Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Semin Cell Dev Biol ; 139: 55-72, 2023 04.
Article in English | MEDLINE | ID: mdl-35292192

ABSTRACT

The presubiculum (PRS) is an integral component of the perforant pathway that has recently been recognised as a relatively unscathed region in clinical Alzheimer's disease (AD), despite neighbouring components of the perforant pathway, CA1 and the entorhinal cortex, responsible for formation of episodic memory and storage, showing severe hallmarks of AD including, amyloid-beta (Aß) plaques, tau tangles and marked gliosis. However, the question remains whether this anatomical resilience translates into functional resilience of the PRS neurons. Using neuroanatomy combined with whole-cell electrophysiological recordings, we investigated whether the unique spatial profile of the PRS was replicable in two knock-in mouse models of AD, APPNL-F/NL-F, and APPNL-F/MAPTHTAU and whether the intrinsic properties and morphological integrity of the PRS principal neurons was maintained compared to the lateral entorhinal cortex (LEC) and hippocampal CA1 principal cells. Our data revealed an age-dependent Aß and tau pathology with neuroinflammation in the LEC and CA1, but a presence of fleece-like Aß deposits with an absence of tau tangles and cellular markers of gliosis in the PRS of the mouse models at 11-16 and 18-22 months. These observations were consistent in human post-mortem AD tissue. This spatial profile also correlated with functional resilience of strong burst firing PRS pyramidal cells that showed unaltered sub- and suprathreshold intrinsic biophysical membrane properties and gross morphology in the AD models that were similar to the properties of pyramidal cells recorded in age-matched wild-type mice (11-14 months). This was in contrast to the LEC and CA1 principal cells which showed altered subthreshold intrinsic properties such as a higher input resistance, longer membrane time constants and hyperexcitability in response to suprathreshold stimulation that correlated with atrophied dendrites in both AD models. In conclusion, our data show for the first time that the unique anatomical profile of the PRS constitutes a diffuse AD pathology that is correlated with the preservation of principal pyramidal cell intrinsic biophysical and morphological properties despite alteration of LEC and CA1 pyramidal cells in two distinct genetic models of AD. Understanding the underlying mechanisms of this resilience could be beneficial in preventing the spread of disease pathology before cognitive deficits are precipitated in AD.


Subject(s)
Alzheimer Disease , Mice , Humans , Animals , Infant , Alzheimer Disease/metabolism , Gliosis/metabolism , Gliosis/pathology , Mice, Transgenic , Parahippocampal Gyrus/metabolism , Parahippocampal Gyrus/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Disease Models, Animal , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism
2.
Cell Rep ; 37(8): 110031, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818557

ABSTRACT

Brain circuits are comprised of distinct interconnected neurons that are assembled by synaptic recognition molecules presented by defined pre- and post-synaptic neurons. This cell-cell recognition process is mediated by varying cellular adhesion molecules, including the latrophilin family of adhesion G-protein-coupled receptors. Focusing on parahippocampal circuitry, we find that latrophilin-2 (Lphn2; gene symbol ADGRL2) is specifically enriched in interconnected subregions of the medial entorhinal cortex (MEC), presubiculum (PrS), and parasubiculum (PaS). Retrograde viral tracing from the Lphn2-enriched region of the MEC reveals unique topographical patterning of inputs arising from the PrS and PaS that mirrors Lphn2 expression. Using a Lphn2 conditional knockout mouse model, we find that deletion of MEC Lphn2 expression selectively impairs retrograde viral labeling of inputs arising from the ipsilateral PrS. Combined with analysis of Lphn2 expression within the MEC, this study reveals Lphn2 to be selectively expressed by defined cell types and essential for MEC-PrS circuit connectivity.


Subject(s)
Entorhinal Cortex/physiology , Receptors, Peptide/genetics , Animals , Entorhinal Cortex/metabolism , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Hippocampus/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/cytology , Neurons/physiology , Parahippocampal Gyrus/metabolism , Receptors, Peptide/metabolism
3.
Acta Neuropathol Commun ; 9(1): 128, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34289895

ABSTRACT

The medial temporal lobe (MTL) is a nidus for neurodegenerative pathologies and therefore an important region in which to study polypathology. We investigated associations between neurodegenerative pathologies and the thickness of different MTL subregions measured using high-resolution post-mortem MRI. Tau, TAR DNA-binding protein 43 (TDP-43), amyloid-ß and α-synuclein pathology were rated on a scale of 0 (absent)-3 (severe) in the hippocampus and entorhinal cortex (ERC) of 58 individuals with and without neurodegenerative diseases (median age 75.0 years, 60.3% male). Thickness measurements in ERC, Brodmann Area (BA) 35 and 36, parahippocampal cortex, subiculum, cornu ammonis (CA)1 and the stratum radiatum lacunosum moleculare (SRLM) were derived from 0.2 × 0.2 × 0.2 mm3 post-mortem MRI scans of excised MTL specimens from the contralateral hemisphere using a semi-automated approach. Spearman's rank correlations were performed between neurodegenerative pathologies and thickness, correcting for age, sex and hemisphere, including all four proteinopathies in the model. We found significant associations of (1) TDP-43 with thickness in all subregions (r = - 0.27 to r = - 0.46), and (2) tau with BA35 (r = - 0.31) and SRLM thickness (r = - 0.33). In amyloid-ß and TDP-43 negative cases, we found strong significant associations of tau with ERC (r = - 0.40), BA35 (r = - 0.55), subiculum (r = - 0.42) and CA1 thickness (r = - 0.47). This unique dataset shows widespread MTL atrophy in relation to TDP-43 pathology and atrophy in regions affected early in Braak stageing and tau pathology. Moreover, the strong association of tau with thickness in early Braak regions in the absence of amyloid-ß suggests a role of Primary Age-Related Tauopathy in neurodegeneration.


Subject(s)
Entorhinal Cortex/diagnostic imaging , Hippocampus/diagnostic imaging , Neurodegenerative Diseases/diagnostic imaging , Temporal Lobe/diagnostic imaging , Adult , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain Cortical Thickness , CA1 Region, Hippocampal/diagnostic imaging , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Case-Control Studies , DNA-Binding Proteins/metabolism , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Female , Frontotemporal Lobar Degeneration/diagnostic imaging , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Lewy Body Disease/diagnostic imaging , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurofibrillary Tangles/pathology , Parahippocampal Gyrus/diagnostic imaging , Parahippocampal Gyrus/metabolism , Parahippocampal Gyrus/pathology , Pick Disease of the Brain/diagnostic imaging , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/pathology , Plaque, Amyloid/pathology , Supranuclear Palsy, Progressive/diagnostic imaging , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/pathology , Temporal Lobe/metabolism , Temporal Lobe/pathology , alpha-Synuclein/metabolism , tau Proteins/metabolism
4.
Sci Rep ; 10(1): 17301, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33057124

ABSTRACT

Social cognition is facilitated by oxytocin receptors (OXTR) in the hippocampus, a brain region that changes dynamically with pregnancy, parturition, and parenting experience. We investigated the impact of parenthood on hippocampal OXTR in male and female titi monkeys, a pair-bonding primate species that exhibits biparental care of offspring. We hypothesized that in postmortem brain tissue, OXTR binding in the hippocampal formation would differ between parents and non-parents, and that OXTR density would correlate with frequencies of observed parenting and affiliative behaviors between partners. Subjects were 10 adult titi monkeys. OXTR binding in the hippocampus (CA1, CA2/3, CA4, dentate gyrus, subiculum) and presubiculum layers (PSB1, PSB3) was determined using receptor autoradiography. The average frequency of partner affiliation (Proximity, Contact, and Tail Twining) and infant carrying were determined from longitudinal observations (5-6 per day). Analyses showed that parents exhibited higher OXTR binding than non-parents in PSB1 (t(8) = - 2.33, p = 0.048), and that OXTR binding in the total presubiculm correlated negatively with Proximity (r = - 0.88) and Contact (r = - 0.91), but not Tail Twining or infant carrying. These results suggest that OXTR binding in the presubiculum supports pair bonding and parenting behavior, potentially by mediating changes in hippocampal plasticity.


Subject(s)
Behavior, Animal/physiology , Hippocampus/metabolism , Hippocampus/physiology , Neuronal Plasticity/physiology , Pair Bond , Parenting/psychology , Receptors, Oxytocin/metabolism , Social Cognition , Animals , Callicebus , Female , Male , Parahippocampal Gyrus/metabolism , Parahippocampal Gyrus/physiology
5.
Mech Ageing Dev ; 190: 111290, 2020 09.
Article in English | MEDLINE | ID: mdl-32603667

ABSTRACT

Diagnosis of Alzheimer's disease (AD) is often difficult because of distinct and subjective clinical features, especially in the early stage. FOXO3a protein present in the cognitive centre of brain in inferior temporal region and parahippocampus. FOXO3a can be a potential novel target against AD. AD, Mild Cognitive impairment (MCI) and Geriatric Control (GC) were recruited after diagnosis by clinical assessment, MRI, TauPET and FDG-PET. We have quantified serum FOXO3a by surface plasmon resonance (SPR) and compare with TauPET between of AD, MCI patients and GC. Serum FOXO3A was significantly lower in AD (1.42 ± 0.09 ng/µl) compare to MCI (1.61 ± 0.14 ng/µl) and GC (1.89 ± 0.07 ng/µl). However, the Tau was higher in AD both in serum and also in PET scan. Serum pTau was significantly over-expressed in AD (0.176 ± 0.03 ng/µl), compare to other groups; MCI (0.16 ± 0.014 ng/µl) and GC (0.15 ± 0.024 ng/µl). Serum FOXO3A could significantly differentiate AD vs MCI, MCI vs GC and AD vs GC. However, Tau protein could only differentiate AD vs GC but not MCI vs GC. Serum FOXO3A may serve as novel blood marker for early detection for AD and target for therapeutic intervention.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Forkhead Box Protein O3/blood , Aged , Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Biomarkers/blood , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/metabolism , Early Diagnosis , Female , Gene Expression Profiling , Geriatric Assessment/methods , Humans , Male , Parahippocampal Gyrus/diagnostic imaging , Parahippocampal Gyrus/metabolism , Positron-Emission Tomography/methods , Surface Plasmon Resonance/methods , Temporal Lobe/diagnostic imaging , Temporal Lobe/metabolism , tau Proteins/blood
6.
Neuroscience ; 424: 102-120, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31705965

