Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Immunity ; 2024 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-39419029

RESUMO

The seeded growth of pathogenic protein aggregates underlies the pathogenesis of Alzheimer's disease (AD), but how this pathological cascade is initiated is not fully understood. Sporadic AD is linked genetically to apolipoprotein E (APOE) and other genes expressed in microglia related to immune, lipid, and endocytic functions. We generated a transgenic knockin mouse expressing HaloTag-tagged APOE and optimized experimental protocols for the biochemical purification of APOE, which enabled us to identify fibrillary aggregates of APOE in mice with amyloid-ß (Aß) amyloidosis and in human AD brain autopsies. These APOE aggregates that stained positive for ß sheet-binding dyes triggered Aß amyloidosis within the endo-lysosomal system of microglia, in a process influenced by microglial lipid metabolism and the JAK/STAT signaling pathway. Taking these observations together, we propose a model for the onset of Aß amyloidosis in AD, suggesting that the endocytic uptake and aggregation of APOE by microglia can initiate Aß plaque formation.

2.
Brain ; 2024 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-39319704

RESUMO

Remyelination is a crucial regenerative process in demyelinating diseases, limiting persisting damage to the central nervous system (CNS). It restores saltatory nerve conduction and ensures trophic support of axons. In multiple sclerosis (MS) patients, remyelination has been observed in both white and grey matter and found to be more efficient in the cortex. Brain-enriched myelin-associated protein 1 (BCAS1) identifies oligodendrocyte lineage cells in the stage of active myelin formation in development and regeneration. Other than in the white matter, BCAS1+ oligodendrocytes are maintained at high densities in the cortex throughout life. Here, we investigated cortical lesions in human biopsy and autopsy tissue from patients with MS in direct comparison to demyelinating mouse models and demonstrate that following a demyelinating insult BCAS1+ oligodendrocytes in remyelinating cortical lesions shift from a quiescent to an activated, internode-forming morphology co-expressing myelin-associated glycoprotein (MAG), necessary for axonal contact formation. Noteworthy, activated BCAS1+ oligodendrocytes are found at early time points of experimental demyelination amidst ongoing inflammation. In human tissue, activated BCAS 1+ oligodendrocytes correlate with the density of myeloid cells, further supporting their involvement in an immediate regenerative response. Furthermore, studying the microscopically normal appearing non demyelinated cortex in patients with chronic MS, we find a shift from quiescent BCAS1+ oligodendrocytes to mature, myelin-maintaining oligodendrocytes, suggesting oligodendrocyte differentiation and limited replenishment of BCAS1+ oligodendrocytes in long-standing disease. We also demonstrate that part of perineuronal satellite oligodendrocytes are BCAS1+ and contribute to remyelination in human and experimental cortical demyelination. In summary, our results provide evidence from human tissue and experimental models that BCAS1+ cells in the adult cortex represent a population of pre-differentiated oligodendrocytes that rapidly react after a demyelinating insult thus enabling immediate myelin regeneration. In addition, our data suggest that limited replenishment of BCAS1+ oligodendrocytes may contribute to the remyelination failure observed in the cortex in chronic MS.

3.
Nat Neurosci ; 27(8): 1468-1474, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38937583

RESUMO

Age-related myelin damage induces inflammatory responses, yet its involvement in Alzheimer's disease remains uncertain, despite age being a major risk factor. Using a mouse model of Alzheimer's disease, we found that amyloidosis itself triggers age-related oligodendrocyte and myelin damage. Mechanistically, CD8+ T cells promote the progressive accumulation of abnormally interferon-activated microglia that display myelin-damaging activity. Thus, our data suggest that immune responses against myelinating oligodendrocytes may contribute to neurodegenerative diseases with amyloidosis.


Assuntos
Doença de Alzheimer , Amiloidose , Modelos Animais de Doenças , Microglia , Bainha de Mielina , Animais , Microglia/patologia , Microglia/metabolismo , Microglia/imunologia , Bainha de Mielina/patologia , Bainha de Mielina/metabolismo , Camundongos , Amiloidose/patologia , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/imunologia , Linfócitos T CD8-Positivos/imunologia , Camundongos Transgênicos , Oligodendroglia/patologia , Oligodendroglia/metabolismo , Camundongos Endogâmicos C57BL
4.
Sci Transl Med ; 16(750): eadj7308, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38838131

RESUMO

Progranulin (PGRN) haploinsufficiency is a major risk factor for frontotemporal lobar degeneration with TAR DNA-binding protein 43 (TDP-43) pathology (FTLD-GRN). Multiple therapeutic strategies are in clinical development to restore PGRN in the CNS, including gene therapy. However, a limitation of current gene therapy approaches aimed to alleviate FTLD-associated pathologies may be their inefficient brain exposure and biodistribution. We therefore developed an adeno-associated virus (AAV) targeting the liver (L) to achieve sustained peripheral expression of a transferrin receptor (TfR) binding, brain-penetrant (b) PGRN variant [AAV(L):bPGRN] in two mouse models of FTLD-GRN, namely, Grn knockout and GrnxTmem106b double knockout mice. This therapeutic strategy avoids potential safety and biodistribution issues of CNS-administered AAVs and maintains sustained concentrations of PGRN in the brain after a single dose. AAV(L):bPGRN treatment reduced several FTLD-GRN-associated pathologies including severe motor function deficits, aberrant TDP-43 phosphorylation, dysfunctional protein degradation, lipid metabolism, gliosis, and neurodegeneration in the brain. The potential translatability of our findings was tested in an in vitro model using cocultured human induced pluripotent stem cell (hiPSC)-derived microglia lacking PGRN and TMEM106B and wild-type hiPSC-derived neurons. As in mice, aberrant TDP-43, lysosomal dysfunction, and neuronal loss were ameliorated after treatment with exogenous TfR-binding protein transport vehicle fused to PGRN (PTV:PGRN). Together, our studies suggest that peripherally administered brain-penetrant PGRN replacement strategies ameliorate FTLD-GRN relevant phenotypes including TDP-43 pathology, neurodegeneration, and behavioral deficits. Our data provide preclinical proof of concept for the use of this AAV platform for treatment of FTLD-GRN and potentially other CNS disorders.


Assuntos
Encéfalo , Dependovirus , Modelos Animais de Doenças , Degeneração Lobar Frontotemporal , Camundongos Knockout , Progranulinas , Animais , Humanos , Camundongos , Encéfalo/metabolismo , Encéfalo/patologia , Dependovirus/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Degeneração Lobar Frontotemporal/metabolismo , Degeneração Lobar Frontotemporal/patologia , Terapia Genética , Fosforilação , Progranulinas/metabolismo , Progranulinas/genética , Receptores da Transferrina/metabolismo
5.
Science ; 381(6655): 285-290, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37471539

RESUMO

Disruption of the physiologic sleep-wake cycle and low melatonin levels frequently accompany cardiac disease, yet the underlying mechanism has remained enigmatic. Immunostaining of sympathetic axons in optically cleared pineal glands from humans and mice with cardiac disease revealed their substantial denervation compared with controls. Spatial, single-cell, nuclear, and bulk RNA sequencing traced this defect back to the superior cervical ganglia (SCG), which responded to cardiac disease with accumulation of inflammatory macrophages, fibrosis, and the selective loss of pineal gland-innervating neurons. Depletion of macrophages in the SCG prevented disease-associated denervation of the pineal gland and restored physiological melatonin secretion. Our data identify the mechanism by which diurnal rhythmicity in cardiac disease is disturbed and suggest a target for therapeutic intervention.


Assuntos
Ritmo Circadiano , Cardiopatias , Macrófagos , Melatonina , Glândula Pineal , Transtornos do Sono do Ritmo Circadiano , Gânglio Cervical Superior , Animais , Humanos , Camundongos , Cardiopatias/fisiopatologia , Melatonina/metabolismo , Glândula Pineal/patologia , Glândula Pineal/fisiopatologia , Sono , Transtornos do Sono do Ritmo Circadiano/fisiopatologia , Gânglio Cervical Superior/patologia , Gânglio Cervical Superior/fisiopatologia , Macrófagos/imunologia , Fibrose
6.
EBioMedicine ; 89: 104456, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36745974

RESUMO

A major evolution from purely clinical diagnoses to biomarker supported clinical diagnosing has been occurring over the past years in neurology. High-throughput methods, such as next-generation sequencing and mass spectrometry-based proteomics along with improved neuroimaging methods, are accelerating this development. This calls for a consensus framework that is broadly applicable and provides a spot-on overview of the clinical validity of novel biomarkers. We propose a harmonized terminology and a uniform concept that stratifies biomarkers according to clinical context of use and evidence levels, adapted from existing frameworks in oncology with a strong focus on (epi)genetic markers and treatment context. We demonstrate that this framework allows for a consistent assessment of clinical validity across disease entities and that sufficient evidence for many clinical applications of protein biomarkers is lacking. Our framework may help to identify promising biomarker candidates and classify their applications by clinical context, aiming for routine clinical use of (protein) biomarkers in neurology.


Assuntos
Doenças do Sistema Nervoso , Humanos , Biomarcadores , Proteômica/métodos , Espectrometria de Massas , Neuroimagem
7.
FASEB J ; 35(11): e21962, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34613632

RESUMO

Proteolytic ectodomain shedding of membrane proteins is a fundamental mechanism to control the communication between cells and their environment. A key protease for membrane protein shedding is ADAM17, which requires a non-proteolytic subunit, either inactive Rhomboid 1 (iRhom1) or iRhom2 for its activity. While iRhom1 and iRhom2 are co-expressed in most tissues and appear to have largely redundant functions, the brain is an organ with predominant expression of iRhom1. Yet, little is known about the spatio-temporal expression of iRhom1 in mammalian brain and about its function in controlling membrane protein shedding in the nervous system. Here, we demonstrate that iRhom1 is expressed in mouse brain from the prenatal stage to adulthood with a peak in early postnatal development. In the adult mouse brain iRhom1 was widely expressed, including in cortex, hippocampus, olfactory bulb, and cerebellum. Proteomic analysis of the secretome of primary neurons using the hiSPECS method and of cerebrospinal fluid, obtained from iRhom1-deficient and control mice, identified several membrane proteins that require iRhom1 for their shedding in vitro or in vivo. One of these proteins was 'multiple-EGF-like-domains protein 10' (MEGF10), a phagocytic receptor in the brain that is linked to the removal of amyloid ß and apoptotic neurons. MEGF10 was further validated as an ADAM17 substrate using ADAM17-deficient mouse embryonic fibroblasts. Taken together, this study discovers a role for iRhom1 in controlling membrane protein shedding in the mouse brain, establishes MEGF10 as an iRhom1-dependent ADAM17 substrate and demonstrates that iRhom1 is widely expressed in murine brain.


Assuntos
Proteína ADAM17/metabolismo , Encéfalo/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Embrionárias Murinas
8.
Front Aging Neurosci ; 13: 661284, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054506

RESUMO

Objectives: In recent years several 18F-labeled amyloid PET (Aß-PET) tracers have been developed and have obtained clinical approval. There is evidence that Aß-PET perfusion can provide surrogate information about neuronal injury in neurodegenerative diseases when compared to conventional blood flow and glucose metabolism assessment. However, this paradigm has not yet been tested in neurodegenerative disorders with cortical and subcortical affection. Therefore, we investigated the performance of early acquisition 18F-flutemetamol Aß-PET in comparison to 18F-fluorodeoxyglucose (FDG)-PET in corticobasal syndrome (CBS). Methods: Subjects with clinically possible or probable CBS were recruited within the prospective Activity of Cerebral Networks, Amyloid and Microglia in Aging and Alzheimer's Disease (ActiGliA) observational study and all CBS cases with an available FDG-PET prior to Aß-PET were selected. Aß-PET was acquired 0-10 min p.i. (early-phase) and 90-110 min p.i. (late-phase) whereas FDG-PET was recorded statically from 30 to 50 min p.i. Semiquantitative regional values and asymmetry indices (AI) were compared between early-phase Aß-PET and FDG-PET. Visual assessments of hypoperfusion and hypometabolism were compared between both methods. Late-phase Aß-PET was evaluated visually for assessment of Aß-positivity. Results: Among 20 evaluated patients with CBS, 5 were Aß-positive. Early-phase Aß-PET and FDG-PET SUVr correlated highly in cortical (mean R = 0.86, range 0.77-0.92) and subcortical brain regions (mean R = 0.84, range 0.79-0.90). Strong asymmetry was observed in FDG-PET for the motor cortex (mean |AI| = 2.9%), the parietal cortex (mean |AI| = 2.9%), and the thalamus (mean |AI| = 5.5%), correlating well with AI of early-phase Aß-PET (mean R = 0.87, range 0.62-0.98). Visual assessments of hypoperfusion and hypometabolism were highly congruent. Conclusion: Early-phase Aß-PET facilitates assessment of neuronal injury in CBS for cortical and subcortical areas. Known asymmetries in CBS are captured by this method, enabling assessment of Aß-status and neuronal injury with a single radiation exposure at a single visit.

9.
EMBO Mol Med ; 12(4): e11227, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32154671

RESUMO

Triggering receptor expressed on myeloid cells 2 (TREM2) is essential for the transition of homeostatic microglia to a disease-associated microglial state. To enhance TREM2 activity, we sought to selectively increase the full-length protein on the cell surface via reducing its proteolytic shedding by A Disintegrin And Metalloproteinase (i.e., α-secretase) 10/17. We screened a panel of monoclonal antibodies against TREM2, with the aim to selectively compete for α-secretase-mediated shedding. Monoclonal antibody 4D9, which has a stalk region epitope close to the cleavage site, demonstrated dual mechanisms of action by stabilizing TREM2 on the cell surface and reducing its shedding, and concomitantly activating phospho-SYK signaling. 4D9 stimulated survival of macrophages and increased microglial uptake of myelin debris and amyloid ß-peptide in vitro. In vivo target engagement was demonstrated in cerebrospinal fluid, where nearly all soluble TREM2 was 4D9-bound. Moreover, in a mouse model for Alzheimer's disease-related pathology, 4D9 reduced amyloidogenesis, enhanced microglial TREM2 expression, and reduced a homeostatic marker, suggesting a protective function by driving microglia toward a disease-associated state.


Assuntos
Anticorpos Monoclonais/farmacologia , Glicoproteínas de Membrana/imunologia , Microglia , Mieloma Múltiplo , Receptores Imunológicos/imunologia , Peptídeos beta-Amiloides , Animais , Linhagem Celular Tumoral , Feminino , Macrófagos , Camundongos , Microglia/patologia , Ratos , Ratos Wistar
10.
J Exp Med ; 217(5)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32078678

RESUMO

Remyelination requires innate immune system function, but how exactly microglia and macrophages clear myelin debris after injury and tailor a specific regenerative response is unclear. Here, we asked whether pro-inflammatory microglial/macrophage activation is required for this process. We established a novel toxin-based spinal cord model of de- and remyelination in zebrafish and showed that pro-inflammatory NF-κB-dependent activation in phagocytes occurs rapidly after myelin injury. We found that the pro-inflammatory response depends on myeloid differentiation primary response 88 (MyD88). MyD88-deficient mice and zebrafish were not only impaired in the degradation of myelin debris, but also in initiating the generation of new oligodendrocytes for myelin repair. We identified reduced generation of TNF-α in lesions of MyD88-deficient animals, a pro-inflammatory molecule that was able to induce the generation of new premyelinating oligodendrocytes. Our study shows that pro-inflammatory phagocytic signaling is required for myelin debris degradation, for inflammation resolution, and for initiating the generation of new oligodendrocytes.


Assuntos
Doenças Desmielinizantes/patologia , Inflamação/patologia , Bainha de Mielina/metabolismo , Oligodendroglia/patologia , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Células Cultivadas , Modelos Animais de Doenças , Larva/efeitos dos fármacos , Lisofosfatidilcolinas/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Mutação/genética , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/patologia , Fator 88 de Diferenciação Mieloide/metabolismo , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Fagócitos/efeitos dos fármacos , Fagócitos/patologia , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Proteoma/metabolismo , Remielinização/efeitos dos fármacos , Medula Espinal/patologia , Fator de Necrose Tumoral alfa/farmacologia , Peixe-Zebra
11.
Nat Neurosci ; 21(9): 1196-1208, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30127427

RESUMO

Mononuclear phagocytes are key regulators of both tissue damage and repair in neuroinflammatory conditions such as multiple sclerosis. To examine divergent phagocyte phenotypes in the inflamed CNS, we introduce an in vivo imaging approach that allows us to temporally and spatially resolve the evolution of phagocyte polarization in a murine model of multiple sclerosis. We show that the initial proinflammatory polarization of phagocytes is established after spinal cord entry and critically depends on the compartment they enter. Guided by signals from the CNS environment, individual phagocytes then switch their phenotype as lesions move from expansion to resolution. Our study thus provides a real-time analysis of the temporospatial determinants and regulatory principles of phagocyte specification in the inflamed CNS.


Assuntos
Leucócitos Mononucleares/patologia , Esclerose Múltipla/patologia , Fagócitos/patologia , Animais , Astrócitos/patologia , Astrócitos/ultraestrutura , Células da Medula Óssea/patologia , Células da Medula Óssea/ultraestrutura , Polaridade Celular , Sistemas Computacionais , Encefalomielite Autoimune Experimental/patologia , Humanos , Inflamação/patologia , Leucócitos Mononucleares/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/patologia , Neuroglia/ultraestrutura , Fagócitos/ultraestrutura , Fagocitose , Fenótipo , Análise de Sequência de RNA , Medula Espinal/patologia , Medula Espinal/ultraestrutura
12.
Cell ; 173(3): 706-719.e13, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29677514

RESUMO

Cytoplasmic FUS aggregates are a pathological hallmark in a subset of patients with frontotemporal dementia (FTD) or amyotrophic lateral sclerosis (ALS). A key step that is disrupted in these patients is nuclear import of FUS mediated by the import receptor Transportin/Karyopherin-ß2. In ALS-FUS patients, this is caused by mutations in the nuclear localization signal (NLS) of FUS that weaken Transportin binding. In FTD-FUS patients, Transportin is aggregated, and post-translational arginine methylation, which regulates the FUS-Transportin interaction, is lost. Here, we show that Transportin and arginine methylation have a crucial function beyond nuclear import-namely to suppress RGG/RG-driven phase separation and stress granule association of FUS. ALS-associated FUS-NLS mutations weaken the chaperone activity of Transportin and loss of FUS arginine methylation, as seen in FTD-FUS, promote phase separation, and stress granule partitioning of FUS. Our findings reveal two regulatory mechanisms of liquid-phase homeostasis that are disrupted in FUS-associated neurodegeneration.


Assuntos
Arginina/química , Proteína FUS de Ligação a RNA/química , beta Carioferinas/química , Transporte Ativo do Núcleo Celular , Motivos de Aminoácidos , Citoplasma/metabolismo , Metilação de DNA , DNA Complementar/metabolismo , Densitometria , Degeneração Lobar Frontotemporal/metabolismo , Células HeLa , Homeostase , Humanos , Carioferinas/química , Espectroscopia de Ressonância Magnética , Metilação , Chaperonas Moleculares/química , Mutação , Doenças Neurodegenerativas/metabolismo , Ligação Proteica , Domínios Proteicos
13.
Sci Transl Med ; 9(419)2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-29212715

RESUMO

Investigations into brain function and disease depend on the precise classification of neural cell types. Cells of the oligodendrocyte lineage differ greatly in their morphology, but accurate identification has thus far only been possible for oligodendrocyte progenitor cells and mature oligodendrocytes in humans. We find that breast carcinoma amplified sequence 1 (BCAS1) expression identifies an oligodendroglial subpopulation in the mouse and human brain. These cells are newly formed, myelinating oligodendrocytes that segregate from oligodendrocyte progenitor cells and mature oligodendrocytes and mark regions of active myelin formation in development and in the adult. We find that BCAS1+ oligodendrocytes are restricted to the fetal and early postnatal human white matter but remain in the cortical gray matter until old age. BCAS1+ oligodendrocytes are reformed after experimental demyelination and found in a proportion of chronic white matter lesions of patients with multiple sclerosis (MS) even in a subset of patients with advanced disease. Our work identifies a means to map ongoing myelin formation in health and disease and presents a potential cellular target for remyelination therapies in MS.


Assuntos
Esclerose Múltipla/metabolismo , Proteínas de Neoplasias/metabolismo , Oligodendroglia/metabolismo , Animais , Doenças Desmielinizantes , Humanos , Camundongos , Esclerose Múltipla/patologia , Bainha de Mielina/metabolismo
14.
Nat Commun ; 7: 13275, 2016 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-27848954

RESUMO

Oligodendrocyte damage is a central event in the pathogenesis of the common neuroinflammatory condition, multiple sclerosis (MS). Where and how oligodendrocyte damage is initiated in MS is not completely understood. Here, we use a combination of light and electron microscopy techniques to provide a dynamic and highly resolved view of oligodendrocyte damage in neuroinflammatory lesions. We show that both in MS and in its animal model structural damage is initiated at the myelin sheaths and only later spreads to the oligodendrocyte cell body. Early myelin damage itself is characterized by the formation of local myelin out-foldings-'myelinosomes'-, which are surrounded by phagocyte processes and promoted in their formation by anti-myelin antibodies and complement. The presence of myelinosomes in actively demyelinating MS lesions suggests that oligodendrocyte damage follows a similar pattern in the human disease, where targeting demyelination by therapeutic interventions remains a major open challenge.


Assuntos
Esclerose Múltipla/patologia , Bainha de Mielina/patologia , Oligodendroglia/patologia , Animais , Anticorpos/metabolismo , Proteínas do Sistema Complemento/metabolismo , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Humanos , Imageamento Tridimensional , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Bainha de Mielina/ultraestrutura , Oligodendroglia/ultraestrutura , Proteínas Opsonizantes/metabolismo , Organelas/metabolismo , Organelas/ultraestrutura
15.
Biophys J ; 110(11): 2441-2450, 2016 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-27276262

RESUMO

Myelin is a multilayered membrane that ensheathes axonal fibers in the vertebrate nervous system, allowing fast propagation of nerve action potentials. It contains densely packed lipids, lacks an actin-based cytocortex, and requires myelin basic protein (MBP) as its major structural component. This protein is the basic constituent of the proteinaceous meshwork that is localized between adjacent cytoplasmic membranes of the myelin sheath. Yet, it is not clear how MBP influences the organization and dynamics of the lipid constituents of myelin. Here, we used optical stimulated emission depletion super-resolution microscopy in combination with fluorescence correlation spectroscopy to assess the characteristics of diffusion of different fluorescent lipid analogs in myelin membrane sheets of cultured oligodendrocytes and in micrometer-sized domains that were induced by MBP in live epithelial PtK2 cells. Lipid diffusion was significantly faster and less anomalous both in oligodendrocytes and inside the MBP-rich domains of PtK2 cells compared with undisturbed live PtK2 cells. Our data show that MBP reorganizes lipid diffusion, possibly by preventing the buildup of an actin-based cytocortex and by preventing most membrane proteins from entering the myelin sheath region. Yet, in contrast to myelin sheets in oligodendrocytes, the MBP-induced domains in epithelial PtK2 cells demonstrate no change in lipid order, indicating that segregation of long-chain lipids into myelin sheets is a process specific to oligodendrocytes.


Assuntos
Membrana Celular/metabolismo , Etanolaminas/metabolismo , Galactosilceramidas/metabolismo , Proteína Básica da Mielina/metabolismo , Esfingomielinas/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Difusão , Células Epiteliais/metabolismo , Corantes Fluorescentes , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Microscopia/métodos , Oligodendroglia/metabolismo , Potoroidae , Espectrometria de Fluorescência
16.
Stem Cells ; 34(8): 2115-29, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27068685

RESUMO

Adult neural stem cells with the ability to generate neurons and glia cells are active throughout life in both the dentate gyrus (DG) and the subventricular zone (SVZ). Differentiation of adult neural stem cells is induced by cell fate determinants like the transcription factor Prox1. Evidence has been provided for a function of Prox1 as an inducer of neuronal differentiation within the DG. We now show that within the SVZ Prox1 induces differentiation into oligodendrocytes. Moreover, we find that loss of Prox1 expression in vivo reduces cell migration into the corpus callosum, where the few Prox1 deficient SVZ-derived remaining cells fail to differentiate into oligodendrocytes. Thus, our work uncovers a novel function of Prox1 as a fate determinant for oligodendrocytes in the adult mammalian brain. These data indicate that the neurogenic and oligodendrogliogenic lineages in the two adult neurogenic niches exhibit a distinct requirement for Prox1, being important for neurogenesis in the DG but being indispensable for oligodendrogliogenesis in the SVZ. Stem Cells 2016;34:2115-2129.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Proteínas de Homeodomínio/metabolismo , Ventrículos Laterais/citologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oligodendroglia/citologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Movimento Celular/genética , Células Cultivadas , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos , Neurogênese/genética , Bulbo Olfatório/citologia , Bulbo Olfatório/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/genética , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Receptores Notch/genética , Receptores Notch/metabolismo
17.
Nat Cell Biol ; 18(1): 132-8, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26571211

RESUMO

Genetic defects in myelin formation and maintenance cause leukodystrophies, a group of white matter diseases whose mechanistic underpinnings are poorly understood. Hypomyelination and congenital cataract (HCC), one of these disorders, is caused by mutations in FAM126A, a gene of unknown function. We show that FAM126A, also known as hyccin, regulates the synthesis of phosphatidylinositol 4-phosphate (PtdIns(4)P), a determinant of plasma membrane identity. HCC patient fibroblasts exhibit reduced PtdIns(4)P levels. FAM126A is an intrinsic component of the plasma membrane phosphatidylinositol 4-kinase complex that comprises PI4KIIIα and its adaptors TTC7 and EFR3 (refs 5,7). A FAM126A-TTC7 co-crystal structure reveals an all-α-helical heterodimer with a large protein-protein interface and a conserved surface that may mediate binding to PI4KIIIα. Absence of FAM126A, the predominant FAM126 isoform in oligodendrocytes, destabilizes the PI4KIIIα complex in mouse brain and patient fibroblasts. We propose that HCC pathogenesis involves defects in PtdIns(4)P production in oligodendrocytes, whose specialized function requires massive plasma membrane expansion and thus generation of PtdIns(4)P and downstream phosphoinositides. Our results point to a role for FAM126A in supporting myelination, an important process in development and also following acute exacerbations in multiple sclerosis.


Assuntos
Membrana Celular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Fosfatos de Fosfatidilinositol/biossíntese , Animais , Humanos , Camundongos , Mutação/genética , Fosfatos de Fosfatidilinositol/genética , Estrutura Terciária de Proteína , Transporte Proteico/genética , Transporte Proteico/fisiologia
18.
Acta Neuropathol ; 129(5): 695-713, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25778619

RESUMO

Extracellular α-Synuclein has been implicated in interneuronal propagation of disease pathology in Parkinson's Disease. How α-Synuclein is released into the extracellular space is still unclear. Here, we show that α-Synuclein is present in extracellular vesicles in the central nervous system. We find that sorting of α-Synuclein in extracellular vesicles is regulated by sumoylation and that sumoylation acts as a sorting factor for targeting of both, cytosolic and transmembrane proteins, to extracellular vesicles. We provide evidence that the SUMO-dependent sorting utilizes the endosomal sorting complex required for transport (ESCRT) by interaction with phosphoinositols. Ubiquitination of cargo proteins is so far the only known determinant for ESCRT-dependent sorting into the extracellular vesicle pathway. Our study reveals a function of SUMO protein modification as a Ubiquitin-independent ESCRT sorting signal, regulating the extracellular vesicle release of α-Synuclein. We deciphered in detail the molecular mechanism which directs α-Synuclein into extracellular vesicles which is of highest relevance for the understanding of Parkinson's disease pathogenesis and progression at the molecular level. We furthermore propose that sumo-dependent sorting constitutes a mechanism with more general implications for cell biology.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Vesículas Extracelulares/metabolismo , Oligodendroglia/citologia , Proteína SUMO-1/metabolismo , Sumoilação/fisiologia , alfa-Sinucleína/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Vesículas Extracelulares/genética , Camundongos , Oligodendroglia/metabolismo , Proteína SUMO-1/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , alfa-Sinucleína/genética
19.
Neurology ; 81(19): e146-7, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24190004

RESUMO

A 21-year-old man presented with headache, hypotonia, hypothermia, and somnolence, deteriorating to a Glasgow Coma Scale score of 3 within days. Hormonal testing revealed panhypopituitarism. His cerebral MRI showed a gadolinium-enhancing lesion in the pituitary gland with adjacent changes to the hypothalamus, midbrain, and basal ganglia (figures 1 and 2). Therapy with prednisolone resulted in rapid improvement. Ma2 antibodies were found in the patient's serum and CSF. FDG-PET demonstrated a tumor mass in the superior mediastinum and histology revealed a mediastinal seminoma. Ma2 antibody-mediated paraneoplastic disease has to be considered as a rare differential diagnosis in patients presenting with acute panhypopituitarism.(1.)


Assuntos
Antígenos de Neoplasias/metabolismo , Encefalite/diagnóstico , Hipopituitarismo/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Córtex Cerebral/patologia , Encefalite/líquido cefalorraquidiano , Encefalite/diagnóstico por imagem , Fluordesoxiglucose F18 , Escala de Coma de Glasgow , Humanos , Hipopituitarismo/diagnóstico , Imageamento por Ressonância Magnética , Masculino , Hipófise/patologia , Tomografia por Emissão de Pósitrons , Adulto Jovem
20.
Nat Commun ; 4: 2123, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23828504

RESUMO

Loss of folate receptor-α function is associated with cerebral folate transport deficiency and childhood-onset neurodegeneration. To clarify the mechanism of cerebral folate transport at the blood-cerebrospinal fluid barrier, we investigate the transport of 5-methyltetrahydrofolate in polarized cells. Here we identify folate receptor-α-positive intralumenal vesicles within multivesicular bodies and demonstrate the directional cotransport of human folate receptor-α, and labelled folate from the basolateral to the apical membrane in rat choroid plexus cells. Both the apical medium of folate receptor-α-transfected rat choroid plexus cells and human cerebrospinal fluid contain folate receptor-α-positive exosomes. Loss of folate receptor-α-expressing cerebrospinal fluid exosomes correlates with severely reduced 5-methyltetrahydrofolate concentration, corroborating the importance of the folate receptor-α-mediated folate transport in the cerebrospinal fluid. Intraventricular injections of folate receptor-α-positive and -negative exosomes into mouse brains demonstrate folate receptor-α-dependent delivery of exosomes into the brain parenchyma. Our results unravel a new pathway of folate receptor-α-dependent exosome-mediated folate delivery into the brain parenchyma and opens new avenues for cerebral drug targeting.


Assuntos
Plexo Corióideo/citologia , Plexo Corióideo/metabolismo , Exossomos/metabolismo , Ácido Fólico/metabolismo , Transcitose , Adolescente , Adulto , Animais , Polaridade Celular/efeitos dos fármacos , Criança , Plexo Corióideo/ultraestrutura , Vesículas Citoplasmáticas/efeitos dos fármacos , Vesículas Citoplasmáticas/metabolismo , Cães , Exossomos/efeitos dos fármacos , Exossomos/ultraestrutura , Feminino , Receptor 1 de Folato/metabolismo , Humanos , Células Madin Darby de Rim Canino , Masculino , Camundongos , Modelos Biológicos , Monensin/farmacologia , Transporte Proteico/efeitos dos fármacos , Transportador de Folato Acoplado a Próton/metabolismo , Ratos , Tetra-Hidrofolatos/metabolismo , Transcitose/efeitos dos fármacos , Transferrina/farmacologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA