Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 40(24): e108662, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34825707

RESUMO

Chronic neuroinflammation is a pathogenic component of Alzheimer's disease (AD) that may limit the ability of the brain to clear amyloid deposits and cellular debris. Tight control of the immune system is therefore key to sustain the ability of the brain to repair itself during homeostasis and disease. The immune-cell checkpoint receptor/ligand pair PD-1/PD-L1, known for their inhibitory immune function, is expressed also in the brain. Here, we report upregulated expression of PD-L1 and PD-1 in astrocytes and microglia, respectively, surrounding amyloid plaques in AD patients and in the APP/PS1 AD mouse model. We observed juxtamembrane shedding of PD-L1 from astrocytes, which may mediate ectodomain signaling to PD-1-expressing microglia. Deletion of microglial PD-1 evoked an inflammatory response and compromised amyloid-ß peptide (Aß) uptake. APP/PS1 mice deficient for PD-1 exhibited increased deposition of Aß, reduced microglial Aß uptake, and decreased expression of the Aß receptor CD36 on microglia. Therefore, ineffective immune regulation by the PD-1/PD-L1 axis contributes to Aß plaque deposition during chronic neuroinflammation in AD.


Assuntos
Doença de Alzheimer/imunologia , Precursor de Proteína beta-Amiloide/genética , Antígeno B7-H1/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Regulação para Cima , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/toxicidade , Animais , Astrócitos/metabolismo , Antígenos CD36/metabolismo , Estudos de Casos e Controles , Modelos Animais de Doenças , Feminino , Deleção de Genes , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Pessoa de Meia-Idade
2.
Acta Neuropathol ; 147(1): 65, 2024 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-38557897

RESUMO

Human microglia are critically involved in Alzheimer's disease (AD) progression, as shown by genetic and molecular studies. However, their role in tau pathology progression in human brain has not been well described. Here, we characterized 32 human donors along progression of AD pathology, both in time-from early to late pathology-and in space-from entorhinal cortex (EC), inferior temporal gyrus (ITG), prefrontal cortex (PFC) to visual cortex (V2 and V1)-with biochemistry, immunohistochemistry, and single nuclei-RNA-sequencing, profiling a total of 337,512 brain myeloid cells, including microglia. While the majority of microglia are similar across brain regions, we identified a specific subset unique to EC which may contribute to the early tau pathology present in this region. We calculated conversion of microglia subtypes to diseased states and compared conversion patterns to those from AD animal models. Targeting genes implicated in this conversion, or their upstream/downstream pathways, could halt gene programs initiated by early tau progression. We used expression patterns of early tau progression to identify genes whose expression is reversed along spreading of spatial tau pathology (EC > ITG > PFC > V2 > V1) and identified their potential involvement in microglia subtype conversion to a diseased state. This study provides a data resource that builds on our knowledge of myeloid cell contribution to AD by defining the heterogeneity of microglia and brain macrophages during both temporal and regional pathology aspects of AD progression at an unprecedented resolution.


Assuntos
Doença de Alzheimer , Animais , Humanos , Doença de Alzheimer/patologia , Proteínas tau/genética , Proteínas tau/metabolismo , Transcriptoma , Encéfalo/patologia , Células Mieloides/patologia , Microglia/patologia , Peptídeos beta-Amiloides/metabolismo
3.
EMBO Rep ; 22(11): e51696, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34569685

RESUMO

Neuroinflammation is a common feature of many neurodegenerative diseases. It fosters a dysfunctional neuron-microglia-astrocyte crosstalk that, in turn, maintains microglial cells in a perniciously reactive state that often enhances neuronal damage. The molecular components that mediate this critical communication are not fully explored. Here, we show that secreted frizzled-related protein 1 (SFRP1), a multifunctional regulator of cell-to-cell communication, is part of the cellular crosstalk underlying neuroinflammation. In mouse models of acute and chronic neuroinflammation, SFRP1, largely astrocyte-derived, promotes and sustains microglial activation, and thus a chronic inflammatory state. SFRP1 promotes the upregulation of components of the hypoxia-induced factor-dependent inflammatory pathway and, to a lower extent, of those downstream of the nuclear factor-kappa B. We thus propose that SFRP1 acts as an astrocyte-to-microglia amplifier of neuroinflammation, representing a potential valuable therapeutic target for counteracting the harmful effect of chronic inflammation in several neurodegenerative diseases.