ABSTRACT

Clinical evidence and pathological studies suggest a bidirectional link between temporal lobe epilepsy and Alzheimer's disease (AD). Data analysis from omic studies offers an excellent opportunity to identify the overlap in molecular alterations between the two pathologies. We have subjected proteomic data sets from a rat model of epileptogenesis to a bioinformatics analysis focused on proteins functionally linked with AD. The data sets have been obtained for hippocampus (HC) and parahippocampal cortex samples collected during the course of epileptogenesis. Our study confirmed a relevant dysregulation of proteins linked with Alzheimer pathogenesis. When comparing the two brain areas, a more prominent regulation was evident in parahippocampal cortex samples as compared to the HC. Dysregulated protein groups comprised those affecting mitochondrial function and calcium homeostasis. Differentially expressed mitochondrial proteins included proteins of the mitochondrial complexes I, III, IV, and V as well as of the accessory subunit of complex I. The analysis also revealed a regulation of the microtubule associated protein Tau in parahippocampal cortex tissue during the latency phase. This was further confirmed by immunohistochemistry. Moreover, we demonstrated a complex epileptogenesis-associated dysregulation of proteins involved in amyloid ß processing and its regulation. Among others, the amyloid precursor protein and the α-secretase alpha disintegrin metalloproteinase 17 were included. Our analysis revealed a relevant regulation of key proteins known to be associated with AD pathogenesis. The analysis provides a comprehensive overview of shared molecular alterations characterizing epilepsy development and manifestation as well as AD development and progression.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Epilepsy/metabolism , Hippocampus/metabolism , Mitochondrial Proteins/metabolism , Parahippocampal Gyrus/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Epilepsy/genetics , Female , Mitochondrial Proteins/genetics , Proteomics/methods , Rats , Rats, Sprague-Dawley
7.
J Alzheimers Dis ; 72(4): 1261-1268, 2019.
Article in English | MEDLINE | ID: mdl-31707367

ABSTRACT

BACKGROUND: Subjective memory complaints (SMC) are a risk factor for Alzheimer's disease. OBJECTIVE: We aimed to explore the association between SMC and regional amyloid-ß (Aß) deposition in mild cognitive impairment (MCI). METHODS: Sixty-eight individuals with MCI were recruited. [18F]Florbetaben PET scans were performed. T1-weighted 3D volumes were also acquired for co-registration with PET and for defining the regions of interest (ROI). Two step exploratory partial correlation analyses between SMC and Aß deposition were performed with covariates of age, sex, education, and depression. Furthermore, for the priori ROI that had the most significant partial correlation, we investigated the correlation between the SMC and regional Aß burden using a multiple linear regression model controlling for depression, age, sex, and education. RESULTS: Significant positive correlations between the SMC and Aß burden was found in the medial temporal ROI (first step) and in the left parahippocampus ROI (second step). In the priori left parahippocampus, we found significant correlation between the SMC and Aß burden (R2 = 0.473, p = 0.014). CONCLUSIONS: Our study suggested that the SMC was associated with amyloid accumulation, especially in the left parahippocampus, in individuals with MCI.


Subject(s)
Cognitive Dysfunction/complications , Memory Disorders/complications , Parahippocampal Gyrus/diagnostic imaging , Plaque, Amyloid/diagnostic imaging , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/metabolism , Female , Humans , Magnetic Resonance Imaging , Male , Memory , Memory Disorders/diagnostic imaging , Memory Disorders/metabolism , Neuropsychological Tests , Parahippocampal Gyrus/metabolism , Plaque, Amyloid/metabolism , Positron-Emission Tomography
8.
Epilepsy Res ; 157: 106189, 2019 11.
Article in English | MEDLINE | ID: mdl-31472401

ABSTRACT

PURPOSE: Type IIB focal cortical dysplasia (FCD) is an important cause of drug-resistant epilepsy. However, balloon cells located in the medial temporal lobe have been seldom reported. We aimed to discuss the clinical and pathological features of Type IIB FCD with balloon cells in the medial temporal lobe (MTLE-FCDIIB) and the differential diagnosis with other types of mesial temporal lobe epilepsy. METHODS: Three MTLE-FCDIIB cases were enrolled from Peking Union Medical College Hospital. Clinical and neuroimaging data were analyzed and histology features observed on hematoxylin-eosin (H&E) staining and immunochemical staining, including vimentin, nestin, S-100, CD34, neuronal nuclei antigen (Neun), glial fibrillary acidic protein (GFAP), neurofilament heavy chain (SMI32), were discussed. RESULTS: All cases involved drug-resistant epilepsy patients with childhood onset. The semiology of the epileptic seizure was a highly frequent partial seizure with or without generalized tonic-clonic seizures. Magnetic resonance imaging showed hyper-intensity in the medial temporal lobe without atrophy, different from mesial temporal sclerosis. Histological examination indicated the presence of balloon cells in the white matter of the para-hippocampal gyrus, subiculum, and cornu ammonis with cortical disorganization, and SMI32 positive dysmorphic neurons in the gray matter. Balloon cells were immunohistochemically stained with vimentin and nestin. Granular cell dispersion and pyramidal cell loss were not found. CONCLUSIONS: The presence of balloon cells in the medial temporal lobe is observed in a rare subgroup of FCD, named MTLE-FCDIIB. It has distinct clinical manifestations, neuroimaging features, pathological changes, and prognosis, which should be differentiated from mesial temporal lobe sclerosis and mesial temporal lobe tumors. Our findings enable more accurate diagnosis of mesial temporal lobe epilepsy.


Subject(s)
Epilepsy, Temporal Lobe/pathology , Epilepsy/pathology , Hippocampus/pathology , Malformations of Cortical Development, Group I/pathology , Parahippocampal Gyrus/pathology , Adolescent , Adult , Antigens, Nuclear/metabolism , Epilepsy/diagnostic imaging , Epilepsy/metabolism , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Humans , Magnetic Resonance Imaging , Malformations of Cortical Development, Group I/diagnostic imaging , Malformations of Cortical Development, Group I/metabolism , Nerve Tissue Proteins/metabolism , Nestin/metabolism , Neuroimaging , Neurons/metabolism , Neurons/pathology , Parahippocampal Gyrus/diagnostic imaging , Parahippocampal Gyrus/metabolism , Tomography, X-Ray Computed , Vimentin/metabolism
9.
Neuroscience ; 415: 44-58, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31319099

ABSTRACT

Temporal lobe epilepsy is triggered by an initial insult, such as status epilepticus, that initiates the process of epilepsy development. Heat shock protein 70 (Hsp70) is a ubiquitously expressed molecular chaperone, involved in the inflammatory response that is upregulated after status epilepticus. Hsp70 has been described as an endogenous intracellular ligand of Toll-like receptor 4. It is released from damaged or necrotic tissue and by activated immune cells after an inflammatory event. So far, the time course and the pattern of epileptogenesis-associated alterations in Hsp70 expression have not been described in detail. Thus, we investigated immunohistochemical expression of Hsp70 in hippocampus, parahippocampal cortex, parietal cortex, amygdala, and thalamus following status epilepticus in a rat model of temporal lobe epilepsy. The impact of status epilepticus on Hsp70 expression varied during different phases of epileptogenesis, displaying a stronger effect in the early post-insult phase, a milder and more localized effect in the latency phase and no relevant effect in the chronic phase. Cellular-level characterization revealed that Hsp70 colocalized with the neuronal marker NeuN and with Toll-like receptor 4. No colocalization with the astrocytic marker GFAP or the microglia marker Iba1 was found. The intense neuronal Hsp70 upregulation during the early post-insult phase might contribute to the onset of excessive inflammation triggering molecular and cellular reorganization and generation of a hyperexcitable epileptic network. Therefore, development of multi-targeting strategies aiming at prevention of epileptogenesis should consider Hsp70 modulation in the early days following an epileptogenic insult.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , HSP70 Heat-Shock Proteins/metabolism , Status Epilepticus/metabolism , Amygdala/metabolism , Animals , Astrocytes/metabolism , Female , Hippocampus/metabolism , Inflammation/metabolism , Microglia/metabolism , Neurons/metabolism , Parahippocampal Gyrus/metabolism , Parietal Lobe/metabolism , Rats , Rats, Sprague-Dawley , Thalamus/metabolism , Toll-Like Receptor 4/metabolism
10.
J Neuroimaging ; 29(5): 592-597, 2019 09.
Article in English | MEDLINE | ID: mdl-31273871

ABSTRACT

BACKGROUND AND PURPOSE: Previous magnetic resonance spectroscopy (MRS) studies have concluded that hippocampal and parahippocampal metabolite concentrations remain stable during healthy adult aging. However, these studies used short repetition times (TR ≤ 2 seconds), which lead to incomplete longitudinal magnetization recovery, and thus, heavily T1 -weighted measurements. It is important to accurately characterize brain metabolites changes with age to enable appropriate interpretations of MRS findings in the context of neurodegenerative diseases. Our goal was to assess hippocampal brain metabolite concentrations in a large cohort of diversely aged healthy volunteers using a longer TR of 4 seconds. METHODS: Left hippocampal MR spectra were collected from 38 healthy volunteers at 3T. Absolute metabolite concentrations were determined for total N-acetyl-aspartate (tNAA), total creatine (tCr), total choline (tCho), glutamate and glutamine (Glx), and myoinositol (mI). Individual partial correlations between each metabolite with age were assessed using demographic information and voxel compartmentation as confounders. RESULTS: Hippocampal tNAA, tCr, tCho, and mI all increased with age (NAA: R2 = .17, P = .041; tCr: R2 = .45, P = .0002; tCho: R2 = .37, P = .001; mI: R2 = .44, P = .0003). There were no relationships between age and signal to noise ratio, linewidth, or scan date, indicating the correlations were not confounded by spectral quality. Furthermore, we did not observe a trend with age in the voxel tissue compartmentations. CONCLUSIONS: We observed increases in hippocampal/parahippocampal metabolite concentrations with age, a finding that is in contrast to previous literature. Our findings illustrate the importance of using a sufficiently long TR in MRS to avoid T1 -relaxation effects influencing the measurement of absolute metabolite concentrations.


Subject(s)
Aging/metabolism , Hippocampus/metabolism , Parahippocampal Gyrus/metabolism , Proton Magnetic Resonance Spectroscopy/methods , Adolescent , Adult , Aspartic Acid/analogs & derivatives , Choline/metabolism , Creatine/metabolism , Female , Glutamic Acid/metabolism , Glutamine/metabolism , Hippocampus/diagnostic imaging , Humans , Inositol/metabolism , Male , Middle Aged , Parahippocampal Gyrus/diagnostic imaging , Protons , Young Adult
11.
Neuroimage Clin ; 23: 101905, 2019.
Article in English | MEDLINE | ID: mdl-31279240

ABSTRACT

OBJECTIVE: To evaluate, in vivo, the impact of ongoing chronic migraine (CM) attacks on the endogenous µ-opioid neurotransmission. BACKGROUND: CM is associated with cognitive-emotional dysfunction. CM is commonly associated with frequent acute medication use, including opioids. METHODS: We scanned 15 migraine patients during the spontaneous headache attack (ictal phase): 7 individuals with CM and 8 with episodic migraine (EM), as well as 7 healthy controls (HC), using positron emission tomography (PET) with the selective µ-opioid receptor (µOR) radiotracer [11C]carfentanil. Migraineurs were scanned in two paradigms, one with thermal pain threshold challenge applied to the site of the headache, and one without thermal challenge. Multivariable analysis was performed between the µ-opioid receptor availability and the clinical data. RESULTS: µOR availability, measured with [11C]carfentanil nondisplaceable binding potential (BPND), in the left thalamus (P-value = 0.005) and left caudate (P-value = 0.003) were decreased in CM patients with thermal pain threshold during the ictal phase relative to HC. Lower µOR BPND in the right parahippocampal region (P-value = 0.001) and right amygdala (P-value = 0.002) were seen in CM relative to EM patients. Lower µOR BPND values indicate either a decrease in µOR concentration or an increase in endogenous µ-opioid release in CM patients. In the right amygdala, 71% of the overall variance in µOR BPND levels was explained by the type of migraine (CM vs. EM: partial-R2 = 0.47, P-value<0.001, Cohen's effect size d = 2.6SD), the severity of the attack (pain area and intensity number summation [P.A.I.N.S.]: partial-R2 = 0.16, P-value = 0.031), and the thermal pain threshold (allodynia: partial-R2 = 0.08). CONCLUSIONS: Increased endogenous µ-opioid receptor-mediated neurotransmission is seen in the limbic system of CM patients, especially in right amygdala, which is highly modulated by the attack frequency, pain severity, and sensitivity. This study demonstrates for the first time the negative impact of chronification and exacerbation of headache attacks on the endogenous µ-opioid mechanisms of migraine patients. ClinicalTrials.gov identifier: NCT03004313.


Subject(s)
Amygdala/metabolism , Migraine Disorders/metabolism , Migraine Disorders/physiopathology , Nociception/physiology , Pain Threshold/physiology , Parahippocampal Gyrus/metabolism , Receptors, Opioid, mu/metabolism , Adult , Amygdala/diagnostic imaging , Analgesics, Opioid/pharmacokinetics , Caudate Nucleus/diagnostic imaging , Caudate Nucleus/metabolism , Chronic Disease , Female , Fentanyl/analogs & derivatives , Fentanyl/pharmacokinetics , Humans , Male , Middle Aged , Migraine Disorders/diagnostic imaging , Parahippocampal Gyrus/diagnostic imaging , Physical Stimulation , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Severity of Illness Index , Thalamus/diagnostic imaging , Thalamus/metabolism , Young Adult
12.
Neuroimage Clin ; 22: 101765, 2019.
Article in English | MEDLINE | ID: mdl-30904824

ABSTRACT

BACKGROUND: Although previous studies have suggested that insulin plays a role in brain function, it still remains unclear whether or not insulin has a region-specific association with neuronal and synaptic activity in the living human brain. We investigated the regional pattern of association between basal blood insulin and resting-state cerebral glucose metabolism (CMglu), a proxy for neuronal and synaptic activity, in older adults. METHOD: A total of 234 nondiabetic, cognitively normal (CN) older adults underwent comprehensive clinical assessment, resting-state 18F-fluodeoxyglucose (FDG)-positron emission tomography (PET) and blood sampling to determine overnight fasting blood insulin and glucose levels, as well as apolipoprotein E (APOE) genotyping. RESULTS: An exploratory voxel-wise analysis of FDG-PET without a priori hypothesis demonstrated a positive association between basal blood insulin levels and resting-state CMglu in specific cerebral cortices and hippocampus, rather than in non-specific overall cerebral regions, even after controlling for the effects of APOE e4 carrier status, vascular risk factor score, body mass index, fasting blood glucose, and demographic variables. Particularly, a positive association of basal blood insulin with CMglu in the right posterior hippocampus and adjacent parahippocampal region as well as in the right inferior parietal region remained significant after multiple comparison correction. Conversely, no region showed negative association between basal blood insulin and CMglu. CONCLUSIONS: Our finding suggests that basal fasting blood insulin may have association with neuronal and synaptic activity in specific cerebral regions, particularly in the hippocampal/parahippocampal and inferior parietal regions.


Subject(s)
Aging/metabolism , Cerebral Cortex/metabolism , Glucose/metabolism , Insulins/blood , Aged , Aging/blood , Cerebral Cortex/diagnostic imaging , Female , Fluorodeoxyglucose F18 , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Humans , Male , Middle Aged , Parahippocampal Gyrus/diagnostic imaging , Parahippocampal Gyrus/metabolism , Parietal Lobe/metabolism , Positron-Emission Tomography
13.
PLoS One ; 13(11): e0206547, 2018.
Article in English | MEDLINE | ID: mdl-30383799

ABSTRACT

A large body of literature links risk of cognitive decline, mild cognitive impairment (MCI) and dementia with Type 2 Diabetes (T2D) or pre-diabetes. Accumulating evidence implicates a close relationship between the brain insulin receptor signaling pathway (IRSP) and the accumulation of amyloid beta and hyperphosphorylated and conformationally abnormal tau. We showed previously that the neuropathological features of Alzheimer's disease (AD were reduced in patients with diabetes who were treated with insulin and oral antidiabetic medications. To understand better the neurobiological substrates of T2D and T2D medications in AD, we examined IRSP and endothelial cell markers in the parahippocampal gyrus of controls (N = 30), of persons with AD (N = 19), and of persons with AD and T2D, who, in turn, had been treated with anti-diabetic drugs (insulin and or oral agents; N = 34). We studied the gene expression of selected members of the IRSP and selective endothelial cell markers in bulk postmortem tissue from the parahippocampal gyrus and in endothelial cell enriched isolates from the same brain region. The results indicated that there are considerable abnormalities and reductions in gene expression (bulk tissue homogenates and endothelial cell isolates) in the parahippocampal gyri of persons with AD that map directly to genes associated with the microvasculature and the IRSP. Our results also showed that the numbers of abnormally expressed microvasculature and IRSP associated genes in diabetic AD donors who had been treated with anti-diabetic agents were reduced significantly. These findings suggest that anti-diabetic treatments may reduce or normalize compromised microvascular and IRSP functions in AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Endothelial Cells/metabolism , Hypoglycemic Agents/therapeutic use , Parahippocampal Gyrus/drug effects , Parahippocampal Gyrus/metabolism , Aged, 80 and over , Cohort Studies , Endothelial Cells/drug effects , Female , Gene Expression/drug effects , Humans , Male , Microvessels/drug effects , Microvessels/metabolism , RNA, Messenger/metabolism , Receptor, Insulin
14.
Acta Neuropathol Commun ; 6(1): 62, 2018 07 20.
Article in English | MEDLINE | ID: mdl-30029687

ABSTRACT

In the majority of affected brain regions the pathological hallmarks of Alzheimer's disease (AD) are ß-amyloid (Aß) deposits in the form of diffuse and neuritic plaques, tau pathology in the form of neurofibrillary tangles, neuropil threads and plaque-associated abnormal neurites in combination with an inflammatory response. However, the anatomical area of the presubiculum, is characterised by the presence of a single large evenly distributed 'lake-like' Aß deposit with minimal tau deposition or accumulation of inflammatory markers. Post-mortem brain samples from sporadic AD (SAD) and familial AD (FAD) and two hereditary cerebral amyloid diseases, familial British dementia (FBD) and familial Danish dementia (FDD) were used to compare the morphology of the extracellular proteins deposited in the presubiculum compared to the entorhinal cortex. The level of tau pathology and the extent of microglial activation were quantitated in the two brain regions in SAD and FAD. Frozen tissue was used to investigate the Aß species and proteomic differences between the two regions. Consistent with our previous investigations of FBD and FDD cases we were able to establish that the 'lake-like' pre-amyloid deposits of the presubiculum were not a unique feature of AD but they also found two non-Aß amyloidosis. Comparing the presubiculum to the entorhinal cortex the number of neurofibrillary tangles and tau load were significantly reduced; there was a reduction in microglial activation; there were differences in the Aß profiles and the investigation of the whole proteome showed significant changes in different protein pathways. In summary, understanding why the presubiculum has a different morphological appearance, biochemical and proteomic makeup compared to surrounding brain regions severely affected by neurodegeneration could lead us to understanding protective mechanisms in neurodegenerative diseases.


Subject(s)
Alzheimer Disease/complications , Entorhinal Cortex/metabolism , Neurodegenerative Diseases/etiology , Parahippocampal Gyrus/metabolism , Aged , Aged, 80 and over , Annexin A1/metabolism , Computational Biology , Cytokines/metabolism , Female , Humans , Laser Capture Microdissection , Macrophage-1 Antigen/metabolism , Male , Mass Spectrometry , Middle Aged , Neurofibrillary Tangles/pathology
15.
Sci Rep ; 8(1): 7194, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29740121

ABSTRACT

Pre-clinical research in rodents provides evidence that the central nervous system (CNS) has functional lymphatic vessels. In-vivo observations in humans, however, are not demonstrated. We here show data on CNS lymphatic drainage to cervical lymph nodes in-vivo by magnetic resonance imaging (MRI) enhanced with an intrathecal contrast agent as a cerebrospinal fluid (CSF) tracer. Standardized MRI of the intracranial compartment and the neck were acquired before and up to 24-48 hours following intrathecal contrast agent administration in 19 individuals. Contrast enhancement was radiologically confirmed by signal changes in CSF nearby inferior frontal gyrus, brain parenchyma of inferior frontal gyrus, parahippocampal gyrus, thalamus and pons, and parenchyma of cervical lymph node, and with sagittal sinus and neck muscle serving as reference tissue for cranial and neck MRI acquisitions, respectively. Time series of changes in signal intensity shows that contrast enhancement within CSF precedes glymphatic enhancement and peaks at 4-6 hours following intrathecal injection. Cervical lymph node enhancement coincides in time with peak glymphatic enhancement, with peak after 24 hours. Our findings provide in-vivo evidence of CSF tracer drainage to cervical lymph nodes in humans. The time course of lymph node enhancement coincided with brain glymphatic enhancement rather than with CSF enhancement.


Subject(s)
Arachnoid Cysts/diagnostic imaging , Glymphatic System/diagnostic imaging , Hydrocephalus/diagnostic imaging , Intracranial Hypertension/diagnostic imaging , Intracranial Hypotension/diagnostic imaging , Lymphatic System/diagnostic imaging , Adult , Aged , Arachnoid Cysts/cerebrospinal fluid , Arachnoid Cysts/physiopathology , Cohort Studies , Contrast Media/administration & dosage , Female , Glymphatic System/metabolism , Glymphatic System/physiopathology , Humans , Hydrocephalus/cerebrospinal fluid , Hydrocephalus/physiopathology , Injections, Spinal , Intracranial Hypertension/cerebrospinal fluid , Intracranial Hypertension/physiopathology , Intracranial Hypotension/cerebrospinal fluid , Intracranial Hypotension/physiopathology , Lymph Nodes/diagnostic imaging , Lymph Nodes/metabolism , Lymph Nodes/physiopathology , Lymphatic System/metabolism , Lymphatic System/physiopathology , Lymphatic Vessels/diagnostic imaging , Lymphatic Vessels/metabolism , Lymphatic Vessels/physiopathology , Lymphography , Magnetic Resonance Imaging , Male , Middle Aged , Organometallic Compounds/administration & dosage , Parahippocampal Gyrus/diagnostic imaging , Parahippocampal Gyrus/metabolism , Parahippocampal Gyrus/physiopathology , Parenchymal Tissue/diagnostic imaging , Parenchymal Tissue/metabolism , Parenchymal Tissue/physiopathology , Pons/diagnostic imaging , Pons/metabolism , Pons/physiopathology , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Thalamus/diagnostic imaging , Thalamus/metabolism , Thalamus/physiopathology
16.
Brain Pathol ; 28(1): 58-71, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27997051

ABSTRACT

Adipocyte enhancer binding protein 1 (AEBP1) activates inflammatory responses via the NF-κB pathway in macrophages and regulates adipogenesis in preadipocytes. Up-regulation of AEBP1 in the hippocampi of patients with Alzheimer's disease (AD) has been revealed by microarray analyses of autopsied brains from the Japanese general population (the Hisayama study). In this study, we compared the expression patterns of AEBP1 in normal and AD brains, including in the hippocampus, using immunohistochemistry. The subjects were 24 AD cases and 52 non-AD cases. Brain specimens were immunostained with antibodies against AEBP1, tau protein, amyloid ß protein, NF-κB, GFAP and Iba-1. In normal brains, AEBP1 immunoreactivity mainly localized to the perikarya of hippocampal pyramidal neurons, and its expression was elevated in the pyramidal neurons and some astrocytes in AD hippocampi. Although AEBP1 immunoreactivity was almost absent in neurons containing neurofibrillary tangles, AEBP1 was highly expressed in neurons with pretangles and in the tau-immunopositive, dystrophic neurites of senile plaques. Nuclear localization of NF-κB was also observed in certain AEBP1-positive neurons in AD cases. Comparison of AD and non-AD cases suggested a positive correlation between the expression level of AEBP1 and the degree of amyloid ß pathology. These findings imply that AEBP1 protein has a role in the progression of AD pathology.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Carboxypeptidases/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Repressor Proteins/metabolism , Aged, 80 and over , Astrocytes/metabolism , Astrocytes/pathology , Blotting, Western , Disease Progression , Female , Humans , Immunohistochemistry , Male , Middle Aged , NF-kappa B p50 Subunit/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Parahippocampal Gyrus/metabolism , Parahippocampal Gyrus/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , RNA, Messenger/metabolism
17.
J Alzheimers Dis ; 60(4): 1285-1294, 2017.
Article in English | MEDLINE | ID: mdl-29036815

ABSTRACT

Hippocampal atrophy and hypometabolism of the posterior cingulate cortex (PCC), early markers of Alzheimer's disease (AD), have been shown to be associated in late mild cognitive impairment and early AD via atrophy of connecting cingulum fibers. Recently, a direct association of hippocampal atrophy and PCC hypometabolism has been shown in cognitively normal elderly. We aimed to investigate if this association might be modulated by partly non-hippocampogenic alterations of parahippocampal cingulum (PHC) integrity. 45 cognitively healthy elderly aged 59 to 89 years were included from the Alzheimer's Disease Neuroimaging Initiative. Hippocampal volumes and PCC glucose metabolism were measured using MRI and FDG-PET. PHC fibers connecting the hippocampus and the PCC were reconstructed using diffusion weighted MRI and measures of diffusivity were calculated. Using robust linear regression, interaction effects of PHC diffusivity and hippocampal volume on PCC metabolism were calculated. For both hemispheres, significant interaction effects were found for PHC mean diffusivity. Interaction effects were such that the association of hippocampal volume and PCC metabolism was higher in subjects with increased mean diffusivity in PHC fibers. In cognitively normal elderly, compromised integrity of the PHC may increase the risk of PCC hypometabolism due to hippocampal atrophy. Spared PHC fiber integrity may protect against PCC hypometabolism due to hippocampal atrophy.


Subject(s)
Aging/metabolism , Aging/pathology , Gyrus Cinguli/metabolism , Hippocampus/pathology , Parahippocampal Gyrus/pathology , Aged , Aged, 80 and over , Atrophy , Brain Mapping , Female , Fluorodeoxyglucose F18 , Glucose/metabolism , Gray Matter/diagnostic imaging , Gray Matter/metabolism , Gray Matter/pathology , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/pathology , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Organ Size , Parahippocampal Gyrus/diagnostic imaging , Parahippocampal Gyrus/metabolism , Positron-Emission Tomography , Radiopharmaceuticals , Regression Analysis
18.
Sci Rep ; 7: 45496, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28374768

ABSTRACT

Hyperphosphorylated tau protein deposits and, inflammatory processes are characteristic components of Alzheimer disease (AD) pathology. We here aimed to visualize in vitro the distribution of tau deposits and activated astrocytes across the cortical layers in autopsy AD brain tissue using the radiotracers 3H-THK5117 and 3H-deprenyl. 3H-THK5117 and 3H-deprenyl autoradiographies were carried out on frozen brain sections from three AD patients and one healthy control. 3H-THK5117 showed a distinct laminar cortical binding similar to 3H-deprenyl autoradiography, with an extensive binding in the superficial and deep layers of the temporal neocortices, whereas the middle frontal gyrus showed an even binding throughout the layers. Globally, eventhough some differences could be observed, AT8 (tau) and GFAP (astrocyte) immunostaining showed a laminar pattern comparable to their corresponding radiotracers within each AD case. Some variability was observed between the AD cases reflecting differences in disease phenotype. The similar laminar cortical brain distribution of tau deposits and activated astrocytes supports the hypothesis of a close pathological interconnection. The difference in regional binding patterns of 3H-THK5117 and AT8 antibody staining suggest additional tau binding sites detectable by 3H-THK5117.


Subject(s)
Alzheimer Disease/pathology , Autoradiography , Propanols/chemistry , Quinolines/chemistry , Selegiline/chemistry , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Astrocytes/cytology , Astrocytes/metabolism , Entorhinal Cortex/metabolism , Entorhinal Cortex/pathology , Female , Humans , Male , Microscopy, Fluorescence , Middle Aged , Parahippocampal Gyrus/metabolism , Parahippocampal Gyrus/pathology , Tritium/chemistry , tau Proteins/chemistry
19.
J Chem Neuroanat ; 77: 187-192, 2016 11.
Article in English | MEDLINE | ID: mdl-27435807

ABSTRACT

The muscarinic M1 receptor plays a significant role in cognition, probably by modulating information processing in key regions such as the hippocampus. To understand how the muscarinic M1 receptor achieves these functions in the hippocampus, it is critical to know the distribution of the receptor within this complex brain region. To date, there are limited data on the distribution of muscarinic M1 receptors in the human hippocampus which may also be confounded because some anti-muscarinic receptor antibodies have been shown to lack specificity. Initially, using Western blotting and immunohistochemistry, we showed the anti-muscarinic M1 receptor antibody to be used in our study bound to a single 62kDa protein that was absent in mice lacking the muscarinic M1 receptor gene. Then, using immunohistochemistry, we determined the distribution of muscarinic M1 receptors in human hippocampus from 10 subjects with no discernible history of a neurological or psychiatric disorder. Our data shows the muscarinic M1 receptor to be predominantly on pyramidal cells in the hippocampus. Muscarinic M1 receptor positive cells were most apparent in the deep polymorphic layer of the dentate gyrus, the pyramidal cell layer of cornu ammonis region 3, the cellular layers of the subiculum, layer II of the presubiculum and layer III and V of the parahippocampal gyrus. Positive cells were less numerous and less intensely stained in the pyramidal layer of cornu ammonis region 2 and were sparse in the molecular layer of the dentate gyrus as well as cornu ammonis region 1. Although immunoreactivity was present in the granular layer of the dentate gyrus, it was difficult to identity individual immunopositive cells, possibly due to the density of cells. This distribution of the muscarinic M1 receptors in human hippocampus, and its localisation on glutamatergic cells, would suggest the receptor has a significant role in modulating excitatory hippocampal neurotransmission.


Subject(s)
Hippocampus/metabolism , Receptor, Muscarinic M1/metabolism , Adult , Aged , Animals , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Female , Hippocampus/anatomy & histology , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Middle Aged , Parahippocampal Gyrus/cytology , Parahippocampal Gyrus/metabolism , Pyramidal Cells/metabolism , Reference Values , Synaptic Transmission , Young Adult
20.
Brain Behav Immun ; 53: 138-158, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26685804

ABSTRACT

Detailed knowledge about the patterns of molecular alterations during epileptogenesis is a presupposition for identifying targets for preventive or disease-modifying approaches, as well as biomarkers of the disease. Large-scale differential proteome analysis can provide unique and novel perspectives based on comprehensive data sets informing about the complex regulation patterns in the disease proteome. Thus, we have completed an elaborate differential proteome analysis based on label-free LC-MS/MS in a rat model of epileptogenesis. Hippocampus and parahippocampal cortex tissues were sampled and analyzed separately at three key time points chosen for monitoring disease development following electrically-induced status epilepticus, namely, the early post-insult phase, the latency phase, and the chronic phase with spontaneous recurrent seizures. We focused the bioinformatics analysis on proteins linked to immune and inflammatory responses, because of the emerging evidence of the specific pathogenic role of inflammatory signalings during epileptogenesis. In the early post-insult and the latency phases, pathway enrichment analysis revealed an extensive over-representation of Toll-like receptor signaling, pro-inflammatory cytokines, heat shock protein regulation, and transforming growth factor beta signaling and leukocyte transendothelial migration. The inflammatory response in the chronic phase proved to be more moderate with differential expression in the parahippocampal cortex exceeding that in the hippocampus. The data sets provide novel information about numerous differentially expressed proteins, which serve as interaction partners or modulators in key disease-associated inflammatory signaling events. Noteworthy, a set of proteins which act as modulators of the ictogenic Toll-like receptor signaling proved to be differentially expressed. In addition, we report novel data demonstrating the regulation of different Toll-like receptor ligands during epileptogenesis. Taken together, the findings deepen our understanding of modulation of inflammatory signaling during epileptogenesis providing an excellent and comprehensive basis for the identification of target and biomarker candidates.


Subject(s)
Epilepsy/metabolism , Inflammation/metabolism , Animals , Biomarkers/metabolism , Cerebral Cortex/metabolism , Cytokines/metabolism , Disease Models, Animal , Epilepsy/etiology , Epilepsy/genetics , Female , Gene Expression Profiling , Hippocampus/metabolism , Inflammation/genetics , Parahippocampal Gyrus/metabolism , Proteome/metabolism , Proteomics/methods , Rats , Rats, Sprague-Dawley , Receptors, Purinergic/metabolism , Signal Transduction , Tandem Mass Spectrometry/methods , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...