Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Psychiatry ; 28(11): 4655-4665, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37730843

RESUMEN

Social hierarchy has a profound impact on social behavior, reward processing, and mental health. Moreover, lower social rank can lead to chronic stress and often more serious problems such as bullying victims of abuse, suicide, or attack to society. However, its underlying mechanisms, particularly their association with glial factors, are largely unknown. In this study, we report that astrocyte-derived amphiregulin plays a critical role in the determination of hierarchical ranks. We found that astrocytes-secreted amphiregulin is directly regulated by cAMP response element-binding (CREB)-regulated transcription coactivator 3 (CRTC3) and CREB. Mice with systemic and astrocyte-specific CRTC3 deficiency exhibited a lower social rank with reduced functional connectivity between the prefrontal cortex, a major social hierarchy center, and the parietal cortex. However, this effect was reversed by astrocyte-specific induction of amphiregulin expression, and the epidermal growth factor domain was critical for this action of amphiregulin. These results provide evidence of the involvement of novel glial factors in the regulation of social dominance and may shed light on the clinical application of amphiregulin in the treatment of various psychiatric disorders.


Asunto(s)
Transducción de Señal , Factores de Transcripción , Animales , Ratones , Anfirregulina/genética , Ratones Noqueados , Predominio Social , Factores de Transcripción/metabolismo
2.
Cell Biosci ; 13(1): 181, 2023 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-37773139

RESUMEN

BACKGROUNDS: The expression of major histocompatibility complex I (MHC-I) in neurons has recently been shown to regulate neurite outgrowth and synaptic plasticity. However, its contribution to neurodegenerative diseases such as Alzheimer's disease (AD) remains largely unknown. METHODS: In this study, we investigated the relationship between impaired MHC-I-ß2M complex and AD in vitro and human AD samples. Interaction between protein was identified by liquid chromatography-tandem mass spectrometry and confirmed by immunoprecipitation. Single-chain trimer of MHC-I-ß2M was generated to study the effect of stabilization of MHC-I-ß2M complex on NCAM1 signaling. RESULTS: MHC-I is destabilized in the brains of AD patients and neuronal cells treated with oligomeric ß-amyloid (Aß). Specifically, Aß oligomers disassemble the MHC-I-ß2-microglobulin (ß2M) complex, leading to reduced interactions with neural cell adhesion molecule 1 (NCAM1), a novel interactor of neuronal MHC-I, and decreased signaling. Inhibition of MHC-I-ß2M complex destabilization by non-dissociable MHC-I-ß2M-peptide complex restored MHC-I-NCAM1 signaling in neuronal cells. CONCLUSIONS: The current study demonstrated that disruption of MHC-1-NCAM1 signaling by Aß induced disassembly of MHC-I-ß2M complex is involved in the pathophysiology of AD. Moreover, our findings suggest modulation of MHC-I stability may be a potential therapeutic target for restoring synaptic function in AD.

3.
J Clin Invest ; 133(8)2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-36917188

RESUMEN

The spatiotemporal pattern of the spread of pathologically modified tau through brain regions in Alzheimer's disease (AD) can be explained by prion-like cell-to-cell seeding and propagation of misfolded tau aggregates. Hence, to develop targeted therapeutic antibodies, it is important to identify the seeding- and propagation-competent tau species. The hexapeptide 275VQIINK280 of tau is a critical region for tau aggregation, and K280 is acetylated in various tauopathies, including AD. However, the mechanism that links tau acetylated on lysine 280 (tau-acK280) to subsequent progression to neurodegenerative disease remains unclear. Here, we demonstrate that tau-acK280 is critical for tau propagation processes including secretion, aggregation, and seeding. We developed an antibody, Y01, that specifically targets tau-acK280 and solved the crystal structure of Y01 in complex with an acK280 peptide. The structure confirmed that Y01 directly recognizes acK280 and the surrounding residues. Strikingly, upon interaction with acetylated tau aggregates, Y01 prevented tauopathy progression and increased neuronal viability in neuron cultures and in tau-Tg mice through antibody-mediated neutralization and phagocytosis, respectively. Based on our observations that tau-acK280 is a core species involved in seeding and propagation activities, the Y01 antibody that specifically recognizes acK280 represents a promising therapeutic candidate for AD and other neurodegenerative diseases associated with tauopathy.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Tauopatías , Ratones , Animales , Anticuerpos Monoclonales/farmacología , Proteínas tau/genética , Proteínas tau/metabolismo , Lisina , Tauopatías/tratamiento farmacológico , Modelos Animales de Enfermedad , Encéfalo/metabolismo
4.
Int J Neurosci ; 133(12): 1394-1398, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35603448

