Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 40(24): e108662, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34825707

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Precursor de Proteína beta-Amiloide/genética , Antígeno B7-H1/metabolismo , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Regulación hacia Arriba , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/toxicidad , Animales , Astrocitos/metabolismo , Antígenos CD36/metabolismo , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Persona de Mediana Edad
2.
Acta Neuropathol ; 147(1): 65, 2024 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-38557897

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Animales , Humanos , Enfermedad de Alzheimer/patología , Proteínas tau/genética , Proteínas tau/metabolismo , Transcriptoma , Encéfalo/patología , Células Mieloides/patología , Microglía/patología , Péptidos beta-Amiloides/metabolismo
3.
EMBO Rep ; 22(11): e51696, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34569685

RESUMEN

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.


Asunto(s)
Astrocitos , Microglía , Animales , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Microglía/metabolismo , Enfermedades Neuroinflamatorias
4.
Nature ; 552(7685): 355-361, 2017 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-29293211

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Microglía/metabolismo , Agregación Patológica de Proteínas , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Anticuerpos/administración & dosificación , Anticuerpos/inmunología , Anticuerpos/farmacología , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Proteínas Adaptadoras de Señalización CARD/química , Proteínas Adaptadoras de Señalización CARD/inmunología , Femenino , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Presenilina-1/deficiencia , Presenilina-1/genética , Dominios Proteicos , Memoria Espacial/fisiología
5.
Nature ; 493(7434): 674-8, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23254930

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/patología , Proteínas Portadoras/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Conducta Animal , Encéfalo/enzimología , Proteínas Portadoras/genética , Caspasa 1/genética , Caspasa 1/metabolismo , Disfunción Cognitiva/enzimología , Disfunción Cognitiva/fisiopatología , Regulación Enzimológica de la Expresión Génica , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fagocitosis/genética
6.
Hum Mol Genet ; 23(24): 6644-58, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25027320

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Apolipoproteína E4/genética , Proteínas Nucleares/genética , Fragmentos de Péptidos/genética , Polimorfismo de Nucleótido Simple , Proteínas de Unión al ARN/genética , Proteínas tau/genética , Anciano , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/líquido cefalorraquídeo , Apolipoproteína E4/líquido cefalorraquídeo , Cognición , Femenino , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Proteínas Nucleares/líquido cefalorraquídeo , Fragmentos de Péptidos/líquido cefalorraquídeo , Fosforilación , Proteínas de Unión al ARN/líquido cefalorraquídeo , Factores de Empalme Serina-Arginina , Transducción de Señal , Proteínas tau/líquido cefalorraquídeo
7.
J Neurosci ; 34(26): 8845-54, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24966384

RESUMEN

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.


Asunto(s)
Neurotoxina Derivada del Eosinófilo/metabolismo , Aprendizaje/fisiología , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/metabolismo , Memoria/fisiología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Neurotoxina Derivada del Eosinófilo/genética , Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Trastornos de la Memoria/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Norepinefrina/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
8.
Gastroenterology ; 146(1): 176-87.e1, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24067878

RESUMEN

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.


Asunto(s)
Motilidad Gastrointestinal/inmunología , Ileus/inmunología , Interleucina-1/inmunología , Músculo Liso/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Plexo Mientérico/inmunología , Neuroglía/inmunología , Complicaciones Posoperatorias/inmunología , Receptores Tipo I de Interleucina-1/inmunología , Animales , Modelos Animales de Enfermedad , Ileus/metabolismo , Interleucina-1/metabolismo , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Plexo Mientérico/metabolismo , Neuroglía/metabolismo , Complicaciones Posoperatorias/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Transducción de Señal , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
9.
J Neurosci ; 32(48): 17321-31, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23197723

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Microglía/efectos de los fármacos , PPAR gamma/agonistas , Fagocitosis/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Cognición/fisiología , Modelos Animales de Enfermedad , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Aprendizaje por Laberinto/fisiología , Ratones , Microglía/metabolismo , Fagocitosis/fisiología , Pioglitazona , Presenilina-1/genética , Presenilina-1/metabolismo , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico
10.
J Neurosci ; 32(49): 17824-9, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23223301

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Calgranulina B/metabolismo , Calgranulina B/fisiología , Microglía/fisiología , Fagocitosis/fisiología , Anciano , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/líquido cefalorraquídeo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Encéfalo/metabolismo , Calgranulina A/metabolismo , Estudios de Casos y Controles , Disfunción Cognitiva/sangre , Disfunción Cognitiva/líquido cefalorraquídeo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Fagocitosis/genética , Presenilina-1/genética
11.
Proc Natl Acad Sci U S A ; 107(13): 6058-63, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20231476

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Microglía/metabolismo , Norepinefrina/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Humanos , Técnicas In Vitro , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Norepinefrina/deficiencia , Norepinefrina/farmacología , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transcripción Genética
12.
J Neurosci ; 31(19): 7049-59, 2011 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-21562267

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Astrocitos/metabolismo , Microglía/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Fagocitosis/fisiología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Apolipoproteínas E/genética , Astrocitos/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Medios de Cultivo Condicionados , Ensayo de Inmunoadsorción Enzimática , Hidrocarburos Fluorados/farmacología , Inmunoensayo , Inmunohistoquímica , Receptores X del Hígado , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Receptores Nucleares Huérfanos/genética , Sulfonamidas/farmacología
13.
J Neurosci ; 31(31): 11159-71, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21813677

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Microglía/fisiología , Células Mieloides/clasificación , Células Mieloides/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/cirugía , Precursor de Proteína beta-Amiloide/genética , Animales , Trasplante de Médula Ósea/métodos , Antígeno CD11b/metabolismo , Muerte Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Antígeno Ki-67/metabolismo , Ratones , Ratones Transgénicos , Microdisección/métodos , Mutación/genética , Receptores CCR2/deficiencia , Receptores CCR2/metabolismo , Irradiación Corporal Total/métodos
14.
J Neurochem ; 116(1): 43-52, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21044080

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Corteza Cerebral/metabolismo , Hipocampo/metabolismo , Fragmentos de Péptidos/metabolismo , Presenilina-1/metabolismo , Estrés Psicológico/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Corteza Cerebral/patología , Hipocampo/patología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/genética , Restricción Física , Estrés Psicológico/patología
15.
Alzheimers Dement ; 7(4): 386-395.e6, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21784349

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/diagnóstico , Biomarcadores/líquido cefalorraquídeo , Control de Calidad , Péptidos beta-Amiloides/líquido cefalorraquídeo , Bioensayo/métodos , Ensayo de Inmunoadsorción Enzimática , Humanos , Fragmentos de Péptidos/líquido cefalorraquídeo , Fosforilación , Reproducibilidad de los Resultados , Suecia , Factores de Tiempo , Proteínas tau/líquido cefalorraquídeo
16.
J Neurosci ; 29(45): 14177-84, 2009 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-19906966

RESUMEN

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.


Asunto(s)
Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/fisiopatología , Óxido Nítrico Sintasa de Tipo II/genética , Sepsis/complicaciones , Sepsis/fisiopatología , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Muerte Celular/efectos de los fármacos , Glucosa/metabolismo , Lipopolisacáridos , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/fisiología , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Mensajero/metabolismo , Cintigrafía , Sepsis/inducido químicamente , Sinapsis/fisiología
17.
J Neuroinflammation ; 7: 61, 2010 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-20937084

RESUMEN

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.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Movimiento Celular/fisiología , Microglía/metabolismo , Fagocitosis/fisiología , Receptores de Bradiquinina/metabolismo , Receptores de Endotelina/metabolismo , Receptores de Somatostatina/metabolismo , Análisis de Varianza , Animales , Western Blotting , Línea Celular , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Inmunohistoquímica , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Neurochem ; 109(6): 1779-90, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19457135

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , PPAR gamma/uso terapéutico , Tretinoina/análogos & derivados , Análisis de Varianza , Animales , Anexina A5/metabolismo , Bromodesoxiuridina/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Ligandos , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica/fisiopatología , PPAR gamma/metabolismo , Pioglitazona , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Receptor gamma X Retinoide/metabolismo , Sales de Tetrazolio , Tiazoles , Tiazolidinedionas/uso terapéutico , Transfección/métodos , Tretinoina/uso terapéutico , Ensayo de Tumor de Célula Madre/métodos , Proteína X Asociada a bcl-2/metabolismo
19.
Mol Neurobiol ; 56(9): 6539-6550, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30852796

RESUMEN

The hypothesis that accumulation of beta-amyloid (Aß) species in the brain represents a major event in Alzheimer's disease (AD) pathogenesis still prevails; nevertheless, an array of additional pathological processes contributes to clinical presentation and disease progression. We sought to identify novel targets for AD within genes related to amyloid precursor protein (APP) processing, innate immune responses, and the catecholamine system. Through a series of bioinformatics analyses, we identified TLR5 and other genes involved in toll-like receptor (TLR) signaling as potential AD targets. It is believed that Aß species induce activation of microglia and astrocytes in AD, with a negative impact on disease progression. The TAM (Tyro3, Axl, Mer) family of receptor tyrosine kinases plays pivotal roles in limiting inflammatory responses upon TLR stimulation, for which we further studied their implication in the TLR5 alterations observed in AD. We validated the up-regulation of TLR5 in the frontal cortex of moderate AD cases. In addition, we observed up-regulation of the TAM ligands protein S (PROS1), galectin-3 (LGALS3), and Tulp-1. Furthermore, we identified an association of the TAM ligand GAS6 with AD progression. In THP-1 cells, co-stimulation with Aß and flagellin for 24 h induced up-regulation of TYRO3 and GAS6, which could be prevented by neutralization of TLR5. Our results underscore the role of TLR dysregulations in AD, suggesting the presence of an immunosuppressive response during moderate disease stages, and implicate TAM signaling in AD immune dysregulation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Progresión de la Enfermedad , Receptor Toll-Like 5/metabolismo , Enfermedad de Alzheimer/genética , Amiloide/metabolismo , Biomarcadores/metabolismo , Humanos , Ligandos , Mapas de Interacción de Proteínas , Transducción de Señal , Células THP-1 , Receptor Toll-Like 5/genética
20.
J Neuroinflammation ; 5: 38, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18793399

RESUMEN

BACKGROUND: Septic encephalopathy is a severe brain dysfunction caused by systemic inflammation in the absence of direct brain infection. Changes in cerebral blood flow, release of inflammatory molecules and metabolic alterations contribute to neuronal dysfunction and cell death. METHODS: To investigate the relation of electrophysiological, metabolic and morphological changes caused by SE, we simultaneously assessed systemic circulation, regional cerebral blood flow and cortical electroencephalography in rats exposed to bacterial lipopolysaccharide. Additionally, cerebral glucose uptake, astro- and microglial activation as well as changes of inflammatory gene transcription were examined by small animal PET using [18F]FDG, immunohistochemistry, and real time PCR. RESULTS: While the systemic hemodynamic did not change significantly, regional cerebral blood flow was decreased in the cortex paralleled by a decrease of alpha activity of the electroencephalography. Cerebral glucose uptake was reduced in all analyzed neocortical areas, but preserved in the caudate nucleus, the hippocampus and the thalamus. Sepsis enhanced the transcription of several pro- and anti-inflammatory cytokines and chemokines including tumor necrosis factor alpha, interleukin-1 beta, transforming growth factor beta, and monocot chemoattractant protein 1 in the cerebrum. Regional analysis of different brain regions revealed an increase in ED1-positive microglia in the cortex, while total and neuronal cell counts decreased in the cortex and the hippocampus. CONCLUSION: Together, the present study highlights the complexity of sepsis induced early impairment of neuronal metabolism and activity. Since our model uses techniques that determine parameters relevant to the clinical setting, it might be a useful tool to develop brain specific therapeutic strategies for human septic encephalopathy.


Asunto(s)
Encéfalo/inmunología , Encéfalo/metabolismo , Inflamación/inmunología , Sepsis/inmunología , Animales , Encéfalo/anatomía & histología , Encéfalo/patología , Circulación Cerebrovascular , Electroencefalografía , Glucosa/metabolismo , Hemodinámica , Humanos , Lipopolisacáridos/inmunología , Masculino , Microglía/metabolismo , Neuronas/citología , Neuronas/fisiología , Tomografía de Emisión de Positrones , Distribución Aleatoria , Ratas , Ratas Wistar , Flujo Sanguíneo Regional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA