Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Alzheimers Dement ; 14(2): 243-252, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28755839

RESUMO

INTRODUCTION: Our previous studies have shown that amyloid ß peptide (Aß) is subject to complement-mediated clearance from the peripheral circulation, and that this mechanism is deficient in Alzheimer's disease. The mechanism should be enhanced by Aß antibodies that form immune complexes (ICs) with Aß, and therefore may be relevant to current Aß immunotherapy approaches. METHODS: Multidisciplinary methods were employed to demonstrate enhanced complement-mediated capture of Aß antibody immune complexes compared with Aß alone in both erythrocytes and THP1-derived macrophages. RESULTS: Aß antibodies dramatically increased complement activation and opsonization of Aß, followed by commensurately enhanced Aß capture by human erythrocytes and macrophages. These in vitro findings were consistent with enhanced peripheral clearance of intravenously administered Aß antibody immune complexes in nonhuman primates. DISCUSSION: Together with our previous results, showing significant Alzheimer's disease deficits in peripheral Aß clearance, the present findings strongly suggest that peripheral mechanisms should not be ignored as contributors to the effects of Aß immunotherapy.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/imunologia , Anticorpos/sangue , Proteínas do Sistema Complemento/metabolismo , Eritrócitos/metabolismo , Imunoterapia/métodos , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/metabolismo , Animais , Adesão Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Feminino , Humanos , Fatores Imunológicos , Macaca fascicularis , Macrófagos/metabolismo , Masculino , Fagocitose , Células THP-1/metabolismo , Células THP-1/patologia
2.
Alzheimers Dement ; 14(11): 1438-1449, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29792870

RESUMO

INTRODUCTION: Genome-wide association studies consistently show that single nucleotide polymorphisms (SNPs) in the complement receptor 1 (CR1) gene modestly but significantly alter Alzheimer's disease (AD) risk. Follow-up research has assumed that CR1 is expressed in the human brain despite a paucity of evidence for its function there. Alternatively, erythrocytes contain >80% of the body's CR1, where, in primates, it is known to bind circulating pathogens. METHODS: Multidisciplinary methods were employed. RESULTS: Conventional Western blots and quantitative polymerase chain reaction failed to detect CR1 in the human brain. Brain immunohistochemistry revealed only vascular CR1. By contrast, erythrocyte CR1 immunoreactivity was readily observed and was significantly deficient in AD, as was CR1-mediated erythrocyte capture of circulating amyloid ß peptide. CR1 SNPs associated with decreased erythrocyte CR1 increased AD risk, whereas a CR1 SNP associated with increased erythrocyte CR1 decreased AD risk. DISCUSSION: SNP effects on erythrocyte CR1 likely underlie the association of CR1 polymorphisms with AD risk.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Polimorfismo de Nucleotídeo Único , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Apolipoproteínas E/genética , Eritrócitos/metabolismo , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Microglia/metabolismo , Neocórtex/metabolismo , Estudos Prospectivos , Isoformas de Proteínas , Receptores de Complemento 3b/química
3.
Alzheimers Dement ; 13(12): 1397-1409, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28475854

RESUMO

INTRODUCTION: Although amyloid ß peptide (Aß) is cleared from the brain to cerebrospinal fluid and the peripheral circulation, mechanisms for its removal from blood remain unresolved. Primates have uniquely evolved a highly effective peripheral clearance mechanism for pathogens, immune adherence, in which erythrocyte complement receptor 1 (CR1) plays a major role. METHODS: Multidisciplinary methods were used to demonstrate immune adherence capture of Aß by erythrocytes and its deficiency in Alzheimer's disease (AD). RESULTS: Aß was shown to be subject to immune adherence at every step in the pathway. Aß dose-dependently activated serum complement. Complement-opsonized Aß was captured by erythrocytes via CR1. Erythrocytes, Aß, and hepatic Kupffer cells were colocalized in the human liver. Significant deficits in erythrocyte Aß levels were found in AD and mild cognitive impairment patients. DISCUSSION: CR1 polymorphisms elevate AD risk, and >80% of human CR1 is vested in erythrocytes to subserve immune adherence. The present results suggest that this pathway is pathophysiologically relevant in AD.


