Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
1.
Alzheimers Dement ; 19(8): 3563-3574, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36825551

RESUMEN

INTRODUCTION: Cerebrospinal fluid (CSF) biomarkers for specific cellular disease processes are lacking for tauopathies. In this translational study we aimed to identify CSF biomarkers reflecting early tau pathology-associated unfolded protein response (UPR) activation. METHODS: We employed mass spectrometry proteomics and targeted immunoanalysis in a combination of biomarker discovery in primary mouse neurons in vitro and validation in patient CSF from two independent large multicentre cohorts (EMIF-AD MBD, n = 310; PRIDE, n = 771). RESULTS: First, we identify members of the protein disulfide isomerase (PDI) family in the neuronal UPR-activated secretome and validate secretion upon tau aggregation in vitro. Next, we demonstrate that PDIA1 and PDIA3 levels correlate with total- and phosphorylated-tau levels in CSF. PDIA1 levels are increased in CSF from AD patients compared to controls and patients with tau-unrelated frontotemporal and Lewy body dementia (LBD). HIGHLIGHTS: Neuronal unfolded protein response (UPR) activation induces the secretion of protein disulfide isomerases (PDIs) in vitro. PDIA1 is secreted upon tau aggregation in neurons in vitro. PDIA1 and PDIA3 levels correlate with total and phosphorylated tau levels in CSF. PDIA1 levels are increased in CSF from Alzheimer's disease (AD) patients compared to controls. PDIA1 levels are not increased in CSF from tau-unrelated frontotemporal dementia (FTD) and Lewy body dementia (LBD) patients.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad por Cuerpos de Lewy , Animales , Ratones , Enfermedad por Cuerpos de Lewy/líquido cefalorraquídeo , Proteínas tau/líquido cefalorraquídeo , Proteína Disulfuro Isomerasas , Péptidos beta-Amiloides/líquido cefalorraquídeo , Fosforilación , Enfermedad de Alzheimer/líquido cefalorraquídeo , Biomarcadores/líquido cefalorraquídeo , Fragmentos de Péptidos/líquido cefalorraquídeo
2.
Acta Neuropathol ; 138(6): 943-970, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31456031

RESUMEN

Granulovacuolar degeneration bodies (GVBs) are membrane-bound vacuolar structures harboring a dense core that accumulate in the brains of patients with neurodegenerative disorders, including Alzheimer's disease and other tauopathies. Insight into the origin of GVBs and their connection to tau pathology has been limited by the lack of suitable experimental models for GVB formation. Here, we used confocal, automated, super-resolution and electron microscopy to demonstrate that the seeding of tau pathology triggers the formation of GVBs in different mouse models in vivo and in primary mouse neurons in vitro. Seeding-induced intracellular tau aggregation, but not seed exposure alone, causes GVB formation in cultured neurons, but not in astrocytes. The extent of tau pathology strongly correlates with the GVB load. Tau-induced GVBs are immunoreactive for the established GVB markers CK1δ, CK1ɛ, CHMP2B, pPERK, peIF2α and pIRE1α and contain a LAMP1- and LIMP2-positive single membrane that surrounds the dense core and vacuole. The proteolysis reporter DQ-BSA is detected in the majority of GVBs, demonstrating that GVBs contain degraded endocytic cargo. GFP-tagged CK1δ accumulates in the GVB core, whereas GFP-tagged tau or GFP alone does not, indicating selective targeting of cytosolic proteins to GVBs. Taken together, we established the first in vitro model for GVB formation by seeding tau pathology in primary neurons. The tau-induced GVBs have the marker signature and morphological characteristics of GVBs in the human brain. We show that GVBs are lysosomal structures distinguished by the accumulation of a characteristic subset of proteins in a dense core.


Asunto(s)
Lisosomas/patología , Neuronas/patología , Tauopatías/patología , Vacuolas/patología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos , Femenino , Humanos , Lisosomas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Tauopatías/metabolismo , Vacuolas/metabolismo , Proteínas tau/genética
3.
Biomolecules ; 10(8)2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32707908

RESUMEN

Neurons are highly specialized cells that continuously and extensively communicate with other neurons, as well as glia cells. During their long lifetime, the post-mitotic neurons encounter many stressful situations that can disrupt protein homeostasis (proteostasis). The importance of tight protein quality control is illustrated by neurodegenerative disorders where disturbed neuronal proteostasis causes neuronal dysfunction and loss. For their unique function, neurons require regulated and long-distance transport of membrane-bound cargo and organelles. This highlights the importance of protein quality control in the neuronal endomembrane system, to which the unfolded protein response (UPR) is instrumental. The UPR is a highly conserved stress response that is present in all eukaryotes. However, recent studies demonstrate the existence of cell-type-specific aspects of the UPR, as well as cell non-autonomous UPR signaling. Here we discuss these novel insights in view of the complex cellular architecture of the brain and the implications for neurodegenerative diseases.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Retículo Endoplásmico/genética , Enfermedades Neurodegenerativas/genética , Proteostasis/genética , Respuesta de Proteína Desplegada/genética , Animales , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Humanos , Modelos Genéticos , Enfermedades Neurodegenerativas/metabolismo , Transducción de Señal/genética
4.
Sci Rep ; 9(1): 1567, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733486

RESUMEN

Accumulation of misfolded proteins in the endoplasmic reticulum (ER), defined as ER stress, results in activation of the unfolded protein response (UPR). UPR activation is commonly observed in neurodegenerative diseases. ER stress can trigger unconventional secretion mediated by Golgi reassembly and stacking proteins (GRASP) relocalization in cell lines. Here we study the regulation of GRASP55 by the UPR upon pharmacological induction of ER stress in primary mouse neurons. We demonstrate that UPR activation induces mRNA and protein expression of GRASP55, but not GRASP65, in cortical neurons. UPR activation does not result in relocalization of GRASP55. UPR-induced GRASP55 expression is reduced by inhibition of the PERK pathway of the UPR and abolished by inhibition of the endonuclease activity of the UPR transducer IRE1. Expression of the IRE1 target XBP1s in the absence of ER stress is not sufficient to increase GRASP55 expression. Knockdown of GRASP55 affects neither induction nor recovery of the UPR. We conclude that the UPR regulates the unconventional secretion factor GRASP55 via a mechanism that requires the IRE1 and the PERK pathway of the UPR in neurons.


Asunto(s)
Proteínas de la Matriz de Golgi/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Estrés del Retículo Endoplásmico , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Expresión Génica , Proteínas de la Matriz de Golgi/genética , Humanos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Proteína 1 de Unión a la X-Box/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda