Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Curr Opin Neurobiol ; 84: 102829, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38128422

RESUMEN

Throughout development, the neuronal epigenome is highly sensitive to external stimuli, yet capable of safeguarding cellular memory for a lifetime. In the adult brain, memories of fearful experiences are rapidly instantiated, yet can last for decades, but the mechanisms underlying such longevity remain unknown. Here, we showcase how fear memory formation and storage - traditionally thought to exclusively affect synapse-based events - elicit profound and enduring changes to the chromatin, proposing epigenetic regulation as a plausible molecular template for mnemonic processes. By comparing these to mechanisms occurring in development and differentiation, we notice that an epigenetic machinery similar to that preserving cellular memories might be employed by brain cells so as to form, store, and retrieve behavioral memories.


Asunto(s)
Epigénesis Genética , Memoria , Memoria/fisiología , Encéfalo/fisiología , Miedo/fisiología , Cromatina
2.
Front Behav Neurosci ; 17: 1181818, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37700912

RESUMEN

A memory engram is thought to be the physical substrate of the memory trace within the brain, which is generally depicted as a neuronal ensemble activated by learning to fire together during encoding and retrieval. It has been postulated that engram cell ensembles are functionally interconnected across multiple brain regions to store a single memory as an "engram complex", but visualizing this engram complex across the whole brain has for long been hindered by technical limitations. With the recent development of tissue clearing techniques, advanced light-sheet microscopy, and automated 3D image analysis, it has now become possible to generate a brain-wide map of engram cells and thereby to visualize the "engram complex". In this review, we first provide a comprehensive summary of brain-wide engram mapping studies to date. We then compile a guide on implementing the optimal tissue clearing technique for engram tagging approaches, paying particular attention to visualize engram reactivation as a critical mnemonic property, for which whole-brain multiplexed immunostaining becomes a challenging prerequisite. Finally, we highlight the potential of tissue clearing to simultaneously shed light on both the circuit connectivity and molecular underpinnings of engram cells in a single snapshot. In doing so, novel brain regions and circuits can be identified for subsequent functional manipulation, thus providing an opportunity to robustly examine the "engram complex" underlying memory storage.

3.
Urol Oncol ; 41(12): 484.e17-484.e26, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37407421

RESUMEN

INTRODUCTION AND OBJECTIVE: BTA stat®, NMP22® BladderChek®, UBC® Rapid Test, and CancerCheck® UBC® rapid VISUAL are urinary-based rapid tests. This multicenter study is the first study comparing all available rapid tests on a large cohort of bladder cancer patients and healthy controls in one setting. METHODS: In total 732 urine samples (second morning urine) in a real-world assessment have been analyzed. We evaluated clinical samples from 464 patients with histologically confirmed urothelial tumors of the urinary bladder (17 solitary CIS, 189 low-grade, 187 high-grade nonmuscle invasive, 71 high-grade muscle invasive), 77 patients with No Evidence of Disease (NED), and from 191 healthy controls. Urine samples were analyzed by the BTA stat®, NMP22® BladderChek®, UBC® Rapid Test point-of-care (POC) system using the concile Omega 100 POC reader, and CancerCheck® UBC® rapid VISUAL. Sensitivities and specificities were calculated by contingency analyses. RESULTS: All investigated urinary markers detected more pathological concentrations in urine of bladder cancer patients compared to tumor-free patients. The calculated diagnostic sensitivities for BTA stat®, NMP22® BladderChek®, UBC® Rapid Test, CancerCheck® UBC® rapid VISUAL, and cytology were 62.4%, 13.4%, 58.2%, 28.6%, 36.2% for low-grade, 83.4%, 49.5%, 84.5%, 63.1%, 71.2% for high-grade nonmuscle invasive, and 95.8%, 35.2%, 76.1%, 50.7%, 67.7% for high-grade muscle-invasive bladder cancer. The specificity was 67.9%, 95.5%, 79.4%, 94.4%, and 83.7%, respectively. The area under the curve (AUC) after receiver operating characteristics (ROC) analysis for high-grade non-muscle-invasive tumors was 0.757, 0.725, 0.819, 0.787, and 0.774, respectively. CONCLUSIONS: The analysis of more than 700 urine samples offers an objective view on urine-based rapid diagnostics. Elevated pathological concentrations of markers in urine of bladder cancer patients were detected in all investigated tests. The highest sensitivities for high-grade non-muscle-invasive tumors were calculated for BTA stat® and UBC® Rapid Test, whereas NMP22® BladderChek®, and cytology showed the highest specificities. BTA stat® and UBC® Rapid Test have the potential to be used as a clinical valuable urinary protein biomarker for the detection of high-grade non-muscle-invasive bladder cancer patients and could be included in the management of these tumors.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Vejiga Urinaria , Humanos , Biomarcadores de Tumor/orina , Neoplasias de la Vejiga Urinaria/patología , Proteínas Nucleares/orina , Sensibilidad y Especificidad
4.
Neurobiol Dis ; 184: 106219, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37422091

RESUMEN

Accumulating evidence indicates that early adverse life experiences may be involved in the pathogenesis of Alzheimer's disease (AD). Prenatal stress (PS) can affect brain maturation and neuroimmune and metabolic interactions, leading to age-dependent cognitive deficits in offspring. However, a multi-faceted cause-and-effect impact of PS on the development of cognitive deficits in the process of physiological ageing and in the APPNL-F/NL-F mouse model of Alzheimer's disease has not yet been evaluated. We have identified age-dependent cognitive learning and memory deficits using male C57BL/6 J (wild type, WT) and the knock-in APPNL-F/NL-F (KI) aged 12, 15, and 18 months. An increase in the Aß42/Aß40 ratio and mouse ApoE levels in the hippocampus and frontal cortex preceded the onset of cognitive deficits in the KI mice. Moreover, dysfunction in insulin signaling, including increased IRS-1 serine phosphorylation in both brain areas and the tyrosine phosphorylation deficit in the frontal cortex, suggested age-dependent insulin/IGF-1 resistance. Resistance was reflected by disturbances in mTOR or ERK1/2 kinase phosphorylation and excessive pro-inflammatory (TNF-α, IL-6, and IL-23) status in the KI mice. Importantly, our study has provided insights into the higher vulnerability to PS-induced exacerbation of age-dependent cognitive deficits and biochemical dysfunction in KI mice than in WT animals. We anticipate our study will lead to future investigation of a multi-faceted cause-and-effect relationship between stress during neurodevelopment and the onset of AD pathology, distinguishing it from changes in the course of dementia during normal ageing.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Femenino , Embarazo , Masculino , Ratones , Animales , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Insulina , Ratones Transgénicos , Ratones Endogámicos C57BL , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo
5.
Nat Neurosci ; 26(5): 820-829, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37024573

RESUMEN

A painful episode can lead to a life-long increase in an individual's experience of pain. Fearful anticipation of imminent pain could play a role in this phenomenon, but the neurobiological underpinnings are unclear because fear can both suppress and enhance pain. Here, we show in mice that long-term associative fear memory stored in neuronal engrams in the prefrontal cortex determines whether a painful episode shapes pain experience later in life. Furthermore, under conditions of inflammatory and neuropathic pain, prefrontal fear engrams expand to encompass neurons representing nociception and tactile sensation, leading to pronounced changes in prefrontal connectivity to fear-relevant brain areas. Conversely, silencing prefrontal fear engrams reverses chronically established hyperalgesia and allodynia. These results reveal that a discrete subset of prefrontal cortex neurons can account for the debilitating comorbidity of fear and chronic pain and show that attenuating the fear memory of pain can alleviate chronic pain itself.


Asunto(s)
Dolor Crónico , Ratones , Animales , Memoria a Largo Plazo , Miedo/fisiología , Encéfalo , Corteza Prefrontal/fisiología , Hiperalgesia , Percepción del Dolor/fisiología
6.
Trends Cogn Sci ; 27(4): 404-416, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36813591

RESUMEN

Traumatic events generate some of the most enduring memories, yet little is known about how long-lasting fear memories can be attenuated. In this review, we collect the surprisingly sparse evidence on remote fear memory attenuation from both animal and human research. What is becoming apparent is twofold: although remote fear memories are more resistant to change compared with recent ones, they can nevertheless be attenuated when interventions are targeted toward the period of memory malleability instigated by memory recall, the reconsolidation window. We describe the physiological mechanisms underlying remote reconsolidation-updating approaches and highlight how they can be enhanced through interventions promoting synaptic plasticity. By capitalizing on an intrinsically relevant phase of memory, reconsolidation-updating harbors the potential to permanently alter remote fear memories.


Asunto(s)
Memoria a Largo Plazo , Memoria , Animales , Humanos , Memoria/fisiología , Miedo/fisiología , Plasticidad Neuronal , Extinción Psicológica/fisiología
7.
Alzheimers Dement ; 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36479795

RESUMEN

Disturbances in the brain's capacity to meet its energy demand increase the risk of synaptic loss, neurodegeneration, and cognitive decline. Nutritional and metabolic interventions that target metabolic pathways combined with diagnostics to identify deficits in cerebral bioenergetics may therefore offer novel therapeutic potential for Alzheimer's disease (AD) prevention and management. Many diet-derived natural bioactive components can govern cellular energy metabolism but their effects on brain aging are not clear. This review examines how nutritional metabolism can regulate brain bioenergetics and mitigate AD risk. We focus on leading mechanisms of cerebral bioenergetic breakdown in the aging brain at the cellular level, as well as the putative causes and consequences of disturbed bioenergetics, particularly at the blood-brain barrier with implications for nutrient brain delivery and nutritional interventions. Novel therapeutic nutrition approaches including diet patterns are provided, integrating studies of the gut microbiome, neuroimaging, and other biomarkers to guide future personalized nutritional interventions.

8.
Elife ; 112022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35838139

RESUMEN

Memory formation and storage rely on multiple interconnected brain areas, the contribution of which varies during memory consolidation. The medial prefrontal cortex, in particular the prelimbic cortex (PL), was traditionally found to be involved in remote memory storage, but recent evidence points toward its implication in early consolidation as well. Nevertheless, the inputs to the PL governing these dynamics remain unknown. Here, we first performed a brain-wide, rabies-based retrograde tracing screen of PL engram cells activated during contextual fear memory formation in male mice to identify relevant PL input regions. Next, we assessed the specific activity pattern of these inputs across different phases of memory consolidation, from fear memory encoding to recent and remote memory recall. Using projection-specific chemogenetic inhibition, we then tested their functional role in memory consolidation, which revealed a hitherto unknown contribution of claustrum to PL inputs at encoding, and of insular cortex to PL inputs at recent memory recall. Both of these inputs further impacted how PL engram cells were reactivated at memory recall, testifying to their relevance for establishing a memory trace in the PL. Collectively, these data identify a spatiotemporal shift in PL inputs important for early memory consolidation, and thereby help to refine the working model of memory formation.


Asunto(s)
Consolidación de la Memoria , Animales , Corteza Cerebral/fisiología , Miedo/fisiología , Masculino , Memoria/fisiología , Consolidación de la Memoria/fisiología , Ratones , Corteza Prefrontal/fisiología
9.
Proc Natl Acad Sci U S A ; 119(22): e2116797119, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35613054

RESUMEN

Long-term memory formation relies on synaptic plasticity, neuronal activity-dependent gene transcription, and epigenetic modifications. Multiple studies have shown that HDAC inhibitor (HDACi) treatments can enhance individual aspects of these processes and thereby act as putative cognitive enhancers. However, their mode of action is not fully understood. In particular, it is unclear how systemic application of HDACis, which are devoid of substrate specificity, can target pathways that promote memory formation. In this study, we explore the electrophysiological, transcriptional, and epigenetic responses that are induced by CI-994, a class I HDACi, combined with contextual fear conditioning (CFC) in mice. We show that CI-994­mediated improvement of memory formation is accompanied by enhanced long-term potentiation in the hippocampus, a brain region recruited by CFC, but not in the striatum, a brain region not primarily implicated in fear learning. Furthermore, using a combination of bulk and single-cell RNA-sequencing, we find that, when paired with CFC, HDACi treatment engages synaptic plasticity-promoting gene expression more strongly in the hippocampus, specifically in the dentate gyrus (DG). Finally, using chromatin immunoprecipitation-sequencing (ChIP-seq) of DG neurons, we show that the combined action of HDACi application and conditioning is required to elicit enhancer histone acetylation in pathways that underlie improved memory performance. Together, these results indicate that systemic HDACi administration amplifies brain region-specific processes that are naturally induced by learning.


Asunto(s)
Benzamidas , Giro Dentado , Inhibidores de Histona Desacetilasas , Memoria a Largo Plazo , Fenilendiaminas , Animales , Benzamidas/farmacología , Comunicación Celular/efectos de los fármacos , Giro Dentado/citología , Giro Dentado/efectos de los fármacos , Giro Dentado/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Memoria a Largo Plazo/efectos de los fármacos , Ratones , Plasticidad Neuronal , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fenilendiaminas/farmacología , RNA-Seq , Análisis de la Célula Individual
10.
Int J Mol Sci ; 22(22)2021 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-34830009

RESUMEN

It is becoming increasingly apparent that long-term memory formation relies on a distributed network of brain areas. While the hippocampus has been at the center of attention for decades, it is now clear that other regions, in particular the medial prefrontal cortex (mPFC), are taking an active part as well. Recent evidence suggests that the mPFC-traditionally implicated in the long-term storage of memories-is already critical for the early phases of memory formation such as encoding. In this review, we summarize these findings, relate them to the functional importance of the mPFC connectivity, and discuss the role of the mPFC during memory consolidation with respect to the different theories of memory storage. Owing to its high functional connectivity to other brain areas subserving memory formation and storage, the mPFC emerges as a central hub across the lifetime of a memory, although much still remains to be discovered.


Asunto(s)
Miedo/fisiología , Hipocampo/fisiología , Memoria a Largo Plazo/fisiología , Corteza Prefrontal/fisiología , Animales , Mapeo Encefálico , Humanos
11.
Nat Neurosci ; 24(7): 964-974, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34017129

RESUMEN

Fear and trauma generate some of the longest-lived memories. Despite the corresponding need to understand how such memories can be attenuated, the underlying brain circuits remain unknown. Here, combining viral tracing, neuronal activity mapping, fiber photometry, chemogenetic and closed-loop optogenetic manipulations in mice, we show that the extinction of remote (30-day-old) fear memories depends on thalamic nucleus reuniens (NRe) inputs to the basolateral amygdala (BLA). We found that remote, but not recent (1-day-old), fear extinction activates NRe-to-BLA inputs, which become potentiated upon fear reduction. Furthermore, both monosynaptic NRe-to-BLA and total NRe activity increase shortly before freezing cessation, suggesting that the NRe registers and transmits safety signals to the BLA. Accordingly, pan-NRe and pathway-specific NRe-to-BLA inhibition impairs, whereas their activation facilitates, remote fear extinction. These findings identify the NRe as a crucial BLA regulator for extinction and provide the first functional description of the circuits underlying the attenuation of consolidated fear memories.


Asunto(s)
Amígdala del Cerebelo/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Memoria a Largo Plazo/fisiología , Tálamo/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología
12.
Curr Opin Neurobiol ; 67: 75-84, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33120188

RESUMEN

Multiple studies have found that increasing histone acetylation by means of histone deacetylase inhibitor (HDACi) treatment can ameliorate memory and rescue cognitive impairments, but their mode of action is not fully understood. In particular, it is unclear how HDACis, applied systemically and devoid of genomic target selectivity, would specifically improve memory-related molecular processes. One theory for such specificity is called cognitive epigenetic priming (CEP), according to which HDACis promote memory by facilitating the expression of neuroplasticity-related genes that have been stimulated by learning itself. In this review, we summarize the experimental evidence in support of CEP, describe newly discovered off-target effects of HDACis and highlight similarities between drug-induced and naturally occurring CEP. Understanding the underlying mechanisms of CEP is important in light of the preclinical premise of HDACis as cognitive enhancers.


Asunto(s)
Inhibidores de Histona Desacetilasas , Histonas , Acetilación , Cognición , Epigénesis Genética , Inhibidores de Histona Desacetilasas/farmacología
13.
Front Mol Neurosci ; 13: 205, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33343294

RESUMEN

Over the last two decades, the explosion of experimental, computational, and high-throughput technologies has led to critical insights into how the brain functions in health and disease. It has become increasingly clear that the vast majority of brain activities result from the complex entanglement of genetic factors, epigenetic changes, and environmental stimuli, which, when altered, can lead to neurodegenerative and neuropsychiatric disorders. Nevertheless, a complete understanding of the molecular mechanisms underlying neuronal activities and higher-order cognitive processes continues to elude neuroscientists. Here, we provide a concise overview of how the interaction between the environment and genetic as well as epigenetic mechanisms shapes complex neuronal processes such as learning, memory, and synaptic plasticity. We then consider how this interaction contributes to the development of neurodegenerative and psychiatric disorders, and how it can be modeled to predict phenotypic variability and disease risk. Finally, we outline new frontiers in neurogenetic and neuroepigenetic research and highlight the challenges these fields will face in their quest to decipher the molecular mechanisms governing brain functioning.

14.
Opt Express ; 28(24): 36643-36655, 2020 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-33379754

RESUMEN

Despite the existence of various neural recording and mapping techniques, there is an open territory for the emergence of novel techniques. The current neural imaging and recording techniques suffer from invasiveness, a time-consuming labeling process, poor spatial/ temporal resolution, and noisy signals. Among others, neuroplasmonics is a label-free and nontoxic recording technique with no issue of photo-bleaching or signal-averaging. We introduced an integrated plasmonic-ellipsometry platform for membrane activity detection with cost-effective and high-quality grating extracted from commercial DVDs. With ellipsometry technique, one can measure both amplitude (intensity) and phase difference of reflected light simultaneously with high signal to noise ratio close to surface plasmon resonances, which leads to the enhancement of sensitivity in plasmonic techniques. We cultured three different types of cells (primary hippocampal neurons, neuroblastoma SH-SY5Y cells, and human embryonic kidney 293 (HEK293) cells) on the grating surface. By introducing KCl solution as a chemical stimulus, we can differentiate the neural activity of distinct cell types and observe the signaling event in a label-free, optical recording platform. This method has potential applications in recording neural signal activity without labeling and stimulation artifacts.


Asunto(s)
Técnicas Biosensibles/métodos , Membrana Celular/fisiología , Hipocampo/citología , Neuroblastoma/patología , Neuronas/citología , Resonancia por Plasmón de Superficie/métodos , Animales , Células HEK293/citología , Humanos , Ratas , Células Tumorales Cultivadas
15.
Clin Epigenetics ; 12(1): 20, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32014019

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a complex disorder caused by a combination of genetic and non-genetic risk factors. In addition, an increasing evidence suggests that epigenetic mechanisms also accompany AD. Genetic and epigenetic factors are not independent, but multiple loci show genetic-epigenetic interactions, the so-called quantitative trait loci (QTLs). Recently, we identified the first QTL association with AD, namely Peptidase M20 Domain Containing 1 (PM20D1). We observed that PM20D1 DNA methylation, RNA expression, and genetic background are correlated and, in turn, associated with AD. We provided mechanistic insights for these correlations and had shown that by genetically increasing and decreasing PM20D1 levels, AD-related pathologies were decreased and accelerated, respectively. However, since the PM20D1 QTL region encompasses also other genes, namely Nuclear Casein Kinase and Cyclin Dependent Kinase Substrate 1 (NUCKS1); RAB7, member RAS oncogene family-like 1 (RAB7L1); and Solute Carrier Family 41 Member 1 (SLC41A1), we investigated whether these genes might also contribute to the described AD association. RESULTS: Here, we report a comprehensive analysis of these QTL genes using a repertoire of in silico methods as well as in vivo and in vitro experimental approaches. First, we analyzed publicly available databases to pinpoint the major QTL correlations. Then, we validated these correlations using a well-characterized set of samples and locus-specific approaches-i.e., Sanger sequencing for the genotype, cloning/sequencing and pyrosequencing for the DNA methylation, and allele-specific and real-time PCR for the RNA expression. Finally, we defined the functional relevance of the observed alterations in the context of AD in vitro. Using this approach, we show that only PM20D1 DNA methylation and expression are significantly correlated with the AD-risk associated background. We find that the expression of SLC41A1 and PM20D1-but not NUCKS1 and RAB7L1-is increased in mouse models and human samples of AD, respectively. However, SLC41A1 and PM20D1 are differentially regulated by AD-related stressors, with only PM20D1 being upregulated by amyloid-ß and reactive oxygen species, and with only PM20D1 being neuroprotective when overexpressed in cell and primary cultures. CONCLUSIONS: Our findings reinforce PM20D1 as the most likely gene responsible of the previously reported PM20D1 QTL association with AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Amidohidrolasas/metabolismo , Metilación de ADN/genética , Sitios de Carácter Cuantitativo/genética , Anciano , Enfermedad de Alzheimer/tratamiento farmacológico , Amidohidrolasas/farmacología , Péptidos beta-Amiloides/metabolismo , Animales , Autopsia , Proteínas de Transporte de Catión/metabolismo , Epigénesis Genética , Epigenómica , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , ARN/genética , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
16.
Mol Psychiatry ; 25(9): 2144-2161, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-30089788

RESUMEN

Aggression is frequently observed in neurodevelopmental psychiatric disorders such as schizophrenia, autism, and bipolar disorder. Due to a lack of understanding of its underlying mechanisms, effective treatments for abnormal aggression are still missing. Recently, genetic variations in Sialyltransferase 2 (St8sia2) have been linked to these disorders and aggression. Here we identify abnormal aggressive behaviors and concomitant blunted fear learning in St8sia2 knockout (-/-) mice. It is worth noting that the amygdala of St8sia2-/- mice shows diminished threat-induced activation, as well as alterations in synaptic structure and function, including impaired GluN2B-containing NMDA receptor-mediated synaptic transmission and plasticity. Pharmacological rescue of NMDA receptor activity in the amygdala of St8sia2-/- mice with the partial agonist D-cycloserine restores synaptic plasticity and normalizes behavioral aberrations. Pathological aggression and associated traits were recapitulated by specific amygdala neonatal St8sia2 silencing. Our results establish a developmental link between St8sia2 deficiency and a pathological aggression syndrome, specify synaptic targets for therapeutic developments, and highlight D-cycloserine as a plausible treatment.


Asunto(s)
Agresión , Amígdala del Cerebelo , Receptores de N-Metil-D-Aspartato , Sialiltransferasas , Amígdala del Cerebelo/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sialiltransferasas/genética
17.
Clin Epigenetics ; 11(1): 198, 2019 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-31878957

RESUMEN

BACKGROUND: Metabolic side effects induced by psychotropic drugs represent a major health issue in psychiatry. CREB-regulated transcription coactivator 1 (CRTC1) gene plays a major role in the regulation of energy homeostasis and epigenetic mechanisms may explain its association with obesity features previously described in psychiatric patients. This prospective study included 78 patients receiving psychotropic drugs that induce metabolic disturbances, with weight and other metabolic parameters monitored regularly. Methylation levels in 76 CRTC1 probes were assessed before and after 1 month of psychotropic treatment in blood samples. RESULTS: Significant methylation changes were observed in three CRTC1 CpG sites (i.e., cg07015183, cg12034943, and cg 17006757) in patients with early and important weight gain (i.e., equal or higher than 5% after 1 month; FDR p value = 0.02). Multivariable models showed that methylation decrease in cg12034943 was more important in patients with early weight gain (≥ 5%) than in those who did not gain weight (p = 0.01). Further analyses combining genetic and methylation data showed that cg12034943 was significantly associated with early weight gain in patients carrying the G allele of rs4808844A>G (p = 0.03), a SNP associated with this methylation site (p = 0.03). CONCLUSIONS: These findings give new insights on psychotropic-induced weight gain and underline the need of future larger prospective epigenetic studies to better understand the complex pathways involved in psychotropic-induced metabolic side effects.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Obesidad/genética , Polimorfismo de Nucleótido Simple , Psicotrópicos/efectos adversos , Factores de Transcripción/genética , Aumento de Peso/genética , Adulto , Edad de Inicio , Alelos , Estudios de Casos y Controles , Islas de CpG/efectos de los fármacos , Epigénesis Genética , Femenino , Estudios de Asociación Genética , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Obesidad/inducido químicamente , Estudios Prospectivos , Psicotrópicos/farmacología
18.
Front Mol Neurosci ; 12: 70, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31057365

RESUMEN

Whether the attenuation of traumatic memories is mediated through the suppression of the original memory trace of fear by a new memory trace of safety, or through an updating of the original fear trace towards safety has been a long-standing question at the interface of neuroscience and psychology. This matter is of particular importance for remote fear memories as they lie at the core of stress- and anxiety-related disorders. Recently, we have found that in the dentate gyrus, the effective attenuation of remote fear memories is accompanied by a reactivation of memory recall-induced neurons and that the continued activity of these neurons is critical for fear reduction. However, whether this also applies to other brain areas implicated in the storage of remote fear memories remains to be determined. Here, we show-by cellular compartment analysis of temporal activity using fluorescence in situ hybridization-that such reactivation also occurs in the basolateral amygdala and the infralimbic cortex, two brain areas known to be involved in fear memory attenuation. These results provide further experimental support for effective traumatic memory attenuation likely being mediated by an updating of the original fear trace towards safety.

19.
Neuron ; 101(2): 198-200, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30653932

RESUMEN

Engram cells can encode and switch between multiple mnemonic functions, but how they intrinsically do so is unknown. Pignatelli, Ryan, and colleagues show that upon memory recall, the engram's excitability is transiently elevated, allowing its bearer to adapt to changing environments.


Asunto(s)
Memoria
20.
Psychopharmacology (Berl) ; 236(1): 369-381, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30116860

RESUMEN

RATIONALE: The experience of strong traumata leads to the formation of enduring fear memories that may degenerate into post-traumatic stress disorder. One of the most successful treatments for this condition consists of extinction training during which the repeated exposure to trauma-inducing stimuli in a safe environment results in an attenuation of the fearful component of trauma-related memories. While numerous studies have investigated the neural substrates of recent (e.g., 1-day-old) fear memory attenuation, much less is known about the neural networks mediating the attenuation of remote (e.g., 30-day-old) fear memories. Since extinction training becomes less effective when applied long after the original encoding of the traumatic memory, this represents an important gap in memory research. OBJECTIVES: Here, we aimed to generate a comprehensive map of brain activation upon effective remote fear memory attenuation in the mouse. METHODS: We developed an efficient extinction training paradigm for 1-month-old contextual fear memory attenuation and performed cFos immunohistochemistry and network connectivity analyses on a set of cortical, amygdalar, thalamic, and hippocampal regions. RESULTS: Remote fear memory attenuation induced cFos in the prelimbic cortex, the basolateral amygdala, the nucleus reuniens of the thalamus, and the ventral fields of the hippocampal CA1 and CA3. All these structures were equally recruited by remote fear memory recall, but not by the recall of a familiar neutral context. CONCLUSION: These results suggest that progressive fear attenuation mediated by repetitive exposure is accompanied by sustained neuronal activation and not reverted to a pre-conditioning brain state. These findings contribute to the identification of brain areas as targets for therapeutic approaches against traumatic memories.


Asunto(s)
Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Miedo/fisiología , Memoria a Largo Plazo/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Trastornos por Estrés Postraumático/fisiopatología , Animales , Mapeo Encefálico , Extinción Psicológica/fisiología , Miedo/psicología , Humanos , Terapia Implosiva , Recuerdo Mental/fisiología , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/fisiopatología , Neuronas/fisiología , Trastornos por Estrés Postraumático/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA