Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Publication year range
1.
Traffic ; 25(5): e12937, 2024 May.
Article in English | MEDLINE | ID: mdl-38777335

ABSTRACT

The polymorphic APOE gene is the greatest genetic determinant of sporadic Alzheimer's disease risk: the APOE4 allele increases risk, while the APOE2 allele is neuroprotective compared with the risk-neutral APOE3 allele. The neuronal endosomal system is inherently vulnerable during aging, and APOE4 exacerbates this vulnerability by driving an enlargement of early endosomes and reducing exosome release in the brain of humans and mice. We hypothesized that the protective effects of APOE2 are, in part, mediated through the endosomal pathway. Messenger RNA analyses showed that APOE2 leads to an enrichment of endosomal pathways in the brain when compared with both APOE3 and APOE4. Moreover, we show age-dependent alterations in the recruitment of key endosomal regulatory proteins to vesicle compartments when comparing APOE2 to APOE3. In contrast to the early endosome enlargement previously shown in Alzheimer's disease and APOE4 models, we detected similar morphology and abundance of early endosomes and retromer-associated vesicles within cortical neurons of aged APOE2 targeted-replacement mice compared with APOE3. Additionally, we observed increased brain extracellular levels of endosome-derived exosomes in APOE2 compared with APOE3 mice during aging, consistent with enhanced endosomal cargo clearance by exosomes to the extracellular space. Our findings thus demonstrate that APOE2 enhances an endosomal clearance pathway, which has been shown to be impaired by APOE4 and which may be protective due to APOE2 expression during brain aging.


Subject(s)
Aging , Apolipoprotein E2 , Brain , Endosomes , Exosomes , Animals , Humans , Mice , Aging/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Apolipoprotein E2/metabolism , Apolipoprotein E2/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E3/genetics , Apolipoprotein E4/metabolism , Apolipoprotein E4/genetics , Brain/metabolism , Endosomes/metabolism , Exosomes/metabolism , Mice, Inbred C57BL , Neurons/metabolism
2.
Brain ; 142(1): 163-175, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30496349

ABSTRACT

In addition to being the greatest genetic risk factor for Alzheimer's disease, expression of the ɛ4 allele of apolipoprotein E can lead to cognitive decline during ageing that is independent of Alzheimer's amyloid-ß and tau pathology. In human post-mortem tissue and mouse models humanized for apolipoprotein E, we examined the impact of apolipoprotein E4 expression on brain exosomes, vesicles that are produced within and secreted from late-endocytic multivesicular bodies. Compared to humans or mice homozygous for the risk-neutral ɛ3 allele we show that the ɛ4 allele, whether homozygous or heterozygous with an ɛ3 allele, drives lower exosome levels in the brain extracellular space. In mice, we show that the apolipoprotein E4-driven change in brain exosome levels is age-dependent: while not present at age 6 months, it is detectable at 12 months of age. Expression levels of the exosome pathway regulators tumor susceptibility gene 101 (TSG101) and Ras-related protein Rab35 (RAB35) were found to be reduced in the brain at the protein and mRNA levels, arguing that apolipoprotein E4 genotype leads to a downregulation of exosome biosynthesis and release. Compromised exosome production is likely to have adverse effects, including diminishing a cell's ability to eliminate materials from the endosomal-lysosomal system. This reduction in brain exosome levels in 12-month-old apolipoprotein E4 mice occurs earlier than our previously reported brain endosomal pathway changes, arguing that an apolipoprotein E4-driven failure in exosome production plays a primary role in endosomal and lysosomal deficits that occur in apolipoprotein E4 mouse and human brains. Disruption of these interdependent endosomal-exosomal-lysosomal systems in apolipoprotein E4-expressing individuals may contribute to amyloidogenic amyloid-ß precursor protein processing, compromise trophic signalling and synaptic function, and interfere with a neuron's ability to degrade material, all of which are events that lead to neuronal vulnerability and higher risk of Alzheimer's disease development. Together, these data suggest that exosome pathway dysfunction is a previously unappreciated component of the brain pathologies that occur as a result of apolipoprotein E4 expression.


Subject(s)
Apolipoprotein E4/biosynthesis , Brain/metabolism , Exosomes/metabolism , Aged , Aged, 80 and over , Aging/metabolism , Alleles , Animals , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , DNA-Binding Proteins/biosynthesis , Down-Regulation , Endosomal Sorting Complexes Required for Transport/biosynthesis , Exosomes/ultrastructure , Extracellular Space/metabolism , Female , Genotype , Humans , Lipid Metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Transcription Factors/biosynthesis , rab GTP-Binding Proteins/biosynthesis
3.
Mol Cell Neurosci ; 99: 103395, 2019 09.
Article in English | MEDLINE | ID: mdl-31422108

ABSTRACT

BACE1 is a transmembrane aspartic protease that cleaves various substrates and it is required for normal brain function. BACE1 expression is high during early development, but it is reduced in adulthood. Under conditions of stress and injury, BACE1 levels are increased; however, the underlying mechanisms that drive BACE1 elevation are not well understood. One mechanism associated with brain injury is the activation of injurious p75 neurotrophin receptor (p75), which can trigger pathological signals. Here we report that within 72 h after controlled cortical impact (CCI) or laser injury, BACE1 and p75 are increased and tightly co-expressed in cortical neurons of mouse brain. Additionally, BACE1 is not up-regulated in p75 null mice in response to focal cortical injury, while p75 over-expression results in BACE1 augmentation in HEK-293 and SY5Y cell lines. A luciferase assay conducted in SY5Y cell line revealed that BACE1 expression is regulated at the transcriptional level in response to p75 transfection. Interestingly, this effect does not appear to be dependent upon p75 ligands including mature and pro-neurotrophins. In addition, BACE1 activity on amyloid precursor protein (APP) is enhanced in SY5Y-APP cells transfected with a p75 construct. Lastly, we found that the activation of c-jun n-terminal kinase (JNK) by p75 contributes to BACE1 up-regulation. This study explores how two injury-induced molecules are intimately connected and suggests a potential link between p75 signaling and the expression of BACE1 after brain injury.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Brain Injuries, Traumatic/metabolism , Receptor, Nerve Growth Factor/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/metabolism , HEK293 Cells , Humans , MAP Kinase Kinase 4/metabolism , Male , Mice , Receptor, Nerve Growth Factor/genetics , Signal Transduction , Up-Regulation
4.
Neuroimage ; 173: 370-383, 2018 06.
Article in English | MEDLINE | ID: mdl-29496611

ABSTRACT

Complex animal behavior is produced by dynamic interactions between discrete regions of the brain. As such, defining functional connections between brain regions is critical in gaining a full understanding of how the brain generates behavior. Evidence suggests that discrete regions of the cerebellar cortex functionally project to the forebrain, mediating long-range communication potentially important in motor and non-motor behaviors. However, the connectivity map remains largely incomplete owing to the challenge of driving both reliable and selective output from the cerebellar cortex, as well as the need for methods to detect region specific activation across the entire forebrain. Here we utilize a paired optogenetic and fMRI (ofMRI) approach to elucidate the downstream forebrain regions modulated by activating a region of the cerebellum that induces stereotypical, ipsilateral forelimb movements. We demonstrate with ofMRI, that activating this forelimb motor region of the cerebellar cortex results in functional activation of a variety of forebrain and midbrain areas of the brain, including the hippocampus and primary motor, retrosplenial and anterior cingulate cortices. We further validate these findings using optogenetic stimulation paired with multi-electrode array recordings and post-hoc staining for molecular markers of activated neurons (i.e. c-Fos). Together, these findings demonstrate that a single discrete region of the cerebellar cortex is capable of influencing motor output and the activity of a number of downstream forebrain as well as midbrain regions thought to be involved in different aspects of behavior.


Subject(s)
Cerebellar Cortex/anatomy & histology , Magnetic Resonance Imaging/methods , Neural Pathways/anatomy & histology , Optogenetics/methods , Prosencephalon/anatomy & histology , Animals , Brain Mapping/methods , Image Processing, Computer-Assisted/methods , Mice , Movement/physiology
5.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 208-225, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28711595

ABSTRACT

Extensive parenchymal and vascular Aß deposits are pathological hallmarks of Alzheimer's disease (AD). Besides classic full-length peptides, biochemical analyses of brain deposits have revealed high degree of Aß heterogeneity likely resulting from the action of multiple proteolytic enzymes. In spite of the numerous studies focusing in Aß, the relevance of N- and C-terminal truncated species for AD pathogenesis remains largely understudied. In the present work, using novel antibodies specifically recognizing Aß species N-terminally truncated at position 4 or C-terminally truncated at position 34, we provide a clear assessment of the differential topographic localization of these species in AD brains and transgenic models. Based on their distinct solubility, brain N- and C-terminal truncated species were extracted by differential fractionation and identified via immunoprecipitation coupled to mass spectrometry analysis. Biochemical/biophysical studies with synthetic homologues further confirmed the different solubility properties and contrasting fibrillogenic characteristics of the truncated species composing the brain Aß peptidome. Aß C-terminal degradation leads to the production of more soluble fragments likely to be more easily eliminated from the brain. On the contrary, N-terminal truncation at position 4 favors the formation of poorly soluble, aggregation prone peptides with high amyloidogenic propensity and the potential to exacerbate the fibrillar deposits, self-perpetuating the amyloidogenic loop. Detailed assessment of the molecular diversity of Aß species composing interstitial fluid and amyloid deposits at different disease stages, as well as the evaluation of the truncation profile during various pharmacologic approaches will provide a comprehensive understanding of the still undefined contribution of Aß truncations to the disease pathogenesis and their potential as novel therapeutic targets.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Aged , Aged, 80 and over , Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/pathology , Case-Control Studies , Female , Humans , Male , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Rabbits
6.
J Neurosci ; 36(15): 4248-58, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-27076423

ABSTRACT

Epidemiological findings suggest that diabetic individuals are at a greater risk for developing Alzheimer's disease (AD). To examine the mechanisms by which diabetes mellitus (DM) may contribute to AD pathology in humans, we examined brain tissue from streptozotocin-treated type 1 diabetic adult male vervet monkeys receiving twice-daily exogenous insulin injections for 8-20 weeks. We found greater inhibitory phosphorylation of insulin receptor substrate 1 in each brain region examined of the diabetic monkeys when compared with controls, consistent with a pattern of brain insulin resistance that is similar to that reported in the human AD brain. Additionally, a widespread increase in phosphorylated tau was seen, including brain areas vulnerable in AD, as well as relatively spared structures, such as the cerebellum. An increase in active ERK1/2 was also detected, consistent with DM leading to changes in tau-kinase activity broadly within the brain. In contrast to these widespread changes, we found an increase in soluble amyloid-ß (Aß) levels that was restricted to the temporal lobe, with the greatest increase seen in the hippocampus. Consistent with this localized Aß increase, a hippocampus-restricted decrease in the protein and mRNA for the Aß-degrading enzyme neprilysin (NEP) was found, whereas various Aß-clearing and -degrading proteins were unchanged. Thus, we document multiple biochemical changes in the insulin-controlled DM monkey brain that can link DM with the risk of developing AD, including dysregulation of the insulin-signaling pathway, changes in tau phosphorylation, and a decrease in NEP expression in the hippocampus that is coupled with a localized increase in Aß. SIGNIFICANCE STATEMENT: Given that diabetes mellitus (DM) appears to increase the risk of developing Alzheimer's disease (AD), understanding the mechanisms by which DM promotes AD is important. We report that DM in a nonhuman primate brain leads to changes in the levels or posttranslational processing of proteins central to AD pathobiology, including tau, amyloid-ß (Aß), and the Aß-degrading protease neprilysin. Additional evidence from this model suggests that alterations in brain insulin signaling occurred that are reminiscent of insulin signaling pathway changes seen in human AD. Thus, in an in vivo model highly relevant to humans, we show multiple alterations in the brain resulting from DM that are mechanistically linked to AD risk.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain Chemistry , Diabetes Mellitus, Type 1/metabolism , Hippocampus/metabolism , Insulin Resistance , Neprilysin/metabolism , tau Proteins/metabolism , Animals , Chlorocebus aethiops , Diabetes Mellitus, Experimental/metabolism , Liver/metabolism , Male , Phosphorylation , Signal Transduction
7.
Pharm Res ; 34(1): 1-6, 2017 01.
Article in English | MEDLINE | ID: mdl-27620174

ABSTRACT

How do we inspire new ideas that could lead to potential treatments for rare or neglected diseases, and allow for serendipity that could help to catalyze them? How many potentially good ideas are lost because they are never tested? What if those ideas could have lead to new therapeutic approaches and major healthcare advances? If a clinician or anyone for that matter, has a new idea they want to test to develop a molecule or therapeutic that they could translate to the clinic, how would they do it without a laboratory or funding? These are not idle theoretical questions but addressing them could have potentially huge economic implications for nations. If we fail to capture the diversity of ideas and test them we may also lose out on the next blockbuster treatments. Many of those involved in the process of ideation may be discouraged and simply not know where to go. We try to address these questions and describe how there are options to raising funding, how even small scale investments can foster preclinical or clinical translation, and how there are several approaches to outsourcing the experiments, whether to collaborators or commercial enterprises. While these are not new or far from complete solutions, they are first steps that can be taken by virtually anyone while we work on other solutions to build a more concrete structure for the "idea-hypothesis testing-proof of concept-translation-breakthrough pathway".


Subject(s)
Drug Discovery , Neglected Diseases/drug therapy , Neglected Diseases/therapy , Animals , Cooperative Behavior , Drug Industry/methods , Humans , Laboratories , Research , Therapeutics/methods
8.
Nurs N Z ; 23(3): 41, 2017 Apr.
Article in English | MEDLINE | ID: mdl-30549576

ABSTRACT

The transition from nursing manager to NZNO lead organiser has proved to be just another way of nursing.


Subject(s)
Labor Unions , Leadership , Nurse Administrators , Societies, Nursing , Humans , New Zealand , Workplace
9.
J Biol Chem ; 288(21): 15352-66, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23589300

ABSTRACT

γ-Secretase plays a pivotal role in the production of neurotoxic amyloid ß-peptides (Aß) in Alzheimer disease (AD) and consists of a heterotetrameric core complex that includes the aspartyl intramembrane protease presenilin (PS). The human genome codes for two presenilin paralogs. To understand the causes for distinct phenotypes of PS paralog-deficient mice and elucidate whether PS mutations associated with early-onset AD affect the molecular environment of mature γ-secretase complexes, quantitative interactome comparisons were undertaken. Brains of mice engineered to express wild-type or mutant PS1, or HEK293 cells stably expressing PS paralogs with N-terminal tandem-affinity purification tags served as biological source materials. The analyses revealed novel interactions of the γ-secretase core complex with a molecular machinery that targets and fuses synaptic vesicles to cellular membranes and with the H(+)-transporting lysosomal ATPase macrocomplex but uncovered no differences in the interactomes of wild-type and mutant PS1. The catenin/cadherin network was almost exclusively found associated with PS1. Another intramembrane protease, signal peptide peptidase, predominantly co-purified with PS2-containing γ-secretase complexes and was observed to influence Aß production.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/immunology , Membrane Proteins/metabolism , Presenilin-2/metabolism , Serine Endopeptidases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Catenins/genetics , Catenins/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Transgenic , Mutation , Presenilin-2/genetics , Protein Binding/genetics , Serine Endopeptidases/genetics , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
10.
Elife ; 122024 Feb 12.
Article in English | MEDLINE | ID: mdl-38345922

ABSTRACT

The cerebellum has been implicated in the regulation of social behavior. Its influence is thought to arise from communication, via the thalamus, to forebrain regions integral in the expression of social interactions, including the anterior cingulate cortex (ACC). However, the signals encoded or the nature of the communication between the cerebellum and these brain regions is poorly understood. Here, we describe an approach that overcomes technical challenges in exploring the coordination of distant brain regions at high temporal and spatial resolution during social behavior. We developed the E-Scope, an electrophysiology-integrated miniature microscope, to synchronously measure extracellular electrical activity in the cerebellum along with calcium imaging of the ACC. This single coaxial cable device combined these data streams to provide a powerful tool to monitor the activity of distant brain regions in freely behaving animals. During social behavior, we recorded the spike timing of multiple single units in cerebellar right Crus I (RCrus I) Purkinje cells (PCs) or dentate nucleus (DN) neurons while synchronously imaging calcium transients in contralateral ACC neurons. We found that during social interactions a significant subpopulation of cerebellar PCs were robustly inhibited, while most modulated neurons in the DN were activated, and their activity was correlated with positively modulated ACC neurons. These distinctions largely disappeared when only non-social epochs were analyzed suggesting that cerebellar-cortical interactions were behaviorally specific. Our work provides new insights into the complexity of cerebellar activation and co-modulation of the ACC during social behavior and a valuable open-source tool for simultaneous, multimodal recordings in freely behaving mice.


Social behaviour is important for many animals, especially humans. It governs interactions between individuals and groups. One of the regions involved in social behaviour is the cerebellum, a part of the brain commonly known for controlling movement. It is likely that the cerebellum connects and influences other socially important areas in the brain, such as the anterior cingulate cortex. How exactly these regions communicate during social interaction is not well understood. One of the challenges studying communication between areas in the brain has been a lack of tools that can measure neural activity in multiple regions at once. To address this problem, Hur et al. developed a device called the E-Scope. The E-Scope can measure brain activity from two places in the brain at the same time. It can simultaneously record imaging and electrophysiological data of the different neurons. It is also small enough to be attached to animals without inhibiting their movements. Hur et al. tested the E-Scope by studying neurons in two regions of the cerebellum, called the right Crus I and the dentate nucleus, and in the anterior cingulate cortex during social interactions in mice. The E-Scope recorded from the animals as they interacted with other mice and compared them with those in mice that interacted with objects. During social interactions, Purkinje cells in the right Crus I were mostly less active, while neurons in the dentate nucleus and anterior cingulate cortex became overall more active. These results suggest that communication between the cerebellum and the anterior cingulate cortex is an important part of how the mouse brain coordinates social behaviour. The study of Hur et al. deepens our understanding of the function of the cerebellum in social behaviour. The E-Scope is an openly available tool to allow researchers to record communication between remote brain areas in small animals. This could be important to researchers trying to understand conditions like autism, which can involve difficulties in social interaction, or injuries to the cerebellum resulting in personality changes.


Subject(s)
Calcium , Gyrus Cinguli , Mice , Animals , Cerebellum , Social Behavior , Prosencephalon
11.
Food Chem ; 441: 138282, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38199108

ABSTRACT

This study has redirected focus towards the untapped potential of millets, exploring their utilization as small-scale vegetables like sprouts and microgreens. This study assessed the metabolite profiles and therapeutic efficacy of barnyard millets as sprouts and microgreens for antioxidant, anti-diabetic, and bioaccessibility properties. Based on the study, sprouts contained 456.52 mg GE/g of starch and microgreens contained 470.04 mg GE/g of carbohydrates, whereas the gastric phase of microgreens showed 426.85 mg BSAE/g, 397.6 mg LE/g, 348.19 g RE/g, and 307.40 g AAE/g of proteins, amino acids, vitamin A and vitamin C respectively. Secondary metabolites were significantly concentrated in the microgreen stage which is responsible for their increased antioxidant and antidiabetic potential than sprouts. This study validated the therapeutic and nutritional value of millet sprouts and microgreens by demonstrating their significant nutritional composition.


Subject(s)
Antioxidants , Echinochloa , Antioxidants/metabolism , Echinochloa/chemistry , Hypoglycemic Agents , Vitamins , Proteins
12.
J Neurosci ; 32(50): 17988-97, 2012 Dec 12.
Article in English | MEDLINE | ID: mdl-23238715

ABSTRACT

Climbing fiber (CF) input to the cerebellum is thought to instruct associative motor memory formation through its effects on multiple sites within the cerebellar circuit. We used adeno-associated viral delivery of channelrhodopsin-2 (ChR2) to inferior olivary neurons to selectively express ChR2 in CFs, achieving nearly complete transfection of CFs in the caudal cerebellar lobules of rats. As expected, optical stimulation of ChR2-expressing CFs generates complex spike responses in individual Purkinje neurons (PNs); in addition we found that such stimulation recruits a network of inhibitory interneurons in the molecular layer. This CF-driven disynaptic inhibition prolongs the postcomplex spike pause observed when spontaneously firing PNs receive direct CF input; such inhibition also elicits pauses in spontaneously firing PNs not receiving direct CF input. Baseline firing rates of PNs are strongly suppressed by low-frequency (2 Hz) stimulation of CFs, and this suppression is partly relieved by blocking synaptic inhibition. We conclude that CF-driven, disynaptic inhibition has a major influence on PN excitability and contributes to the widely observed negative correlation between complex and simple spike rates. Because they receive input from many CFs, molecular layer interneurons are well positioned to detect the spatiotemporal patterns of CF activity believed to encode error signals. Together, our findings suggest that such inhibition may bind together groups of Purkinje neurons to provide instructive signals to downstream sites in the cerebellar circuit.


Subject(s)
Neural Pathways/physiology , Neurons/physiology , Olivary Nucleus/physiology , Purkinje Cells/physiology , Animals , Female , Male , Neural Pathways/cytology , Neurons/cytology , Olivary Nucleus/cytology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
13.
Proc Natl Acad Sci U S A ; 107(4): 1630-5, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20080541

ABSTRACT

An additional copy of the beta-amyloid precursor protein (APP) gene causes early-onset Alzheimer's disease (AD) in trisomy 21 (DS). Endosome dysfunction develops very early in DS and AD and has been implicated in the mechanism of neurodegeneration. Here, we show that morphological and functional endocytic abnormalities in fibroblasts from individuals with DS are reversed by lowering the expression of APP or beta-APP-cleaving enzyme 1 (BACE-1) using short hairpin RNA constructs. By contrast, endosomal pathology can be induced in normal disomic (2N) fibroblasts by overexpressing APP or the C-terminal APP fragment generated by BACE-1 (betaCTF), all of which elevate the levels of betaCTFs. Expression of a mutant form of APP that cannot undergo beta-cleavage had no effect on endosomes. Pharmacological inhibition of APP gamma-secretase, which markedly reduced Abeta production but raised betaCTF levels, also induced AD-like endosome dysfunction in 2N fibroblasts and worsened this pathology in DS fibroblasts. These findings strongly implicate APP and the betaCTF of APP, and exclude Abeta and the alphaCTF, as the cause of endocytic pathway dysfunction in DS and AD, underscoring the potential multifaceted value of BACE-1 inhibition in AD therapeutics.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Aspartic Acid Endopeptidases/metabolism , Down Syndrome/metabolism , Endosomes/metabolism , RNA Interference , Adolescent , Adult , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/genetics , Cells, Cultured , Child , Child, Preschool , Down Syndrome/complications , Down Syndrome/genetics , Fibroblasts/metabolism , Humans , Infant , Protein Transport , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Young Adult
14.
Proc Natl Acad Sci U S A ; 107(17): 7969-74, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20385796

ABSTRACT

Familial Danish dementia (FDD) is a progressive neurodegenerative disease with cerebral deposition of Dan-amyloid (ADan), neuroinflammation, and neurofibrillary tangles, hallmark characteristics remarkably similar to those in Alzheimer's disease (AD). We have generated transgenic (tg) mouse models of familial Danish dementia that exhibit the age-dependent deposition of ADan throughout the brain with associated amyloid angiopathy, microhemorrhage, neuritic dystrophy, and neuroinflammation. Tg mice are impaired in the Morris water maze and exhibit increased anxiety in the open field. When crossed with TauP301S tg mice, ADan accumulation promotes neurofibrillary lesions, in all aspects similar to the Tau lesions observed in crosses between beta-amyloid (Abeta)-depositing tg mice and TauP301S tg mice. Although these observations argue for shared mechanisms of downstream pathophysiology for the sequence-unrelated ADan and Abeta peptides, the lack of codeposition of the two peptides in crosses between ADan- and Abeta-depositing mice points also to distinguishing properties of the peptides. Our results support the concept of the amyloid hypothesis for AD and related dementias, and suggest that different proteins prone to amyloid formation can drive strikingly similar pathogenic pathways in the brain.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Dementia/metabolism , Disease Models, Animal , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing , Alzheimer Disease/etiology , Animals , Blotting, Western , Dementia/etiology , Histological Techniques , Immunoassay , Membrane Glycoproteins , Mice , Mice, Transgenic , Neuropsychological Tests
15.
Aging Brain ; 4: 100102, 2023.
Article in English | MEDLINE | ID: mdl-38058491

ABSTRACT

Human apolipoprotein E (APOE) is the greatest determinant of genetic risk for memory deficits and Alzheimer's disease (AD). While APOE4 drives memory loss and high AD risk, APOE2 leads to healthy brain aging and reduced AD risk compared to the common APOE3 variant. We examined brain APOE protein levels in humanized mice homozygous for these alleles and found baseline levels to be age- and isoform-dependent: APOE2 levels were greater than APOE3, which were greater than APOE4. Despite the understanding that APOE lipoparticles do not traverse the blood-brain barrier, we show that brain APOE levels are responsive to dietary fat intake. Challenging mice for 6 months on a Western diet high in fat and cholesterol increased APOE protein levels in an allele-dependent fashion with a much greater increase within blood plasma than within the brain. In the brain, APOE2 levels responded most to the Western diet challenge, increasing by 20 % to 30 %. While increased lipoparticles are generally deleterious in the periphery, we propose that higher brain APOE2 levels may represent a readily available pool of beneficial lipid particles for neurons.

16.
bioRxiv ; 2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37066345

ABSTRACT

The cerebellum has been implicated in the regulation of social behavior. Its influence is thought to arise from communication, via the thalamus, to forebrain regions integral in the expression of social interactions, including the anterior cingulate cortex (ACC). However, the signals encoded or the nature of the communication between the cerebellum and these brain regions is poorly understood. Here, we describe an approach that overcomes technical challenges in exploring the coordination of distant brain regions at high temporal and spatial resolution during social behavior. We developed the E-Scope, an electrophysiology-integrated miniature microscope, to synchronously measure extracellular electrical activity in the cerebellum along with calcium imaging of the ACC. This single coaxial cable device combined these data streams to provide a powerful tool to monitor the activity of distant brain regions in freely behaving animals. During social behavior, we recorded the spike timing of multiple single units in cerebellar right Crus I (RCrus I) Purkinje cells (PCs) or dentate nucleus (DN) neurons while synchronously imaging calcium transients in contralateral ACC neurons. We found that during social interactions a significant subpopulation of cerebellar PCs were robustly inhibited, while most modulated neurons in the DN were activated, and their activity was correlated with positively modulated ACC neurons. These distinctions largely disappeared when only non-social epochs were analyzed suggesting that cerebellar-cortical interactions were behaviorally specific. Our work provides new insights into the complexity of cerebellar activation and co-modulation of the ACC during social behavior and a valuable open-source tool for simultaneous, multimodal recordings in freely behaving mice.

17.
Appl Biochem Biotechnol ; 195(11): 6790-6808, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36930407

ABSTRACT

Miliusa nilagirica, a rare tree species of Western Ghats, belongs to the Annonaceae family, a family with potential antioxidant and antidiabetic properties. This study is designed vividly to establish the relationship between the constituent phytochemicals and their hyperglycemic effects through the antioxidant traits of M. nilagirica in vitro. Phytochemical tests were conducted on dry powdered leaves and extracts of various methods to determine the existence of various constituents. The antidiabetic potential of leaf extracts was estimated by using the α-amylase inhibitory model and the antioxidant potential was estimated with various assays. The quantitative phytochemical screening of leaf parts shows the presence of carbohydrates (88.74 ± 0.65 mg GE/g sample), proteins (82.17 ± 2.52 mg BSAE/g sample), phenolics (40.44 ± 0.43 GAE/100 g), and flavonoids (66.05 ± 0.48 mg RE/g extract). Methanol extract of Soxhlet of M. nilagirica registered the strongest antioxidant activity in all assays, 75.66% inhibition (DPPH assay), 795.01 µmol/g (ABTS˙+ radical scavenging), 994.33 µmol/g (FRAP assay), 362.02 mg AAE/g extract (TAC assay), 47% inhibition (NO scavenging assay). In vitro α-amylase inhibition showed a highly noticeable reduction in ethyl acetate extract from Soxhlet (75.19%). HPLC and FTIR analyses on the extracts added strengths to the obtained results on the potentiality of M. nilagirica. From the results, it is evident that phytochemicals from M. nilagirica can be studied further, isolated, and incorporated as an alternative to synthetic supplements for hyperglycemia.


Subject(s)
Hypoglycemic Agents , Plant Extracts , Hypoglycemic Agents/pharmacology , Plant Extracts/pharmacology , Plant Extracts/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Phytochemicals/pharmacology , Phytochemicals/chemistry , alpha-Amylases , Plant Leaves
18.
J Neurosci ; 31(5): 1837-49, 2011 Feb 02.
Article in English | MEDLINE | ID: mdl-21289194

ABSTRACT

Deposition of amyloid ß peptides (Aßs) in extracellular amyloid plaques within the human brain is a hallmark of Alzheimer's disease (AD). Aß derives from proteolytic processing of the amyloid precursor protein (APP) by ß- and γ-secretases. The initial cleavage by ß-secretase results in shedding of the APP ectodomain and generation of APP C-terminal fragments (APP-CTFs), which can then be further processed within the transmembrane domain by γ-secretase, resulting in release of Aß. Here, we demonstrate that accumulation of sphingolipids (SLs), as occurs in lysosomal lipid storage disorders (LSDs), decreases the lysosome-dependent degradation of APP-CTFs and stimulates γ-secretase activity. Together, this results in increased generation of both intracellular and secreted Aß. Notably, primary fibroblasts from patients with different SL storage diseases show strong accumulation of potentially amyloidogenic APP-CTFs. By using biochemical, cell biological, and genetic approaches, we demonstrate that SL accumulation affects autophagic flux and impairs the clearance of APP-CTFs. Thus, accumulation of SLs might not only underlie the pathogenesis of LSDs, but also trigger increased generation of Aß and contribute to neurodegeneration in sporadic AD.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Autophagy , Fibroblasts/metabolism , Lysosomes/metabolism , Sphingolipids/metabolism , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fibroblasts/ultrastructure , Immunohistochemistry , Lysosomes/genetics , Lysosomes/ultrastructure , Mice , Mice, Knockout , Microscopy, Electron , Peptide Fragments/metabolism , Sphingolipids/genetics , Transfection
19.
J Neurosci ; 31(34): 12083-93, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21865451

ABSTRACT

The proteolytic machinery comprising metalloproteases and γ-secretase, an intramembrane aspartyl protease involved in Alzheimer's disease, cleaves several substrates in addition to the extensively studied amyloid precursor protein. Some of these substrates, such as N-cadherin, are synaptic proteins involved in synapse remodeling and maintenance. Here we show, in rats and mice, that metalloproteases and γ-secretase are physiologic regulators of synapses. Both proteases are synaptic, with γ-secretase tethered at the synapse by δ-catenin, a synaptic scaffolding protein that also binds to N-cadherin and, through scaffolds, to AMPA receptor and a metalloprotease. Activity-dependent proteolysis by metalloproteases and γ-secretase takes place at both sides of the synapse, with the metalloprotease cleavage being NMDA receptor-dependent. This proteolysis decreases levels of synaptic proteins and diminishes synaptic transmission. Our results suggest that activity-dependent substrate cleavage by synaptic metalloproteases and γ-secretase modifies synaptic transmission, providing a novel form of synaptic autoregulation.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Hippocampus/enzymology , Homeostasis/physiology , Metalloproteases/physiology , Synapses/enzymology , Synaptic Transmission/physiology , Animals , Catenins/deficiency , Catenins/genetics , Cells, Cultured , Female , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Synaptic Membranes/enzymology , Synaptic Membranes/ultrastructure , Delta Catenin
20.
Brain ; 134(Pt 1): 258-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21186265

ABSTRACT

Autophagy, a major degradative pathway for proteins and organelles, is essential for survival of mature neurons. Extensive autophagic-lysosomal pathology in Alzheimer's disease brain contributes to Alzheimer's disease pathogenesis, although the underlying mechanisms are not well understood. Here, we identified and characterized marked intraneuronal amyloid-ß peptide/amyloid and lysosomal system pathology in the Alzheimer's disease mouse model TgCRND8 similar to that previously described in Alzheimer's disease brains. We further establish that the basis for these pathologies involves defective proteolytic clearance of neuronal autophagic substrates including amyloid-ß peptide. To establish the pathogenic significance of these abnormalities, we enhanced lysosomal cathepsin activities and rates of autophagic protein turnover in TgCRND8 mice by genetically deleting cystatin B, an endogenous inhibitor of lysosomal cysteine proteases. Cystatin B deletion rescued autophagic-lysosomal pathology, reduced abnormal accumulations of amyloid-ß peptide, ubiquitinated proteins and other autophagic substrates within autolysosomes/lysosomes and reduced intraneuronal amyloid-ß peptide. The amelioration of lysosomal function in TgCRND8 markedly decreased extracellular amyloid deposition and total brain amyloid-ß peptide 40 and 42 levels, and prevented the development of deficits of learning and memory in fear conditioning and olfactory habituation tests. Our findings support the pathogenic significance of autophagic-lysosomal dysfunction in Alzheimer's disease and indicate the potential value of restoring normal autophagy as an innovative therapeutic strategy for Alzheimer's disease.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Autophagy/physiology , Brain/pathology , Memory Disorders/physiopathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Analysis of Variance , Animals , Blotting, Western , Brain/metabolism , Brain/physiopathology , Conditioning, Psychological , Enzyme-Linked Immunosorbent Assay , Fear , Habituation, Psychophysiologic , Immunohistochemistry , Lysosomes/metabolism , Lysosomes/pathology , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL