Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
J Alzheimers Dis ; 93(3): 1065-1081, 2023.
Article de Anglais | MEDLINE | ID: mdl-37212118

RÉSUMÉ

BACKGROUND: The tau protein phosphorylated at Thr181 (p-tau181) in cerebrospinal fluid and blood is a sensitive biomarker for Alzheimer's disease (AD). Increased p-tau181 levels correlate well with amyloid-ß (Aß) pathology and precede neurofibrillary tangle formation in the early stage of AD; however, the relationship between p-tau181 and Aß-mediated pathology is less well understood. We recently reported that p-tau181 represents axonal abnormalities in mice with Aß pathology (AppNLGF). However, from which neuronal subtype(s) these p-tau181-positive axons originate remains elusive. OBJECTIVE: The main purpose of this study is to differentiate neuronal subtype(s) and elucidate damage associated with p-tau181-positive axons by immunohistochemical analysis of AppNLGF mice brains. METHODS: Colocalization between p-tau181 and (1) unmyelinated axons positive for vesicular acetylcholine transporter or norepinephrine transporter and (2) myelinated axons positive for vesicular glutamate transporter, vesicular GABA transporter, or parvalbumin in the brains of 24-month-old AppNLGF and control mice without Aß pathology were analyzed. The density of these axons was also compared. RESULTS: Unmyelinated axons of cholinergic or noradrenergic neurons did not overlap with p-tau181. By contrast, p-tau181 signals colocalized with myelinated axons of parvalbumin-positive GABAergic interneurons but not of glutamatergic neurons. Interestingly, the density of unmyelinated axons was significantly decreased in AppNLGF mice, whereas that of glutamatergic, GABAergic, or p-tau181-positive axons was less affected. Instead, myelin sheaths surrounding p-tau181-positive axons were significantly reduced in AppNLGF mice. CONCLUSION: This study demonstrates that p-tau181 signals colocalize with axons of parvalbumin-positive GABAergic interneurons with disrupted myelin sheaths in the brains of a mouse model of Aß pathology.


Sujet(s)
Maladie d'Alzheimer , Animaux , Souris , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Axones/anatomopathologie , Marqueurs biologiques/liquide cérébrospinal , Interneurones , Parvalbumines/métabolisme , Protéines tau/métabolisme
2.
iScience ; 26(2): 105968, 2023 Feb 17.
Article de Anglais | MEDLINE | ID: mdl-36718365

RÉSUMÉ

Drosophila Toll-9 is most closely related to mammalian Toll-like receptors; however, physiological functions of Toll-9 remain elusive. We examined the roles of Toll-9 in fly brains in aging and neurodegeneration. Toll-9 mRNA levels were increased in aged fly heads accompanied by activation of nuclear factor-kappa B (NF-kB) and stress-activated protein kinase (SAPK) signaling, and many of these changes were modulated by Toll-9 in glial cells. The loss of Toll-9 did not affect lifespan or brain integrity, whereas it exacerbated hydrogen peroxide-induced lethality. Toll-9 expression was also induced by nerve injury but did not affect acute stress response or glial engulfment activity, suggesting Toll-9 may modulate subsequent neurodegeneration. In a fly tauopathy model, Toll-9 deficiency enhanced neurodegeneration and disease-related tau phosphorylation with reduced SAPK activity, and blocking SAPK enhanced tau phosphorylation and neurodegeneration. In sum, Toll-9 is induced upon aging and nerve injury and affects neurodegeneration by modulating stress kinase signaling.

3.
Brain Commun ; 4(6): fcac286, 2022.
Article de Anglais | MEDLINE | ID: mdl-36440096

RÉSUMÉ

Phospho-tau 217, phospho-tau 231 and phospho-tau 181 in cerebrospinal fluid and plasma are promising biomarkers for the diagnosis of Alzheimer's disease. All these p-tau proteins are detected in neurofibrillary tangles in brains obtained post-mortem from Alzheimer's disease patients. However, increases in p-tau levels in cerebrospinal fluid and plasma during the preclinical stage of Alzheimer's disease correlate with amyloid-ß burden and precede neurofibrillary tangles in brains, suggesting that these p-tau proteins are indicative of amyloid-ß-mediated brain pathology. In addition, phospho-tau 217 has greater sensitivity than phospho-tau 181, though it is unclear whether each of these p-tau variants contributes to the same or a different type of neuropathology prior to neurofibrillary tangle formation. In this study, we evaluated the intracerebral localization of p-tau in App knock-in mice with amyloid-ß plaques without neurofibrillary tangle pathology (AppNLGF ), in App knock-in mice with increased amyloid-ß levels without amyloid-ß plaques (AppNL ) and in wild-type mice. Immunohistochemical analysis showed that phospho-tau 217 and phospho-tau 231 were detected only in AppNLGF mice as punctate structures around amyloid-ß plaques, overlapping with the tau pathology marker, AT8 epitope phospho-tau 202/205/208. Moreover, phospho-tau 217 and phospho-tau 202/205/208 colocalized with the postsynaptic marker PSD95 and with a major tau kinase active, GSK3ß. In contrast and similar to total tau, phospho-tau 181 signals were readily detectable as fibre structures in wild-type and AppNL mice and colocalized with an axonal marker neurofilament light chain. In AppNLGF mice, these phospho-tau 181-positive structures were disrupted around amyloid-ß plaques and only partially overlapped with phospho-tau 217. These results indicate that phospho-tau 217, phospho-tau 231 and a part of phospho-tau 181 signals are markers of postsynaptic pathology around amyloid-ß plaques, with phospho-tau 181 also being a marker of axonal abnormality caused by amyloid-ß burden in brains.

4.
J Alzheimers Dis ; 82(4): 1513-1530, 2021.
Article de Anglais | MEDLINE | ID: mdl-34180416

RÉSUMÉ

BACKGROUND: The locus coeruleus (LC), a brainstem nucleus comprising noradrenergic neurons, is one of the earliest regions affected by Alzheimer's disease (AD). Amyloid-ß (Aß) pathology in the cortex in AD is thought to exacerbate the age-related loss of LC neurons, which may lead to cortical tau pathology. However, mechanisms underlying LC neurodegeneration remain elusive. OBJECTIVE: Here, we aimed to examine how noradrenergic neurons are affected by cortical Aß pathology in AppNL-G-F/NL-G-F knock-in mice. METHODS: The density of noradrenergic axons in LC-innervated regions and the LC neuron number were analyzed by an immunohistochemical method. To explore the potential mechanisms for LC degeneration, we also examined the occurrence of tau pathology in LC neurons, the association of reactive gliosis with LC neurons, and impaired trophic support in the brains of AppNL-G-F/NL-G-F mice. RESULTS: We observed a significant reduction in the density of noradrenergic axons from the LC in aged AppNL-G-F/NL-G-F mice without neuron loss or tau pathology, which was not limited to areas near Aß plaques. However, none of the factors known to be related to the maintenance of LC neurons (i.e., somatostatin/somatostatin receptor 2, brain-derived neurotrophic factor, nerve growth factor, and neurotrophin-3) were significantly reduced in AppNL-G-F/NL-G-F mice. CONCLUSION: This study demonstrates that cortical Aß pathology induces noradrenergic neurodegeneration, and further elucidation of the underlying mechanisms will reveal effective therapeutics to halt AD progression.


Sujet(s)
Neurones adrénergiques , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Amyloïdose/métabolisme , Locus ceruleus/anatomopathologie , Dégénérescence nerveuse/métabolisme , Animaux , Encéphale/anatomopathologie , Modèles animaux de maladie humaine , Techniques de knock-in de gènes , Humains , Mâle , Souris , Souris transgéniques
5.
STAR Protoc ; 2(2): 100501, 2021 06 18.
Article de Anglais | MEDLINE | ID: mdl-33997815

RÉSUMÉ

For decades, the fruit fly Drosophila melanogaster has been an efficient genetic model to investigate many aspects of human neurodegenerative diseases. Through genetic and pharmacologic approaches, these studies have revealed the molecular mechanisms underlying disease pathogenesis and provided therapeutic implications. Here, we describe a protocol for assessing Alzheimer's disease-related amyloid-ß toxicity in a transgenic fly model through biochemical, histological, and behavioral analyses. We also discuss the advantages and limitations of our protocols. For complete details on the use and execution of this protocol, please refer to Wang et al. (2021).


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/génétique , Peptides bêta-amyloïdes/métabolisme , Animaux , Animal génétiquement modifié/génétique , Animal génétiquement modifié/métabolisme , Modèles animaux de maladie humaine , Drosophila melanogaster , Humains
6.
Neuron ; 109(2): 257-272.e14, 2021 01 20.
Article de Anglais | MEDLINE | ID: mdl-33238137

RÉSUMÉ

To identify the molecular mechanisms and novel therapeutic targets of late-onset Alzheimer's Disease (LOAD), we performed an integrative network analysis of multi-omics profiling of four cortical areas across 364 donors with varying cognitive and neuropathological phenotypes. Our analyses revealed thousands of molecular changes and uncovered neuronal gene subnetworks as the most dysregulated in LOAD. ATP6V1A was identified as a key regulator of a top-ranked neuronal subnetwork, and its role in disease-related processes was evaluated through CRISPR-based manipulation in human induced pluripotent stem cell-derived neurons and RNAi-based knockdown in Drosophila models. Neuronal impairment and neurodegeneration caused by ATP6V1A deficit were improved by a repositioned compound, NCH-51. This study provides not only a global landscape but also detailed signaling circuits of complex molecular interactions in key brain regions affected by LOAD, and the resulting network models will serve as a blueprint for developing next-generation therapeutic agents against LOAD.


Sujet(s)
Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/thérapie , Encéphale/physiologie , Bases de données génétiques , Réseaux de régulation génique/physiologie , Transduction du signal/physiologie , Maladie d'Alzheimer/anatomopathologie , Animaux , Animal génétiquement modifié , Encéphale/anatomopathologie , Bases de données génétiques/tendances , Drosophila melanogaster , Femelle , Humains , Cellules souches pluripotentes induites/physiologie , Mâle , Analyse de séquence d'ARN/méthodes
7.
Hum Mol Genet ; 29(5): 817-833, 2020 03 27.
Article de Anglais | MEDLINE | ID: mdl-31942999

RÉSUMÉ

The molecular biological mechanisms of Alzheimer's disease (AD) involve disease-associated crosstalk through many genes and include a loss of normal as well as a gain of abnormal interactions among genes. A protein domain network (PDN) is a collection of physical bindings that occur between protein domains, and the states of the PDNs in patients with AD are likely to be perturbed compared to those in normal healthy individuals. To identify PDN changes that cause neurodegeneration, we analysed the PDNs that occur among genes co-expressed in each of three brain regions at each stage of AD. Our analysis revealed that the PDNs collapsed with the progression of AD stage and identified five hub genes, including Rac1, as key players in PDN collapse. Using publicly available as well as our own gene expression data, we confirmed that the mRNA expression level of the RAC1 gene was downregulated in the entorhinal cortex (EC) of AD brains. To test the causality of these changes in neurodegeneration, we utilized Drosophila as a genetic model and found that modest knockdown of Rac1 in neurons was sufficient to cause age-dependent behavioural deficits and neurodegeneration. Finally, we identified a microRNA, hsa-miR-101-3p, as a potential regulator of RAC1 in AD brains. As the Braak neurofibrillary tangle (NFT) stage progressed, the expression levels of hsa-miR-101-3p were increased specifically in the EC. Furthermore, overexpression of hsa-miR-101-3p in the human neuronal cell line SH-SY5Y caused RAC1 downregulation. These results highlight the utility of our integrated network approach for identifying causal changes leading to neurodegeneration in AD.


Sujet(s)
Maladie d'Alzheimer/anatomopathologie , Régulation de l'expression des gènes , Réseaux de régulation génique , Maladies neurodégénératives/étiologie , Enchevêtrements neurofibrillaires/anatomopathologie , Protéine G rac1/métabolisme , Maladie d'Alzheimer/complications , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Animaux , Évolution de la maladie , Drosophila melanogaster , Humains , microARN/génétique , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie , Enchevêtrements neurofibrillaires/métabolisme , Motifs et domaines d'intéraction protéique , Protéine G rac1/génétique
8.
BMC Neurosci ; 20(1): 13, 2019 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-30894120

RÉSUMÉ

BACKGROUND: Knock-in (KI) mouse models of Alzheimer's disease (AD) that endogenously overproduce Aß without non-physiological overexpression of amyloid precursor protein (APP) provide important insights into the pathogenic mechanisms of AD. Previously, we reported that AppNL-G-F mice, which harbor three familial AD mutations (Swedish, Beyreuther/Iberian, and Arctic) exhibited emotional alterations before the onset of definitive cognitive deficits. To determine whether these mice exhibit deficits in learning and memory at more advanced ages, we compared the Morris water maze performance of AppNL-G-F and AppNL mice, which harbor only the Swedish mutation, with that of wild-type (WT) C57BL/6J mice at the age of 24 months. To correlate cognitive deficits and neuroinflammation, we also examined Aß plaque formation and reactive gliosis in these mice. RESULTS: In the Morris water maze, a spatial task, 24-month-old AppNL-G-F/NL-G-F mice exhibited significantly poorer spatial learning than WT mice during the hidden training sessions, but similarly to WT mice during the visible training sessions. Not surprisingly, AppNL-G-F/NL-G-F mice also exhibited spatial memory deficits both 1 and 7 days after the last training session. By contrast, 24-month-old AppNL/NL mice had intact spatial learning and memory relative to WT mice. Immunohistochemical analyses revealed that 24-month-old AppNL-G-F/NL-G-F mice developed massive Aß plaques and reactive gliosis (microgliosis and astrocytosis) throughout the brain, including the cortex and hippocampus. By contrast, we observed no detectable brain pathology in AppNL/NL mice despite overproduction of human Aß40 and Aß42 in their brains. CONCLUSIONS: Aß plaque formation, followed by sustained neuroinflammation, is necessary for the induction of definitive cognitive deficits in App-KI mouse models of AD. Our data also indicate that introduction of the Swedish mutation alone in endogenous APP is not sufficient to produce either AD-related brain pathology or cognitive deficits in mice.


