Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.447
Filter
1.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(8): 953-956, 2024 Aug 10.
Article in Chinese | MEDLINE | ID: mdl-39097278

ABSTRACT

OBJECTIVE: To explore the clinical, imaging, and genetic characteristics of an adult patient with sporadic Neuronal intranuclear inclusion disease (NIID). METHODS: A patient who had visited the First People's Hospital of Chenzhou on August 6, 2023 was selected as the study subject. Results of clinical examination, neuroimaging, and genetic testing were retrospectively analyzed along with a literature review. The number of GGC trinucleotide repeats in the 5'-untranslated region of the NOTCH2NLC gene was determined by GC-PCR. RESULTS: The patient had presented with episodic encephalopathy, with enhanced magnetic resonance imaging showing enhancement features of the posterior cerebral cortex during the period of acute episode. Genetic testing revealed an increased number of GGC repeats (n = 97) in the 5'- untranslated region of the NOTCH2NLC gene, which confirmed the diagnosis of NIID. CONCLUSION: Clinical attention should be paid to the enhanced MRI findings of patients with adult-onset NIID, for whom posterior cortical enhancement may be characteristic manifestation during the acute phase of encephalopathy-like episode.


Subject(s)
Intranuclear Inclusion Bodies , Neurodegenerative Diseases , Humans , Intranuclear Inclusion Bodies/genetics , Neurodegenerative Diseases/genetics , Magnetic Resonance Imaging , Male , Genetic Testing , Female , Middle Aged , Receptor, Notch2/genetics , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Brain/diagnostic imaging , Brain/pathology , Adult
2.
Cereb Cortex ; 34(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39106177

ABSTRACT

Fibromyalgia (FM) is a central sensitization syndrome that is strongly associated with the cerebral cortex. This study used bidirectional two-sample Mendelian randomization (MR) analysis to investigate the bidirectional causality between FM and the cortical surface area and cortical thickness of 34 brain regions. Inverse variance weighted (IVW) was used as the primary method for this study, and sensitivity analyses further supported the results. The forward MR analysis revealed that genetically determined thinner cortical thickness in the parstriangularis (OR = 0.0567 mm, PIVW = 0.0463), caudal middle frontal (OR = 0.0346 mm, PIVW = 0.0433), and rostral middle frontal (OR = 0.0285 mm, PIVW = 0.0463) was associated with FM. Additionally, a reduced genetically determined cortical surface area in the pericalcarine (OR = 0.9988 mm2, PIVW = 0.0085) was associated with an increased risk of FM. Conversely, reverse MR indicated that FM was associated with cortical thickness in the caudal middle frontal region (ß = -0.0035 mm, PIVW = 0.0265), fusiform region (ß = 0.0024 mm, SE = 0.0012, PIVW = 0.0440), the cortical surface area in the supramarginal (ß = -9.3938 mm2, PIVW = 0.0132), and postcentral regions (ß = -6.3137 mm2, PIVW = 0.0360). Reduced cortical thickness in the caudal middle frontal gyrus is shown to have a significant relationship with FM prevalence in a bidirectional causal analysis.


Subject(s)
Cerebral Cortex , Fibromyalgia , Humans , Fibromyalgia/genetics , Fibromyalgia/diagnostic imaging , Fibromyalgia/pathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Mendelian Randomization Analysis , Magnetic Resonance Imaging , Female , Genetic Predisposition to Disease/genetics , Male , Polymorphism, Single Nucleotide
4.
PLoS One ; 19(8): e0295086, 2024.
Article in English | MEDLINE | ID: mdl-39159236