Assuntos
Doença de Alzheimer/sangue , Peptídeos beta-Amiloides/metabolismo , Disfunção Cognitiva/sangue , Eritrócitos/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores de Complemento/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/farmacologia , Animais , Estudos de Casos e Controles , Disfunção Cognitiva/patologia , Disfunção Cognitiva/fisiopatologia , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Feminino , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/ultraestrutura , Macaca fascicularis/sangue , Masculino , Testes de Estado Mental e Demência , Microscopia Eletrônica , Pessoa de Meia-Idade , Fragmentos de Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Receptores de Complemento/genética
4.
J Neurosci ; 34(17): 5882-94, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24760848

RESUMO

A persistent and nonresolving inflammatory response to accumulating Aß peptide species is a cardinal feature in the development of Alzheimer's disease (AD). In response to accumulating Aß peptide species, microglia, the innate immune cells of the brain, generate a toxic inflammatory response that accelerates synaptic and neuronal injury. Many proinflammatory signaling pathways are linked to progression of neurodegeneration. However, endogenous anti-inflammatory pathways capable of suppressing Aß-induced inflammation represent a relatively unexplored area. Here we report that signaling through the prostaglandin-E2 (PGE2) EP4 receptor potently suppresses microglial inflammatory responses to Aß42 peptides. In cultured microglial cells, EP4 stimulation attenuated levels of Aß42-induced inflammatory factors and potentiated phagocytosis of Aß42. Microarray analysis demonstrated that EP4 stimulation broadly opposed Aß42-driven gene expression changes in microglia, with enrichment for targets of IRF1, IRF7, and NF-κB transcription factors. In vivo, conditional deletion of microglial EP4 in APPSwe-PS1ΔE9 (APP-PS1) mice conversely increased inflammatory gene expression, oxidative protein modification, and Aß deposition in brain at early stages of pathology, but not at later stages, suggesting an early anti-inflammatory function of microglial EP4 signaling in the APP-PS1 model. Finally, EP4 receptor levels decreased significantly in human cortex with progression from normal to AD states, suggesting that early loss of this beneficial signaling system in preclinical AD development may contribute to subsequent progression of pathology.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/farmacologia , Inflamação/metabolismo , Microglia/metabolismo , Fragmentos de Peptídeos/farmacologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Humanos , Inflamação/patologia , Éteres Metílicos/farmacologia , Microglia/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP4/agonistas , Transdução de Sinais/efeitos dos fármacos
5.
J Neurosci ; 33(40): 16016-32, 2013 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-24089506

RESUMO

Prostaglandin E2 (PGE2), a potent lipid signaling molecule, modulates inflammatory responses through activation of downstream G-protein coupled EP(1-4) receptors. Here, we investigated the cell-specific in vivo function of PGE2 signaling through its E-prostanoid 2 (EP2) receptor in murine innate immune responses systemically and in the CNS. In vivo, systemic administration of lipopolysaccharide (LPS) resulted in a broad induction of cytokines and chemokines in plasma that was significantly attenuated in EP2-deficient mice. Ex vivo stimulation of peritoneal macrophages with LPS elicited proinflammatory responses that were dependent on EP2 signaling and that overlapped with in vivo plasma findings, suggesting that myeloid-lineage EP2 signaling is a major effector of innate immune responses. Conditional deletion of the EP2 receptor in myeloid lineage cells in Cd11bCre;EP2(lox/lox) mice attenuated plasma inflammatory responses and transmission of systemic inflammation to the brain was inhibited, with decreased hippocampal inflammatory gene expression and cerebral cortical levels of IL-6. Conditional deletion of EP2 significantly blunted microglial and astrocytic inflammatory responses to the neurotoxin MPTP and reduced striatal dopamine turnover. Suppression of microglial EP2 signaling also increased numbers of dopaminergic (DA) neurons in the substantia nigra independent of MPTP treatment, suggesting that microglial EP2 may influence development or survival of DA neurons. Unbiased microarray analysis of microglia isolated from adult Cd11bCre;EP2(lox/lox) and control mice demonstrated a broad downregulation of inflammatory pathways with ablation of microglial EP2 receptor. Together, these data identify a cell-specific proinflammatory role for macrophage/microglial EP2 signaling in innate immune responses systemically and in brain.


Assuntos
Encéfalo/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Microglia/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Inflamação/induzido quimicamente , Inflamação/genética , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP2/genética
6.
Ann Neurol ; 72(5): 788-98, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22915243

RESUMO

OBJECTIVE: There is significant evidence for a central role of inflammation in the development of Alzheimer disease (AD). Epidemiological studies indicate that chronic use of nonsteroidal anti-inflammatory drugs (NSAIDs) reduces the risk of developing AD in healthy aging populations. As NSAIDs inhibit the enzymatic activity of the inflammatory cyclooxygenases COX-1 and COX-2, these findings suggest that downstream prostaglandin signaling pathways function in the preclinical development of AD. Here, we investigate the function of prostaglandin E(2) (PGE(2) ) signaling through its EP3 receptor in the neuroinflammatory response to Aß peptide. METHODS: The function of PGE(2) signaling through its EP3 receptor was examined in vivo in a model of subacute neuroinflammation induced by administration of Aß(42) peptides. Our findings were then confirmed in young adult APPSwe-PS1ΔE9 transgenic mice. RESULTS: Deletion of the PGE(2) EP3 receptor in a model of Aß(42) peptide-induced neuroinflammation reduced proinflammatory gene expression, cytokine production, and oxidative stress. In the APPSwe-PS1ΔE9 model of familial AD, deletion of the EP3 receptor blocked induction of proinflammatory gene and protein expression and lipid peroxidation. In addition, levels of Aß peptides were significantly decreased, as were ß-secretase and ß C-terminal fragment levels, suggesting that generation of Aß peptides may be increased as a result of proinflammatory EP3 signaling. Finally, deletion of EP3 receptor significantly reversed the decline in presynaptic proteins seen in APPSwe-PS1ΔE9 mice. INTERPRETATION: Our findings identify the PGE(2) EP3 receptor as a novel proinflammatory, proamyloidogenic, and synaptotoxic signaling pathway, and suggest a role for COX-PGE(2) -EP3 signaling in the development of AD.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Dinoprostona/metabolismo , Encefalite/metabolismo , Regulação da Expressão Gênica/genética , Transdução de Sinais/fisiologia , Fatores Etários , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Animais Recém-Nascidos , Ácido Aspártico Endopeptidases/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Proteínas de Ligação ao Cálcio , Células Cultivadas , Disfunção Cognitiva/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Encefalite/induzido quimicamente , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Fragmentos de Peptídeos/farmacologia , Prostaglandina-Endoperóxido Sintases/metabolismo , RNA Mensageiro/metabolismo , Receptores de Prostaglandina E Subtipo EP3/deficiência , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Proteína 2 Associada à Membrana da Vesícula/metabolismo
7.
J Biol Chem ; 286(32): 28026-40, 2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21690086

RESUMO

Fast neurotransmission and slower hormone release share the same core fusion machinery consisting of SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins. In evoked neurotransmission, interactions between SNAREs and the Munc18-1 protein, a member of the Sec1/Munc18 (SM) protein family, are essential for exocytosis, whereas other SM proteins are dispensable. To address if the exclusivity of Munc18-1 demonstrated in neuroexocytosis also applied to fast insulin secretion, we characterized the presence and function of Munc18-1 and its closest homologue Munc18-2 in ß-cell stimulus-secretion coupling. We show that pancreatic ß-cells express both Munc18-1 and Munc18-2. The two Munc18 homologues exhibit different subcellular localization, and only Munc18-1 redistributes in response to glucose stimulation. However, both Munc18-1 and Munc18-2 augment glucose-stimulated hormone release. Ramp-like photorelease of caged Ca(2+) and high resolution whole-cell patch clamp recordings show that Munc18-1 and Munc18-2 overexpression shift the Ca(2+) sensitivity of the fastest phase of insulin exocytosis differently. In addition, we reveal that Ca(2+) sensitivity of exocytosis in ß-cells depends on the phosphorylation status of the Munc18 proteins. Even though Munc18-1 emerges as the key SM-protein determining the Ca(2+) threshold for triggering secretory activity in a stimulated ß-cell, Munc18-2 has the ability to increase Ca(2+) sensitivity and thus mediates the release of fusion-competent granules requiring a lower cytoplasmic-free Ca(2+) concentration, [Ca(2+)](i)(.) Hence, Munc18-1 and Munc18-2 display distinct subcellular compartmentalization and can coordinate the insulin exocytotic process differently as a consequence of the actual [Ca(2+)](i).


