Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 322, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39078420

RESUMEN

Transmembrane protein 9 (TMEM9) is a transmembrane protein that regulates lysosomal acidification by interacting with the v-type ATPase complex. However, the role of TMEM9 in the lysosome-dependent autophagy machinery has yet to be identified. In this study, we demonstrate that the lysosomal protein TMEM9, which is involved in vesicle acidification, regulates Rab9-dependent alternative autophagy through its interaction with Beclin1. The cytosolic domain of TMEM9 interacts with Beclin1 via its Bcl-2-binding domain. This interaction between TMEM9 and Beclin1 dissociates Bcl-2, an autophagy-inhibiting partner, from Beclin1, thereby activating LC3-independent and Rab9-dependent alternative autophagy. Late endosomal and lysosomal TMEM9 apparently colocalizes with Rab9 but not with LC3. Furthermore, we show that multiple glycosylation of TMEM9, essential for lysosomal localization, is essential for its interaction with Beclin1 and the activation of Rab9-dependent alternative autophagy. These findings reveal that TMEM9 recruits and activates the Beclin1 complex at the site of Rab9-dependent autophagosome to induce alternative autophagy.


Asunto(s)
Autofagia , Beclina-1 , Lisosomas , Proteínas de la Membrana , Proteínas de Unión al GTP rab , Beclina-1/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de Unión al GTP rab/metabolismo , Lisosomas/metabolismo , Células HEK293 , Unión Proteica , Células HeLa , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Autofagosomas/metabolismo
2.
Brain Behav Immun ; 121: 291-302, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39098437

RESUMEN

In Alzheimer's disease, chronic neuroinflammation is accompanied by amyloid and tau pathologies. Especially, aberrant microglial activation is known to precede the regional tau pathology development, but the mechanisms how microglia affect tau spread remain largely unknown. Here, we found that toll-like receptor 2 (TLR2) in microglia recognizes oligomeric tau as a pathogenic ligand and induces inflammatory responses. Knockout of TLR2 reduced tau pathology and microglial activation in rTg4510 tau transgenic mice. Treatment of oligomeric tau induced TLR2 activation and increased inflammatory responses in microglial cells. TLR2 further mediated the tau-induced microglial activation and promoted tau uptake into neurons in neuron-microglia co-culture system and in mouse hippocampus after intracranial tau injection. Importantly, treatment with anti-TLR2 monoclonal antibody Tomaralimab blocked TLR2 activation and inflammatory responses in a dose-dependent manner, and significantly reduced tau spread and memory loss in rTg4510 mice. These results suggest that TLR2 plays a crucial role in tau spread by causing aberrant microglial activation in response to pathological tau, and blocking TLR2 with immunotherapy may ameliorate tau pathogenesis in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Inmunoterapia , Trastornos de la Memoria , Ratones Transgénicos , Microglía , Enfermedades Neuroinflamatorias , Neuronas , Receptor Toll-Like 2 , Proteínas tau , Animales , Receptor Toll-Like 2/metabolismo , Ratones , Proteínas tau/metabolismo , Microglía/metabolismo , Enfermedad de Alzheimer/metabolismo , Inmunoterapia/métodos , Enfermedades Neuroinflamatorias/metabolismo , Trastornos de la Memoria/metabolismo , Neuronas/metabolismo , Hipocampo/metabolismo , Modelos Animales de Enfermedad , Ratones Noqueados , Inflamación/metabolismo , Ratones Endogámicos C57BL
3.
Hum Mol Genet ; 29(2): 228-237, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31814000

