Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 302, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39008111

RESUMO

DNAX-activating protein of 12 kDa (DAP12) is a transmembrane adapter protein expressed in lymphoid and myeloid lineage cells. It interacts with several immunoreceptors forming functional complexes that trigger intracellular signaling pathways. One of the DAP12 associated receptors is the triggering receptor expressed on myeloid cells 2 (TREM2). Mutations in both DAP12 and TREM2 have been linked to neurodegenerative diseases. However, mechanisms involved in the regulation of subcellular trafficking and turnover of these proteins are not well understood. Here, we demonstrate that proteasomal degradation of DAP12 is increased in the absence of TREM2. Interestingly, unassembled DAP12 is also retained in early secretory compartments, including the endoplasmic reticulum (ER) and the ER-Golgi intermediate compartment (ERGIC), thereby preventing its transport to the plasma membrane. We also show that unassembled DAP12 interacts with the retention in ER sorting receptor 1 (RER1). The deletion of endogenous RER1 decreases expression of functional TREM2-DAP12 complexes and membrane proximal signaling, and resulted in almost complete inhibition of phagocytic activity in THP-1 differentiated macrophage-like cells. These results indicate that RER1 acts as an important regulator of DAP12 containing immunoreceptor complexes and immune cell function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Retículo Endoplasmático , Glicoproteínas de Membrana , Receptores Imunológicos , Via Secretória , Humanos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Retículo Endoplasmático/metabolismo , Via Secretória/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Células HEK293 , Transdução de Sinais , Fagocitose/genética , Macrófagos/metabolismo , Transporte Proteico , Ligação Proteica , Animais , Complexo de Golgi/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/genética , Membrana Celular/metabolismo
2.
Int J Mol Sci ; 25(10)2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38791456

RESUMO

Presenilin proteins (PS1 and PS2) represent the catalytic subunit of γ-secretase and play a critical role in the generation of the amyloid ß (Aß) peptide and the pathogenesis of Alzheimer disease (AD). However, PS proteins also exert multiple functions beyond Aß generation. In this study, we examine the individual roles of PS1 and PS2 in cellular cholesterol metabolism. Deletion of PS1 or PS2 in mouse models led to cholesterol accumulation in cerebral neurons. Cholesterol accumulation was also observed in the lysosomes of embryonic fibroblasts from Psen1-knockout (PS1-KO) and Psen2-KO (PS2-KO) mice and was associated with decreased expression of the Niemann-Pick type C1 (NPC1) protein involved in intracellular cholesterol transport in late endosomal/lysosomal compartments. Mass spectrometry and complementary biochemical analyses also revealed abnormal N-glycosylation of NPC1 and several other membrane proteins in PS1-KO and PS2-KO cells. Interestingly, pharmacological inhibition of N-glycosylation resulted in intracellular cholesterol accumulation prominently in lysosomes and decreased NPC1, thereby resembling the changes in PS1-KO and PS2-KO cells. In turn, treatment of PS1-KO and PS2-KO mouse embryonic fibroblasts (MEFs) with the chaperone inducer arimoclomol partially normalized NPC1 expression and rescued lysosomal cholesterol accumulation. Additionally, the intracellular cholesterol accumulation in PS1-KO and PS2-KO MEFs was prevented by overexpression of NPC1. Collectively, these data indicate that a loss of PS function results in impaired protein N-glycosylation, which eventually causes decreased expression of NPC1 and intracellular cholesterol accumulation. This mechanism could contribute to the neurodegeneration observed in PS KO mice and potentially to the pathogenesis of AD.


Assuntos
Colesterol , Fibroblastos , Lisossomos , Proteína C1 de Niemann-Pick , Presenilina-1 , Presenilina-2 , Animais , Camundongos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Colesterol/metabolismo , Fibroblastos/metabolismo , Glicosilação , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lisossomos/metabolismo , Camundongos Knockout , Neurônios/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Presenilina-2/metabolismo , Presenilina-2/genética
3.
Brain ; 145(10): 3558-3570, 2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36270003

RESUMO

Alzheimer's disease is neuropathologically characterized by the deposition of the amyloid ß-peptide (Aß) as amyloid plaques. Aß plaque pathology starts in the neocortex before it propagates into further brain regions. Moreover, Aß aggregates undergo maturation indicated by the occurrence of post-translational modifications. Here, we show that propagation of Aß plaques is led by presumably non-modified Aß followed by Aß aggregate maturation. This sequence was seen neuropathologically in human brains and in amyloid precursor protein transgenic mice receiving intracerebral injections of human brain homogenates from cases varying in Aß phase, Aß load and Aß maturation stage. The speed of propagation after seeding in mice was best related to the Aß phase of the donor, the progression speed of maturation to the stage of Aß aggregate maturation. Thus, different forms of Aß can trigger propagation/maturation of Aß aggregates, which may explain the lack of success when therapeutically targeting only specific forms of Aß.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Animais , Humanos , Camundongos , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Placa Amiloide/metabolismo , Camundongos Transgênicos , Encéfalo/patologia , Modelos Animais de Doenças
4.
Nature ; 552(7685): 355-361, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29293211

RESUMO

The spreading of pathology within and between brain areas is a hallmark of neurodegenerative disorders. In patients with Alzheimer's disease, deposition of amyloid-ß is accompanied by activation of the innate immune system and involves inflammasome-dependent formation of ASC specks in microglia. ASC specks released by microglia bind rapidly to amyloid-ß and increase the formation of amyloid-ß oligomers and aggregates, acting as an inflammation-driven cross-seed for amyloid-ß pathology. Here we show that intrahippocampal injection of ASC specks resulted in spreading of amyloid-ß pathology in transgenic double-mutant APPSwePSEN1dE9 mice. By contrast, homogenates from brains of APPSwePSEN1dE9 mice failed to induce seeding and spreading of amyloid-ß pathology in ASC-deficient APPSwePSEN1dE9 mice. Moreover, co-application of an anti-ASC antibody blocked the increase in amyloid-ß pathology in APPSwePSEN1dE9 mice. These findings support the concept that inflammasome activation is connected to seeding and spreading of amyloid-ß pathology in patients with Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Microglia/metabolismo , Agregação Patológica de Proteínas , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Anticorpos/administração & dosagem , Anticorpos/imunologia , Anticorpos/farmacologia , Proteínas Adaptadoras de Sinalização CARD/antagonistas & inibidores , Proteínas Adaptadoras de Sinalização CARD/química , Proteínas Adaptadoras de Sinalização CARD/imunologia , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Presenilina-1/deficiência , Presenilina-1/genética , Domínios Proteicos , Memória Espacial/fisiologia
5.
J Biol Chem ; 296: 100631, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33823153

RESUMO

TREM2 is a pattern recognition receptor, expressed on microglia and myeloid cells, detecting lipids and Aß and inducing an innate immune response. Missense mutations (e.g., R47H) of TREM2 increase risk of Alzheimer's disease (AD). The soluble ectodomain of wild-type TREM2 (sTREM2) has been shown to protect against AD in vivo, but the underlying mechanisms are unclear. We show that Aß oligomers bind to cellular TREM2, inducing shedding of the sTREM2 domain. Wild-type sTREM2 bound to Aß oligomers (measured by single-molecule imaging, dot blots, and Bio-Layer Interferometry) inhibited Aß oligomerization and disaggregated preformed Aß oligomers and protofibrils (measured by transmission electron microscopy, dot blots, and size-exclusion chromatography). Wild-type sTREM2 also inhibited Aß fibrillization (measured by imaging and thioflavin T fluorescence) and blocked Aß-induced neurotoxicity (measured by permeabilization of artificial membranes and by loss of neurons in primary neuronal-glial cocultures). In contrast, the R47H AD-risk variant of sTREM2 is less able to bind and disaggregate oligomeric Aß but rather promotes Aß protofibril formation and neurotoxicity. Thus, in addition to inducing an immune response, wild-type TREM2 may protect against amyloid pathology by the Aß-induced release of sTREM2, which blocks Aß aggregation and neurotoxicity. In contrast, R47H sTREM2 promotes Aß aggregation into protofibril that may be toxic to neurons. These findings may explain how wild-type sTREM2 apparently protects against AD in vivo and why a single copy of the R47H variant gene is associated with increased AD risk.


Assuntos
Peptídeos beta-Amiloides/química , Amiloide/química , Glicoproteínas de Membrana/fisiologia , Proteínas Mutantes/metabolismo , Mutação , Neurônios/patologia , Síndromes Neurotóxicas/patologia , Receptores Imunológicos/fisiologia , Doença de Alzheimer , Amiloide/metabolismo , Animais , Camundongos , Camundongos Knockout , Proteínas Mutantes/genética , Neurônios/metabolismo , Síndromes Neurotóxicas/etiologia
6.
Glia ; 70(12): 2290-2308, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35912412

RESUMO

The receptor Triggering Receptor Expressed on Myeloid cells 2 (TREM2) is associated with several neurodegenerative diseases including Alzheimer's Disease and TREM2 stimulation represents a novel therapeutic opportunity. TREM2 can be activated by antibodies targeting the stalk region, most likely through receptor dimerization. Endogenous ligands of TREM2 are suggested to be negatively charged apoptotic bodies, mimicked by phosphatidylserine incorporated in liposomes and other polyanionic molecules likely binding to TREM2 IgV fold. However, there has been much discrepancy in the literature on the nature of phospholipids (PLs) that can activate TREM2 and on the stability of the corresponding liposomes over time. We describe optimized liposomes as robust agonists selective for TREM2 over TREM1 in cellular system. The detailed structure/activity relationship studies of lipid polar heads indicate that negatively charged lipid heads are required for activity and we identified the shortest maximally active PL sidechain. Optimized liposomes are active on both TREM2 common variant and TREM2 R47H mutant. Activity and selectivity were further confirmed in different native TREM2 expressing cell types including on integrated cellular responses such as stimulation of phagocytic activity. Such tool agonists will be useful in further studies of TREM2 biology in cellular systems alongside antibodies, and in the design of small molecule synthetic TREM2 agonists.


Assuntos
Doença de Alzheimer , Lipossomos , Glicoproteínas de Membrana/imunologia , Receptores Imunológicos/imunologia , Doença de Alzheimer/metabolismo , Anticorpos/metabolismo , Encéfalo/metabolismo , Humanos , Ligantes , Microglia/metabolismo , Células Mieloides/metabolismo , Fosfatidilserinas/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo
7.
Glia ; 69(5): 1126-1139, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33314333

RESUMO

The triggering receptor expressed on myeloid cells 2 (TREM2) is an immune receptor expressed on myeloid-derived cell types. The extracellular immunoglobulin-like domain of TREM2 binds anionic ligands including Apolipoprotein E and Amyloid-ß. The transmembrane domain interacts with its adaptor protein DAP12/TYROBP that is responsible for propagation of downstream signaling upon ligand interaction. Several sequence variants of TREM2 have been linked to different neurodegenerative diseases including Alzheimer's disease. Here, we generated HEK 293 Flp-In cell lines stably expressing human TREM2 and DAP12 using a bicistronic construct with a T2A linker sequence allowing initial expression of both proteins in stoichiometric amounts. Cell biological and biochemical analyses revealed transport of TREM2 to the cell surface, and canonical sequential proteolytic processing and shedding of TREM2 (sTREM2). The functionality of this cell system was demonstrated by detection of phosphorylated spleen tyrosine kinase (SYK) upon stimulation of TREM2 with the anionic membrane lipid phosphatidylserine or anti-TREM2 antibodies. Using this cell model, we demonstrated impaired signaling of disease associated TREM2 variants. We also identified a monoclonal antibody against the stalk region of TREM2 with agonistic activity. Activation of TREM2-DAP12 signaling with the monoclonal antibody and the partial loss of function of disease associated variants were recapitulated in induced pluripotent stem cell derived microglia. Thus, this reporter cell model represents a suitable experimental system to investigate signaling of TREM2 variants, and for the identification of ligands and compounds that modulate TREM2-DAP12 signaling. MAIN POINTS: Disease associated variants impair the signaling activity of TREM2 by distinct mechanisms. Targeting the stalk region of TREM2 with bivalent antibodies activates TREM2 signaling.


Assuntos
Doença de Alzheimer , Microglia , Anticorpos Monoclonais , Proteínas de Transporte , Células HEK293 , Humanos , Ligantes , Glicoproteínas de Membrana/genética , Células Mieloides , Receptores Imunológicos/genética
8.
Glia ; 69(12): 2917-2932, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34427354

RESUMO

Rare coding variants of the microglial triggering receptor expressed on myeloid cells 2 (TREM2) confer an increased risk for Alzheimer's disease (AD) characterized by the progressive accumulation of aggregated forms of amyloid ß peptides (Aß). Aß peptides are generated by proteolytic processing of the amyloid precursor protein (APP). Heterogeneity in proteolytic cleavages and additional post-translational modifications result in the production of several distinct Aß variants that could differ in their aggregation behavior and toxic properties. Here, we sought to assess whether post-translational modifications of Aß affect the interaction with TREM2. Biophysical and biochemical methods revealed that TREM2 preferentially interacts with oligomeric Aß, and that phosphorylation of Aß increases this interaction. Phosphorylation of Aß also affected the TREM2 dependent interaction and phagocytosis by primary microglia and in APP transgenic mouse models. Thus, TREM2 function is important for sensing phosphorylated Aß variants in distinct aggregation states and reduces the accumulation and deposition of these toxic Aß species in preclinical models of Alzheimer's disease.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Microglia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
9.
Biochem Biophys Res Commun ; 570: 137-142, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34280617

RESUMO

γ-Secretase is a protease catalysing the proteolysis of type-I membrane proteins usually after precedent ectodomain shedding of the respective protein substrates. Since proteolysis of membrane proteins is involved in fundamental cellular signaling pathways, dysfunction of γ-secretase can have significant impact on cellular metabolism and differentiation. Here, we examined the role of γ-secretase in cellular lipid metabolism using neuronally differentiated human SH-SY5Y cells. The pharmacological inhibition of γ-secretase induced lipid droplet (LD) accumulation. The LD accumulation was significantly attenuated by preventing the accumulation of C-terminal fragment of the amyloid precursor protein (APP-CTF), which is a direct substrate of γ-secretase. Additionally, LD accumulation upon γ-secretase inhibition was not induced in APP-knock out (APP-KO) mouse embryonic fibroblasts (MEFs), suggesting significant involvement of APP-CTF accumulation in LD accumulation upon γ-secretase inhibition. On the other hand, γ-secretase inhibition-dependent cholesterol accumulation was not attenuated by inhibition of APP-CTF accumulation in the differentiated SH-SY5Y cells nor in APP-KO MEFs. These results suggest that γ-secretase inhibition can induce accumulation of LD and cholesterol differentially via APP-CTF accumulation.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Gotículas Lipídicas/metabolismo , Fragmentos de Peptídeos/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Colesterol/metabolismo , Camundongos
10.
Hum Mutat ; 41(1): 169-181, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31464095

RESUMO

Rare coding variants in the triggering receptor expressed on myeloid cells-2 (TREM2) gene have been associated with Alzheimer disease (AD) and homozygous TREM2 loss-of-function variants have been reported in families with monogenic frontotemporal-like dementia with/without bone abnormalities. In a whole-exome sequencing study of a family with probable AD-type dementia without pathogenic variants in known autosomal dominant dementia disease genes and negative for the apolipoprotein E (APOE) ε4 allele, we identified an extremely rare TREM2 coding variant, that is, a glycine-to-tryptophan substitution at amino acid position 145 (NM_018965.3:c.433G>T/p.[Gly145Trp]). This alteration is found in only 1 of 251,150 control alleles in gnomAD. It was present in both severely affected as well as in another putatively affected and one 61 years old as yet unaffected family member suggesting incomplete penetrance and/or a variable age of onset. Gly145 maps to an intrinsically disordered region (IDR) of TREM2 between the immunoglobulin-like and transmembrane domain. Subsequent cellular studies showed that the variant led to IDR shortening and structural changes of the mutant protein resulting in an impairment of cellular responses upon receptor activation. Our results, suggest that a p.(Gly145Trp)-induced structural disturbance and functional impairment of TREM2 may contribute to the pathogenesis of an AD-like form of dementia.


Assuntos
Demência/diagnóstico , Demência/genética , Predisposição Genética para Doença , Variação Genética , Heterozigoto , Proteínas Intrinsicamente Desordenadas/genética , Glicoproteínas de Membrana/genética , Receptores Imunológicos/genética , Idoso , Alelos , Animais , Linhagem Celular , Feminino , Estudos de Associação Genética , Humanos , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Fases de Leitura Aberta/genética , Linhagem , Fenótipo , Transporte Proteico , Receptores Imunológicos/metabolismo , Transdução de Sinais , Sequenciamento do Exoma
11.
Acta Neuropathol ; 140(6): 811-830, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32926214

RESUMO

Alzheimer's disease (AD) is characterized by amyloid-beta (Aß) deposits, which come in myriad morphologies with varying clinical relevance. Previously, we observed an atypical Aß deposit, referred to as the coarse-grained plaque. In this study, we evaluate the plaque's association with clinical disease and perform in-depth immunohistochemical and morphological characterization. The coarse-grained plaque, a relatively large (Ø ≈ 80 µm) deposit, characterized as having multiple cores and Aß-devoid pores, was prominent in the neocortex. The plaque was semi-quantitatively scored in the middle frontal gyrus of Aß-positive cases (n = 74), including non-demented cases (n = 15), early-onset (EO)AD (n = 38), and late-onset (LO)AD cases (n = 21). The coarse-grained plaque was only observed in cases with clinical dementia and more frequently present in EOAD compared to LOAD. This plaque was associated with a homozygous APOE ε4 status and cerebral amyloid angiopathy (CAA). In-depth characterization was done by studying the coarse-grained plaque's neuritic component (pTau, APP, PrPC), Aß isoform composition (Aß40, Aß42, AßN3pE, pSer8Aß), its neuroinflammatory component (C4b, CD68, MHC-II, GFAP), and its vascular attribution (laminin, collagen IV, norrin). The plaque was compared to the classic cored plaque, cotton wool plaque, and CAA. Similar to CAA but different from classic cored plaques, the coarse-grained plaque was predominantly composed of Aß40. Furthermore, the coarse-grained plaque was distinctly associated with both intense neuroinflammation and vascular (capillary) pathology. Confocal laser scanning microscopy (CLSM) and 3D analysis revealed for most coarse-grained plaques a particular Aß40 shell structure and a direct relation with vessels. Based on its morphological and biochemical characteristics, we conclude that the coarse-grained plaque is a divergent Aß plaque-type associated with EOAD. Differences in Aß processing and aggregation, neuroinflammatory response, and vascular clearance may presumably underlie the difference between coarse-grained plaques and other Aß deposits. Disentangling specific Aß deposits between AD subgroups may be important in the search for disease-mechanistic-based therapies.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Angiopatia Amiloide Cerebral/patologia , Placa Amiloide/patologia , Idade de Início , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Capilares/patologia , Angiopatia Amiloide Cerebral/genética , Feminino , Humanos , Masculino , Neuritos/patologia
12.
Glia ; 67(3): 539-550, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30548312

RESUMO

The microglial triggering receptor expressed on myeloid cells 2 (TREM2) signals via the activatory membrane adaptor molecule TYROBP. Genetic variants or mutations of TREM2 or TYROBP have been linked to inflammatory neurodegenerative diseases associated with aging. The typical aging process goes along with microglial changes and mild neuronal loss, but the exact contribution of TREM2 is still unclear. Aged TREM2 knock-out mice showed decreased age-related neuronal loss in the substantia nigra and the hippocampus. Transcriptomic analysis of the brains of 24 months old TREM2 knock-out mice revealed 211 differentially expressed genes mostly downregulated and associated with complement activation and oxidative stress response pathways. Consistently, 24 months old TREM2 knock-out mice showed lower transcription of microglial (Aif1 and Tmem119), oxidative stress markers (Inos, Cyba, and Cybb) and complement components (C1qa, C1qb, C1qc, C3, C4b, Itgam, and Itgb2), decreased microglial numbers and expression of the microglial activation marker Cd68, as well as accumulation of oxidized lipids. Cultured microglia of TREM2 knock-out mice showed reduced phagocytosis and oxidative burst. Thus, microglial TREM2 contributes to age-related microglial changes, phagocytic oxidative burst, and loss of neurons with possible detrimental effects during physiological aging.


Assuntos
Envelhecimento/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Receptores Imunológicos/metabolismo , Fatores Etários , Envelhecimento/genética , Animais , Hipocampo/citologia , Hipocampo/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Microglia/citologia , Neurônios/citologia , Estresse Oxidativo/fisiologia , Fagocitose/fisiologia , Receptores Imunológicos/genética , Substância Negra/citologia , Substância Negra/metabolismo
13.
J Biol Chem ; 291(9): 4334-41, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26694609

RESUMO

The triggering receptor expressed on myeloid cells (TREM) 2 is a member of the immunoglobulin superfamily of receptors and mediates signaling in immune cells via engagement of its co-receptor DNAX-activating protein of 12 kDa (DAP12). Homozygous mutations in TREM2 or DAP12 cause Nasu-Hakola disease, which is characterized by bone abnormalities and dementia. Recently, a variant of TREM2 has also been associated with an increased risk for Alzheimer disease. The selective expression of TREM2 on immune cells and its association with different forms of dementia indicate a contribution of this receptor in common pathways of neurodegeneration.


Assuntos
Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Células Mieloides/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Receptores Imunológicos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Humanos , Lipodistrofia/genética , Lipodistrofia/imunologia , Lipodistrofia/metabolismo , Lipodistrofia/patologia , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , Mutação , Células Mieloides/imunologia , Células Mieloides/patologia , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia , Neurônios/imunologia , Neurônios/patologia , Osteocondrodisplasias/genética , Osteocondrodisplasias/imunologia , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patologia , Receptores Imunológicos/genética , Panencefalite Esclerosante Subaguda/genética , Panencefalite Esclerosante Subaguda/imunologia , Panencefalite Esclerosante Subaguda/metabolismo , Panencefalite Esclerosante Subaguda/patologia , Receptor Gatilho 1 Expresso em Células Mieloides
14.
J Biol Chem ; 291(20): 10528-40, 2016 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-26957541

RESUMO

The disaccharide trehalose is commonly considered to stimulate autophagy. Cell treatment with trehalose could decrease cytosolic aggregates of potentially pathogenic proteins, including mutant huntingtin, α-synuclein, and phosphorylated tau that are associated with neurodegenerative diseases. Here, we demonstrate that trehalose also alters the metabolism of the Alzheimer disease-related amyloid precursor protein (APP). Cell treatment with trehalose decreased the degradation of full-length APP and its C-terminal fragments. Trehalose also reduced the secretion of the amyloid-ß peptide. Biochemical and cell biological experiments revealed that trehalose alters the subcellular distribution and decreases the degradation of APP C-terminal fragments in endolysosomal compartments. Trehalose also led to strong accumulation of the autophagic marker proteins LC3-II and p62, and decreased the proteolytic activation of the lysosomal hydrolase cathepsin D. The combined data indicate that trehalose decreases the lysosomal metabolism of APP by altering its endocytic vesicular transport.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Trealose/metabolismo , Trealose/farmacologia , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Células HEK293 , Células Hep G2 , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Fragmentos de Peptídeos/metabolismo , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Serina-Treonina Quinases TOR/metabolismo
15.
J Biol Chem ; 291(31): 16059-67, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27252381

RESUMO

Neurodegeneration is characterized by the ubiquitous presence of modifications in protein deposits. Despite their potential significance in the initiation and progression of neurodegenerative diseases, the effects of posttranslational modifications on the molecular properties of protein aggregates are largely unknown. Here, we study the Alzheimer disease-related amyloid-ß (Aß) peptide and investigate how phosphorylation at serine 8 affects the structure of Aß aggregates. Serine 8 is shown to be located in a region of high conformational flexibility in monomeric Aß, which upon phosphorylation undergoes changes in local conformational dynamics. Using hydrogen-deuterium exchange NMR and fluorescence quenching techniques, we demonstrate that Aß phosphorylation at serine 8 causes structural changes in the N-terminal region of Aß aggregates in favor of less compact conformations. Structural changes induced by serine 8 phosphorylation can provide a mechanistic link between phosphorylation and other biological events that involve the N-terminal region of Aß aggregates. Our data therefore support an important role of posttranslational modifications in the structural polymorphism of amyloid aggregates and their modulatory effect on neurodegeneration.


Assuntos
Peptídeos beta-Amiloides/química , Agregação Patológica de Proteínas , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Medição da Troca de Deutério , Humanos , Ressonância Magnética Nuclear Biomolecular , Fosforilação , Serina/química , Serina/metabolismo
16.
Glia ; 65(7): 1103-1118, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28370426

RESUMO

The Eph-ephrin system plays pivotal roles in cell adhesion and migration. The receptor-like functions of the ephrin ligands allow the regulation of intracellular processes via reverse signaling. γ-Secretase mediated processing of ephrin-B has previously been linked to activation of Src, a kinase crucial for focal adhesion and podosome phosphorylation. Here, we analyzed the role of γ-secretase in the stimulation of reverse ephrin-B2 signaling in the migration of mouse embryonic stem cell derived microglia. The proteolytic generation of the ephrin-B2 intracellular domain (ICD) by γ-secretase stimulates Src and focal adhesion kinase (FAK). Inhibition of γ-secretase decreased the phosphorylation of Src and FAK, and reduced cell motility. These effects were associated with enlargement of the podosomal surface. Interestingly, expression of ephrin-B2 ICD could rescue these effects, indicating that this proteolytic fragment mediates the activation of Src and FAK, and thereby regulates podosomal dynamics in microglial cells. Together, these results identify γ-secretase as well as ephrin-B2 as regulators of microglial migration.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Movimento Celular/fisiologia , Citoplasma/metabolismo , Efrina-B2/metabolismo , Microglia/citologia , Microglia/fisiologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Animais Recém-Nascidos , Movimento Celular/genética , Embrião de Mamíferos , Efrina-B2/genética , Quinase 1 de Adesão Focal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Receptor EphB1/metabolismo , Transdução de Sinais/genética , Células-Tronco/fisiologia
17.
J Neurochem ; 143(4): 445-454, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28940294

RESUMO

γ-Secretase is an intramembrane cleaving protease involved in the generation of the Alzheimer's disease (AD)-associated amyloid ß peptide (Aß). γ-Secretase is ubiquitously expressed in different organs, and also in different cell types of the human brain. Besides the involvement in the proteolytic generation of Aß from the amyloid precursor protein, γ-secretase cleaves many additional protein substrates, suggesting pleiotropic functions under physiological and pathophysiological conditions. Microglia exert important functions during brain development and homeostasis in adulthood, and accumulating evidence indicates that microglia and neuroinflammatory processes contribute to the pathogenesis of neurodegenerative diseases. Recent studies demonstrate functional implications of γ-secretase in microglia, suggesting that alterations in γ-secretase activity could contribute to AD pathogenesis by modulation of microglia and related neuroinflammatory processes during neurodegeneration. In this review, we discuss the involvement of γ-secretase in the regulation of microglial functions, and the potential relevance of these processes under physiological and pathophysiological conditions. This article is part of the series "Beyond Amyloid".


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/enzimologia , Microglia/enzimologia , Doenças Neurodegenerativas/enzimologia , Animais , Humanos , Inflamação/patologia , Microglia/patologia , Doenças Neurodegenerativas/patologia
18.
J Neuroinflammation ; 13: 17, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26792193

RESUMO

BACKGROUND: Triggering receptor expressed on myeloid cells-2 (TREM2) exerts important functions in the regulation of monocytes, like dendritic cells, osteoclasts, tissue macrophages, and microglia. Mutations in TREM2 are associated with several diseases, including Nasu-Hakola disease, frontotemporal dementia, and Alzheimer's disease (AD). TREM2 undergoes sequential proteolytic processing by ectodomain shedding and intramembrane proteolysis. FINDINGS: We show that inhibition of γ-secretase-dependent cleavage of the TREM2 C-terminal fragment in cellular membranes interferes with TREM2-dependent signaling and cellular function. Inhibition of γ-secretase decreases membrane-proximal signaling and intracellular Ca(2+) response. Decreased signaling alters morphological changes and phagocytic activity of cells upon selective stimulation of TREM2. CONCLUSIONS: The data demonstrate the importance of γ-secretase-dependent intramembrane processing in TREM2-mediated signaling and, thus, a functional relation of two AD-associated proteins.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Glicoproteínas de Membrana/metabolismo , Células Mieloides/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais/fisiologia , Animais , Células COS , Sinalização do Cálcio/fisiologia , Linhagem Celular Transformada , Chlorocebus aethiops , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Microglia/metabolismo , Modelos Biológicos , Fagocitose/fisiologia , Receptores Imunológicos/genética , Fatores de Tempo , Transfecção
19.
Biol Chem ; 397(8): 777-90, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27078672

RESUMO

Proteolytic processing of the amyloid precursor protein (APP) leads to amyloid-ß (Aß) peptides. So far, the mechanism of APP processing is insufficiently characterized at the molecular level. Whereas the knowledge of Aß generation by several proteases has been expanded, the contribution of the Kunitz-type protease inhibitor domain (KPI) present in two major APP isoforms to the complex proteolytic processing of APP is poorly understood. In this study, we have identified KPI-containing APP as a very potent, slow-binding inhibitor for the membrane-bound proteolytic regulator of iron homeostasis matriptase-2 by forming stable complexes with its target protease in HEK cells. Inhibition and complex formation depend on the intact KPI domain. By inhibiting matriptase-2, KPI-containing APP is protected from matriptase-2-mediated proteolysis within the Aß region, thus preventing the generation of N-terminally truncated Aß.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Sequência de Aminoácidos , Precursor de Proteína beta-Amiloide/análise , Células Cultivadas , Células HEK293 , Humanos , Cinética , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA