Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21.947
1.
Nat Commun ; 15(1): 3872, 2024 May 08.
Article En | MEDLINE | ID: mdl-38719797

The gut microbiota and microglia play critical roles in Alzheimer's disease (AD), and elevated Bacteroides is correlated with cerebrospinal fluid amyloid-ß (Aß) and tau levels in AD. We hypothesize that Bacteroides contributes to AD by modulating microglia. Here we show that administering Bacteroides fragilis to APP/PS1-21 mice increases Aß plaques in females, modulates cortical amyloid processing gene expression, and down regulates phagocytosis and protein degradation microglial gene expression. We further show that administering Bacteroides fragilis to aged wild-type male and female mice suppresses microglial uptake of Aß1-42 injected into the hippocampus. Depleting murine Bacteroidota with metronidazole decreases amyloid load in aged 5xFAD mice, and activates microglial pathways related to phagocytosis, cytokine signaling, and lysosomal degradation. Taken together, our study demonstrates that members of the Bacteroidota phylum contribute to AD pathogenesis by suppressing microglia phagocytic function, which leads to impaired Aß clearance and accumulation of amyloid plaques.


Alzheimer Disease , Amyloid beta-Peptides , Disease Models, Animal , Mice, Transgenic , Microglia , Phagocytosis , Plaque, Amyloid , Animals , Microglia/metabolism , Microglia/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Female , Mice , Male , Bacteroides fragilis/metabolism , Gastrointestinal Microbiome , Humans , Mice, Inbred C57BL , Hippocampus/metabolism , Hippocampus/pathology
2.
Sci Rep ; 14(1): 10622, 2024 05 09.
Article En | MEDLINE | ID: mdl-38724691

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.


Depressive Disorder, Major , Hippocampus , Hypothalamus , Magnetic Resonance Imaging , Positron-Emission Tomography , Humans , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/pathology , Male , Female , Hypothalamus/metabolism , Hypothalamus/diagnostic imaging , Adult , Hippocampus/metabolism , Hippocampus/diagnostic imaging , Hippocampus/pathology , Middle Aged , Double-Blind Method , Positron-Emission Tomography/methods , Dentate Gyrus/metabolism , Dentate Gyrus/diagnostic imaging , Dentate Gyrus/pathology , Citalopram/therapeutic use , Hypothalamo-Hypophyseal System/metabolism , Organ Size
3.
Cells ; 13(9)2024 Apr 28.
Article En | MEDLINE | ID: mdl-38727294

Information on long-term effects of postovulatory oocyte aging (POA) on offspring is limited. Whether POA affects offspring by causing oxidative stress (OS) and mitochondrial damage is unknown. Here, in vivo-aged (IVA) mouse oocytes were collected 9 h after ovulation, while in vitro-aged (ITA) oocytes were obtained by culturing freshly ovulated oocytes for 9 h in media with low, moderate, or high antioxidant potential. Oocytes were fertilized in vitro and blastocysts transferred to produce F1 offspring. F1 mice were mated with naturally bred mice to generate F2 offspring. Both IVA and the ITA groups in low antioxidant medium showed significantly increased anxiety-like behavior and impaired spatial and fear learning/memory and hippocampal expression of anxiolytic and learning/memory-beneficial genes in both male and female F1 offspring. Furthermore, the aging in both groups increased OS and impaired mitochondrial function in oocytes, blastocysts, and hippocampus of F1 offspring; however, it did not affect the behavior of F2 offspring. It is concluded that POA caused OS and damaged mitochondria in aged oocytes, leading to defects in anxiety-like behavior and learning/memory of F1 offspring. Thus, POA is a crucial factor that causes psychological problems in offspring, and antioxidant measures may be taken to ameliorate the detrimental effects of POA on offspring.


Behavior, Animal , Mitochondria , Oocytes , Oxidative Stress , Animals , Oocytes/metabolism , Mitochondria/metabolism , Female , Mice , Male , Ovulation , Anxiety/metabolism , Anxiety/pathology , Antioxidants/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Blastocyst/metabolism , Cellular Senescence , Memory
4.
Int Immunopharmacol ; 133: 112158, 2024 May 30.
Article En | MEDLINE | ID: mdl-38691917

BACKGROUND: The prevalence of depression is higher in patients with inflammatory bowel disease (IBD) than in the general population. Inflammatory cytokines and the kynurenine pathway (KP) play important roles in IBD and associated depression. Aripiprazole (ARP), an atypical antipsychotic, shows various anti-inflammatory properties and may be useful in treating major depressive disorder. This study aimed to evaluate the protective effects of ARP on TNBS-induced colitis and subsequent depression in rats, highlighting the role of the KP. MATERIAL AND METHODS: Fifty-six male Wistar rats were used, and all groups except for the normal and sham groups received a single dose of intra-rectal TNBS. Three different doses of ARP and dexamethasone were injected intraperitoneally for two weeks in treatment groups. On the 15th day, behavioral tests were performed to evaluate depressive-like behaviors. Colon ulcer index and histological changes were assessed. The tissue levels of inflammatory cytokines, KP markers, lipopolysaccharide (LPS), nuclear factor-kappa-B (NF-κB), and zonula occludens (ZO-1) were evaluated in the colon and hippocampus. RESULTS: TNBS effectively induced intestinal damages and subsequent depressive-like symptoms in rats. TNBS treatment significantly elevated the intestinal content of inflammatory cytokines and NF-κB expression, dysregulated the KP markers balance in both colon and hippocampus tissues, and increased the serum levels of LPS. However, treatment with ARP for 14 days successfully reversed these alterations, particularly at higher doses. CONCLUSION: ARP could alleviate IBD-induced colon damage and associated depressive-like behaviors mainly via suppressing inflammatory cytokines activity, serum LPS concentration, and affecting the NF-κB/kynurenine pathway.


Anti-Inflammatory Agents , Aripiprazole , Colitis , Cytokines , Depression , Kynurenine , NF-kappa B , Rats, Wistar , Trinitrobenzenesulfonic Acid , Animals , Male , Kynurenine/metabolism , Kynurenine/blood , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Aripiprazole/therapeutic use , Aripiprazole/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Depression/drug therapy , Depression/chemically induced , Depression/metabolism , Rats , NF-kappa B/metabolism , Cytokines/metabolism , Signal Transduction/drug effects , Colon/pathology , Colon/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Disease Models, Animal , Humans
5.
Cell Commun Signal ; 22(1): 269, 2024 May 14.
Article En | MEDLINE | ID: mdl-38745240

BACKGROUND: The pathway involving PTEN-induced putative kinase 1 (PINK1) and PARKIN plays a crucial role in mitophagy, a process activated by artesunate (ART). We propose that patients with anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis exhibit insufficient mitophagy, and ART enhances mitophagy via the PINK1/PARKIN pathway, thereby providing neuroprotection. METHODS: Adult female mice aged 8-10 weeks were selected to create a passive transfer model of anti-NMDAR encephalitis. We conducted behavioral tests on these mice within a set timeframe. Techniques such as immunohistochemistry, immunofluorescence, and western blotting were employed to assess markers including PINK1, PARKIN, LC3B, p62, caspase3, and cleaved caspase3. The TUNEL assay was utilized to detect neuronal apoptosis, while transmission electron microscopy (TEM) was used to examine mitochondrial autophagosomes. Primary hippocampal neurons were cultured, treated, and then analyzed through immunofluorescence for mtDNA, mtROS, TMRM. RESULTS: In comparison to the control group, mitophagy levels in the experimental group were not significantly altered, yet there was a notable increase in apoptotic neurons. Furthermore, markers indicative of mitochondrial leakage and damage were found to be elevated in the experimental group compared to the control group, but these markers showed improvement following ART treatment. ART was effective in activating the PINK1/PARKIN pathway, enhancing mitophagy, and diminishing neuronal apoptosis. Behavioral assessments revealed that ART ameliorated symptoms in mice with anti-NMDAR encephalitis in the passive transfer model (PTM). The knockdown of PINK1 led to a reduction in mitophagy levels, and subsequent ART intervention did not alleviate symptoms in the anti-NMDAR encephalitis PTM mice, indicating that ART's therapeutic efficacy is mediated through the activation of the PINK1/PARKIN pathway. CONCLUSIONS: At the onset of anti-NMDAR encephalitis, mitochondrial damage is observed; however, this damage is mitigated by the activation of mitophagy via the PINK1/PARKIN pathway. This regulatory feedback mechanism facilitates the removal of damaged mitochondria, prevents neuronal apoptosis, and consequently safeguards neural tissue. ART activates the PINK1/PARKIN pathway to enhance mitophagy, thereby exerting neuroprotective effects and may achieve therapeutic goals in treating anti-NMDAR encephalitis.


Anti-N-Methyl-D-Aspartate Receptor Encephalitis , Artesunate , Disease Models, Animal , Neuroprotective Agents , Protein Kinases , Animals , Artesunate/pharmacology , Artesunate/therapeutic use , Mice , Female , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/pathology , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/drug therapy , Protein Kinases/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Microscopy, Electron, Transmission , Mitophagy/drug effects , Apoptosis/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Hippocampus/pathology , Hippocampus/drug effects , Hippocampus/metabolism
6.
Mol Biol Rep ; 51(1): 607, 2024 May 05.
Article En | MEDLINE | ID: mdl-38704801

BACKGROUND: Intracerebral hemorrhage (ICH) is a critical neurological condition with few treatment options, where secondary immune responses and specific cell death forms, like pyroptosis, worsen brain damage. Pyroptosis involves gasdermin-mediated membrane pores, increasing inflammation and neural harm, with the NLRP3/Caspase-1/GSDMD pathway being central to this process. Peroxiredoxin II (Prx II), recognized for its mitochondrial protection and reactive oxygen species (ROS) scavenging abilities, appears as a promising neuronal pyroptosis modulator. However, its exact role and action mechanisms need clearer definition. This research aims to explore Prx II impact on neuronal pyroptosis and elucidate its mechanisms, especially regarding endoplasmic reticulum (ER) stress and oxidative stress-induced neuronal damage modulation. METHODS AND RESULTS: Utilizing MTT assays, Microscopy, Hoechst/PI staining, Western blotting, and immunofluorescence, we found Prx II effectively reduces LPS/ATP-induced pyroptosis and neuroinflammation in HT22 hippocampal neuronal cells. Our results indicate Prx II's neuroprotective actions are mediated through PI3K/AKT activation and ER stress pathway inhibition, diminishing mitochondrial dysfunction and decreasing neuronal pyroptosis through the ROS/MAPK/NF-κB pathway. These findings highlight Prx II potential therapeutic value in improving intracerebral hemorrhage outcomes by lessening secondary brain injury via critical signaling pathway modulation involved in neuronal pyroptosis. CONCLUSIONS: Our study not only underlines Prx II importance in neuroprotection but also opens new therapeutic intervention avenues in intracerebral hemorrhage, stressing the complex interplay between redox regulation, ER stress, and mitochondrial dynamics in neuroinflammation and cell death management.


Endoplasmic Reticulum Stress , Neurons , Neuroprotective Agents , Oxidative Stress , Peroxiredoxins , Pyroptosis , Reactive Oxygen Species , Pyroptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Animals , Oxidative Stress/drug effects , Neurons/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Mice , Reactive Oxygen Species/metabolism , Peroxiredoxins/metabolism , Signal Transduction/drug effects , Cell Line , Mitochondria/metabolism , Mitochondria/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/complications
7.
BMC Neurol ; 24(1): 159, 2024 May 11.
Article En | MEDLINE | ID: mdl-38734615

BACKGROUND: Carbon monoxide (CO) poisoning is now one of the leading causes of poisoning-related mortality worldwide. The central nervous system is the most vulnerable structure in acute CO poisoning. MRI is of great significance in the diagnosis and prognosis of CO toxic encephalopathy. The imaging features of CO poisoning are diverse. We report atypical hippocampal lesions observed on MRI in four patients after acute CO exposure. CASE PRESENTATIONS: We report four patients who presented to the emergency department with loss of consciousness. The diagnosis of CO poisoning was confirmed on the basis of their detailed history, physical examination and laboratory tests. Brain MRI in all of these patients revealed abnormal signal intensity in hippocampi bilaterally. They all received hyperbaric oxygen therapy. The prognosis of all four patients was poor. CONCLUSION: Hippocampi, as a relatively rare lesion on MRI of CO poisoning, is of important significance both in the early and delayed stages of acute CO poisoning. In this article, we summarize the case reports of hippocampal lesions on MRI in patients with CO poisoning in recent years, in order to provide reference for the diagnosis and prognosis of CO poisoning.


Carbon Monoxide Poisoning , Hippocampus , Magnetic Resonance Imaging , Humans , Carbon Monoxide Poisoning/diagnostic imaging , Carbon Monoxide Poisoning/complications , Hippocampus/pathology , Hippocampus/diagnostic imaging , Magnetic Resonance Imaging/methods , Male , Female , Adult , Middle Aged
8.
Cell Mol Life Sci ; 81(1): 215, 2024 May 13.
Article En | MEDLINE | ID: mdl-38739166

Down syndrome (DS) is a genetic disease characterized by a supernumerary chromosome 21. Intellectual deficiency (ID) is one of the most prominent features of DS. Central nervous system defects lead to learning disabilities, motor and language delays, and memory impairments. At present, a prenatal treatment for the ID in DS is lacking. Subcutaneous administration of synthetic preimplantation factor (sPIF, a peptide with a range of biological functions) in a model of severe brain damage has shown neuroprotective and anti-inflammatory properties by directly targeting neurons and microglia. Here, we evaluated the effect of PIF administration during gestation and until weaning on Dp(16)1Yey mice (a mouse model of DS). Possible effects at the juvenile stage were assessed using behavioral tests and molecular and histological analyses of the brain. To test the influence of perinatal sPIF treatment at the adult stage, hippocampus-dependent memory was evaluated on postnatal day 90. Dp(16)1Yey pups showed significant behavioral impairment, with impaired neurogenesis, microglial cell activation and a low microglial cell count, and the deregulated expression of genes linked to neuroinflammation and cell cycle regulation. Treatment with sPIF restored early postnatal hippocampal neurogenesis, with beneficial effects on astrocytes, microglia, inflammation, and cell cycle markers. Moreover, treatment with sPIF restored the level of DYRK1A, a protein that is involved in cognitive impairments in DS. In line with the beneficial effects on neurogenesis, perinatal treatment with sPIF was associated with an improvement in working memory in adult Dp(16)1Yey mice. Perinatal treatment with sPIF might be an option for mitigating cognitive impairments in people with DS.


Disease Models, Animal , Down Syndrome , Neurogenesis , Animals , Down Syndrome/drug therapy , Down Syndrome/pathology , Down Syndrome/metabolism , Down Syndrome/complications , Down Syndrome/genetics , Neurogenesis/drug effects , Mice , Female , Pregnancy , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/drug effects , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Dyrk Kinases , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Male , Cognition Disorders/drug therapy , Cognition Disorders/pathology
9.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 319-326, 2024 Apr.
Article Zh | MEDLINE | ID: mdl-38710516

Objective To investigate the impact of the cannabinoid receptor agonist arachidonyl-2'-chloroethylamide (ACEA) on cognitive function in mice with sepsis-associated encephalopathy (SAE). Methods C57BL/6 mice were randomly divided into artificial cerebrospinal fluid (ACSF) and lipopolysaccharide (LPS) groups. The SAE model was established by intraventricular injection of LPS. The severity of sepsis in mice was assessed by sepsis severity score (MSS) and body mass changes. Behavioral paradigms were used to evaluate motor ability (open field test) and cognitive function (contextual fear conditioning test, Y-maze test). To evaluate the effects of ACEA intervention on SAE, mice were randomly assigned to ACSF group, ACEA intervention combined with ACSF group, LPS group, and ACEA intervention combined with LPS group. The dosage of ACEA intervention was 1.5 mg/kg. Real-time quantitative PCR was used to measure the mRNA expression levels of interleukin 1ß (IL-1ß), IL-6, and tumor necrosis factor α (TNF-α) in mouse hippocampal tissues. Western blot analysis was used to assess the protein levels of IL-6 and TNF-α in the hippocampus. Nissl staining was performed to examine neuronal damage in the CA1 region of the mouse hippocampus. Behavioral paradigms were again employed to evaluate motor ability and cognitive function. Results Three days after intraventricular LPS injection, mice exhibited significant cognitive dysfunction, confirming SAE modeling. Compared to the control group, the LPS group showed significant increases in mRNA of inflammatory factors such as IL-6, TNF-α, and IL-1ß, together with significant increases in IL-6 and TNF-α protein levels in the hippocampus, a decrease in Nissl bodies in the CA1 region, and significant cognitive dysfunction. Compared to the LPS group, the ACEA intervention group showed a significant decrease in the mRNA of IL-6, TNF-α, and IL-1ß, a significant reduction in IL-6 and TNF-α protein levels, an increase in Nissl bodies, and improved cognitive function. Conclusion ACEA improves cognitive function in SAE mice by inhibiting the expression levels of inflammatory factors IL-6 and TNF-α.


Arachidonic Acids , Mice, Inbred C57BL , Sepsis-Associated Encephalopathy , Animals , Sepsis-Associated Encephalopathy/drug therapy , Sepsis-Associated Encephalopathy/metabolism , Mice , Male , Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Lipopolysaccharides/adverse effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/agonists , Cognition/drug effects , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism
10.
Sci Rep ; 14(1): 10054, 2024 05 02.
Article En | MEDLINE | ID: mdl-38698053

ß-Thalassaemia is one of the most common genetic diseases worldwide. During the past few decades, life expectancy of patients has increased significantly owing to advance in medical treatments. Cognitive impairment, once has been neglected, has gradually become more documented. Cognitive impairment in ß-thalassaemia patients is associated with natural history of the disease and socioeconomic factors. Herein, to determined effect of ß-thalassaemia intrinsic factors, 22-month-old ß-thalassaemia mouse was used as a model to assess cognitive impairment and to investigate any aberrant brain pathology in ß-thalassaemia. Open field test showed that ß-thalassaemia mice had decreased motor function. However, no difference of neuronal degeneration in primary motor cortex, layer 2/3 area was found. Interestingly, impaired learning and memory function accessed by a Morris water maze test was observed and correlated with a reduced number of living pyramidal neurons in hippocampus at the CA3 region in ß-thalassaemia mice. Cognitive impairment in ß-thalassaemia mice was significantly correlated with several intrinsic ß-thalassaemic factors including iron overload, anaemia, damaged red blood cells (RBCs), phosphatidylserine (PS)-exposed RBC large extracellular vesicles (EVs) and PS-exposed medium EVs. This highlights the importance of blood transfusion and iron chelation in ß-thalassaemia patients. In addition, to improve patients' quality of life, assessment of cognitive functions should become part of routine follow-up.


Cognitive Dysfunction , Disease Models, Animal , Hippocampus , beta-Thalassemia , Animals , beta-Thalassemia/pathology , beta-Thalassemia/complications , beta-Thalassemia/genetics , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Mice , Hippocampus/pathology , Hippocampus/metabolism , Male , Neurons/metabolism , Neurons/pathology , Iron Overload/pathology , Iron Overload/metabolism , Iron Overload/complications , Extracellular Vesicles/metabolism , Erythrocytes/metabolism , Erythrocytes/pathology , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Maze Learning
11.
Zh Nevrol Psikhiatr Im S S Korsakova ; 124(4. Vyp. 2): 56-63, 2024.
Article Ru | MEDLINE | ID: mdl-38696152

The most common cause of severe cognitive impairment in adults is Alzheimer's disease (AD). Depending on the age of onset, AD is divided into early (<65 years) and late (≥65 years) forms. Early-onset AD (EOAD) is significantly less common than later-onset AD (LOAD) and accounts for only about 5-10% of cases. However, its medical and social significance, as a disease leading to loss of ability to work and legal capacity, as well as premature death in patients aged 40-64 years, is extremely high. Patients with EOAD compared with LOAD have a greater number of atypical clinical variants - 25% and 6-12.5%, respectively, which complicates the differential diagnosis of EOAD with other neurodegenerative diseases. However, the typical classical amnestic variant predominates in both EOAD and LOAD. Also, patients with EOAD have peculiarities according to neuroimaging data: when performing MRI of the brain, patients with EOAD often have more pronounced parietal atrophy and less pronounced hippocampal atrophy compared to patients with LOAD. The article pays attention to the features of the clinical and neuroimaging data in patients with EOAD; a case of a patient with EOAD is presented.


Age of Onset , Alzheimer Disease , Magnetic Resonance Imaging , Neuroimaging , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/diagnosis , Neuroimaging/methods , Middle Aged , Atrophy/diagnostic imaging , Diagnosis, Differential , Male , Brain/diagnostic imaging , Brain/pathology , Female , Hippocampus/diagnostic imaging , Hippocampus/pathology
12.
CNS Neurosci Ther ; 30(4): e14706, 2024 04.
Article En | MEDLINE | ID: mdl-38584347

OBJECTIVE: This study aimed to investigate whether spontaneous brain activity can be used as a prospective indicator to identify cognitive impairment in patients with Parkinson's disease (PD). METHODS: Resting-state functional magnetic resonance imaging (RS-fMRI) was performed on PD patients. The cognitive level of patients was assessed by the Montreal Cognitive Assessment (MoCA) scale. The fractional amplitude of low-frequency fluctuation (fALFF) was applied to measure the strength of spontaneous brain activity. Correlation analysis and between-group comparisons of fMRI data were conducted using Rest 1.8. By overlaying cognitively characterized brain regions and defining regions of interest (ROIs) based on their spatial distribution for subsequent cognitive stratification studies. RESULTS: A total of 58 PD patients were enrolled in this study. They were divided into three groups: normal cognition (NC) group (27 patients, average MoCA was 27.96), mild cognitive impairment (MCI) group (21 patients, average MoCA was 23.52), and severe cognitive impairment (SCI) group (10 patients, average MoCA was 17.3). It is noteworthy to mention that those within the SCI group exhibited the most advanced chronological age, with an average of 74.4 years, whereas the MCI group displayed a higher prevalence of male participants at 85.7%. It was found hippocampal regions were a stable representative brain region of cognition according to the correlation analysis between the fALFF of the whole brain and cognition, and the comparison of fALFF between different cognitive groups. The parahippocampal gyrus was the only region with statistically significant differences in fALFF among the three cognitive groups, and it was also the only brain region to identify MCI from NC, with an AUC of 0.673. The paracentral lobule, postcentral gyrus was the region that identified SCI from NC, with an AUC of 0.941. The midbrain, hippocampus, and parahippocampa gyrus was the region that identified SCI from MCI, with an AUC of 0.926. CONCLUSION: The parahippocampal gyrus was the potential brain region for recognizing cognitive impairment in PD, specifically for identifying MCI. Thus, the fALFF of parahippocampal gyrus is expected to contribute to future study as a multimodal fingerprint for early warning.


Cognitive Dysfunction , Parkinson Disease , Humans , Male , Aged , Female , Parkinson Disease/complications , Parkinson Disease/diagnostic imaging , Parkinson Disease/pathology , Prospective Studies , Brain/pathology , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Magnetic Resonance Imaging/methods , Hippocampus/pathology
13.
Mult Scler Relat Disord ; 86: 105520, 2024 Jun.
Article En | MEDLINE | ID: mdl-38582026

BACKGROUND: Previous studies have shown that thalamic and hippocampal neurodegeneration is associated with clinical decline in Multiple Sclerosis (MS). However, contributions of the specific thalamic nuclei and hippocampal subfields require further examination. OBJECTIVE: Using 7 Tesla (7T) magnetic resonance imaging (MRI), we investigated the cross-sectional associations between functionally grouped thalamic nuclei and hippocampal subfields volumes and T1 relaxation times (T1-RT) and subsequent clinical outcomes in MS. METHODS: High-resolution T1-weighted and T2-weighted images were acquired at 7T (n=31), preprocessed, and segmented using the Thalamus Optimized Multi Atlas Segmentation (THOMAS, for thalamic nuclei) and the Automatic Segmentation of Hippocampal Subfields (ASHS, for hippocampal subfields) packages. We calculated Pearson correlations between hippocampal subfields and thalamic nuclei volumes and T1-RT and subsequent multi-modal rater-determined and patient-reported clinical outcomes (∼2.5 years after imaging acquisition), correcting for confounders and multiple tests. RESULTS: Smaller volume bilaterally in the anterior thalamus region correlated with worse performance in gait function, as measured by the Patient Determined Disease Steps (PDDS). Additionally, larger volume in most functional groups of thalamic nuclei correlated with better visual information processing and cognitive function, as measured by the Symbol Digit Modalities Test (SDMT). In bilateral medial and left posterior thalamic regions, there was an inverse association between volumes and T1-RT, potentially indicating higher tissue degeneration in these regions. We also observed marginal associations between the right hippocampal subfields (both volumes and T1-RT) and subsequent clinical outcomes, though they did not survive correction for multiple testing. CONCLUSION: Ultrahigh field MRI identified markers of structural damage in the thalamic nuclei associated with subsequently worse clinical outcomes in individuals with MS. Longitudinal studies will enable better understanding of the role of microstructural integrity in these brain regions in influencing MS outcomes.


Hippocampus , Magnetic Resonance Imaging , Multiple Sclerosis , Thalamic Nuclei , Humans , Hippocampus/diagnostic imaging , Hippocampus/pathology , Male , Female , Adult , Thalamic Nuclei/diagnostic imaging , Thalamic Nuclei/pathology , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/pathology , Middle Aged , Cross-Sectional Studies
14.
J Neuroimmunol ; 390: 578349, 2024 May 15.
Article En | MEDLINE | ID: mdl-38669915

Depression is a psychiatric disorder associated with multiple factors including microglia-mediated neuroinflammation. Although atractylodin exerted a variety of biological activities, however the effect of atractylodin on neuroinflammation-related depression was still unclear. In this study, the lipopolysaccharide (LPS)-induced mouse model was used to explore the antidepressant effects and molecular mechanisms of atractylodin. The results showed that atractylodin increased sugar preference, also reduced immobility time in FST and TST. Further study showed atractylodin reduced the oxidative stress and the activation of microglia in mouse hippocampus, also inhibited the level of cytokine release, especially IL-1ß. The results of western blotting showed that atractylodin significantly inhibited the expression of NLRP3 and pro-IL1ß via inhibition of NF-κB pathway. Our studies showed that atractylodin upregulated BDNF/Akt pathway in mouse hippocampus. Therefore, this study firstly indicated that atractylodin can ameliorate lipopolysaccharide-induced depressive-like behaviors in mice through reducing neuroinflammation and neuronal damage, and its molecular mechanism may be associated with the decrease of the expression of NLRP3 inflammasome and upregulation of BDNF/Akt pathway.


Depression , Furans , Lipopolysaccharides , Neuroinflammatory Diseases , Animals , Mice , Lipopolysaccharides/toxicity , Male , Furans/pharmacology , Furans/therapeutic use , Depression/drug therapy , Depression/chemically induced , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/chemically induced , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism
15.
Prostaglandins Other Lipid Mediat ; 172: 106836, 2024 Jun.
Article En | MEDLINE | ID: mdl-38599513

Dravet syndrome is an intractable epilepsy with a high seizure burden that is resistant to current anti-seizure medications. There is evidence that neuroinflammation plays a role in epilepsy and seizures, however few studies have specifically examined neuroinflammation in Dravet syndrome under conditions of a higher seizure burden. Here we used an established genetic mouse model of Dravet syndrome (Scn1a+/- mice), to examine whether a higher seizure burden impacts the number and morphology of microglia in the hippocampus. Moreover, we examined whether a high seizure burden influences classical inflammatory mediators in this brain region. Scn1a+/- mice with a high seizure burden induced by thermal priming displayed a localised reduction in microglial cell density in the granule cell layer and subgranular zone of the dentate gyrus, regions important to postnatal neurogenesis. However, microglial cell number and morphology remained unchanged in other hippocampal subfields. The high seizure burden in Scn1a+/- mice did not affect hippocampal mRNA expression of classical inflammatory mediators such as interleukin 1ß and tumour necrosis factor α, but increased cyclooxygenase 2 (COX-2) expression. We then quantified hippocampal levels of prostanoids that arise from COX-2 mediated metabolism of fatty acids and found that Scn1a+/- mice with a high seizure burden displayed increased hippocampal concentrations of numerous prostaglandins, notably PGF2α, PGE2, PGD2, and 6-K-PGF1A, compared to Scn1a+/- mice with a low seizure burden. In conclusion, a high seizure burden increased hippocampal concentrations of various prostaglandin mediators in a mouse model of Dravet syndrome. Future studies could interrogate the prostaglandin pathways to further better understand their role in the pathophysiology of Dravet syndrome.


Disease Models, Animal , Epilepsies, Myoclonic , Hippocampus , NAV1.1 Voltage-Gated Sodium Channel , Prostaglandins , Seizures , Animals , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/metabolism , Epilepsies, Myoclonic/pathology , Mice , Hippocampus/metabolism , Hippocampus/pathology , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Seizures/metabolism , Seizures/genetics , Seizures/pathology , Prostaglandins/metabolism , Male , Microglia/metabolism , Microglia/pathology
16.
Int J Mol Sci ; 25(8)2024 Apr 18.
Article En | MEDLINE | ID: mdl-38674042

An important component contributing to the onset of epilepsy is the death of hippocampal neurons. Several studies have shown that Dravet syndrome model mice: Scn1a KO mice have a high number of apoptotic neurons following seizures, but the precise mechanism underlying this remains unclear. The aim of this research was to elucidate the potential molecular mechanism of neuronal apoptosis in Scn1a KO mice by integrating proteomics and transcriptomics, with the ultimate goal of offering better neuroprotection. We found that apoptotic processes were enriched in both proteomic and transcriptomic GO analyses, and KEGG results also indicated that differential proteins and genes play a role in neurotransmission, the cell cycle, apoptosis, and neuroinflammation. Then, we examined the upstream and downstream KGML interactions of the pathways to determine the relationship between the two omics, and we found that the HIF-1 signaling pathway plays a significant role in the onset and apoptosis of epilepsy. Meanwhile, the expression of the apoptosis-related protein VHL decreased in this pathway, and the expression of p21 was upregulated. Therefore, this study suggests that VHL/HIF-1α/p21 might be involved in the apoptosis of hippocampal neurons in Scn1a KO mice.


Apoptosis , Disease Models, Animal , Epilepsies, Myoclonic , Hippocampus , Mice, Knockout , NAV1.1 Voltage-Gated Sodium Channel , Neurons , Proteomics , Transcriptome , Animals , Epilepsies, Myoclonic/metabolism , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/pathology , Hippocampus/metabolism , Hippocampus/pathology , Apoptosis/genetics , Mice , Neurons/metabolism , Neurons/pathology , NAV1.1 Voltage-Gated Sodium Channel/genetics , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Proteomics/methods , Signal Transduction , Gene Expression Profiling , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
17.
Neuroreport ; 35(8): 529-535, 2024 May 15.
Article En | MEDLINE | ID: mdl-38606637

Physical activity (PA) is a promising therapeutic for Alzheimer's disease (AD). Only a handful of meta-analyses have studied the impact of PA interventions on regional brain volumes, and none to date has solely included studies on effect of PA on regional brain volumes in individuals with cognitive impairment (CI). In this meta-analysis, we examined whether there is support for the hypothesis that PA interventions positively impact hippocampal volume (HV) in individuals with CI. We also assessed whether the level of CI [mild CI (MCI) vs. AD] impacted this relationship. We identified six controlled trials that met inclusion criteria. These included 236 participants with AD, MCI, or preclinical AD. Data were extracted and analyzed following Cochrane guidelines. We used a random-effects model to estimate the mean change in HV pre- and post-exercise intervention. Forest plots, Hedges' g funnel plots, and Egger's test were used to assess unbiasedness and visualize intervention effects, and Tau 2 , Cochran's Q, and I 2 were calculated to assess heterogeneity. The primary analysis revealed a significant positive effect of PA on total HV. However, sub-group analyses indicated a significant preservation of HV only in individuals with MCI, but not in those with AD. Egger's test indicated no evidence of publication bias. Subgroup analyses also revealed significant heterogeneity only within the MCI cohort for the total and left HV. PA demonstrated a moderate, significant effect in preserving HV among individuals with MCI, but not AD, highlighting a therapeutic benefit, particularly in earlier disease stages.


Alzheimer Disease , Atrophy , Cognitive Dysfunction , Exercise , Hippocampus , Humans , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Cognitive Dysfunction/therapy , Hippocampus/pathology , Hippocampus/diagnostic imaging , Exercise/physiology , Exercise Therapy/methods
18.
Ecotoxicol Environ Saf ; 277: 116314, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38642409

Fine particulate matter (PM2.5) has been extensively implicated in the pathogenesis of neurodevelopmental disorders, but the underlying mechanism remains unclear. Recent studies have revealed that PM2.5 plays a role in regulating iron metabolism and redox homeostasis in the brain, which is closely associated with ferroptosis. In this study, the role and underlying mechanism of ferroptosis in PM2.5-induced neurotoxicity were investigated in mice, primary hippocampal neurons, and HT22 cells. Our findings demonstrated that exposure to PM2.5 could induce abnormal behaviors, neuroinflammation, and neuronal loss in the hippocampus of mice. These effects may be attributed to ferroptosis induced by PM2.5 exposure in hippocampal neurons. RNA-seq analysis revealed that the upregulation of iron metabolism-related protein Heme Oxygenase 1 (HO-1) and the activation of mitophagy might play key roles in PM2.5-induced ferroptosis in HT22 cells. Subsequent in vitro experiments showed that PM2.5 exposure significantly upregulated HO-1 in primary hippocampal neurons and HT22 cells. Moreover, PM2.5 exposure activated mitophagy in HT22 cells, leading to the loss of mitochondrial membrane potential, alterations in the expression of autophagy-related proteins LC3, P62, and mTOR, as well as an increase in mitophagy-related protein PINK1 and PARKIN. As a heme-degradation enzyme, the upregulation of HO-1 promotes the release of excess iron, genetically inhibiting the upregulation of HO-1 in HT22 cells could prevent both PM2.5-induced mitophagy and ferroptosis. Furthermore, pharmacological inhibition of mitophagy in HT22 cells reduced levels of ferrous ions and lipid peroxides, thereby preventing ferroptosis. Collectively, this study demonstrates that HO-1 mediates PM2.5-induced mitophagy-dependent ferroptosis in hippocampal neurons, and inhibiting mitophagy or ferroptosis may be a key therapeutic target to ameliorate neurotoxicity following PM2.5 exposure.


Ferroptosis , Heme Oxygenase-1 , Hippocampus , Mitophagy , Neurons , Particulate Matter , Up-Regulation , Animals , Particulate Matter/toxicity , Ferroptosis/drug effects , Mitophagy/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Neurons/drug effects , Neurons/pathology , Mice , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Up-Regulation/drug effects , Male , Mice, Inbred C57BL , Air Pollutants/toxicity , Membrane Proteins
19.
Alzheimers Dement ; 20(5): 3687-3695, 2024 May.
Article En | MEDLINE | ID: mdl-38574400

INTRODUCTION: Cerebral small vessel disease (SVD) and amyloid beta (Aß) pathology frequently co-exist. The impact of concurrent pathology on the pattern of hippocampal atrophy, a key substrate of memory impacted early and extensively in dementia, remains poorly understood. METHODS: In a unique cohort of mixed Alzheimer's disease and moderate-severe SVD, we examined whether total and regional neuroimaging measures of SVD, white matter hyperintensities (WMH), and Aß, as assessed by 18F-AV45 positron emission tomography, exert additive or synergistic effects on hippocampal volume and shape. RESULTS: Frontal WMH, occipital WMH, and Aß were independently associated with smaller hippocampal volume. Frontal WMH had a spatially distinct impact on hippocampal shape relative to Aß. In contrast, hippocampal shape alterations associated with occipital WMH spatially overlapped with Aß-vulnerable subregions. DISCUSSION: Hippocampal degeneration is differentially sensitive to SVD and Aß pathology. The pattern of hippocampal atrophy could serve as a disease-specific biomarker, and thus guide clinical diagnosis and individualized treatment strategies for mixed dementia.


Alzheimer Disease , Amyloid beta-Peptides , Cerebral Small Vessel Diseases , Hippocampus , Positron-Emission Tomography , Humans , Hippocampus/pathology , Hippocampus/diagnostic imaging , Cerebral Small Vessel Diseases/pathology , Cerebral Small Vessel Diseases/diagnostic imaging , Male , Aged , Female , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , White Matter/pathology , White Matter/diagnostic imaging , Atrophy/pathology , Magnetic Resonance Imaging , Aged, 80 and over , Neuroimaging , Cohort Studies
20.
Clin Exp Pharmacol Physiol ; 51(6): e13863, 2024 Jun.
Article En | MEDLINE | ID: mdl-38650114

Chronic hyperglycaemia is a chief feature of diabetes mellitus and complicates with many systematic anomalies. Non-human primates (NHPs) are excellent for studying hyperglycaemia or diabetes and associated comorbidities, but lack behavioural observation. In the study, behavioural, brain imaging and histological analysis were performed in a case of spontaneously hyperglycaemic (HGM) Macaca fascicularis. The results were shown that the HGM monkey had persistent body weight loss, long-term hyperglycaemia, insulin resistance, dyslipidemia, but normal concentrations of insulin, C-peptide, insulin autoantibody, islet cell antibody and glutamic acid decarboxylase antibody. Importantly, an impaired working memory in a delayed response task and neurological dysfunctions were found in the HGM monkey. The tendency for atrophy in hippocampus was observed by magnetic resonance imaging. Lenticular opacification, lens fibres disruptions and vacuole formation also occurred to the HGM monkey. The data suggested that the spontaneous HGM monkey might present diabetes-like characteristics and associated neurobehavioral anomalies in this case. This study first reported cognitive deficits in a spontaneous hyperglycaemia NHPs, which might provide evidence to use macaque as a promising model for translational research in diabetes and neurological complications.


Cataract , Hyperglycemia , Macaca fascicularis , Animals , Hyperglycemia/metabolism , Cataract/pathology , Male , Cognition Disorders/etiology , Cognition Disorders/pathology , Nervous System Diseases , Hippocampus/pathology , Hippocampus/metabolism
...