Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 902
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.
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
3.
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
4.
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
5.
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
6.
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
7.
Neuroimage ; 292: 120607, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38614372

RESUMEN

INTRODUCTION: In Alzheimer's disease (AD), early diagnosis facilitates treatment options and leads to beneficial outcomes for patients, their carers and the healthcare system. The neuropsychological battery of the Uniform Data Set (UDSNB3.0) assesses cognition in ageing and dementia, by measuring scores across different cognitive domains such as attention, memory, processing speed, executive function and language. However, its neuroanatomical correlates have not been investigated using 7 Tesla MRI (7T MRI). METHODS: We used 7T MRI to investigate the correlations between hippocampal subfield volumes and the UDSNB3.0 in 24 individuals with Amyloidß-status AD and 18 age-matched controls, with respective age ranges of 60 (42-76) and 62 (52-79) years. AD participants with a Medial Temporal Atrophy scale of higher than 2 on 3T MRI were excluded from the study. RESULTS: A significant difference in the entire hippocampal volume was observed in the AD group compared to healthy controls (HC), primarily influenced by CA1, the largest hippocampal subfield. Notably, no significant difference in whole brain volume between the groups implied that hippocampal volume loss was not merely reflective of overall brain atrophy. UDSNB3.0 cognitive scores showed significant differences between AD and HC, particularly in Memory, Language, and Visuospatial domains. The volume of the Dentate Gyrus (DG) showed a significant association with the Memory and Executive domain scores in AD patients as assessed by the UDSNB3.0.. The data also suggested a non-significant trend for CA1 volume associated with UDSNB3.0 Memory, Executive, and Language domain scores in AD. In a reassessment focusing on hippocampal subfields and MoCA memory subdomains in AD, associations were observed between the DG and Cued, Uncued, and Recognition Memory subscores, whereas CA1 and Tail showed associations only with Cued memory. DISCUSSION: This study reveals differences in the hippocampal volumes measured using 7T MRI, between individuals with early symptomatic AD compared with healthy controls. This highlights the potential of 7T MRI as a valuable tool for early AD diagnosis and the real-time monitoring of AD progression and treatment efficacy. CLINICALTRIALS: GOV: ID NCT04992975 (Clinicaltrial.gov 2023).


Asunto(s)
Enfermedad de Alzheimer , Región CA1 Hipocampal , Giro Dentado , Imagen por Resonancia Magnética , Trastornos de la Memoria , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/patología , Masculino , Imagen por Resonancia Magnética/métodos , Femenino , Anciano , Giro Dentado/diagnóstico por imagen , Giro Dentado/patología , Persona de Mediana Edad , Región CA1 Hipocampal/diagnóstico por imagen , Región CA1 Hipocampal/patología , Trastornos de la Memoria/diagnóstico por imagen , Trastornos de la Memoria/patología , Adulto , Péptidos beta-Amiloides/metabolismo
8.
J Alzheimers Dis ; 99(1): 121-143, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38640149

RESUMEN

Background: Previous work from our group has shown that chronic exposure to Vanadium pentoxide (V2O5) causes cytoskeletal alterations suggesting that V2O5 can interact with cytoskeletal proteins through polymerization and tyrosine phosphatases inhibition, causing Alzheimer's disease (AD)-like hippocampal cell death. Objective: This work aims to characterize an innovative AD experimental model through chronic V2O5 inhalation, analyzing the spatial memory alterations and the presence of neurofibrillary tangles (NFTs), amyloid-ß (Aß) senile plaques, cerebral amyloid angiopathy, and dendritic spine loss in AD-related brain structures. Methods: 20 male Wistar rats were divided into control (deionized water) and experimental (0.02 M V2O5 1 h, 3/week for 6 months) groups (n = 10). The T-maze test was used to assess spatial memory once a month. After 6 months, histological alterations of the frontal and entorhinal cortices, CA1, subiculum, and amygdala were analyzed by performing Congo red, Bielschowsky, and Golgi impregnation. Results: Cognitive results in the T-maze showed memory impairment from the third month of V2O5 inhalation. We also noted NFTs, Aß plaque accumulation in the vascular endothelium and pyramidal neurons, dendritic spine, and neuronal loss in all the analyzed structures, CA1 being the most affected. Conclusions: This model characterizes neurodegenerative changes specific to AD. Our model is compatible with Braak AD stage IV, which represents a moment where it is feasible to propose therapies that have a positive impact on stopping neuronal damage.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Modelos Animales de Enfermedad , Memoria Espacial , Compuestos de Vanadio , Animales , Masculino , Administración por Inhalación , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/patología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/patología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Angiopatía Amiloide Cerebral/inducido químicamente , Angiopatía Amiloide Cerebral/patología , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/patología , Corteza Entorrinal/efectos de los fármacos , Corteza Entorrinal/patología , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/patología , Aprendizaje por Laberinto/efectos de los fármacos , Ovillos Neurofibrilares/efectos de los fármacos , Ovillos Neurofibrilares/patología , Placa Amiloide/inducido químicamente , Placa Amiloide/patología , Ratas Wistar , Memoria Espacial/efectos de los fármacos , Compuestos de Vanadio/administración & dosificación , Compuestos de Vanadio/toxicidad
9.
Neuroscience ; 526: 204-222, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37385335

RESUMEN

Current data suggest a hypothesis of vascular pathogenesis for the development and progression of Alzheimer's disease (AD). To investigate this, we studied the association of apolipoprotein E4 (APOE4) gene on microvessels in human autopsy-confirmed AD with and without APOE4, compared with age/sex-matched control (AC) hippocampal CA1 stratum radiatum. AD arterioles (without APOE4 gene) had mild oxidative stress and loss of vascular endothelial growth factor (VEGF) and endothelial cell density, reflecting aging progression. In AD + APOE4, an increase in strong oxidative DNA damage marker 8-hydroxy-2'-deoxyguanosine (8-OHdG), VEGF, and endothelial cell density were associated with increased diameter of arterioles and perivascular space dilation. In cultured human brain microvascular cells (HBMECs), treatment of ApoE4 protein plus amyloid-ß (Aß) oligomers increased superoxide production and the apoptotic marker cleaved caspase 3, sustained hypoxia inducible factor-1α (HIF-1α) stability that was associated with an increase in MnSOD, VEGF, and cell density. This cell over-proliferation was inhibited with the antioxidants N-acetyl cysteine and MnTMPyP, the HIF-1α inhibitor echinomycin, the VEGFR-2 receptor blocker SU1498, the protein kinase C (PKC) ε knock-down (KD) and the extracellular signal-regulated kinase 1/2 (ERK) inhibitor FR180204. The PKCε KD and echinomycin decreased VEGF and/or ERK. In conclusion, AD capillaries and arterioles in hippocampal CA1 stratum radiatum of non-APOE4 carriers are related with aging, while those in APOE4 carriers with AD are related with pathogenesis of cerebrovascular disease.


Asunto(s)
Enfermedad de Alzheimer , Equinomicina , Humanos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E , Región CA1 Hipocampal/patología , Equinomicina/metabolismo , Hipocampo/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Exp Neurol ; 365: 114406, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37062352

RESUMEN

Structural and functional deficits in the hippocampus are a prominent feature of moderate-severe traumatic brain injury (TBI). In this work, we investigated the potential of Quantitative Susceptibility Imaging (QSM) to reveal the temporal changes in myelin integrity in a mouse model of concussion (mild TBI). We employed a cross-sectional design wherein we assigned 43 mice to cohorts undergoing either a concussive impact or a sham procedure, with QSM imaging at day 2, 7, or 14 post-injury, followed by Luxol Fast Blue (LFB) myelin staining to assess the structural integrity of hippocampal white matter (WM). We assessed spatial learning in the mice using the Active Place Avoidance Test (APA), recording their ability to use visual cues to locate and avoid zone-dependent mild electrical shocks. QSM and LFB staining indicated changes in the stratum lacunosum-molecular layer of the hippocampus in the concussion groups, suggesting impairment of this key relay between the entorhinal cortex and the CA1 regions. These imaging and histology findings were consistent with demyelination, namely increased magnetic susceptibility to MR imaging and decreased LFB staining. In the APA test, sham animals showed fewer entries into the shock zone compared to the concussed cohort. Thus, we present radiological, histological, and behavioral findings that concussion can induce significant and alterations in hippocampal integrity and function that evolve over time after the injury.


Asunto(s)
Conmoción Encefálica , Enfermedades Desmielinizantes , Modelos Animales de Enfermedad , Hipocampo , Fenómenos Magnéticos , Animales , Ratones , Conmoción Encefálica/patología , Estudios Transversales , Enfermedades Desmielinizantes/patología , Hipocampo/patología , Electrochoque , Aprendizaje Espacial , Sustancia Blanca/patología , Corteza Entorrinal/patología , Reacción de Prevención , Señales (Psicología) , Estimulación Luminosa , Región CA1 Hipocampal/patología , Masculino , Axones/patología , Región CA3 Hipocampal/patología
11.
J Neuropathol Exp Neurol ; 82(5): 402-411, 2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-36881691

RESUMEN

Inflammatory responses in the brain contribute to cognitive deficits. Nuclear factor-κB (NF-κB), a critical transcription factor in inflammatory responses, is activated in post-stroke cognitive deficit. Baihui (DU20) and Shenting (DU24) acupoints, the main acupoints of Du Meridian, are widely used to improve cognitive deficits in Chinese patients with stroke. It has been reported that post-stroke cognitive deficits can be treated by electroacupuncture (EA) but the underlying mechanisms of these effects are unclear. Using the rat middle cerebral artery occlusion cerebral ischemia-reperfusion injury model, we found that EA at these 2 acupoints improved neurological function, decreased cerebral infarct lesion volumes, and ameliorated the inflammatory response in the hippocampal CA1 region. The treatment also ameliorated memory and learning deficits by inhibiting the NF-κB signaling pathway in the ischemic hippocampal CA 1 region. This coincided with downregulation of interleukin-1ß, interleukin-6, CD45, and tumor necrosis factor-α. We conclude that EA at these 2 acupoints ameliorates memory and learning deficits following experimental cerebral infarction by inhibiting NF-κB-mediated inflammatory injury in the hippocampal CA1 region.


Asunto(s)
Isquemia Encefálica , Electroacupuntura , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , Accidente Cerebrovascular , Ratas , Animales , FN-kappa B/metabolismo , Accidente Cerebrovascular Isquémico/complicaciones , Accidente Cerebrovascular Isquémico/terapia , Ratas Sprague-Dawley , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/terapia , Isquemia Encefálica/complicaciones , Isquemia Encefálica/terapia , Isquemia Encefálica/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/terapia , Infarto de la Arteria Cerebral Media/metabolismo , Región CA1 Hipocampal/patología , Daño por Reperfusión/complicaciones , Daño por Reperfusión/terapia , Daño por Reperfusión/metabolismo
12.
Brain Pathol ; 33(1): e13109, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35794855

RESUMEN

Epigenetic modification contributes to the pathogenesis of cerebral ischemia. Piwil2 belongs to the PIWI proteins subfamily and has a key role in the regulation of gene transcription through epigenetics. However, the roles of Piwil2 in cerebral ischemia have not been investigated. In this study, we aim to elucidate the roles and the underlying molecular mechanisms of Piwil2 in ischemic tolerance induced by hypoxic postconditioning (HPC) against transient global cerebral ischemia (tGCI). We found that the expression of Piwil2 in CA1 was downregulated by HPC after tGCI. Silencing Piwil2 with antisense oligodeoxynucleotide (AS-ODN) in CA1 after tGCI decreased the expression of apoptosis-related proteins and exerted neuroprotective effects. Opposite results were observed after overexpression of Piwil2 induced by administration of Piwil2-carried lentivirus. Furthermore, we revealed differentially expressed Piwil2-interacting piRNAs in CA1 between HPC and tGCI groups by RNA binding protein immunoprecipitation (RIP) assay. Moreover, downregulating Piwil2 induced by HPC or AS-ODN after tGCI caused a marked reduction of DNA methyltransferase 3A (DNMT3A), which in turn abolished the tGCI-induced increase in the DNA methylation of cyclic AMP response element-binding 2 (CREB2), thus increasing mRNA and protein of CREB2. Finally, downregulating Piwil2 restored dendritic complexity and length, prevented the loss of dentritic spines, thereby improving cognitive function after tGCI. These data firstly reveal that Piwil2 plays an important part in HPC-mediated neuroprotection against cerebral ischemia through epigenetic regulation of CREB2.


Asunto(s)
Isquemia Encefálica , Ataque Isquémico Transitorio , Animales , Ratas , Isquemia Encefálica/patología , Región CA1 Hipocampal/patología , Infarto Cerebral/patología , Epigénesis Genética , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/prevención & control , Metilación , Ratas Wistar , Proteínas de Unión al ARN/metabolismo
13.
J Alzheimers Dis ; 90(4): 1501-1521, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36278355

RESUMEN

BACKGROUND: Many identified mechanisms could be upstream of the prominent amyloid-ß (Aß) plaques in Alzheimer's disease (AD). OBJECTIVE: To profile the progression of pathology in AD. METHODS: We monitored metabolic signaling, redox stress, intraneuronal amyloid-ß (iAß) accumulation, and extracellular plaque deposition in the brains of 3xTg-AD mice across the lifespan. RESULTS: Intracellular accumulation of aggregated Aß in the CA1 pyramidal cells at 9 months preceded extracellular plaques that first presented in the CA1 at 16 months of age. In biochemical assays, brain glutathione (GSH) declined with age in both 3xTg-AD and non-transgenic controls, but the decline was accelerated in 3xTg-AD brains from 2 to 4 months. The decline in GSH correlated exponentially with the rise in iAß. Integrated metabolic signaling as the ratio of phospho-Akt (pAkt) to total Akt (tAkt) in the PI3kinase and mTOR pathway declined at 6, 9, and 12 months, before rising at 16 and 20 months. These pAkt/tAkt ratios correlated with both iAß and GSH levels in a U-shaped relationship. Selective vulnerability of age-related AD-genotype-specific pAkt changes was greatest in the CA1 pyramidal cell layer. To demonstrate redox causation, iAß accumulation was lowered in cultured middle-age adult 3xTg-AD neurons by treatment of the oxidized redox state in the neurons with exogenous cysteine. CONCLUSION: The order of pathologic progression in the 3xTg-AD mouse was loss of GSH (oxidative redox shift) followed by a pAkt/tAkt metabolic shift in CA1, iAß accumulation in CA1, and extracellular Aß deposition. Upstream targets may prove strategically more effective for therapy before irreversible changes.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Ratones Transgénicos , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Placa Amiloide/patología , Modelos Animales de Enfermedad , Región CA1 Hipocampal/patología , Glutatión/metabolismo
14.
Neurobiol Dis ; 172: 105820, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35843448

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder with growing major health impacts, particularly in countries with aging populations. The examination of neural circuit mechanisms in AD mouse models is a recent focus for identifying new AD treatment strategies. We hypothesize that age-progressive changes of both long-range and local hippocampal neural circuit connectivity occur in AD. Recent advancements in viral-genetic technologies provide new opportunities for semi-quantitative mapping of cell-type-specific neural circuit connections in AD mouse models. We applied a recently developed monosynaptic rabies tracing method to hippocampal neural circuit mapping studies in AD model mice to determine how local and global circuit connectivity to hippocampal CA1 excitatory neurons may be altered in the single APP knock-in (APP-KI) AD mouse model. To determine age-related AD progression, we measured circuit connectivity in age-matched littermate control and AD model mice at two different ages (3-4 vs. 10-11 months old). We quantitatively mapped the connectivity strengths of neural circuit inputs to hippocampal CA1 excitatory neurons from brain regions including hippocampal subregions, medial septum, subiculum and entorhinal cortex, comparing different age groups and genotypes. We focused on hippocampal CA1 because of its clear relationship with learning and memory and that the hippocampal formation shows clear neuropathological changes in human AD. Our results reveal alterations in circuit connectivity of hippocampal CA1 in AD model mice. Overall, we find weaker extrinsic CA1 input connectivity strengths in AD model mice compared with control mice, including sex differences of reduced subiculum to CA1 inputs in aged female AD mice compared with aged male AD mice. Unexpectedly, we find a connectivity pattern shift with an increased proportion of inputs from the CA3 region to CA1 excitatory neurons when comparing young and old AD model mice, as well as old wild-type mice and old AD model mice. These unexpected shifts in CA3-CA1 input proportions in this AD mouse model suggest the possibility that compensatory circuit increases may occur in response to connectivity losses in other parts of the hippocampal circuits. We expect that this work provides new insights into the neural circuit mechanisms of AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Virus de la Rabia , Anciano , Enfermedad de Alzheimer/patología , Animales , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Corteza Entorrinal/patología , Femenino , Hipocampo/patología , Humanos , Lactante , Masculino , Ratones , Ratones Transgénicos
15.
J Vasc Surg ; 75(1): 223-228, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34478810

RESUMEN

OBJECTIVE: We sought to determine whether extracranial carotid atherosclerotic disease (ECAD) is associated with increased key neurodegenerative pathology such as neurofibrillary tangle (NFT), beta-amyloid plaque, or cerebral amyloid angiopathy (CAA) accumulation, findings associated with Alzheimer's disease (AD) and other dementias. METHODS: Our prospective, longitudinal, clinicopathologic study, the AZSAND (Arizona study of aging and neurodegenerative disorders) and Brain and Body Donation Program, recorded the presence or absence of clinically diagnosed ECAD and performed semiquantitative density estimates of NFT, beta-amyloid plaque, and CAA at death. After adjusting for potential confounding factors determined by logistic regression analysis, histopathology density scores were evaluated in individuals with ECAD (n = 66) and those without ECAD (n = 125). RESULTS: We found that the presence of ECAD was associated with a 21% greater NFT burden at death compared with no ECAD (P = .02). Anatomically, an increased NFT burden was seen throughout the brain regions evaluated but was significant in the temporal lobe (P < .05) and entorhinal cortex (P = .02). In addition, we found that subjects who had undergone carotid endarterectomy (CEA), the surgical treatment of ECAD (n = 32), had decreased NFT densities compared with those with ECAD who had not undergone CEA (n = 66; P = .04). In contrast to NFT, ECAD was not associated with beta-amyloid plaques or CAA density. CONCLUSIONS: These findings indicate that ECAD is associated with the NFT burden in the temporal lobe and entorhinal cortex, which has clinical significance for AD and non-AD dementias and cognitive dysfunction. Further understanding of whether ECAD increases the risk of neurodegenerative brain changes is highly relevant because ECAD is a treatable disease that has not, otherwise, been evaluated for nor specifically treated as a dementia risk factor.


Asunto(s)
Enfermedad de Alzheimer/epidemiología , Enfermedades de las Arterias Carótidas/epidemiología , Angiopatía Amiloide Cerebral/epidemiología , Disfunción Cognitiva/epidemiología , Ovillos Neurofibrilares/patología , Placa Amiloide/epidemiología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/patología , Región CA1 Hipocampal/patología , Angiopatía Amiloide Cerebral/diagnóstico , Angiopatía Amiloide Cerebral/patología , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/patología , Corteza Entorrinal/patología , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Placa Amiloide/diagnóstico , Placa Amiloide/patología , Estudios Prospectivos , Medición de Riesgo/estadística & datos numéricos , Factores de Riesgo
16.
Cell Rep ; 37(10): 110094, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34879272

RESUMEN

Cognitive impairment (CI) is a disabling concomitant of multiple sclerosis (MS) with a complex and controversial pathogenesis. The cytokine interleukin-17A (IL-17A) is involved in the immune pathogenesis of MS, but its possible effects on synaptic function and cognition are still largely unexplored. In this study, we show that the IL-17A receptor (IL-17RA) is highly expressed by hippocampal neurons in the CA1 area and that exposure to IL-17A dose-dependently disrupts hippocampal long-term potentiation (LTP) through the activation of its receptor and p38 mitogen-activated protein kinase (MAPK). During experimental autoimmune encephalomyelitis (EAE), IL-17A overexpression is paralleled by hippocampal LTP dysfunction. An in vivo behavioral analysis shows that visuo-spatial learning abilities are preserved when EAE is induced in mice lacking IL-17A. Overall, this study suggests a key role for the IL-17 axis in the neuro-immune cross-talk occurring in the hippocampal CA1 area and its potential involvement in synaptic dysfunction and MS-related CI.


Asunto(s)
Conducta Animal , Región CA1 Hipocampal/metabolismo , Cognición , Encefalomielitis Autoinmune Experimental/metabolismo , Interleucina-17/metabolismo , Plasticidad Neuronal , Receptores de Interleucina-17/metabolismo , Sinapsis/metabolismo , Animales , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Encefalomielitis Autoinmune Experimental/psicología , Interleucina-17/genética , Potenciación a Largo Plazo , Masculino , Ratones Biozzi , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-17/genética , Transducción de Señal , Aprendizaje Espacial , Sinapsis/patología , Proteínas Quinasas p38 Activadas por Mitógenos
17.
Stem Cell Reports ; 16(12): 3005-3019, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34861165

RESUMEN

New neurons are abnormal in the adult hippocampus of Alzheimer's disease (AD) mouse models. The effects of modulating adult neurogenesis on AD pathogenesis differ from study to study. We reported recently that ablation of adult neural stem cells (aNSCs) was associated with improved memory in AD models. Here, we found that long-term potentiation (LTP) was improved in the hippocampus of APP/PS1 mice after ablation of aNSCs. This effect was confirmed in hAPP-J20 mice, a second AD mouse model. On the other hand, we found that exposure to enriched environment (EE) dramatically increased the number of DCX+ neurons, promoted dendritic growth, and affected the location of newborn neurons in the dentate gyrus of APP/PS1 mice, and EE exposure significantly ameliorated memory deficits in APP/PS1 mice. Together, our data suggest that both inhibiting abnormal adult neurogenesis and enhancing healthy adult neurogenesis could be beneficial for AD, and they are not mutually exclusive.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/fisiopatología , Cognición/fisiología , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Potenciación a Largo Plazo , Ratones Transgénicos , Células-Madre Neurales/metabolismo , Presenilina-1/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de GABA-A/metabolismo , Memoria Espacial
18.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830337

RESUMEN

The hippocampus is a primary area for contextual memory, known to process spatiotemporal information within a specific episode. Long-term strengthening of glutamatergic transmission is a mechanism of contextual learning in the dorsal cornu ammonis 1 (CA1) area of the hippocampus. CA1-specific immobilization or blockade of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptor delivery can impair learning performance, indicating a causal relationship between learning and receptor delivery into the synapse. Moreover, contextual learning also strengthens GABAA (gamma-aminobutyric acid) receptor-mediated inhibitory synapses onto CA1 neurons. Recently we revealed that strengthening of GABAA receptor-mediated inhibitory synapses preceded excitatory synaptic plasticity after contextual learning, resulting in a reduced synaptic excitatory/inhibitory (E/I) input balance that returned to pretraining levels within 10 min. The faster plasticity at inhibitory synapses may allow encoding a contextual memory and prevent cognitive dysfunction in various hippocampal pathologies. In this review, we focus on the dynamic changes of GABAA receptor mediated-synaptic currents after contextual learning and the intracellular mechanism underlying rapid inhibitory synaptic plasticity. In addition, we discuss that several pathologies, such as Alzheimer's disease, autism spectrum disorders and epilepsy are characterized by alterations in GABAA receptor trafficking, synaptic E/I imbalance and neuronal excitability.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Trastorno del Espectro Autista/metabolismo , Región CA1 Hipocampal/metabolismo , Epilepsia/metabolismo , Receptores AMPA/genética , Receptores de GABA-A/genética , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Región CA1 Hipocampal/patología , Cognición/fisiología , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Epilepsia/patología , Regulación de la Expresión Génica , Humanos , Aprendizaje/fisiología , Plasticidad Neuronal/genética , Neuronas , Nootrópicos/uso terapéutico , Transporte de Proteínas , Receptores AMPA/metabolismo , Receptores de GABA-A/metabolismo , Sinapsis , Transmisión Sináptica
19.
Int J Mol Sci ; 22(20)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34681766

RESUMEN

G protein-gated inwardly rectifying K+ (GIRK) channels are the main targets controlling excitability and synaptic plasticity on hippocampal neurons. Consequently, dysfunction of GIRK-mediated signalling has been implicated in the pathophysiology of Alzheimer´s disease (AD). Here, we provide a quantitative description on the expression and localisation patterns of GIRK2 in two transgenic mice models of AD (P301S and APP/PS1 mice), combining histoblots and immunoelectron microscopic approaches. The histoblot technique revealed differences in the expression of GIRK2 in the two transgenic mice models. The expression of GIRK2 was significantly reduced in the hippocampus of P301S mice in a laminar-specific manner at 10 months of age but was unaltered in APP/PS1 mice at 12 months compared to age-matched wild type mice. Ultrastructural approaches using the pre-embedding immunogold technique, demonstrated that the subcellular localisation of GIRK2 was significantly reduced along the neuronal surface of CA1 pyramidal cells, but increased in its frequency at cytoplasmic sites, in both P301S and APP/PS1 mice. We also found a decrease in plasma membrane GIRK2 channels in axon terminals contacting dendritic spines of CA1 pyramidal cells in P301S and APP/PS1 mice. These data demonstrate for the first time a redistribution of GIRK channels from the plasma membrane to intracellular sites in different compartments of CA1 pyramidal cells. Altogether, the pre- and post-synaptic reduction of GIRK2 channels suggest that GIRK-mediated alteration of the excitability in pyramidal cells could contribute to the cognitive dysfunctions as described in the two AD animal models.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Hipocampo/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Hipocampo/patología , Masculino , Ratones Transgénicos , Plasticidad Neuronal/fisiología , Presenilina-1/genética , Proteínas tau/genética
20.
Int J Mol Sci ; 22(19)2021 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-34638676

RESUMEN

Cerebral ischemia induces an inhibition of protein synthesis and causes cell death and neuronal deficits. These deleterious effects do not occur in resilient areas of the brain, where protein synthesis is restored. In cellular stress conditions, as brain ischemia, translational repressors named eukaryotic initiation factor (eIF) 4E-binding proteins (4E-BPs) specifically bind to eIF4E and are critical in the translational control. We previously described that 4E-BP2 protein, highly expressed in brain, can be a molecular target for the control of cell death or survival in the reperfusion after ischemia in an animal model of transient cerebral ischemia. Since these previous studies showed that phosphorylation would not be the regulation that controls the binding of 4E-BP2 to eIF4E under ischemic stress, we decided to investigate the differential detection of 4E-BP2-interacting proteins in two brain regions with different vulnerability to ischemia-reperfusion (IR) in this animal model, to discover new potential 4E-BP2 modulators and biomarkers of cerebral ischemia. For this purpose, 4E-BP2 immunoprecipitates from the resistant cortical region and the vulnerable hippocampal cornu ammonis 1 (CA1) region were analyzed by two-dimensional (2-D) fluorescence difference in gel electrophoresis (DIGE), and after a biological variation analysis, 4E-BP2-interacting proteins were identified by matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) mass spectrometry. Interestingly, among the 4E-BP2-interacting proteins identified, heat shock 70 kDa protein-8 (HSC70), dihydropyrimidinase-related protein-2 (DRP2), enolase-1, ubiquitin carboxyl-terminal hydrolase isozyme-L1 (UCHL1), adenylate kinase isoenzyme-1 (ADK1), nucleoside diphosphate kinase-A (NDKA), and Rho GDP-dissociation inhibitor-1 (Rho-GDI), were of notable interest, showing significant differences in their association with 4E-BP2 between resistant and vulnerable regions to ischemic stress. Our data contributes to the first characterization of the 4E-BP2 interactome, increasing the knowledge in the molecular basis of the protection and vulnerability of the ischemic regions and opens the way to detect new biomarkers and therapeutic targets for diagnosis and treatment of cerebral ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Muerte Celular/fisiología , Factores Eucarióticos de Iniciación/metabolismo , Neuronas/metabolismo , Daño por Reperfusión/metabolismo , Animales , Isquemia Encefálica/patología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Masculino , Neuronas/patología , Fosfoproteínas/metabolismo , Fosforilación/fisiología , Unión Proteica/fisiología , Biosíntesis de Proteínas/fisiología , Ratas , Ratas Wistar , Daño por Reperfusión/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA