Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Cell Neurosci ; 15: 669280, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149361

RESUMO

The fine processes of single astrocytes can contact many thousands of synapses whose function they can modulate through bi-directional signaling. The spatial arrangement of astrocytic processes and neuronal structures is relevant for such interactions and for the support of neuronal signaling by astrocytes. At the same time, the geometry of perisynaptic astrocyte processes is variable and dynamically regulated. Studying these fine astrocyte processes represents a technical challenge, because many of them cannot be fully resolved by diffraction-limited microscopy. Therefore, we have established two indirect parameters of astrocyte morphology, which, while not fully resolving local geometry by design, provide statistical measures of astrocyte morphology: the fraction of tissue volume that astrocytes occupy and the density of resolvable astrocytic processes. Both are straightforward to obtain using widely available microscopy techniques. We here present the approach and demonstrate its robustness across various experimental conditions using mainly two-photon excitation fluorescence microscopy in acute slices and in vivo as well as modeling. Using these indirect measures allowed us to analyze the morphology of relatively large populations of astrocytes. Doing so we captured the heterogeneity of astrocytes within and between the layers of the hippocampal CA1 region and the developmental profile of astrocyte morphology. This demonstrates that volume fraction (VF) and segment density are useful parameters for describing the structure of astrocytes. They are also suitable for online monitoring of astrocyte morphology with widely available microscopy techniques.

2.
Nature ; 586(7830): E28, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33005053

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Front Physiol ; 11: 871, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32903427

RESUMO

Astrocytes are the main cell type responsible for the regulation of brain homeostasis, including the maintenance of ion gradients and neurotransmitter clearance. These processes are tightly coupled to changes in the intracellular sodium (Na+) concentration. While activation of the sodium-potassium-ATPase (NKA) in response to an elevation of extracellular K+ may decrease intracellular Na+, the cotransport of transmitters, such as glutamate, together with Na+ results in an increase in astrocytic Na+. This increase in intracellular Na+ can modulate, for instance, metabolic downstream pathways. Thereby, astrocytes are capable to react on a fast time scale to surrounding neuronal activity via intracellular Na+ fluctuations and adjust energy production to the demand of their environment. Beside the well-documented conventional roles of Na+ signaling mainly mediated through changes in its electrochemical gradient, several recent studies have identified more atypical roles for Na+, including protein interactions leading to changes in their biochemical activity or Na+-dependent regulation of gene expression. In this review, we will address both the conventional as well as the atypical functions of astrocytic Na+ signaling, presenting the role of transporters and channels involved and their implications for physiological processes in the central nervous system (CNS). We will also discuss how these important functions are affected under pathological conditions, including stroke and migraine. We postulate that Na+ is an essential player not only in the maintenance of homeostatic processes but also as a messenger for the fast communication between neurons and astrocytes, adjusting the functional properties of various cellular interaction partners to the needs of the surrounding network.

4.
Cell Rep ; 30(10): 3466-3477.e4, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32160550

RESUMO

Astroglia regulate neurovascular coupling while engaging in signal exchange with neurons. The underlying cellular machinery is thought to rely on astrocytic Ca2+ signals, but what controls their amplitude and waveform is poorly understood. Here, we employ time-resolved two-photon excitation fluorescence imaging in acute hippocampal slices and in cortex in vivo to find that resting [Ca2+] predicts the scale (amplitude) and the maximum (peak) of astroglial Ca2+ elevations. We bidirectionally manipulate resting [Ca2+] by uncaging intracellular Ca2+ or Ca2+ buffers and use ratiometric imaging of a genetically encoded Ca2+ indicator to establish that alterations in resting [Ca2+] change co-directionally the peak level and anti-directionally the amplitude of local Ca2+ transients. This relationship holds for spontaneous and for induced (for instance by locomotion) Ca2+ signals. Our findings uncover a basic generic rule of Ca2+ signal formation in astrocytes, thus also associating the resting Ca2+ level with the physiological "excitability" state of astroglia.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio , Cálcio/metabolismo , Animais , Fluorescência , Locomoção , Camundongos , Frações Subcelulares
5.
EMBO Mol Med ; 11(2)2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30617153

RESUMO

Reactive astrogliosis is a hallmark of Alzheimer's disease (AD), but its role for disease initiation and progression has remained incompletely understood. We here show that the transcription factor Stat3 (signal transducer and activator of transcription 3), a canonical inducer of astrogliosis, is activated in an AD mouse model and human AD Therefore, using a conditional knockout approach, we deleted Stat3 specifically in astrocytes in the APP/PS1 model of AD We found that Stat3-deficient APP/PS1 mice show decreased ß-amyloid levels and plaque burden. Plaque-close microglia displayed a more complex morphology, internalized more ß-amyloid, and upregulated amyloid clearance pathways in Stat3-deficient mice. Moreover, astrocyte-specific Stat3-deficient APP/PS1 mice showed decreased pro-inflammatory cytokine activation and lower dystrophic neurite burden, and were largely protected from cerebral network imbalance. Finally, Stat3 deletion in astrocytes also strongly ameliorated spatial learning and memory decline in APP/PS1 mice. Importantly, these protective effects on network dysfunction and cognition were recapitulated in APP/PS1 mice systemically treated with a preclinical Stat3 inhibitor drug. In summary, our data implicate Stat3-mediated astrogliosis as an important therapeutic target in AD.


Assuntos
Doença de Alzheimer/patologia , Astrócitos/patologia , Proliferação de Células , Fator de Transcrição STAT3/análise , Animais , Modelos Animais de Doenças , Técnicas de Inativação de Genes , Humanos , Camundongos , Camundongos Knockout , Fator de Transcrição STAT3/deficiência
6.
J Exp Med ; 215(6): 1649-1663, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29724785

RESUMO

Astrocytic hyperactivity is an important contributor to neuronal-glial network dysfunction in Alzheimer's disease (AD). We have previously shown that astrocyte hyperactivity is mediated by signaling through the P2Y1 purinoreceptor (P2Y1R) pathway. Using the APPPS1 mouse model of AD, we here find that chronic intracerebroventricular infusion of P2Y1R inhibitors normalizes astroglial and neuronal network dysfunction, as measured by in vivo two-photon microscopy, augments structural synaptic integrity, and preserves hippocampal long-term potentiation. These effects occur independently from ß-amyloid metabolism or plaque burden but are associated with a higher morphological complexity of periplaque reactive astrocytes, as well as reduced dystrophic neurite burden and greater plaque compaction. Importantly, APPPS1 mice chronically treated with P2Y1R antagonists, as well as APPPS1 mice carrying an astrocyte-specific genetic deletion (Ip3r2-/-) of signaling pathways downstream of P2Y1R activation, are protected from the decline of spatial learning and memory. In summary, our study establishes the restoration of network homoeostasis by P2Y1R inhibition as a novel treatment target in AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Cognição , Rede Nervosa/fisiopatologia , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Receptores Purinérgicos P2Y1/metabolismo , Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/farmacologia , Difosfato de Adenosina/uso terapêutico , Doença de Alzheimer/patologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Hipocampo/patologia , Humanos , Memória/efeitos dos fármacos , Camundongos Transgênicos , Rede Nervosa/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Placa Amiloide/metabolismo , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
7.
Nature ; 554(7690): 102-105, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29364869

RESUMO

Here we describe the honeycomb maze, a behavioural paradigm for the study of spatial navigation in rats. The maze consists of 37 platforms that can be raised or lowered independently. Place navigation requires an animal to go to a goal platform from any of several start platforms via a series of sequential choices. For each, the animal is confined to a raised platform and allowed to choose between two of the six adjacent platforms, the correct one being the platform with the smallest angle to the goal-heading direction. Rats learn rapidly and their choices are influenced by three factors: the angle between the two choice platforms, the distance from the goal, and the angle between the correct platform and the direction of the goal. Rats with hippocampal damage are impaired in learning and their performance is affected by all three factors. The honeycomb maze represents a marked improvement over current spatial navigation tests, such as the Morris water maze, because it controls the choices of the animal at each point in the maze, provides the ability to assess knowledge of the goal direction from any location, enables the identification of factors influencing task performance and provides the possibility for concomitant single-cell recording.


Assuntos
Objetivos , Hipocampo/fisiologia , Aprendizagem em Labirinto/fisiologia , Navegação Espacial/fisiologia , Animais , Eletrofisiologia/instrumentação , Córtex Entorrinal/patologia , Córtex Entorrinal/fisiopatologia , Córtex Entorrinal/cirurgia , Hipocampo/patologia , Hipocampo/fisiopatologia , Hipocampo/cirurgia , Masculino , Ratos , Análise de Célula Única/instrumentação
8.
Elife ; 52016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27897969

RESUMO

SATB2 is a risk locus for schizophrenia and encodes a DNA-binding protein that regulates higher-order chromatin configuration. In the adult brain Satb2 is almost exclusively expressed in pyramidal neurons of two brain regions important for memory formation, the cerebral cortex and the CA1-hippocampal field. Here we show that Satb2 is required for key hippocampal functions since deletion of Satb2 from the adult mouse forebrain prevents the stabilization of synaptic long-term potentiation and markedly impairs long-term fear and object discrimination memory. At the molecular level, we find that synaptic activity and BDNF up-regulate Satb2, which itself binds to the promoters of coding and non-coding genes. Satb2 controls the hippocampal levels of a large cohort of miRNAs, many of which are implicated in synaptic plasticity and memory formation. Together, our findings demonstrate that Satb2 is critically involved in long-term plasticity processes in the adult forebrain that underlie the consolidation and stabilization of context-linked memory.


Assuntos
Regulação da Expressão Gênica , Hipocampo/fisiologia , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Memória de Longo Prazo , MicroRNAs/biossíntese , Fatores de Transcrição/metabolismo , Animais , Técnicas de Inativação de Genes , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos , Camundongos Knockout , Fatores de Transcrição/genética
9.
Acta Neuropathol Commun ; 4(1): 76, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27487766

RESUMO

Vascular cognitive impairment is the second most common form of dementia. The pathogenic pathways leading to vascular cognitive impairment remain unclear but clinical and experimental data have shown that chronic reactive astrogliosis occurs within white matter lesions, indicating that a sustained pro-inflammatory environment affecting the white matter may contribute towards disease progression. To model vascular cognitive impairment, we induced prolonged mild cerebral hypoperfusion in mice by bilateral common carotid artery stenosis. This chronic hypoperfusion resulted in reactive gliosis of astrocytes and microglia within white matter tracts, demyelination and axonal degeneration, consecutive spatial memory deficits, and loss of white matter integrity, as measured by ultra high-field magnetic resonance diffusion tensor imaging. White matter astrogliosis was accompanied by activation of the pro-inflammatory transcription factor nuclear factor (NF)-kB in reactive astrocytes. Using mice expressing a dominant negative inhibitor of NF-kB under the control of the astrocyte-specific glial fibrillary acid protein (GFAP) promoter (GFAP-IkBα-dn), we found that transgenic inhibition of astroglial NF-kB signaling ameliorated gliosis and axonal loss, maintained white matter structural integrity, and preserved memory function. Collectively, our results imply that pro-inflammatory changes in white matter astrocytes may represent an important detrimental component in the pathogenesis of vascular cognitive impairment, and that targeting these pathways may lead to novel therapeutic strategies.


Assuntos
Astrócitos/metabolismo , Disfunção Cognitiva/imunologia , Demência Vascular/imunologia , NF-kappa B/metabolismo , Substância Branca/imunologia , Animais , Astrócitos/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/imunologia , Encéfalo/patologia , Estenose das Carótidas , Disfunção Cognitiva/diagnóstico por imagem , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Citocinas/metabolismo , Demência Vascular/diagnóstico por imagem , Demência Vascular/patologia , Demência Vascular/psicologia , Doenças Desmielinizantes/diagnóstico por imagem , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/psicologia , Modelos Animais de Doenças , Gliose/diagnóstico por imagem , Gliose/imunologia , Gliose/patologia , Gliose/psicologia , Masculino , Camundongos Transgênicos , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/antagonistas & inibidores , Substância Branca/diagnóstico por imagem , Substância Branca/patologia
10.
Nat Commun ; 5: 5422, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25406732

RESUMO

Astrocytic network alterations have been reported in Alzheimer's disease (AD), but the underlying pathways have remained undefined. Here we measure astrocytic calcium, cerebral blood flow and amyloid-ß plaques in vivo in a mouse model of AD using multiphoton microscopy. We find that astrocytic hyperactivity, consisting of single-cell transients and calcium waves, is most pronounced in reactive astrogliosis around plaques and is sometimes associated with local blood flow changes. We show that astroglial hyperactivity is reduced after P2 purinoreceptor blockade or nucleotide release through connexin hemichannels, but is augmented by increasing cortical ADP concentration. P2X receptor blockade has no effect, but inhibition of P2Y1 receptors, which are strongly expressed by reactive astrocytes surrounding plaques, completely normalizes astrocytic hyperactivity. Our data suggest that astroglial network dysfunction is mediated by purinergic signalling in reactive astrocytes, and that intervention aimed at P2Y1 receptors or hemichannel-mediated nucleotide release may help ameliorate network dysfunction in AD.


Assuntos
Doença de Alzheimer/metabolismo , Astrócitos/metabolismo , Placa Amiloide/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Trifosfato de Adenosina/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Sinalização do Cálcio , Circulação Cerebrovascular , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica , Transdução de Sinais
11.
PLoS Biol ; 12(1): e1001763, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24453941

RESUMO

Nogo-A is a membrane protein of the central nervous system (CNS) restricting neurite growth and synaptic plasticity via two extracellular domains: Nogo-66 and Nogo-A-Δ20. Receptors transducing Nogo-A-Δ20 signaling remained elusive so far. Here we identify the G protein-coupled receptor (GPCR) sphingosine 1-phosphate receptor 2 (S1PR2) as a Nogo-A-Δ20-specific receptor. Nogo-A-Δ20 binds S1PR2 on sites distinct from the pocket of the sphingolipid sphingosine 1-phosphate (S1P) and signals via the G protein G13, the Rho GEF LARG, and RhoA. Deleting or blocking S1PR2 counteracts Nogo-A-Δ20- and myelin-mediated inhibition of neurite outgrowth and cell spreading. Blockade of S1PR2 strongly enhances long-term potentiation (LTP) in the hippocampus of wild-type but not Nogo-A(-/-) mice, indicating a repressor function of the Nogo-A/S1PR2 axis in synaptic plasticity. A similar increase in LTP was also observed in the motor cortex after S1PR2 blockade. We propose a novel signaling model in which a GPCR functions as a receptor for two structurally unrelated ligands, a membrane protein and a sphingolipid. Elucidating Nogo-A/S1PR2 signaling platforms will provide new insights into regulation of synaptic plasticity.


Assuntos
Hipocampo/metabolismo , Córtex Motor/metabolismo , Proteínas da Mielina/genética , Plasticidade Neuronal/genética , Receptores de Lisoesfingolipídeo/genética , Animais , Proliferação de Células , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Hipocampo/citologia , Potenciação de Longa Duração , Lisofosfolipídeos/metabolismo , Camundongos , Camundongos Knockout , Córtex Motor/citologia , Proteínas da Mielina/deficiência , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Neuritos/metabolismo , Proteínas Nogo , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Sinapses/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
12.
Nature ; 493(7434): 674-8, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23254930

RESUMO

Alzheimer's disease is the world's most common dementing illness. Deposition of amyloid-ß peptide drives cerebral neuroinflammation by activating microglia. Indeed, amyloid-ß activation of the NLRP3 inflammasome in microglia is fundamental for interleukin-1ß maturation and subsequent inflammatory events. However, it remains unknown whether NLRP3 activation contributes to Alzheimer's disease in vivo. Here we demonstrate strongly enhanced active caspase-1 expression in human mild cognitive impairment and brains with Alzheimer's disease, suggesting a role for the inflammasome in this neurodegenerative disease. Nlrp3(-/-) or Casp1(-/-) mice carrying mutations associated with familial Alzheimer's disease were largely protected from loss of spatial memory and other sequelae associated with Alzheimer's disease, and demonstrated reduced brain caspase-1 and interleukin-1ß activation as well as enhanced amyloid-ß clearance. Furthermore, NLRP3 inflammasome deficiency skewed microglial cells to an M2 phenotype and resulted in the decreased deposition of amyloid-ß in the APP/PS1 model of Alzheimer's disease. These results show an important role for the NLRP3/caspase-1 axis in the pathogenesis of Alzheimer's disease, and suggest that NLRP3 inflammasome inhibition represents a new therapeutic intervention for the disease.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Proteínas de Transporte/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Comportamento Animal , Encéfalo/enzimologia , Proteínas de Transporte/genética , Caspase 1/genética , Caspase 1/metabolismo , Disfunção Cognitiva/enzimologia , Disfunção Cognitiva/fisiopatologia , Regulação Enzimológica da Expressão Gênica , Humanos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Memória , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Óxido Nítrico Sintase Tipo II/metabolismo , Fagocitose/genética
13.
EMBO J ; 30(11): 2266-80, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21522131

RESUMO

Despite its key role in Alzheimer pathogenesis, the physiological function(s) of the amyloid precursor protein (APP) and its proteolytic fragments are still poorly understood. Previously, we generated APPsα knock-in (KI) mice expressing solely the secreted ectodomain APPsα. Here, we generated double mutants (APPsα-DM) by crossing APPsα-KI mice onto an APLP2-deficient background and show that APPsα rescues the postnatal lethality of the majority of APP/APLP2 double knockout mice. Surviving APPsα-DM mice exhibited impaired neuromuscular transmission, with reductions in quantal content, readily releasable pool, and ability to sustain vesicle release that resulted in muscular weakness. We show that these defects may be due to loss of an APP/Mint2/Munc18 complex. Moreover, APPsα-DM muscle showed fragmented post-synaptic specializations, suggesting impaired postnatal synaptic maturation and/or maintenance. Despite normal CNS morphology and unaltered basal synaptic transmission, young APPsα-DM mice already showed pronounced hippocampal dysfunction, impaired spatial learning and a deficit in LTP that could be rescued by GABA(A) receptor inhibition. Collectively, our data show that APLP2 and APP are synergistically required to mediate neuromuscular transmission, spatial learning and synaptic plasticity.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/deficiência , Animais , Cruzamentos Genéticos , Aprendizagem , Camundongos , Camundongos Knockout , Junção Neuromuscular/fisiologia , Plasticidade Neuronal , Transmissão Sináptica
14.
Proc Natl Acad Sci U S A ; 108(6): 2569-74, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21262805

RESUMO

Whereas the role of NogoA in limiting axonal fiber growth and regeneration following an injury of the mammalian central nervous system (CNS) is well known, its physiological functions in the mature uninjured CNS are less well characterized. NogoA is mainly expressed by oligodendrocytes, but also by subpopulations of neurons, in particular in plastic regions of the CNS, e.g., in the hippocampus where it is found at synaptic sites. We analyzed synaptic transmission as well as long-term synaptic plasticity (long-term potentiation, LTP) in the presence of function blocking anti-NogoA or anti-Nogo receptor (NgR) antibodies and in NogoA KO mice. Whereas baseline synaptic transmission, short-term plasticity and long-term depression were not affected by either approach, long-term potentiation was significantly increased following NogoA or NgR1 neutralization. Synaptic potentiation thus seems to be restricted by NogoA. Surprisingly, synaptic weakening was not affected by interfering with NogoA signaling. Mechanistically of interest is the observation that by blockade of the GABA(A) receptors normal synaptic strengthening reoccurred in the absence of NogoA signaling. The present results show a unique role of NogoA expressed in the adult hippocampus in restricting physiological synaptic plasticity on a very fast time scale. NogoA could thus serve as an important negative regulator of functional and structural plasticity in mature neuronal networks.


Assuntos
Hipocampo/metabolismo , Potenciação de Longa Duração/fisiologia , Proteínas da Mielina/metabolismo , Plasticidade Neuronal/fisiologia , Oligodendroglia/metabolismo , Sinapses/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Hipocampo/citologia , Potenciação de Longa Duração/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteínas da Mielina/antagonistas & inibidores , Proteínas da Mielina/genética , Plasticidade Neuronal/efeitos dos fármacos , Proteínas Nogo , Oligodendroglia/citologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sinapses/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA