Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.616
Filtrar
1.
PLoS Biol ; 22(7): e3002687, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38991663

RESUMEN

Reactive astrocytes are associated with neuroinflammation and cognitive decline in diverse neuropathologies; however, the underlying mechanisms are unclear. We used optogenetic and chemogenetic tools to identify the crucial roles of the hippocampal CA1 astrocytes in cognitive decline. Our results showed that repeated optogenetic stimulation of the hippocampal CA1 astrocytes induced cognitive impairment in mice and decreased synaptic long-term potentiation (LTP), which was accompanied by the appearance of inflammatory astrocytes. Mechanistic studies conducted using knockout animal models and hippocampal neuronal cultures showed that lipocalin-2 (LCN2), derived from reactive astrocytes, mediated neuroinflammation and induced cognitive impairment by decreasing the LTP through the reduction of neuronal NMDA receptors. Sustained chemogenetic stimulation of hippocampal astrocytes provided similar results. Conversely, these phenomena were attenuated by a metabolic inhibitor of astrocytes. Fiber photometry using GCaMP revealed a high level of hippocampal astrocyte activation in the neuroinflammation model. Our findings suggest that reactive astrocytes in the hippocampus are sufficient and required to induce cognitive decline through LCN2 release and synaptic modulation. This abnormal glial-neuron interaction may contribute to the pathogenesis of cognitive disturbances in neuroinflammation-associated brain conditions.


Asunto(s)
Astrocitos , Disfunción Cognitiva , Hipocampo , Lipocalina 2 , Potenciación a Largo Plazo , Enfermedades Neuroinflamatorias , Neuronas , Animales , Astrocitos/metabolismo , Astrocitos/patología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/etiología , Disfunción Cognitiva/patología , Lipocalina 2/metabolismo , Lipocalina 2/genética , Ratones , Hipocampo/metabolismo , Hipocampo/patología , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/metabolismo , Neuronas/metabolismo , Neuronas/patología , Ratones Noqueados , Masculino , Ratones Endogámicos C57BL , Receptores de N-Metil-D-Aspartato/metabolismo , Optogenética , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/metabolismo , Modelos Animales de Enfermedad
2.
Int J Mol Sci ; 25(13)2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-39000499

RESUMEN

General anesthetics may accelerate the neuropathological changes related to Alzheimer's disease (AD), of which amyloid beta (Aß)-induced toxicity is one of the main causes. However, the interaction of general anesthetics with different Aß-isoforms remains unclear. In this study, we investigated the effects of sevoflurane (0.4 and 1.2 maximal alveolar concentration (MAC)) on four Aß species-induced changes on dendritic spine density (DSD) in hippocampal brain slices of Thy1-eGFP mice and multiple epidermal growth factor-like domains 10 (MEGF10)-related astrocyte-mediated synaptic engulfment in hippocampal brain slices of C57BL/6 mice. We found that both sevoflurane and Aß downregulated CA1-dendritic spines. Moreover, compared with either sevoflurane or Aß alone, pre-treatment with Aß isoforms followed by sevoflurane application in general further enhanced spine loss. This enhancement was related to MEGF10-related astrocyte-dependent synaptic engulfment, only in AßpE3 + 1.2 MAC sevoflurane and 3NTyrAß + 1.2 MAC sevoflurane condition. In addition, removal of sevoflurane alleviated spine loss in Aß + sevoflurane. In summary, these results suggest that both synapses and astrocytes are sensitive targets for sevoflurane; in the presence of 3NTyrAß, 1.2 MAC sevoflurane alleviated astrocyte-mediated synaptic engulfment and exerted a lasting effect on dendritic spine remodeling.