ABSTRACT

Sustained compressive injury (SCI) in the brain is observed in numerous injury and pathological scenarios, including tumors, ischemic stroke, and traumatic brain injury-related tissue swelling. Sustained compressive injury is characterized by tissue loading over time, and currently, there are few in vitro models suitable to study neural cell responses to strain-dependent sustained compressive injury. Here, we present an in vitro model of sustained compressive neural injury via centrifugation. Spheroids were made from neonatal rat cortical cells seeded at 4000 cells/spheroid and cultured for 14 days in vitro. A subset of spheroids was centrifuged at 104, 209, 313 or 419 rads/s for 2 minutes. Modeling the physical deformation of the spheroids via finite element analyses, we found that spheroids centrifuged at the aforementioned angular velocities experienced pressures of 10, 38, 84 and 149 kPa, respectively, and compressive (resp. tensile) strains of 10% (5%), 18% (9%), 27% (14%) and 35% (18%), respectively. Quantification of LIVE-DEAD assay and Hoechst 33342 nuclear staining showed that centrifuged spheroids subjected to pressures above 10 kPa exhibited significantly higher DNA damage than control spheroids at 2, 8, and 24 hours post-injury. Immunohistochemistry of ß3-tubulin networks at 2, 8, and 24 hours post-centrifugation injury showed increasing degradation of microtubules over time with increasing strain. Our findings show that cellular injuries occur as a result of specific levels and timings of sustained tissue strains. This experimental SCI model provides a high throughput in vitro platform to examine cellular injury, to gain insights into brain injury that could be targeted with therapeutic strategies.


Subject(s)
Cell Survival , Neurites , Spheroids, Cellular , Animals , Spheroids, Cellular/pathology , Rats , Neurites/metabolism , Neurites/pathology , Stress, Mechanical , Cerebral Cortex/pathology , Cells, Cultured , Rats, Sprague-Dawley , DNA Damage , Centrifugation
5.
Brain Behav ; 14(8): e3634, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39169605

ABSTRACT

PURPOSE: The purpose of this prospective longitudinal study was to evaluate the changes in brain surface gyrification in older long-term breast cancer survivors 5-15 years after chemotherapy treatment. METHODS: Older breast cancer survivors aged ≥ 65 years treated with chemotherapy (C+) or without chemotherapy (C-) 5-15 years prior and age- and sex-matched healthy controls (HC) were recruited (time point 1 (TP1)) and followed up for 2 years (time point 2 (TP2)). Study assessments for both time points included neuropsychological (NP) testing with the NIH Toolbox cognition battery and cortical gyrification analysis based on brain MRI. RESULTS: The study cohort with data for both TP1 and TP2 consisted of the following: 10 participants for the C+ group, 12 participants for the C- group, and 13 participants for the HC group. The C+ group had increased gyrification in six local gyral regions including the right fusiform, paracentral, precuneus, superior, middle temporal gyri and left pars opercularis gyrus, and it had decreased gyrification in two local gyral regions from TP1 to TP2 (p < .05, Bonferroni corrected). The C- and HC groups showed decreased gyrification only (p < .05, Bonferroni corrected). In the C+ group, changes in right paracentral gyrification and crystalized composite scores were negatively correlated (R = -0.76, p = .01). CONCLUSIONS: Altered gyrification could be the neural correlate of cognitive changes in older chemotherapy-treated long-term breast cancer survivors.


Subject(s)
Breast Neoplasms , Cancer Survivors , Magnetic Resonance Imaging , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/diagnostic imaging , Aged , Longitudinal Studies , Prospective Studies , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Antineoplastic Agents/adverse effects , Neuropsychological Tests , Aged, 80 and over
6.
Proc Natl Acad Sci U S A ; 121(34): e2409343121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39136994

ABSTRACT

Mutations in the PSEN genes are the major cause of familial Alzheimer's disease, and presenilin (PS) is the catalytic subunit of γ-secretase, which cleaves type I transmembrane proteins, including the amyloid precursor protein (APP) to release Aß peptides. While PS plays an essential role in the protection of neuronal survival, PSEN mutations also increase the ratio of Aß42/Aß40. Thus, it remains unresolved whether PSEN mutations cause AD via a loss of its essential function or increases of Aß42/Aß40. Here, we test whether the knockin (KI) allele of Psen1 L435F, the most severe FAD mutation located closest to the active site of γ-secretase, causes age-dependent cortical neurodegeneration independent of Aß by crossing various Psen mutant mice to the App-null background. We report that removing Aß completely through APP deficiency has no impact on the age-dependent neurodegeneration in Psen mutant mice, as shown by the absence of effects on the reduced cortical volume and decreases of cortical neurons at the ages of 12 and 18 mo. The L435F KI allele increases Aß42/Aß40 in the cerebral cortex while decreasing de novo production and steady-state levels of Aß42 and Aß40 in the presence of APP. Furthermore, APP deficiency does not alleviate elevated apoptotic cell death in the cerebral cortex of Psen mutant mice at the ages of 2, 12, and 18 mo, nor does it affect the progressive microgliosis in these mice. Our findings demonstrate that Psen1 mutations cause age-dependent neurodegeneration independent of Aß, providing further support for a loss-of-function pathogenic mechanism underlying PSEN mutations.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Cerebral Cortex , Mutation , Presenilin-1 , Presenilin-1/genetics , Presenilin-1/metabolism , Animals , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Mice , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Neurons/metabolism , Neurons/pathology , Mice, Transgenic , Humans
7.
Cells ; 13(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39120293

ABSTRACT

Major depressive disorder (MDD) is a complex and devastating illness that affects people of all ages. Despite the large use of antidepressants in current medical practice, neither their mechanisms of action nor the aetiology of MDD are completely understood. Experimental evidence supports the involvement of Parvalbumin-positive GABAergic neurons (PV-neurons) in the pathogenesis of MDD. DLX5 and DLX6 (DLX5/6) encode two homeodomain transcription factors involved in cortical GABAergic differentiation and function. In the mouse, the level of expression of these genes is correlated with the cortical density of PV-neurons and with anxiety-like behaviours. The same genomic region generates the lncRNA DLX6-AS1, which, in humans, participates in the GABAergic regulatory module downregulated in schizophrenia and ASD. Here, we show that the expression levels of Dlx5/6 in the adult mouse brain are correlated with the immobility time in the forced swim test, which is used to measure depressive-like behaviours. We show that the administration of the antidepressant fluoxetine (Flx) to normal mice induces, within 24 h, a rapid and stable reduction in Dlx5, Dlx6 and Dlx6-AS1 expression in the cerebral cortex through the activation of the TrkB-CREB pathway. Experimental Dlx5 overexpression counteracts the antidepressant effects induced by Flx treatment. Our findings show that one of the short-term effects of Flx administration is the reduction in Dlx5/6 expression in GABAergic neurons, which, in turn, has direct consequences on PV expression and on behavioural profiles. Variants in the DLX5/6 regulatory network could be implicated in the predisposition to depression and in the variability of patients' response to antidepressant treatment.


Subject(s)
Antidepressive Agents , Cerebral Cortex , Fluoxetine , GABAergic Neurons , Homeodomain Proteins , Receptor, trkB , Animals , GABAergic Neurons/metabolism , GABAergic Neurons/drug effects , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Mice , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/metabolism , Receptor, trkB/metabolism , Receptor, trkB/genetics , Male , Signal Transduction/drug effects , Mice, Inbred C57BL , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , Depressive Disorder, Major/genetics
8.
Cereb Cortex ; 34(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39123310

ABSTRACT

Structural covariance networks and causal effects within can provide critical information on gray matter reorganization and disease-related hierarchical changes. Based on the T1WI data of 43 classical trigeminal neuralgia patients and 45 controls, we constructed morphological similarity networks of cortical thickness, sulcal depth, fractal dimension, and gyrification index. Moreover, causal structural covariance network analyses were conducted in regions with morphological abnormalities or altered nodal properties, respectively. We found that patients showed reduced sulcal depth, gyrification index, and fractal dimension, especially in the salience network and the default mode network. Additionally, the integration of the fractal dimension and sulcal depth networks was significantly reduced, accompanied by decreased nodal efficiency of the bilateral temporal poles, and right pericalcarine cortex within the sulcal depth network. Negative causal effects existed from the left insula to the right caudal anterior cingulate cortex in the gyrification index map, also from bilateral temporal poles to right pericalcarine cortex within the sulcal depth network. Collectively, patients exhibited impaired integrity of the covariance networks in addition to the abnormal gray matter morphology in the salience network and default mode network. Furthermore, the patients may experience progressive impairment in the salience network and from the limbic system to the sensory system in network topology, respectively.


Subject(s)
Cerebral Cortex , Magnetic Resonance Imaging , Trigeminal Neuralgia , Humans , Trigeminal Neuralgia/pathology , Trigeminal Neuralgia/diagnostic imaging , Trigeminal Neuralgia/physiopathology , Female , Male , Middle Aged , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Aged , Nerve Net/diagnostic imaging , Nerve Net/pathology , Adult , Gray Matter/diagnostic imaging , Gray Matter/pathology , Brain Mapping
9.
PLoS One ; 19(8): e0308464, 2024.
Article in English | MEDLINE | ID: mdl-39110702

ABSTRACT

Neuronal loss is a hallmark of stroke and other neurodegenerative diseases, and as such, neuronal loss caused by microglia has been thought to be a contributing factor to disease progression. Here, we show that microglia indeed contribute significantly to neuronal loss in a mouse model of stroke, but this microglial-dependent process of neuronal clearance specifically targets stressed and degenerating neurons in the ischemic cortical region and not healthy non-ischemic neurons. Nonspecific stimulation of microglia decreased the density of neurons in the ischemic cortical region, whereas specific inhibition of MFG-E8 signaling, which is required for microglial phagocytosis of neurons, had the opposite effect. In both scenarios, the effects were microglia specific, as the same treatments had no effect in mice whose microglia were depleted prior to stroke. Finally, even though the inhibition of MFG-E8 signaling increased neuronal density in the ischemic brain region, it substantially exacerbated the development of cortical infarction. In conclusion, microglia through MFG-E8 signaling contribute to the loss of ischemic neurons and, in doing so, minimize the development of cortical infarction after stroke.


Subject(s)
Antigens, Surface , Microglia , Milk Proteins , Neurons , Signal Transduction , Stroke , Animals , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Mice , Milk Proteins/metabolism , Antigens, Surface/metabolism , Stroke/metabolism , Stroke/pathology , Stroke/complications , Male , Mice, Inbred C57BL , Disease Models, Animal , Cerebral Infarction/pathology , Cerebral Infarction/metabolism , Cerebral Infarction/etiology , Brain/metabolism , Brain/pathology , Phagocytosis , Cerebral Cortex/metabolism , Cerebral Cortex/pathology
10.
Neuropharmacology ; 258: 110094, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39094830

ABSTRACT

Traumatic brain injuries (TBI) of varying severity are becoming more frequent all over the world. The process of neuroinflammation, in which macrophages and microglia are key players, underlies all types of brain damage. The present study focuses on evaluating the therapeutic potential of N-docosahexaenoylethanolamine (DHEA, synaptamide), which is an endogenous metabolite of docosahexaenoic acid in traumatic brain injury. Previously, several in vitro and in vivo models have shown significant anti-neuroinflammatory and synaptogenic activity of synaptamide. The results of the present study show that synaptamide by subcutaneous administration (10 mg/kg/day, 7 days) exerts anti-inflammatory and anti-apoptotic effects in the thalamus and cerebral cortex of experimental animals (male C57BL/6 mice). Were analyzed the dynamics of changes in the activity of Iba-1- and CD68-positive microglia/macrophages, the level of production of pro-inflammatory cytokines (IL1ß, IL6, TNFα) and pro-apoptotic proteins (Bad, Bax), the expression of pro- and anti-inflammatory markers (CD68, CD206, arg-1). ATF3 transcription factor distribution and neuronal state in the thalamus and cerebral cortex of animals with craniotomy, traumatic brain injury, and therapy are quantitatively assessed. The obtained data showed that synaptamide: (1) has no effect on the total pool of microglia/macrophages; (2) inhibits the activity of pro-inflammatory microglia/macrophages and cytokines they produce; (3) increases the expression of CD206 but not arg-1; (4) has anti-apoptotic effect and (5) improves the morphological state of neurons. The results obtained confirm the high therapeutic potential of synaptamide in the therapy of traumatic brain injury.


Subject(s)
Apoptosis , Brain Injuries, Traumatic , Cerebral Cortex , Mice, Inbred C57BL , Microglia , Neurons , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Apoptosis/drug effects , Mice , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Thalamus/drug effects , Thalamus/metabolism , Thalamus/pathology , Cytokines/metabolism , Ethanolamines/pharmacology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism
11.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125656

ABSTRACT

Abnormal shifts in global climate, leading to extreme weather, significantly threaten the safety of individuals involved in outdoor activities. Hypothermia-induced coma or death frequently occurs in clinical and forensic settings. Despite this, the precise mechanism of central nervous system injury due to hypothermia remains unclear, hindering the development of targeted clinical treatments and specific forensic diagnostic indicators. The GEO database was searched to identify datasets related to hypothermia. Post-bioinformatics analyses, DEGs, and ferroptosis-related DEGs (FerrDEGs) were intersected. GSEA was then conducted to elucidate the functions of the Ferr-related genes. Animal experiments conducted in this study demonstrated that hypothermia, compared to the control treatment, can induce significant alterations in iron death-related genes such as PPARG, SCD, ADIPOQ, SAT1, EGR1, and HMOX1 in cerebral cortex nerve cells. These changes lead to iron ion accumulation, lipid peroxidation, and marked expression of iron death-related proteins. The application of the iron death inhibitor Ferrostatin-1 (Fer-1) effectively modulates the expression of these genes, reduces lipid peroxidation, and improves the expression of iron death-related proteins. Severe hypothermia disrupts the metabolism of cerebral cortex nerve cells, causing significant alterations in ferroptosis-related genes. These genetic changes promote ferroptosis through multiple pathways.


Subject(s)
Cerebral Cortex , Ferroptosis , Hypothermia , Neurons , Ferroptosis/genetics , Animals , Hypothermia/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neurons/metabolism , Iron/metabolism , Lipid Peroxidation , Male , Rats , Phenylenediamines/pharmacology , Cyclohexylamines
12.
CNS Neurosci Ther ; 30(8): e14901, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39097922

ABSTRACT

BACKGROUND: It has been demonstrated that progressive supranuclear palsy (PSP) correlates with structural abnormalities in several distinct regions of the brain. However, whether there are changes in the morphological similarity network (MSN) and the relationship between changes in brain structure and gene expression remain largely unknown. METHODS: We used two independent cohorts (discovery dataset: PSP: 51, healthy controls (HC): 82; replication dataset: PSP: 53, HC: 55) for MSN analysis and comparing the longitudinal changes in the MSN of PSP. Then, we applied partial least squares regression to determine the relationships between changes in MSN and spatial transcriptional features and identified specific genes associated with MSN differences in PSP. We further investigated the biological processes enriched in PSP-associated genes and the cellular characteristics of these genes, and finally, we performed an exploratory analysis of the relationship between MSN changes and neurotransmitter receptors. RESULTS: We found that the MSN in PSP patients was mainly decreased in the frontal and temporal cortex but increased in the occipital cortical region. This difference is replicable. In longitudinal studies, MSN differences are mainly manifested in the frontal and parietal regions. Furthermore, the expression pattern associated with MSN changes in PSP involves genes implicated in astrocytes and excitatory and inhibitory neurons and is functionally enriched in neuron-specific biological processes related to synaptic signaling. Finally, we found that the changes in MSN were mainly negatively correlated with the levels of serotonin, norepinephrine, and opioid receptors. CONCLUSIONS: These results have enhanced our understanding of the microscale genetic and cellular mechanisms responsible for large-scale morphological abnormalities in PSP patients, suggesting potential targets for future therapeutic trials.


Subject(s)
Supranuclear Palsy, Progressive , Humans , Supranuclear Palsy, Progressive/genetics , Supranuclear Palsy, Progressive/pathology , Supranuclear Palsy, Progressive/metabolism , Female , Male , Aged , Middle Aged , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cohort Studies , Longitudinal Studies
13.
Int J Mol Sci ; 25(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39126055

ABSTRACT

Rasmussen's encephalitis (RE) stands as a rare neurological disorder marked by progressive cerebral hemiatrophy and epilepsy resistant to medical treatment. Despite extensive study, the primary cause of RE remains elusive, while its histopathological features encompass cortical inflammation, neuronal degeneration, and gliosis. The underlying molecular mechanisms driving disease progression remain largely unexplored. In this case study, we present a patient with RE who underwent hemispherotomy and has remained seizure-free for over six months, experiencing gradual motor improvement. Furthermore, we conducted molecular analysis on the excised brain tissue, unveiling a decrease in the expression of cell-cycle-associated genes coupled with elevated levels of BDNF and TNF-α proteins. These findings suggest the potential involvement of cell cycle regulators in the progression of RE.


Subject(s)
Encephalitis , Humans , Encephalitis/genetics , Encephalitis/pathology , Encephalitis/metabolism , Male , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Brain/pathology , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/metabolism , Female , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Cell Cycle/genetics
14.
Cereb Cortex ; 34(7)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39051658

ABSTRACT

Behavioral addiction (BA) is a conceptually new addictive phenotype characterized by compulsive reward-seeking behaviors despite adverse consequences. Currently, its underlying neurogenetic mechanism remains unclear. Here, this study aimed to investigate the association between cortical thickness (CTh) and genetic phenotypes in BA. We conducted a systematic search in five databases and extracted gene expression data from the Allen Human Brain Atlas. Meta-analysis of 10 studies (343 addicted individuals and 355 controls) revealed that the BA group showed thinner CTh in the precuneus, postcentral gyrus, orbital-frontal cortex, and dorsolateral prefrontal cortex (P < 0.005). Meta-regression showed that the CTh in the precuneus and postcentral gyrus were negatively associated with the addiction severity (P < 0.0005). More importantly, the CTh phenotype of BA was spatially correlated with the expression of 12 genes (false discovery rate [FDR] < 0.05), and the dopamine D2 receptor had the highest correlation (rho = 0.55). Gene enrichment analysis further revealed that the 12 genes were involved in the biological processes of behavior regulation and response to stimulus (FDR < 0.05). In conclusion, our findings demonstrated the thinner CTh in cognitive control-related brain areas in BA, which could be associated with the expression of genes involving dopamine metabolism and behavior regulation.


Subject(s)
Behavior, Addictive , Cerebral Cortex , Humans , Behavior, Addictive/genetics , Behavior, Addictive/diagnostic imaging , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Male , Adult , Female , Brain Cortical Thickness , Receptors, Dopamine D2/genetics , Magnetic Resonance Imaging
15.
Biomolecules ; 14(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39062523

ABSTRACT

Microglia, the resident macrophages of the central nervous system, exhibit altered gene expression in response to various neurological conditions. This study investigates the relationship between West Nile Virus infection and microglial senescence, focusing on the role of LGALS3BP, a protein implicated in both antiviral responses and aging. Using spatial transcriptomics, RNA sequencing and flow cytometry, we characterized changes in microglial gene signatures in adult and aged mice following recovery from WNV encephalitis. Additionally, we analyzed Lgals3bp expression and generated Lgals3bp-deficient mice to assess the impact on neuroinflammation and microglial phenotypes. Our results show that WNV-activated microglia share transcriptional signatures with aged microglia, including upregulation of genes involved in interferon response and inflammation. Lgals3bp was broadly expressed in the CNS and robustly upregulated during WNV infection and aging. Lgals3bp-deficient mice exhibited reduced neuroinflammation, increased homeostatic microglial numbers, and altered T cell populations without differences in virologic control or survival. These data indicate that LGALS3BP has a role in regulating neuroinflammation and microglial activation and suggest that targeting LGALS3BP might provide a potential route for mitigating neuroinflammation-related cognitive decline in aging and post-viral infections.


Subject(s)
Cerebral Cortex , Glycoproteins , Microglia , West Nile Fever , West Nile virus , Animals , Male , Mice , Aging/genetics , Aging/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cellular Senescence/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/virology , Microglia/pathology , Phenotype , West Nile Fever/genetics , West Nile Fever/virology , West Nile Fever/pathology , West Nile Fever/metabolism , West Nile virus/physiology , Glycoproteins/genetics , Glycoproteins/metabolism
16.
Mol Neurodegener ; 19(1): 58, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080744

ABSTRACT

BACKGROUND: It is not fully established whether plasma ß-amyloid(Aß)42/Aß40 and phosphorylated Tau181 (p-Tau181) can effectively detect Alzheimer's disease (AD) pathophysiology in older Chinese adults and how these biomarkers correlate with astrocyte reactivity, Aß plaque deposition, tau tangle aggregation, and neurodegeneration. METHODS: We recruited 470 older adults and analyzed plasma Aß42/Aß40, p-Tau181, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) using the Simoa platform. Among them, 301, 195, and 70 underwent magnetic resonance imaging, Aß and tau positron emission tomography imaging. The plasma Aß42/Aß40 and p-Tau181 thresholds were defined as ≤0.0609 and ≥2.418 based on the receiver operating characteristic curve analysis using the Youden index by comparing Aß-PET negative cognitively unimpaired individuals and Aß-PET positive cognitively impaired patients. To evaluate the feasibility of using plasma Aß42/Aß40 (A) and p-Tau181 (T) to detect AD and understand how astrocyte reactivity affects this process, we compared plasma GFAP, Aß plaque, tau tangle, plasma NfL, hippocampal volume, and temporal-metaROI cortical thickness between different plasma A/T profiles and explored their relations with each other using general linear models, including age, sex, APOE-ε4, and diagnosis as covariates. RESULTS: Plasma A+/T + individuals showed the highest levels of astrocyte reactivity, Aß plaque, tau tangle, and axonal degeneration, and the lowest hippocampal volume and temporal-metaROI cortical thickness. Lower plasma Aß42/Aß40 and higher plasma p-Tau181 were independently and synergistically correlated with higher plasma GFAP and Aß plaque. Elevated plasma p-Tau181 and GFAP concentrations were directly and interactively associated with more tau tangle formation. Regarding neurodegeneration, higher plasma p-Tau181 and GFAP concentrations strongly correlated with more axonal degeneration, as measured by plasma NfL, and lower plasma Aß42/Aß40 and higher plasma p-Tau181 were related to greater hippocampal atrophy. Higher plasma GFAP levels were associated with thinner cortical thickness and significantly interacted with lower plasma Aß42/Aß40 and higher plasma p-Tau181 in predicting more temporal-metaROI cortical thinning. Voxel-wise imaging analysis confirmed these findings. DISCUSSION: This study provides a valuable reference for using plasma biomarkers to detect AD in the Chinese community population and offers novel insights into how astrocyte reactivity contributes to AD progression, highlighting the importance of targeting reactive astrogliosis to prevent AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Astrocytes , tau Proteins , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Female , tau Proteins/metabolism , Male , Aged , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Biomarkers/blood , Positron-Emission Tomography/methods , Aged, 80 and over , Middle Aged , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/blood , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , Magnetic Resonance Imaging/methods , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/metabolism , Plaque, Amyloid/pathology
17.
Eat Weight Disord ; 29(1): 47, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39028377

ABSTRACT

PURPOSE: This study investigated the association between childhood eating behaviors and cortical morphology, in relation to sex and age, in a community sample. METHODS: Neuroimaging data of 71 children (mean age = 9.9 ± 1.4 years; 39 boys/32 girls) were obtained from the Nathan Kline Institute-Rockland Sample. Emotional overeating, food fussiness, and emotional undereating were assessed using the Children's Eating Behavior Questionnaire. Cortical thickness was obtained at 81,924 vertices covering the entire cortex. Generalized Linear Mixed Models were used for statistical analysis. RESULTS: There was a significant effect of sex in the association between cortical thickness and emotional overeating (localized at the right postcentral and bilateral superior parietal gyri). Boys with more emotional overeating presented cortical thickening, whereas the opposite was observed in girls (p < 0.05). Different patterns of association were identified between food fussiness and cortical thickness (p < 0.05). The left rostral middle frontal gyrus displayed a positive correlation with food fussiness from 6 to 8 years, but a negative correlation from 12 to 14 years. Emotional undereating was associated with cortical thickening at the left precuneus, left middle temporal gyrus, and left insula (p < 0.05) with no effect of sex or age. CONCLUSIONS: Leveraging on a community sample, findings support distinct patterns of associations between eating behaviors and cortical thickness, depending on sex and age.


Subject(s)
Cerebral Cortex , Feeding Behavior , Feeding and Eating Disorders , Magnetic Resonance Imaging , Humans , Male , Female , Child , Feeding Behavior/psychology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Adolescent , Feeding and Eating Disorders/psychology , Age Factors , Sex Factors , Emotions/physiology , Child Behavior/psychology
18.
Neurochem Int ; 178: 105799, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38950625

ABSTRACT

Alumunium usage and toxicity has been a global concern especially an increased use of nanoparticulated aluminum (Al-NPs) products from the environment and the workplace. Al degrades in to nanoparticulate form in the environment due to the routine process of bioremediation in human body. Al-NPs toxicity plays key role in the pathophysiology of neurodegeneration which is characterised by the development of neurofibrillary tangles and neuritic plaques which correlates to the Alzheimer's disease. This study evaluated the Al-NPs induced neurodegeneration and causative behavioral alterations due to oxidative stress, inflammation, DNA damage, ß-amyloid aggregation, and histopathological changes in mice. Furthermore, the preventive effect of naringenin (NAR) as a potent neuroprotective flavonoid against Al-NPs induced neurodegeneration was assessed. Al-NPs were synthesized and examined using FTIR, XRD, TEM, and particle size analyzer. Mice were orally administered with Al-NPs (6 mg/kg b.w.) followed by NAR treatment (10 mg/kg b.w. per day) for 66 days. The spatial working memory was determined by novel object recognition, T-maze, Y-maze, and Morris Water Maze tests. We measured nitric oxide, advanced oxidation of protein products, protein carbonylation, lipid peroxidation, superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase, reduced glutathione, oxidised glutathione, and acetylcholine esterase, as well as cytokines analysis, immunohistochemistry, and DNA damage. Al-NPs significantly reduced the learning memory power, increased oxidative stress, reduced antioxidant enzymatic activity, increased DNA damage, altered the levels of cytokines, and increased ß-amyloid aggregation in the cortex and hippocampus regions of the mice brain. These neurobehavioral impairments, neuronal oxidative stress, and histopathological alterations were significantly attenuated by NAR supplementation. In conclusion, Al-NPs may be potent neurotoxic upon exposure and that NAR could serve as a potential preventive measure in the treatment and management of neuronal degeneration.


Subject(s)
Aluminum , Flavanones , Hippocampus , Oxidative Stress , Animals , Flavanones/pharmacology , Flavanones/therapeutic use , Oxidative Stress/drug effects , Mice , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Aluminum/toxicity , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Down-Regulation/drug effects , Nerve Degeneration/drug therapy , Nerve Degeneration/pathology , Nerve Degeneration/chemically induced , Nerve Degeneration/metabolism , Metal Nanoparticles
19.
Behav Brain Res ; 471: 115137, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38971432

ABSTRACT

Hyperexcitability of neuronal networks is central to the pathogenesis of Alzheimer's disease (AD). Pharmacological activation of Kv7 channels is an effective way to reduce neuronal firing. Our results showed that that pharmacologically activating the Kv7 channel with Retigabine (RTG) can alleviate cognitive impairment in mice without affecting spontaneous activity. RTG could also ameliorate damage to the Nissl bodies in cortex and hippocampal CA and DG regions in 9-month-old APP/PS1 mice. Additionally, RTG could reduce the Aß plaque number in the hippocampus and cortex of both 6-month-old and 9-month-old mice. By recordings of electroencephalogram, we showed that a decrease in the number of abnormal discharges in the brains of the AD model mice when the Kv7 channel was opened. Moreover, Western blot analysis revealed a reduction in the expression of the p-Tau protein in both the hippocampus and cortex upon Kv7 channel opening. These findings suggest that Kv7 channel opener RTG may ameliorate cognitive impairment in AD, most likely by reducing brain excitability.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Carbamates , Cognitive Dysfunction , Disease Models, Animal , Hippocampus , Mice, Transgenic , Phenylenediamines , Animals , Phenylenediamines/pharmacology , Carbamates/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Presenilin-1/genetics , Male , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , tau Proteins/metabolism , Behavior, Animal/drug effects , Mice, Inbred C57BL
20.
Cereb Cortex ; 34(7)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39077916

ABSTRACT

The lifetime effects of repetitive head impacts have captured considerable public and scientific interest over the past decade, yet a knowledge gap persists in our understanding of midlife neurological well-being, particularly in amateur level athletes. This study aimed to identify the effects of lifetime exposure to sports-related head impacts on brain morphology in retired, amateur athletes. This cross-sectional study comprised of 37 former amateur contact sports athletes and 21 age- and sex-matched noncontact athletes. High-resolution anatomical, T1 scans were analyzed for the cortical morphology, including cortical thickness, sulcal depth, and sulcal curvature, and cognitive function was assessed using the Dementia Rating Scale-2. Despite no group differences in cognitive functions, the contact group exhibited significant cortical thinning particularly in the bilateral frontotemporal regions and medial brain regions, such as the cingulate cortex and precuneus, compared to the noncontact group. Deepened sulcal depth and increased sulcal curvature across all four lobes of the brain were also notable in the contact group. These data suggest that brain morphology of middle-aged former amateur contact athletes differs from that of noncontact athletes and that lifetime exposure to repetitive head impacts may be associated with neuroanatomical changes.


Subject(s)
Athletes , Cerebral Cortex , Magnetic Resonance Imaging , Humans , Male , Female , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/anatomy & histology , Cross-Sectional Studies , Middle Aged , Athletic Injuries/pathology , Athletic Injuries/diagnostic imaging , Aged , Brain Concussion/pathology , Brain Concussion/diagnostic imaging , Cognition/physiology
SELECTION OF CITATIONS
SEARCH DETAIL