Assuntos
Astrócitos , Microglia , Animais , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Microglia/metabolismo , Doenças Neuroinflamatórias
4.
Nature ; 552(7685): 355-361, 2017 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-29293211

RESUMO

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


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Microglia/metabolismo , Agregação Patológica de Proteínas , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Anticorpos/administração & dosagem , Anticorpos/imunologia , Anticorpos/farmacologia , Proteínas Adaptadoras de Sinalização CARD/antagonistas & inibidores , Proteínas Adaptadoras de Sinalização CARD/química , Proteínas Adaptadoras de Sinalização CARD/imunologia , Feminino , Hipocampo/citologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Presenilina-1/deficiência , Presenilina-1/genética , Domínios Proteicos , Memória Espacial/fisiologia
5.
Acta Neuropathol ; 141(5): 681-696, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33609158

RESUMO

Alzheimer's disease (AD) is the most prevalent form of dementia and is characterized by abnormal extracellular aggregates of amyloid-ß and intraneuronal hyperphosphorylated tau tangles and neuropil threads. Microglia, the tissue-resident macrophages of the central nervous system (CNS), are important for CNS homeostasis and implicated in AD pathology. In amyloid mouse models, a phagocytic/activated microglia phenotype has been identified. How increasing levels of amyloid-ß and tau pathology affect human microglia transcriptional profiles is unknown. Here, we performed snRNAseq on 482,472 nuclei from non-demented control brains and AD brains containing only amyloid-ß plaques or both amyloid-ß plaques and tau pathology. Within the microglia population, distinct expression profiles were identified of which two were AD pathology-associated. The phagocytic/activated AD1-microglia population abundance strongly correlated with tissue amyloid-ß load and localized to amyloid-ß plaques. The AD2-microglia abundance strongly correlated with tissue phospho-tau load and these microglia were more abundant in samples with overt tau pathology. This full characterization of human disease-associated microglia phenotypes provides new insights in the pathophysiological role of microglia in AD and offers new targets for microglia-state-specific therapeutic strategies.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Microglia/patologia , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Feminino , Humanos , Masculino
6.
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
7.
Hum Mol Genet ; 23(24): 6644-58, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25027320

RESUMO

Cerebrospinal fluid amyloid-beta 1-42 (Aß1-42) and phosphorylated Tau at position 181 (pTau181) are biomarkers of Alzheimer's disease (AD). We performed an analysis and meta-analysis of genome-wide association study data on Aß1-42 and pTau181 in AD dementia patients followed by independent replication. An association was found between Aß1-42 level and a single-nucleotide polymorphism in SUCLG2 (rs62256378) (P = 2.5×10(-12)). An interaction between APOE genotype and rs62256378 was detected (P = 9.5 × 10(-5)), with the strongest effect being observed in APOE-ε4 noncarriers. Clinically, rs62256378 was associated with rate of cognitive decline in AD dementia patients (P = 3.1 × 10(-3)). Functional microglia experiments showed that SUCLG2 was involved in clearance of Aß1-42.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Apolipoproteína E4/genética , Proteínas Nucleares/genética , Fragmentos de Peptídeos/genética , Polimorfismo de Nucleotídeo Único , Proteínas de Ligação a RNA/genética , Proteínas tau/genética , Idoso , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Apolipoproteína E4/líquido cefalorraquidiano , Cognição , Feminino , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Masculino , Proteínas Nucleares/líquido cefalorraquidiano , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fosforilação , Proteínas de Ligação a RNA/líquido cefalorraquidiano , Fatores de Processamento de Serina-Arginina , Transdução de Sinais , Proteínas tau/líquido cefalorraquidiano
8.
J Neurosci ; 34(26): 8845-54, 2014 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-24966384

RESUMO

To assess the consequences of locus ceruleus (LC) degeneration and subsequent noradrenaline (NA) deficiency in early Alzheimer's disease (AD), mice overexpressing mutant amyloid precursor protein and presenilin-1 (APP/PS1) were crossed with Ear2(-/-) mice that have a severe loss of LC neurons projecting to the hippocampus and neocortex. Testing spatial memory and hippocampal long-term potentiation revealed an impairment in APP/PS1 Ear2(-/-) mice, whereas APP/PS1 or Ear2(-/-) mice showed only minor changes. These deficits were associated with distinct synaptic changes including reduced expression of the NMDA 2A subunit and increased levels of NMDA receptor 2B in APP/PS1 Ear2(-/-) mice. Acute pharmacological replacement of NA by L-threo-DOPS partially restored phosphorylation of ß-CaMKII and spatial memory performance in APP/PS1 Ear2(-/-) mice. These changes were not accompanied by altered APP processing or amyloid ß peptide (Aß) deposition. Thus, early LC degeneration and subsequent NA reduction may contribute to cognitive deficits via CaMKII and NMDA receptor dysfunction independent of Aß and suggests that NA supplementation could be beneficial in treating AD.


Assuntos
Neurotoxina Derivada de Eosinófilo/metabolismo , Aprendizagem/fisiologia , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/metabolismo , Memória/fisiologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Neurotoxina Derivada de Eosinófilo/genética , Locus Cerúleo/metabolismo , Locus Cerúleo/patologia , Transtornos da Memória/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Norepinefrina/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
9.
Gastroenterology ; 146(1): 176-87.e1, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24067878

RESUMO

BACKGROUND & AIMS: Postoperative ileus (POI) is a common consequence of abdominal surgery that increases the risk of postoperative complications and morbidity. We investigated the cellular mechanisms and immune responses involved in the pathogenesis of POI. METHODS: We studied a mouse model of POI in which intestinal manipulation leads to inflammation of the muscularis externa and disrupts motility. We used C57BL/6 (control) mice as well as mice deficient in Toll-like receptors (TLRs) and cytokine signaling components (TLR-2(-/-), TLR-4(-/-), TLR-2/4(-/-), MyD88(-/-), MyD88/TLR adaptor molecule 1(-/-), interleukin-1 receptor [IL-1R1](-/-), and interleukin (IL)-18(-/-) mice). Bone marrow transplantation experiments were performed to determine which cytokine receptors and cell types are involved in the pathogenesis of POI. RESULTS: Development of POI did not require TLRs 2, 4, or 9 or MyD88/TLR adaptor molecule 2 but did require MyD88, indicating a role for IL-1R1. IL-1R1(-/-) mice did not develop POI; however, mice deficient in IL-18, which also signals via MyD88, developed POI. Mice given injections of an IL-1 receptor antagonist (anakinra) or antibodies to deplete IL-1α and IL-1ß before intestinal manipulation were protected from POI. Induction of POI activated the inflammasome in muscularis externa tissues of C57BL6 mice, and IL-1α and IL-1ß were released in ex vivo organ bath cultures. In bone marrow transplantation experiments, the development of POI required activation of IL-1 receptor in nonhematopoietic cells. IL-1R1 was expressed by enteric glial cells in the myenteric plexus layer, and cultured primary enteric glia cells expressed IL-6 and the chemokine monocyte chemotactic protein 1 in response to IL-1ß stimulation. Immunohistochemical analysis of human small bowel tissue samples confirmed expression of IL-1R1 in the ganglia of the myenteric plexus. CONCLUSIONS: IL-1 signaling, via IL-1R1 and MyD88, is required for development of POI after intestinal manipulation in mice. Agents that interfere with the IL-1 signaling pathway are likely to be effective in the treatment of POI.


Assuntos
Motilidade Gastrointestinal/imunologia , Íleus/imunologia , Interleucina-1/imunologia , Músculo Liso/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Plexo Mientérico/imunologia , Neuroglia/imunologia , Complicações Pós-Operatórias/imunologia , Receptores Tipo I de Interleucina-1/imunologia , Animais , Modelos Animais de Doenças , Íleus/metabolismo , Interleucina-1/metabolismo , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-18/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Plexo Mientérico/metabolismo , Neuroglia/metabolismo , Complicações Pós-Operatórias/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
10.
J Neurosci ; 32(48): 17321-31, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23197723

RESUMO

Alzheimer's disease (AD) is characterized by the extracellular deposition of amyloid-ß (Aß), neurofibrillary tangle formation, and a microglial-driven inflammatory response. Chronic inflammatory activation compromises microglial clearance functions. Because peroxisome proliferator-activated receptor γ (PPARγ) agonists suppress inflammatory gene expression, we tested whether activation of PPARγ would also result in improved microglial Aß phagocytosis. The PPARγ agonist pioglitazone and a novel selective PPARα/γ modulator, DSP-8658, currently in clinical development for the treatment of type 2 diabetes, enhanced the microglial uptake of Aß in a PPARγ-dependent manner. This PPARγ-stimulated increase of Aß phagocytosis was mediated by the upregulation of scavenger receptor CD36 expression. In addition, combined treatment with agonists for the heterodimeric binding partners of PPARγ, the retinoid X receptors (RXRs), showed additive enhancement of the Aß uptake that was mediated by RXRα activation. Evaluation of DSP-8658 in the amyloid precursor protein/presenilin 1 mouse model confirmed an increased microglial Aß phagocytosis in vivo, which subsequently resulted in a reduction of cortical and hippocampal Aß levels. Furthermore, DSP-8658-treated mice showed improved spatial memory performance. Therefore, stimulation of microglial clearance by simultaneous activation of the PPARγ/RXRα heterodimer may prove beneficial in prevention of AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Microglia/efeitos dos fármacos , PPAR gama/agonistas , Fagocitose/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Cognição/efeitos dos fármacos , Cognição/fisiologia , Modelos Animais de Doenças , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Aprendizagem em Labirinto/fisiologia , Camundongos , Microglia/metabolismo , Fagocitose/fisiologia , Pioglitazona , Presenilina-1/genética , Presenilina-1/metabolismo , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico
11.
J Neurosci ; 32(49): 17824-9, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23223301

RESUMO

Neuroinflammation plays a fundamental role in the pathogenesis of Alzheimer's disease (AD), resulting in the extensive activation of microglial and astroglial cells. Here we describe the role of myeloid-related protein Mrp14, a recently described amplifier of inflammation, in Alzheimer's disease and in the related amyloid precursor protein/presenilin1 (APP/PS1) mouse model. Detection of Mrp14 in control, mildly cognitive impaired, and AD patients revealed a strong induction of Mrp14 in protein extracts as well as in the cerebrospinal fluid, but not in blood plasma. In APP/PS1 mice, Mrp14 and its heterodimeric partner Mrp8 was found to be upregulated in microglial cells surrounding amyloid plaques. Functionally, loss of Mrp14 led to increased phagocytosis of fibrillar amyloid ß (Aß) in microglia cells in vitro and in vivo. Generating APP/PS1-transgenic mice deficient for Mrp14, we observed a decrease of key cytokines involved in APP processing, a reduction of BACE1 expression and activity, and consequently overall Aß deposition. We therefore conclude that Mrp14 promotes APP processing and Aß accumulation under neuroinflammatory conditions.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Calgranulina B/metabolismo , Calgranulina B/fisiologia , Microglia/fisiologia , Fagocitose/fisiologia , Idoso , Doença de Alzheimer/sangue , Doença de Alzheimer/líquido cefalorraquidiano , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/metabolismo , Calgranulina A/metabolismo , Estudos de Casos e Controles , Disfunção Cognitiva/sangue , Disfunção Cognitiva/líquido cefalorraquidiano , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Fagocitose/genética , Presenilina-1/genética
12.
Proc Natl Acad Sci U S A ; 107(13): 6058-63, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-20231476

RESUMO

Locus ceruleus (LC)-supplied norepinephrine (NE) suppresses neuroinflammation in the brain. To elucidate the effect of LC degeneration and subsequent NE deficiency on Alzheimer's disease pathology, we evaluated NE effects on microglial key functions. NE stimulation of mouse microglia suppressed Abeta-induced cytokine and chemokine production and increased microglial migration and phagocytosis of Abeta. Induced degeneration of the locus ceruleus increased expression of inflammatory mediators in APP-transgenic mice and resulted in elevated Abeta deposition. In vivo laser microscopy confirmed a reduced recruitment of microglia to Abeta plaque sites and impaired microglial Abeta phagocytosis in NE-depleted APP-transgenic mice. Supplying the mice the norepinephrine precursor L-threo-DOPS restored microglial functions in NE-depleted mice. This indicates that decrease of NE in locus ceruleus projection areas facilitates the inflammatory reaction of microglial cells in AD and impairs microglial migration and phagocytosis, thereby contributing to reduced Abeta clearance. Consequently, therapies targeting microglial phagocytosis should be tested under NE depletion.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Locus Cerúleo/metabolismo , Locus Cerúleo/patologia , Microglia/metabolismo , Norepinefrina/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Quimiocinas/biossíntese , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Humanos , Técnicas In Vitro , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/patologia , Norepinefrina/deficiência , Norepinefrina/farmacologia , Fagocitose/efeitos dos fármacos , Fagocitose/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transcrição Gênica
13.
J Neurosci ; 31(19): 7049-59, 2011 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-21562267

RESUMO

Liver X receptors (LXRs) regulate immune cell function and cholesterol metabolism, both factors that are critically involved in Alzheimer's disease (AD). To investigate the therapeutic potential of long-term LXR activation in amyloid-ß (Aß) peptide deposition in an AD model, 13-month-old, amyloid plaque-bearing APP23 mice were treated with the LXR agonist TO901317. Postmortem analysis demonstrated that TO901317 efficiently crossed the blood-brain barrier. Insoluble and soluble Aß levels in the treated APP23 mice were reduced by 80% and 40%, respectively, compared with untreated animals. Amyloid precursor protein (APP) processing, however, was hardly changed by the compound, suggesting that the observed effects were instead mediated by Aß disposal. Despite the profound effect on Aß levels, spatial learning in the Morris water maze was only slightly improved by the treatment. ABCA1 (ATP-binding cassette transporter 1) and apolipoprotein E (ApoE) protein levels were increased and found to be primarily localized in astrocytes. Experiments using primary microglia demonstrated that medium derived from primary astrocytes exposed to TO901317 stimulated phagocytosis of fibrillar Aß. Conditioned medium from TO901317-treated ApoE(-/-) or LXRα(-/-) astrocytes did not increase phagocytosis of Aß. In APP23 mice, long-term treatment with TO901317 strongly increased the association of microglia and Aß plaques. Short-term treatment of APP/PS1 mice with TO901317 also increased this association, which was dependent on the presence of LXRα and was accompanied by increased ApoE lipidation. Together, these data suggest that astrocytic LXRα activation and subsequent release of ApoE by astrocytes is critical for the ability of microglia to remove fibrillar Aß in response to treatment with TO901317.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Astrócitos/metabolismo , Microglia/metabolismo , Receptores Nucleares Órfãos/metabolismo , Fagocitose/fisiologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Apolipoproteínas E/genética , Astrócitos/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Meios de Cultivo Condicionados , Ensaio de Imunoadsorção Enzimática , Hidrocarbonetos Fluorados/farmacologia , Imunoensaio , Imuno-Histoquímica , Receptores X do Fígado , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Receptores Nucleares Órfãos/genética , Sulfonamidas/farmacologia
14.
J Neurosci ; 31(31): 11159-71, 2011 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-21813677

RESUMO

Mononuclear phagocytes are important modulators of Alzheimer's disease (AD), but the specific functions of resident microglia, bone marrow-derived mononuclear cells, and perivascular macrophages have not been resolved. To elucidate the spatiotemporal roles of mononuclear phagocytes during disease, we targeted myeloid cell subsets from different compartments and examined disease pathogenesis in three different mouse models of AD (APP(swe/PS1), APP(swe), and APP23 mice). We identified chemokine receptor 2 (CCR2)-expressing myeloid cells as the population that was preferentially recruited to ß-amyloid (Aß) deposits. Unexpectedly, AD brains with dysfunctional microglia and devoid of parenchymal bone marrow-derived phagocytes did not show overt changes in plaque pathology and Aß load. In contrast, restriction of CCR2 deficiency to perivascular myeloid cells drastically impaired ß-amyloid clearance and amplified vascular Aß deposition, while parenchymal plaque deposition remained unaffected. Together, our data advocate selective functions of CCR2-expressing myeloid subsets, which could be targeted specifically to modify disease burden in AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Microglia/fisiologia , Células Mieloides/classificação , Células Mieloides/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/cirurgia , Precursor de Proteína beta-Amiloide/genética , Animais , Transplante de Medula Óssea/métodos , Antígeno CD11b/metabolismo , Morte Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Transgênicos , Microdissecção/métodos , Mutação/genética , Receptores CCR2/deficiência , Receptores CCR2/metabolismo , Irradiação Corporal Total/métodos
15.
J Neurochem ; 116(1): 43-52, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21044080

RESUMO

Both hypercortisolemia and hippocampal damage are features found in patients diagnosed of Alzheimer's disease (AD) and epidemiological evidence supports a role for stress as a risk factor for AD. It is known that immobilization stress is followed by accumulation of oxidative/nitrosative mediators in brain after the release of proinflammatory cytokines, nuclear factor kappa B activation, nitric oxide synthase-2 and cyclooxygenase-2 expression. Long-term exposure to elevated corticosteroid levels is known to affect the hippocampus which plays a central role in the regulation of the hypothalamic-pituitary-adrenal axis. We therefore studied the effect of chronic immobilization stress on amyloid precursor protein/PS1 mice. Stress exposure increased AD-induced neuroinflammation characterized by astrogliosis, increased inflammatory gene transcription and lipid peroxidation. Importantly, immobilization stress did not increase the soluble or insoluble amyloid ß levels suggesting that increased cortisol levels lower the threshold for a neuroinflammatory response, independently from amyloid ß. Since inflammation may act as a factor that contributes disease progression, the stress-inflammation relation described here may be relevant to understand the initial mechanisms in underlying the risk enhancing action of stress on AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Fragmentos de Peptídeos/metabolismo , Presenilina-1/metabolismo , Estresse Psicológico/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Córtex Cerebral/patologia , Hipocampo/patologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Presenilina-1/genética , Restrição Física , Estresse Psicológico/patologia
16.
Alzheimers Dement ; 7(4): 386-395.e6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21784349

RESUMO

BACKGROUND: The cerebrospinal fluid (CSF) biomarkers amyloid ß (Aß)-42, total-tau (T-tau), and phosphorylated-tau (P-tau) demonstrate good diagnostic accuracy for Alzheimer's disease (AD). However, there are large variations in biomarker measurements between studies, and between and within laboratories. The Alzheimer's Association has initiated a global quality control program to estimate and monitor variability of measurements, quantify batch-to-batch assay variations, and identify sources of variability. In this article, we present the results from the first two rounds of the program. METHODS: The program is open for laboratories using commercially available kits for Aß, T-tau, or P-tau. CSF samples (aliquots of pooled CSF) are sent for analysis several times a year from the Clinical Neurochemistry Laboratory at the Mölndal campus of the University of Gothenburg, Sweden. Each round consists of three quality control samples. RESULTS: Forty laboratories participated. Twenty-six used INNOTEST enzyme-linked immunosorbent assay kits, 14 used Luminex xMAP with the INNO-BIA AlzBio3 kit (both measure Aß-(1-42), P-tau(181P), and T-tau), and 5 used Meso Scale Discovery with the Aß triplex (AßN-42, AßN-40, and AßN-38) or T-tau kits. The total coefficients of variation between the laboratories were 13% to 36%. Five laboratories analyzed the samples six times on different occasions. Within-laboratory precisions differed considerably between biomarkers within individual laboratories. CONCLUSIONS: Measurements of CSF AD biomarkers show large between-laboratory variability, likely caused by factors related to analytical procedures and the analytical kits. Standardization of laboratory procedures and efforts by kit vendors to increase kit performance might lower variability, and will likely increase the usefulness of CSF AD biomarkers.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/diagnóstico , Biomarcadores/líquido cefalorraquidiano , Controle de Qualidade , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Bioensaio/métodos , Ensaio de Imunoadsorção Enzimática , Humanos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fosforilação , Reprodutibilidade dos Testes , Suécia , Fatores de Tempo , Proteínas tau/líquido cefalorraquidiano
17.
J Neurosci ; 29(45): 14177-84, 2009 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-19906966

RESUMO

To date, long-term consequences of septic encephalopathy on cerebral metabolism, cognition, learning, and memory capabilities and factors involved are poorly understood. In this study, we used a murine sepsis model to demonstrate that bacterial lipopolysaccharide (LPS) causes long-term cognitive deficits in mice. Two months after LPS treatment, wild-type mice committed more working and reference memory errors than controls. The behavioral impairment was independent of the cerebral glucose uptake as evidenced by (18)F-Fluordeoxyglucose small animal positron emission tomography. In contrast, mice deficient for the inducible nitric oxide synthase gene (NOS2-/-) did not show any cognitive changes when challenged with LPS. Immunohistochemical analysis demonstrated that LPS did not lead to neuronal cell death but caused sustained microglial activation in wild-type as compared to NOS2-/- mice. Expression analysis showed that LPS-treated NOS2-/- mice had lower brain mRNA levels for proinflammatory factors compared with wild-type mice. Expression analysis demonstrated distinct changes in the content of synaptic proteins in wild-type mice, which were not observed in the NOS2-/- mice. Together, this data set outlines the importance of the NOS2 activation for long-term cerebral changes after severe sepsis.


Assuntos
Transtornos Cognitivos/etiologia , Transtornos Cognitivos/fisiopatologia , Óxido Nítrico Sintase Tipo II/genética , Sepse/complicações , Sepse/fisiopatologia , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/fisiopatologia , Morte Celular/efeitos dos fármacos , Glucose/metabolismo , Lipopolissacarídeos , Aprendizagem em Labirinto/fisiologia , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/fisiologia , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/metabolismo , Cintilografia , Sepse/induzido quimicamente , Sinapses/fisiologia
18.
J Neuroinflammation ; 7: 61, 2010 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-20937084

RESUMO

Microglial activation plays an integral role in the development and course of neurodegeneration. Although neuropeptides such as bradykinin (BK), somatostatin (SST), and endothelin (ET) are known to be important mediators of inflammation in the periphery, evidence of a similar function in brain is scarce. Using immunocytochemistry, we demonstrate the expression of receptors for BK (B1, B2 subtypes), ET (ETA, ETB subtypes) and SST (SST 2, 3, 4 subtypes) in primary microglia and microglial cell lines. Exposure of BV2 and N9, as well as primary microglial cells to BK or SST increased Aß uptake in a concentration-dependent manner, whereas endothelin decreased Aß uptake. This was caused by increased phagocytosis of Aß since the rate of intracellular Aß degradation remained unaffected. All neuropeptides increased chemotactic activity of microglia. In addition, BK reduced Aß-induced expression of proinflammatory genes including iNOS and COX-2. ET decreased the Aß-induced expression of monocyte chemoattractant protein 1 and interleukin-6. These results suggest that neuropeptides play an important role in chemotaxis and Aß clearance and modulate the brain's response to neuroinflammatory processes.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Movimento Celular/fisiologia , Microglia/metabolismo , Fagocitose/fisiologia , Receptores da Bradicinina/metabolismo , Receptores de Endotelina/metabolismo , Receptores de Somatostatina/metabolismo , Análise de Variância , Animais , Western Blotting , Linhagem Celular , Células Cultivadas , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Imuno-Histoquímica , Camundongos , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
J Neural Transm (Vienna) ; 117(8): 919-47, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20632195

RESUMO

Generation of neurotoxic amyloid beta peptides and their deposition along with neurofibrillary tangle formation represent key pathological hallmarks in Alzheimer's disease (AD). Recent evidence suggests that inflammation may be a third important component which, once initiated in response to neurodegeneration or dysfunction, may actively contribute to disease progression and chronicity. Various neuroinflammatory mediators including complement activators and inhibitors, chemokines, cytokines, radical oxygen species and inflammatory enzyme systems are expressed and released by microglia, astrocytes and neurons in the AD brain. Degeneration of aminergic brain stem nuclei including the locus ceruleus and the nucleus basalis of Meynert may facilitate the occurrence of inflammation in their projection areas given the antiinflammatory and neuroprotective action of their key transmitters norepinephrine and acetylcholine. While inflammation has been thought to arise secondary to degeneration, recent experiments demonstrated that inflammatory mediators may stimulate amyloid precursor protein processing by various means and therefore can establish a vicious cycle. Despite the fact that some aspects of inflammation may even be protective for bystander neurons, antiinflammatory treatment strategies should therefore be considered. Non-steroidal anti-inflammatory drugs have been shown to reduce the risk and delay the onset to develop AD. While, the precise molecular mechanism underlying this effect is still unknown, a number of possible mechanisms including cyclooxygenase 2 or gamma-secretase inhibition and activation of the peroxisome proliferator activated receptor gamma may alone or, more likely, in concert account for the epidemiologically observed protection.


Assuntos
Doença de Alzheimer , Encéfalo/patologia , Inflamação/etiologia , Inflamação/patologia , Doença de Alzheimer/complicações , Doença de Alzheimer/imunologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/imunologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/imunologia , Cromogranina A/imunologia , Cromogranina A/metabolismo , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/uso terapêutico , Emaranhados Neurofibrilares/imunologia
20.
J Neurochem ; 109(6): 1779-90, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19457135

RESUMO

Glioblastoma represent the most common primary brain tumor in adults and are currently considered incurable. We investigated antiproliferative and anti-invasive mechanisms of 6-OH-11-O-hydroxyfenantrene (IIF), a retinoid X receptor ligand, and pioglitazone (PGZ), a peroxisome proliferator-activated receptor gamma activator, in three different glioblastoma cell lines. A dose-dependent reduction of tumor invasion and strong decrease of matrix metalloproteinases 2 and 9 expression was observed, especially when a combination therapy of IIF and PGZ was administered. Combined treatment also markedly reduced proliferation and induced apoptosis in all glioma cell lines tested. This was in particular accompanied by decrease of antiapoptotic proteins Bcl2 and p53, while simultaneously pro-apoptotic cytochrome c, cleaved caspase 3, Bax and Bad levels increased. These in vitro findings were further substantiated in a murine glioma model in vivo, where oral administration of PGZ and IIF resulted in significantly reduced tumor volume and proliferation. Of note, treatment with nuclear receptor ligands was not only effective when the treatment was initiated shortly after the intraparenchymal seeding of the glioma cells, but even when initiated in the last third of the observation period. Collectively, our results demonstrate the effectiveness of a combined treatment of ligands of proliferator-activated receptor and retinoid X receptor against glioblastoma.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/tratamento farmacológico , PPAR gama/uso terapêutico , Tretinoína/análogos & derivados , Análise de Variância , Animais , Anexina A5/metabolismo , Bromodesoxiuridina/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Citocromos c/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Ligantes , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica/fisiopatologia , PPAR gama/metabolismo , Pioglitazona , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Receptor X Retinoide gama/metabolismo , Sais de Tetrazólio , Tiazóis , Tiazolidinedionas/uso terapêutico , Transfecção/métodos , Tretinoína/uso terapêutico , Ensaio Tumoral de Célula-Tronco/métodos , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA