Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.522
Filtrar
1.
CNS Neurosci Ther ; 30(8): e14924, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39143678

RESUMO

AIMS: Postoperative cognitive dysfunction (POCD) is prevalent among the elderly, characterized primarily by cognitive decline after surgery. This study aims to explore how extracellular vesicles (EVs) derived from BV2 microglial cells, with and without the C-C chemokine receptor type 5 (CCR5), affect neuroinflammation, neuronal integrity, and cognitive function in a POCD mouse model. METHODS: We collected EVs from LPS-stimulated BV2 cells expressing CCR5 (EVsM1) and from BV2 cells with CCR5 knockdown (EVsM1-CCR5). These were administered to POCD-induced mice. Protein interactions between CCR5, G-protein-coupled receptors (GPCRs), and Ras were analyzed using structure-based docking and co-immunoprecipitation (Co-IP). We assessed the phosphorylation of p38 and Erk, the expression of synaptic proteins PSD95 and MAP2, and conducted Morris Water Maze tests to evaluate cognitive function. RESULTS: Structure-based docking and Co-IP confirmed interactions between CCR5, GPR, and Ras, suggesting a CCR5-GPCRs-Ras-MAPK pathway involvement in neuroinflammation. EVsM1 heightened neuroinflammation, reduced synaptic integrity, and impaired cognitive function in POCD mice. In contrast, EVsM1-CCR5 reduced neuroinflammatory markers, preserved synaptic proteins, enhanced dendritic spine structure, and improved cognitive outcomes. CONCLUSION: EVsM1 induced neuroinflammation via the CCR5-GPCRs-Ras-MAPK pathway, with EVsM1-CCR5 showing protective effects on POCD progression, suggesting a new therapeutic strategy for POCD management via targeted modification of microglial EVs.


Assuntos
Camundongos Endogâmicos C57BL , Microglia , Doenças Neuroinflamatórias , Complicações Cognitivas Pós-Operatórias , Receptores CCR5 , Animais , Microglia/metabolismo , Camundongos , Receptores CCR5/metabolismo , Doenças Neuroinflamatórias/metabolismo , Complicações Cognitivas Pós-Operatórias/metabolismo , Masculino , Vesículas Extracelulares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas ras/metabolismo , Cognição/fisiologia , Cognição/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Disfunção Cognitiva/metabolismo
2.
CNS Neurosci Ther ; 30(8): e14799, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39107952

RESUMO

We investigate the mechanism of action of astragalin (AST) in the treatment of Alzheimer's disease (AD). Network pharmacology was conducted to analyze the relationships among AST, AD, and neuroinflammation, The APP/PS1 transgenic mice with AD were used in the experiments; to be specific, the influence of AST on the behavior of mice was analyzed by Morris water maze and eight-arm radial maze tests, the tissue inflammatory factor levels were detected by ELISA, and pathological changes were analyzed by H&E and immunohistochemical staining. Analysis results of network pharmacology suggested that AST exerted the multi-target effect on neuroinflammation in AD. Through molecular docking and dynamics analyses, COX2 might be the target of AST. Moreover, animal experimental results demonstrated that AST improved the behavior of AD mice, and enhanced the motor and memory abilities, meanwhile, it suppressed the expression of inflammatory factors in tissues and the activation of microglial cells. this study discovers that AST can suppress microglial cell activation via COX2 to improve neuroinflammation in AD.


Assuntos
Doença de Alzheimer , Quempferóis , Camundongos Transgênicos , Simulação de Acoplamento Molecular , Farmacologia em Rede , Animais , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Camundongos , Quempferóis/farmacologia , Quempferóis/uso terapêutico , Aprendizagem em Labirinto/efeitos dos fármacos , Masculino , Ciclo-Oxigenase 2/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo
3.
Fluids Barriers CNS ; 21(1): 63, 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39113115

RESUMO

Cognitive decline covers a broad spectrum of disorders, not only resulting from brain diseases but also from systemic diseases, which seriously influence the quality of life and life expectancy of patients. As a highly selective anatomical and functional interface between the brain and systemic circulation, the blood-brain barrier (BBB) plays a pivotal role in maintaining brain homeostasis and normal function. The pathogenesis underlying cognitive decline may vary, nevertheless, accumulating evidences support the role of BBB disruption as the most prevalent contributing factor. This may mainly be attributed to inflammation, metabolic dysfunction, cell senescence, oxidative/nitrosative stress and excitotoxicity. However, direct evidence showing that BBB disruption causes cognitive decline is scarce, and interestingly, manipulation of the BBB opening alone may exert beneficial or detrimental neurological effects. A broad overview of the present literature shows a close relationship between BBB disruption and cognitive decline, the risk factors of BBB disruption, as well as the cellular and molecular mechanisms underlying BBB disruption. Additionally, we discussed the possible causes leading to cognitive decline by BBB disruption and potential therapeutic strategies to prevent BBB disruption or enhance BBB repair. This review aims to foster more investigations on early diagnosis, effective therapeutics, and rapid restoration against BBB disruption, which would yield better cognitive outcomes in patients with dysregulated BBB function, although their causative relationship has not yet been completely established.


Assuntos
Barreira Hematoencefálica , Disfunção Cognitiva , Humanos , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/metabolismo , Animais
4.
Fluids Barriers CNS ; 21(1): 64, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39118132

RESUMO

Given the persistent challenge of differentiating idiopathic Normal Pressure Hydrocephalus (iNPH) from similar clinical entities, we conducted an in-depth proteomic study of cerebrospinal fluid (CSF) in 28 shunt-responsive iNPH patients, 38 Mild Cognitive Impairment (MCI) due to Alzheimer's disease, and 49 healthy controls. Utilizing the Olink Explore 3072 panel, we identified distinct proteomic profiles in iNPH that highlight significant downregulation of synaptic markers and cell-cell adhesion proteins. Alongside vimentin and inflammatory markers upregulation, these results suggest ependymal layer and transependymal flow dysfunction. Moreover, downregulation of multiple proteins associated with congenital hydrocephalus (e.g., L1CAM, PCDH9, ISLR2, ADAMTSL2, and B4GAT1) points to a possible shared molecular foundation between congenital hydrocephalus and iNPH. Through orthogonal partial least squares discriminant analysis (OPLS-DA), a panel comprising 13 proteins has been identified as potential diagnostic biomarkers of iNPH, pending external validation. These findings offer novel insights into the pathophysiology of iNPH, with implications for improved diagnosis.


Assuntos
Hidrocefalia de Pressão Normal , Proteômica , Humanos , Hidrocefalia de Pressão Normal/líquido cefalorraquidiano , Proteômica/métodos , Masculino , Feminino , Idoso , Biomarcadores/líquido cefalorraquidiano , Disfunção Cognitiva/líquido cefalorraquidiano , Disfunção Cognitiva/metabolismo , Idoso de 80 Anos ou mais , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/metabolismo , Pessoa de Meia-Idade
5.
JCI Insight ; 9(14)2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39133647

RESUMO

The aggregation and prion-like propagation of tau are the hallmarks of Alzheimer's disease (AD) and other tauopathies. However, the molecular mechanisms underlying the assembly and spread of tau pathology remain elusive. Epidemiological data show that exposure to fine particulate matter (PM2.5) is associated with an increased risk of AD. However, the molecular mechanisms remain unknown. Here, we showed that PM2.5 triggered the aggregation of tau and promoted the formation of tau fibrils. Injection of PM2.5-induced tau preformed fibrils (PFFs) into the hippocampus of tau P301S transgenic mice promoted the aggregation of tau and induced cognitive deficits and synaptic dysfunction. Furthermore, intranasal administration of PM2.5 exacerbated tau pathology and induced cognitive impairment in tau P301S mice. In conclusion, our results indicated that PM2.5 exposure promoted tau pathology and induced cognitive impairments. These results provide mechanistic insight into how PM2.5 increases the risk of AD.


Assuntos
Modelos Animais de Doenças , Hipocampo , Camundongos Transgênicos , Material Particulado , Tauopatias , Proteínas tau , Animais , Material Particulado/toxicidade , Proteínas tau/metabolismo , Camundongos , Tauopatias/metabolismo , Tauopatias/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/efeitos dos fármacos , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/etiologia , Agregação Patológica de Proteínas/metabolismo , Humanos , Masculino
6.
Transl Neurodegener ; 13(1): 41, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39123214

RESUMO

The rising prevalence of diabetes mellitus has casted a spotlight on one of its significant sequelae: cognitive impairment. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally developed for diabetes management, are increasingly studied for their cognitive benefits. These benefits may include reduction of oxidative stress and neuroinflammation, decrease of amyloid burdens, enhancement of neuronal plasticity, and improved cerebral glucose utilization. The multifaceted effects and the relatively favorable side-effect profile of SGLT2 inhibitors render them a promising therapeutic candidate for cognitive disorders. Nonetheless, the application of SGLT2 inhibitors for cognitive impairment is not without its limitations, necessitating more comprehensive research to fully determine their therapeutic potential for cognitive treatment. In this review, we discuss the role of SGLT2 in neural function, elucidate the diabetes-cognition nexus, and synthesize current knowledge on the cognitive effects of SGLT2 inhibitors based on animal studies and clinical evidence. Research gaps are proposed to spur further investigation.


Assuntos
Disfunção Cognitiva , Inibidores do Transportador 2 de Sódio-Glicose , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Humanos , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/psicologia , Disfunção Cognitiva/metabolismo , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico
7.
J Neuroinflammation ; 21(1): 192, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39095838

RESUMO

BACKGROUND: Type 2 diabetes mellitus (T2D) is associated with an increased risk of cognitive dysfunction. Angiopoietin-like protein 8 (ANGPTL8) is an important regulator in T2D, but the role of ANGPTL8 in diabetes-associated cognitive dysfunction remains unknown. Here, we explored the role of ANGPTL8 in diabetes-associated cognitive dysfunction through its interaction with paired immunoglobulin-like receptor B (PirB) in the central nervous system. METHODS: The levels of ANGPTL8 in type 2 diabetic patients with cognitive dysfunction and control individuals were measured. Mouse models of diabetes-associated cognitive dysfunction were constructed to investigate the role of ANGPTL8 in cognitive function. The cognitive function of the mice was assessed by the Barnes Maze test and the novel object recognition test, and levels of ANGPTL8, synaptic and axonal markers, and pro-inflammatory cytokines were measured. Primary neurons and microglia were treated with recombinant ANGPTL8 protein (rA8), and subsequent changes were examined. In addition, the changes induced by ANGPTL8 were validated after blocking PirB and its downstream pathways. Finally, mice with central nervous system-specific knockout of Angptl8 and PirB-/- mice were generated, and relevant in vivo experiments were performed. RESULTS: Here, we demonstrated that in the diabetic brain, ANGPTL8 was secreted by neurons into the hippocampus, resulting in neuroinflammation and impairment of synaptic plasticity. Moreover, neuron-specific Angptl8 knockout prevented diabetes-associated cognitive dysfunction and neuroinflammation. Mechanistically, ANGPTL8 acted in parallel to neurons and microglia via its receptor PirB, manifesting as downregulation of synaptic and axonal markers in neurons and upregulation of proinflammatory cytokine expression in microglia. In vivo, PirB-/- mice exhibited resistance to ANGPTL8-induced neuroinflammation and synaptic damage. CONCLUSION: Taken together, our findings reveal the role of ANGPTL8 in the pathogenesis of diabetes-associated cognitive dysfunction and identify the ANGPTL8-PirB signaling pathway as a potential target for the management of this condition.


Assuntos
Proteína 8 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Disfunção Cognitiva , Diabetes Mellitus Tipo 2 , Camundongos Knockout , Receptores Imunológicos , Transdução de Sinais , Animais , Camundongos , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/prevenção & controle , Disfunção Cognitiva/etiologia , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Semelhantes a Angiopoietina/metabolismo , Proteínas Semelhantes a Angiopoietina/genética , Humanos , Masculino , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Camundongos Endogâmicos C57BL , Sinapses/metabolismo , Sinapses/patologia , Sinapses/efeitos dos fármacos , Hormônios Peptídicos/metabolismo , Pessoa de Meia-Idade , Feminino
9.
ACS Chem Neurosci ; 15(16): 3064-3077, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39119909

RESUMO

Polycystic ovary syndrome (PCOS) is an intricate endocrine disorder that targets millions of women globally. Recent research has drawn attention to its association with cognitive impairment and Alzheimer's disease (AD) risk, yet the exact mechanism remains elusive. This study aimed to explore the potential role of PCOS-associated insulin resistance (IR) and inflammation in linking PCOS to AD pathogenesis. It additionally investigated the therapeutic merits of pterostilbene (PTS) in ameliorating PCOS and associated cognitive deficits in comparison to metformin (MET). Rats were divided into five groups; vehicle group, PTS group [30 mg/kg, per os (p.o.) for 13 days], and the remaining three groups received letrozole (1 mg/kg, p.o. for 21 days) to represent the PCOS, PCOS + MET (300 mg/kg, p.o. for 13 days), and PCOS + PTS groups, respectively. Behavioral tests were conducted, along with a histopathological investigation of brains and ovaries. Assessment of serum hormonal profile and hippocampal IRS-1/PI3K/AKT/GSK-3ß insulin signaling pathway components were performed. PTS rats exhibited improved insulin sensitivity and hormonal profile, besides enhanced neurobehavioral tests performance and histopathological findings. These effects may be attributed to modulation of the IRS-1/PI3K/AKT/GSK-3ß pathway, reducing GSK-3ß activity, and mitigating Tau hyperphosphorylation and Aß accumulation in the brain. Likewise, PTS attenuated nuclear factor kappa B-mediated inflammation and reversed AChE elevation, suggesting multifaceted neuroprotective effects. Comparatively, PTS showed outcomes similar to those of MET in most parameters. The obtained findings validated that dysregulated insulin signaling in PCOS rats detrimentally affects cognitive function, which is halted by PTS, unveiling the potential of PTS as a novel therapy for PCOS and related cognitive deficits.


Assuntos
Disfunção Cognitiva , Glicogênio Sintase Quinase 3 beta , Proteínas Substratos do Receptor de Insulina , Resistência à Insulina , Metformina , Fosfatidilinositol 3-Quinases , Síndrome do Ovário Policístico , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Estilbenos , Animais , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo , Feminino , Metformina/farmacologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Ratos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estilbenos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Modelos Animais de Doenças , Hipoglicemiantes/farmacologia , Ratos Wistar
10.
Behav Brain Res ; 473: 115193, 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39122091

RESUMO

BACKGROUND & AIMS: Growing evidence suggests that neurotransmitters may be associated with cognitive decline in MDD. This study primarily investigated the differences in cognitive functions between MDD patients and healthy controls, and explored the potential association between neurotransmitters and cognitive function of MDD patients. METHODS: This cross-sectional study enrolled 87 first-diagnosed and drug-naïve patients with MDD and 50 healthy controls. Neurotransmitters (glutamine, glutamic acid, γ-2Aminobutiric acid, kainate, vanillylmandelic acid (VMA), 3-methoxy 4-hydroxyphenyl ethylene glycol (MHPG), noradrenaline (NE), homovanillic acid, dihydroxy-phenyl acetic acid (DOPAC), dopamine (DA), tryptophane, kynurenine, 5-HT, 5-hydroxyindoleacetic acid) were measured using LC-MS/MS and cognitive functions were assessed by the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). Then associative analyses with adjustment (female, age, BMI, education) by multiple linear regression between neurotransmitters and cognitive functions especially in MDD patients were performed. RESULTS: MDD patients had lower RBANS scores in immediate memory, delayed memory and RBANS scores after adjustment. Neurotransmitters were associated with the cognitive levels of MDD patients after adjustment: DOPAC and DOPAC/DA had positive association with immediate memory score; DOPAC, DOPAC/DA and (VMA+MHPG)/NE were positively associated with attention score; NE was negatively associated with language score; DOPAC/DA was positively associated with both delayed memory and RBANS scores. CONCLUSION: Patients had greater cognitive impairment especially in memory. Furthermore, plasma neurotransmitter may be related to MDD and play an important role in cognitive impairment in MDD, especially in memory and attention.


Assuntos
Cognição , Transtorno Depressivo Maior , Neurotransmissores , Humanos , Feminino , Masculino , Adulto , Neurotransmissores/sangue , Neurotransmissores/metabolismo , Transtorno Depressivo Maior/metabolismo , Transtorno Depressivo Maior/sangue , Estudos Transversais , Cognição/fisiologia , Pessoa de Meia-Idade , Adulto Jovem , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/sangue , Testes Neuropsicológicos
11.
Alzheimers Res Ther ; 16(1): 189, 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39160609

RESUMO

BACKGROUND: As a currently incurable but preventable disease, the prevention and early diagnosis of Alzheimer's disease (AD) has long been a research hotspot. Amyloid deposition has been shown to be a major pathological feature of AD. Notably, not all the people with amyloid-beta (Aß) pathology will have significant cognitive declines and eventually develop AD. Therefore, the aim of this study was to explore the risk factors for cognitive decline in Aß-positive participants. METHODS: We included 650 non-demented participants who were Aß-positive at baseline from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Mixed effects and COX regression models were applied to assess 37 potential risk factors. Mixed effects models were employed to assess the temporal associations between potential risk factors and four cognitive assessment scales. COX regression models were used to assess the impact of potential risk factors on cognitive diagnosis conversion. Univariate and multivariate analyses were applied to the above models. Additionally, we used the Cochran-Armitage trend test to examine whether the incidence of cognitive decline increased with the number concurrent of risk factors. RESULTS: Six factors (low diastolic pressure, low body mass index, retired status, a history of drug abuse, Parkinsonism, and depression) were the identified risk factors and four factors (a history of urinary disease, musculoskeletal diseases, no major surgical history, and no prior dermatologic-connective tissue diseases) were found to be suggestive risk factors. The incidence of cognitive decline in the Aß-positive participants gradually increased as the number of concurrent risk factors increased (p for trend = 0.0005). CONCLUSIONS: Our study may facilitate the understanding of the potential pathological processes in AD and provide novel targets for the prevention of cognitive decline among participants with Aß positivity.


Assuntos
Peptídeos beta-Amiloides , Disfunção Cognitiva , Humanos , Feminino , Masculino , Disfunção Cognitiva/epidemiologia , Disfunção Cognitiva/metabolismo , Fatores de Risco , Peptídeos beta-Amiloides/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/epidemiologia , Testes Neuropsicológicos
12.
Int J Mol Sci ; 25(15)2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39126094

RESUMO

Chronic and continuous alcohol consumption increases the risk of cognitive decline and may lead to alcohol-related dementia. We investigated the potential of Heracleum moellendorffii Hance root extract (HME) for treating alcohol-related cognitive impairment. Behavioral tests evaluated the effects of HME on cognitive function and depression. Changes in hippocampus and liver tissues were evaluated by Western blotting and H&E staining. The group treated with HME 200 mg/kg showed a significant increase in spontaneous alternation in Y-maze and a decrease in immobility in a forced swimming test (FST) compared to the vehicle-treated group. These results suggest that HME can restore memory deficits and reverse depressive symptoms caused by chronic alcohol consumption. The HME-treated group also upregulated brain-derived neurotrophic factor (BDNF), phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), and phosphorylated cAMP response element-binding protein (CREB) in the hippocampus. Additionally, it reduced lipid vacuolation in the liver and increased the expression of aldehyde dehydrogenase 1 (ADH1). The administration of HME improves cognitive impairment and reverses depressive symptoms due to alcohol consumption, restoring neural plasticity in the hippocampus and alcohol metabolism in the liver. These findings suggest that HME is a promising treatment for alcohol-related brain disorders. Molecular mechanisms underlying the therapeutic effects of HME and its active ingredients should be investigated further.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Disfunção Cognitiva , Hipocampo , Extratos Vegetais , Animais , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/etiologia , Camundongos , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Masculino , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Etanol/efeitos adversos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fígado/metabolismo , Fígado/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Depressão/tratamento farmacológico , Depressão/metabolismo , Modelos Animais de Doenças
13.
J Neuroinflammation ; 21(1): 201, 2024 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-39135052

RESUMO

BACKGROUND: Gender is a significant risk factor for late-onset Alzheimer's disease (AD), often attributed to the decline of estrogen. The plant estrogen secoisolariciresinol diglucoside (SDG) has demonstrated anti-inflammatory and neuroprotective effects. However, the protective effects and mechanisms of SDG in female AD remain unclear. METHODS: Ten-month-old female APPswe/PSEN1dE9 (APP/PS1) transgenic mice were treated with SDG to assess its potential ameliorative effects on cognitive impairments in a female AD model through a series of behavioral and biochemical experiments. Serum levels of gut microbial metabolites enterodiol (END) and enterolactone (ENL) were quantified using HPLC-MS. Correlation analysis and broad-spectrum antibiotic cocktail (ABx) treatment were employed to demonstrate the involvement of END and ENL in SDG's cognitive improvement effects in female APP/PS1 mice. Additionally, an acute neuroinflammation model was constructed in three-month-old C57BL/6J mice treated with lipopolysaccharide (LPS) and subjected to i.c.v. injection of G15, an inhibitor of G protein-coupled estrogen receptor (GPER), to investigate the mediating role of the estrogen receptor GPER in the cognitive benefits conferred by SDG. RESULTS: SDG administration resulted in significant improvements in spatial, recognition, and working memory in female APP/PS1 mice. Neuroprotective effects were observed, including enhanced expression of CREB/BDNF and PSD-95, reduced ß-amyloid (Aß) deposition, and decreased levels of TNF-α, IL-6, and IL-10. SDG also altered gut microbiota composition, increasing serum levels of END and ENL. Correlation analysis indicated significant associations between END, ENL, cognitive performance, hippocampal Aß-related protein mRNA expression, and cortical neuroinflammatory cytokine levels. The removal of gut microbiota inhibited END and ENL production and eliminated the neuroprotective effects of SDG. Furthermore, GPER was found to mediate the inhibitory effects of SDG on neuroinflammatory responses. CONCLUSION: These findings suggest that SDG promotes the production of gut microbial metabolites END and ENL, which inhibit cerebral ß-amyloid deposition, activate GPER to enhance CREB/BDNF signaling pathways, and suppress neuroinflammatory responses. Consequently, SDG exerts neuroprotective effects and ameliorates cognitive impairments associated with AD in female mice.


Assuntos
Doença de Alzheimer , Fator Neurotrófico Derivado do Encéfalo , Butileno Glicóis , Disfunção Cognitiva , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Microbioma Gastrointestinal , Glucosídeos , Camundongos Transgênicos , Doenças Neuroinflamatórias , Receptores de Estrogênio , Receptores Acoplados a Proteínas G , Animais , Feminino , Doença de Alzheimer/metabolismo , Doença de Alzheimer/tratamento farmacológico , Glucosídeos/farmacologia , Glucosídeos/uso terapêutico , Camundongos , Microbioma Gastrointestinal/efeitos dos fármacos , Butileno Glicóis/farmacologia , Butileno Glicóis/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Modelos Animais de Doenças
14.
Alzheimers Res Ther ; 16(1): 185, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39148136

RESUMO

BACKGROUND: The cholinergic neurotransmitter system is crucial to cognitive function, with the basal forebrain (BF) being particularly susceptible to Alzheimer's disease (AD) pathology. However, the interaction of white matter hyperintensities (WMH) in cholinergic pathways and BF atrophy without amyloid pathology remains poorly understood. METHODS: We enrolled patients who underwent neuropsychological tests, magnetic resonance imaging, and 18F-florbetaben positron emission tomography due to cognitive impairment at the teaching university hospital from 2015 to 2022. Among these, we selected patients with negative amyloid scans and additionally excluded those with Parkinson's dementia that may be accompanied by BF atrophy. The WMH burden of cholinergic pathways was quantified by the Cholinergic Pathways Hyperintensities Scale (CHIPS) score, and categorized into tertile groups because the CHIPS score did not meet normal distribution. Segmentation of the BF on volumetric T1-weighted MRI was performed using FreeSurfer, then was normalized for total intracranial volume. Multivariable regression analysis was performed to investigate the association between BF volumes and CHIPS scores. RESULTS: A total of 187 patients were enrolled. The median CHIPS score was 12 [IQR 5.0; 24.0]. The BF volume of the highest CHIPS tertile group (mean ± SD, 3.51 ± 0.49, CHIPSt3) was significantly decreased than those of the lower CHIPS tertile groups (3.75 ± 0.53, CHIPSt2; 3.83 ± 0.53, CHIPSt1; P = 0.02). In the univariable regression analysis, factors showing significant associations with the BF volume were the CHIPSt3 group, age, female, education, diabetes mellitus, smoking, previous stroke history, periventricular WMH, and cerebral microbleeds. In multivariable regression analysis, the CHIPSt3 group (standardized beta [ßstd] = -0.25, P = 0.01), female (ßstd = 0.20, P = 0.04), and diabetes mellitus (ßstd = -0.22, P < 0.01) showed a significant association with the BF volume. Sensitivity analyses showed a negative correlation between CHIPS score and normalized BF volume, regardless of WMH severity. CONCLUSIONS: We identified a significant correlation between strategic WMH burden in the cholinergic pathway and BF atrophy independently of amyloid positivity and WMH severity. These results suggest a mechanism of cholinergic neuronal loss through the dying-back phenomenon and provide a rationale that strategic WMH assessment may help identify target groups that may benefit from acetylcholinesterase inhibitor treatment.


Assuntos
Prosencéfalo Basal , Imageamento por Ressonância Magnética , Tomografia por Emissão de Pósitrons , Substância Branca , Humanos , Feminino , Masculino , Idoso , Substância Branca/diagnóstico por imagem , Substância Branca/patologia , Prosencéfalo Basal/diagnóstico por imagem , Prosencéfalo Basal/patologia , Pessoa de Meia-Idade , Testes Neuropsicológicos , Disfunção Cognitiva/diagnóstico por imagem , Disfunção Cognitiva/patologia , Disfunção Cognitiva/metabolismo , Atrofia/patologia , Idoso de 80 Anos ou mais
15.
J Neuroinflammation ; 21(1): 194, 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39097742

RESUMO

Chronic neuroinflammation and microglial activation are key mediators of the secondary injury cascades and cognitive impairment that follow exposure to repetitive mild traumatic brain injury (r-mTBI). Peroxisome proliferator-activated receptor-γ (PPARγ) is expressed on microglia and brain resident myeloid cell types and their signaling plays a major anti-inflammatory role in modulating microglial responses. At chronic timepoints following injury, constitutive PPARγ signaling is thought to be dysregulated, thus releasing the inhibitory brakes on chronically activated microglia. Increasing evidence suggests that thiazolidinediones (TZDs), a class of compounds approved from the treatment of diabetes mellitus, effectively reduce neuroinflammation and chronic microglial activation by activating the peroxisome proliferator-activated receptor-γ (PPARγ). The present study used a closed-head r-mTBI model to investigate the influence of the TZD Pioglitazone on cognitive function and neuroinflammation in the aftermath of r-mTBI exposure. We revealed that Pioglitazone treatment attenuated spatial learning and memory impairments at 6 months post-injury and reduced the expression of reactive microglia and astrocyte markers in the cortex, hippocampus, and corpus callosum. We then examined whether Pioglitazone treatment altered inflammatory signaling mechanisms in isolated microglia and confirmed downregulation of proinflammatory transcription factors and cytokine levels. To further investigate microglial-specific mechanisms underlying PPARγ-mediated neuroprotection, we generated a novel tamoxifen-inducible microglial-specific PPARγ overexpression mouse line and examined its influence on microglial phenotype following injury. Using RNA sequencing, we revealed that PPARγ overexpression ameliorates microglial activation, promotes the activation of pathways associated with wound healing and tissue repair (such as: IL10, IL4 and NGF pathways), and inhibits the adoption of a disease-associated microglia-like (DAM-like) phenotype. This study provides insight into the role of PPARγ as a critical regulator of the neuroinflammatory cascade that follows r-mTBI in mice and demonstrates that the use of PPARγ agonists such as Pioglitazone and newer generation TZDs hold strong therapeutic potential to prevent the chronic neurodegenerative sequelae of r-mTBI.


Assuntos
Disfunção Cognitiva , Microglia , PPAR gama , Pioglitazona , Animais , Masculino , Camundongos , Concussão Encefálica/metabolismo , Concussão Encefálica/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Pioglitazona/farmacologia , Pioglitazona/uso terapêutico , PPAR gama/metabolismo
16.
CNS Neurosci Ther ; 30(7): e14887, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39073013

RESUMO

AIMS: Neuroinflammation is a recognized contributor to cognitive disorders like Alzheimer's disease, with ferroptosis emerging as a novel mechanism underlying cognitive dysfunction associated with neuroinflammation. Insulin, pivotal in the central nervous system, holds promise for cognitive function enhancement. This study aimed to establish a cognitive impairment model through intracerebroventricular injection of lipopolysaccharide (LPS) and explore the impact of intracerebroventricular insulin injection on cognitive function in mice. METHODS: We employed diverse experimental techniques, including animal behavior testing, molecular assays, targeted metabolomics, nuclear medicine, and electron microscopy, to assess neurodegenerative changes, brain insulin resistance (IR), glucose uptake and metabolism, and ferroptosis. The model of cognitive impairment was induced via intracerebroventricular injection of LPS, followed by intracerebroventricular administration of insulin to evaluate its effects. RESULTS: Insulin treatment effectively mitigated LPS-induced cognitive decline and safeguarded against neuronal degeneration. Furthermore, insulin alleviated LPS-induced insulin resistance, enhanced glucose uptake in the hippocampus, and promoted the Pentose Phosphate Pathway (PPP) and nicotinamide adenine dinucleotide phosphate (NADPH) production. Additionally, insulin activated the glutathione (GSH)-glutathione peroxidase 4 (GPX4) pathway, reducing lipid peroxidation, and mitochondrial damage characteristic of LPS-induced ferroptosis in the hippocampus. CONCLUSION: Our findings underscore the therapeutic potential of insulin in alleviating LPS-induced cognitive impairment and ferroptosis by modulating glucose metabolism. This study offers a promising avenue for future interventions targeting cognitive decline.


Assuntos
Disfunção Cognitiva , Ferroptose , Glucose , Hipocampo , Insulina , Lipopolissacarídeos , Animais , Ferroptose/efeitos dos fármacos , Ferroptose/fisiologia , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Lipopolissacarídeos/toxicidade , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Camundongos , Masculino , Glucose/metabolismo , Camundongos Endogâmicos C57BL , Resistência à Insulina/fisiologia
17.
Behav Brain Res ; 471: 115123, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38972485

RESUMO

Apolipoprotein-E4 (ApoE4) is an important genetic risk factor for Alzheimer's disease. The development of targeted-replacement human ApoE knock-in mice facilitates research into mechanisms by which ApoE4 affects the brain. We performed meta-analyses and meta-regression analyses to examine differences in cognitive performance between ApoE4 and ApoE3 mice. We included 61 studies in which at least one of the following tests was assessed: Morris Water Maze (MWM), novel object location (NL), novel object recognition (NO) and Fear Conditioning (FC) test. ApoE4 vs. ApoE3 mice performed significantly worse on the MWM (several outcomes, 0.17 ≤ g ≤ 0.60), NO (exploration, g=0.33; index, g=0.44) and FC (contextual, g=0.49). ApoE4 vs. ApoE3 differences were not systematically related to sex or age. We conclude that ApoE4 knock-in mice in a non-AD condition show some, but limited cognitive deficits, regardless of sex and age. These effects suggest an intrinsic vulnerability in ApoE4 mice that may become more pronounced under additional brain load, as seen in neurodegenerative diseases.


Assuntos
Apolipoproteína E4 , Disfunção Cognitiva , Técnicas de Introdução de Genes , Animais , Apolipoproteína E4/genética , Camundongos , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Humanos , Camundongos Transgênicos , Modelos Animais de Doenças , Apolipoproteína E3/genética
18.
Kaohsiung J Med Sci ; 40(8): 732-743, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38963317

RESUMO

An increasing number of elderly individuals are experiencing postoperative cognitive dysfunction (POCD) problems after undergoing hip replacement surgery, with gut microbiota metabolites playing a role in its pathogenesis. Among these, the specific effects of trimethylamine N-oxide (TMAO) on POCD are still unclear. This study aimed to explore the role of TMAO on cognitive dysfunction and underlying mechanisms in mice. The POCD model was created through femoral fracture surgery in elderly mice, followed by cognitive function assessments using the Morris Water Maze and Novel Object Recognition tests. The gut microbiota depletion and fecal microbiota transplantation were performed to examine the relationship between TMAO levels and cognitive outcomes. The effects of TMAO treatment on cognitive dysfunction, microglial activation, and inflammatory cytokine levels in the brain were also evaluated, with additional assessment of the role of microglial ablation in reducing TMAO-induced cognitive impairment. Elevated TMAO levels were found to be associated with cognitive decline in mice following femoral fracture surgery, with gut microbiota depletion mitigating both TMAO elevation and cognitive dysfunction. In contrast, fecal microbiota transplantation from postoperative mice resulted in accelerated cognitive dysfunction and TMAO accumulation in germ-free mice. Furthermore, TMAO treatment worsened cognitive deficits, neuroinflammation, and promoted microglial activation, which were reversed through the ablation of microglia. TMAO exacerbates cognitive dysfunction and neuroinflammation in POCD mice, with microglial activation playing a crucial role in this process. Our findings may provide new therapeutic strategies for managing TMAO-related POCD and improving the quality of life for elderly patients.


Assuntos
Disfunção Cognitiva , Modelos Animais de Doenças , Fraturas do Fêmur , Microbioma Gastrointestinal , Metilaminas , Animais , Metilaminas/metabolismo , Metilaminas/efeitos adversos , Camundongos , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/etiologia , Masculino , Fraturas do Fêmur/metabolismo , Fraturas do Fêmur/complicações , Microglia/metabolismo , Transplante de Microbiota Fecal , Camundongos Endogâmicos C57BL , Complicações Cognitivas Pós-Operatórias/metabolismo , Complicações Cognitivas Pós-Operatórias/etiologia , Citocinas/metabolismo , Encéfalo/metabolismo
19.
PLoS One ; 19(7): e0306930, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39012854

RESUMO

BACKGROUND: Cryptochrome-2 (CRY2) is a core rhythm gene that plays a crucial role in DNA damage repair. The present study investigated the potential role of CRY2 in mediating sleep deprivation-induced cognitive decline in 5xFAD mice. METHODS: To assess the effects of SD on different brain regions of the mouse brain, we used 18F FDG PET-CT. Cognitive function was evaluated using the Morris water maze test and Y-maze. Lentivirus was used for the overexpression of CRY2, and small interfering RNA (siRNA) was used for the downregulation of CRY2 to verify the effect of CRY2. We used qRT‒PCR and Western blotting to identify the downstream factors of CRY2 and evaluated the cognitive function of mice to confirm the effects of these factors. RESULTS: The AD mice exhibited cognitive decline after 21 days of SD and had higher expression of CRY2 compared to AD mice with normal sleep. Overexpression of CRY2 led to decreased cognitive function in AD mice, and the downregulation of CRY2 attenuated the SD-induced cognitive decline in AD mice. CRY2 reduced the expression and function of CISH, which reduced the inhibition of STAT1 phosphorylation and led to synaptic dysfunction. CISH overexpression attenuated the impairing effect of sleep deprivation on cognitive function in AD mice. Furthermore, 18F FDG PET-CT revealed that SD significantly reduced glucose metabolism in different brain regions of AD mice. CONCLUSION: Our study demonstrated that sleep deprivation upregulated CRY2 in the hippocampus of AD mice, which resulted in synaptic dysfunction by decreasing CISH-mediated STAT1 phosphorylation.


Assuntos
Disfunção Cognitiva , Criptocromos , Camundongos Transgênicos , Privação do Sono , Animais , Privação do Sono/metabolismo , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/genética , Disfunção Cognitiva/etiologia , Camundongos , Criptocromos/metabolismo , Criptocromos/genética , Masculino , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Modelos Animais de Doenças , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Aprendizagem em Labirinto , Encéfalo/metabolismo , Encéfalo/diagnóstico por imagem
20.
Nat Commun ; 15(1): 5830, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38992057

RESUMO

Impaired ion channels regulating Golgi pH lead to structural alterations in the Golgi apparatus, such as fragmentation, which is found, along with cognitive impairment, in Alzheimer's disease. However, the causal relationship between altered Golgi structure and cognitive impairment remains elusive due to the lack of understanding of ion channels in the Golgi apparatus of brain cells. Here, we identify that a transmembrane protein TMEM87A, renamed Golgi-pH-regulating cation channel (GolpHCat), expressed in astrocytes and neurons that contributes to hippocampus-dependent memory. We find that GolpHCat displays unique voltage-dependent currents, which is potently inhibited by gluconate. Additionally, we gain structural insights into the ion conduction through GolpHCat at the molecular level by determining three high-resolution cryogenic-electron microscopy structures of human GolpHCat. GolpHCat-knockout mice show fragmented Golgi morphology and altered protein glycosylation and functions in the hippocampus, leading to impaired spatial memory. These findings suggest a molecular target for Golgi-related diseases and cognitive impairment.


Assuntos
Complexo de Golgi , Hipocampo , Camundongos Knockout , Neurônios , Animais , Humanos , Masculino , Camundongos , Astrócitos/metabolismo , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/patologia , Microscopia Crioeletrônica , Glicosilação , Complexo de Golgi/metabolismo , Células HEK293 , Hipocampo/metabolismo , Concentração de Íons de Hidrogênio , Canais Iônicos/metabolismo , Canais Iônicos/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Memória/fisiologia , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Memória Espacial/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA