Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Mol Neurobiol ; 61(1): 487-497, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37626270

ABSTRACT

Histone post-translational modifications play an important role in the regulation of long-term memory and modulation of expression of neuronal immediate early genes (IEGs). The lysine methyltransferase KMT1A/ Suv39h1 (a mammalian ortholog of the Drosophila melanogaster SU (VAR) 3-9) aids in the methylation of histone H3 at lysine 9. We previously reported that age-related memory decline is associated with an increase in Suv39h1 expression in the hippocampus of male mice. The scopolamine-induced amnesic mouse model is a well-known animal model of memory impairment. In the current study, we have made an attempt to find a link between the changes in the H3K9 trimethylation pattern and memory decline during scopolamine-induced amnesia. It was followed by checking the effect of siRNA-mediated silencing of hippocampal Suv39h1 on memory and expression of neuronal IEGs. Scopolamine treatment significantly increased global levels of H3K9me3 and Suv39h1 in the amnesic hippocampus. Suv39h1 silencing in amnesic mice reduced H3K9me3 levels at the neuronal IEGs (Arc and BDNF) promoter, increased the expression of Arc and BDNF in the hippocampus, and improved recognition memory. Thus, these findings suggest that the silencing of Suv39h1 alone or in combination with other epigenetic drugs might be effective for treating memory decline during amnesia.


Subject(s)
Brain-Derived Neurotrophic Factor , Scopolamine , Animals , Male , Mice , Amnesia/chemically induced , Amnesia/drug therapy , Amnesia/genetics , Brain-Derived Neurotrophic Factor/metabolism , Drosophila melanogaster/metabolism , Histones/metabolism , Lysine/metabolism , Mammals/metabolism , Memory Disorders/drug therapy
2.
Int J Mol Sci ; 24(8)2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37108481

ABSTRACT

Despite several antidepressant treatments being available in clinics, they are not effective in all patients. In recent years, N-acetylcysteine (NAC) has been explored as adjunctive therapy for many psychiatric disorders, including depression, for its antioxidant properties. Given the promising efficacy of this compound for the treatment of such pathologies, it is fundamental to investigate, at the preclinical level, the ability of the drug to act in the modulation of neuroplastic mechanisms in basal conditions and during challenging events in order to highlight the potential features of the drug useful for clinical efficacy. To this aim, adult male Wistar rats were treated with the antidepressant venlafaxine (VLX) (10 mg/kg) or NAC (300 mg/kg) for 21 days and then subjected to 1 h of acute restraint stress (ARS). We found that NAC enhanced the expression of several immediate early genes, markers of neuronal plasticity in the ventral and dorsal hippocampus, prefrontal cortex and amygdala, and in particular it mediated the acute-stress-induced upregulation of Nr4a1 expression more than VLX. These data suggested the ability of NAC to induce coping strategies to face external challenges, highlighting its potential for the improvement of neuroplastic mechanisms for the promotion of resilience, in particular via the modulation of Nr4a1.


Subject(s)
Acetylcysteine , Genes, Immediate-Early , Animals , Male , Rats , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Antidepressive Agents/therapeutic use , Rats, Wistar , Venlafaxine Hydrochloride/pharmacology , Venlafaxine Hydrochloride/therapeutic use
3.
Int J Mol Sci ; 24(2)2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36674969

ABSTRACT

Rett syndrome (RTT) is a severe neurodevelopmental disease caused almost exclusively by mutations to the MeCP2 gene. This disease may be regarded as a synaptopathy, with impairments affecting synaptic plasticity, inhibitory and excitatory transmission and network excitability. The complete understanding of the mechanisms behind how the transcription factor MeCP2 so profoundly affects the mammalian brain are yet to be determined. What is known, is that MeCP2 involvement in activity-dependent expression programs is a critical link between this protein and proper neuronal activity, which allows the correct maturation of connections in the brain. By using RNA-sequencing analysis, we found several immediate-early genes (IEGs, key mediators of activity-dependent responses) directly bound by MeCP2 at the chromatin level and upregulated in the hippocampus and prefrontal cortex of the Mecp2-KO mouse. Quantification of the IEGs response to stimulus both in vivo and in vitro detected an aberrant expression pattern in MeCP2-deficient neurons. Furthermore, altered IEGs levels were found in RTT patient's peripheral blood and brain regions of post-mortem samples, correlating with impaired expression of downstream myelination-related genes. Altogether, these data indicate that proper IEGs expression is crucial for correct synaptic development and that MeCP2 has a key role in the regulation of IEGs.


Subject(s)
Rett Syndrome , Mice , Animals , Rett Syndrome/genetics , Rett Syndrome/metabolism , Genes, Immediate-Early , Methyl-CpG-Binding Protein 2/metabolism , Brain/metabolism , Neurons/metabolism , Hippocampus/metabolism , Disease Models, Animal , Mammals/metabolism
4.
Front Psychol ; 13: 1005726, 2022.
Article in English | MEDLINE | ID: mdl-36211859

ABSTRACT

In this review, we discuss the functional equivalence of the avian and mammalian hippocampus, based mostly on our own research in domestic chicks, which provide an important developmental model (most research on spatial cognition in other birds relies on adult animals). In birds, like in mammals, the hippocampus plays a central role in processing spatial information. However, the structure of this homolog area shows remarkable differences between birds and mammals. To understand the evolutionary origin of the neural mechanisms for spatial navigation, it is important to test how far theories developed for the mammalian hippocampus can also be applied to the avian hippocampal formation. To address this issue, we present a brief overview of studies carried out in domestic chicks, investigating the direct involvement of chicks' hippocampus homolog in spatial navigation.

5.
Hippocampus ; 32(11-12): 839-856, 2022 11.
Article in English | MEDLINE | ID: mdl-36314648

ABSTRACT

Memory is vital to our daily existence. Although a large number of studies have suggested that the hippocampus is dedicated to long-term memory, understanding how memory is anatomically encoded within the hippocampal neuronal network is still lacking. Previously our laboratory showed that hippocampal pyramidal cells are organized in cell clusters to encode both spatial and episodic memory. Based on these findings, we hypothesized that "cluster-type" is a functional organization principal in the hippocampus to encode all types of memory. Here, we tested whether contextual fear, another hippocampus-dependent memory, is also organized in cell clusters. We further investigated the possibility that post-learning sleep may affect functional organization. Cluster formation was examined by assessing the topographic localization of active cells using immediate early gene (IEG, Zif268) imaging methods. The first experiment provides evidence of a cluster-type organization in the hippocampus for fear memory by showing a spatial distribution of adjacent Zif268 positive cells. Exposure to the context itself, without electric shocks, induced a similar cellular formation; however, the degree of clustering was significantly lower. The second experiment provides evidence that sleep plays a role in the refinement and long-term stability of the clusters. The present results confirm the existence of a cluster-type topographic functional neuronal organization in the hippocampus for memory, and further suggest that post-learning sleep enhances the cluster-type organization.


Subject(s)
Early Growth Response Protein 1 , Hippocampus , Early Growth Response Protein 1/metabolism , Hippocampus/physiology , Fear/physiology , Genes, Immediate-Early , Sleep
6.
Front Integr Neurosci ; 16: 933426, 2022.
Article in English | MEDLINE | ID: mdl-36118115

ABSTRACT

Developmental neuroscience research has not yet fully unveiled the dynamics involved in human birth. The trigger of the first breath, often assumed to be the marker of human life, has not been characterized nor has the process entailing brain modification and activation at birth been clarified yet. To date, few researchers only have investigated the impact of the extrauterine environment, with its strong stimuli, on birth. This 'hypothesis and theory' article assumes the role of a specific stimulus activating the central nervous system (CNS) at human birth. This stimulus must have specific features though, such as novelty, efficacy, ubiquity, and immediacy. We propose light as a robust candidate for the CNS activation via the retina. Available data on fetal and neonatal neurodevelopment, in particular with reference to retinal light-responsive pathways, will be examined together with the GABA functional switch, and the subplate disappearance, which, at an experimental level, differentiate the neonatal brain from the fetal brain. In this study, we assume how a very rapid activation of retinal photoreceptors at birth initiates a sudden brain shift from the prenatal pattern of functions to the neonatal setup. Our assumption implies the presence of a photoreceptor capable of capturing and transducing light/photon stimulus, transforming it into an effective signal for the activation of new brain functions at birth. Opsin photoreception or, more specifically, melanopsin-dependent photoreception, which is provided by intrinsically photosensitive retinal ganglion cells (ipRGCs), is considered as a valid candidate. Although what is assumed herein cannot be verified in humans based on knowledge available so far, proposing an important and novel function can trigger a broad range of diversified research in different domains, from neurophysiology to neurology and psychiatry.

7.
Virology ; 575: 1-9, 2022 10.
Article in English | MEDLINE | ID: mdl-35987078

ABSTRACT

Coronavirus infection of cells differentially regulates the expression of host genes and their related pathways. In this study, we present the transcriptomic profile of cells infected with gammacoronavirus infectious bronchitis virus (IBV). In IBV-infected human non-small cell lung carcinoma cells (H1299 cells), a total of 1162 differentially expressed genes (DEGs), including 984 upregulated and 178 downregulated genes, was identified. These DEGs were mainly enriched in MAPK and Wnt signaling pathways, and 5 out of the 10 top upregulated genes in all transcripts were immediate-early response genes (IEGs). In addition, the induction of 11 transcripts was validated in IBV-infected H1299 and Vero cells by RT-qPCR. The accuracy, reliability and genericity of the transcriptomic data were demonstrated by functional characterization of these IEGs in cells infected with different coronaviruses in our previous publications. This study provides a reliable transcriptomic profile of host genes and pathways regulated by coronavirus infection.


Subject(s)
Coronavirus Infections , Infectious bronchitis virus , Animals , Chickens/genetics , Chlorocebus aethiops , Coronavirus Infections/pathology , Humans , Infectious bronchitis virus/physiology , Reproducibility of Results , Signal Transduction , Transcriptome , Vero Cells
8.
Mol Neurobiol ; 59(9): 5722-5733, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35789976

ABSTRACT

Amnesia is the inability to store new information and recall old memories. After the postulation of cholinergic hypothesis of geriatric memory dysfunction, the cholinergic signaling became a popular target to understand the underlying molecular mechanism of amnesia and its recovery. Scopolamine is a non-selective cholinergic receptor antagonist and induces amnesia through downregulation of synaptic plasticity genes including immediate early genes (IEGs). Scopolamine-induced amnesic mouse model is widely used to study the memory impairment that mimics the pathophysiology of aging, neurodegenerative, and neuropsychiatric disorders. However, a detailed understanding of cholinergic signaling-mediated regulation of plasticity-related gene expression remains elusive. Therefore, we have investigated the role of muscarinic acetylcholine receptors (mAChRs) and their downstream mediator protein kinase C (PKC) in the regulation of IEGs expression in amnesic mice hippocampus. Pilocarpine, a mAChRs agonist, was used to activate the cholinergic signaling in scopolamine-induced amnesia. Further, a PKC activator bryostatin 1 was used to understand the sole involvement of PKC as a downstream mediator of mAChRs-mediated signaling. Pilocarpine treatment significantly restored the scopolamine-induced impaired recognition memory and downregulated hippocampal IEGs expression and phosphorylation of ERK1/2 (extracellular signal-regulated kinase 1/2) and CREB (cAMP response element-binding protein). On the other hand, the bryostatin 1-mediated activation of PKC in scopolamine-induced amnesia selectively restored the hippocampal IEGs expression, recognition memory, and phosphorylation of ERK1/2 and CREB. Taken together, our findings suggest that mAChRs and their downstream mediator PKC regulate the hippocampal IEGs expression and ERK1/2-mediated CREB phosphorylation in scopolamine-induced amnesic mice.


Subject(s)
Genes, Immediate-Early , Scopolamine , Amnesia/genetics , Animals , Cholinergic Agents/pharmacology , Cyclic AMP Response Element-Binding Protein , Hippocampus/metabolism , Memory Disorders , Mice , Phosphorylation , Pilocarpine , Protein Kinase C/metabolism , Receptors, Muscarinic/metabolism , Scopolamine/pharmacology
9.
Front Physiol ; 13: 897931, 2022.
Article in English | MEDLINE | ID: mdl-35694389

ABSTRACT

In chickens, the sense of taste plays an important role in detecting nutrients and choosing feed. The molecular mechanisms underlying the taste-sensing system of chickens are well studied, but the neural mechanisms underlying taste reactivity have received less attention. Here we report the short-term taste behaviour of chickens towards umami and bitter (quinine) taste solutions and the associated neural activity in the nucleus taeniae of the amygdala, nucleus accumbens and lateral septum. We found that chickens had more contact with and drank greater volumes of umami than bitter or a water control, and that chicks displayed increased head shaking in response to bitter compared to the other tastes. We found that there was a higher neural activity, measured as c-Fos activation, in response to umami taste in the right hemisphere of the nucleus taeniae of the amygdala. In the left hemisphere, there was a higher c-Fos activation of the nucleus taeniae of the amygdala in response to bitter than in the right hemisphere. Our findings provide clear evidence that chickens respond differently to umami and bitter tastes, that there is a lateralised response to tastes at the neural level, and reveals a new function of the avian nucleus taeniae of the amygdala as a region processing reward information.

10.
Mol Brain ; 14(1): 160, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34715888

ABSTRACT

Songbirds are one of the few animal taxa that possess vocal learning abilities. Different species of songbirds exhibit species-specific learning programs during song acquisition. Songbirds with open-ended vocal learning capacity, such as the canary, modify their songs during adulthood. Nevertheless, the neural molecular mechanisms underlying open-ended vocal learning are not fully understood. We investigated the singing-driven expression of neural activity-dependent genes (Arc, Egr1, c-fos, Nr4a1, Sik1, Dusp6, and Gadd45ß) in the canary to examine a potential relationship between the gene expression level and the degree of seasonal vocal plasticity at different ages. The expression of these genes was differently regulated throughout the critical period of vocal learning in the zebra finch, a closed-ended song learner. In the canary, the neural activity-dependent genes were induced by singing in the song nuclei throughout the year. However, in the vocal motor nucleus, the robust nucleus of the arcopallium (RA), all genes were regulated with a higher induction rate by singing in the fall than in the spring. The singing-driven expression of these genes showed a similar induction rate in the fall between the first year juvenile and the second year adult canaries, suggesting a seasonal, not age-dependent, regulation of the neural activity-dependent genes. By measuring seasonal vocal plasticity and singing-driven gene expression, we found that in RA, the induction intensity of the neural activity-dependent genes was correlated with the state of vocal plasticity. These results demonstrate a correlation between vocal plasticity and the singing-driven expression of neural activity-dependent genes in RA through song development, regardless of whether a songbird species possesses an open- or closed-ended vocal learning capacity.


Subject(s)
Canaries/genetics , Gene Expression Regulation/physiology , Learning/physiology , Nerve Tissue Proteins/biosynthesis , Seasons , Vocalization, Animal/physiology , Aging/physiology , Animals , Canaries/physiology , Male , Nerve Tissue Proteins/genetics , Neural Pathways/physiology
11.
Mol Cell ; 81(19): 4041-4058.e15, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34624217

ABSTRACT

Deregulation of oncogenic signals in cancer triggers replication stress. Immediate early genes (IEGs) are rapidly and transiently expressed following stressful signals, contributing to an integrated response. Here, we find that the orphan nuclear receptor NR4A1 localizes across the gene body and 3' UTR of IEGs, where it inhibits transcriptional elongation by RNA Pol II, generating R-loops and accessible chromatin domains. Acute replication stress causes immediate dissociation of NR4A1 and a burst of transcriptionally poised IEG expression. Ectopic expression of NR4A1 enhances tumorigenesis by breast cancer cells, while its deletion leads to massive chromosomal instability and proliferative failure, driven by deregulated expression of its IEG target, FOS. Approximately half of breast and other primary cancers exhibit accessible chromatin domains at IEG gene bodies, consistent with this stress-regulatory pathway. Cancers that have retained this mechanism in adapting to oncogenic replication stress may be dependent on NR4A1 for their proliferation.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Immediate-Early Proteins/metabolism , Mitosis , Neoplastic Cells, Circulating/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , 3' Untranslated Regions , Animals , Antineoplastic Agents/pharmacology , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chromatin Assembly and Disassembly , Female , Gene Expression Regulation, Neoplastic , Genomic Instability , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Indoles/pharmacology , MCF-7 Cells , Mice, Inbred NOD , Mice, SCID , Mitosis/drug effects , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phenylacetates/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , R-Loop Structures , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Signal Transduction , Transcription Elongation, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Cells ; 10(9)2021 09 03.
Article in English | MEDLINE | ID: mdl-34571950

ABSTRACT

A tight regulation of the balance between inhibitory and excitatory synaptic transmission is a prerequisite for synaptic plasticity in neuronal networks. In this context, the neurite growth inhibitor membrane protein Nogo-A modulates synaptic plasticity, strength, and neurotransmitter receptor dynamics. However, the molecular mechanisms underlying these actions are unknown. We show that Nogo-A loss-of-function in primary mouse hippocampal cultures by application of a function-blocking antibody leads to higher excitation following a decrease in GABAARs at inhibitory and an increase in the GluA1, but not GluA2 AMPAR subunit at excitatory synapses. This unbalanced regulation of AMPAR subunits results in the incorporation of Ca2+-permeable GluA2-lacking AMPARs and increased intracellular Ca2+ levels due to a higher Ca2+ influx without affecting its release from the internal stores. Increased neuronal activation upon Nogo-A loss-of-function prompts the phosphorylation of the transcription factor CREB and the expression of c-Fos. These results contribute to the understanding of the molecular mechanisms underlying the regulation of the excitation/inhibition balance and thereby of plasticity in the brain.


Subject(s)
Calcium/metabolism , Hippocampus/metabolism , Neurons/metabolism , Nogo Proteins/metabolism , Animals , Excitatory Postsynaptic Potentials/physiology , Female , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Synapses/metabolism , Synaptic Transmission/physiology
13.
Front Mol Neurosci ; 14: 689952, 2021.
Article in English | MEDLINE | ID: mdl-34211369

ABSTRACT

Increasing evidence supports a model whereby memories are encoded by sparse ensembles of neurons called engrams, activated during memory encoding and reactivated upon recall. An engram consists of a network of cells that undergo long-lasting modifications of their transcriptional programs and connectivity. Ground-breaking advancements in this field have been made possible by the creative exploitation of the characteristic transcriptional responses of neurons to activity, allowing both engram labeling and manipulation. Nevertheless, numerous aspects of engram cell-type composition and function remain to be addressed. As recent transcriptomic studies have revealed, memory encoding induces persistent transcriptional and functional changes in a plethora of neuronal subtypes and non-neuronal cells, including glutamatergic excitatory neurons, GABAergic inhibitory neurons, and glia cells. Dissecting the contribution of these different cellular classes to memory engram formation and activity is quite a challenging yet essential endeavor. In this review, we focus on the role played by the GABAergic inhibitory component of the engram through two complementary lenses. On one hand, we report on available physiological evidence addressing the involvement of inhibitory neurons to different stages of memory formation, consolidation, storage and recall. On the other, we capitalize on a growing number of transcriptomic studies that profile the transcriptional response of inhibitory neurons to activity, revealing important clues on their potential involvement in learning and memory processes. The picture that emerges suggests that inhibitory neurons are an essential component of the engram, likely involved in engram allocation, in tuning engram excitation and in storing the memory trace.

14.
Mol Brain ; 14(1): 102, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34187543

ABSTRACT

Identification of neurons undergoing plasticity in response to external stimuli is one of the pertinent problems in neuroscience. Immediate early genes (IEGs) are widely used as a marker for neuronal plasticity. Here, we model the dynamics of IEG expression as a consecutive, irreversible first-order reaction with a limiting substrate. First, we develop an analytical framework to show that such a model, together with two-photon in vivo imaging of IEG expression, can be used to identify distinct neuronal subsets representing multiple memories. Using the above combination, we show that the expression kinetics, rather than intensity threshold, can be used to identify neuronal ensembles responding to the presentation of two events in vivo. The analytical expression allowed us to segregate the neurons based on their temporal response to one specific behavioural event, thereby improving the ability to detect plasticity related neurons. We image the retrosplenial cortex (RSc) of cfos-GFP transgenic mice to follow the dynamics of cellular changes resulting from contextual fear conditioning behaviour, enabling us to establish a representation of context in RSc at the cellular scale following memory acquisition. Thus, we obtain a general method that distinguishes neurons that took part in multiple temporally separated events by measuring fluorescence of individual neurons in live mice.


Subject(s)
Gene Expression Regulation , Genes, Immediate-Early , Imaging, Three-Dimensional , Memory/physiology , Neurons/metabolism , Animals , Behavior, Animal , Fluorescence , Green Fluorescent Proteins/metabolism , Kinetics , Mice, Transgenic , Neuronal Plasticity/genetics , Proto-Oncogene Proteins c-fos/metabolism , Time Factors
15.
Prog Brain Res ; 261: 341-378, 2021.
Article in English | MEDLINE | ID: mdl-33785135

ABSTRACT

Layer V pyramidal neurons constitute principle output neurons of the medial prefrontal cortex (mPFC)/neocortex to subcortical regions including the intralaminar/midline thalamic nuclei, amygdala, basal ganglia, brainstem nuclei and the spinal cord. The effects of 5-hydroxytryptamine (5-HT) on layer V pyramidal cells primarily reflect a range of excitatory influences through 5-HT2A receptors and inhibitory influences through non-5-HT2A receptors, including 5-HT1A receptors. While the 5-HT2A receptor is primarily a postsynaptic receptor on throughout the apical dendritic field of 5-HT2A receptors, activation of a minority of 5-HT2A receptors also appears to increase spontaneous excitatory postsynaptic currents/potentials (EPSCs/EPSPs) via a presynaptic effect on thalamocortical terminals arising from the midline and intralaminar thalamic nuclei. Activation of 5-HT2A receptors by the phenethylamine hallucinogen also appears to increase asynchronous release of glutamate upon the layer V pyramidal dendritic field, an effect that is suppressed by 5-HT itself through non-5-HT2A receptors. Serotonergic hallucinogens acting on 5-HT2A receptors also appears to increase gene expression of immediate early genes (iEG) and other receptors appearing to induce an iEG-like response like BDNF. Psychedelic hallucinogens acting on 5-HT2A receptors also induce head twitches in rodents that appear related to induction of glutamate release. These electrophysiological, biochemical and behavioral effects of serotonergic hallucinogens appear to be related to modulating glutamatergic thalamocortical neurotransmission and/or shifting the balance toward 5-HT2A receptor activation and away from non-5-HT2A receptor activation. These 5-HT2A receptor induced responses are modulated by feedback homeostatic mechanisms through mGlu2, mGlu4, and mGlu8 presynaptic receptors on thalamocortical terminals. These 5-HT2A receptor and glutamatergic interactions also appear to play a role on higher cortical functions of the mPFC such as motoric impulsivity and antidepressant-like behavioral responses on the differential-reinforcement-of low rate 72-s (DRL 72-s schedule). These mutually opposing effects between 5-HT2A receptor and mGlu autoreceptor activation (e.g., blocking 5-HT2A receptors and enhancing activity at mGlu2 receptors) may play a clinical role with respect to currently prescribed or novel antidepressant drugs. Thus, there is an important balance between 5-HT2A receptor activation and activation of mGlu autoreceptors on prefrontal cortical layer V pyramidal cells with respect to the electrophysiological, biochemical and behavioral effects serotonergic hallucinogenic drugs.


Subject(s)
Pyramidal Cells , Excitatory Postsynaptic Potentials , Glutamic Acid , Prefrontal Cortex , Serotonin
16.
Trends Food Sci Technol ; 104: 219-234, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32836826

ABSTRACT

BACKGROUND: Garlic (Allium sativum L.) is a common herb consumed worldwide as functional food and traditional remedy for the prevention of infectious diseases since ancient time. Garlic and its active organosulfur compounds (OSCs) have been reported to alleviate a number of viral infections in pre-clinical and clinical investigations. However, so far no systematic review on its antiviral effects and the underlying molecular mechanisms exists. SCOPE AND APPROACH: The aim of this review is to systematically summarize pre-clinical and clinical investigations on antiviral effects of garlic and its OSCs as well as to further analyse recent findings on the mechanisms that underpin these antiviral actions. PubMed, Cochrane library, Google Scholar and Science Direct databases were searched and articles up to June 2020 were included in this review. KEY FINDINGS AND CONCLUSIONS: Pre-clinical data demonstrated that garlic and its OSCs have potential antiviral activity against different human, animal and plant pathogenic viruses through blocking viral entry into host cells, inhibiting viral RNA polymerase, reverse transcriptase, DNA synthesis and immediate-early gene 1(IEG1) transcription, as well as through downregulating the extracellular-signal-regulated kinase (ERK)/mitogen activated protein kinase (MAPK) signaling pathway. The alleviation of viral infection was also shown to link with immunomodulatory effects of garlic and its OSCs. Clinical studies further demonstrated a prophylactic effect of garlic in the prevention of widespread viral infections in humans through enhancing the immune response. This review highlights that garlic possesses significant antiviral activity and can be used prophylactically in the prevention of viral infections.

17.
Cell Rep ; 30(11): 3717-3728.e6, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187544

ABSTRACT

Understanding the mechanisms of activity-dependent gene transcription underlying adaptive behaviors is challenging at neuronal-subtype resolution. Using cell-type specific molecular analysis in agouti-related peptide (AgRP) neurons, we reveal that the profound hunger-induced transcriptional changes greatly depend on plant homeodomain finger protein 6 (PHF6), a transcriptional repressor enriched in AgRP neurons. Loss of PHF6 in the satiated mice results in a hunger-state-shifting transcriptional profile, while hunger fails to further induce a rapid and robust activity-dependent gene transcription in PHF6-deficient AgRP neurons. We reveal that PHF6 binds to the promoters of a subset of immediate-early genes (IEGs) and that this chromatin binding is dynamically regulated by hunger state. Depletion of PHF6 decreases hunger-driven feeding motivation and makes the mice resistant to body weight gain under repetitive fasting-refeeding conditions. Our work identifies a neuronal subtype-specific transcriptional repressor that modulates transcriptional profiles in different nutritional states and enables adaptive eating behavior.


Subject(s)
Chromatin/metabolism , Gene Regulatory Networks/genetics , Hunger/physiology , Neurons/metabolism , Repressor Proteins/metabolism , Agouti-Related Protein/metabolism , Animals , Diet , Down-Regulation/genetics , Feeding Behavior , Gene Ontology , Genes, Immediate-Early , Hypothalamus/metabolism , Mice, Inbred C57BL , Motivation , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/genetics , Satiety Response , Weight Gain
18.
Biogerontology ; 21(2): 175-189, 2020 04.
Article in English | MEDLINE | ID: mdl-31760560

ABSTRACT

With advancing age, memory declines through different mechanisms including dysregulation of expression of synaptic plasticity genes in hippocampus. Increasing evidences suggest that these synaptic plasticity genes are regulated through epigenetic modifications. Recently we have reported that the neuronal immediate early genes (IEGs) are regulated by DNA methylation and histone acetylation, and their expression is downregulated in the hippocampus of old male mice, which subsequently results in decline of memory. These modifications do not work in isolation but act synergistically and lead to distinct regulation of gene expression. Therefore, in the present study, we have explored whether these genes are also regulated by histone methylation and this has any correlation with memory decline during aging. This study for the first time reports involvement of H3K9me3 in the regulation of neuronal IEGs during aging. Using novel object recognition and Y-maze test, the recognition and spatial memory was checked in male mice of different ages and it was found to decline in old. We have examined the expression of H3K9me3 specific histone methyltransferases and noted that only SUV39H1 (suppressor of variegation 3-9 homolog 1) increased significantly in old. Also the global H3K9me3 level was high in the hippocampus of old male mice. Further, chromatin immunoprecipitation assay revealed rise in H3K9me3 level at the promoter of IEGs in old as compared to young male mice. The immunofluorescence analysis also showed varying pattern of H3K9me3 expression in different subregions of hippocampus with aging. These findings showed negative correlation of increase in hippocampal histone H3K9me3 with memory decline in old male mice. Diagram here represents that during aging, there is increase in expression of SUV39H1. Such increased enzyme upregulates global and gene specific methylation in hippocampus of old male mice. H3K9me3 level increases at the promoter of neuronal IEGs leading to heterochromatisation and hence decrease in their expression and ultimately decline in memory during aging.


Subject(s)
Aging/metabolism , Behavior, Animal , DNA Methylation , Exploratory Behavior , Genes, Immediate-Early , Hippocampus/metabolism , Histones/metabolism , Spatial Memory , Age Factors , Aging/genetics , Animals , Male , Mice , Open Field Test , Promoter Regions, Genetic , Sex Factors
19.
Front Cell Neurosci ; 14: 593309, 2020.
Article in English | MEDLINE | ID: mdl-33390905

ABSTRACT

Motherhood entails changes in behavior with increased motivation for pups, induced in part by pregnancy hormones acting upon the brain. This work explores whether this alters sensory processing of pup-derived chemosignals. To do so, we analyse the expression of immediate early genes (IEGs) in the vomeronasal organ (VNO; Egr1) and centers of the olfactory and vomeronasal brain pathways (cFos) in virgin and late-pregnant females exposed to pups, as compared to buttons (socially neutral control). In pup-exposed females, we quantified diverse behaviors including pup retrieval, sniffing, pup-directed attack, nest building and time in nest or on nest, as well as time off nest. Pups induce Egr1 expression in the VNO of females, irrespective of their physiological condition, thus suggesting the existence of VNO-detected pup chemosignals. A similar situation is found in the accessory olfactory bulb (AOB) and posteromedial part of the medial bed nucleus of the stria terminalis (BSTMPM). By contrast, in the medial amygdala and posteromedial cortical amygdala (PMCo), responses to pups-vs-buttons are different in virgin and late-pregnant females, thus suggesting altered sensory processing during late pregnancy. The olfactory system also shows changes in sensory processing with pregnancy. In the main olfactory bulbs, as well as the anterior and posterior piriform cortex, buttons activate cFos expression in virgins more than in pregnant females. By contrast, in the anterior and especially posterior piriform cortex, pregnant females show more activation by pups than buttons. Correlation between IEGs expression and behavior suggests the existence of two vomeronasal subsystems: one associated to pup care (with PMCo as its main center) and another related to pup-directed aggression observed in some pregnant females (with the BSTMPM as the main nucleus). Our data also suggest a coactivation of the olfactory and vomeronasal systems during interaction with pups in pregnant females.

20.
Bioessays ; 41(8): e1900045, 2019 08.
Article in English | MEDLINE | ID: mdl-31237359

ABSTRACT

Consolidation of long-term memory is a highly and precisely regulated multistep process. The transcription regulator cAMP response element-binding protein (CREB) plays a key role in initiating memory consolidation. With time processing, first the cofactors are changed and, secondly, CREB gets dispensable. This ultimately changes the expressed gene program to genes required to maintain the memory. Regulation of memory consolidation also requires epigenetic mechanisms and control at the RNA level. At the neuronal circuit level, oscillation in the activity of CREB and downstream factor define engram cells. Together the combination of all regulation mechanisms allows correct memory processing while keeping the process dynamic and flexible to adjust to different contexts. Also see the video abstract here https://youtu.be/BhSCSmorpEc.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation/physiology , Gene Regulatory Networks/physiology , Memory Consolidation/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cytoskeletal Proteins/metabolism , Epigenesis, Genetic/physiology , Humans , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/genetics , Neurons/metabolism , Phosphorylation , RNA, Messenger/metabolism , Signal Transduction/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL