Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Glia ; 69(8): 1852-1881, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33634529

RESUMEN

Astrocytes regulate synaptic communication and are essential for proper brain functioning. In Alzheimer's disease (AD) astrocytes become reactive, which is characterized by an increased expression of intermediate filament proteins and cellular hypertrophy. Reactive astrocytes are found in close association with amyloid-beta (Aß) deposits. Synaptic communication and neuronal network function could be directly modulated by reactive astrocytes, potentially contributing to cognitive decline in AD. In this review, we focus on reactive astrocytes as treatment targets in AD in the APPswePS1dE9 AD mouse model, a widely used model to study amyloidosis and gliosis. We first give an overview of the model; that is, how it was generated, which cells express the transgenes, and the effect of its genetic background on Aß pathology. Subsequently, to determine whether modifying reactive astrocytes in AD could influence pathogenesis and cognition, we review studies using this mouse model in which interventions were directly targeted at reactive astrocytes or had an indirect effect on reactive astrocytes. Overall, studies specifically targeting astrocytes to reduce astrogliosis showed beneficial effects on cognition, which indicates that targeting astrocytes should be included in developing novel therapies for AD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Gliosis/patología , Ratones , Ratones Transgénicos
2.
Front Cell Neurosci ; 13: 503, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31798416

RESUMEN

Glia alterations in the anterior cingulate cortex (ACC) and dorsolateral prefrontal cortex (DLPFC) have been postulated to play an important role in the pathophysiology of psychiatric disorders. Astroglia is the most abundant type of glial cells in the central nervous system. The expression levels of astrocyte markers (glial fibrillary acidic protein (GFAP), synemin-α, synemin-ß, vimentin, nestin) in isolated gray matter from postmortem ACC and DLPFC were determined to investigate the possible involvement of astrocytes in depression. Donors were aged non-suicidal subjects with bipolar disorder (BPD) or major depressive disorder (MDD), and matched controls. GFAP mRNA levels were significantly increased in the ACC of BPD patients. However, GFAP immunohistochemistry showed that the area fraction of GFAP immunoreactive astrocytes was decreased in the ACC of BPD patients, while there were no changes in the cell density and integrated optical density (IOD), indicating that there might be a reduction of GFAP-positive astrocyte processes and remodeling of the astrocyte network in BPD. Furthermore, in controls, DLPFC GFAP mRNA levels were significantly lower with a time of death at daytime (08:01-20:00 h) compared to nighttime (20:01-08:00 h). In depression, such a diurnal pattern was not present. These findings in BPD and MDD subjects warrant further studies given the crucial roles of astrocytes in the central nervous system.

3.
J Exp Biol ; 222(Pt 17)2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31371403

RESUMEN

The timing of breeding is under selection in wild populations as a result of climate change, and understanding the underlying physiological processes mediating this timing provides insight into the potential rate of adaptation. Current knowledge on this variation in physiology is, however, mostly limited to males. We assessed whether individual differences in the timing of breeding in females are reflected in differences in candidate gene expression and, if so, whether these differences occur in the upstream (hypothalamus) or downstream (ovary and liver) parts of the neuroendocrine system. We used 72 female great tits from two generations of lines artificially selected for early and late egg laying, which were housed in climate-controlled aviaries and went through two breeding cycles within 1 year. In the first breeding season we obtained individual egg-laying dates, while in the second breeding season, using the same individuals, we sampled several tissues at three time points based on the timing of the first breeding attempt. For each tissue, mRNA expression levels were measured using qPCR for a set of candidate genes associated with the timing of reproduction and subsequently analysed for differences between generations, time points and individual timing of breeding. We found differences in gene expression between generations in all tissues, with the most pronounced differences in the hypothalamus. Differences between time points, and early- and late-laying females, were found exclusively in the ovary and liver. Altogether, we show that fine-tuning of the seasonal timing of breeding, and thereby the opportunity for adaptation in the neuroendocrine system, is regulated mostly downstream in the neuro-endocrine system.


Asunto(s)
Expresión Génica , Comportamiento de Nidificación , Reproducción , Pájaros Cantores/fisiología , Animales , Variación Biológica Individual , Femenino , Hipotálamo/fisiología , Hígado/fisiología , Ovario/fisiología , Estaciones del Año , Pájaros Cantores/genética
4.
eNeuro ; 6(1)2019.
Artículo en Inglés | MEDLINE | ID: mdl-30671537

RESUMEN

Many brain regions go through critical periods of development during which plasticity is enhanced. These critical periods are associated with extensive growth and retraction of thalamocortical and intracortical axons. Here, we investigated whether a signaling pathway that is central in Wallerian axon degeneration also regulates critical period plasticity in the primary visual cortex (V1). Wallerian degeneration is characterized by rapid disintegration of axons once they are separated from the cell body. This degenerative process is initiated by reduced presence of cytoplasmic nicotinamide mononucleotide adenylyltransferases (NMNATs) and is strongly delayed in mice overexpressing cytoplasmic NMNAT proteins, such as WldS mutant mice producing a UBE4b-NMNAT1 fusion protein or NMNAT3 transgenic mice. Here, we provide evidence that in WldS mice and NMNAT3 transgenic mice, ocular dominance (OD) plasticity in the developing visual cortex is reduced. This deficit is only observed during the second half of the critical period. Additionally, we detect an early increase of visual acuity in the V1 of WldS mice. We do not find evidence for Wallerian degeneration occurring during OD plasticity. Our findings suggest that NMNATs do not only regulate Wallerian degeneration during pathological conditions but also control cellular events that mediate critical period plasticity during the physiological development of the cortex.


Asunto(s)
Plasticidad Neuronal/fisiología , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Corteza Visual/crecimiento & desarrollo , Corteza Visual/metabolismo , Degeneración Walleriana/metabolismo , Animales , Expresión Génica , Ratones Endogámicos C57BL , Ratones Transgénicos , Nicotinamida-Nucleótido Adenililtransferasa/genética , Sinapsis/metabolismo , Técnicas de Cultivo de Tejidos , Agudeza Visual/fisiología
5.
Cereb Cortex ; 28(4): 1183-1194, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28184425

RESUMEN

The formation, plasticity and maintenance of synaptic connections is regulated by molecular and electrical signals. ß-Catenin is an important protein in these events and regulates cadherin-mediated cell adhesion and the recruitment of pre- and postsynaptic proteins in an activity-dependent fashion. Mutations in the ß-catenin gene can cause cognitive disability and autism, with life-long consequences. Understanding its synaptic function may thus be relevant for the treatment of these disorders. So far, ß-catenin's function has been studied predominantly in cell culture and during development but knowledge on its function in adulthood is limited. Here, we show that ablating ß-catenin in excitatory neurons of the adult visual cortex does not cause the same synaptic deficits previously observed during development. Instead, it reduces NMDA-receptor currents and impairs visual processing. We conclude that ß-catenin remains important for adult cortical function but through different mechanisms than during development.


Asunto(s)
Receptores de N-Metil-D-Aspartato/metabolismo , Corteza Visual/metabolismo , beta Catenina/metabolismo , 2-Amino-5-fosfonovalerato/análogos & derivados , 2-Amino-5-fosfonovalerato/farmacología , Animales , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , N-Metilaspartato/metabolismo , Parvalbúminas/metabolismo , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Privación Sensorial , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/genética , Corteza Visual/efectos de los fármacos , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/fisiología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo , beta Catenina/genética
6.
Oncotarget ; 8(50): 88104-88121, 2017 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-29152145

RESUMEN

Astrocytomas are the most common malignant brain tumours and are to date incurable. It is unclear how astrocytomas progress into higher malignant grades. The intermediate filament cytoskeleton is emerging as an important regulator of malignancy in several tumours. The majority of the astrocytomas express the intermediate filament protein Glial Fibrillary Acidic Protein (GFAP). Several GFAP splice variants have been identified and the main variants expressed in human astrocytoma are the GFAPα and GFAPδ isoforms. Here we show a significant downregulation of GFAPα in grade IV astrocytoma compared to grade II and III, resulting in an increased GFAPδ/α ratio. Mimicking this increase in GFAPδ/α ratio in astrocytoma cell lines and comparing the subsequent transcriptomic changes with the changes in the patient tumours, we have identified a set of GFAPδ/α ratio-regulated high-malignant and low-malignant genes. These genes are involved in cell proliferation and protein phosphorylation, and their expression correlated with patient survival. We additionally show that changing the ratio of GFAPδ/α, by targeting GFAP expression, affected expression of high-malignant genes. Our data imply that regulating GFAP expression and splicing are novel therapeutic targets that need to be considered as a treatment for astrocytoma.

7.
Neurobiol Aging ; 55: 115-122, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28434692

RESUMEN

Alzheimer's disease (AD) is strongly associated with microglia-induced neuroinflammation. Particularly, Aß plaque-associated microglia take on an "activated" morphology. However, the function and phenotype of these Aß plaque-associated microglia are not well understood. We show hyperreactivity of Aß plaque-associated microglia upon systemic inflammation in transgenic AD mouse models (i.e., 5XFAD and APP23). Gene expression profiling of Aß plaque-associated microglia (major histocompatibility complex II+ microglia) isolated from 5XFAD mice revealed a proinflammatory phenotype. The upregulated genes involved in the biological processes (gene ontology terms) included: "immune response to external stimulus" such as Axl, Cd63, Egr2, and Lgals3, "cell motility", such as Ccl3, Ccl4, Cxcr4, and Sdc3, "cell differentiation", and "system development", such as St14, Trpm1, and Spp1. In human AD tissue with similar Braak stages, expression of phagocytic markers and AD-associated genes, including HLA-DRA, APOE, AXL, TREM2, and TYROBP, was higher in laser-captured early-onset AD (EOAD) plaques than in late-onset AD plaques. Interestingly, the nonplaque parenchyma of both EOAD and late-onset AD brains, the expression of above-mentioned markers were similarly low. Here, we provide evidence that Aß plaque-associated microglia are hyperreactive in their immune response and phagocytosis in the transgenic AD mice as well as in EOAD brain tissue. We suggest that Aß plaque-associated microglia are the primary source of neuroinflammation related to AD pathology.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Microglía/inmunología , Placa Amiloide/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Animales , Apolipoproteínas E , Encéfalo/inmunología , Diferenciación Celular/genética , Movimiento Celular/genética , Movimiento Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Inflamación/genética , Inflamación/inmunología , Masculino , Glicoproteínas de Membrana , Ratones Transgénicos , Persona de Mediana Edad , Fagocitosis/genética , Fagocitosis/inmunología , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Receptores Inmunológicos , Tirosina Quinasa del Receptor Axl
8.
Glia ; 65(1): 50-61, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27615381

RESUMEN

Huntington's disease (HD) is an autosomal dominant inherited neurodegenerative disorder that is caused by a CAG expansion in the Huntingtin (HTT) gene, leading to HTT inclusion formation in the brain. The mutant huntingtin protein (mHTT) is ubiquitously expressed and therefore nuclear inclusions could be present in all brain cells. The effects of nuclear inclusion formation have been mainly studied in neurons, while the effect on glia has been comparatively disregarded. Astrocytes, microglia, and oligodendrocytes are glial cells that are essential for normal brain function and are implicated in several neurological diseases. Here we examined the number of nuclear mHTT inclusions in both neurons and various types of glia in the two brain areas that are the most affected in HD, frontal cortex, and striatum. We compared nuclear mHTT inclusion body formation in three HD mouse models that express either full-length HTT or an N-terminal exon1 fragment of mHTT, and we observed nuclear inclusions in neurons, astrocytes, oligodendrocytes, and microglia. When studying the frequency of cells with nuclear inclusions in mice, we found that half of the population of neurons contained nuclear inclusions at the disease end stage, whereas the proportion of GFAP-positive astrocytes and oligodendrocytes having a nuclear inclusion was much lower, while microglia hardly showed any nuclear inclusions. Nuclear inclusions were also present in neurons and all studied glial cell types in human patient material. This is the first report to compare nuclear mHTT inclusions in glia and neurons in different HD mouse models and HD patient brains. GLIA 2016;65:50-61.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Enfermedad de Huntington/metabolismo , Masculino , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo
9.
Biochim Biophys Acta ; 1862(10): 1847-60, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27425031

RESUMEN

Amyloid plaques in Alzheimer's disease (AD) mice are surrounded by activated microglia. The functional role of microglia activation in AD is not well understood; both detrimental and beneficial effects on AD progression have been reported. Here we show that the population of activated microglia in the cortex of the APPswe/PS1dE9 mouse AD model is divided into a CD11c-positive and a CD11c-negative subpopulation. Cd11c transcript levels and number of CD11c-positive microglia increase sharply when plaques start to occur and both parameters continue to rise in parallel with the age-related increasing plaque load. CD11c cells are localized near plaques at all stages of the disease development and constitute 23% of all activated microglia. No differences between these two populations were found in terms of proliferation, immunostaining intensity of Iba1, MHC class II, CD45, or immunoproteasome subunit LMP7/ß5i. Comparison of the transcriptome of isolated CD11c-positive and CD11c-negative microglia from the cortex of aged APPswe/PS1dE9 with WT microglia showed that gene expression changes had a similar general pattern. However, a differential expression was found for genes involved in immune signaling (Il6, S100a8/Mrp8, S100a9/Mrp14, Spp1, Igf1), lysosome activation, and carbohydrate- and cholesterol/lipid-metabolism (Apoe). In addition, the increased expression of Gpnmb/DC-HIL, Tm7sf4/DC-STAMP, and Gp49a/Lilrb4, suggests a suppressive/tolerizing influence of CD11c cells. We show that amyloid plaques in the APP/PS1 model are associated with two distinct populations of activated microglia: CD11c-positive and CD11c-negative cells. Our findings imply that CD11c-positive microglia can potentially counteract amyloid deposition via increased Aß-uptake and degradation, and by containing the inflammatory response.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Antígeno CD11c/metabolismo , Regulación de la Expresión Génica , Microglía/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Antígeno CD11c/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , Microglía/patología , Proteínas del Tejido Nervioso/genética
10.
Prog Neurobiol ; 144: 121-41, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26797041

RESUMEN

Alzheimer's disease is the main cause of dementia in the elderly and begins with a subtle decline in episodic memory followed by a more general decline in overall cognitive abilities. Though the exact trigger for this cascade of events remains unknown the presence of the misfolded amyloid-beta protein triggers reactive gliosis, a prominent neuropathological feature in the brains of Alzheimer's patients. The cytoskeletal and morphological changes of astrogliosis are its evident features, while changes in oxidative stress defense, cholesterol metabolism, and gene transcription programs are less manifest. However, these latter molecular changes may underlie a disruption in homeostatic regulation that keeps the brain environment balanced. Astrocytes in Alzheimer's disease show changes in glutamate and GABA signaling and recycling, potassium buffering, and in cholinergic, purinergic, and calcium signaling. Ultimately the dysregulation of homeostasis maintained by astrocytes can have grave consequences for the stability of microcircuits within key brain regions. Specifically, altered inhibition influenced by astrocytes can lead to local circuit imbalance with farther reaching consequences for the functioning of larger neuronal networks. Healthy astrocytes have a role in maintaining and modulating normal neuronal communication, synaptic physiology and energy metabolism, astrogliosis interferes with these functions. This review considers the molecular and functional changes occurring during astrogliosis in Alzheimer's disease, and proposes that astrocytes are key players in the development of dementia.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Astrocitos/metabolismo , Gliosis/metabolismo , Enfermedad de Alzheimer/patología , Animales , Gliosis/patología , Humanos
11.
PLoS One ; 10(9): e0138107, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26418157

RESUMEN

Gaucher disease is characterized by lysosomal accumulation of glucosylceramide due to deficient activity of lysosomal glucocerebrosidase (GBA). In cells, glucosylceramide is also degraded outside lysosomes by the enzyme glucosylceramidase 2 (GBA2) of which inherited deficiency is associated with ataxias. The interest in GBA and glucosylceramide metabolism in the brain has grown following the notion that mutations in the GBA gene impose a risk factor for motor disorders such as α-synucleinopathies. We earlier developed a ß-glucopyranosyl-configured cyclophellitol-epoxide type activity based probe (ABP) allowing in vivo and in vitro visualization of active molecules of GBA with high spatial resolution. Labeling occurs through covalent linkage of the ABP to the catalytic nucleophile residue in the enzyme pocket. Here, we describe a method to visualize active GBA molecules in rat brain slices using in vivo labeling. Brain areas related to motor control, like the basal ganglia and motor related structures in the brainstem, show a high content of active GBA. We also developed a ß-glucopyranosyl cyclophellitol-aziridine ABP allowing in situ labeling of GBA2. Labeled GBA2 in brain areas can be identified and quantified upon gel electrophoresis. The distribution of active GBA2 markedly differs from that of GBA, being highest in the cerebellar cortex. The histological findings with ABP labeling were confirmed by biochemical analysis of isolated brain areas. In conclusion, ABPs offer sensitive tools to visualize active GBA and to study the distribution of GBA2 in the brain and thus may find application to establish the role of these enzymes in neurodegenerative disease conditions such as α-synucleinopathies and cerebellar ataxia.


Asunto(s)
Encéfalo/enzimología , Enfermedad de Gaucher/genética , Glucosilceramidasa/metabolismo , Glucosilceramidas/metabolismo , Enfermedades Neurodegenerativas/genética , Animales , Astrocitos/enzimología , Astrocitos/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/patología , Técnica del Anticuerpo Fluorescente , Colorantes Fluorescentes/química , Enfermedad de Gaucher/patología , Glucosilceramidasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/enzimología , Microglía/metabolismo , Microscopía Confocal , Enfermedades Neurodegenerativas/patología , Células de Purkinje/metabolismo , Ratas , Ratas Wistar
12.
Biomarkers ; 20(3): 196-201, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26220620

RESUMEN

ADAM10 is a potential biomarker for Alzheimer's disease (AD). ADAM10 protein levels are reduced in platelets of AD patients. The aim was to verify the total blood and platelet ADAM10 gene expression in AD patients and to compare with mild cognitive impairment (MCI) and healthy subjects. No significant differences in ADAM10 gene expression were observed. Therefore, the decrease of ADAM10 protein in platelets of AD patients is not caused by a reduction in ADAM10 mRNA. Further studies must be performed to investigate other pathways in the down regulation of ADAM10 protein.


Asunto(s)
Proteínas ADAM/genética , Enfermedad de Alzheimer/sangre , Secretasas de la Proteína Precursora del Amiloide/genética , Disfunción Cognitiva/sangre , Proteínas de la Membrana/genética , ARN Mensajero/genética , Proteínas ADAM/sangre , Proteína ADAM10 , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Secretasas de la Proteína Precursora del Amiloide/sangre , Biomarcadores/sangre , Plaquetas/metabolismo , Plaquetas/patología , Estudios de Casos y Controles , Disfunción Cognitiva/genética , Disfunción Cognitiva/fisiopatología , Femenino , Expresión Génica , Humanos , Masculino , Proteínas de la Membrana/sangre , Persona de Mediana Edad , Pruebas Neuropsicológicas , ARN Mensajero/sangre
13.
Acta Neuropathol Commun ; 3: 31, 2015 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-26001565

RESUMEN

INTRODUCTION: Microglia are tissue macrophages of the central nervous system that monitor brain homeostasis and react upon neuronal damage and stress. Aging and neurodegeneration induce a hypersensitive, pro-inflammatory phenotype, referred to as primed microglia. To determine the gene expression signature of priming, the transcriptomes of microglia in aging, Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS) mouse models were compared using Weighted Gene Co-expression Network Analysis (WGCNA). RESULTS: A highly consistent consensus transcriptional profile of up-regulated genes was identified, which prominently differed from the acute inflammatory gene network induced by lipopolysaccharide (LPS). Where the acute inflammatory network was significantly enriched for NF-κB signaling, the primed microglia profile contained key features related to phagosome, lysosome, antigen presentation, and AD signaling. In addition, specific signatures for aging, AD, and ALS were identified. CONCLUSION: Microglia priming induces a highly conserved transcriptional signature with aging- and disease-specific aspects.


Asunto(s)
Envejecimiento/genética , Inflamación/genética , Microglía/inmunología , Enfermedades Neurodegenerativas/genética , Transducción de Señal/genética , Transcriptoma/genética , Envejecimiento/inmunología , Enfermedad de Alzheimer/genética , Animales , Humanos , Ratones , FN-kappa B/genética , Enfermedades Neurodegenerativas/inmunología , Regulación hacia Arriba
14.
Glia ; 63(6): 1036-56, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25731615

RESUMEN

Reactive astrocytes with an increased expression of intermediate filament (IF) proteins Glial Fibrillary Acidic Protein (GFAP) and Vimentin (VIM) surround amyloid plaques in Alzheimer's disease (AD). The functional consequences of this upregulation are unclear. To identify molecular pathways coupled to IF regulation in reactive astrocytes, and to study the interaction with microglia, we examined WT and APPswe/PS1dE9 (AD) mice lacking either GFAP, or both VIM and GFAP, and determined the transcriptome of cortical astrocytes and microglia from 15- to 18-month-old mice. Genes involved in lysosomal degradation (including several cathepsins) and in inflammatory response (including Cxcl5, Tlr6, Tnf, Il1b) exhibited a higher AD-induced increase when GFAP, or VIM and GFAP, were absent. The expression of Aqp4 and Gja1 displayed the same pattern. The downregulation of neuronal support genes in astrocytes from AD mice was absent in GFAP/VIM null mice. In contrast, the absence of IFs did not affect the transcriptional alterations induced by AD in microglia, nor was the cortical plaque load altered. Visualizing astrocyte morphology in GFAP-eGFP mice showed no clear structural differences in GFAP/VIM null mice, but did show diminished interaction of astrocyte processes with plaques. Microglial proliferation increased similarly in all AD groups. In conclusion, absence of GFAP, or both GFAP and VIM, alters AD-induced changes in gene expression profile of astrocytes, showing a compensation of the decrease of neuronal support genes and a trend for a slightly higher inflammatory expression profile. However, this has no consequences for the development of plaque load, microglial proliferation, or microglial activation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/deficiencia , Microglía/metabolismo , Vimentina/deficiencia , Anciano , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Proliferación Celular/fisiología , Quimiocina CXCL5/metabolismo , Modelos Animales de Enfermedad , Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Presenilina-1/genética , Presenilina-1/metabolismo , Vimentina/genética
15.
Neurobiol Aging ; 35(12): 2746-2760, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25002035

RESUMEN

Reactive astrocytes and microglia are associated with amyloid plaques in Alzheimer's disease (AD). Yet, not much is known about the molecular alterations underlying this reactive phenotype. To get an insight into the molecular changes underlying AD induced astrocyte and microglia reactivity, we performed a transcriptional analysis on acutely isolated astrocytes and microglia from the cortex of aged controls and APPswe/PS1dE9 AD mice. As expected, both cell types acquired a proinflammatory phenotype, which confirms the validity of our approach. Interestingly, we observed that the immune alteration in astrocytes was relatively more pronounced than in microglia. Concurrently, our data reveal that astrocytes display a reduced expression of neuronal support genes and genes involved in neuronal communication. The microglia showed a reduced expression of phagocytosis and/or endocytosis genes. Co-expression analysis of a human AD expression data set and the astrocyte and microglia data sets revealed that the inflammatory changes in astrocytes were remarkably comparable in mouse and human AD, whereas the microglia changes showed less similarity. Based on these findings we argue that chronically proinflammatory astrocyte and microglia phenotypes, showing a reduction of genes involved in neuronal support and neuronal signaling, are likely to contribute to the neuronal dysfunction and cognitive decline in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Astrocitos/patología , Inflamación/genética , Inflamación/patología , Microglía/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/psicología , Animales , Astrocitos/inmunología , Astrocitos/fisiología , Separación Celular , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/patología , Cognición , Modelos Animales de Enfermedad , Endocitosis/genética , Expresión Génica , Humanos , Ratones Transgénicos , Microglía/inmunología , Microglía/fisiología , Fagocitosis/genética , Transmisión Sináptica/genética
16.
Acta Neuropathol Commun ; 2: 76, 2014 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-24974208

RESUMEN

Alzheimer's disease is caused by increased production or reduced clearance of amyloid-ß, which results in the formation amyloid-ß plaques and triggers a cascade of downstream events leading to progressive neurodegeneration. The earliest clinical symptoms of Alzheimer's disease, i.e., memory loss, are however poorly understood from a molecular and cellular perspective. Here we used APPswe/PS1dE9 (APP/PS1) transgenic mice to study the early pre-pathological effects of increased amyloid-ß levels on hippocampal synaptic plasticity and memory. Using an unbiased proteomics approach we show that the early increase in amyloid-ß levels in APP/PS1 mice at three months of age coincides with a robust and significant upregulation of several protein components of the extracellular matrix in hippocampal synaptosome preparations. This increase in extracellular matrix levels occurred well before the onset of plaque formation and was paralleled by impairments in hippocampal long-term potentiation and contextual memory. Direct injection into the hippocampus of the extracellular matrix inactivating enzyme chondroitinase ABC restored both long-term potentiation and contextual memory performance. These findings indicate an important role for the extracellular matrix in causing early memory loss in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Hipocampo/metabolismo , Trastornos de la Memoria/metabolismo , Enfermedad de Alzheimer/patología , Animales , Condroitina ABC Liasa/farmacología , Condicionamiento Psicológico , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide , Proteómica
17.
J Neurochem ; 130(6): 805-15, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24749759

RESUMEN

Enteric glial cells (EGCs) are in many respects similar to astrocytes of the central nervous system and express similar proteins including glial fibrillary acidic protein (GFAP). Changes in GFAP expression and/or phosphorylation have been reported during brain damage or central nervous system degeneration. As in Parkinson's disease (PD) the enteric neurons accumulate α-synuclein, and thus are showing PD-specific pathological features, we undertook the present survey to study whether the enteric glia in PD become reactive by assessing the expression and phosphorylation levels of GFAP in colonic biopsies. Twenty-four PD, six progressive supranuclear palsy (PSP), six multiple system atrophy (MSA) patients, and 21 age-matched healthy controls were included. The expression levels and the phosphorylation state of GFAP were analyzed in colonic biopsies by western blot. Additional experiments were performed using real-time PCR for a more precise analysis of the GFAP isoforms expressed by EGCs. We showed that GFAPκ was the main isoform expressed in EGCs. As compared to control subjects, patients with PD, but not PSP and MSA, had significant higher GFAP expression levels in their colonic biopsies. The phosphorylation level of GFAP at serine 13 was significantly lower in PD patients compared to control subjects. By contrast, no change in GFAP phosphorylation was observed between PSP, MSA and controls. Our findings provide evidence that enteric glial reaction occurs in PD and further reinforce the role of the enteric nervous system in the initiation and/or the progression of the disease. We showed that GFAP is over-expressed and hypophosphorylated in the enteric glial cells (EGCs) of Parkinson's disease (PD) patients as compared to healthy subjects and patients with atypical parkinsonism (MSA, multiple system atrophy and PSP, progressive supranuclear palsy). Our findings provide evidence that enteric glial reaction occurs in PD but not in PSP and MSA and further reinforce the role of the enteric nervous system in the pathophysiology of PD.


Asunto(s)
Proteína Ácida Fibrilar de la Glía/biosíntesis , Enfermedad de Parkinson/metabolismo , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Western Blotting , Química Encefálica/efectos de los fármacos , Línea Celular , Colon/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Neuroglía/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Serina/metabolismo
18.
Neurobiol Aging ; 35(1): 1-14, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23954174

RESUMEN

Astrocytes and microglia become reactive in many neurological disorders resulting in phenotypic and functional alterations. Both cell types might also display functional changes during normal aging. To identify gene signatures and changes in basal cellular functions of astrocytes and microglia in relation to aging, we isolated viable astrocytes and microglia from young adult and aged mouse cortices and determined their gene expression profile. Aged astrocytes, compared with young astrocytes, showed an increased inflammatory phenotype and increased 'zinc ion binding.' Young astrocytes showed higher expression of genes involved in 'neuronal differentiation' and hemoglobin synthesis. Astrocyte expression of genes involved in neuronal signaling remains high throughout age. Aged microglia had higher expression of genes involved in 'vesicle release,' 'zinc ion binding,' and genes within the tumor necrosis factor-ligand family and young microglia had increased transcript levels of C-C motif chemokines. These data provide a transcriptome database of cell-type enriched genes of astrocytes and microglia from adult mice and give insight into the differential gene signature of astrocytes and microglia in relation to normal aging.


Asunto(s)
Envejecimiento/genética , Astrocitos/fisiología , Separación Celular/métodos , Corteza Cerebral/citología , Microglía/fisiología , Transducción de Señal/genética , Transcriptoma/genética , Animales , Astrocitos/citología , Astrocitos/patología , Diferenciación Celular/genética , Células Cultivadas , Quimiocinas CC/genética , Hemoglobinas/biosíntesis , Inflamación/genética , Ratones , Microglía/citología , Microglía/patología , Vesículas Secretoras/genética , Transducción de Señal/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Transcripción Genética/genética , Zinc/metabolismo
19.
Neurobiol Aging ; 35(3): 492-510, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24269023

RESUMEN

In Alzheimer's disease (AD), amyloid plaques are surrounded by reactive astrocytes with an increased expression of intermediate filaments including glial fibrillary acidic protein (GFAP). Different GFAP isoforms have been identified that are differentially expressed by specific subpopulations of astrocytes and that impose different properties to the intermediate filament network. We studied transcript levels and protein expression patterns of all known GFAP isoforms in human hippocampal AD tissue at different stages of the disease. Ten different transcripts for GFAP isoforms were detected at different abundancies. Transcript levels of most isoforms increased with AD progression. GFAPδ-immunopositive astrocytes were observed in subgranular zone, hilus, and stratum-lacunosum-moleculare. GFAPδ-positive cells also stained for GFAPα. In AD donors, astrocytes near plaques displayed increased staining of both GFAPα and GFAPδ. The reading-frame-shifted isoform, GFAP(+1), staining was confined to a subset of astrocytes with long processes, and their number increased in the course of AD. In conclusion, the various GFAP isoforms show differential transcript levels and are upregulated in a concerted manner in AD. The GFAP(+1) isoform defines a unique subset of astrocytes, with numbers increasing with AD progression. These data indicate the need for future exploration of underlying mechanisms concerning the functions of GFAPδ and GFAP(+1) isoforms in astrocytes and their possible role in AD pathology.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Astrocitos/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Placa Amiloide/metabolismo , Células Cultivadas , Progresión de la Enfermedad , Expresión Génica , Hipocampo/metabolismo , Humanos , Isoformas de Proteínas , Índice de Severidad de la Enfermedad , Transcripción Genética , Regulación hacia Arriba
20.
Prog Neurobiol ; 111: 1-16, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23994259

RESUMEN

The research field of adult neurogenesis is rapidly expanding with more and more information becoming available on the identity of the cells located within the subventricular zone (SVZ). Much of our understanding is based on rodent studies. The SVZ is comprised of several different cell types including B1 astrocytes, transit amplifying progenitor cells (C cells), and neuroblasts (A cells). B1 astrocytes are the quiescent neural stem cells that continue to divide throughout a lifespan. They give rise to a progenitor cell, termed a C cell, which in turn, generates neuroblasts destined for the olfactory bulb. There is much controversy over how to distinguish various SVZ cell types. This review summarizes the known markers for rodent SVZ cell types, with particular attention paid towards B1 astrocytes and C cells. Unfortunately, there is no perfect stem cell marker. B1 astrocytes, C cells, and neuroblasts gain and lose marker expression patterns throughout their lineage progression. These expression patterns often overlap at the transition from one cell type to another. The SVZ cell lineage must be seen as a continuum, rather than a static and inert system. This view will aid in understanding the mechanisms underlying marker expression and cellular behavior in the SVZ.


Asunto(s)
Células Madre Adultas/fisiología , Ventrículos Cerebrales/citología , Neurogénesis/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Neuroglía/fisiología , Neuronas/fisiología , Roedores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA