Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
EMBO J ; 40(24): e108662, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34825707

RESUMO

Chronic neuroinflammation is a pathogenic component of Alzheimer's disease (AD) that may limit the ability of the brain to clear amyloid deposits and cellular debris. Tight control of the immune system is therefore key to sustain the ability of the brain to repair itself during homeostasis and disease. The immune-cell checkpoint receptor/ligand pair PD-1/PD-L1, known for their inhibitory immune function, is expressed also in the brain. Here, we report upregulated expression of PD-L1 and PD-1 in astrocytes and microglia, respectively, surrounding amyloid plaques in AD patients and in the APP/PS1 AD mouse model. We observed juxtamembrane shedding of PD-L1 from astrocytes, which may mediate ectodomain signaling to PD-1-expressing microglia. Deletion of microglial PD-1 evoked an inflammatory response and compromised amyloid-ß peptide (Aß) uptake. APP/PS1 mice deficient for PD-1 exhibited increased deposition of Aß, reduced microglial Aß uptake, and decreased expression of the Aß receptor CD36 on microglia. Therefore, ineffective immune regulation by the PD-1/PD-L1 axis contributes to Aß plaque deposition during chronic neuroinflammation in AD.


Assuntos
Doença de Alzheimer/imunologia , Precursor de Proteína beta-Amiloide/genética , Antígeno B7-H1/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Regulação para Cima , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Astrócitos/metabolismo , Antígenos CD36/metabolismo , Estudos de Casos e Controles , Modelos Animais de Doenças , Feminino , Deleção de Genes , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Pessoa de Meia-Idade
2.
Biochemistry ; 60(37): 2773-2780, 2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34469142

RESUMO

The prevailing opinion is that prefibrillar ß-amyloid (Aß) species, rather than end-stage amyloid fibrils, cause neuronal dysfunction in Alzheimer's disease, although the mechanisms behind Aß neurotoxicity remain to be elucidated. Luminescent conjugated oligothiophenes (LCOs) exhibit spectral properties upon binding to amyloid proteins and have previously been reported to change the toxicity of Aß1-42 and prion protein. In a previous study, we showed that an LCO, pentamer formyl thiophene acetic acid (p-FTAA), changed the toxicity of Aß1-42. Here we investigated whether an LCO, heptamer formyl thiophene acetic acid (h-FTAA), could change the toxicity of Aß1-42 by comparing its behavior with that of p-FTAA. Moreover, we investigated the effects on toxicity when Aß with the Arctic mutation (AßArc) was aggregated with both LCOs. Cell viability assays on SH-SY5Y neuroblastoma cells demonstrated that h-FTAA has a stronger impact on Aß1-42 toxicity than does p-FTAA. Interestingly, h-FTAA, but not p-FTAA, rescued the AßArc-mediated toxicity. Aggregation kinetics and binding assay experiments with Aß1-42 and AßArc when aggregated with both LCOs showed that h-FTAA and p-FTAA either interact with different species or affect the aggregation in different ways. In conclusion, h-FTAA protects against Aß1-42 and AßArc toxicity, thus showing h-FTAA to be a useful tool for improving our understanding of the process of Aß aggregation linked to cytotoxicity.


Assuntos
Acetatos/química , Precursor de Proteína beta-Amiloide/metabolismo , Tiofenos/química , Acetatos/metabolismo , Amiloide/química , Peptídeos beta-Amiloides/química , Precursor de Proteína beta-Amiloide/fisiologia , Precursor de Proteína beta-Amiloide/toxicidade , Proteínas Amiloidogênicas/química , Corantes Fluorescentes/química , Humanos , Cinética , Luminescência , Fragmentos de Peptídeos/metabolismo , Agregados Proteicos/efeitos dos fármacos , Agregados Proteicos/fisiologia , Coloração e Rotulagem/métodos , Tiofenos/metabolismo
3.
J Neurosci ; 41(24): 5315-5329, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33980545

RESUMO

Impairment of episodic memory, a class of memory for spatiotemporal context of an event, is an early symptom of Alzheimer's disease. Both spatial and temporal information are encoded and represented in the hippocampal neurons, but how these representations are impaired under amyloid ß (Aß) pathology remains elusive. We performed chronic imaging of the hippocampus in awake male amyloid precursor protein (App) knock-in mice behaving in a virtual reality environment to simultaneously monitor spatiotemporal representations and the progression of Aß depositions. We found that temporal representation is preserved, whereas spatial representation is significantly impaired in the App knock-in mice. This is because of the overall reduction of active place cells, but not time cells, and compensatory hyperactivation of remaining place cells near Aß aggregates. These results indicate the differential impact of Aß aggregates on two major modalities of episodic memory, suggesting different mechanisms for forming and maintaining these two representations in the hippocampus.


Assuntos
Doença de Alzheimer/patologia , Região CA1 Hipocampal/patologia , Transtornos da Memória/patologia , Neurônios/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Modelos Animais de Doenças , Masculino , Memória Episódica , Camundongos
4.
Neurobiol Dis ; 147: 105146, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33122075

RESUMO

Pathological accumulations of amyloid-beta (Aß) peptide are found in retina early in Alzheimer's disease, yet its effects on retinal neuronal structure remain unknown. To investigate this, we injected fibrillized Aß1-42 protein into the eye of adult C57BL/6 J mice and analyzed the retina, optic nerve (ON), and the superior colliculus (SC), the primary retinal target in mice. We found that retinal Aß exposure stimulated microglial activation and retinal ganglion cell (RGC) loss as early as 1-week post-injection. Pathology was not limited to the retina, but propagated into other areas of the central nervous system. Microgliosis spread throughout the retinal projection (retina, ON, and SC), with multiplex protein quantitation demonstrating an increase in endogenously produced Aß in the ON and SC corresponding to the injected retinas. Surprisingly, this pathology spread to the opposite side, with unilateral Aß eye injections driving increased Aß levels, neuroinflammation, and RGC death in the opposite, un-injected retinal projection. As Aß-mediated microglial activation has been shown to propagate Aß pathology, we also investigated the role of the Aß-binding microglial scavenger receptor CD36 in this pathology. Transgenic mice lacking the CD36 receptor were resistant to Aß-induced inflammation and RGC death up to 2 weeks following exposure. These results indicate that Aß pathology drives regional neuropathology in the retina and does not remain isolated to the affected eye, but spreads throughout the nervous system. Further, CD36 may serve as a promising target to prevent Aß-mediated inflammatory damage.


Assuntos
Precursor de Proteína beta-Amiloide/toxicidade , Gliose/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Animais , Antígenos CD36/metabolismo , Feminino , Humanos , Injeções Intravítreas , Masculino , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/patologia , Retina/efeitos dos fármacos , Retina/patologia , Colículos Superiores/efeitos dos fármacos , Colículos Superiores/patologia
5.
BMC Mol Cell Biol ; 21(1): 58, 2020 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-32731849

RESUMO

BACKGROUND: Endosomal trafficking and amyloidogenic cleavage of amyloid precursor protein (APP) is believed to play a role in the neurodegeneration observed in Alzheimer's disease (AD). Recent evidence has suggested that packaging and secretion of APP and its amyloidogenic cleaved products into small extracellular vesicles (EVs) may facilitate uptake of these neurotoxic factors during disease progression. However, the molecular mechanisms underlying trafficking of APP into EVs are poorly understood. RESULTS: In this study, the mechanism and impact of APP trafficking into extracellular vesicles (EVs) were assessed by a series of inducible gene knockdowns. We demonstrate that vesicle-associated proteins Alix and Syntenin-1 are essential for proper subcellular localization and efficient EV secretion of APP via an endosomal sorting complexes required for transport (ESCRT)-independent pathway. The neurotoxic C-terminal fragment (CTFß) of APP is similarly secreted in association with small vesicles. These mechanisms are conserved in terminally differentiated neuron-like cells. Furthermore, knockdown of Alix and Syntenin-1 alters the subcellular localization of APP, sequestering the precursor protein to endoplasmic reticulum and endolysosomal compartments, respectively. Finally, transfer of small EVs containing mutant APP confers an increase in reactive oxygen species production and neurotoxicity to human induced pluripotent stem cell-derived cortical neurons and naïve primary neurons, an effect that is ameliorated by Alix and Syntenin-1 depletion. CONCLUSIONS: Altogether these findings elucidate a novel mechanism for understanding the intracellular trafficking of APP and CTFß into secreted extracellular vesicles, and the resultant potential impact on neurotoxicity in the context of Alzheimer's disease amyloidopathy.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Vesículas Extracelulares/metabolismo , Sinteninas/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/ultraestrutura , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotoxinas/toxicidade , Transporte Proteico/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
6.
Brain Res ; 1746: 147022, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32707043

RESUMO

Alzheimer's disease (AD) is the most common cause of dementia. Neuropathological processes, including the accumulation of amyloid-ß (Aß) plaques and neurofibrillary tangles, and neuroinflammation, lead to cognitive impairment at middle and eventually later stages of AD progression. Over the last decade, focused efforts have explored repurposed drug approaches for AD pathophysiological mechanisms. Recently, auranofin, an anti-inflammatory drug, was shown to have therapeutic potential in a number of diseases in addition to rheumatoid arthritis. Surprisingly, no data regarding the effects of auranofin on cognitive deficits in AD mice or the influence of auranofin on Aß pathology and neuroinflammatory processes are available. In the present study, we used 14-month-old transgenic male APPNL-G-F/NL-G-F mice to assess the effects of subchronic administration of auranofin at low doses (1 and 5 mg/kg, intraperitoneal) on spatial memory, Aß pathology and the expression of cortical and hippocampal proteins (glial fibrillary acidic protein (GFAP), ionized calcium binding adaptor molecule-1 (Iba-1)) and proteins related to synaptic plasticity (glutamic acid decarboxylase 67 (GAD67), homer proteins homologue-1 (Homer-1)). The data demonstrated that auranofin significantly decreased Aß deposition in the hippocampus and the number of Aß plaques in the cingulate cortex, but it did not have memory-enhancing effects or induce changes in the expression of the studied proteins. Our current results highlight the importance of considering further pre-clinical research to investigate the possible beneficial effects of auranofin on the other pathological aspects of AD.


Assuntos
Doença de Alzheimer/patologia , Anti-Inflamatórios/farmacologia , Auranofina/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Transgênicos
7.
Cell Rep ; 31(13): 107839, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32610140

RESUMO

Amyloid ß-protein (Aß) toxicity is hypothesized to play a seminal role in Alzheimer's disease (AD) pathogenesis. However, it remains unclear how Aß causes synaptic dysfunction and synapse loss. We hypothesize that one mechanism of Aß-induced synaptic injury is related to the cleavage of amyloid ß precursor protein (APP) at position D664 by caspases that release the putatively cytotoxic C31 peptide. In organotypic slice cultures derived from mice with a knock-in mutation in the APP gene (APP D664A) to inhibit caspase cleavage, Aß-induced synaptic injury is markedly reduced in two models of Aß toxicity. Loss of dendritic spines is also attenuated in mice treated with caspase inhibitors. Importantly, the time-dependent dendritic spine loss is correlated with localized activation of caspase-3 but is absent in APP D664A cultures. We propose that the APP cytosolic domain plays an essential role in Aß-induced synaptic damage in the injury pathway mediated by localized caspase activation.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Caspase 3/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Inibidores de Caspase/farmacologia , Espinhas Dendríticas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Técnicas de Introdução de Genes , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Mutação/genética , Neuroproteção/efeitos dos fármacos
8.
Folia Neuropathol ; 58(1): 10-21, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32337953

RESUMO

INTRODUCTION: Alzheimer's disease (AD) is still the fifth leading cause of death and most common dementia worldwide. To date, there is no efficient strategy that can slow down the progression of AD owing to delayed diagnosis and limited therapies. MiR-143-3p is up-regulated in serum of AD patients, yet the exact role it plays in AD pathology is still poorly understood. The aim of this study was to investigate the effect of miR-143-3p on neuronal survival. MATERIAL AND METHODS: We induced neuronal differentiation in SH-SY5Y cells using all-trans-retinoic acid (RA), and Aß1-42 was used to establish the in vitro AD cell model. The expression of tubulin ß III and neuregulin-1 (NRG1) was evaluated by immunofluorescence. TUNEL assay was performed to assess cell apoptosis. Cell viability was evaluated using the Cell Counting Kit-8 assay. The binding interaction between miR-143-3p and NRG1 was verified using the luciferase reporter assay. RESULTS: Typical neuronal-like axons were observed in RA-induced SH-SY5Y cells, followed by increased tubulin ß III. A dramatically increased apoptotic rate and reduced cell viability were observed in the AD cell model. Then we silenced the miR-143-3p expression, and Aß1-42 induced cell apoptosis was alleviated after miR-143-3p inhibition, accompanied by decreased cleaved caspase-3 and cleaved caspase-9 levels. Additionally, NRG1 was confirmed to be a downstream target of miR-143-3p, increased cell viability and suppressed cell apoptosis after miR-143-3p inhibition was abolished by NRG1 knockdown. CONCLUSIONS: Our findings reveal that miR-143-3p inhibition promotes neuronal survival in an in vitro cell model via targeting NRG1, and the miR-143-3p/NRG1 axis is a potential therapeutic target and promising biomarker for AD treatment.


Assuntos
Doença de Alzheimer , MicroRNAs/metabolismo , Neuregulina-1/metabolismo , Neurônios/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Humanos , Neurônios/patologia
9.
Oxid Med Cell Longev ; 2020: 3515609, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33425207

RESUMO

BACKGROUND: Acorus tatarinowii Schott [Shi Chang Pu in Chinese (SCP)] is a traditional Chinese medicine frequently used in the clinical treatment of dementia, amnesia, epilepsy, and other mental disorders. Previous studies have shown the potential efficacy of SCP against Alzheimer's disease (AD). Nevertheless, the active constituents and the modes of action of SCP in AD treatment have not been fully elucidated. PURPOSE: The aim of this study was to investigate the protective effects of SCP on abnormal proteins and clarify its molecular mechanisms in the treatment of AD by using a Caenorhabditis elegans (C. elegans) model. METHODS: This study experimentally assessed the effect of SCP-Oil in CL4176 strains expressing human Aß in muscle cells and CL2355 strains expressing human Aß in pan-neurons. Western blotting, qRT-PCR, and fluorescence detection were performed to determine the oxidative stress and signaling pathways affected by SCP-Oil in nematodes. RESULTS: SCP-Oil could significantly reduce the deposition of misfolded Aß and polyQ proteins and improved serotonin sensitivity and olfactory learning skill in worms. The analysis of pharmacological action mechanism of SCP-Oil showed that its maintaining protein homeostasis is dependent on the autophagy pathway regulated partly by hsf-1 and sir-2.1 genes. CONCLUSION: Our results provide new insights to develop treatment strategy for AD by targeting autophagy, and SCP-Oil could be an alternative drug for anti-AD.


Assuntos
Acorus/metabolismo , Precursor de Proteína beta-Amiloide/biossíntese , Precursor de Proteína beta-Amiloide/toxicidade , Autofagia/efeitos dos fármacos , Caenorhabditis elegans/efeitos dos fármacos , Óleos Voláteis/farmacologia , Extratos Vegetais/farmacologia , Doença de Alzheimer/tratamento farmacológico , Animais , Quimiotaxia , Cromatografia Líquida de Alta Pressão , Modelos Animais de Doenças , Homeostase , Peptídeos/química , Dobramento de Proteína , Transdução de Sinais , Especificidade da Espécie
10.
Aging Cell ; 19(2): e13070, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31777182

RESUMO

Dysregulation of autophagy is associated with the neurodegenerative processes in Alzheimer's disease (AD), yet it remains controversial whether autophagy is a cause or consequence of AD. We have previously expressed the full-length human APP in Drosophila and established a fly AD model that exhibits multiple AD-like symptoms. Here we report that depletion of CHIP effectively palliated APP-induced pathological symptoms, including morphological, behavioral, and cognitive defects. Mechanistically, CHIP is required for APP-induced autophagy dysfunction, which promotes Aß production via increased expression of BACE and Psn. Our findings suggest that aberrant autophagy is not only a consequence of abnormal APP activity, but also contributes to dysregulated APP metabolism and subsequent AD pathogenesis.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Autofagia/genética , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/metabolismo , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Regulação para Baixo , Proteínas de Drosophila/genética , Olho/crescimento & desenvolvimento , Olho/metabolismo , Deficiências da Aprendizagem/genética , Locomoção/genética , Longevidade/genética , Proteínas Nucleares/genética , Presenilinas/metabolismo , Interferência de RNA , Asas de Animais/metabolismo , Asas de Animais/patologia
11.
Sci Rep ; 9(1): 5476, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30940901

RESUMO

The aggregation of insoluble amyloid beta (Aß) peptides in the brain is known to trigger the onset of neurodegenerative diseases, such as Alzheimer's disease. In spite of the massive number of investigations, the underlying mechanisms to destabilize the Aß aggregates are still poorly understood. Some studies indicate the importance of oxidation to destabilize the Aß aggregates. In particular, oxidation induced by cold atmospheric plasma (CAP) has demonstrated promising results in eliminating these toxic aggregates. In this paper, we investigate the effect of oxidation on the stability of an Aß pentamer. By means of molecular dynamics simulations and umbrella sampling, we elucidate the conformational changes of Aß pentamer in the presence of oxidized residues, and we estimate the dissociation free energy of the terminal peptide out of the pentamer form. The calculated dissociation free energy of the terminal peptide is also found to decrease with increasing oxidation. This indicates that Aß pentamer aggregation becomes less favorable upon oxidation. Our study contributes to a better insight in one of the potential mechanisms for inhibition of toxic Aß peptide aggregation, which is considered to be the main culprit to Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/toxicidade , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Oxirredução , Agregados Proteicos , Conformação Proteica , Multimerização Proteica , Estabilidade Proteica
12.
Curr Alzheimer Res ; 16(5): 453-457, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30907322

RESUMO

BACKGROUND: Alzheimer's disease (AD) is associated with extracellular accumulation and aggregation of amyloid ß (Aß) peptides ultimately seeding in senile plaques. Recent data show that their direct precursor C99 (ßCTF) also accumulates in AD-affected brain as well as in AD-like mouse models. C99 is consistently detected much earlier than Aß, suggesting that this metabolite could be an early contributor to AD pathology. C99 accumulates principally within endolysosomal and autophagic structures and its accumulation was described as both a consequence and one of the causes of endolysosomalautophagic pathology, the occurrence of which has been documented as an early defect in AD. C99 was also accompanied by C99-derived C83 (αCTF) accumulation occurring within the same intracellular organelles. Both these CTFs were found to dimerize leading to the generation of higher molecular weight CTFs, which were immunohistochemically characterized in situ by means of aggregate-specific antibodies. DISCUSSION: Here, we discuss studies demonstrating a direct link between the accumulation of C99 and C99-derived APP-CTFs and early neurotoxicity. We discuss the role of C99 in endosomal-lysosomalautophagic dysfunction, neuroinflammation, early brain network alterations and synaptic dysfunction as well as in memory-related behavioral alterations, in triple transgenic mice as well as in newly developed AD animal models. CONCLUSION: This review summarizes current evidence suggesting a potential role of the ß -secretasederived APP C-terminal fragment C99 in Alzheimer's disease etiology.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/toxicidade , Doença de Alzheimer/metabolismo , Animais , Humanos
13.
J Neurochem ; 149(4): 499-517, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30040874

RESUMO

Changes in glycerophosphocholine metabolism are observed in Alzheimer's disease; however, it is not known whether these metabolic disruptions are linked to cognitive decline. Here, using unbiased lipidomic approaches and direct biochemical assessments, we profiled Land's cycle lipid remodeling in the hippocampus, frontal cortex, and temporal-parietal-entorhinal cortices of human amyloid beta precursor protein (ΑßPP) over-expressing mice. We identified a cortex-specific hypo-metabolic signature at symptomatic onset and a cortex-specific hyper-metabolic signature of Land's cycle glycerophosphocholine remodeling over the course of progressive behavioral decline. When N5 TgCRND8 and ΑßPPSwe /PSIdE9 mice first exhibited deficits in the Morris Water Maze, levels of lyso-phosphatidylcholines, LPC(18:0/0:0), LPC(16:0/0:0), LPC(24:6/0:0), LPC(25:6/0:0), the lyso-platelet-activating factor (PAF), LPC(O-18:0/0:0), and the PAF, PC(O-22:6/2:0), declined as a result of reduced calcium-dependent cytosolic phospholipase A2 α (cPLA2 α) activity in all cortices but not hippocampus. Chronic intermittent hypoxia, an environmental risk factor that triggers earlier learning memory impairment in ΑßPPSwe /PSIdE9 mice, elicited these same metabolic changes in younger animals. Thus, this lipidomic signature of phenoconversion appears age-independent. By contrast, in symptomatic N5 TgCRND8 mice, cPLA2 α activity progressively increased; overall Lyso-phosphatidylcholines (LPC) and LPC(O) and PC(O-18:1/2:0) levels progressively rose. Enhanced cPLA2 α activity was only detected in transgenic mice; however, age-dependent increases in the PAF acetylhydrolase 1b α1 to α2 expression ratio, evident in both transgenic and non-transgenic mice, reduced PAF hydrolysis thereby contributing to PAF accumulation. Taken together, these data identify distinct age-independent and age-dependent disruptions in Land's cycle metabolism linked to symptomatic onset and progressive behavioral decline in animals with pre-existing Αß pathology. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Assuntos
Doença de Alzheimer/metabolismo , Córtex Cerebral/metabolismo , Fosfatidilcolinas/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Modelos Animais de Doenças , Progressão da Doença , Humanos , Camundongos
14.
Mol Neurobiol ; 55(10): 8103-8123, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29508282

RESUMO

Hyperactivated microglia plays a key role in regulating neuroinflammatory responses which cause damage to neurons. In recent years, substantial attention has been paid in identifying new strategies to abrogate neuroinflammation. Tiliroside, a natural dietary glycosidic flavonoid, is known to inhibit neuroinflammation. This study was aimed at investigating the molecular mechanisms involved in the inhibition of neuroinflammation and neurotoxicity by tiliroside. The effects of tiliroside on Nrf2 and SIRT1 activities in BV2 microglia and HT22 hippocampal neurons were investigated using immunoblotting and DNA binding assays. The roles of Nrf2 and SIRT1 in the anti-inflammatory activity of tiliroside were further investigated using RNA interference experiments. HT22 neuronal viability was determined by XTT, calcium influx, DNA fragmentation assays. The effect of tiliroside on MAP2 protein expression in HT22 neurons was investigated using western blotting and immunofluorescence. We also studied the impact of tiliroside on DNA fragmentation and ROS generation in APPSwe-transfected 3D neuronal stem cells. Results show that tiliroside increased protein levels of Nrf2, HO-1 and NQO1, indicating an activation of the Nrf2 protective mechanisms in the microglia. Furthermore, transfection of BV2 cells with Nrf2 siRNA resulted in the loss of anti-inflammatory activity by tiliroside. Tiliroside reduced protein levels of acetylated-NF-κB-p65, and increased SIRT1 in LPS/IFNγ-activated BV2 microglia. RNAi experiments revealed that inhibition of neuroinflammation by tiliroside was not affected by silencing SIRT1 gene. Results of neurotoxicity experiments revealed that neuroinflammation-induced toxicity, DNA fragmentation, ROS generation and calcium accumulation in HT22 neurons were significantly reduced by tiliroside treatment. In addition, the compound also protected differentiated human neural progenitor cells by blocking ROS generation and DNA fragmentation. Overall, this study has established that tiliroside protected BV2 microglia from LPS/IFNγ-induced neuroinflammation and HT22 neuronal toxicity by targeting Nrf2 antioxidant mechanisms. The compound also produced inhibition of NF-κB acetylation through activation of SIRT1, as well as increasing SIRT1 activity in mouse hippocampal neurons. Results from this study have further established the mechanisms involved in the anti-neuroinflammatory and neuroprotective activities of tiliroside.


Assuntos
Dieta , Flavonoides/farmacologia , Fator 2 Relacionado a NF-E2/farmacologia , Neuroproteção/efeitos dos fármacos , Transdução de Sinais , Acetilação , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Elementos de Resposta Antioxidante/genética , Antioxidantes/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Citocinas/metabolismo , Heme Oxigenase-1/metabolismo , Humanos , Inflamação/patologia , Interferon gama/metabolismo , Lipopolissacarídeos , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , NF-kappa B/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/toxicidade , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/metabolismo , Regulação para Cima/efeitos dos fármacos
15.
J Exp Med ; 214(12): 3669-3685, 2017 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-29114064

RESUMO

Sortilin-related receptor with LDLR class A repeats (SORLA, SORL1, or LR11) is a genetic risk factor associated with Alzheimer's disease (AD). Although SORLA is known to regulate trafficking of the amyloid ß (Aß) precursor protein to decrease levels of proteotoxic Aß oligomers, whether SORLA can counteract synaptic dysfunction induced by Aß oligomers remains unclear. Here, we show that SORLA interacts with the EphA4 receptor tyrosine kinase and attenuates ephrinA1 ligand-induced EphA4 clustering and activation to limit downstream effects of EphA4 signaling in neurons. Consistent with these findings, SORLA transgenic mice, compared with WT mice, exhibit decreased EphA4 activation and redistribution to postsynaptic densities, with milder deficits in long-term potentiation and memory induced by Aß oligomers. Importantly, we detected elevated levels of active EphA4 in human AD brains, where EphA4 activation is inversely correlated with SORLA/EphA4 association. These results demonstrate a novel role for SORLA as a physiological and pathological EphA4 modulator, which attenuates synaptotoxic EphA4 activation and cognitive impairment associated with Aß-induced neurodegeneration in AD.


Assuntos
Precursor de Proteína beta-Amiloide/toxicidade , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Degeneração Neural/patologia , Receptor EphA4/metabolismo , Receptores de LDL/metabolismo , Doença de Alzheimer/patologia , Animais , Efrinas/farmacologia , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/metabolismo , Células HEK293 , Humanos , Ligantes , Potenciação de Longa Duração/efeitos dos fármacos , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Mutação/genética , Degeneração Neural/metabolismo , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Receptores de LDL/química , Receptores de LDL/genética , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia
16.
Biomed Res Int ; 2017: 7464872, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29057264

RESUMO

Icariin (ICA), a prenylated flavanol glycoside present in abundant quantities in Epimedium sagittatum, has shown promise in the treatment and prevention of Alzheimer's disease. Damage to synaptic plasticity induced by amyloid-beta-mediated neurotoxicity is considered a main pathological mechanism driving the learning and memory deficits present in patients with Alzheimer's disease. This study investigated the neuroprotective effects of icariin in an Aß1-42-induced rat model of Alzheimer's disease. Our results showed that Aß1-42 injection induced loss of learning and memory behaviour in the Morris water maze, which could be reversed with intragastric administration of ICA. Furthermore, ICA reversed decreases in PSD-95, BDNF, pTrkB, pAkt, and pCREB expressions and prevented deterioration of synaptic interface structure. These findings indicate that ICA may improve synaptic plasticity through the BDNF/TrkB/Akt pathway and provide further evidence for its clinical application to improve learning and memory in patients with Alzheimer's disease.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Disfunção Cognitiva/tratamento farmacológico , Flavonoides/administração & dosagem , Transtornos da Memória/tratamento farmacológico , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Disfunção Cognitiva/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Transtornos da Memória/patologia , Plasticidade Neuronal/efeitos dos fármacos , Fármacos Neuroprotetores , Fragmentos de Peptídeos/toxicidade , Ratos
17.
Neurosci Bull ; 33(4): 405-412, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28555357

RESUMO

Accumulation and aggregation of ß-amyloid (Aß) peptides result in neuronal death, leading to cognitive dysfunction in Alzheimer's disease. The self-assembled Aß molecules form various intermediate aggregates including oligomers that are more toxic to neurons than the mature aggregates, including fibrils. Thus, one strategy to alleviate Aß toxicity is to facilitate the conversion of Aß intermediates to larger aggregates such as fibrils. In this study, we designed a peptide named A3 that significantly enhanced the formation of amorphous aggregates of Aß by accelerating the aggregation kinetics. Thioflavin T fluorescence experiments revealed an accelerated aggregation of Aß monomers, accompanying reduced Aß cytotoxicity. Transgenic Caenorhabditis elegans over-expressing amyloid precursor protein exhibited paralysis due to the accumulation of Aß oligomers, and this phenotype was attenuated by feeding the animals with A3 peptide. These findings suggest that the Aß aggregation-promotion effect can potentially be useful for developing strategies to reduce Aß toxicity.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/induzido quimicamente , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/ultraestrutura , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Cinética , Neuroblastoma/patologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/ultraestrutura , Agregação Patológica de Proteínas/genética , Transfecção
18.
Sci Rep ; 6: 35865, 2016 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-27775057

RESUMO

The pathological hallmark of misfolded protein diseases and aging is the accumulation of proteotoxic aggregates. However, the mechanisms of proteotoxicity and the dynamic changes in fiber formation and dissemination remain unclear, preventing a cure. Here we adopted a reductionist approach and used atomic force microscopy to define the temporal and spatial changes of amyloid aggregates, their modes of dissemination and the biochemical changes that may influence their growth. We show that pre-amyloid oligomers (PAO) mature to form linear and circular protofibrils, and amyloid fibers, and those can break reforming PAO that can migrate invading neighbor structures. Simulating the effect of immunotherapy modifies the dynamics of PAO formation. Anti-fibers as well as anti-PAO antibodies fragment the amyloid fibers, however the fragmentation using anti-fibers antibodies favored the migration of PAO. In conclusion, we provide evidence for the mechanisms of misfolded protein maturation and propagation and the effects of interventions on the resolution and dissemination of amyloid pathology.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Agregados Proteicos , Agregação Patológica de Proteínas , Dobramento de Proteína , Multimerização Proteica , Humanos , Cinética , Microscopia de Força Atômica , Análise Espaço-Temporal
19.
Hum Mol Genet ; 24(21): 6093-105, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26253732

RESUMO

Both active and passive immunotherapy protocols decrease insoluble amyloid-ß42 (Aß42) peptide in animal models, suggesting potential therapeutic applications against the main pathological trigger in Alzheimer's disease (AD). However, recent clinical trials have reported no significant benefits from humanized anti-Aß42 antibodies. Engineered single-chain variable fragment antibodies (scFv) are much smaller and can easily penetrate the brain, but identifying the most effective scFvs in murine AD models is slow and costly. We show here that scFvs against the N- and C-terminus of Aß42 (scFv9 and scFV42.2, respectively) that decrease insoluble Aß42 in CRND mice are neuroprotective in Drosophila models of Aß42 and amyloid precursor protein neurotoxicity. Both scFv9 and scFv42.2 suppress eye toxicity, reduce cell death in brain neurons, protect the structural integrity of dendritic terminals in brain neurons and delay locomotor dysfunction. Additionally, we show for the first time that co-expression of both anti-Aß scFvs display synergistic neuroprotective activities, suggesting that combined therapies targeting distinct Aß42 epitopes can be more effective than targeting a single epitope. Overall, we demonstrate the feasibility of using Drosophila as a first step for characterizing neuroprotective anti-Aß scFvs in vivo and identifying scFv combinations with synergistic neuroprotective activities.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/imunologia , Modelos Animais de Doenças , Drosophila , Fragmentos de Peptídeos/imunologia , Anticorpos de Cadeia Única/imunologia , Doença de Alzheimer/imunologia , Doença de Alzheimer/prevenção & controle , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Animais Geneticamente Modificados , Encéfalo/metabolismo , Encéfalo/patologia , Olho Composto de Artrópodes/imunologia , Avaliação Pré-Clínica de Medicamentos/métodos , Sinergismo Farmacológico , Feminino , Masculino , Atividade Motora , Neurônios/metabolismo , Proteínas Recombinantes
20.
Adv Exp Med Biol ; 855: 67-94, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26149926

RESUMO

Aggregation of amyloid-beta (Aß) peptide is the major event underlying neuronal damage in Alzheimer's disease (AD). Specific lipids and their homeostasis play important roles in this and other neurodegenerative disorders. The complex interplay between the lipids and the generation, clearance or deposition of Aß has been intensively investigated and is reviewed in this chapter. Membrane lipids can have an important influence on the biogenesis of Aß from its precursor protein. In particular, increased cholesterol in the plasma membrane augments Aß generation and shows a strong positive correlation with AD progression. Furthermore, apolipoprotein E, which transports cholesterol in the cerebrospinal fluid and is known to interact with Aß or compete with it for the lipoprotein receptor binding, significantly influences Aß clearance in an isoform-specific manner and is the major genetic risk factor for AD. Aß is an amphiphilic peptide that interacts with various lipids, proteins and their assemblies, which can lead to variation in Aß aggregation in vitro and in vivo. Upon interaction with the lipid raft components, such as cholesterol, gangliosides and phospholipids, Aß can aggregate on the cell membrane and thereby disrupt it, perhaps by forming channel-like pores. This leads to perturbed cellular calcium homeostasis, suggesting that Aß-lipid interactions at the cell membrane probably trigger the neurotoxic cascade in AD. Here, we overview the roles of specific lipids, lipid assemblies and apolipoprotein E in Aß processing, clearance and aggregation, and discuss the contribution of these factors to the neurotoxicity in AD.


Assuntos
Precursor de Proteína beta-Amiloide/química , Lipídeos/química , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/toxicidade , Apolipoproteínas E/metabolismo , Humanos , Lipídeos/toxicidade , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...