Asunto(s)
Péptidos beta-Amiloides , Astrocitos , Región CA1 Hipocampal , Espinas Dendríticas , Ratones Endogámicos C57BL , Sevoflurano , Sinapsis , Sevoflurano/farmacología , Animales , Espinas Dendríticas/metabolismo , Espinas Dendríticas/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Ratones , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/citología , Masculino , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Anestésicos por Inhalación/farmacología
3.
Sci Adv ; 10(26): eadl2675, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38941473

RESUMEN

Declined memory is a hallmark of Alzheimer's disease (AD). Experiments in rodents and human postmortem studies suggest that serotonin (5-hydroxytryptamine, 5-HT) plays a role in memory, but the underlying mechanisms are unknown. Here, we investigate the role of 5-HT 2C receptor (5-HT2CR) in regulating memory. Transgenic mice expressing a humanized HTR2C mutation exhibit impaired plasticity of hippocampal ventral CA1 (vCA1) neurons and reduced memory. Further, 5-HT neurons project to and synapse onto vCA1 neurons. Disruption of 5-HT synthesis in vCA1-projecting neurons or deletion of 5-HT2CRs in the vCA1 impairs neural plasticity and memory. We show that a selective 5-HT2CR agonist, lorcaserin, improves synaptic plasticity and memory in an AD mouse model. Cumulatively, we demonstrate that hippocampal 5-HT2CR signaling regulates memory, which may inform the use of 5-HT2CR agonists in the treatment of dementia.


Asunto(s)
Enfermedad de Alzheimer , Memoria , Ratones Transgénicos , Plasticidad Neuronal , Receptor de Serotonina 5-HT2C , Animales , Humanos , Receptor de Serotonina 5-HT2C/metabolismo , Receptor de Serotonina 5-HT2C/genética , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Plasticidad Neuronal/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Serotonina/metabolismo , Modelos Animales de Enfermedad , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Agonistas del Receptor de Serotonina 5-HT2/farmacología
4.
STAR Protoc ; 5(2): 103110, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38843398

RESUMEN

The hippocampus has a major role in processing spatial information but has been found to encode non-spatial information from multisensory modalities in recent studies. Here, we present a protocol for recording non-spatial stimuli (visual, auditory, and a combination) that evoked calcium activity of hippocampal CA1 neuronal ensembles in C57BL/6 mice using a miniaturized fluorescence microscope. We describe steps for experimental apparatus setup, surgical procedures, software development, and neuronal population activity analysis. For complete details on the use and execution of this protocol, please refer to Sun et al.1.


Asunto(s)
Región CA1 Hipocampal , Calcio , Ratones Endogámicos C57BL , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Ratones , Calcio/metabolismo , Calcio/análisis , Microscopía Fluorescente/métodos , Neuronas/metabolismo , Neuronas/citología , Neuronas/fisiología , Masculino
5.
Bull Exp Biol Med ; 176(6): 736-742, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38907060

RESUMEN

Intranasal administration of total bovine brain gangliosides (6 mg/kg) to rats protected the CA1 hippocampal neurons from the death caused by two-vessel occlusion model (with hypotension) of forebrain ischemia/reperfusion injury. The immunohistochemical reaction of specific antibodies to marker proteins of activated microglia (Iba1) and astrocytes (GFAP) in hippocampal slices revealed the neuroprotective effect of exogenous gangliosides which can be mostly explained by their ability to suppress neuroinflammation and gliosis. The expression of neurotrophic factor BDNF in the CA1 region of hippocampus did not differ in sham-operated rats and animals exposed to ischemia/reperfusion. However, the administration of gangliosides increased the BDNF expression in both control and ischemic groups. The intranasal route of administration allows using lower concentrations of gangliosides preventing the death of hippocampal neurons.


Asunto(s)
Administración Intranasal , Factor Neurotrófico Derivado del Encéfalo , Región CA1 Hipocampal , Gangliósidos , Neuronas , Fármacos Neuroprotectores , Daño por Reperfusión , Animales , Daño por Reperfusión/patología , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Gangliósidos/farmacología , Ratas , Masculino , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/administración & dosificación , Ratas Wistar , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Microfilamentos/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Isquemia Encefálica/metabolismo , Prosencéfalo/efectos de los fármacos , Prosencéfalo/patología , Prosencéfalo/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad
6.
Behav Brain Res ; 470: 115094, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-38844057

RESUMEN

Estrogen receptor (ER) activation by 17-ß estradiol (E2) can attenuate neuronal injury and behavioral impairments following global cerebral ischemia (GCI) in rodents. This study sought to further examine the discrete roles of ERs through characterization of the effects of selective ER activation on post-ischemic pro-inflammatory microglial activation, hippocampal neuronal injury, and anxiety-like behaviors. Forty-six ovariectomized (OVX) adult female Wistar rats received daily s.c injections (100 µg/kg/day) of propylpyrazole triol (PPT; ERα agonist), diarylpropionitrile (DPN; ERß agonist), G-1 (G-protein coupled ER agonist; GPER), E2 (activating all receptors), or vehicle solution (VEH) for 21 days. After final injection, rats underwent GCI via 4-vessel occlusion (n=8 per group) or sham surgery (n=6, vehicle injections). The Open Field Test (OFT), Elevated Plus Maze (EPM), and Hole Board Test (HBT) assessed anxiety-like behaviors. Microglial activation (Iba1, CD68, CD86) in the basolateral amygdala (BLA), CA1 of the hippocampus, and paraventricular nucleus of the hypothalamus (PVN) was determined 8 days post-ischemia. Compared to sham rats, Iba1 activation and CA1 neuronal injury were increased in all ischemic groups except DPN-treated rats, with PPT-treated ischemic rats also showing increased PVN Iba1-ir expression. Behaviorally, VEH ischemic rats showed slightly elevated anxiety in the EPM compared to sham counterparts, with no significant effects of agonists. While no changes were observed in the OFT, emotion regulation via grooming in the HBT was increased in G-1 rats compared to E2 rats. Our findings support selective ER activation to regulate post-ischemic microglial activation and coping strategies in the HBT, despite minimal impact on hippocampal injury.


Asunto(s)
Ansiedad , Isquemia Encefálica , Región CA1 Hipocampal , Microglía , Fenoles , Pirazoles , Ratas Wistar , Animales , Femenino , Microglía/metabolismo , Microglía/efectos de los fármacos , Ratas , Ansiedad/metabolismo , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Isquemia Encefálica/metabolismo , Pirazoles/farmacología , Fenoles/farmacología , Ovariectomía , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Propionatos/farmacología , Propionatos/administración & dosificación , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Estradiol/farmacología , Modelos Animales de Enfermedad , Receptores de Estrógenos/metabolismo , Nitrilos/farmacología
7.
Nat Commun ; 15(1): 4531, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866749

RESUMEN

Individuals with autism spectrum disorder (ASD) have a higher prevalence of social memory impairment. A series of our previous studies revealed that hippocampal ventral CA1 (vCA1) neurons possess social memory engram and that the neurophysiological representation of social memory in the vCA1 neurons is disrupted in ASD-associated Shank3 knockout mice. However, whether the dysfunction of Shank3 in vCA1 causes the social memory impairment observed in ASD remains unclear. In this study, we found that vCA1-specific Shank3 conditional knockout (cKO) by the adeno-associated virus (AAV)- or specialized extracellular vesicle (EV)- mediated in vivo gene editing was sufficient to recapitulate the social memory impairment in male mice. Furthermore, the utilization of EV-mediated Shank3-cKO allowed us to quantitatively examine the role of Shank3 in social memory. Our results suggested that there is a certain threshold for the proportion of Shank3-cKO neurons required for social memory disruption. Thus, our study provides insight into the population coding of social memory in vCA1, as well as the pathological mechanisms underlying social memory impairment in ASD.


Asunto(s)
Trastorno del Espectro Autista , Región CA1 Hipocampal , Edición Génica , Memoria , Ratones Noqueados , Proteínas del Tejido Nervioso , Conducta Social , Animales , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Región CA1 Hipocampal/metabolismo , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Ratones , Memoria/fisiología , Neuronas/metabolismo , Dependovirus/genética , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Ratones Endogámicos C57BL
8.
eNeuro ; 11(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38886063

RESUMEN

Persistent activity in excitatory pyramidal cells (PYRs) is a putative mechanism for maintaining memory traces during working memory. We have recently demonstrated persistent interruption of firing in fast-spiking parvalbumin-expressing interneurons (PV-INs), a phenomenon that could serve as a substrate for persistent activity in PYRs through disinhibition lasting hundreds of milliseconds. Here, we find that hippocampal CA1 PV-INs exhibit type 2 excitability, like striatal and neocortical PV-INs. Modeling and mathematical analysis showed that the slowly inactivating potassium current KV1 contributes to type 2 excitability, enables the multiple firing regimes observed experimentally in PV-INs, and provides a mechanism for robust persistent interruption of firing. Using a fast/slow separation of times scales approach with the KV1 inactivation variable as a bifurcation parameter shows that the initial inhibitory stimulus stops repetitive firing by moving the membrane potential trajectory onto a coexisting stable fixed point corresponding to a nonspiking quiescent state. As KV1 inactivation decays, the trajectory follows the branch of stable fixed points until it crosses a subcritical Hopf bifurcation (HB) and then spirals out into repetitive firing. In a model describing entorhinal cortical PV-INs without KV1, interruption of firing could be achieved by taking advantage of the bistability inherent in type 2 excitability based on a subcritical HB, but the interruption was not robust to noise. Persistent interruption of firing is therefore broadly applicable to PV-INs in different brain regions but is only made robust to noise in the presence of a slow variable, KV1 inactivation.


Asunto(s)
Interneuronas , Modelos Neurológicos , Parvalbúminas , Parvalbúminas/metabolismo , Interneuronas/fisiología , Interneuronas/metabolismo , Animales , Potenciales de Acción/fisiología , Región CA1 Hipocampal/fisiología , Región CA1 Hipocampal/metabolismo , Inhibición Neural/fisiología , Células Piramidales/fisiología , Células Piramidales/metabolismo , Canales de Potasio de la Superfamilia Shaker/metabolismo , Corteza Entorrinal/fisiología , Corteza Entorrinal/metabolismo , Masculino
9.
Mol Autism ; 15(1): 28, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877552

RESUMEN

BACKGROUND: Mutations in the X-linked gene cyclin-dependent kinase-like 5 (CDKL5) cause a severe neurological disorder characterised by early-onset epileptic seizures, autism and intellectual disability (ID). Impaired hippocampal function has been implicated in other models of monogenic forms of autism spectrum disorders and ID and is often linked to epilepsy and behavioural abnormalities. Many individuals with CDKL5 deficiency disorder (CDD) have null mutations and complete loss of CDKL5 protein, therefore in the current study we used a Cdkl5-/y rat model to elucidate the impact of CDKL5 loss on cellular excitability and synaptic function of CA1 pyramidal cells (PCs). We hypothesised abnormal pre and/or post synaptic function and plasticity would be observed in the hippocampus of Cdkl5-/y rats. METHODS: To allow cross-species comparisons of phenotypes associated with the loss of CDKL5, we generated a loss of function mutation in exon 8 of the rat Cdkl5 gene and assessed the impact of the loss of CDLK5 using a combination of extracellular and whole-cell electrophysiological recordings, biochemistry, and histology. RESULTS: Our results indicate that CA1 hippocampal long-term potentiation (LTP) is enhanced in slices prepared from juvenile, but not adult, Cdkl5-/y rats. Enhanced LTP does not result from changes in NMDA receptor function or subunit expression as these remain unaltered throughout development. Furthermore, Ca2+ permeable AMPA receptor mediated currents are unchanged in Cdkl5-/y rats. We observe reduced mEPSC frequency accompanied by increased spine density in basal dendrites of CA1 PCs, however we find no evidence supporting an increase in silent synapses when assessed using a minimal stimulation protocol in slices. Additionally, we found no change in paired-pulse ratio, consistent with normal release probability at Schaffer collateral to CA1 PC synapses. CONCLUSIONS: Our data indicate a role for CDKL5 in hippocampal synaptic function and raise the possibility that altered intracellular signalling rather than synaptic deficits contribute to the altered plasticity. LIMITATIONS: This study has focussed on the electrophysiological and anatomical properties of hippocampal CA1 PCs across early postnatal development. Studies involving other brain regions, older animals and behavioural phenotypes associated with the loss of CDKL5 are needed to understand the pathophysiology of CDD.


Asunto(s)
Modelos Animales de Enfermedad , Potenciación a Largo Plazo , Proteínas Serina-Treonina Quinasas , Receptores AMPA , Receptores de N-Metil-D-Aspartato , Espasmos Infantiles , Animales , Masculino , Ratas , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Síndromes Epilépticos/genética , Síndromes Epilépticos/metabolismo , Potenciales Postsinápticos Excitadores , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Hipocampo/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Células Piramidales/metabolismo , Células Piramidales/patología , Receptores AMPA/metabolismo , Receptores AMPA/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Espasmos Infantiles/genética , Espasmos Infantiles/metabolismo , Sinapsis/metabolismo
10.
J Alzheimers Dis ; 99(4): 1303-1316, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38759018

RESUMEN

Background: Anxiety and social withdrawal are highly prevalent among patients with Alzheimer's disease (AD). However, the neural circuit mechanisms underlying these symptoms remain elusive, and there is a need for effective prevention strategies. Objective: This study aims to elucidate the neural circuitry mechanisms underlying social anxiety in AD. Methods: We utilized 5xFAD mice and conducted a series of experiments including optogenetic manipulation, Tandem Mass Tag-labeled proteome analysis, behavioral assessments, and immunofluorescence staining. Results: In 5xFAD mice, we observed significant amyloid-ß (Aß) accumulation in the anterior part of basolateral amygdala (aBLA). Behaviorally, 6-month-old 5xFAD mice displayed excessive social avoidance during social interaction. Concurrently, the pathway from aBLA to ventral hippocampal CA1 (vCA1) was significantly activated and exhibited a disorganized firing patterns during social interaction. By optogenetically inhibiting the aBLA-vCA1 pathway, we effectively improved the social ability of 5xFAD mice. In the presence of Aß accumulation, we identified distinct changes in the protein network within the aBLA. Following one month of administration of Urolithin A (UA), we observed significant restoration of the abnormal protein network within the aBLA. UA treatment also attenuated the disorganized firings of the aBLA-vCA1 pathway, leading to an improvement in social ability. Conclusions: The aBLA-vCA1 circuit is a vulnerable pathway in response to Aß accumulation during the progression of AD and plays a crucial role in Aß-induced social anxiety. Targeting the aBLA-vCA1 circuit and UA administration are both effective strategies for improving the Aß-impaired social ability.


Asunto(s)
Péptidos beta-Amiloides , Complejo Nuclear Basolateral , Región CA1 Hipocampal , Cumarinas , Ratones Transgénicos , Animales , Ratones , Péptidos beta-Amiloides/metabolismo , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Complejo Nuclear Basolateral/metabolismo , Complejo Nuclear Basolateral/efectos de los fármacos , Cumarinas/farmacología , Enfermedad de Alzheimer/metabolismo , Masculino , Conducta Social , Modelos Animales de Enfermedad , Ansiedad/metabolismo , Interacción Social/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Optogenética
11.
Am J Physiol Regul Integr Comp Physiol ; 327(1): R35-R45, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38708544

RESUMEN

Pregnancy is associated with neural and behavioral plasticity, systemic inflammation, and oxidative stress, yet the impact of inflammation and oxidative stress on maternal neural and behavioral plasticity during pregnancy is unclear. We hypothesized that healthy pregnancy transiently reduces learning and memory and these deficits are associated with pregnancy-induced elevations in inflammation and oxidative stress. Cognitive performance was tested with novel object recognition (recollective memory), Morris water maze (spatial memory), and open field (anxiety-like) behavior tasks in female Sprague-Dawley rats of varying reproductive states [nonpregnant (nulliparous), pregnant (near term), and 1-2 mo after pregnancy (primiparous); n = 7 or 8/group]. Plasma and CA1 proinflammatory cytokines were measured with a MILLIPLEX magnetic bead assay. Plasma oxidative stress was measured via advanced oxidation protein products (AOPP) assay. CA1 markers of oxidative stress, neuronal activity, and apoptosis were quantified via Western blot analysis. Our results demonstrate that CA1 oxidative stress-associated markers were elevated in pregnant compared with nulliparous rats (P ≤ 0.017) but there were equivalent levels in pregnant and primiparous rats. In contrast, reproductive state did not impact CA1 inflammatory cytokines, neuronal activity, or apoptosis. Likewise, there was no effect of reproductive state on recollective or spatial memory. Even so, spatial learning was impaired (P ≤ 0.007) whereas anxiety-like behavior (P ≤ 0.034) was reduced in primiparous rats. Overall, our data suggest that maternal hippocampal CA1 is protected from systemic inflammation but vulnerable to peripartum oxidative stress. Peripartum oxidative stress elevations, such as in pregnancy complications, may contribute to peripartum neural and behavioral plasticity.NEW & NOTEWORTHY Healthy pregnancy is associated with elevated maternal systemic and brain oxidative stress. During postpregnancy, brain oxidative stress remains elevated whereas systemic oxidative stress is resolved. This sustained maternal brain oxidative stress is associated with learning impairments and decreased anxiety-like behavior during the postpregnancy period.


Asunto(s)
Estrés Oxidativo , Ratas Sprague-Dawley , Animales , Femenino , Embarazo , Ratas , Inflamación/metabolismo , Inflamación/fisiopatología , Memoria , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiopatología , Memoria Espacial , Citocinas/metabolismo , Citocinas/sangre , Ansiedad/metabolismo , Neuronas/metabolismo , Aprendizaje por Laberinto , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/sangre
12.
Bioessays ; 46(7): e2400006, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38693811

RESUMEN

Long-term potentiation (LTP) of excitatory synapses is a leading model to explain the concept of information storage in the brain. Multiple mechanisms contribute to LTP, but central amongst them is an increased sensitivity of the postsynaptic membrane to neurotransmitter release. This sensitivity is predominantly determined by the abundance and localization of AMPA-type glutamate receptors (AMPARs). A combination of AMPAR structural data, super-resolution imaging of excitatory synapses, and an abundance of electrophysiological studies are providing an ever-clearer picture of how AMPARs are recruited and organized at synaptic junctions. Here, we review the latest insights into this process, and discuss how both cytoplasmic and extracellular receptor elements cooperate to tune the AMPAR response at the hippocampal CA1 synapse.


Asunto(s)
Potenciación a Largo Plazo , Receptores AMPA , Sinapsis , Receptores AMPA/metabolismo , Animales , Humanos , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología
13.
Sci Rep ; 14(1): 11713, 2024 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778177

RESUMEN

The development of neurons is regulated by several spatiotemporally changing factors, which are crucial to give the ability of neurons to form functional networks. While external physical stimuli may impact the early developmental stages of neurons, the medium and long-term consequences of these influences have yet to be thoroughly examined. Using an animal model, this study focuses on the morphological and transcriptome changes of the hippocampus that may occur as a consequence of fetal ultrasound examination. We selectively labeled CA1 neurons of the hippocampus with in-utero electroporation to analyze their morphological features. Furthermore, certain samples also went through RNA sequencing after repetitive ultrasound exposure. US exposure significantly changed several morphological properties of the basal dendritic tree. A notable increase was also observed in the density of spines on the basal dendrites, accompanied by various alterations in individual spine morphology. Transcriptome analysis revealed several up or downregulated genes, which may explain the molecular background of these alterations. Our results suggest that US-derived changes in the dendritic trees of CA1 pyramidal cells might be connected to modification of the transcriptome of the hippocampus and may lead to an increased dendritic input.


Asunto(s)
Región CA1 Hipocampal , Dendritas , Transcriptoma , Animales , Región CA1 Hipocampal/metabolismo , Dendritas/metabolismo , Femenino , Embarazo , Células Piramidales/metabolismo , Ratones , Hipocampo/metabolismo , Perfilación de la Expresión Génica , Espinas Dendríticas/metabolismo , Ultrasonografía Prenatal
14.
Cell Mol Biol Lett ; 29(1): 79, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783169

RESUMEN

BACKGROUND: Postoperative cognitive dysfunction (POCD) is a common complication after anesthesia/surgery, especially among elderly patients, and poses a significant threat to their postoperative quality of life and overall well-being. While it is widely accepted that elderly patients may experience POCD following anesthesia/surgery, the exact mechanism behind this phenomenon remains unclear. Several studies have indicated that the interaction between silent mating type information regulation 2 homologue 1 (SIRT1) and brain-derived neurotrophic factor (BDNF) is crucial in controlling cognitive function and is strongly linked to neurodegenerative disorders. Hence, this research aims to explore how SIRT1/BDNF impacts cognitive decline caused by anesthesia/surgery in aged mice. METHODS: Open field test (OFT) was used to determine whether anesthesia/surgery affected the motor ability of mice, while the postoperative cognitive function of 18 months old mice was evaluated with Novel object recognition test (NORT), Object location test (OLT) and Fear condition test (FC). The expressions of SIRT1 and other molecules were analyzed by western blot and immunofluorescence staining. The hippocampal synaptic plasticity was detected by Golgi staining and Long-term potentiation (LTP). The effects of SIRT1 and BDNF overexpression as well as chemogenetic activation of glutamatergic neurons in hippocampal CA1 region of 18 months old vesicular glutamate transporter 1 (VGLUT1) mice on POCD were further investigated. RESULTS: The research results revealed that older mice exhibited cognitive impairment following intramedullary fixation of tibial fracture. Additionally, a notable decrease in the expression of SIRT1/BDNF and neuronal excitability in hippocampal CA1 glutamatergic neurons was observed. By increasing levels of SIRT1/BDNF or enhancing glutamatergic neuron excitability in the CA1 region, it was possible to effectively mitigate synaptic plasticity impairment and ameliorate postoperative cognitive dysfunction. CONCLUSIONS: The decline in SIRT1/BDNF levels leading to changes in synaptic plasticity and neuronal excitability in older mice could be a significant factor contributing to cognitive impairment after anesthesia/surgery.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Región CA1 Hipocampal , Regulación hacia Abajo , Plasticidad Neuronal , Neuronas , Complicaciones Cognitivas Postoperatorias , Sirtuina 1 , Animales , Sirtuina 1/metabolismo , Sirtuina 1/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Ratones , Neuronas/metabolismo , Complicaciones Cognitivas Postoperatorias/metabolismo , Complicaciones Cognitivas Postoperatorias/etiología , Región CA1 Hipocampal/metabolismo , Masculino , Ratones Endogámicos C57BL , Potenciación a Largo Plazo , Ácido Glutámico/metabolismo , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología
15.
Eur J Pharmacol ; 975: 176638, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38734297

RESUMEN

The underlying mechanisms of macamide's neuroprotective effects in Alzheimer's disease (AD) were investigated in the paper. Macamides are considered as unique ingredients in maca. Improvement effects and mechanisms of macamide on cognitive impairment have not been revealed. In this study, Vina 1.1.2 was used for docking to evaluate the binding abilities of 12 main macamides to acetylcholinesterase (AChE). N-benzyl-(9Z,12Z)-octadecadienamide (M 18:2) was selected to study the following experiments because it can stably bind to AChE with a strong binding energy. The animal experiments showed that M 18:2 prevented the scopolamine (SCP)-induced cognitive impairment and neurotransmitter disorders, increased the positive rates of Nrf2 and HO-1 in hippocampal CA1, improved the synaptic plasticity by maintaining synaptic morphology and increasing the synapse density. Moreover, the contents of IL-1ß, IL-6, and TNF-α in the hippocampus, serum, and colon were reduced by M 18:2. Furthermore, M 18:2 promoted colonic epithelial integrity and partially restored the composition of the gut microbiota to normal, including decreased genera Clostridiales_unclassified and Lachnospiraceae_unclassified, as well as increased genera Muribaculaceae_unclassified, Muribaculum, Alistipes, and Bacteroides, which may be the possible biomarkers of cognitive aging. In summary, M 18:2 exerted neuroprotective effects on SCP-induced AD mice possibly via activating the Nrf2/HO-1 signaling pathway and modulating the gut microbiota.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Factor 2 Relacionado con NF-E2 , Fármacos Neuroprotectores , Transducción de Señal , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Microbioma Gastrointestinal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratones , Masculino , Acetilcolinesterasa/metabolismo , Escopolamina , Plasticidad Neuronal/efectos de los fármacos , Simulación del Acoplamiento Molecular , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología
16.
PLoS Comput Biol ; 20(5): e1012085, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38709845

RESUMEN

Alzheimer's Disease (AD) is characterized by a range of behavioral alterations, including memory loss and psychiatric symptoms. While there is evidence that molecular pathologies, such as amyloid beta (Aß), contribute to AD, it remains unclear how this histopathology gives rise to such disparate behavioral deficits. One hypothesis is that Aß exerts differential effects on neuronal circuits across brain regions, depending on the neurophysiology and connectivity of different areas. To test this, we recorded from large neuronal populations in dorsal CA1 (dCA1) and ventral CA1 (vCA1), two hippocampal areas known to be structurally and functionally diverse, in the APP/PS1 mouse model of amyloidosis. Despite similar levels of Aß pathology, dCA1 and vCA1 showed distinct disruptions in neuronal population activity as animals navigated a virtual reality environment. In dCA1, pairwise correlations and entropy, a measure of the diversity of activity patterns, were decreased in APP/PS1 mice relative to age-matched C57BL/6 controls. However, in vCA1, APP/PS1 mice had increased pair-wise correlations and entropy as compared to age matched controls. Finally, using maximum entropy models, we connected the microscopic features of population activity (correlations) to the macroscopic features of the population code (entropy). We found that the models' performance increased in predicting dCA1 activity, but decreased in predicting vCA1 activity, in APP/PS1 mice relative to the controls. Taken together, we found that Aß exerts distinct effects across different hippocampal regions, suggesting that the various behavioral deficits of AD may reflect underlying heterogeneities in neuronal circuits and the different disruptions that Aß pathology causes in those circuits.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Región CA1 Hipocampal , Animales , Masculino , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiopatología , Región CA1 Hipocampal/patología , Biología Computacional , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Presenilina-1/genética , Presenilina-1/metabolismo
17.
J Neurosci Res ; 102(4): e25333, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38656542

RESUMEN

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.


Asunto(s)
Conducta Exploratoria , Hipocampo , Plasticidad Neuronal , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo , Péptido Intestinal Vasoactivo , Animales , Masculino , Ratas , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Conducta Exploratoria/fisiología , Hipocampo/metabolismo , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Plasticidad Neuronal/fisiología , Ratas Wistar , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
18.
Neurobiol Aging ; 139: 20-29, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38583392

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Calcio , Disfunción Cognitiva , Modelos Animales de Enfermedad , Própolis , Animales , Própolis/uso terapéutico , Própolis/administración & dosificación , Própolis/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/psicología , Enfermedad de Alzheimer/etiología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/prevención & control , Disfunción Cognitiva/tratamiento farmacológico , Calcio/metabolismo , Ratones Transgénicos , Señalización del Calcio/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Péptidos beta-Amiloides/metabolismo , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Ratones
19.
Life Sci ; 346: 122618, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38614306

RESUMEN

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.


Asunto(s)
Región CA1 Hipocampal , Miedo , Proteinas GADD45 , Ratones Endogámicos C57BL , Animales , Masculino , Miedo/fisiología , Ratones , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Cognición/fisiología , Memoria/fisiología , Potenciación a Largo Plazo/fisiología , Aprendizaje por Laberinto/fisiología , Plasticidad Neuronal/fisiología , Antígenos de Diferenciación/metabolismo , Antígenos de Diferenciación/genética , Técnicas de Silenciamiento del Gen
20.
Mar Drugs ; 22(4)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38667787

RESUMEN

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.


Asunto(s)
Región CA1 Hipocampal , Gerbillinae , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedades Neuroinflamatorias , Fármacos Neuroprotectores , Daño por Reperfusión , Sefarosa/análogos & derivados , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Masculino , Daño por Reperfusión/tratamiento farmacológico , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Modelos Animales de Enfermedad , Microglía/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Polisacáridos/farmacología , Neuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA