Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.594
Filter
1.
Neurobiol Aging ; 139: 20-29, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38583392

ABSTRACT

Brazilian green propolis (propolis) is a chemically complex resinous substance that is a potentially viable therapeutic agent for Alzheimer's disease. Herein, propolis induced a transient increase in intracellular Ca2+ concentration ([Ca2+]i) in Neuro-2A cells; moreover, propolis-induced [Ca2+]i elevations were suppressed prior to 24-h pretreatment with amyloid-ß. To reveal the effect of [Ca2+]i elevation on impaired cognition, we performed memory-related behavioral tasks in APP-KI mice relative to WT mice at 4 and 12 months of age. Propolis, at 300-1000 mg/kg/d for 8 wk, significantly ameliorated cognitive deficits in APP-KI mice at 4 months, but not at 12 months of age. Consistent with behavioral observations, injured hippocampal long-term potentiation was markedly ameliorated in APP-KI mice at 4 months of age following repeated propolis administration. In addition, repeated administration of propolis significantly activated intracellular calcium signaling pathway in the CA1 region of APP-KI mice. These results suggest a preventive effect of propolis on cognitive decline through the activation of intracellular calcium signaling pathways in CA1 region of AD mice model.


Subject(s)
Alzheimer Disease , Calcium , Cognitive Dysfunction , Disease Models, Animal , Propolis , Animals , Propolis/therapeutic use , Propolis/administration & dosage , Propolis/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Alzheimer Disease/psychology , Alzheimer Disease/etiology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/drug therapy , Calcium/metabolism , Mice, Transgenic , Calcium Signaling/drug effects , Long-Term Potentiation/drug effects , Male , Amyloid beta-Peptides/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/drug effects , Mice
2.
Pharmacol Res ; 203: 107176, 2024 May.
Article in English | MEDLINE | ID: mdl-38583687

ABSTRACT

Cannabidiol (CBD), a non-psychotomimetic constituent of Cannabis sativa, has been recently approved for epileptic syndromes often associated with Autism spectrum disorder (ASD). However, the putative efficacy and mechanism of action of CBD in patients suffering from ASD and related comorbidities remain debated, especially because of the complex pharmacology of CBD. We used pharmacological, immunohistochemical and biochemical approaches to investigate the effects and mechanisms of action of CBD in the recently validated Fmr1-Δexon 8 rat model of ASD, that is also a model of Fragile X Syndrome (FXS), the leading monogenic cause of autism. CBD rescued the cognitive deficits displayed by juvenile Fmr1-Δexon 8 animals, without inducing tolerance after repeated administration. Blockade of CA1 hippocampal GPR55 receptors prevented the beneficial effect of both CBD and the fatty acid amide hydrolase (FAAH) inhibitor URB597 in the short-term recognition memory deficits displayed by Fmr1-Δexon 8 rats. Thus, CBD may exert its beneficial effects through CA1 hippocampal GPR55 receptors. Docking analysis further confirmed that the mechanism of action of CBD might involve competition for brain fatty acid binding proteins (FABPs) that deliver anandamide and related bioactive lipids to their catabolic enzyme FAAH. These findings demonstrate that CBD reduced cognitive deficits in a rat model of FXS and provide initial mechanistic insights into its therapeutic potential in neurodevelopmental disorders.


Subject(s)
Cannabidiol , Disease Models, Animal , Fragile X Syndrome , Hippocampus , Receptors, Cannabinoid , Recognition, Psychology , Animals , Fragile X Syndrome/drug therapy , Fragile X Syndrome/metabolism , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Receptors, Cannabinoid/metabolism , Male , Recognition, Psychology/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Fragile X Mental Retardation Protein/metabolism , Fragile X Mental Retardation Protein/genetics , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Memory/drug effects , Receptors, G-Protein-Coupled/metabolism , Molecular Docking Simulation
3.
Life Sci ; 346: 122618, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38614306

ABSTRACT

AIMS: This study was designed to investigate the role of growth arrest and DNA damage-inducible ß (GADD45B) in modulating fear memory acquisition and elucidate its underlying mechanisms. MAIN METHODS: Adeno-associated virus (AAV) that knockdown or overexpression GADD45B were injected into ventral hippocampal CA1 (vCA1) by stereotactic, and verified by fluorescence and Western blot. The contextual fear conditioning paradigm was employed to examine the involvement of GADD45B in modulating aversive memory acquisition. The Y-maze and novel location recognition (NLR) tests were used to examine non-aversive cognition. The synaptic plasticity and electrophysiological properties of neurons were measured by slice patch clamp. KEY FINDINGS: Knockdown of GADD45B in the vCA1 significantly enhanced fear memory acquisition, accompanied by an upregulation of long-term potentiation (LTP) expression and intrinsic excitability of vCA1 pyramidal neurons (PNs). Conversely, overexpression of GADD45B produced the opposite effects. Notably, silencing the activity of vCA1 neurons abolished the impact of GADD45B knockdown on fear memory development. Moreover, mice with vCA1 GADD45B overexpression exhibited impaired spatial cognition, whereas mice with GADD45B knockdown did not display such impairment. SIGNIFICANCE: These results provided compelling evidence for the crucial involvement of GADD45B in the formation of aversive memory and spatial cognition.


Subject(s)
CA1 Region, Hippocampal , Fear , GADD45 Proteins , Mice, Inbred C57BL , Animals , Male , Fear/physiology , Mice , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Cognition/physiology , Memory/physiology , Long-Term Potentiation/physiology , Maze Learning/physiology , Neuronal Plasticity/physiology , Antigens, Differentiation/metabolism , Antigens, Differentiation/genetics , Gene Knockdown Techniques
4.
Cell Rep ; 43(4): 114115, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38607918

ABSTRACT

In the CA1 hippocampus, vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) play a prominent role in disinhibitory circuit motifs. However, the specific behavioral conditions that lead to circuit disinhibition remain uncertain. To investigate the behavioral relevance of VIP-IN activity, we employed wireless technologies allowing us to monitor and manipulate their function in freely behaving mice. Our findings reveal that, during spatial exploration in new environments, VIP-INs in the CA1 hippocampal region become highly active, facilitating the rapid encoding of novel spatial information. Remarkably, both VIP-INs and pyramidal neurons (PNs) exhibit increased activity when encountering novel changes in the environment, including context- and object-related alterations. Concurrently, somatostatin- and parvalbumin-expressing inhibitory populations show an inverse relationship with VIP-IN and PN activity, revealing circuit disinhibition that occurs on a timescale of seconds. Thus, VIP-IN-mediated disinhibition may constitute a crucial element in the rapid encoding of novelty and the acquisition of recognition memory.


Subject(s)
CA1 Region, Hippocampal , Interneurons , Recognition, Psychology , Vasoactive Intestinal Peptide , Animals , Interneurons/metabolism , Interneurons/physiology , Vasoactive Intestinal Peptide/metabolism , CA1 Region, Hippocampal/physiology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/cytology , Mice , Male , Recognition, Psychology/physiology , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Mice, Inbred C57BL , Memory/physiology , Parvalbumins/metabolism , Exploratory Behavior/physiology , Somatostatin/metabolism
5.
J Neurosci Res ; 102(4): e25333, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38656542

ABSTRACT

Novelty influences hippocampal-dependent memory through metaplasticity. Mismatch novelty detection activates the human hippocampal CA1 area and enhances rat hippocampal-dependent learning and exploration. Remarkably, mismatch novelty training (NT) also enhances rodent hippocampal synaptic plasticity while inhibition of VIP interneurons promotes rodent exploration. Since VIP, acting on VPAC1 receptors (Rs), restrains hippocampal LTP and depotentiation by modulating disinhibition, we now investigated the impact of NT on VPAC1 modulation of hippocampal synaptic plasticity in male Wistar rats. NT enhanced both CA1 hippocampal LTP and depotentiation unlike exploring an empty holeboard (HT) or a fixed configuration of objects (FT). Blocking VIP VPAC1Rs with PG 97269 (100 nM) enhanced both LTP and depotentiation in naïve animals, but this effect was less effective in NT rats. Altered endogenous VIP modulation of LTP was absent in animals exposed to the empty environment (HT). HT and FT animals showed mildly enhanced synaptic VPAC1R levels, but neither VIP nor VPAC1R levels were altered in NT animals. Conversely, NT enhanced the GluA1/GluA2 AMPAR ratio and gephyrin synaptic content but not PSD-95 excitatory synaptic marker. In conclusion, NT influences hippocampal synaptic plasticity by reshaping brain circuits modulating disinhibition and its control by VIP-expressing hippocampal interneurons while upregulation of VIP VPAC1Rs is associated with the maintenance of VIP control of LTP in FT and HT animals. This suggests VIP receptor ligands may be relevant to co-adjuvate cognitive recovery therapies in aging or epilepsy, where LTP/LTD imbalance occurs.


Subject(s)
Exploratory Behavior , Hippocampus , Neuronal Plasticity , Receptors, Vasoactive Intestinal Polypeptide, Type I , Vasoactive Intestinal Peptide , Animals , Male , Rats , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Exploratory Behavior/physiology , Hippocampus/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Rats, Wistar , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Vasoactive Intestinal Peptide/metabolism
6.
Mar Drugs ; 22(4)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38667787

ABSTRACT

Porphyran, a sulfated polysaccharide found in various species of marine red algae, has been demonstrated to exhibit diverse bioactivities, including anti-inflammatory effects. However, the protective effects of porphyran against cerebral ischemia and reperfusion (IR) injury have not been investigated. The aim of this study was to examine the neuroprotective effects of porphyran against brain IR injury and its underlying mechanisms using a gerbil model of transient forebrain ischemia (IR in the forebrain), which results in pyramidal cell (principal neuron) loss in the cornu ammonis 1 (CA1) subregion of the hippocampus on day 4 after IR. Porphyran (25 and 50 mg/kg) was orally administered daily for one week prior to IR. Pretreatment with 50 mg/kg of porphyran, but not 25 mg/kg, significantly attenuated locomotor hyperactivity and protected pyramidal cells located in the CA1 area from IR injury. The pretreatment with 50 mg/kg of porphyran significantly suppressed the IR-induced activation and proliferation of microglia in the CA1 subregion. Additionally, the pretreatment significantly inhibited the overexpressions of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing protein-3 (NLRP3) inflammasome complex, and pro-inflammatory cytokines (interleukin 1 beta and interleukin 18) induced by IR in the CA1 subregion. Overall, our findings suggest that porphyran exerts neuroprotective effects against brain IR injury, potentially by reducing the reaction (activation) and proliferation of microglia and reducing NLRP3 inflammasome-mediated neuroinflammation.


Subject(s)
CA1 Region, Hippocampal , Gerbillinae , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Neuroinflammatory Diseases , Neuroprotective Agents , Reperfusion Injury , Sepharose/analogs & derivatives , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Neuroprotective Agents/pharmacology , Male , Reperfusion Injury/drug therapy , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/metabolism , Neuroinflammatory Diseases/drug therapy , Disease Models, Animal , Microglia/drug effects , Brain Ischemia/drug therapy , Polysaccharides/pharmacology , Neurons/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism
7.
J Neurosci ; 44(19)2024 May 08.
Article in English | MEDLINE | ID: mdl-38565288

ABSTRACT

Excitotoxicity and the concurrent loss of inhibition are well-defined mechanisms driving acute elevation in excitatory/inhibitory (E/I) balance and neuronal cell death following an ischemic insult to the brain. Despite the high prevalence of long-term disability in survivors of global cerebral ischemia (GCI) as a consequence of cardiac arrest, it remains unclear whether E/I imbalance persists beyond the acute phase and negatively affects functional recovery. We previously demonstrated sustained impairment of long-term potentiation (LTP) in hippocampal CA1 neurons correlating with deficits in learning and memory tasks in a murine model of cardiac arrest/cardiopulmonary resuscitation (CA/CPR). Here, we use CA/CPR and an in vitro ischemia model to elucidate mechanisms by which E/I imbalance contributes to ongoing hippocampal dysfunction in male mice. We reveal increased postsynaptic GABAA receptor (GABAAR) clustering and function in the CA1 region of the hippocampus that reduces the E/I ratio. Importantly, reduced GABAAR clustering observed in the first 24 h rebounds to an elevation of GABAergic clustering by 3 d postischemia. This increase in GABAergic inhibition required activation of the Ca2+-permeable ion channel transient receptor potential melastatin-2 (TRPM2), previously implicated in persistent LTP and memory deficits following CA/CPR. Furthermore, we find Ca2+-signaling, likely downstream of TRPM2 activation, upregulates Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, thereby driving the elevation of postsynaptic inhibitory function. Thus, we propose a novel mechanism by which inhibitory synaptic strength is upregulated in the context of ischemia and identify TRPM2 and CaMKII as potential pharmacological targets to restore perturbed synaptic plasticity and ameliorate cognitive function.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Mice, Inbred C57BL , Signal Transduction , TRPM Cation Channels , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Male , Mice , TRPM Cation Channels/metabolism , Signal Transduction/physiology , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Receptors, GABA-A/metabolism , Hippocampus/metabolism , Neural Inhibition/physiology , GABAergic Neurons/metabolism , Heart Arrest/complications , Heart Arrest/metabolism
8.
J Neurosci ; 44(17)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38514181

ABSTRACT

The initiation of abstinence after chronic drug self-administration is stressful. Cocaine-seeking behavior on the first day of the absence of the expected drug (Extinction Day 1, ED1) is reduced by blocking 5-HT signaling in dorsal hippocampal cornu ammonis 1 (CA1) in both male and female rats. We hypothesized that the experience of ED1 can substantially influence later relapse behavior and that dorsal raphe (DR) serotonin (5-HT) input to CA1 may be involved. We inhibited 5-HT1A/1B receptors (WAY-100635 plus GR-127935), or DR input (chemogenetics), in CA1 on ED1 to test the role of this pathway on cocaine-seeking persistence 2 weeks later. We also inhibited 5-HT1A or 5-HT1B receptors in CA1 during conditioned place preference (CPP) for cocaine, to examine mechanisms involved in the persistent effects of ED1 manipulations. Inhibition of DR inputs, or 5-HT1A/1B signaling, in CA1 decreased drug seeking on ED1 and decreased cocaine seeking 2 weeks later revealing that 5-HT signaling in CA1 during ED1 contributes to persistent drug seeking during abstinence. In addition, 5-HT1B antagonism alone transiently decreased drug-associated memory performance when given prior to a CPP test, whereas similar antagonism of 5-HT1A alone had no such effect but blocked CPP retrieval on a test 24 h later. These CPP findings are consistent with prior work showing that DR inputs to CA1 augment recall of the drug-associated context and drug seeking via 5-HT1B receptors and prevent consolidation of the updated nondrug context via 5-HT1A receptors. Thus, treatments that modulate 5-HT-dependent memory mechanisms in CA1 during initial abstinence may facilitate later maintenance of abstinence.


Subject(s)
Cocaine , Drug-Seeking Behavior , Oxadiazoles , Serotonin , Animals , Male , Drug-Seeking Behavior/physiology , Drug-Seeking Behavior/drug effects , Rats , Serotonin/metabolism , Female , Cocaine/administration & dosage , Cocaine/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , Pyridines/pharmacology , Serotonin Antagonists/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Piperazines/pharmacology , Rats, Sprague-Dawley , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/psychology , Self Administration , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Receptor, Serotonin, 5-HT1B/metabolism , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism
9.
Neuroscience ; 545: 148-157, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38513764

ABSTRACT

In this study, the electrophysiological and biochemical consequences of repeated exposure to morphine in male rats on glutamatergic synaptic transmission, synaptic plasticity, the expression of GABA receptors and glutamate receptors at the temporoammonic-CA1 synapse along the longitudinal axis of the hippocampus (dorsal, intermediate, ventral, DH, IH, VH, respectively) were investigated. Slice electrophysiological methods, qRT-PCR, and western blotting techniques were used to characterize synaptic plasticity properties. We showed that repeated morphine exposure (RME) reduced excitatory synaptic transmission and ability for long-term potentiation (LTP) in the VH as well as eliminated the dorsoventral difference in paired-pulse responses. A decreased expression of NR2B subunit in the VH and an increased expression GABAA receptor of α1 and α5 subunits in the DH were observed following RME. Furthermore, RME did not affect the expression of NR2A, AMPA receptor subunits, and γ2GABAA and GABAB receptors in either segment of the hippocampus. In sum, the impact of morphine may differ depending on the region of the hippocampus studied. A distinct change in the short- and long-term synaptic plasticity along the hippocampus long axis due to repeated morphine exposure, partially mediated by a change in the expression profile of glutamatergic receptor subunits. These findings can be useful in further understanding the cellular mechanism underlying deficits in information storage and, more generally, cognitive processes resulting from chronic opioid abuse.


Subject(s)
Morphine , Neuronal Plasticity , Rats, Sprague-Dawley , Animals , Male , Morphine/pharmacology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Rats , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Narcotics/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Receptors, GABA/metabolism , Receptors, GABA/drug effects
10.
Exp Neurol ; 376: 114752, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38484863

ABSTRACT

Dendritic spines play a pivotal role in synaptic communication and are crucial for learning and memory processes. Abnormalities in spine morphology and plasticity are observed in neurodevelopmental and neuropsychiatric disorders, yet the underlying signaling mechanisms remain poorly understood. The microtubule affinity regulating kinase 1 (MARK1) has been implicated in neurodevelopmental disorders, and the MARK1 gene shows accelerated evolution in the human lineage suggesting a role in cognition. However, the in vivo role of MARK1 in synaptogenesis and cognitive functions remains unknown. Here we show that forebrain-specific conditional knockout (cKO) of Mark1 in mice causes defects in dendritic spine morphogenesis in hippocampal CA1 pyramidal neurons with a significant reduction in spine density. In addition, we found loss of MARK1 causes synaptic accumulation of GKAP and GluA2. Furthermore, we found that MARK1 cKO mice show defects in spatial learning in the Morris water maze and reduced anxiety-like behaviors in the elevated plus maze. Taken together, our data show a novel role for MARK1 in regulating dendritic spine morphogenesis and cognitive functions in vivo.


Subject(s)
Cognition , Dendritic Spines , Mice, Knockout , Protein Serine-Threonine Kinases , Animals , Mice , Protein Serine-Threonine Kinases/genetics , Cognition/physiology , Maze Learning/physiology , Morphogenesis/physiology , Morphogenesis/genetics , Pyramidal Cells/metabolism , CA1 Region, Hippocampal/growth & development , CA1 Region, Hippocampal/metabolism , Male , Mice, Inbred C57BL
11.
Brain Res Bull ; 208: 110890, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38302069

ABSTRACT

Cognitive impairment is a major complication of cerebral ischemia-reperfusion (CIR) injury and has an important impact on the quality of life of patients. However, the precise mechanisms underlying cognitive impairment after CIR injury remain elusive. In the current study, we investigated the role of interleukin 17 A (IL-17A) on CIR injury-induced cognitive impairment in wild-type and IL-17A knockout mice using RNA sequencing analysis, neurological assessments, Golgi-Cox staining, dendritic spine analysis, immunofluorescence assay, and western blot analysis. RNA sequencing identified 195 CIR-induced differentially expressed genes (83 upregulated and 112 downregulated), highlighting several enriched biological processes (negative regulation of phosphorylation, transcription regulator complex, and receptor ligand activity) and signaling pathways (mitogen-activated protein kinase [MAPK], tumor necrosis factor, and IL-17 signaling pathways). We also injected adeno-associated virus into the bilateral hippocampal CA1 regions of CIR mice to upregulate or downregulate cyclic AMP response element-binding protein. IL-17A knockout activated the extracellular signal-regulated kinase (ERK)/MAPK signaling pathway and further improved synaptic plasticity, structure, and function in CIR mice. Together, our findings suggest that IL-17A deficiency alleviates CIR injury by activating the ERK/MAPK signaling pathway and enhancing hippocampal synaptic plasticity.


Subject(s)
Brain Ischemia , Reperfusion Injury , Humans , Animals , Mice , CA1 Region, Hippocampal/metabolism , Interleukin-17/metabolism , Quality of Life , Extracellular Signal-Regulated MAP Kinases/metabolism , Reperfusion Injury/metabolism
12.
Synapse ; 78(1): e22285, 2024 01.
Article in English | MEDLINE | ID: mdl-38287475

ABSTRACT

Agents that positively modulate the activity of α7nAChRs are used as cognitive enhancers and for the treatment of hippocampus-dependent functional decline. However, it is not known whether the expression and the effects of α7nAChRs apply to the entire longitudinal axis of the hippocampus equally. Given that cholinergic system-involving hippocampal functions are not equally distributed along the hippocampus, we comparatively examined the expression and the effects of α7nAChRs on excitatory synaptic transmission between the dorsal and the ventral hippocampal slices from adult rats. We found that α7nAChRs are equally expressed in the CA1 field of the two segments of the hippocampus. However, activation of α7nAChRs by their highly selective agonist PNU 282987 induced a gradually developing increase in field excitatory postsynaptic potential only in the dorsal hippocampus. This long-term potentiation was not reversed upon application of nonselective nicotinic receptor antagonist mecamylamine, but the induction of potentiation was prevented by prior blockade of α7nAChRs by their antagonist MG 624. In contrast to the long-term synaptic plasticity, we found that α7nAChRs did not modulate short-term synaptic plasticity in either the dorsal or the ventral hippocampus. These results may have implications for the role that α7nAChRs play in specifically modulating functions that depend on the normal function of the dorsal hippocampus. We propose that hippocampal functions that rely on a direct α7 nAChR-mediated persistent enhancement of glutamatergic synaptic transmission are preferably supported by dorsal but not ventral hippocampal synapses.


Subject(s)
Receptors, Nicotinic , alpha7 Nicotinic Acetylcholine Receptor , Rats , Animals , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Hippocampus/metabolism , CA1 Region, Hippocampal/metabolism , Receptors, Nicotinic/metabolism , Synaptic Transmission/physiology
13.
Brain Struct Funct ; 229(3): 639-655, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37690045

ABSTRACT

Hippocampal afferent inputs, terminating on proximal and distal subfields of the cornus ammonis (CA), enable the functional discrimination of 'what' (item identity) and 'where' (spatial location) elements of a spatial representation. This kind of information is supported by structures such as the retrosplenial cortex (RSC). Spatial content learning promotes the expression of hippocampal synaptic plasticity, particularly long-term depression (LTD). In the CA1 region, this is specifically facilitated by the learning of item-place features of a spatial environment. Gene-tagging, by means of time-locked fluorescence in situ hybridization (FISH) to detect nuclear expression of immediate early genes, can reveal neuronal populations that engage in experience-dependent information encoding. In the current study, using FISH, we examined if learning-facilitated LTD results in subfield-specific information encoding in the hippocampus and RSC. Rats engaged in novel exploration of small items during stimulation of Schaffer collateral-CA1 synapses. This resulted in LTD (> 24 h). FISH, to detect nuclear expression of Homer1a, revealed that the distal-CA1 and proximal-CA3 subcompartments were particularly activated by this event. By contrast, all elements of the proximodistal cornus ammonis-axis showed equal nuclear Homer1a expression following LTD induction solely by means of afferent stimulation. The RSC exhibited stronger nuclear Homer1a expression in response to learning-facilitated LTD, and to novel item-place experience, compared to LTD induced by sole afferent stimulation in CA1. These results show that both the cornus ammonis and RSC engage in differentiated information encoding of item-place learning that is salient enough, in its own right, to drive the expression of hippocampal LTD. These results also reveal a novel role of the RSC in item-place learning.


Subject(s)
Gyrus Cinguli , Long-Term Synaptic Depression , Rats , Animals , In Situ Hybridization, Fluorescence , Long-Term Synaptic Depression/physiology , Hippocampus/metabolism , Spatial Learning/physiology , Neuronal Plasticity , Synapses , Gene Expression , Long-Term Potentiation/physiology , CA1 Region, Hippocampal/metabolism
14.
Mol Neurobiol ; 61(4): 2336-2356, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37875707

ABSTRACT

Our previous study has proved that the Klotho up-regulation participated in cerebral ischemic preconditioning (CIP)-induced brain ischemic tolerance. However, the exact neuroprotective mechanism of Klotho in CIP remains unclear. We explored the hypothesis that STAT4-mediated Klotho up-regulation contributes to the CIP-induced brain ischemic tolerance via inhibiting neuronal pyroptosis. Firstly, the expressions of pyroptosis-associated proteins (i.e., NLRP3, GSDMD, pro-caspase-1, and cleaved caspase-1) in hippocampal CA1 region were determined during the process of brain ischemic tolerance. We found the expression of pyroptosis-associated proteins was significantly up-regulated in the ischemic insult (II) group, and showed no significant changes in the CIP group. The expression level of each pyroptosis-associated proteins was lower in the CIP + II group than that in the II group. Inhibition of Klotho expression increased the expression of pyroptosis-associated proteins in the CIP + II group and blocked the CIP-induced brain ischemic tolerance. Injection of Klotho protein decreased the expression of pyroptosis-associated proteins in the II group, and protected neurons from ischemic injury. Secondly, the transcription factor STAT4 of Klotho was identified by bioinformatic analysis. Double luciferase reporter gene assay and chromatin immunoprecipitation assay showed STAT4 can bind to the site between nt - 881 and - 868 on the Klotho promoter region and positively regulates Klotho expression. Moreover, we found CIP significantly enhanced the expression of STAT4. Knockdown STAT4 suppressed Klotho up-regulation after CIP and blocked the CIP-induced brain ischemic tolerance. Collectively, it can be concluded that STAT4-mediated the up-regulation of Klotho contributed to the brain ischemic tolerance induced by CIP via inhibiting pyroptosis.


Subject(s)
Brain Ischemia , Ischemic Preconditioning , Rats , Animals , Rats, Wistar , Up-Regulation , Pyroptosis , STAT4 Transcription Factor/metabolism , Brain Ischemia/metabolism , CA1 Region, Hippocampal/metabolism , Neurons/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
15.
CNS Neurosci Ther ; 30(4): e14508, 2024 04.
Article in English | MEDLINE | ID: mdl-37864452

ABSTRACT

AIMS: Exposure to crystalline silica (CS) in occupational settings induces chronic inflammation in the respiratory system and, potentially, the brain. Some workers are frequently concurrently exposed to both CS and nicotine. Here, we explored the impact of nicotine on CS-induced neuroinflammation in the mouse hippocampus. METHODS: In this study, we established double-exposed models of CS and nicotine in C57BL/6 mice. To assess depression-like behavior, experiments were conducted at 3, 6, and 9 weeks. Serum inflammatory factors were analyzed by ELISA. Hippocampus was collected for RNA sequencing analysis and examining the gene expression patterns linked to inflammation and cell death. Microglia and astrocyte activation and hippocampal neuronal death were assessed using immunohistochemistry and immunofluorescence staining. Western blotting was used to analyze the NF-κB expression level. RESULTS: Mice exposed to CS for 3 weeks showed signs of depression. This was accompanied by elevated IL-6 in blood, destruction of the blood-brain barrier, and activation of astrocytes caused by an increased NF-κB expression in the CA1 area of the hippocampus. The elevated levels of astrocyte-derived Lcn2 and upregulated genes related to inflammation led to higher neuronal mortality. Moreover, nicotine mitigated the NF-κB expression, astrocyte activation, and neuronal death, thereby ameliorating the associated symptoms. CONCLUSION: Silica exposure induces neuroinflammation and neuronal death in the mouse hippocampal CA1 region and depressive behavior. However, nicotine inhibits CS-induced neuroinflammation and neuronal apoptosis, alleviating depressive-like behaviors in mice.


Subject(s)
NF-kappa B , Nicotine , Mice , Animals , NF-kappa B/metabolism , Nicotine/pharmacology , Nicotine/metabolism , Astrocytes/metabolism , Neuroinflammatory Diseases , Mice, Inbred C57BL , Hippocampus/metabolism , CA1 Region, Hippocampal/metabolism , Inflammation/metabolism , Apoptosis , Microglia/metabolism
16.
Behav Pharmacol ; 35(2-3): 103-113, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37934654

ABSTRACT

Stress-induced antinociception (SIA) is due to the activation of several neural pathways and neurotransmitters that often suppress pain perception. Studies have shown that the orexin neuropeptide system is essential in pain modulation. Therefore, this study aimed to investigate the role of orexinergic receptors in the hippocampal CA1 region in modulating SIA response during the formalin test as an animal model of inflammatory pain. The orexin-1 receptor (OX1r) antagonist, SB334867, at 1, 3, 10, and 30 nmol or TCS OX2 29 as an orexin-2 receptor (OX2r) antagonist at the same doses were microinjected into the CA1 region in rats. Five minutes later, rats were exposed to restraint stress (RS) for 3 h, and pain-related behaviors were monitored in 5-min blocks for the 60-min test period in the formalin test. Results showed that applying RS for 3 h reduced pain responses in the early and late phases of the formalin test. The main findings showed that intra-CA1 injection of orexin receptor antagonists reduced the antinociception caused by stress in both phases of the formalin test. In addition, the contribution of OX2r in mediating the antinociceptive effect of stress was more prominent than that of OX1r in the early phase of the formalin test. However, in the late phase, both receptors worked similarly. Accordingly, the orexin system and its two receptors in the CA1 region of the hippocampus regulate SIA response to this animal model of pain in formalin test.


Subject(s)
CA1 Region, Hippocampal , Pain , Rats , Animals , Orexins/metabolism , Orexin Receptors/metabolism , Rats, Wistar , Pain Measurement , Carbachol/pharmacology , Pain/drug therapy , Pain/metabolism , CA1 Region, Hippocampal/metabolism , Orexin Receptor Antagonists/pharmacology
17.
Mol Brain ; 16(1): 73, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37848907

ABSTRACT

Calcium ions (Ca2+) play pivotal roles in regulating diverse brain functions, including cognition, emotion, locomotion, and learning and memory. These functions are intricately regulated by a variety of Ca2+-dependent cellular processes, encompassing synaptic plasticity, neuro/gliotransmitter release, and gene expression. In our previous work, we developed 'monster OptoSTIM1' (monSTIM1), an improved OptoSTIM1 that selectively activates Ca2+-release-activated Ca2+ (CRAC) channels in the plasma membrane through blue light, allowing precise control over intracellular Ca2+ signaling and specific brain functions. However, the large size of the coding sequence of monSTIM1 poses a limitation for its widespread use, as it exceeds the packaging capacity of adeno-associated virus (AAV). To address this constraint, we have introduced monSTIM1 variants with reduced coding sequence sizes and established AAV-based systems for expressing them in neurons and glial cells in the mouse brain. Upon expression by AAVs, these monSTIM1 variants significantly increased the expression levels of cFos in neurons and astrocytes in the hippocampal CA1 region following non-invasive light illumination. The use of monSTIM1 variants offers a promising avenue for investigating the spatiotemporal roles of Ca2+-mediated cellular activities in various brain functions. Furthermore, this toolkit holds potential as a therapeutic strategy for addressing brain disorders associated with aberrant Ca2+ signaling.


Subject(s)
Calcium Channels , Dependovirus , Mice , Animals , Dependovirus/metabolism , Optogenetics , CA1 Region, Hippocampal/metabolism , Learning , Calcium/metabolism , Calcium Signaling/physiology
18.
Aging (Albany NY) ; 15(20): 11227-11243, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37857016

ABSTRACT

Effective preventive measures against postoperative cognitive dysfunction in older adults are urgently needed. In this study, we investigated the effect of electroacupuncture (EA) on anesthesia and surgery-induced cognitive decline in aged rats by RNA-seq analysis, behavioral testing, Golgi-Cox staining, dendritic spine analysis, immunofluorescence assay and western blot analysis. EA ameliorated anesthesia and surgery induced-cognitive decline. RNA-seq analysis identified numerous differentially-expressed genes, including 353 upregulated genes and 563 downregulated genes, after pretreatment with EA in aged rats with postoperative cognitive dysfunction. To examine the role of CREB in EA, we injected adeno-associated virus (AAV) into the CA1 region of the hippocampus bilaterally into the aged rats to downregulate the transcription factor. EA improved synaptic plasticity, structurally and functionally, by activating the MAPK/ERK/CREB signaling pathway in aged rats. Together, our findings suggest that EA protects against anesthesia and surgery-induced cognitive decline in aged rats by activating the MAPK/ERK/CREB signaling pathway and enhancing hippocampal synaptic plasticity.


Subject(s)
Cognitive Dysfunction , Electroacupuncture , Postoperative Cognitive Complications , Rats , Animals , CA1 Region, Hippocampal/metabolism , Rats, Sprague-Dawley , Postoperative Cognitive Complications/metabolism , Hippocampus/metabolism , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/metabolism
19.
Nat Commun ; 14(1): 6100, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37773230

ABSTRACT

Memory consolidation involves discrete patterns of transcriptional events in the hippocampus. Despite the emergence of single-cell transcriptomic profiling techniques, mapping the transcriptomic signature across subregions of the hippocampus has remained challenging. Here, we utilized unbiased spatial sequencing to delineate transcriptome-wide gene expression changes across subregions of the dorsal hippocampus of male mice following learning. We find that each subregion of the hippocampus exhibits distinct yet overlapping transcriptomic signatures. The CA1 region exhibited increased expression of genes related to transcriptional regulation, while the DG showed upregulation of genes associated with protein folding. Importantly, our approach enabled us to define the transcriptomic signature of learning within two less-defined hippocampal subregions, CA1 stratum radiatum, and oriens. We demonstrated that CA1 subregion-specific expression of a transcription factor subfamily has a critical functional role in the consolidation of long-term memory. This work demonstrates the power of spatial molecular approaches to reveal simultaneous transcriptional events across the hippocampus during memory consolidation.


Subject(s)
Memory Consolidation , Transcriptome , Male , Mice , Animals , Transcriptome/genetics , Hippocampus/physiology , CA1 Region, Hippocampal/metabolism , Learning
20.
Int J Mol Sci ; 24(18)2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37762124

ABSTRACT

Chronic pain is a significant health problem worldwide. Recent evidence has suggested that the ventral hippocampus is dysfunctional in humans and rodents, with decreased neuronal excitability and connectivity with other brain regions, parallel pain chronicity, and persistent nociceptive hypersensitivity. But the molecular mechanisms underlying hippocampal modulation of pain remain poorly elucidated. In this study, we used ex vivo whole-cell patch-clamp recording, immunofluorescence staining, and behavioral tests to examine whether hyperpolarization-activated cyclic nucleotide-gated channels 2 (HCN2) in the ventral hippocampal CA1 (vCA1) were involved in regulating nociceptive perception and CFA-induced inflammatory pain in mice. Reduced sag potential and firing rate of action potentials were observed in vCA1 pyramidal neurons from CFA-injected mice. Moreover, the expression of HCN2, but not HCN1, in vCA1 decreased in mice injected with CFA. HCN2 knockdown in vCA1 pyramidal neurons induced thermal hypersensitivity, whereas overexpression of HCN2 alleviated thermal hyperalgesia induced by intraplantar injection of CFA in mice. Our findings suggest that HCN2 in the vCA1 plays an active role in pain modulation and could be a promising target for the treatment of chronic pain.


Subject(s)
Chronic Pain , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Potassium Channels , Animals , Mice , Action Potentials , Cyclic Nucleotide-Gated Cation Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Nociception , Potassium Channels/genetics , Potassium Channels/metabolism , CA1 Region, Hippocampal/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...