Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.506
Filtrar
1.
Sci Rep ; 14(1): 10622, 2024 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-38724691

RESUMO

Reduced hippocampal volume occurs in major depressive disorder (MDD), potentially due to elevated glucocorticoids from an overactivated hypothalamus-pituitary-adrenal (HPA) axis. To examine this in humans, hippocampal volume and hypothalamus (HPA axis) metabolism was quantified in participants with MDD before and after antidepressant treatment. 65 participants (n = 24 males, n = 41 females) with MDD were treated in a double-blind, randomized clinical trial of escitalopram. Participants received simultaneous positron emission tomography (PET)/magnetic resonance imaging (MRI) before and after treatment. Linear mixed models examined the relationship between hippocampus/dentate gyrus volume and hypothalamus metabolism. Chi-squared tests and multivariable logistic regression examined the association between hippocampus/dentate gyrus volume change direction and hypothalamus activity change direction with treatment. Multiple linear regression compared these changes between remitter and non-remitter groups. Covariates included age, sex, and treatment type. No significant linear association was found between hippocampus/dentate gyrus volume and hypothalamus metabolism. 62% (38 of 61) of participants experienced a decrease in hypothalamus metabolism, 43% (27 of 63) of participants demonstrated an increase in hippocampus size (51% [32 of 63] for the dentate gyrus) following treatment. No significant association was found between change in hypothalamus activity and change in hippocampus/dentate gyrus volume, and this association did not vary by sex, medication, or remission status. As this multimodal study, in a cohort of participants on standardized treatment, did not find an association between hypothalamus metabolism and hippocampal volume, it supports a more complex pathway between hippocampus neurogenesis and hypothalamus metabolism changes in response to treatment.


Assuntos
Transtorno Depressivo Maior , Hipocampo , Hipotálamo , Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Humanos , Transtorno Depressivo Maior/metabolismo , Transtorno Depressivo Maior/diagnóstico por imagem , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/patologia , Masculino , Feminino , Hipotálamo/metabolismo , Hipotálamo/diagnóstico por imagem , Adulto , Hipocampo/metabolismo , Hipocampo/diagnóstico por imagem , Hipocampo/patologia , Pessoa de Meia-Idade , Método Duplo-Cego , Tomografia por Emissão de Pósitrons/métodos , Giro Denteado/metabolismo , Giro Denteado/diagnóstico por imagem , Giro Denteado/patologia , Citalopram/uso terapêutico , Sistema Hipotálamo-Hipofisário/metabolismo , Tamanho do Órgão
2.
J Physiol Sci ; 74(1): 30, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773373

RESUMO

Experiments measuring evoked potentials require flexible and rapid adjustment of stimulation and recording parameters. In this study, we have developed a recording system and an associated Android application that allow making such adjustments wirelessly. The system consists of 3 units: for stimulation, recording and control. Most of the modules in this system are custom made, although the stimulator and tablet are off-the-shelf products. When installed on the tablet, our Android application allows wireless communication with the control unit from a distance of 5 m. In testing, the recording unit had low internal noise and displayed signals faithfully. Upon receiving commands from the control unit, the stimulation unit produced precisely timed pulse outputs. Using this system, we were able to record evoked field potentials in the dentate gyrus of a rat; responses increased as expected with increasing stimulation pulse amplitude and duration.


Assuntos
Potenciais Evocados , Tecnologia sem Fio , Animais , Tecnologia sem Fio/instrumentação , Ratos , Potenciais Evocados/fisiologia , Masculino , Estimulação Elétrica/métodos , Giro Denteado/fisiologia
3.
Nat Commun ; 15(1): 4047, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38744873

RESUMO

Human hippocampal organoids (hHOs) derived from human induced pluripotent stem cells (hiPSCs) have emerged as promising models for investigating neurodegenerative disorders, such as schizophrenia and Alzheimer's disease. However, obtaining the electrical information of these free-floating organoids in a noninvasive manner remains a challenge using commercial multi-electrode arrays (MEAs). The three-dimensional (3D) MEAs developed recently acquired only a few neural signals due to limited channel numbers. Here, we report a hippocampal cyborg organoid (cyb-organoid) platform coupling a liquid metal-polymer conductor (MPC)-based mesh neuro-interface with hHOs. The mesh MPC (mMPC) integrates 128-channel multielectrode arrays distributed on a small surface area (~2*2 mm). Stretchability (up to 500%) and flexibility of the mMPC enable its attachment to hHOs. Furthermore, we show that under Wnt3a and SHH activator induction, hHOs produce HOPX+ and PAX6+ progenitors and ZBTB20+PROX1+ dentate gyrus (DG) granule neurons. The transcriptomic signatures of hHOs reveal high similarity to the developing human hippocampus. We successfully detect neural activities from hHOs via the mMPC from this cyb-organoid. Compared with traditional planar devices, our non-invasive coupling offers an adaptor for recording neural signals from 3D models.


Assuntos
Hipocampo , Células-Tronco Pluripotentes Induzidas , Organoides , Humanos , Organoides/metabolismo , Organoides/citologia , Hipocampo/citologia , Hipocampo/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Neurônios/metabolismo , Neurônios/citologia , Metais/química , Transcriptoma , Giro Denteado/citologia , Giro Denteado/metabolismo
4.
Neuroimage ; 292: 120607, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38614372

RESUMO

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).


Assuntos
Doença de Alzheimer , Região CA1 Hipocampal , Giro Denteado , Imageamento por Ressonância Magnética , Transtornos da Memória , Humanos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/patologia , Masculino , Imageamento por Ressonância Magnética/métodos , Feminino , Idoso , Giro Denteado/diagnóstico por imagem , Giro Denteado/patologia , Pessoa de Meia-Idade , Região CA1 Hipocampal/diagnóstico por imagem , Região CA1 Hipocampal/patologia , Transtornos da Memória/diagnóstico por imagem , Transtornos da Memória/patologia , Adulto , Peptídeos beta-Amiloides/metabolismo
5.
Sci Rep ; 14(1): 8104, 2024 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-38582752

RESUMO

GCaMP is a genetically encoded calcium indicator (GECI) widely used in neuroscience research. It measures intracellular Ca2+ level by fluorescence changes as it directly binds to Ca2+. In this process, the effect of this calcium buffer on the intracellular calcium signaling and cell physiology is often not taken into consideration. However, growing evidence from calcium imaging studies shows GCaMP expression under certain conditions can generate aberrant activity, such as seizures. In this study, we examined the effect of GCaMP6 expression in the dentate gyrus (DG) on epileptogenesis. We found that viral expression of GCaMP6s but not GCaMP6f in the DG induces tonic-clonic seizures several weeks after viral injection. Cell-type specific expression of GCaMP6s revealed the granule cells (GCs) as the key player in GCaMP6s-induced epilepsy. Finally, by using slice electrophysiology, we demonstrated that GCaMP6s expression increases neuronal excitability in the GCs. Together, this study highlights the ability of GCaMP6s in DG-associated epileptogenesis.


Assuntos
Cálcio , Neurônios , Humanos , Cálcio/metabolismo , Neurônios/metabolismo , Convulsões/genética , Convulsões/metabolismo , Sinalização do Cálcio , Cálcio da Dieta/metabolismo , Giro Denteado/metabolismo
6.
Cells ; 13(8)2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38667284

RESUMO

This study investigates the combined effects of the neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31-Pro34]NPY at a dose of 132 µg and Ketamine at 10 mg/Kg on cognitive functions and neuronal proliferation, against a backdrop where neurodegenerative diseases present an escalating challenge to global health systems. Utilizing male Sprague-Dawley rats in a physiological model, this research employed a single-dose administration of these compounds and assessed their impact 24 h after treatment on object-in-place memory tasks, alongside cellular proliferation within the dorsal hippocampus dentate gyrus. Methods such as the in situ proximity ligation assay and immunohistochemistry for proliferating a cell nuclear antigen (PCNA) and doublecortin (DCX) were utilized. The results demonstrated that co-administration significantly enhanced memory consolidation and increased neuronal proliferation, specifically neuroblasts, without affecting quiescent neural progenitors and astrocytes. These effects were mediated by the potential formation of NPY1R-TrkB heteroreceptor complexes, as suggested by receptor co-localization studies, although further investigation is required to conclusively prove this interaction. The findings also highlighted the pivotal role of brain-derived neurotrophic factor (BDNF) in mediating these effects. In conclusion, this study presents a promising avenue for enhancing cognitive functions and neuronal proliferation through the synergistic action of the NPY1R agonist and Ketamine, potentially via NPY1R-TrkB heteroreceptor complex formation, offering new insights into therapeutic strategies for neurodegenerative diseases.


Assuntos
Proliferação de Células , Cognição , Proteína Duplacortina , Ketamina , Neurônios , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G , Receptores de Neuropeptídeo Y , Receptores de Neuropeptídeos , Animais , Masculino , Ketamina/farmacologia , Ketamina/administração & dosagem , Cognição/efeitos dos fármacos , Ratos , Receptores de Neuropeptídeo Y/agonistas , Receptores de Neuropeptídeo Y/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proliferação de Células/efeitos dos fármacos , Receptor trkB/agonistas , Receptor trkB/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Giro Denteado/efeitos dos fármacos , Giro Denteado/metabolismo , Neurogênese/efeitos dos fármacos
7.
Biomolecules ; 14(4)2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38672524

RESUMO

Neuronal plasticity is a crucial mechanism for an adapting nervous system to change. It is shown to be regulated by perineuronal nets (PNNs), the condensed forms of the extracellular matrix (ECM) around neuronal bodies. By assessing the changes in the number, intensity, and structure of PNNs, the ultrastructure of the PNN mesh, and the expression of inhibitory and excitatory synaptic inputs on these neurons, we aimed to clarify the role of an ECM glycoprotein, tenascin-C (TnC), in the dorsal hippocampus. To enhance neuronal plasticity, TnC-deficient (TnC-/-) and wild-type (TnC+/+) young adult male mice were reared in an enriched environment (EE) for 8 weeks. Deletion of TnC in TnC-/- mice showed an ultrastructural reduction of the PNN mesh and an increased inhibitory input in the dentate gyrus (DG), and an increase in the number of PNNs with a rise in the inhibitory input in the CA2 region. EE induced an increased inhibitory input in the CA2, CA3, and DG regions; in DG, the change was also followed by an increased intensity of PNNs. No changes in PNNs or synaptic expression were found in the CA1 region. We conclude that the DG and CA2 regions emerged as focal points of alterations in PNNs and synaptogenesis with EE as mediated by TnC.


Assuntos
Matriz Extracelular , Hipocampo , Plasticidade Neuronal , Sinapses , Tenascina , Animais , Tenascina/metabolismo , Tenascina/genética , Masculino , Camundongos , Hipocampo/metabolismo , Matriz Extracelular/metabolismo , Sinapses/metabolismo , Camundongos Knockout , Neurônios/metabolismo , Camundongos Endogâmicos C57BL , Giro Denteado/metabolismo
8.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38673966

RESUMO

Activity-dependent neuroprotective protein (ADNP) is a neuroprotective protein essential for embryonic development, proper brain development, and neuronal plasticity. Its mutation causes the autism-like ADNP syndrome (also called the Helsmoortel-Van der Aa syndrome), characterized by neural developmental disorders and motor dysfunctions. Similar to the ADNP syndrome, the ADNP haploinsufficient mouse shows low synapse density, leading to motor and cognitive ability delays. Moderate physical activity (PA) has several neuroprotective and cognitive benefits, promoting neuronal survival, differentiation, neurogenesis, and plasticity. Until now, no study has investigated the effect of moderate exercise on ADNP expression and distribution in the rat brain. The aim of the current investigation was to study the effects of moderate exercise on the ADNP expression and neuronal activation measured by the microtubule protein ß-Tubulin III. In pursuit of this objective, twenty-four rats were selected and evenly distributed into two categories: sedentary control rats and rats exposed to moderate physical activity on a treadmill over a span of 12 weeks. Our results showed that moderate PA increases the expression of ADNP and ß-Tubulin III in the dentate gyrus (DG) hippocampal region and cerebellum. Moreover, we found a co-localization of ADNP and ß-Tubulin III in both DG and cerebellum, suggesting a direct association of ADNP with adult neuronal activation induced by moderate PA.


Assuntos
Encéfalo , Proteínas do Tecido Nervoso , Condicionamento Físico Animal , Animais , Masculino , Ratos , Encéfalo/metabolismo , Cerebelo/metabolismo , Giro Denteado/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética , Ratos Wistar
9.
Curr Biol ; 34(9): 2011-2019.e7, 2024 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-38636511

RESUMO

Environmental enrichment (EE) improves memory, particularly the ability to discriminate similar past experiences.1,2,3,4,5,6 The hippocampus supports this ability via pattern separation, the encoding of similar events using dissimilar memory representations.7 This is carried out in the dentate gyrus (DG) and CA3 subfields.8,9,10,11,12 Upregulation of adult neurogenesis in the DG improves memory through enhanced pattern separation.1,2,3,4,5,6,11,13,14,15,16 Adult-born granule cells (abGCs) in DG are suggested to contribute to pattern separation by driving inhibition in regions such as CA3,13,14,15,16,17,18 leading to sparser, nonoverlapping representations of similar events (although a role for abGCs in driving excitation in the hippocampus has also been reported16). Place cells in the hippocampus contribute to pattern separation by remapping to spatial and contextual alterations to the environment.19,20,21,22,23,24,25,26,27 How spatial responses in CA3 are affected by EE and input from increased numbers of abGCs in DG is, however, unknown. Here, we investigate the neural mechanisms facilitating improved memory following EE using associative recognition memory tasks that model the automatic and integrative nature of episodic memory. We find that EE-dependent improvements in difficult discriminations are related to increased neurogenesis and sparser memory representations across the hippocampus. Additionally, we report for the first time that EE changes how CA3 place cells discriminate similar contexts. CA3 place cells of enriched rats show greater spatial tuning, increased firing rates, and enhanced remapping to contextual changes. These findings point to more precise and flexible CA3 memory representations in enriched rats, which provides a putative mechanism for EE-dependent improvements in fine memory discrimination.


Assuntos
Região CA3 Hipocampal , Meio Ambiente , Animais , Ratos , Região CA3 Hipocampal/fisiologia , Masculino , Neurogênese/fisiologia , Ratos Long-Evans , Memória/fisiologia , Giro Denteado/fisiologia
10.
Expert Opin Ther Targets ; 28(4): 309-322, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38626283

RESUMO

BACKGROUND: Major Depressive Disorder (MDD) poses a significant challenge to global health, with current treatments often limited by efficacy and onset delays. This study explores the synergistic antidepressant-like effects of an NPY1R agonist and Ketamine, targeting their neurobiological interactions within the ventral hippocampus. RESEARCH DESIGN AND METHODS: Utilizing a preclinical model, this study administered Neuropeptide Y receptor 1 (NPY1R) agonist and Ketamine, both separately and in combination, through intracerebroventricular (icv) and intranasal (i.n.) routes. The Forced Swimming Test (FST) was employed to assess antidepressant-like activity, while in situ Proximity Ligation Assay and immunohistochemistry were used to examine NPY1R/TrkB heteroreceptor complexes and BDNF expression in the ventral dentate gyrus (DG), along with neurogenesis markers. RESULTS: The combined treatment significantly reduced immobility in the FST, indicative of enhanced antidepressant-like effects, correlated with increased formation of NPY1R/TrkB complex and brain-derived neurotrophic factor (BDNF) expression in the ventral DG. These molecular alterations were associated with increased neurogenesis. CONCLUSIONS: The coadministration of an NPY1R agonist and Ketamine in a rodent model demonstrated potentiated antidepressant responses through synergistic neurobiological pathways, including TrkB signaling and hippocampal neurogenesis. This indicates a novel therapeutic strategy for MDD, warranting further clinical investigation to fully understand its implications.


Assuntos
Antidepressivos , Fator Neurotrófico Derivado do Encéfalo , Transtorno Depressivo Maior , Sinergismo Farmacológico , Hipocampo , Ketamina , Neurogênese , Receptor trkB , Receptores de Neuropeptídeo Y , Transdução de Sinais , Animais , Neurogênese/efeitos dos fármacos , Antidepressivos/farmacologia , Antidepressivos/administração & dosagem , Masculino , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptores de Neuropeptídeo Y/agonistas , Receptores de Neuropeptídeo Y/metabolismo , Transtorno Depressivo Maior/tratamento farmacológico , Ketamina/farmacologia , Ketamina/administração & dosagem , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Receptor trkB/agonistas , Receptor trkB/metabolismo , Modelos Animais de Doenças , Ratos , Camundongos , Ratos Sprague-Dawley , Giro Denteado/efeitos dos fármacos , Giro Denteado/metabolismo , Natação
11.
Prog Neurobiol ; 237: 102612, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38642602

RESUMO

Recurrent seizures lead to accumulation of the activity-dependent transcription factor ∆FosB in hippocampal dentate granule cells in both mouse models of epilepsy and mouse models of Alzheimer's disease (AD), which is also associated with increased incidence of seizures. In patients with AD and related mouse models, the degree of ∆FosB accumulation corresponds with increasing severity of cognitive deficits. We previously found that ∆FosB impairs spatial memory in mice by epigenetically regulating expression of target genes such as calbindin that are involved in synaptic plasticity. However, the suppression of calbindin in conditions of neuronal hyperexcitability has been demonstrated to provide neuroprotection to dentate granule cells, indicating that ∆FosB may act over long timescales to coordinate neuroprotective pathways. To test this hypothesis, we used viral-mediated expression of ∆JunD to interfere with ∆FosB signaling over the course of several months in transgenic mice expressing mutant human amyloid precursor protein (APP), which exhibit spontaneous seizures and develop AD-related neuropathology and cognitive deficits. Our results demonstrate that persistent ∆FosB activity acts through discrete modes of hippocampal target gene regulation to modulate neuronal excitability, limit recurrent seizure activity, and provide neuroprotection to hippocampal dentate granule cells in APP mice.


Assuntos
Precursor de Proteína beta-Amiloide , Giro Denteado , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-fos , Convulsões , Animais , Giro Denteado/metabolismo , Camundongos , Convulsões/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Neuroproteção/fisiologia , Modelos Animais de Doenças , Doença de Alzheimer/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Humanos
12.
eNeuro ; 11(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38688719

RESUMO

Glutamatergic mossy cells (MCs) mediate associational and commissural connectivity, exhibiting significant heterogeneity along the septotemporal axis of the mouse dentate gyrus (DG). However, it remains unclear whether the neuronal features of MCs are conserved across mammals. This study compares the neuroanatomy of MCs in the DG of mice and monkeys. The MC marker, calretinin, distinguishes two subpopulations: septal and temporal. Dual-colored fluorescence labeling is utilized to compare the axonal projection patterns of these subpopulations. In both mice and monkeys, septal and temporal MCs project axons across the longitudinal axis of the ipsilateral DG, indicating conserved associational projections. However, unlike in mice, no MC subpopulations in monkeys make commissural projections to the contralateral DG. In monkeys, temporal MCs send associational fibers exclusively to the inner molecular layer, while septal MCs give rise to wide axonal projections spanning multiple molecular layers, akin to equivalent MC subpopulations in mice. Despite conserved septotemporal heterogeneity, interspecies differences are observed in the topological organization of septal MCs, particularly in the relative axonal density in each molecular layer along the septotemporal axis of the DG. In summary, this comparative analysis sheds light on both conserved and divergent features of MCs in the DG of mice and monkeys. These findings have implications for understanding functional differentiation along the septotemporal axis of the DG and contribute to our knowledge of the anatomical evolution of the DG circuit in mammals.


Assuntos
Axônios , Calbindina 2 , Giro Denteado , Camundongos Endogâmicos C57BL , Animais , Masculino , Giro Denteado/citologia , Giro Denteado/anatomia & histologia , Calbindina 2/metabolismo , Fibras Musgosas Hipocampais/fisiologia , Camundongos , Especificidade da Espécie , Feminino
13.
Hippocampus ; 34(6): 278-283, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38501294

RESUMO

Evidence suggests that individual hippocampal subfields are preferentially involved in various memory-related processes. Here, we demonstrated dissociations in these memory processes in two unique individuals with near-selective bilateral damage within the hippocampus, affecting the dentate gyrus (DG) in case BL and the cornu ammonis 1 (CA1) subfield in case BR. BL was impaired in discriminating highly similar objects in memory (i.e., mnemonic discrimination) but exhibited preserved overall recognition of studied objects, regardless of similarity. Conversely, BR demonstrated impaired general recognition. These results provide evidence for the DG in discrimination processes, likely related to underlying pattern separation computations, and the CA1 in retention/retrieval.


Assuntos
Região CA1 Hipocampal , Giro Denteado , Discriminação Psicológica , Giro Denteado/fisiologia , Humanos , Região CA1 Hipocampal/fisiologia , Masculino , Discriminação Psicológica/fisiologia , Reconhecimento Psicológico/fisiologia , Feminino , Pessoa de Meia-Idade , Idoso , Memória/fisiologia
14.
Cell Rep ; 43(4): 114000, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38527063

RESUMO

Fear overgeneralization is a maladaptive response to traumatic stress that is associated with the inability to discriminate between threat and safety contexts, a hallmark feature of post-traumatic stress disorder (PTSD). However, the neural mechanisms underlying this deficit remain unclear. Here, we show that traumatic stress exposure impairs contextual discrimination between threat and safety contexts in the learned helplessness (LH) model. Mossy cells (MCs) in the dorsal hippocampus are suppressed in response to traumatic stress. Bidirectional manipulation of MC activity in the LH model reveals that MC inhibition is causally linked to impaired contextual discrimination. Mechanistically, MC inhibition increases the number of active granule cells in a given context, significantly overlapping context-specific ensembles. Our study demonstrates that maladaptive inhibition of MCs after traumatic stress is a substantial mechanism underlying fear overgeneralization with contextual discrimination deficit, suggesting a potential therapeutic target for cognitive symptoms of PTSD.


Assuntos
Giro Denteado , Transtornos de Estresse Pós-Traumáticos , Animais , Masculino , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Medo/fisiologia , Fibras Musgosas Hipocampais/patologia , Desamparo Aprendido
15.
Neuropharmacology ; 250: 109908, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38492883

RESUMO

Decreased hippocampal synaptic plasticity is an important pathological change in stress-related mood disorders, including major depressive disorder. However, the underlying mechanism is unclear. PGC-1α, a transcriptional coactivator, is a key factor in synaptic plasticity. We investigated the relationships between changes in hippocampal PGC-1α expression and depressive-like and stress-coping behaviours, and whether they are related to hippocampal synapses. Adeno-associated virus was used to alter hippocampal PGC-1α expression in male C57BL/6 mice. The sucrose preference test and forced swimming test were used to assess their depressive-like and stress-coping behaviours, respectively. Immunohistochemistry and stereology were used to calculate the total number of excitatory synapses in each hippocampal subregion (the cornu ammonis (CA) 1, CA3, and dentate gyrus). Immunofluorescence was used to visualize the changes in dendritic structure. Western blotting was used to detect the expression of hippocampal PGC-1α and mitochondrial-associated proteins, such as UCP2, NRF1 and mtTFAs. Our results showed that mice with downregulated PGC-1α expression in the hippocampus exhibited depressive-like and passive stress-coping behaviours, while mice with upregulated PGC-1α in the hippocampus exhibited increased stress-coping behaviours. Moreover, the downregulation of hippocampal PGC-1α expression resulted in a decrease in the number of excitatory synapses in the DG and in the protein expression of UCP2 in the hippocampus. Alternatively, upregulation of hippocampal PGC-1α yielded the opposite results. This suggests that hippocampal PGC-1α is involved in regulating depressive-like and stress-coping behaviours and modulating the number of excitatory synapses in the DG. This provides new insight for the development of antidepressants.


Assuntos
Capacidades de Enfrentamento , Transtorno Depressivo Maior , Animais , Masculino , Camundongos , Giro Denteado , Transtorno Depressivo Maior/metabolismo , Hipocampo/metabolismo , Camundongos Endogâmicos C57BL , Sinapses/metabolismo
16.
Artigo em Inglês | MEDLINE | ID: mdl-38514038

RESUMO

Multiple lines of evidence suggest that the trace amine-associated receptor 1 (TAAR1) holds promise as a potential target for stress-related disorders, such as treating major depressive disorder (MDD). The role of TAAR1 in the regulation of adult neurogenesis is recently supported by transcriptomic data. However, it remains unknown whether TAAR1 in dentate gyrus (DG) mediate chronic stress-induced negative effects on hippocampal plasticity and related behavior in mice. The present study consisted of a series of experiments using RNAscope, genetic approaches, behavioral tests, immunohistochemical staining, Golgi-Cox technique to unravel the effects of TAAR1 on alterations of dentate neuronal plasticity and cognitive function in the chronic social defeat stress model. The mice subjected to chronic defeat stress exhibited a noteworthy decrease in the mRNA level of TAAR1 in DG. Additionally, they exhibited compromised social memory and spatial object recognition memory, as well as impaired proliferation and maturation of adult-born dentate granule cells. Moreover, the selective knockout TAAR1 in DG mostly mimicked the cognitive function deficits and neurogenesis impairment induced by chronic stress. Importantly, the administration of the selective TAAR1 partial agonist RO5263397 during stress exposure attenuated the adverse effects of chronic stress on cognitive function, adult neurogenesis, dendritic arborization, and the synapse number of dentate neurons in DG. In summary, our findings suggest that TAAR1 plays a crucial role in mediating the detrimental effects of chronic stress on hippocampal plasticity and cognition. TAAR1 agonists exhibit therapeutic potential for individuals suffering from cognitive impairments associated with MDD.


Assuntos
Giro Denteado , Transtorno Depressivo Maior , Receptores Acoplados a Proteínas G , Animais , Camundongos , Giro Denteado/fisiologia , Hipocampo/fisiologia , Cognição/fisiologia , Plasticidade Neuronal/fisiologia , Neurogênese
17.
J Neurosci ; 44(18)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38503495

RESUMO

Repetitive firing of granule cells (GCs) in the dentate gyrus (DG) facilitates synaptic transmission to the CA3 region. This facilitation can gate and amplify the flow of information through the hippocampus. High-frequency bursts in the DG are linked to behavior and plasticity, but GCs do not readily burst. Under normal conditions, a single shock to the perforant path in a hippocampal slice typically drives a GC to fire a single spike, and only occasionally more than one spike is seen. Repetitive spiking in GCs is not robust, and the mechanisms are poorly understood. Here, we used a hybrid genetically encoded voltage sensor to image voltage changes evoked by cortical inputs in many mature GCs simultaneously in hippocampal slices from male and female mice. This enabled us to study relatively infrequent double and triple spikes. We found GCs are relatively homogeneous and their double spiking behavior is cell autonomous. Blockade of GABA type A receptors increased multiple spikes and prolonged the interspike interval, indicating inhibitory interneurons limit repetitive spiking and set the time window for successive spikes. Inhibiting synaptic glutamate release showed that recurrent excitation mediated by hilar mossy cells contributes to, but is not necessary for, multiple spiking. Blockade of T-type Ca2+ channels did not reduce multiple spiking but prolonged interspike intervals. Imaging voltage changes in different GC compartments revealed that second spikes can be initiated in either dendrites or somata. Thus, pharmacological and biophysical experiments reveal roles for both synaptic circuitry and intrinsic excitability in GC repetitive spiking.


Assuntos
Potenciais de Ação , Giro Denteado , Animais , Giro Denteado/fisiologia , Giro Denteado/citologia , Masculino , Camundongos , Feminino , Potenciais de Ação/fisiologia , Sinapses/fisiologia , Neurônios/fisiologia , Camundongos Endogâmicos C57BL , Transmissão Sináptica/fisiologia , Camundongos Transgênicos
18.
Sci Rep ; 14(1): 6915, 2024 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-38519576

RESUMO

Neuronal aging may be, in part, related to a change in DNA methylation. Thus, methyl donors, like folate and methionine, may play a role in cognitive changes associated to neuronal aging. To test the role of these metabolites, we performed stereotaxic microinjection of these molecules into the dentate gyrus (DG) of aged mice (an average age of 21 month). Folate, but not S-Adenosyl-Methionine (SAM), enhances cognition in aged mice. In the presence of folate, we observed partial rejuvenation of DG cells, characterized by the expression of juvenile genes or reorganization of extracellular matrix. Here, we have also tried to identify the mechanism independent of DNA methylation, that involve folate effects on cognition. Our analyses indicated that folate binds to folate receptor α (FRα) and, upon folate binding, FRα is transported to cell nucleus, where it is acting as transcription factor for expressing genes like SOX2 or GluN2B. In this work, we report that a FRα binding peptide also replicates the folate effect on cognition, in aged mice. Our data suggest that such effect is not sex-dependent. Thus, we propose the use of this peptide to improve cognition since it lacks of folate-mediated side effects. The use of synthetic FRα binding peptides emerge as a future strategy for the study of brain rejuvenation.


Assuntos
Receptor 1 de Folato , Rejuvenescimento , Animais , Camundongos , Cognição , Giro Denteado/metabolismo , Receptor 1 de Folato/metabolismo , Ácido Fólico/metabolismo , Metionina , Peptídeos/metabolismo , S-Adenosilmetionina
19.
Biomolecules ; 14(3)2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38540798

RESUMO

Although granule cell dispersion (GCD) in the hippocampus is known to be an important feature associated with epileptic seizures in temporal lobe epilepsy (TLE), the endogenous molecules that regulate GCD are largely unknown. In the present study, we have examined whether there is any change in AEG-1 expression in the hippocampus of a kainic acid (KA)-induced mouse model of TLE. In addition, we have investigated whether the modulation of astrocyte elevated gene-1 (AEG-1) expression in the dentate gyrus (DG) by intracranial injection of adeno-associated virus 1 (AAV1) influences pathological phenotypes such as GCD formation and seizure susceptibility in a KA-treated mouse. We have identified that the protein expression of AEG-1 is upregulated in the DG of a KA-induced mouse model of TLE. We further demonstrated that AEG-1 upregulation by AAV1 delivery in the DG-induced anticonvulsant activities such as the delay of seizure onset and inhibition of spontaneous recurrent seizures (SRS) through GCD suppression in the mouse model of TLE, while the inhibition of AEG-1 expression increased susceptibility to seizures. The present observations suggest that AEG-1 is a potent regulator of GCD formation and seizure development associated with TLE, and the significant induction of AEG-1 in the DG may have therapeutic potential against epilepsy.


Assuntos
Epilepsia do Lobo Temporal , Epilepsia , Animais , Camundongos , Astrócitos/metabolismo , Giro Denteado/metabolismo , Epilepsia/metabolismo , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/genética , Epilepsia do Lobo Temporal/tratamento farmacológico , Hipocampo/metabolismo , Ácido Caínico/efeitos adversos , Ácido Caínico/metabolismo , Convulsões/induzido quimicamente , Convulsões/genética , Convulsões/metabolismo
20.
J Cell Physiol ; 239(5): e31249, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38501376

RESUMO

The hippocampal dentate gyrus, responds to diverse pathological stimuli through neurogenesis. This phenomenon, observed following brain injury or neurodegeneration, is postulated to contribute to neuronal repair and functional recovery, thereby presenting an avenue for endogenous neuronal restoration. This study investigated the extent of regenerative response in hippocampal neurogenesis by leveraging the well-established kainic acid-induced status epilepticus model in vivo. In our study, we observed the activation and proliferation of neuronal progenitors or neural stem cell (NSC) and their subsequent migration to the injury sites following the seizure. At the injury sites, new neurons (Tuj1+BrdU+ and NeuN+BrdU+) have been generated indicating regenerative and reparative roles of the progenitor cells. We further detected whether this transient neurogenic burst, which might be a response towards an attempt to repair the brain, is associated with persistent long-term exhaustion of the dentate progenitor cells and impairment of adult neurogenesis marked by downregulation of Ki67, HoPX, and Sox2 with BrdU+ cell in the later part of life. Our studies suggest that the adult brain has the constitutive endogenous regenerative potential for brain repair to restore the damaged neurons, meanwhile, in the long term, it accelerates the depletion of the finite NSC pool in the hippocampal neurogenic niche by changing its proliferative and neurogenic capacity. A thorough understanding of the impact of modulating adult neurogenesis will eventually be required to design novel therapeutics to stimulate or assist brain repair while simultaneously preventing the adverse effects of early robust neurogenesis on the proliferative potential of endogenous neuronal progenitors.


Assuntos
Hipocampo , Células-Tronco Neurais , Neurogênese , Animais , Células-Tronco Neurais/metabolismo , Hipocampo/patologia , Hipocampo/metabolismo , Proliferação de Células , Masculino , Nicho de Células-Tronco , Giro Denteado/patologia , Giro Denteado/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Ácido Caínico/toxicidade , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/patologia , Estado Epiléptico/metabolismo , Regeneração Nervosa , Modelos Animais de Doenças , Camundongos , Movimento Celular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...