RESUMEN

PURPOSE/AIM OF THE STUDY: Accumulation of hyperphosphorylated tau is a key pathological finding of Alzheimer's disease. Recently, acetylation of tau is emerging as another key pathogenic modification, especially regarding the acetylation of tau at K280 of the hexapeptide 275VQIINK280, a critical sequence in driving tau aggregation. However, the relationship between these two key post-translational modifications is not well known. In this study, effect of acetylation of tau at K280 on tau phosphorylation profile was investigated. MATERIALS AND METHODS: The human neuroblastoma cell line, SH-SY5Y, was transfected with p300 acetyltransferase and tau to induce acetylation of tau. Phosphorylation profile after acetylation was evaluated on western blot. K280A-mutant tau was transfected to investigate the effect of acetylation of tau at K280 on tau phosphorylation profile. RESULTS: Overexpression of p300 acetyltransferase in tau-transfected SH-SY5Y human neuroblastoma cells increased acetylation of tau. Meanwhile, tau and its phosphorylation also increased at various sites such as S199/202, S202/T205, T231, and S422, but not at S396. However, blocking acetylation only at K280 with K280A-mutant tau reversed the increased phosphorylation of tau at S202/T205, T231, and S422, but not at S199/202 or S396. CONCLUSION: Here we identified tau phosphorylation profile in the context of p300-induced acetylation and K280A-mutant tau, demonstrating that tau acetylation affects phosphorylation differently by residues and that acetylation at K280 is a determinant of phosphorylation at some residues in the context of pathologic acetyltransferase activity. Yet, our results suggest there is a complex interplay yet to be explored between tau acetylation with tau phosphorylation.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Humanos , Fosforilación , Proteínas tau/metabolismo , Acetilación , Procesamiento Proteico-Postraduccional , Enfermedad de Alzheimer/metabolismo
5.
Biochem Biophys Res Commun ; 524(3): 764-771, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32037089

RESUMEN

ß-Amyloid (Aß) plaque in the brains of patients with Alzheimer's disease (AD) is mainly caused by impaired clearance of Aß by glial cells, including microglia and astrocytes. Because microglia play an important protective role in the central nervous system, many efforts have been made to identify agents that effectively improve microglial Aß phagocytosis. This study found that TLQP-21, which is cleaved from VGF (VGF nerve growth factor inducible) precursor protein, enhanced Aß phagocytosis and degradation by microglial BV2 cells. TLQP-21 also improved microglial phagocytic activity and promoted fibrillar amyloid-ß (fAß) uptake by microglial BV2 cells via a C3AR1-dependent mechanism. Moreover, TLQP-21 stimulated Aß degradation by enhancing lysosome activity, thereby enhancing fAß clearance. These results suggest that treatment with TLQP-21 may be a novel therapeutic strategy to efficiently enhance microglial Aß clearance in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Espacio Extracelular/metabolismo , Microglía/metabolismo , Fragmentos de Péptidos/farmacología , Amiloide/efectos de los fármacos , Animales , Línea Celular , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Microglía/efectos de los fármacos , Neuropéptidos/farmacología , Fagocitosis/efectos de los fármacos , Proteolisis/efectos de los fármacos , Receptores de Complemento/metabolismo
6.
Aging Cell ; 19(2): e13078, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31800167

RESUMEN

A promising new therapeutic target for the treatment of Alzheimer's disease (AD) is the circadian system. Although patients with AD are known to have abnormal circadian rhythms and suffer sleep disturbances, the role of the molecular clock in regulating amyloid-beta (Aß) pathology is still poorly understood. Here, we explored how the circadian repressors REV-ERBα and ß affected Aß clearance in mouse microglia. We discovered that, at Circadian time 4 (CT4), microglia expressed higher levels of the master clock protein BMAL1 and more rapidly phagocytosed fibrillary Aß1-42 (fAß1-42 ) than at CT12. BMAL1 directly drives transcription of REV-ERB proteins, which are implicated in microglial activation. Interestingly, pharmacological inhibition of REV-ERBs with the small molecule antagonist SR8278 or genetic knockdown of REV-ERBs-accelerated microglial uptake of fAß1-42 and increased transcription of BMAL1. SR8278 also promoted microglia polarization toward a phagocytic M2-like phenotype with increased P2Y12 receptor expression. Finally, constitutive deletion of Rev-erbα in the 5XFAD model of AD decreased amyloid plaque number and size and prevented plaque-associated increases in disease-associated microglia markers including TREM2, CD45, and Clec7a. Altogether, our work suggests a novel strategy for controlling Aß clearance and neuroinflammation by targeting REV-ERBs and provides new insights into the role of REV-ERBs in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Relojes Circadianos/genética , Microglía/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Placa Amiloide/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción ARNTL/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/síntesis química , Animales , Proteínas CLOCK/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Isoquinolinas/farmacología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Placa Amiloide/genética , Placa Amiloide/patología , ARN Interferente Pequeño , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores Purinérgicos P2Y12/efectos de los fármacos , Receptores Purinérgicos P2Y12/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Sinapsis/genética , Sinapsis/metabolismo , Tiofenos/farmacología
7.
Neurobiol Dis ; 130: 104519, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31233882

RESUMEN

The intraneuronal aggregates of hyperphosphorylated and misfolded tau (neurofibrillary tangles, NFTs) cause a stereotypical spatiotemporal Alzheimer's disease (AD) progression that correlates with the severity of the associated cognitive decline. Kinase activity contributes to the balance between neuron survival and cell death. Hyperactivation of kinases including the conventional protein kinase C (PKC) is a defective molecular event accompanying associative memory loss, tau phosphorylation, and progression of AD or related neurodegenerative diseases. Here, we investigated the ability of small therapeutic compounds (a custom library) to improve tau-induced rough-eye phenotype in a Drosophila melanogaster model of frontotemporal dementia. We also assessed the tau phosphorylation in vivo and selected hit compounds. Among the potential hits, we investigated Ro 31-8220, described earlier as a potent PKCα inhibitor. Ro 31-8220 robustly improved the rough-eye phenotype, reduced phosphorylated tau species in vitro and in vivo, reversed tau-induced memory impairment, and improved the fly motor functions. In a human neuroblastoma cell line, Ro 31-8220 reduced the PKC activity and the tau phosphorylation pattern, but we also have to acknowledge the compound's wide range of biological activity. Nevertheless, Ro 31-8220 is a novel therapeutic mitigator of tau-induced neurotoxocity.


Asunto(s)
Demencia Frontotemporal/metabolismo , Indoles/farmacología , Ovillos Neurofibrilares/efectos de los fármacos , Neuronas/efectos de los fármacos , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Drosophila melanogaster , Evaluación Preclínica de Medicamentos , Ovillos Neurofibrilares/metabolismo , Neuronas/metabolismo , Fosforilación/efectos de los fármacos
8.
Neurobiol Dis ; 129: 182-194, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31121321

RESUMEN

The link between Val232Met variant of phospholipase D3 (PLD3) and late-onset Alzheimer's disease (AD) is still obscure. While it may not affect directly the amyloid precursor protein function, PLD3 could be regulating multiple cellular compartments. Here, we investigated the function of wild-type human PLD3 (PLD3WT) and the Val232Met variant (PLD3VM) in the presence of ß-amyloid (Aß) in a Drosophila melanogaster model of AD. We expressed PLD3WT in CNS of the Aß-model flies and monitored its effect on the ER stress, cell apoptosis and recovery the Aß-induced cognitive impairment. The expression reduced ER stress and neuronal apoptosis, which resulted in normalized antioxidative phospholipids levels and brain protection. A specific O-glycosylation at pT271 in PLD3 is essential for its normal trafficking and cellular localization. The V232 M substitution impairs this O-glycosylation, leading to enlarged lysosomes and plausibly aberrant protein recycling. PLD3VM was less neuroprotective, and while, PLD3WT expression enhances the lysosomal functions, V232 M attenuated PLD3's trafficking to the lysosomes. Thus, the V232 M mutation may affect AD pathogenesis. Further understanding of the mechanistic role of PLD3 in AD could lead to developing novel therapeutic agents.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Neuroprotección/fisiología , Fosfolipasa D/genética , Fosfolipasa D/metabolismo , Animales , Animales Modificados Genéticamente , Drosophila melanogaster , Predisposición Genética a la Enfermedad , Glicosilación , Humanos , Mutación , Neuronas/metabolismo , Neuronas/patología , Transporte de Proteínas
9.
Mol Cell Neurosci ; 95: 13-24, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30594669