RESUMEN

The transient receptor potential vanilloid 1 (TRPV1) protein is a pain receptor that elicits a hot sensation when an organism eats the capsaicin of red chili peppers. This calcium (Ca2+)-permeable cation channel is mostly expressed in the peripheral nervous system sensory neurons but also in the central nervous system (e.g. hippocampus and cortex). Preclinical studies found that TRPV1 mediates behaviors associated with anxiety and depression. Loss of TRPV1 functionality increases expression of genes related to synaptic plasticity and neurogenesis. Thus, we hypothesized that TRPV1 deficiency may modulate Alzheimer's disease (AD). We generated a triple-transgenic AD mouse model (3xTg-AD+/+) with wild-type (TRPV1+/+), hetero (TRPV1+/-) and knockout (TRPV1-/-) TRPV1 to investigate the role of TRPV1 in AD pathogenesis. We analyzed the animals' memory function, hippocampal Ca2+ levels and amyloid-ß (Aß) and tau pathologies when they were 12 months old. We found that compared with 3xTg-AD-/-/TRPV1+/+ mice, 3xTg-AD+/+/TRPV1+/+ mice had memory impairment and increased levels of hippocampal Ca2+, Aß and total and phosphorylated tau. However, 3xTg-AD+/+/TRPV1-/- mice had better memory function and lower levels of hippocampal Ca2+, Aß, tau and p-tau, compared with 3xTg-AD+/+/TRPV1+/+ mice. Examination of 3xTg-AD-derived primary neuronal cultures revealed that the intracellular Ca2+ chelator BAPTA/AM and the TRPV1 antagonist capsazepine decreased the production of Aß, tau and p-tau. Taken together, these results suggested that TRPV1 deficiency had anti-AD effects and promoted resilience to memory loss. These findings suggest that drugs or food components that modulate TRPV1 could be exploited as therapeutics to prevent or treat AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Calcio/metabolismo , Trastornos de la Memoria/metabolismo , Canales Catiónicos TRPV/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Canales de Calcio/metabolismo , Capsaicina/análogos & derivados , Capsaicina/farmacología , Quelantes/farmacología , Modelos Animales de Enfermedad , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Hipocampo/metabolismo , Aprendizaje/efectos de los fármacos , Trastornos de la Memoria/genética , Ratones , Ratones Noqueados , Nociceptores/metabolismo , Nociceptores/patología , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Proteínas tau/genética
4.
Mol Psychiatry ; 26(10): 5542-5556, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33452442

RESUMEN

Proteinopathy in neurodegenerative diseases is typically characterized by deteriorating activity of specific protein aggregates. In tauopathies, including Alzheimer's disease (AD), tau protein abnormally accumulates and induces dysfunction of the affected neurons. Despite active identification of tau modifications responsible for tau aggregation, a critical modulator inducing tau proteinopathy by affecting its protein degradation flux is not known. Here, we report that anaplastic lymphoma kinase (ALK), a receptor tyrosine kinase, is crucial for the tau-mediated AD pathology. ALK caused abnormal accumulation of highly phosphorylated tau in the somatodendritic region of neurons through its tyrosine kinase activity. ALK-induced LC3-positive axon swelling and loss of spine density, leading to tau-dependent neuronal degeneration. Notably, ALK activation in neurons impaired Stx17-dependent autophagosome maturation and this defect was reversed by a dominant-negative Grb2. In a Drosophila melanogaster model, transgenic flies neuronally expressing active Drosophila Alk exhibited the aggravated tau rough eye phenotype with retinal degeneration and shortened lifespan. In contrast, expression of kinase-dead Alk blocked these phenotypes. Consistent with the previous RNAseq analysis showing upregulation of ALK expression in AD [1], ALK levels were significantly elevated in the brains of AD patients showing autophagosomal defects. Injection of an ALK.Fc-lentivirus exacerbated memory impairment in 3xTg-AD mice. Conversely, pharmacologic inhibition of ALK activity with inhibitors reversed the memory impairment and tau accumulation in both 3xTg-AD and tauC3 (caspase-cleaved tau) transgenic mice. Together, we propose that aberrantly activated ALK is a bona fide mediator of tau proteinopathy that disrupts autophagosome maturation and causes tau accumulation and aggregation, leading to neuronal dysfunction in AD.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Enfermedad de Alzheimer/genética , Quinasa de Linfoma Anaplásico/genética , Animales , Drosophila melanogaster , Humanos , Ratones , Ratones Transgénicos , Tauopatías/genética , Proteínas tau/genética
5.
FASEB J ; 34(2): 2944-2957, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31908024

RESUMEN

Mitochondrial quality control maintains mitochondrial function by regulating mitochondrial dynamics and mitophagy. Despite the identification of mitochondrial quality control factors, little is known about the crucial regulators coordinating both mitochondrial fission and mitophagy. Through a cell-based functional screening assay, FK506 binding protein 8 (FKBP8) was identified to target microtubule-associated protein 1 light chain 3 (LC3) to the mitochondria and to change mitochondrial morphology. Microscopy analysis revealed that the formation of tubular and enlarged mitochondria was observed in FKBP8 knockdown HeLa cells and the cortex of Fkbp8 heterozygote-knockout mouse embryos. Under iron depletion-induced stress, FKBP8 was recruited to the site of mitochondrial division through budding and colocalized with LC3. FKBP8 was also found to be required for mitochondrial fragmentation and mitophagy under hypoxic stress. Conversely, FKBP8 overexpression induced mitochondrial fragmentation in HeLa cells, human fibroblasts and mouse embryo fibroblasts (MEFs), and this fragmentation occurred in Drp1 knockout MEF cells, FIP200 knockout HeLa cells and BNIP3/NIX double knockout HeLa cells, but not in Opa1 knockout MEFs. Interestingly, we found an LIR motif-like sequence (LIRL), as well as an LIR motif, at the N-terminus of FKBP8 and LIRL was essential for both inducing mitochondrial fragmentation and binding of FKBP8 to OPA1. Together, we suggest that FKBP8 plays an essential role in mitochondrial fragmentation through LIRL during mitophagy and this activity of FKBP8 together with LIR is required for mitophagy under stress conditions.


Asunto(s)
Fibroblastos/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Estrés Fisiológico , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/genética , Proteínas de Unión a Tacrolimus/genética
6.
FASEB J ; 33(3): 4300-4313, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30540497

RESUMEN

SRC-family kinases (SFKs) have been implicated in Alzheimer's disease (AD), but their mode of action was scarcely understood. Here, we show that LYN plays an essential role in amyloid ß (Aß)-triggered neurotoxicity and tau hyperphosphorylation by phosphorylating Fcγ receptor IIb2 (FcγRIIb2). We found that enzyme activity of LYN was increased in the brain of AD patients and was promoted in neuronal cells exposed to Aß 1-42 (Aß1-42). Knockdown of LYN expression inhibited Aß1-42-induced neuronal cell death. Of note, LYN interacted with FcγRIIb2 upon exposure to Aß1-42 and phosphorylated FcγRIIb2 at Tyr273 within immunoreceptor tyrosine-based inhibitory motif in neuronal cells. With the use of the structure-based drug design, we isolated KICG2576, an ATP-competitive inhibitor of LYN. Determination of cocrystal structure illustrated that KICG2576 bound to the cleft in the LYN kinase domain and inhibited LYN with a half-maximal inhibitory concentration value of 0.15 µM. KICG2576 inhibited Aß- or FcγRIIb2-induced cell death, and this effect was better than pyrazolopyrimidine 1, a widely used inhibitor of SFK. Upon exposure to Aß, KICG2576 blocked the phosphorylation of FcγRIIb2 and translocation of phosphatidylinositol 3,4,5-trisphosphate 5-phosphatase 2, a binding protein to the phosphorylated FcγRIIb2, to the plasma membrane, resulting in the inhibition of tau hyperphosphorylation, the downstream event of Aß1-42-FcγRIIb2 binding. Furthermore, intracerebroventricular injection of KICG2576 into mice ameliorated Aß-induced memory impairment. These results suggest that LYN plays a crucial role in Aß1-42-mediated neurotoxicity and tau pathology, providing a therapeutic potential of LYN in AD.-Gwon, Y., Kim, S.-H., Kim, H. T., Kam, T.-I., Park, J., Lim, B., Cha, H., Chang, H.-J., Hong, Y. R., Jung, Y.-K. Amelioration of amyloid ß-FcγRIIb neurotoxicity and tau pathologies by targeting LYN.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Neuronas/metabolismo , Receptores de IgG/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Hipocampo/metabolismo , Humanos , Trastornos de la Memoria/metabolismo , Ratones , Fragmentos de Péptidos/metabolismo , Fosfatidilinositoles/metabolismo , Fosforilación/fisiología , Ratas , Familia-src Quinasas/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(28): 7361-7366, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28652364

RESUMEN

Cell death-inducing DFF45-like effector (CIDE) domains, initially identified in apoptotic nucleases, form a family with diverse functions ranging from cell death to lipid homeostasis. Here we show that the CIDE domains of Drosophila and human apoptotic nucleases Drep2, Drep4, and DFF40 all form head-to-tail helical filaments. Opposing positively and negatively charged interfaces mediate the helical structures, and mutations on these surfaces abolish nuclease activation for apoptotic DNA fragmentation. Conserved filamentous structures are observed in CIDE family members involved in lipid homeostasis, and mutations on the charged interfaces compromise lipid droplet fusion, suggesting that CIDE domains represent a scaffold for higher-order assembly in DNA fragmentation and other biological processes such as lipid homeostasis.


Asunto(s)
Fragmentación del ADN , Desoxirribonucleasas/química , Proteínas de Unión a Poli-ADP-Ribosa/química , Proteínas/química , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/química , Sitios de Unión , Muerte Celular , Cristalografía por Rayos X , Proteínas de Drosophila/química , Drosophila melanogaster , Homeostasis , Lípidos/química , Ratones , Microscopía Electrónica de Transmisión , Conformación Molecular , Mutación , Dominios Proteicos , Multimerización de Proteína , Proteínas/genética
8.
J Neurosci ; 38(42): 9001-9018, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30185465

RESUMEN

Emerging evidences suggest that intraneuronal Aß correlates with the onset of Alzheimer's disease (AD) and highly contributes to neurodegeneration. However, critical mediator responsible for Aß uptake in AD pathology needs to be clarified. Here, we report that FcγRIIb2, a variant of Fcγ-receptor IIb (FcγRIIb), functions in neuronal uptake of pathogenic Aß. Cellular accumulation of oligomeric Aß1-42, not monomeric Aß1-42 or oligomeric Aß1-40, was blocked by Fcgr2b knock-out in neurons and partially in astrocytes. Aß1-42 internalization was FcγRIIb2 di-leucine motif-dependent and attenuated by TOM1, a FcγRIIb2-binding protein that repressed the receptor recycling. TOM1 expression was downregulated in the hippocampus of male 3xTg-AD mice and AD patients, and regulated by miR-126-3p in neuronal cells after exposure to Aß1-42 In addition, memory impairments in male 3xTg-AD mice were rescued by the lentiviral administration of TOM1 gene. Augmented Aß uptake into lysosome caused its accumulation in cytoplasm and mitochondria. Moreover, neuronal accumulation of Aß in both sexes of 3xTg-AD mice and memory deficits in male 3xTg-AD mice were ameliorated by forebrain-specific expression of Aß-uptake-defective Fcgr2b mutant. Our findings suggest that FcγRIIb2 is essential for neuropathic uptake of Aß in AD.SIGNIFICANCE STATEMENT Accumulating evidences suggest that intraneuronal Aß is found in the early step of AD brain and is implicated in the pathogenesis of AD. However, the critical mediator involved in these processes is uncertain. Here, we describe that the FcγRIIb2 variant is responsible for both neuronal uptake and intraneuronal distribution of pathogenic Aß linked to memory deficits in AD mice, showing a pathologic significance of the internalized Aß. Further, Aß internalization is attenuated by TOM1, a novel FcγRIIb2-binding protein. Together, we provide a molecular mechanism responsible for neuronal uptake of pathogenic Aß found in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Proteínas/metabolismo , Receptores de IgG/metabolismo , Animales , Astrocitos/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones Noqueados , MicroARNs/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores de IgG/genética
9.
J Neurochem ; 142(5): 734-746, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28632947

RESUMEN

In neurodegenerative diseases like Alzheimer's disease (AD), tau is hyperphosphorylated and forms aggregates and neurofibrillary tangles in affected neurons. Autophagy is critical to clear the aggregates of disease-associated proteins and is often altered in patients and animal models of AD. Because mechanistic target of rapamycin (mTOR) negatively regulates autophagy and is hyperactive in the brains of patients with AD, mTOR is an attractive therapeutic target for AD. However, pharmacological strategies to increase autophagy by targeting mTOR inhibition cause various side effects. Therefore, autophagy activation mediated by non-mTOR pathways is a new option for autophagy-based AD therapy. Here, we report that pimozide activates autophagy to rescue tau pathology in an AD model. Pimozide increased autophagic flux through the activation of the AMPK-Unc-51 like autophagy activating kinase 1 (ULK1) axis, but not of mTOR, in neuronal cells, and this function was independent of dopamine D2 receptor inhibition. Pimozide reduced levels of abnormally phosphorylated tau aggregates in neuronal cells. Further, daily intraperitoneal (i.p.) treatment of pimozide led to a recovery from memory deficits of TauC3 mice expressing a caspase-cleaved form of tau. In the brains of these mice, we found increased phosphorylation of AMPK1 and ULK1, and reduced levels of the soluble oligomers and NP40-insoluble aggregates of abnormally phosphorylated tau. Together, these results suggest that pimozide rescues memory impairments in TauC3 mice and reduces tau aggregates by increasing autophagic flux through the mTOR-independent AMPK-ULK1 axis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia/fisiología , Pimozida/farmacología , Proteínas tau/metabolismo , Animales , Autofagia/efectos de los fármacos , Células Cultivadas , Antagonistas de Dopamina/farmacología , Antagonistas de Dopamina/uso terapéutico , Femenino , Células HeLa , Humanos , Masculino , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Pimozida/uso terapéutico , Proteínas tau/antagonistas & inhibidores
10.
Br J Cancer ; 116(3): 370-381, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28081539

RESUMEN

BACKGROUND: Normal cells are sensitive to anoikis, which is a cell detachment-induced apoptosis. However, cancer cells acquire anoikis resistance that is essential for successful metastasis. This study aimed to demonstrate the function and potential mechanism of NADPH oxidase 4 (NOX4) and EGFR activation in regulating anoikis resistance in lung cancer. METHODS: Cells were cultured either in the attached or suspended condition. Cell viability was measured by cell counting and live and dead cell staining. Expression levels of NOX4 and EGFR were measured by PCR and immunoblotting. Reactive oxygen species (ROS) levels were measured by flow cytometry. Effects of NOX4 overexpression or NOX4 knockdown by si-NOX4 on anoikis sensitivity were explored. Levels of NOX4 and EGFR in lung cancer tissues were evaluated by IHC staining. RESULTS: NOX4 was upregulated but EGFR decreased in suspended cells compared with attached cells. Accordingly, ROS levels were increased in suspended cells, resulting in the activation of Src and EGFR. NOX4 knockdown decreased activation of Src and EGFR, and thus sensitised cells to anoikis. NOX4 overexpression increased EGFR levels and attenuated anoikis. NOX4 expression is upregulated and is positively correlated with EGFR levels in the lung cancer patient tissues. CONCLUSIONS: NOX4 upregulation confers anoikis resistance by ROS-mediated activation of EGFR and Src, and by maintaining EGFR levels, which is critical for cell survival.


Asunto(s)
Anoicis/genética , Receptores ErbB/fisiología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , NADPH Oxidasas/fisiología , Células A549 , Anoicis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Receptores ErbB/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , Metástasis de la Neoplasia , ARN Interferente Pequeño/farmacología , Células Tumorales Cultivadas
11.
Proc Natl Acad Sci U S A ; 111(1): E54-61, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24367089

RESUMEN

APIP, Apaf-1 interacting protein, has been known to inhibit two main types of programmed cell death, apoptosis and pyroptosis, and was recently found to be associated with cancers and inflammatory diseases. Distinct from its inhibitory role in cell death, APIP was also shown to act as a 5-methylthioribulose-1-phosphate dehydratase, or MtnB, in the methionine salvage pathway. Here we report the structural and enzymatic characterization of human APIP as an MtnB enzyme with a Km of 9.32 µM and a Vmax of 1.39 µmol min(-1) mg(-1). The crystal structure was determined at 2.0-Å resolution, revealing an overall fold similar to members of the zinc-dependent class II aldolase family. APIP/MtnB exists as a tetramer in solution and exhibits an assembly with C4 symmetry in the crystal lattice. The pocket-shaped active site is located at the end of a long cleft between two adjacent subunits. We propose an enzymatic reaction mechanism involving Glu139* as a catalytic acid/base, as supported by enzymatic assay, substrate-docking study, and sequence conservation analysis. We explored the relationship between two distinct functions of APIP/MtnB, cell death inhibition, and methionine salvage, by measuring the ability of enzymatic mutants to inhibit cell death, and determined that APIP/MtnB functions as a cell death inhibitor independently of its MtnB enzyme activity for apoptosis induced by either hypoxia or etoposide, but dependently for caspase-1-induced pyroptosis. Our results establish the structural and biochemical groundwork for future mechanistic studies of the role of APIP/MtnB in modulating cell death and inflammation and in the development of related diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Apoptosis , Muerte Celular , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis/metabolismo , Bacillus subtilis/metabolismo , Caspasa 1/metabolismo , Caspasa 9/metabolismo , Dominio Catalítico , Células HeLa , Humanos , Inflamación/metabolismo , Metionina/metabolismo , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Neoplasias/metabolismo , Homología de Secuencia de Aminoácido
12.
J Neurochem ; 137(5): 770-81, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26924229

RESUMEN

Amyloid beta peptide (Aß) is a pathological hallmark of Alzheimer's disease (AD) and is generated through the sequential cleavage of amyloid precursor protein (APP) by ß- and γ-secretases. Hypoxia is a known risk factor for AD and stimulates Aß generation by γ-secretase; however, the underlying mechanisms remain unclear. In this study, we showed that dual-specificity phosphatase 26 (DUSP26) regulates Aß generation through changes in subcellular localization of the γ-secretase complex and its substrate C99 under hypoxic conditions. DUSP26 was identified as a novel γ-secretase regulator from a genome-wide functional screen using a cDNA expression library. The phosphatase activity of DUSP26 was required for the increase in Aß42 generation through γ-secretase, but this regulation did not affect the amount of the γ-secretase complex. Interestingly, DUSP26 induced the accumulation of C99 in the axons by stimulating anterograde transport of C99-positive vesicles. Additionally, DUSP26 induced c-Jun N-terminal kinase (JNK) activation for APP processing and axonal transport of C99. Under hypoxic conditions, DUSP26 expression levels were elevated together with JNK activation, and treatment with JNK inhibitor SP600125, or the DUSP26 inhibitor NSC-87877, reduced hypoxia-induced Aß generation by diminishing vesicle trafficking of C99 to the axons. Finally, we observed enhanced DUSP26 expression and JNK activation in the hippocampus of AD patients. Our results suggest that DUSP26 mediates hypoxia-induced Aß generation through JNK activation, revealing a new regulator of γ-secretase-mediated APP processing under hypoxic conditions. We propose the role of phosphatase dual-specificity phosphatase 26 (DUSP26) in the selective regulation of Aß42 production in neuronal cells under hypoxic stress. Induction of DUSP26 causes JNK-dependent shift in the subcellular localization of γ-secretase and C99 from the cell body to axons for Aß42 generation. These findings provide a new strategy for developing new therapeutic targets to arrest AD progression.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Transporte Axonal/fisiología , Fosfatasas de Especificidad Dual/biosíntesis , Fosfatasas de Especificidad Dual/farmacología , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/biosíntesis , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/farmacología , Fragmentos de Péptidos/biosíntesis , Enfermedad de Alzheimer/metabolismo , Transporte Axonal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Células HEK293 , Humanos , Técnicas de Cultivo de Órganos
13.
Neurobiol Dis ; 87: 19-28, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26704708

RESUMEN

In neurodegenerative diseases like AD, tau forms neurofibrillary tangles, composed of tau protein. In the AD brain, activated caspases cleave tau at the 421th Asp, generating a caspase-cleaved form of tau, TauC3. Although TauC3 is known to assemble rapidly into filaments in vitro, a role of TauC3 in vivo remains unclear. Here, we generated a transgenic mouse expressing human TauC3 using a neuron-specific promoter. In this mouse, we found that human TauC3 was expressed in the hippocampus and cortex. Interestingly, TauC3 mice showed drastic learning and spatial memory deficits and reduced synaptic density at a young age (2-3months). Notably, tau oligomers as well as tau aggregates were found in TauC3 mice showing memory deficits. Further, i.p. or i.c.v. injection with methylene blue or Congo red, inhibitors of tau aggregation in vitro, and i.p. injection with rapamycin significantly reduced the amounts of tau oligomers in the hippocampus, rescued spine density, and attenuated memory impairment in TauC3 mice. Together, these results suggest that TauC3 facilitates early memory impairment in transgenic mice accompanied with tau oligomer formation, providing insight into the role of TauC3 in the AD pathogenesis associated with tau oligomers and a useful AD model to test drug candidates.


Asunto(s)
Caspasas/metabolismo , Trastornos de la Memoria/metabolismo , Proteínas tau/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/patología , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nootrópicos/farmacología , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/fisiología , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Sirolimus/farmacología , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Proteínas tau/genética
14.
J Neurochem ; 132(3): 276-85, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25418874

RESUMEN

Methyl-ß-cyclodextrin (MßCD) is a reagent that depletes cholesterol and disrupts lipid rafts, a type of cholesterol-enriched cell membrane microdomain. Lipid rafts are essential for neuronal functions such as synaptic transmission and plasticity, which are sensitive to even low doses of MßCD. However, how MßCD changes synaptic function, such as N-methyl-d-aspartate receptor (NMDA-R) activity, remains unclear. We monitored changes in synaptic transmission and plasticity after disrupting lipid rafts with MßCD. At low concentrations (0.5 mg/mL), MßCD decreased basal synaptic transmission and miniature excitatory post-synaptic current without changing NMDA-R-mediated synaptic transmission and the paired-pulse facilitation ratio. Interestingly, low doses of MßCD failed to deplete cholesterol or affect α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPA-R) and NMDA-R levels, while clearly reducing GluA1 levels selectively in the synaptosomal fraction. Low doses of MßCD decreased the inhibitory effects of NASPM, an inhibitor for GluA2-lacking AMPA-R. MßCD successfully decreased NMDA-R-mediated long-term potentiation but did not affect the formation of either NMDA-R-mediated or group I metabotropic glutamate receptor-dependent long-term depression. MßCD inhibited de-depression without affecting de-potentiation. These results suggest that MßCD regulates GluA1-dependent synaptic potentiation but not synaptic depression in a cholesterol-independent manner.


Asunto(s)
Receptores AMPA/fisiología , Sinapsis/efectos de los fármacos , beta-Ciclodextrinas/farmacología , Animales , Colesterol/metabolismo , Técnicas In Vitro , Masculino , Microdominios de Membrana/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores AMPA/efectos de los fármacos , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/efectos de los fármacos , Sinaptosomas/efectos de los fármacos
15.
Biochem Biophys Res Commun ; 462(2): 112-8, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25930997

RESUMEN

In Alzheimer's disease and other tauopathy, abnormal Tau proteins form intracellular aggregates and Tau filaments. However, the mechanisms that regulate Tau aggregation are not fully understood. In this paper, we show that POLDIP2 is a novel regulator of Tau aggregation. From a cell-based screening using cDNA expression library, we isolated POLDIP2 which increased Tau aggregation. Expression of POLDIP2 was increased in neuronal cells by the multiple stresses, including Aß, TNF-α and H2O2. Accordingly, ectopic expression of POLDIP2 enhanced the formation of Tau aggregates without affecting Tau phosphorylation, while down-regulation of POLDIP2 alleviated ROS-induced Tau aggregation. Interestingly, we found that POLDIP2 overexpression induced impairments of autophagy activity and partially proteasome activity and this activities were retained in DUF525 domain of POLDIP2. In a drosophila model of human tauopathy, knockdown of the drosophila POLDIP2 homolog, CG12162, attenuated rough eye phenotype induced by Tau overexpression. Further, the lifespan of neural-Tau(R406W) transgenic files were recovered by CG12162 knockdown. Together, these observations indicate that POLDIP2 plays a crucial role in Tau aggregation via the impairment of autophagy activity, providing insight into Tau aggregation in Tau pathology.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas tau/metabolismo , Animales , Animales Modificados Genéticamente , Autofagia , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Técnicas de Silenciamiento del Gen , Genes de Insecto , Células HEK293 , Células HeLa , Humanos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas tau/química , Proteínas tau/genética
16.
Cell Mol Life Sci ; 71(24): 4803-13, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25151011

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disease. Although a major cause of AD is the accumulation of amyloid-ß (Aß) peptide that induces neuronal loss and cognitive impairments, our understanding of its neurotoxic mechanisms is limited. Recent studies have identified putative Aß-binding receptors that mediate Aß neurotoxicity in cells and models of AD. Once Aß interacts with a receptor, a toxic signal is transduced into neurons, resulting in cellular defects including endoplasmic reticulum stress and mitochondrial dysfunction. In addition, Aß can also be internalized into neurons through unidentified Aß receptors and induces malfunction of subcellular organelles, which explains some part of Aß neurotoxicity. Understanding the neurotoxic signaling initiated by Aß-receptor binding and cellular defects provide insight into new therapeutic windows for AD. In the present review, we summarize the findings on Aß-binding receptors and the neurotoxicity of oligomeric Aß.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Neuronas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Humanos , Modelos Biológicos , Neuronas/patología , Unión Proteica , Transducción de Señal
17.
Cell Mol Life Sci ; 71(13): 2561-76, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24270855

RESUMEN

The gamma (γ)-secretase holoenzyme is composed of four core proteins and cleaves APP to generate amyloid beta (Aß), a key molecule that causes major neurotoxicity during the early stage of Alzheimer's disease (AD). However, despite its important role in Aß production, little is known about the regulation of γ-secretase. OCIAD2, a novel modulator of γ-secretase that stimulates Aß production, and which was isolated from a genome-wide functional screen using cell-based assays and a cDNA library comprising 6,178 genes. Ectopic expression of OCIAD2 enhanced Aß production, while reduction of OCIAD2 expression suppressed it. OCIAD2 expression facilitated the formation of an active γ-secretase complex and enhanced subcellular localization of the enzyme components to lipid rafts. OCIAD2 interacted with nicastrin to stimulate γ-secretase activity. OCIAD2 also increased the interaction of nicastrin with C99 and stimulated APP processing via γ-secretase activation, but did not affect Notch processing. In addition, a cell-permeable Tat-OCIAD2 peptide that interfered with the interaction of OCIAD2 with nicastrin interrupted the γ-secretase-mediated AICD production. Finally, OCIAD2 expression was significantly elevated in the brain of AD patients and PDAPP mice. This study identifies OCIAD2 as a selective activator of γ-secretase to increase Aß generation.


Asunto(s)
Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/biosíntesis , Animales , Fibroblastos/metabolismo , Biblioteca de Genes , Humanos , Glicoproteínas de Membrana/genética , Microdominios de Membrana/metabolismo , Ratones , Ratones Noqueados/metabolismo , Proteínas de Neoplasias/genética , Receptores Notch/metabolismo
18.
Hum Mol Genet ; 21(12): 2725-37, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22419736

RESUMEN

Abnormally hyperphosphorylated tau is often caused by tau kinases, such as GSK3ß and Cdk5. Such occurrence leads to neurofibrillary tangle formation and neuronal degeneration in tauopathy, including Alzheimer's disease (AD). However, little is known about the signaling cascade underlying the pathologic phosphorylation of tau by Aß(42). In this study, we show that adenylate kinase 1 (AK1) is a novel regulator of abnormal tau phosphorylation. AK1 expression is markedly increased in the brains of AD patients and AD model mice and is significantly induced by Aß(42) in the primary neurons. Ectopic expression of AK1 alone augments the pathologic phosphorylation of tau at PHF1, CP13 and AT180 epitopes and enhances the formation of tau aggregates. Inversely, downregulation of AK1 alleviates Aß(42)-induced hyperphosphorylation of tau. AK1 plays a role in Aß(42)-induced impairment of AMPK activity and GSK3ß activation in the primary neurons. Pharmacologic studies show that treatment with an AMPK inhibitor activates GSK3ß, and a GSK3ß inhibitor attenuates AK1-mediated tau phosphorylation. In a Drosophila model of human tauopathy, the retinal expression of human AK1 severely exacerbates rough eye phenotype and increases abnormal tau phosphorylation. Further, neural expression of AK1 reduces the lifespan of tau transgenic files. Taken together, these observations indicate that the neuronal expression of AK1 is induced by Aß(42) to increase abnormal tau phosphorylation via AMPK-GSK3ß and contributes to tau-mediated neurodegeneration, providing a new upstream modulator of GSK3ß in the pathologic phosphorylation of tau.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adenilato Quinasa/metabolismo , Enfermedad de Alzheimer/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas tau/metabolismo , Adenilato Quinasa/genética , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Modificados Genéticamente , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Fosforilación/efectos de los fármacos , Interferencia de ARN , Proteínas tau/genética
19.
Hum Mol Genet ; 21(1): 101-14, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21954231

RESUMEN

Huntington's disease (HD), an inherited neurodegenerative disorder, is caused by an expansion of cytosine-adenine-guanine repeats in the huntingtin gene. The aggregation of mutant huntingtin (mtHTT) and striatal cell loss are representative features to cause uncontrolled movement and cognitive defect in HD. However, underlying mechanism of mtHTT aggregation and cell toxicity remains still elusive. Here, to find new genes modulating mtHTT aggregation, we performed cell-based functional screening using the cDNA expression library and isolated IRE1 gene, one of endoplasmic reticulum (ER) stress sensors. Ectopic expression of IRE1 led to its self-activation and accumulated detergent-resistant mtHTT aggregates. Treatment of neuronal cells with ER stress insults, tunicamycin and thapsigargin, increased mtHTT aggregation via IRE1 activation. The kinase activity of IRE1, but not the endoribonuclease activity, was necessary to stimulate mtHTT aggregation and increased death of neuronal cells, including SH-SY5Y and STHdhQ111/111 huntingtin knock-in striatal cells. Interestingly, ER stress impaired autophagy flux via IRE1-TRAF2 pathway, thus enhancing cellular accumulation of mtHTT. Atg5 deficiency in M5-7 cells increased mtHTT aggregation but blocked ER stress-induced mtHTT aggregation. Further, ER stress markers including p-IRE1 and autophagy markers such as p62 were up-regulated exclusively in the striatal tissues of HD mouse models and in HD patients. Moreover, down-regulation of IRE1 expression rescues the rough-eye phenotype by mtHTT in a HD fly model. These results suggest that IRE1 plays an essential role in ER stress-mediated aggregation of mtHTT via the inhibition of autophagy flux and thus neuronal toxicity of mtHTT aggregates in HD.


Asunto(s)
Autofagia , Regulación hacia Abajo , Estrés del Retículo Endoplásmico , Endorribonucleasas/metabolismo , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/fisiopatología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Células Cultivadas , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Endorribonucleasas/genética , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Proteínas de la Membrana/genética , Ratones , Mutación , Neuronas/enzimología , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA