic synaptic transmission. Together, these findings suggest a link between PKMzeta-mediated synaptic plasticity and memory impairment in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Sistema Límbico/metabolismo , Proteína Quinasa C/biosíntesis , Receptores AMPA/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Western Blotting , Femenino , Humanos , Inmunohistoquímica , Isoenzimas/biosíntesis , Sistema Límbico/patología , Potenciación a Largo Plazo/fisiología , Masculino , Microscopía Confocal , Persona de Mediana Edad , Neuritas/metabolismo , Neuritas/patología , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología
19.
J Biol Chem ; 278(41): 40305-16, 2003 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-12857744

RESUMEN

Protein kinase M zeta (PKM zeta) is a newly described form of PKC that is necessary and sufficient for the maintenance of hippocampal long term potentiation (LTP) and the persistence of memory in Drosophila. PKM zeta is the independent catalytic domain of the atypical PKC zeta isoform and produces long term effects at synapses because it is persistently active, lacking autoinhibition from the regulatory domain of PKC zeta. PKM has been thought of as a proteolytic fragment of PKC. Here we report that brain PKM zeta is a new PKC isoform, synthesized from a PKM zeta mRNA encoding a PKC zeta catalytic domain without a regulatory domain. Multiple zeta-specific antisera show that PKM zeta is expressed in rat forebrain as the major form of zeta in the near absence of full-length PKC zeta. A PKC zeta knockout mouse, in which the regulatory domain was disrupted and catalytic domain spared, still expresses brain PKM zeta, indicating that this form of PKM is not a PKC zeta proteolytic fragment. Furthermore, the distribution of brain PKM zeta does not correlate with PKC zeta mRNA but instead with an alternate zeta RNA transcript thought incapable of producing protein. In vitro translation of this RNA, however, generates PKM zeta of the same molecular weight as that in brain. Metabolic labeling of hippocampal slices shows increased de novo synthesis of PKM zeta in LTP. Because PKM zeta is a kinase synthesized in an autonomously active form and is necessary and sufficient for maintaining LTP, it serves as an example of a link coupling gene expression directly to synaptic plasticity.


Asunto(s)
Encéfalo/enzimología , Memoria/fisiología , Proteína Quinasa C/biosíntesis , Proteína Quinasa C/genética , ARN Mensajero/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Dominio Catalítico/genética , ADN Complementario/genética , Expresión Génica , Humanos , Técnicas In Vitro , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Modelos Neurológicos , Datos de Secuencia Molecular , Plasticidad Neuronal , Biosíntesis de Proteínas , Proteína Quinasa C/química , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Homología de Secuencia de Ácido Nucleico , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...