RESUMEN

The number of neurofibrillary tangles containing abnormal hyperphosphorylated tau protein correlates with the degree of dementia in Alzheimer's disease (AD). In addition, autophagosome accumulation and disturbance of autophagy, the process by which toxic aggregate proteins are degraded in the cytosol, are also found in AD models. These indicate that regulation of the autophagy-lysosome system may be a potential therapeutic target for AD. Activation of transcription factor EB (TFEB), a master regulator of autophagy-lysosome system gene transcription, reduces the amount of tau in APP mice. Here, to identify potential therapeutic compounds for AD, we performed two types of screening to determine pharmacologically active compounds that increase 1) neuronal viability in okadaic acid-induced tau hyperphosphorylation-related neurodegeneration models and 2) nuclear localization of TFEB in high-contents screening. Ouabain, a cardiac glycoside, was discovered as a common hit compound in both screenings. It also exhibited a significant protective effect in tau transgenic fly and mouse models in vivo. This work demonstrates that ouabain enhances activation of TFEB through inhibition of the mTOR pathway and induces downstream autophagy-lysosomal gene expression and cellular restorative properties. Therefore, therapeutic approaches using ouabain reduce the accumulation of abnormal toxic tau in vitro and in vivo.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Ouabaína/farmacología , Animales , Células Cultivadas , Drosophila melanogaster , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
10.
J Allergy Clin Immunol ; 141(3): 938-950.e12, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29225084

RESUMEN

BACKGROUND: Eosinophilic inflammation is a major pathologic feature of chronic rhinosinusitis (CRS) and is frequently associated with severe refractory disease. Prostaglandin (PG) D2 levels are increased in patients with CRS, and PGD2 is an important contributing factor to eosinophilic inflammation. Autophagy has a pleiotropic effect on immune responses and disease pathogenesis. Recent studies suggest the potential involvement of autophagy in patients with CRS and the PG pathway. OBJECTIVE: We sought to investigate whether altered function of autophagy is associated with eosinophilic inflammation and dysregulated production of PGD2 in patients with CRS. METHODS: We used myeloid cell-specific deletion of autophagy-related gene 7 (Atg7), which is vital for autophagy, and investigated the effects of impaired autophagy on eosinophilic inflammation in a murine model of eosinophilic chronic rhinosinusitis (ECRS). The effect of autophagy on PGD2 production and gene expression profiles associated with allergy and the PG pathway were assessed. RESULTS: We found that impaired autophagy in myeloid cells aggravated eosinophilia, epithelial hyperplasia, and mucosal thickening in mice with ECRS. This aggravation was associated with gene expression profiles that favor eosinophilic inflammation, TH2 response, mast cell infiltration, and PGD2 dysregulation. Supporting this, PGD2 production was also increased significantly by impaired autophagy. Among other myeloid cells, macrophages were associated with autophagy deficiency, leading to increased IL-1ß levels. Macrophage depletion or blockade of IL-1 receptor led to alleviation of eosinophilic inflammation and sinonasal anatomic abnormalities associated with autophagy deficiency. CONCLUSION: Our results suggest that impaired autophagy in myeloid cells, particularly macrophages, has a causal role in eosinophilic inflammation and ECRS pathogenesis.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/inmunología , Autofagia/inmunología , Eosinofilia/inmunología , Rinitis/inmunología , Transducción de Señal/inmunología , Sinusitis/inmunología , Animales , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/genética , Modelos Animales de Enfermedad , Eosinofilia/genética , Eosinofilia/patología , Inflamación , Ratones , Ratones Transgénicos , Rinitis/genética , Rinitis/patología , Transducción de Señal/genética , Sinusitis/genética , Sinusitis/patología
11.
J Alzheimers Dis ; 58(1): 253-261, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28387679

RESUMEN

BACKGROUND: Intracranial accumulation of amyloid-ß (Aß) is a characteristic finding of Alzheimer's disease (AD). It is thought to be the result of Aß overproduction by neurons and impaired clearance by several systems, including degradation by microglia. Sleep disturbance is now considered a risk factor for AD, but studies focusing on how sleep modulates microglial handling of Aß have been scarce. OBJECTIVE: To determine whether phagocytosis and degradation of extracellular Aß fibrils by BV2 microglial cells were impaired by treatment with orexin-A/B, a major modulator of the sleep-wake cycle, which may mimic sleep deprivation conditions. METHODS: BV2 cells were treated with orexin and Aß for various durations and phagocytic and autophagic processes for degradation of extracellular Aß were examined. RESULTS: After treatment with orexin, the formation of actin filaments around Aß fibrils, which is needed for phagocytosis, was impaired, and phagocytosis regulating molecules such as PI3K, Akt, and p38-MAPK were downregulated in BV2 cells. Orexin also suppressed autophagic flux, through disruption of the autophagosome-lysosome fusion process, resulting in impaired Aß degradation in BV2 cells. CONCLUSIONS: Our results demonstrate that orexin can hinder clearance of Aß through the suppression of phagocytosis and autophagic flux in microglia. This is a novel mechanism linking AD and sleep, and suggests that attenuated microglial function, due to sleep deprivation, may increase Aß accumulation in the brain.


Asunto(s)
Amiloide/metabolismo , Microglía/efectos de los fármacos , Orexinas/farmacología , Fagocitosis/efectos de los fármacos , Proteolisis/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular Transformada , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Alzheimers Res Ther ; 9(1): 4, 2017 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-28109317

RESUMEN

BACKGROUND: Amyloid precursor protein (APP) is cleaved by ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) to produce ß-amyloid (Aß), a critical pathogenic peptide in Alzheimer's disease (AD). Aß generation can be affected by the intracellular trafficking of APP or its related secretases, which is thus important to understanding its pathological alterations. Although sorting nexin (SNX) family proteins regulate this trafficking, the relevance and role of sorting nexin-4 (SNX4) regarding AD has not been studied yet. METHODS: In this study, human brain tissue and APP/PS1 mouse brain tissue were used to check the disease relevance of SNX4. To investigate the role of SNX4 in AD pathogenesis, several experiments were done, such as coimmunoprecipitation, Western blotting, immunohistochemistry, and gradient fractionation. RESULTS: We found that SNX4 protein levels changed in the brains of patients with AD and of AD model mice. Overexpression of SNX4 significantly increased the levels of BACE1 and Aß. Downregulation of SNX4 had the opposite effect. SNX4 interacts with BACE1 and prevents BACE1 trafficking to the lysosomal degradation system, resulting in an increased half-life of BACE1 and increased production of Aß. CONCLUSIONS: We show that SNX4 regulates BACE1 trafficking. Our findings suggest novel therapeutic implications of modulating SNX4 to regulate BACE1-mediated ß-processing of APP and subsequent Aß generation.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Nexinas de Clasificación/metabolismo , Anciano , Anciano de 80 o más Años , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Membrana Celular/metabolismo , Membrana Celular/patología , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Presenilina-1/genética , Presenilina-1/metabolismo
13.
Oncotarget ; 7(24): 35577-35591, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27229537

RESUMEN

Obesity-induced insulin resistance and diabetes are significantly associated with infiltrates of inflammatory cells in adipose tissue. Previous studies recognized the involvement of autophagy in the regulation of metabolism in multiple tissues, including ß-cells, hepatocytes, myocytes, and adipocytes. However, despite the importance of macrophages in obesity-induced insulin resistance, the role of macrophage autophagy in regulating insulin sensitivity is seldom addressed. In the present study, we show that macrophage autophagy is important for the regulation of systemic insulin sensitivity. We found that macrophage autophagy is downregulated by both acute and chronic inflammatory stimuli, and blockade of autophagy significantly increased accumulation of reactive oxygen species (ROS) in macrophages. Macrophage-specific Atg7 knockout mice displayed a shift in the proportion to pro-inflammatory M1 macrophages and impairment of insulin sensitivity and glucose homeostasis under high-fat diet conditions. Furthermore, inhibition of ROS in macrophages with antioxidant recovered adipocyte insulin sensitivity. Our results provide evidence of the underlying mechanism of how macrophage autophagy regulates inflammation and insulin sensitivity. We anticipate our findings will serve as a basis for development of therapeutics for inflammatory diseases, including diabetes.


Asunto(s)
Tejido Adiposo/patología , Autofagia/efectos de los fármacos , Inflamación/patología , Resistencia a la Insulina , Macrófagos/metabolismo , Obesidad/complicaciones , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia/genética , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Glucosa/metabolismo , Insulina/metabolismo , Macrólidos/farmacología , Macrófagos/citología , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo
14.
Brain Res ; 1649(Pt B): 201-209, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27016058

RESUMEN

Autophagy is a process to degrade and recycle cellular constituents via the lysosome for regulating cellular homeostasis. Its dysfunction is now considered to be involved in many diseases, including neurodegenerative diseases. Many features reflecting autophagy impairment, such as autophagosome accumulation and lysosomal dysfunction, have been also revealed to be involved in Alzheimer's disease (AD). Recent genetic studies such as genome-wide association studies in AD have identified a number of novel genes associated with AD. Some of the identified genes have demonstrated dysfunction in autophagic processes in AD, while others remain under investigation. Since autophagy is strongly regarded to be one of the major pathogenic mechanisms of AD, it is necessary to review how the AD-associated genes are related to autophagy. We anticipate our current review to be a starting point for future studies regarding AD-associated genes and autophagy. This article is part of a Special Issue entitled SI:Autophagy.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Autofagia , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
15.
BMB Rep ; 49(7): 376-81, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26839154

RESUMEN

Several lines of evidence have revealed that phosphorylation of amyloid precursor protein (APP) at Thr668 is involved in the pathogenesis of Alzheimer's disease (AD). Okadaic acid (OA), a protein phosphatase-2A inhibitor, has been used in AD research models to increase tau phosphorylation and induce neuronal death. We previously showed that OA increased levels of APP and induced accumulation of APP in axonal swellings. In this study, we found that in OA-treated neurons, phosphorylation of APP at Thr668 increased and accumulated in axonal swellings by c-jun N-terminal kinase (JNK), and not by Cdk5 or ERK/MAPK. These results suggest that JNK may be one of therapeutic targets for the treatment of AD. [BMB Reports 2016; 49(7): 376-381].


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Ácido Ocadaico/toxicidad , Enfermedad de Alzheimer/patología , Animales , Antracenos/farmacología , Western Blotting , Células Cultivadas , Inmunohistoquímica , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Neuronas/citología , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Regulación hacia Arriba/efectos de los fármacos
16.
BMB Rep ; 49(6): 337-42, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26779997

RESUMEN

Understanding of trafficking, processing, and degradation mechanisms of amyloid precursor protein (APP) is important because APP can be processed to produce ß-amyloid (Aß), a key pathogenic molecule in Alzheimer's disease (AD). Here, we found that APP contains KFERQ motif at its C-terminus, a consensus sequence for chaperone-mediated autophagy (CMA) or microautophagy which are another types of autophagy for degradation of pathogenic molecules in neurodegenerative diseases. Deletion of KFERQ in APP increased C-terminal fragments (CTFs) and secreted N-terminal fragments of APP and kept it away from lysosomes. KFERQ deletion did not abolish the interaction of APP or its cleaved products with heat shock cognate protein 70 (Hsc70), a protein necessary for CMA or microautophagy. These findings suggest that KFERQ motif is important for normal processing and degradation of APP to preclude the accumulation of APP-CTFs although it may not be important for CMA or microautophagy. [BMB Reports 2016; 49(6): 337-342].


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Procesamiento Proteico-Postraduccional , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Línea Celular Tumoral , Proteínas del Choque Térmico HSC70/metabolismo , Humanos , Lisosomas/metabolismo , Fosforilación , Unión Proteica , Relación Estructura-Actividad , Proteínas tau/metabolismo
17.
Front Neurosci ; 10: 618, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28149270

RESUMEN

The R47H coding variant of the triggering receptor expressed on myeloid cells-2 (TREM2) increases the risk of Alzheimer's disease (AD) similar to apolipoprotein E4. TREM2 R47H has recently been shown to have impaired binding to damage-associated lipid or apolipoprotein ligands. However, it is not known how this R47H variant affects the biochemical characteristics of TREM2 and alters the pathogenesis of AD. We previously reported that TREM2-R47H has a slightly different glycosylation pattern from wild-type. A more detailed characterization in our present study confirms that TREM2 R47H has an altered glycosylation pattern and reduced stability. TREM2 R47H shows different glycosylation profiles from analysis using monensin or kifunensine treatment which were confirmed by mass spectrometry. The solubility of TREM2 R47H and its cleaved products such as intracellular domain (ICD) is also decreased, increasing its proteasomal and lysosomal degradation. The different biochemical characteristics of TREM2 R47H, including glycosylation, solubility and processing, may offer insights into a future therapeutic strategy for AD.

18.
Aging Cell ; 14(5): 878-86, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26178297

RESUMEN

Toxicity induced by aberrant protein aggregates in Alzheimer's disease (AD) causes synaptic disconnection and concomitant progressive neurodegeneration that eventually impair cognitive function. cAMP-response element-binding protein (CREB) is a transcription factor involved in the molecular switch that converts short-term to long-term memory. Although disturbances in CREB function have been suggested to cause memory deficits in both AD and AD animal models, the mechanism of CREB dysfunction is still unclear. Here, we show that the dopamine- and cAMP-regulated phosphoprotein 32 kDa (DARPP-32), a key inhibitor of protein phosphate-1 (PP-1) that regulates CREB phosphorylation, is cleaved by activated calpain in both AD brains and neuronal cells treated with amyloid-ß or okadaic acid, a protein phosphatase-2A inhibitor that induces tau hyperphosphorylation and neuronal death. We found that DARPP-32 is mainly cleaved at Thr(153) by calpain and that this cleavage of DARPP-32 reduces CREB phosphorylation via loss of its inhibitory function on PP1. Our results suggest a novel mechanism of DARPP-32-CREB signalling dysregulation in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Calpaína/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/enzimología , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Femenino , Humanos , Masculino , Ratones , Fosforilación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
19.
Traffic ; 16(5): 510-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25615530

RESUMEN

The triggering receptor expressed on myeloid cells 2 (TREM2) is an immune-modulatory receptor involved in phagocytosis and inflammation. Mutations of Q33X, Y38C and T66M cause Nasu-Hakola disease (NHD) which is characterized by early onset of dementia and bone cysts. A recent, genome-wide association study also revealed that single nucleotide polymorphism of TREM2, such as R47H, increased the risk of Alzheimer's disease (AD) similar to ApoE4. However, how these mutations affect the trafficking of TREM2, which may affect the normal functions of TREM2, was not known. In this study, we show that TREM2 with NHD mutations are impaired in the glycosylation with complex oligosaccharides in the Golgi apparatus, in the trafficking to plasma membrane and further processing by γ-secretase. Although R47H mutation in AD affected the glycosylation and normal trafficking of TREM2 less, the detailed pattern of glycosylated TREM2 differs from that of the wild type, thus suggesting that precise regulation of TREM2 glycosylation is impaired when arginine at 47 is mutated to histidine. Our results suggest that the impaired glycosylation and trafficking of TREM2 from endoplasmic reticulum/Golgi to plasma membrane by mutations may inhibit its normal functions in the plasma membrane, which may contribute to the disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Aparato de Golgi/metabolismo , Lipodistrofia/metabolismo , Glicoproteínas de Membrana/genética , Mutación , Oligosacáridos/metabolismo , Osteocondrodisplasias/metabolismo , Receptores Inmunológicos/genética , Panencefalitis Esclerosante Subaguda/metabolismo , Enfermedad de Alzheimer/genética , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Glicosilación , Células HeLa , Humanos , Lipodistrofia/genética , Glicoproteínas de Membrana/metabolismo , Osteocondrodisplasias/genética , Polimorfismo de Nucleótido Simple , Transporte de Proteínas , Receptores Inmunológicos/metabolismo , Panencefalitis Esclerosante Subaguda/genética
20.
J Antibiot (Tokyo) ; 68(2): 76-80, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25138142

RESUMEN

Systemic inflammatory response syndrome (SIRS) is a serious condition that can cause organ failure as an exaggerated immunoresponse to the infection or other causes. Recently, autophagy was reported as a key process that regulates inflammatory responses in macrophages. Vancomycin is one of the most commonly prescribed antibiotics for sepsis treatment or following surgery. However, there are no studies on how vancomycin affects autophagy or inflammation. Here, we treated macrophage cell lines with vancomycin and lipopolysaccharides and found that vancomycin blocks autophagy and increases inflammatory responses. This finding suggests that vancomycin should be more cautiously administered in order to prevent unwanted SIRS during sepsis.


Asunto(s)
Antibacterianos/farmacología , Autofagia/efectos de los fármacos , Interleucina-1beta/efectos de los fármacos , Macrófagos/efectos de los fármacos , Vancomicina/farmacología , Animales , Línea Celular , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/inmunología , Ratones , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...