Sujet(s)
Maladie d'Alzheimer/métabolisme , Précurseur de la protéine bêta-amyloïde/métabolisme , Dysfonctionnement cognitif/étiologie , Dysfonctionnement cognitif/métabolisme , Gliose/métabolisme , Plaque amyloïde/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/psychologie , Peptides bêta-amyloïdes/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Animaux , Encéphale/métabolisme , Encéphale/anatomopathologie , Dysfonctionnement cognitif/anatomopathologie , Modèles animaux de maladie humaine , Techniques de knock-in de gènes , Gliose/anatomopathologie , Gliose/psychologie , Humains , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/psychologie , Mâle , Apprentissage du labyrinthe/physiologie , Souris de lignée C57BL , Souris transgéniques , Fragments peptidiques/métabolisme , Plaque amyloïde/anatomopathologie , Plaque amyloïde/psychologie , Mémoire spatiale/physiologie
9.
BMC Neurosci ; 19(1): 46, 2018 07 28.
Article de Anglais | MEDLINE | ID: mdl-30055565

RÉSUMÉ

BACKGROUND: Alzheimer's disease (AD), the most common cause of dementia, is characterized by the progressive deposition of amyloid-ß (Aß) peptides and neurofibrillary tangles. Mouse models of Aß amyloidosis generated by knock-in (KI) of a humanized Aß sequence provide distinct advantages over traditional transgenic models that rely on overexpression of amyloid precursor protein (APP). In App-KI mice, three familial AD-associated mutations were introduced into the endogenous mouse App locus to recapitulate Aß pathology observed in AD: the Swedish (NL) mutation, which elevates total Aß production; the Beyreuther/Iberian (F) mutation, which increases the Aß42/Aß40 ratio; and the Arctic (G) mutation, which promotes Aß aggregation. AppNL-G-F mice harbor all three mutations and develop progressive Aß amyloidosis and neuroinflammatory response in broader brain areas, whereas AppNL mice carrying only the Swedish mutation exhibit no overt AD-related pathological changes. To identify behavioral alterations associated with Aß pathology, we assessed emotional and cognitive domains of AppNL-G-F and AppNL mice at different time points, using the elevated plus maze, contextual fear conditioning, and Barnes maze tasks. RESULTS: Assessments of emotional domains revealed that, in comparison with wild-type (WT) C57BL/6J mice, AppNL-G-F/NL-G-F mice exhibited anxiolytic-like behavior that was detectable from 6 months of age. By contrast, AppNL/NL mice exhibited anxiogenic-like behavior from 15 months of age. In the contextual fear conditioning task, both AppNL/NL and AppNL-G-F/NL-G-F mice exhibited intact learning and memory up to 15-18 months of age, whereas AppNL-G-F/NL-G-F mice exhibited hyper-reactivity to painful stimuli. In the Barnes maze task, AppNL-G-F/NL-G-F mice exhibited a subtle decline in spatial learning ability at 8 months of age, but retained normal memory functions. CONCLUSION: AppNL/NL and AppNL-G-F/NL-G-F mice exhibit behavioral changes associated with non-cognitive, emotional domains before the onset of definitive cognitive deficits. Our observations consistently indicate that AppNL-G-F/NL-G-F mice represent a model for preclinical AD. These mice are useful for the study of AD prevention rather than treatment after neurodegeneration.


Sujet(s)
Peptides bêta-amyloïdes/génétique , Amyloïdose/génétique , Comportement animal/physiologie , Émotions/physiologie , Techniques de knock-in de gènes , Maladie d'Alzheimer/génétique , Précurseur de la protéine bêta-amyloïde/génétique , Animaux , Troubles de la cognition/génétique , Dysfonctionnement cognitif/génétique , Modèles animaux de maladie humaine , Souris transgéniques
10.
Genome Med ; 10(1): 26, 2018 03 29.
Article de Anglais | MEDLINE | ID: mdl-29598827

RÉSUMÉ

BACKGROUND: Cerebral amyloidosis, neuroinflammation, and tauopathy are key features of Alzheimer's disease (AD), but interactions among these features remain poorly understood. Our previous multiscale molecular network models of AD revealed TYROBP as a key driver of an immune- and microglia-specific network that was robustly associated with AD pathophysiology. Recent genetic studies of AD further identified pathogenic mutations in both TREM2 and TYROBP. METHODS: In this study, we systematically examined molecular and pathological interactions among Aß, tau, TREM2, and TYROBP by integrating signatures from transgenic Drosophila models of AD and transcriptome-wide gene co-expression networks from two human AD cohorts. RESULTS: Glial expression of TREM2/TYROBP exacerbated tau-mediated neurodegeneration and synergistically affected pathways underlying late-onset AD pathology, while neuronal Aß42 and glial TREM2/TYROBP synergistically altered expression of the genes in synaptic function and immune modules in AD. CONCLUSIONS: The comprehensive pathological and molecular data generated through this study strongly validate the causal role of TREM2/TYROBP in driving molecular networks in AD and AD-related phenotypes in flies.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Maladie d'Alzheimer/génétique , Peptides bêta-amyloïdes/métabolisme , Protéines de Drosophila/métabolisme , Drosophila melanogaster/génétique , Glycoprotéines membranaires/métabolisme , Protéines membranaires/métabolisme , Récepteurs immunologiques/métabolisme , Protéines tau/métabolisme , Animaux , Animal génétiquement modifié , Encéphale/métabolisme , Encéphale/anatomopathologie , Modèles animaux de maladie humaine , Protéines de Drosophila/génétique , Femelle , Régulation de l'expression des gènes , Réseaux de régulation génique , Humains , Mâle , Dégénérescence nerveuse/génétique , Dégénérescence nerveuse/anatomopathologie , Névroglie/métabolisme , Neurones/métabolisme , Transduction du signal/génétique , Synapses/métabolisme
11.
PLoS Genet ; 14(1): e1007196, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29357349

RÉSUMÉ

Wolfram syndrome (WS), caused by loss-of-function mutations in the Wolfram syndrome 1 gene (WFS1), is characterized by juvenile-onset diabetes mellitus, bilateral optic atrophy, and a wide spectrum of neurological and psychiatric manifestations. WFS1 encodes an endoplasmic reticulum (ER)-resident transmembrane protein, and mutations in this gene lead to pancreatic ß-cell death induced by high levels of ER stress. However, the mechanisms underlying neurodegeneration caused by WFS1 deficiency remain elusive. Here, we investigated the role of WFS1 in the maintenance of neuronal integrity in vivo by knocking down the expression of wfs1, the Drosophila homolog of WFS1, in the central nervous system. Neuronal knockdown of wfs1 caused age-dependent behavioral deficits and neurodegeneration in the fly brain. Knockdown of wfs1 in neurons and glial cells resulted in premature death and significantly exacerbated behavioral deficits in flies, suggesting that wfs1 has important functions in both cell types. Although wfs1 knockdown alone did not promote ER stress, it increased the susceptibility to oxidative stress-, excitotoxicity- or tauopathy-induced behavioral deficits, and neurodegeneration. The glutamate release inhibitor riluzole significantly suppressed premature death phenotypes induced by neuronal and glial knockdown of wfs1. This study highlights the protective role of wfs1 against age-associated neurodegeneration and furthers our understanding of potential disease-modifying factors that determine susceptibility and resilience to age-associated neurodegenerative diseases.


Sujet(s)
Drosophila melanogaster/génétique , Protéines membranaires/génétique , Troubles mentaux/génétique , Dégénérescence nerveuse/génétique , Système nerveux/métabolisme , Vieillissement/génétique , Vieillissement/anatomopathologie , Animaux , Animal génétiquement modifié , Cellules cultivées , Techniques de knock-down de gènes , Prédisposition génétique à une maladie , Humains , Neurones/métabolisme , Similitude de séquences , Stress psychologique/complications , Stress psychologique/génétique , Stress psychologique/physiopathologie , Syndrome de Wolfram/génétique
12.
Dev Cell ; 41(6): 652-664.e5, 2017 06 19.
Article de Anglais | MEDLINE | ID: mdl-28633019

RÉSUMÉ

The unfolded protein response (UPR), which protects cells against accumulation of misfolded proteins in the ER, is induced in several age-associated degenerative diseases. However, sustained UPR activation has negative effects on cellular functions and may worsen disease symptoms. It remains unknown whether and how UPR components can be utilized to counteract chronic ER proteinopathies. We found that promotion of ER-associated degradation (ERAD) through upregulation of ERAD-enhancing α-mannosidase-like proteins (EDEMs) protected against chronic ER proteinopathy without inducing toxicity in a Drosophila model. ERAD activity in the brain decreased with aging, and upregulation of EDEMs suppressed age-dependent behavioral decline and extended the lifespan without affecting the UPR gene expression network. Intriguingly, EDEM mannosidase activity was dispensable for these protective effects. Therefore, upregulation of EDEM function in the ERAD protects against ER proteinopathy in vivo and thus represents a potential therapeutic target for chronic diseases.


Sujet(s)
Dégradation associée au réticulum endoplasmique/physiologie , Réticulum endoplasmique/métabolisme , Expression des gènes/physiologie , Glycoprotéines/métabolisme , Protéines membranaires/métabolisme , Animaux , Lignée cellulaire , Drosophila melanogaster/métabolisme , Pliage des protéines
13.
PLoS Genet ; 12(3): e1005917, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-27023670

RÉSUMÉ

Abnormal accumulation of the microtubule-interacting protein tau is associated with neurodegenerative diseases including Alzheimer's disease (AD). ß-amyloid (Aß) lies upstream of abnormal tau behavior, including detachment from microtubules, phosphorylation at several disease-specific sites, and self-aggregation into toxic tau species in AD brains. To prevent the cascade of events leading to neurodegeneration in AD, it is essential to elucidate the mechanisms underlying the initial events of tau mismetabolism. Currently, however, these mechanisms remain unclear. In this study, using transgenic Drosophila co-expressing human tau and Aß, we found that tau phosphorylation at AD-related Ser262/356 stabilized microtubule-unbound tau in the early phase of tau mismetabolism, leading to neurodegeneration. Aß increased the level of tau detached from microtubules, independent of the phosphorylation status at GSK3-targeted SP/TP sites. Such mislocalized tau proteins, especially the less phosphorylated species, were stabilized by phosphorylation at Ser262/356 via PAR-1/MARK. Levels of Ser262 phosphorylation were increased by Aß42, and blocking this stabilization of tau suppressed Aß42-mediated augmentation of tau toxicity and an increase in the levels of tau phosphorylation at the SP/TP site Thr231, suggesting that this process may be involved in AD pathogenesis. In contrast to PAR-1/MARK, blocking tau phosphorylation at SP/TP sites by knockdown of Sgg/GSK3 did not reduce tau levels, suppress tau mislocalization to the cytosol, or diminish Aß-mediated augmentation of tau toxicity. These results suggest that stabilization of microtubule-unbound tau by phosphorylation at Ser262/356 via the PAR-1/MARK may act in the initial steps of tau mismetabolism in AD pathogenesis, and that such tau species may represent a potential therapeutic target for AD.


Sujet(s)
Maladie d'Alzheimer/génétique , Peptides bêta-amyloïdes/génétique , Glycogen Synthase Kinase 3/génétique , Récepteur de type PAR-1/génétique , Protéines tau/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Animaux , Animal génétiquement modifié , Modèles animaux de maladie humaine , Drosophila/génétique , Drosophila/métabolisme , Glycogen Synthase Kinase 3/métabolisme , Glycogen synthase kinase 3 beta , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Humains , Microtubules/métabolisme , Microtubules/anatomopathologie , Neurones/métabolisme , Neurones/anatomopathologie , Phosphorylation , Récepteur de type PAR-1/métabolisme , Tubuline/génétique , Tubuline/métabolisme , Protéines tau/métabolisme
14.
J Proteome Res ; 12(6): 2654-65, 2013 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-23550703

RÉSUMÉ

Hyperphosphorylation of microtubule-associated protein tau is thought to contribute to Alzheimer's disease (AD) pathogenesis. We previously showed that DNA damage-activated cell cycle checkpoint kinases Chk1 and Chk2 phosphorylate tau at an AD-related site and enhance tau toxicity, suggesting potential roles of these kinases in AD. The purpose of this study is to systematically identify which sites in tau are directly phosphorylated by Chk1 and Chk2. Using recombinant human tau phosphorylated by Chk1 and Chk2 in vitro, we first analyzed tau phosphorylation at the AD-related sites by Western blot with phospho-tau-specific antibodies. Second, to globally identify phosphorylated sites in tau, liquid chromatography-tandem mass spectrometry (LC-MS(3)) was employed. These systematic analyses identified a total of 27 Ser/Thr residues as Chk1- or Chk2- target sites. None of them were proline-directed kinase targets. Many of these sites are located within the microtubule-binding domain and C-terminal domain, whose phosphorylation has been shown to reduce tau binding to microtubules and/or has been implicated in tau toxicity. Among these 27 sites, 13 sites have been identified to be phosphorylated in AD brains. Since DNA damage is accumulated in diseased brains, Chk1 and Chk2 may be involved in tau phosphorylation and toxicity in AD pathogenesis.


Sujet(s)
Checkpoint kinase 2/métabolisme , Protein kinases/métabolisme , Protéines recombinantes/métabolisme , Protéines tau/composition chimique , Protéines tau/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Checkpoint kinase 1 , Chromatographie en phase liquide , Altération de l'ADN , Humains , Microtubules/métabolisme , Microtubules/anatomopathologie , Phosphorylation , Liaison aux protéines , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Spectrométrie de masse en tandem , Protéines tau/génétique
15.
J Nat Med ; 67(3): 626-35, 2013 Jul.
Article de Anglais | MEDLINE | ID: mdl-23203627

RÉSUMÉ

The intestinal epithelial cells sit at the interface between a lumen and a lamina propria or lymph nodes such as Peyer's patches, where they play important roles in maintaining intestinal homeostasis through chemokine secretion. This study investigated the effect of Hochuekkito (TJ-41)-a traditional Japanese herbal (Kampo) formula used as a tonic for weakness-on chemokine expression in intestinal epithelial cells in order to explore the mechanism of its modulating effect against mucosal immunity. When cells from the rat normal small intestinal epithelial cell-line IEC-6 were stimulated with TJ-41, mRNA expression of CC chemokine ligand (CCL) 11 (eotaxin), CCL20 (MIP-3α) and CCL25 (TECK) was enhanced. Oral administration of TJ-41 to methotrexate-treated mice enhanced mRNA expression of CCL25 and keratinocyte growth factor in the jejunum with, decreasing mRNA expression of the inflammatory marker tumor necrosis factor (TNF)-α. Although oral administration of TJ-41 did not affect CCL20 mRNA expression in villus epithelium of methotrexate-treated mice, enhancement of CCL20 mRNA expression was observed in Peyer's patches. Immunohistochemical analysis detected dense staining with anti-CCL20 antibody in the follicle-associated epithelium region of Peyer's patches in mice administered TJ-41. Analysis of active ingredients indicates that polysaccharide-containing macromolecules in TJ-41 contribute to the enhancement of CCL20 mRNA expression through an intracellular signal cascade via nuclear factor kappa B (NF-κB) activation.


Sujet(s)
Anti-inflammatoires/pharmacologie , Chimiokines/métabolisme , Médicaments issus de plantes chinoises/pharmacologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Muqueuse intestinale/effets des médicaments et des substances chimiques , Jéjunum/effets des médicaments et des substances chimiques , Médecine kampo , Administration par voie orale , Animaux , Anti-inflammatoires/administration et posologie , Lignée cellulaire , Chimiokine CCL11/métabolisme , Chimiokine CCL20/métabolisme , Chimiokines/génétique , Chimiokines CC/métabolisme , Médicaments issus de plantes chinoises/administration et posologie , Cellules épithéliales/immunologie , Cellules épithéliales/métabolisme , Femelle , Facteur de croissance fibroblastique de type 7/métabolisme , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , Jéjunum/immunologie , Jéjunum/métabolisme , Souris , Souris de lignée BALB C , Facteur de transcription NF-kappa B/métabolisme , Plaques de Peyer/effets des médicaments et des substances chimiques , Plaques de Peyer/métabolisme , Phytothérapie , Plantes médicinales , ARN messager/métabolisme , Rats , Transduction du signal , Facteurs temps , Facteur de nécrose tumorale alpha/métabolisme
16.
J Agric Food Chem ; 61(3): 569-78, 2013 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-23256460

RÉSUMÉ

A high proportion of pelargonidin 3-O-ß-D-glucopyranoside (Pg3G) is metabolized to glucuronides and excreted in mammal urine after ingestion of strawberry fruit, suggesting that these metabolites play important functional roles in vivo. The aim of the present study was to elucidate the structures and determine the biological fate of the two dominant metabolites of Pg3G in rats to enable an accurate discussion of the biological properties of anthocyanins. Authentic Pg3G was orally administered to rats. One pelargonidin monoglucuronide and three Pg3G-monoglucuronides (glucuronides of the glucoside) were identified together with intact Pg3G in both blood plasma and urine samples. The structures of the two dominant metabolites were elucidated as pelargonidin 3-O-ß-D-glucuronide (Pg3GlcA) and pelargonidin 3-O-ß-D-glucuronyl-(1→2)-ß-D-glucoside by means of (1)H and (13)C nuclear magnetic resonance spectroscopy and heteronuclear multiple-bond connective spectroscopy. The bioavailability of Pg3G in its intact form was 0.31% of the orally administered dose, and 0.65% was absorbed in the Pg3GlcA form.


Sujet(s)
Anthocyanes/composition chimique , Absorption , Administration par voie orale , Animaux , Anthocyanes/sang , Anthocyanes/urine , Biodisponibilité , Relation dose-effet des médicaments , Fragaria/composition chimique , Fruit/composition chimique , Glucuronides/sang , Glucuronides/urine , Spectroscopie par résonance magnétique , Mâle , Rats , Rat Wistar
17.
PLoS Genet ; 8(8): e1002918, 2012.
Article de Anglais | MEDLINE | ID: mdl-22952452

RÉSUMÉ

Abnormal phosphorylation and toxicity of a microtubule-associated protein tau are involved in the pathogenesis of Alzheimer's disease (AD); however, what pathological conditions trigger tau abnormality in AD is not fully understood. A reduction in the number of mitochondria in the axon has been implicated in AD. In this study, we investigated whether and how loss of axonal mitochondria promotes tau phosphorylation and toxicity in vivo. Using transgenic Drosophila expressing human tau, we found that RNAi-mediated knockdown of milton or Miro, an adaptor protein essential for axonal transport of mitochondria, enhanced human tau-induced neurodegeneration. Tau phosphorylation at an AD-related site Ser262 increased with knockdown of milton or Miro; and partitioning defective-1 (PAR-1), the Drosophila homolog of mammalian microtubule affinity-regulating kinase, mediated this increase of tau phosphorylation. Tau phosphorylation at Ser262 has been reported to promote tau detachment from microtubules, and we found that the levels of microtubule-unbound free tau increased by milton knockdown. Blocking tau phosphorylation at Ser262 site by PAR-1 knockdown or by mutating the Ser262 site to unphosphorylatable alanine suppressed the enhancement of tau-induced neurodegeneration caused by milton knockdown. Furthermore, knockdown of milton or Miro increased the levels of active PAR-1. These results suggest that an increase in tau phosphorylation at Ser262 through PAR-1 contributes to tau-mediated neurodegeneration under a pathological condition in which axonal mitochondria is depleted. Intriguingly, we found that knockdown of milton or Miro alone caused late-onset neurodegeneration in the fly brain, and this neurodegeneration could be suppressed by knockdown of Drosophila tau or PAR-1. Our results suggest that loss of axonal mitochondria may play an important role in tau phosphorylation and toxicity in the pathogenesis of AD.


Sujet(s)
Maladie d'Alzheimer , Protéines de Drosophila , Drosophila , Glycogen Synthase Kinase 3 , Protéines G rho , Protéines tau , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Animaux , Animal génétiquement modifié , Axones/métabolisme , Axones/anatomopathologie , Drosophila/génétique , Drosophila/métabolisme , Drosophila/physiologie , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Techniques de knock-down de gènes , Glycogen Synthase Kinase 3/génétique , Glycogen Synthase Kinase 3/métabolisme , Humains , Microtubules/métabolisme , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Dégénérescence nerveuse/métabolisme , Dégénérescence nerveuse/anatomopathologie , Phosphorylation , Protéines G rho/génétique , Protéines G rho/métabolisme , Protéines tau/génétique , Protéines tau/métabolisme
18.
Article de Anglais | MEDLINE | ID: mdl-19965961

RÉSUMÉ

A traditional Japanese herbal (Kampo) medicine, Hochuekkito (Bu-Zhong-Yi-Qi-Tang in Chinese, TJ-41) is a well-known Kampo formula, and has been found to enhance antigen-specific antibody response in not only local mucosal immune system in upper respiratory tract, but also systemic immune system through upper respiratory mucosal immune system. Although this immunopharmacological effect has been proposed to express by modulation of intestinal immune system including Peyer's patches and intestinal epithelial cells, active ingredients are not known. TJ-41 directly affected the production of bone marrow cell-proliferative growth factors from murine Peyer's patch immunocompetent cells in vitro. Among low molecular, intermediate size and macromolecular weight fractions prepared from TJ-41, only fraction containing macromolecular weight ingredients showed Peyer's patch-mediated bone marrow cell-proliferation enhancing activity. Anion-exchange chromatography and gel filtration gave 17 subfractions comprising polysaccharides and lignins from the macromolecular weight fraction of TJ-41, and some of the subfractions showed significant enhancing activities having different degrees. Some of the subfractions also expressed stimulating activity on G-CSF-production from colonic epithelial cells, and statistically significant positive correlation was observed among enhancing activities of the subfractions against Peyer's patch immunocompetent cells and epithelial cells. Among the fractions from TJ-41 oral administration of macromolecular weight ingredient fraction to mice succeeded to enhance antigen-specific antibody response in systemic immune system through upper respiratory mucosal immune system, but all the separated fractions failed to enhance the in vivo antibody response in upper respiratory tract.

19.
Bioorg Med Chem ; 18(8): 2964-75, 2010 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-20363142

RÉSUMÉ

Forty-one derivatives of papyriferic acid were prepared based on our previous finding that methyl papyriferate (3) showed potent reversing effect on cytotoxicity of colchicine against multidrug resistance (MDR) human cancer cells (KB-C2), and evaluated for their cytotoxicity and effect on reversing P-gp-mediated MDR against KB-C2 cells. 3-O-(Morpholino-beta-oxopropanoyl)-12beta-acetoxy-3alpha,25-dihydroxy-(20S,24R)-epoxydammarane (37) significantly increased the sensitivity of colchicine against KB-C2 cells by 185-fold at 5microg/mL (7.4microM), and the cytotoxicity of colchicine was recovered to nearly that of sensitive (KB) cells. The other several new amide derivatives also exhibited potent reversal activity comparable to or more potent than methyl papyriferate and verapamil.


Sujet(s)
Antinéoplasiques/composition chimique , Malonates/composition chimique , Morpholines/composition chimique , Triterpènes/composition chimique , Glycoprotéine P/métabolisme , Antinéoplasiques/synthèse chimique , Antinéoplasiques/toxicité , Lignée cellulaire tumorale , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Humains , Malonates/synthèse chimique , Malonates/toxicité , Morpholines/synthèse chimique , Morpholines/pharmacologie , Triterpènes/synthèse chimique , Triterpènes/pharmacologie , Triterpènes/toxicité
20.
Phytochemistry ; 70(11-12): 1456-61, 2009.
Article de Anglais | MEDLINE | ID: mdl-19766276

RÉSUMÉ

A xanthonolignoid, 2-O-demethylkielcorin, and a phenylxanthone, chinexanthone A, were isolated from stems of Hypericum chinense together with four known xanthonolignoids and seven known xanthones. Their structures were established by spectroscopic analysis, as their optical properties and absolute stereochemistry determined. The cytotoxicities of the isolated xanthone derivatives as well as additional 32 xanthones against a panel of human cancer cell lines were also evaluated.


Sujet(s)
Antinéoplasiques d'origine végétale/isolement et purification , Hypericum/composition chimique , Tumeurs/traitement médicamenteux , Extraits de plantes/composition chimique , Xanthones/isolement et purification , Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques d'origine végétale/usage thérapeutique , Lignée cellulaire tumorale , Humains , Structure moléculaire , Phytothérapie , Extraits de plantes/pharmacologie , Extraits de plantes/usage thérapeutique , Tiges de plante , Xanthones/composition chimique , Xanthones/pharmacologie , Xanthones/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...