Assuntos
Cálcio/metabolismo , Exocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Munc18/metabolismo , Animais , Linhagem Celular Tumoral , Exocitose/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Humanos , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Proteínas Munc18/genética , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Edulcorantes/farmacologia
8.
PLoS Genet ; 4(11): e1000278, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19043548

RESUMO

Alternative splicing is an evolutionary innovation to create functionally diverse proteins from a limited number of genes. SNAP-25 plays a central role in neuroexocytosis by bridging synaptic vesicles to the plasma membrane during regulated exocytosis. The SNAP-25 polypeptide is encoded by a single copy gene, but in higher vertebrates a duplication of exon 5 has resulted in two mutually exclusive splice variants, SNAP-25a and SNAP-25b. To address a potential physiological difference between the two SNAP-25 proteins, we generated gene targeted SNAP-25b deficient mouse mutants by replacing the SNAP-25b specific exon with a second SNAP-25a equivalent. Elimination of SNAP-25b expression resulted in developmental defects, spontaneous seizures, and impaired short-term synaptic plasticity. In adult mutants, morphological changes in hippocampus and drastically altered neuropeptide expression were accompanied by severe impairment of spatial learning. We conclude that the ancient exon duplication in the Snap25 gene provides additional SNAP-25-function required for complex neuronal processes in higher eukaryotes.


Assuntos
Éxons/genética , Duplicação Gênica , Neurônios/fisiologia , Proteína 25 Associada a Sinaptossoma/genética , Processamento Alternativo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas do Domínio Duplacortina , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo
9.
J Neuroimmune Pharmacol ; 12(2): 292-304, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27734267

RESUMO

Inflammation is a ubiquitous factor accompanying normal aging and neurodegeneration, and recent studies indicate a major contribution of inducible cyclooxygenase (COX-2) and its downstream prostaglandin signaling pathways in modulating neuroinflammatory responses and neuronal function. We have previously shown that the prostaglandin PGE2 receptor EP4 suppresses innate immune responses in models of systemic inflammation. Here we investigated the role of the EP4 receptor in models of Parkinson's disease (PD). Systemic co-administration of the EP4 agonist ONO-AE1-329 with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) prevented loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) without significant changes in glial activation, suggesting a potent neuroprotective effect of EP4 signaling in this acute model of DA neuronal loss. Cell-specific conditional ablation of EP4 in Cd11bCre;EP4lox/lox mice exacerbated MPTP-associated glial activation and T-cell infiltration in SNpc, consistent with anti-inflammatory functions of microglial EP4 signaling. In vitro, in primary microglia stimulated with oligomeric α-synuclein, EP4 receptor activation suppressed generation of pro-inflammatory and oxidative stress factors. Taken together, these findings suggest a dual neuroprotective and anti-inflammatory mechanism of action by the EP4 receptor in models of PD.


Assuntos
Anti-Inflamatórios/uso terapêutico , Dinoprostona/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Transtornos Parkinsonianos/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais/fisiologia , Animais , Anti-Inflamatórios/farmacologia , Células Cultivadas , Masculino , Éteres Metílicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Transtornos Parkinsonianos/prevenção & controle , Receptores de Prostaglandina E Subtipo EP4/agonistas , Transdução de Sinais/efeitos dos fármacos
10.
Brain Res Mol Brain Res ; 138(2): 215-27, 2005 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-15908038

RESUMO

Cyclin-dependent kinase 5 (Cdk5) has emerged as a key coordinator of cell signaling in neurite outgrowth. Cdk5 needs to associate with one of the regulatory proteins p35 or p39 to be an active enzyme. To investigate if Cdk5 plays a role in the establishment of functional synapses, we have characterized the expression of Cdk5, p35, and p39 in the neuroblastoma-glioma cell line NG108-15, and recorded postsynaptic activity in myotubes in response to presynaptic overexpression of Cdk5, p35, and p39. Endogenous Cdk5 and p35 protein levels increased with cellular differentiation and preferentially distributed to soluble pools, whereas the level of p39 protein remained low and primarily was present in membrane and cytoskeletal fractions. Transient transfection of a dominant-negative mutant of Cdk5 in NG108-15 cells and subsequent culturing on differentiating muscle cells resulted in a significant reduction in synaptic activity, as measured by postsynaptic miniature endplate potentials (mEPPs). Overexpression of either Cdk5/p35 or Cdk5/p39 resulted in a substantial increase in synaptic structures that displayed postsynaptic activities, as well as mEPP frequency. These findings demonstrate that Cdk5, p35, and p39 are endogenously expressed in NG108-15 cells, exhibit distinct subcellular localizations, and that both Cdk5/p35 and Cdk5/p39 are central in formation of functional synapses.


Assuntos
Sistema Nervoso Central/enzimologia , Quinases Ciclina-Dependentes/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/enzimologia , Sinapses/enzimologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Membrana Celular/enzimologia , Sistema Nervoso Central/embriologia , Quinase 5 Dependente de Ciclina , Quinases Ciclina-Dependentes/genética , Citoesqueleto/enzimologia , Ativação Enzimática/fisiologia , Camundongos , Fibras Musculares Esqueléticas/enzimologia , Vias Neurais/embriologia , Junção Neuromuscular/embriologia , Junção Neuromuscular/enzimologia , Ratos , Transmissão Sináptica/genética , Regulação para Cima/fisiologia
11.
Curr Immunol Rev ; 11(2): 125-131, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28413375

RESUMO

The inflammatory response is a fundamental driving force in the pathogenesis of Alzheimer's disease (AD). In the setting of accumulating immunogenic Aß peptide assemblies, microglia, the innate immune cells of the brain, generate a non-resolving immune response and fail to adequately clear accumulating Aß peptides, accelerating neuronal and synaptic injury. Pathological, biomarker, and imaging studies point to a prominent role of the innate immune response in AD development, and the molecular components of this response are beginning to be unraveled. The inflammatory cyclooxygenase-PGE2 pathway is implicated in pre-clinical development of AD, both in epidemiology of normal aging populations and in transgenic mouse models of Familial AD. The cyclooxygenase-PGE2 pathway modulates the inflammatory response to accumulating Aß peptides through actions of specific E-prostanoid G-protein coupled receptors.

12.
Genom Data ; 5: 268-271, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26251825

RESUMO

Amyloid-ß (Aß) peptides accumulate in the brains of patients with Alzheimer's disease (AD), where they generate a persistent inflammatory response from microglia, the innate immune cells of the brain. The immune modulatory cyclooxygenase/prostaglandin E2 (COX/PGE2) pathway has been implicated in preclinical AD development, both in human epidemiology studies1 and in transgenic rodent models of AD2, 3. PGE2 signals through four G-protein-coupled receptors, including the EP2 receptor that has been investigated for its role in mediating the inflammatory and phagocytic responses to Aß4. To identify transcriptional differences in microglia lacking the EP2 receptor, we examined mice with EP2 conditionally deleted in Cd11b-expressing immune cells. We injected Aß peptides or saline vehicle into the brains of adult mice, isolated primary microglia, and analyzed RNA expression by microarray. The resulting datasets were analyzed in two studies5, 6, one describing the basal status of microglia with or without EP2 deletion, and the second study analyzing the microglial response to Aß. Here we describe in detail the experimental design and data analyses. The raw data from these studies are deposited in GEO, accession GSE57181 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE57181).

13.
J Clin Invest ; 125(1): 350-64, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25485684

RESUMO

Microglia, the innate immune cells of the CNS, perform critical inflammatory and noninflammatory functions that maintain normal neural function. For example, microglia clear misfolded proteins, elaborate trophic factors, and regulate and terminate toxic inflammation. In Alzheimer's disease (AD), however, beneficial microglial functions become impaired, accelerating synaptic and neuronal loss. Better understanding of the molecular mechanisms that contribute to microglial dysfunction is an important objective for identifying potential strategies to delay progression to AD. The inflammatory cyclooxygenase/prostaglandin E2 (COX/PGE2) pathway has been implicated in preclinical AD development, both in human epidemiology studies and in transgenic rodent models of AD. Here, we evaluated murine models that recapitulate microglial responses to Aß peptides and determined that microglia-specific deletion of the gene encoding the PGE2 receptor EP2 restores microglial chemotaxis and Aß clearance, suppresses toxic inflammation, increases cytoprotective insulin-like growth factor 1 (IGF1) signaling, and prevents synaptic injury and memory deficits. Our findings indicate that EP2 signaling suppresses beneficial microglia functions that falter during AD development and suggest that inhibition of the COX/PGE2/EP2 immune pathway has potential as a strategy to restore healthy microglial function and prevent progression to AD.


Assuntos
Doença de Alzheimer/metabolismo , Microglia/imunologia , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Transdução de Sinais , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Quimiotaxia/imunologia , Dinoprostona/fisiologia , Feminino , Expressão Gênica , Hipocampo/imunologia , Hipocampo/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/imunologia , Terminações Pré-Sinápticas/metabolismo , Memória Espacial , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA