Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.969
Filter
1.
Nat Aging ; 4(5): 625-637, 2024 May.
Article in English | MEDLINE | ID: mdl-38664576

ABSTRACT

Autopsy studies indicated that the locus coeruleus (LC) accumulates hyperphosphorylated tau before allocortical regions in Alzheimer's disease. By combining in vivo longitudinal magnetic resonance imaging measures of LC integrity, tau positron emission tomography imaging and cognition with autopsy data and transcriptomic information, we examined whether LC changes precede allocortical tau deposition and whether specific genetic features underlie LC's selective vulnerability to tau. We found that LC integrity changes preceded medial temporal lobe tau accumulation, and together these processes were associated with lower cognitive performance. Common gene expression profiles between LC-medial temporal lobe-limbic regions map to biological functions in protein transport regulation. These findings advance our understanding of the spatiotemporal patterns of initial tau spreading from the LC and LC's selective vulnerability to Alzheimer's disease pathology. LC integrity measures can be a promising indicator for identifying the time window when individuals are at risk of disease progression and underscore the importance of interventions mitigating initial tau spread.


Subject(s)
Alzheimer Disease , Cognition , Locus Coeruleus , Positron-Emission Tomography , tau Proteins , Locus Coeruleus/metabolism , Locus Coeruleus/diagnostic imaging , Locus Coeruleus/pathology , Humans , tau Proteins/metabolism , tau Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Cognition/physiology , Male , Female , Aged , Magnetic Resonance Imaging , Aged, 80 and over , Temporal Lobe/metabolism , Temporal Lobe/diagnostic imaging , Temporal Lobe/pathology
2.
Alzheimers Res Ther ; 16(1): 89, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654300

ABSTRACT

BACKGROUND: Association of medial temporal lobe (MTL) metabolism with Alzheimer's disease (AD) and dementia with Lewy bodies (DLB) has not been evaluated considering their mixed disease (MD). METHODS: 131 patients with AD, 133 with DLB, 122 with MD, and 28 normal controls (NCs) underwent neuropsychological tests, assessments for parkinsonism, cognitive fluctuation (CF), and visual hallucinations (VH), and 18F-fluorodeoxyglucose PET to quantify MTL metabolism in the amygdala, hippocampus, and entorhinal cortex. The effects of AD and DLB on MTL metabolism were evaluated using general linear models (GLMs). Associations between MTL metabolism, cognition, and clinical features were evaluated using GLMs or logistic regression models separately performed for the AD spectrum (NC + AD + MD), DLB spectrum (NC + DLB + MD), and disease groups (AD + DLB + MD). Covariates included age, sex, and education. RESULTS: AD was associated with hippocampal/entorhinal hypometabolism, whereas DLB was associated with relative amygdalar/hippocampal hypermetabolism. Relative MTL hypermetabolism was associated with lower attention/visuospatial/executive scores and severe parkinsonism in both the AD and DLB spectra and disease groups. Left hippocampal/entorhinal hypometabolism was associated with lower verbal memory scores, whereas right hippocampal hypometabolism was associated with lower visual memory scores in both the AD spectrum and disease groups. Relative MTL hypermetabolism was associated with an increased risk of CF and VH in the disease group, and relative amygdalar hypermetabolism was associated with an increased risk of VH in the DLB spectrum. CONCLUSIONS: Entorhinal-hippocampal hypometabolism and relative amygdala-hippocampal hypermetabolism could be characteristics of AD- and DLB-related neurodegeneration, respectively.


Subject(s)
Alzheimer Disease , Fluorodeoxyglucose F18 , Lewy Body Disease , Neuropsychological Tests , Positron-Emission Tomography , Temporal Lobe , Humans , Lewy Body Disease/metabolism , Lewy Body Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Female , Male , Aged , Temporal Lobe/metabolism , Temporal Lobe/diagnostic imaging , Aged, 80 and over , Middle Aged
3.
Neuroscience ; 546: 75-87, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38552733

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which there are very limited treatment options. Dysfunction of the excitatory neurotransmitter system is thought to play a major role in the pathogenesis of this condition. Vesicular glutamate transporters (VGLUTs) are key to controlling the quantal release of glutamate. Thus, expressional changes in disease can have implications for aberrant neuronal activity, raising the possibility of a therapeutic target. There is no information regarding the expression of VGLUTs in the human medial temporal lobe in AD, one of the earliest and most severely affected brain regions. This study aimed to quantify and compare the layer-specific expression of VGLUT1 and VGLUT2 between control and AD cases in the hippocampus, subiculum, entorhinal cortex, and superior temporal gyrus. Free-floating fluorescent immunohistochemistry was used to label VGLUT1 and VGLUT2 in the hippocampus, subiculum, entorhinal cortex, and superior temporal gyrus. Sections were imaged using laser-scanning confocal microscopy and transporter densitometric analysis was performed. VGLUT1 density was not significantly different in AD tissue, except lower staining density observed in the dentate gyrus stratum moleculare (p = 0.0051). VGLUT2 expression was not altered in the hippocampus and entorhinal cortex of AD cases but was significantly lower in the subiculum (p = 0.015) and superior temporal gyrus (p = 0.0023). This study indicates a regionally specific vulnerability of VGLUT1 and VGLUT2 expression in the medial temporal lobe and superior temporal gyrus in AD. However, the causes and functional consequences of these disturbances need to be further explored to assess VGLUT1 and VGLUT2 as viable therapeutic targets.


Subject(s)
Alzheimer Disease , Temporal Lobe , Vesicular Glutamate Transport Protein 1 , Vesicular Glutamate Transport Protein 2 , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Temporal Lobe/metabolism , Temporal Lobe/pathology , Male , Vesicular Glutamate Transport Protein 1/metabolism , Aged , Female , Vesicular Glutamate Transport Protein 2/metabolism , Aged, 80 and over , Middle Aged , Immunohistochemistry
4.
Nature ; 625(7994): 345-351, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38057661

ABSTRACT

Frontotemporal lobar degeneration (FTLD) causes frontotemporal dementia (FTD), the most common form of dementia after Alzheimer's disease, and is often also associated with motor disorders1. The pathological hallmarks of FTLD are neuronal inclusions of specific, abnormally assembled proteins2. In the majority of cases the inclusions contain amyloid filament assemblies of TAR DNA-binding protein 43 (TDP-43) or tau, with distinct filament structures characterizing different FTLD subtypes3,4. The presence of amyloid filaments and their identities and structures in the remaining approximately 10% of FTLD cases are unknown but are widely believed to be composed of the protein fused in sarcoma (FUS, also known as translocated in liposarcoma). As such, these cases are commonly referred to as FTLD-FUS. Here we used cryogenic electron microscopy (cryo-EM) to determine the structures of amyloid filaments extracted from the prefrontal and temporal cortices of four individuals with FTLD-FUS. Surprisingly, we found abundant amyloid filaments of the FUS homologue TATA-binding protein-associated factor 15 (TAF15, also known as TATA-binding protein-associated factor 2N) rather than of FUS itself. The filament fold is formed from residues 7-99 in the low-complexity domain (LCD) of TAF15 and was identical between individuals. Furthermore, we found TAF15 filaments with the same fold in the motor cortex and brainstem of two of the individuals, both showing upper and lower motor neuron pathology. The formation of TAF15 amyloid filaments with a characteristic fold in FTLD establishes TAF15 proteinopathy in neurodegenerative disease. The structure of TAF15 amyloid filaments provides a basis for the development of model systems of neurodegenerative disease, as well as for the design of diagnostic and therapeutic tools targeting TAF15 proteinopathy.


Subject(s)
Frontotemporal Lobar Degeneration , TATA-Binding Protein Associated Factors , Humans , Amyloid/chemistry , Amyloid/metabolism , Amyloid/ultrastructure , Brain Stem/metabolism , Brain Stem/pathology , Cryoelectron Microscopy , Frontotemporal Dementia/etiology , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Frontotemporal Lobar Degeneration/complications , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Motor Cortex/metabolism , Motor Cortex/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , TATA-Binding Protein Associated Factors/ultrastructure , Temporal Lobe/metabolism , Temporal Lobe/pathology
5.
Epilepsia Open ; 9(1): 187-199, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37881152

ABSTRACT

OBJECTIVE: The study investigated metabolic connectivity (MC) differences between patients with unilateral drug-resistant mesial temporal lobe epilepsy (MTLE) with hippocampal sclerosis (HS) and healthy controls (HCs), based on [18 F]-fluorodeoxyglucose (FDG)-PET data. We focused on the MC changes dependent on the lateralization of the epileptogenic lobe and on correlations with postoperative outcomes. METHODS: FDG-PET scans of 47 patients with unilateral MTLE with histopathologically proven HS and 25 HC were included in the study. All the patients underwent a standard anterior temporal lobectomy and were more than 2 years after the surgery. MC changes were compared between the two HS groups (left HS, right HS) and HC. Differences between the metabolic network of seizure-free and non-seizure-free patients after surgery were depicted afterward. Network changes were correlated with clinical characteristics. RESULTS: The study showed widespread metabolic network changes in the HS patients as compared to HC. The changes were more extensive in the right HS than in the left HS. Unfavorable surgical outcomes were found in patients with decreased MC within the network including both the lesional and contralesional hippocampus, ipsilesional frontal operculum, and contralesional insula. Favorable outcomes correlated with decreased MC within the network involving both orbitofrontal cortices and the ipsilesional temporal lobe. SIGNIFICANCE: There are major differences in the metabolic networks of left and right HS, with more extensive changes in right HS. The changes within the metabolic network could help predict surgical outcomes in patients with HS. MC may identify patients with potentially unfavorable outcomes and direct them to a more detailed presurgical evaluation. PLAIN LANGUAGE SUMMARY: Metabolic connectivity is a promising method for metabolic network mapping. Metabolic networks in mesial temporal lobe epilepsy are dependent on lateralization of the epileptogenic lobe and could predict surgical outcomes.


Subject(s)
Drug Resistant Epilepsy , Epilepsy, Temporal Lobe , Humans , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/surgery , Fluorodeoxyglucose F18/metabolism , Temporal Lobe/metabolism , Hippocampus/surgery , Hippocampus/metabolism , Treatment Outcome
6.
BMC Med Imaging ; 23(1): 185, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37964218

ABSTRACT

BACKGROUND: 1H magnetic resonance spectroscopy (1H-MRS) can be used to study neurological disorders because it can be utilized to examine the concentrations of related metabolites. However, the diagnostic utility of different field strengths for temporal lobe epilepsy (TLE) remains unclear. The purpose of this study is to make quantitative comparisons of metabolites of TLE at 1.5T and 3.0T and evaluate their efficacy. METHODS: Our retrospective collections included the single-voxel 1H-MRS of 23 TLE patients and 17 healthy control volunteers (HCs) with a 1.5T scanner, as well as 29 TLE patients and 17 HCs with a 3.0T scanner. Particularly, HCs were involved both the scans with 1.5T and 3.0T scanners, respectively. The metabolites, including the N-acetylaspartate (NAA), creatine (Cr), and choline (Cho), were measured in the left or right temporal pole of brain. To analyze the ratio of brain metabolites, including NAA/Cr, NAA/Cho, NAA/(Cho + Cr) and Cho/Cr, four controlled experiments were designed to evaluate the diagnostic utility of TLE on 1.5T and 3.0T MRS, included: (1) 1.5T TLE group vs. 1.5T HCs by the Mann-Whitney U Test, (2) 3.0T TLE group vs. 3.0T HCs by the Mann-Whitney U Test, (3) the power analysis for the 1.5T and 3.0T scanner, and (4) 3.0T HCs vs. 1.5T HCs by Paired T-Test. RESULTS: Three metabolite ratios (NAA/Cr, NAA/Cho, and NAA/(Cho + Cr) showed the same statistical difference (p < 0.05) in distinguishing the TLE from HCs in the bilateral temporal poles when using 1.5T or 3.0T scanners. Similarly, the power analysis demonstrated that four metabolite ratios (NAA/Cr, NAA/Cho, NAA/(Cho + Cr), Cho/Cr) had similar distinction abilities between 1.5T and 3.0T scanner, denoting both 1.5T and 3.0T scanners were provided with similar sensitivities and reproducibilities for metabolites detection. Moreover, the metabolite ratios of the same healthy volunteers were not statistically different between 1.5T and 3.0T scanners, except for NAA/Cho (p < 0.05). CONCLUSIONS: 1.5T and 3.0T scanners may have comparable diagnostic potential when 1H-MRS was used to diagnose patients with TLE.


Subject(s)
Epilepsy, Temporal Lobe , Humans , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/metabolism , Magnetic Resonance Imaging , Retrospective Studies , Magnetic Resonance Spectroscopy/methods , Temporal Lobe/metabolism , Creatine/metabolism , Choline
7.
Hum Brain Mapp ; 44(18): 6364-6374, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37846762

ABSTRACT

Alzheimer's disease (AD) is one of the most prevalent forms of dementia in older individuals. Convergent evidence suggests structural connectome abnormalities in specific brain regions are linked to AD progression. The biological basis underpinnings of these connectome changes, however, have remained elusive. We utilized an individual regional mean connectivity strength (RMCS) derived from a regional radiomics similarity network to capture altered morphological connectivity in 1654 participants (605 normal controls, 766 mild cognitive impairment [MCI], and 283 AD). Then, we also explored the biological basis behind these morphological changes through gene enrichment analysis and cell-specific analysis. We found that RMCS probes of the hippocampus and medial temporal lobe were significantly altered in AD and MCI, with these differences being spatially related to the expression of AD-risk genes. In addition, gene enrichment analysis revealed that the modulation of chemical synaptic transmission is the most relevant biological process associated with the altered RMCS in AD. Notably, neuronal cells were found to be the most pertinent cells in the altered RMCS. Our findings shed light on understanding the biological basis of structural connectome changes in AD, which may ultimately lead to more effective diagnostic and therapeutic strategies for this devastating disease.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Connectome , Humans , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Magnetic Resonance Imaging , Brain/diagnostic imaging , Brain/metabolism , Temporal Lobe/diagnostic imaging , Temporal Lobe/metabolism , Cognitive Dysfunction/diagnostic imaging , Transcription, Genetic
8.
Int J Mol Sci ; 24(19)2023 Sep 23.
Article in English | MEDLINE | ID: mdl-37833937

ABSTRACT

The European Commission of the International League Against Epilepsy (ILAE) has identified glial mechanisms of seizures and epileptogenesis as top research priorities. The aim of our study was to conduct a comparative analysis of the expression levels of cytoskeletal proteins (glial fibrillar acidic protein (GFAP) and vimentin), protective protein S100, and proapoptotic caspase-3 protein in patients with drug-resistant epilepsy (DRE) associated with focal cortical dysplasia (FCD). We aimed to investigate how the expression levels of these proteins depend on age (both in children and adults), gender, and disease duration, using immunohistochemistry. Nonparametric statistical methods were employed for data analysis. In the epileptic focus area of the cortex and white matter in patients with FCD-associated temporal lobe DRE, a higher level of expression of these proteins was observed. Age and gender differences were found for vimentin and S100. In the early stages of disease development, there was a compensatory sequential increase in the expression of cytoskeletal and protective proteins. In patients with DRE, depending on the disease duration, patterns of development of neurodegeneration were noted, which is accompanied by apoptosis of gliocytes. These results provide insights into epilepsy mechanisms and may contribute to improving diagnostic and treatment approaches.


Subject(s)
Drug Resistant Epilepsy , Epilepsy, Temporal Lobe , Epilepsy , Focal Cortical Dysplasia , Humans , Adult , Child , Epilepsy, Temporal Lobe/metabolism , Vimentin/genetics , Vimentin/metabolism , Cytoskeletal Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Epilepsy/metabolism , Temporal Lobe/metabolism , Glial Fibrillary Acidic Protein/metabolism , Retrospective Studies
9.
Science ; 382(6667): eadf2359, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824649

ABSTRACT

Single-cell transcriptomic studies have identified a conserved set of neocortical cell types from small postmortem cohorts. We extended these efforts by assessing cell type variation across 75 adult individuals undergoing epilepsy and tumor surgeries. Nearly all nuclei map to one of 125 robust cell types identified in the middle temporal gyrus. However, we found interindividual variance in abundances and gene expression signatures, particularly in deep-layer glutamatergic neurons and microglia. A minority of donor variance is explainable by age, sex, ancestry, disease state, and cell state. Genomic variation was associated with expression of 150 to 250 genes for most cell types. This characterization of cellular variation provides a baseline for cell typing in health and disease.


Subject(s)
Temporal Lobe , Transcriptome , Adult , Humans , Epilepsy/metabolism , Gene Expression Profiling , Neurons/metabolism , Temporal Lobe/cytology , Temporal Lobe/metabolism , Nervous System Diseases/genetics , Mental Disorders/genetics
10.
J Alzheimers Dis ; 96(1): 313-328, 2023.
Article in English | MEDLINE | ID: mdl-37742643

ABSTRACT

BACKGROUND: In Alzheimer's disease (AD), the gradual accumulation of amyloid-ß (Aß) and tau proteins may underlie alterations in empathy. OBJECTIVE: To assess whether tau aggregation in the medial temporal lobes related to differences in cognitive empathy (the ability to take others' perspectives) and emotional empathy (the ability to experience others' feelings) in AD. METHODS: Older adults (n = 105) completed molecular Aß positron emission tomography (PET) scans. Sixty-eight of the participants (35 women) were Aß positive and symptomatic with diagnoses of mild cognitive impairment, dementia of the Alzheimer's type, logopenic variant primary progressive aphasia, or posterior cortical atrophy. The remaining 37 (22 women) were asymptomatic Aß negative healthy older controls. Using the Interpersonal Reactivity Index, we compared current levels of informant-rated cognitive empathy (Perspective-Taking subscale) and emotional empathy (Empathic Concern subscale) in the Aß positive and negative participants. The Aß positive participants also underwent molecular tau-PET scans, which were used to investigate whether regional tau burden in the bilateral medial temporal lobes related to empathy. RESULTS: Aß positive participants had lower perspective-taking and higher empathic concern than Aß negative healthy controls. Medial temporal tau aggregation in the Aß positive participants had divergent associations with cognitive and emotional empathy. Whereas greater tau burden in the amygdala predicted lower perspective-taking, greater tau burden in the entorhinal cortex predicted greater empathic concern. Tau burden in the parahippocampal cortex did not predict either form of empathy. CONCLUSIONS: Across AD clinical syndromes, medial temporal lobe tau aggregation is associated with lower perspective-taking yet higher empathic concern.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Female , Aged , Alzheimer Disease/metabolism , Empathy , tau Proteins/metabolism , Temporal Lobe/diagnostic imaging , Temporal Lobe/metabolism , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/psychology , Positron-Emission Tomography/methods , Cognition
11.
Eur J Nucl Med Mol Imaging ; 51(1): 168-179, 2023 12.
Article in English | MEDLINE | ID: mdl-37707571

ABSTRACT

PURPOSE: Temporal lobe epilepsy (TLE) is a common, polygenic epilepsy syndrome that involves glucose hypometabolism in the epileptogenic zone. However, the transcriptional and cellular signatures underlying the metabolism in TLE remain unclear. METHODS: In this retrospective study, 2-[18F]-fluoro-2-deoxy-D-glucose ([18F]FDG) positron emission tomography (PET) scans of TLE patients (n = 104) who underwent anterior temporal lobectomy were consecutively collected between 2016 and 2021. The transcriptional profiles of TLE risk genes across the brain were identified by the gene expression analyses from six TLE patients and twelve postmortem donors (six from the Allen Human Brain Atlas). Integrating the neuroimaging and transcriptomic data, we examined the relationship between the expression of TLE-associated genes and metabolic alterations in TLE. Furthermore, we performed functional enrichment analyses of the genes with higher weight in partial least squares regression using Metascape. RESULTS: A total of 104 patients with TLE (mean age 29 ± 9 years, 50% male) and 30 healthy controls (HCs) (mean age 31 ± 6 years, 53% male) were enrolled. Compared to that of HCs, patients with TLE showed hypometabolism in the temporal lobes and adjacent structures but hypermetabolism in the thalamus and basal ganglia. The cortical map of inter-group differences in cerebral metabolism was spatially correlated with the expression of a weighted combination of genes enriched in ontology terms and pathways related to neurovascular unit (NVU) integrity and synaptic plasticity. DISCUSSION: Our findings, combined with the analysis of neuroimaging and transcriptional data, suggest that genes related to NVU integrity and synaptic plasticity may drive alterations to brain metabolism that mediate the genetic risk of TLE.


Subject(s)
Epilepsy, Temporal Lobe , Humans , Male , Young Adult , Adult , Female , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/genetics , Retrospective Studies , Brain/diagnostic imaging , Brain/metabolism , Temporal Lobe/diagnostic imaging , Temporal Lobe/metabolism , Positron-Emission Tomography/methods , Fluorodeoxyglucose F18/metabolism , Glucose/metabolism , Magnetic Resonance Imaging
12.
Acta Neuropathol Commun ; 11(1): 130, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37563653

ABSTRACT

Right temporal variant frontotemporal dementia, also called right-predominant semantic dementia, often has an unclear position within the framework of the updated diagnostic criteria for behavioral variant frontotemporal dementia or primary progressive aphasia. Recent studies have suggested that this population may be clinically, neuropathologically, and genetically distinct from those with behavioral variant frontotemporal dementia or left-predominant typical semantic variant primary progressive aphasia. Here we describe a Japanese case of right temporal variant frontotemporal dementia with novel heterozygous MAPT mutation Adenine to Thymidine in intervening sequence (IVS) 9 at position -7 from 3' splicing site of intron 9/exon 10 boundary (MAPT IVS9-7A > T). Postmortem neuropathological analysis revealed a predominant accumulation of 4 repeat tau, especially in the temporal lobe, amygdala, and substantia nigra, but lacked astrocytic plaques or tufted astrocytes. Immunoelectron microscopy of the tau filaments extracted from the brain revealed a ribbon-like structure. Moreover, a cellular MAPT splicing assay confirmed that this novel variant promoted the inclusion of exon 10, resulting in the predominant production of 4 repeat tau. These data strongly suggest that the MAPT IVS9-7 A > T variant found in our case is a novel mutation that stimulates the inclusion of exon 10 through alternative splicing of MAPT transcript and causes predominant 4 repeat tauopathy which clinically presents as right temporal variant frontotemporal dementia.


Subject(s)
Aphasia, Primary Progressive , Frontotemporal Dementia , Pick Disease of the Brain , Tauopathies , Humans , Aphasia, Primary Progressive/pathology , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Introns/genetics , Mutation , Pick Disease of the Brain/pathology , tau Proteins/genetics , tau Proteins/metabolism , Tauopathies/genetics , Tauopathies/pathology , Temporal Lobe/metabolism
13.
Int J Mol Sci ; 24(13)2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37446307

ABSTRACT

Temporal lobe epilepsy (TLE) is the most common form of epilepsy in adults. Tissue reorganization at the site of the epileptogenic focus is accompanied by changes in the expression patterns of protein molecules. The study of mRNA and its corresponding proteins is crucial for understanding the pathogenesis of the disease. Protein expression profiles do not always directly correlate with the levels of their transcripts; therefore, it is protein profiling that is no less important for understanding the molecular mechanisms and biological processes of TLE. The study and annotation of proteins that are statistically significantly different in patients with TLE is an approach to search for biomarkers of this disease, various stages of its development, as well as a method for searching for specific targets for the development of a further therapeutic strategy. When writing a systematic review, the following aggregators of scientific journals were used: MDPI, PubMed, ScienceDirect, Springer, and Web of Science. Scientific articles were searched using the following keywords: "proteomic", "mass-spectrometry", "protein expression", "temporal lobe epilepsy", and "biomarkers". Publications from 2003 to the present have been analyzed. Studies of brain tissues, experimental models of epilepsy, as well as biological fluids, were analyzed. For each of the groups, aberrantly expressed proteins found in various studies were isolated. Most of the studies omitted important characteristics of the studied patients, such as: duration of illness, type and response to therapy, gender, etc. Proteins that overlap across different tissue types and different studies have been highlighted: DPYSL, SYT1, STMN1, APOE, NME1, and others. The most common biological processes for them were the positive regulation of neurofibrillary tangle assembly, the regulation of amyloid fibril formation, lipoprotein catabolic process, the positive regulation of vesicle fusion, the positive regulation of oxidative stress-induced intrinsic apoptotic signaling pathway, removal of superoxide radicals, axon extension, and the regulation of actin filament depolymerization. MS-based proteomic profiling for a relevant study must accept a number of limitations, the most important of which is the need to compare different types of neurological and, in particular, epileptic disorders. Such a criterion could increase the specificity of the search work and, in the future, lead to the discovery of biomarkers for a particular disease.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Adult , Humans , Epilepsy, Temporal Lobe/metabolism , Epilepsy/metabolism , Proteins/metabolism , Mass Spectrometry , Biomarkers/metabolism , Temporal Lobe/metabolism
14.
Int J Mol Sci ; 24(14)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37511361

ABSTRACT

Limited comparative data exist on the molecular spectrum of amyloid-beta (Aß) and tau deposition in individuals with Down syndrome (DS) and sporadic Alzheimer's disease (sAD). We assessed Aß and tau deposition severity in the temporal lobe and cerebellum of ten DS and ten sAD cases. Immunohistochemistry was performed using antibodies against eight different Aß epitopes (6F/3D, Aß38, Aß39, Aß40, Aß42, Aß43, pyroglutamate Aß at third glutamic acid (AßNp3E), phosphorylated- (p-)Aß at 8th serine (AßpSer8)), and six different pathological tau epitopes (p-Ser202/Thr205, p-Thr231, p-Ser396, Alz50, MC1, GT38). Findings were evaluated semi-quantitatively and quantitatively using digital pathology. DS cases had significantly higher neocortical parenchymal deposition (Aß38, Aß42, and AßpSer8), and cerebellar parenchymal deposition (Aß40, Aß42, AßNp3E, and AßpSer8) than sAD cases. Furthermore, DS cases had a significantly larger mean plaque size (6F/3D, Aß42, AßNp3E) in the temporal lobe, and significantly greater deposition of cerebral and cerebellar Aß42 than sAD cases in the quantitative analysis. Western blotting corroborated these findings. Regarding tau pathology, DS cases had significantly more severe cerebral tau deposition than sAD cases, especially in the white matter (p-Ser202/Thr205, p-Thr231, Alz50, and MC1). Greater total tau deposition in the white matter (p-Ser202/Thr205, p-Thr231, and Alz50) of DS cases was confirmed by quantitative analysis. Our data suggest that the Aß and tau molecular signatures in DS are distinct from those in sAD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Down Syndrome , tau Proteins , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Cerebellum/metabolism , Down Syndrome/genetics , Down Syndrome/metabolism , Down Syndrome/pathology , Peptide Fragments , tau Proteins/genetics , tau Proteins/metabolism , Temporal Lobe/metabolism
15.
Epilepsy Behav ; 145: 109354, 2023 08.
Article in English | MEDLINE | ID: mdl-37473654

ABSTRACT

BACKGROUND: Studies have shown conflicting results in the correlation between serotonin-1A (5-HT1A) receptor binding levels in the brain and temporal lobe epilepsy (TLE). There is a need to systematically evaluate the correlation between the 5-HT1A binding level and TLE from the perspective of the brain using molecular imaging. METHODS: Chinese and English databases, such as the China National Knowledge Infrastructure (CNKI), the China Science and Technology Journal Database (VIP), WanFang, the Chinese Biomedical Literature Service System (SinoMed), PubMed and Web of Science, were searched. RESULTS: Two evaluators independently screened the literature, extracted data, and evaluated the risk of bias in the included studies according to the inclusion and exclusion criteria. RevMan 5.4.1 was used to analyze the data. A total of 196 participants were included; of these, 95 had TLE and 131 were healthy controls who had never had a seizure before participating in the study. Meta-analysis results suggested that 1) decreased 5-HT1A binding was found on the affected side of patients with TLE (standard mean difference (SMD) = -1.45, 95% confidence interval (CI) [-2.27, -0.64], Z = 3.48, P = 0.0005); 2) decreased 5-HT1A binding was found in the ipsilateral hippocampus of patients with TLE (SMD = -1.76, 95% CI [-2.51, -1.00], Z = 4.57, P<0.00001); 3) decreased 5-HT1A binding was found in the ipsilateral temporal lobe cortex of patients with TLE (SMD = -0.46, 95% CI [-0.80, -0.12], Z = 2.66, P = 0.008); 4) decreased 5-HT1A binding was found in the ipsilateral amygdala in patients with TLE (SMD = -1.36, 95% CI [-2.48, -0.23], Z = 2.37, P = 0.02); and 5) decreased 5-HT1A binding was found in the frontal lobe of patients with TLE(SMD = -0.75, 95% CI [-1.29, -0.20], Z = 2.67, P = 0.008). CONCLUSION: A reduction in 5-HT1A binding in the hippocampus, temporal cortex, amygdala, and frontal lobe was observed on the affected side of patients with TLE. The decrease in 5-HT1A binding can be considered related to TLE. Potentially relevant factors should be considered in future molecular imaging studies.


Subject(s)
Epilepsy, Temporal Lobe , Humans , Brain/diagnostic imaging , Brain/metabolism , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Magnetic Resonance Imaging/methods , Seizures/metabolism , Temporal Lobe/metabolism
16.
Mol Neurobiol ; 60(10): 5755-5769, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37341859

ABSTRACT

The purpose of this study was to identify and validate new putative lead drug targets in drug-resistant mesial temporal lobe epilepsy (mTLE) starting from differentially expressed genes (DEGs) previously identified in mTLE in humans by transcriptome analysis. We identified consensus DEGs among two independent mTLE transcriptome datasets and assigned them status as "lead target" if they (1) were involved in neuronal excitability, (2) were new in mTLE, and (3) were druggable. For this, we created a consensus DEG network in STRING and annotated it with information from the DISEASES database and the Target Central Resource Database (TCRD). Next, we attempted to validate lead targets using qPCR, immunohistochemistry, and Western blot on hippocampal and temporal lobe neocortical tissue from mTLE patients and non-epilepsy controls, respectively. Here we created a robust, unbiased list of 113 consensus DEGs starting from two lists of 3040 and 5523 mTLE significant DEGs, respectively, and identified five lead targets. Next, we showed that CACNB3, a voltage-gated Ca2+ channel subunit, was significantly regulated in mTLE at both mRNA and protein level. Considering the key role of Ca2+ currents in regulating neuronal excitability, this suggested a role for CACNB3 in seizure generation. This is the first time changes in CACNB3 expression have been associated with drug-resistant epilepsy in humans, and since efficient therapeutic strategies for the treatment of drug-resistant mTLE are lacking, our finding might represent a step toward designing such new treatment strategies.


Subject(s)
Drug Resistant Epilepsy , Epilepsy, Temporal Lobe , Humans , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/complications , Temporal Lobe/metabolism , Seizures/metabolism , Hippocampus/metabolism , Drug Resistant Epilepsy/genetics , Drug Resistant Epilepsy/metabolism
17.
Ann Neurol ; 94(1): 1-12, 2023 07.
Article in English | MEDLINE | ID: mdl-37183762

ABSTRACT

The anatomical distribution of most neurodegenerative diseases shows considerable interindividual variations. In contrast, frontotemporal lobar degeneration with transactive response DNA-binding protein type C (TDP-C) shows a consistent predilection for the anterior temporal lobe (ATL). The relatively selective atrophy of ATL in TDP-C patients has highlighted the importance of this region for complex cognitive and behavioral functions. This review includes observations on 28 TDP-C patients, 18 with semantic primary progressive aphasia and 10 with other syndromes. Longitudinal imaging allowed the delineation of progression trajectories. At post-mortem examination, the pathognomonic feature of TDP-C consisted of long, thick neurites found predominantly in superficial cortical layers. These neurites may represent dystrophic apical dendrites of layer III and V pyramidal neurons that are known to play pivotal roles in complex cortical computations. Other types of frontotemporal lobar degeneration TDP, such as TDP-A and TDP-B, are not associated with long dystrophic neurites in the cerebral cortex, and do not show similar predilection patterns for ATL. Research is beginning to identify molecular, structural, and immunological differences between pathological TDP-43 in TDP-C versus TDP-A and B. Parallel investigations based on proteomics, somatic mutations, and genome-wide association studies are detecting molecular features that could conceivably mediate the selective vulnerability of ATL to TDP-C. Future work will focus on characterizing the distinctive features of the abnormal TDP-C neurites, the mechanisms of neurotoxicity, initial cellular targets within the ATL, trajectory of spread, and the nature of ATL-specific markers that modulate vulnerability to TDP-C. ANN NEUROL 2023;94:1-12.


Subject(s)
Frontotemporal Dementia , Frontotemporal Lobar Degeneration , Humans , Genome-Wide Association Study , Brain/pathology , Frontotemporal Dementia/metabolism , Temporal Lobe/metabolism , Frontotemporal Lobar Degeneration/pathology , Atrophy/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
18.
Neurology ; 101(4): e370-e385, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37258299

ABSTRACT

BACKGROUND AND OBJECTIVES: Sleep disordered breathing (SDB) has been related to amyloid deposition and an increased dementia risk. However, how SDB relates to medial temporal lobe neurodegeneration and subsequent episodic memory impairment is unclear. Our objective was to investigate the impact of amyloid positivity on the associations between SDB severity, medial temporal lobe subregions, and episodic memory performance in cognitively unimpaired older adults. METHODS: Data were acquired between 2016 and 2020 in the context of the Age-Well randomized controlled trial of the Medit-Aging European project. Participants older than 65 years who were free of neurologic, psychiatric, or chronic medical diseases were recruited from the community. They completed a neuropsychological evaluation, in-home polysomnography, a Florbetapir PET, and an MRI, including a specific high-resolution assessment of the medial temporal lobe and hippocampal subfields. Multiple linear regressions were conducted to test interactions between amyloid status and SDB severity on the volume of MTL subregions, controlling for age, sex, education, and the ApoE4 status. Secondary analyses aimed at investigating the links between SDB, MTL subregional atrophy, and episodic memory performance at baseline and at a mean follow-up of 20.66 months in the whole cohort and in subgroups stratified according to amyloid status. RESULTS: We included 122 cognitively intact community-dwelling older adults (mean age ± SD: 69.40 ± 3.85 years, 77 women, 26 Aß+ individuals) in baseline analyses and 111 at follow-up. The apnea-hypopnea index interacted with entorhinal (ß = -0.81, p < 0.001, pη2 = 0.19), whole hippocampal (ß = -0.61, p < 0.001, pη2 = 0.10), subiculum (ß = -0.56, p = 0.002, pη2 = 0.08), CA1 (ß = -0.55, p = 0.002, pη2 = 0.08), and DG (ß = -0.53, p = 0.003, pη2 = 0.08) volumes such that a higher sleep apnea severity was related to lower MTL subregion volumes in amyloid-positive individuals, but not in those who were amyloid negative. In the whole cohort, lower whole hippocampal (r = 0.27, p = 0.005) and CA1 (r = 0.28, p = 0.003) volumes at baseline were associated with worse episodic memory performance at follow-up. DISCUSSION: Overall, we showed that SDB was associated with MTL atrophy in cognitively asymptomatic older adults engaged in the Alzheimer continuum, which may increase the risk of developing memory impairment over time. TRIAL REGISTRATION INFORMATION: ClinicalTrials.gov Identifier: NCT02977819.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Female , Aged , Temporal Lobe/metabolism , Acrylates , Amyloid/metabolism , Magnetic Resonance Imaging , Amyloidogenic Proteins , Atrophy , Positron-Emission Tomography , Amyloid beta-Peptides/metabolism
19.
Neuropathol Appl Neurobiol ; 49(3): e12913, 2023 06.
Article in English | MEDLINE | ID: mdl-37209022

ABSTRACT

AIM: The functions of the interlaminar astrocytes in layer I of the human cortex are currently unknown. Here, we aimed to explore whether there is any morphological remodelling of interlaminar astrocytes in layer I of the temporal cortex in epilepsy. METHODS: Tissues were obtained from 17 epilepsy surgery patients and 17 post-mortem age-matched controls. In addition, 10 Alzheimer's disease (AD) patients and 10 age-matched controls were used as the disease control group. Paraffin sections (6 µm) and frozen sections (35 or 150 µm) of inferior temporal gyrus tissue were used for immunohistochemistry. With the use of tissue transparency, 3D reconstruction and hierarchical clustering, we performed a quantitative morphological analysis of astrocytes. RESULTS: Upper and lower zones were identified in layer I of the human cortex. Compared with the astrocytes in layers IV-V, layer I interlaminar astrocytes occupied a significantly smaller volume and exhibited shorter and fewer process intersections. Increased Chaslin's gliosis (consisting of types I and II subpial interlaminar astrocytes) and number of glial fibrillary acidic protein (GFAP)-immunoreactive interlaminar astrocytes in layer I of the temporal cortex were confirmed in patients with epilepsy. There was no difference in the number of interlaminar astrocytes in layer I between AD and age-matched control groups. Using tissue transparency and 3D reconstruction technology, the astrocyte domain in the human temporal cortex was classified into four clusters, among which the interlaminar astrocytes in cluster II were more abundant in epilepsy, showing specific topological structures in patients with epilepsy. Furthermore, there was a significant increase in the astrocyte domain of interlaminar cells in layer I of the temporal cortex in patients with epilepsy. CONCLUSION: The observed significant astrocytic structural remodelling in the temporal cortex of epilepsy patients showed that the astrocyte domain in layer I may play an important role in temporal lobe epilepsy.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Humans , Astrocytes/metabolism , Epilepsy/metabolism , Temporal Lobe/metabolism , Cerebral Cortex/metabolism , Epilepsy, Temporal Lobe/metabolism
20.
J Psychopharmacol ; 37(5): 510-519, 2023 05.
Article in English | MEDLINE | ID: mdl-37070169

ABSTRACT

BACKGROUND: Childhood trauma (CT) has been linked to increased risk for mental illness in adulthood. Although work in experimental animals has shown that early life stressors can affect inhibitory and excitatory neurotransmission in adult rodents, with possible excitotoxic effects on local grey matter volumes (GMV), the neurobiological mechanisms that mediate this relationship in humans remain poorly understood. AIM: To examine glutamate and gamma-aminobutyric acid (GABA) metabolite concentrations and potential excitotoxic effects on GMV, in adults who experienced CT. METHODS: Fifty-six young adults (Mage = 20.41) were assigned to High CT (n = 29) and Low CT (n = 27) groups (by using the CT questionnaire) and underwent magnetic resonance spectroscopy (1H-MRS) to measure temporal lobe metabolite concentrations and volumetric imaging to measure GMV. RESULTS: Glutamate concentrations did not differ between groups; however, relative to the Low CT group, participants in the High CT group had reduced GABA concentrations in the left superior temporal gyrus (STG) voxel. Furthermore, logistic regression showed that participants with low left STG GABA concentrations and low left STG volumes were significantly more likely to be in the high CT group. CONCLUSIONS: This study provides the first evidence that both low GABA concentrations and its interaction with GMV in the left STG are associated with high levels of CT and suggest that altered inhibitory neurotransmission/metabolism may be linked to a lower GMV in the left STG in adults who experienced CT. Future studies are warranted to establish if utilizing these measures can stratify clinical high-risk and predict future clinical outcomes in high CT individuals.


Subject(s)
Adverse Childhood Experiences , Neurochemistry , Humans , Young Adult , Adult , Magnetic Resonance Imaging , Temporal Lobe/diagnostic imaging , Temporal Lobe/metabolism , Temporal Lobe/pathology , Glutamic Acid/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...