Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Biol Cell ; 33(11): ar99, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35731557

RESUMEN

Microglia are the primary resident innate immune cells of the CNS. They possess branched, motile cell processes that are important for their cellular functions. To study the pathways that control microglial morphology and motility under physiological and disease conditions, it is necessary to quantify microglial morphology and motility precisely and reliably. Several image analysis approaches are available for the quantification of microglial morphology and motility. However, they are either not automated, not freely accessible, and/or limited in the number of morphology and motility parameters that can be assessed. Thus, we have developed MotiQ, an open-source, freely accessible software for automated quantification of microglial motility and morphology. MotiQ allows quantification of a diverse set of cellular motility and morphology parameters, including the parameters that have become the gold standard in the microglia field. We demonstrate that MotiQ can be applied to in vivo, ex vivo, and in vitro data from confocal, epifluorescence, or two-photon microscopy, and we compare its results to other analysis approaches. We suggest MotiQ as a versatile and customizable tool to study microglia.


Asunto(s)
Microglía , Movimiento Celular/fisiología , Microglía/metabolismo
2.
Cells ; 12(1)2022 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-36611872

RESUMEN

Amyloid-ß (Aß) deposition is an initiating factor in Alzheimer's disease (AD). Microglia are the brain immune cells that surround and phagocytose Aß plaques, but their phagocytic capacity declines in AD. This is in agreement with studies that associate AD risk loci with genes regulating the phagocytic function of immune cells. Immunotherapies are currently pursued as strategies against AD and there are increased efforts to understand the role of the immune system in ameliorating AD pathology. Here, we evaluated the effect of the Aß targeting ACI-24 vaccine in reducing AD pathology in an amyloidosis mouse model. ACI-24 vaccination elicited a robust and sustained antibody response in APPPS1 mice with an accompanying reduction of Aß plaque load, Aß plaque-associated ApoE and dystrophic neurites as compared to non-vaccinated controls. Furthermore, an increased number of NLRP3-positive plaque-associated microglia was observed following ACI-24 vaccination. In contrast to this local microglial activation at Aß plaques, we observed a more ramified morphology of Aß plaque-distant microglia compared to non-vaccinated controls. Accordingly, bulk transcriptomic analysis revealed a trend towards the reduced expression of several disease-associated microglia (DAM) signatures that is in line with the reduced Aß plaque load triggered by ACI-24 vaccination. Our study demonstrates that administration of the Aß targeting vaccine ACI-24 reduces AD pathology, suggesting its use as a safe and cost-effective AD therapeutic intervention.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Ratones , Animales , Microglía/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ratones Transgénicos , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/metabolismo , Amiloidosis/metabolismo , Placa Amiloide/metabolismo , Fenotipo , Vacunación
3.
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
5.
Front Neurol ; 12: 654850, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054698

RESUMEN

Purinergic signaling regulates neuronal and glial cell functions in the healthy CNS. In neurodegenerative diseases, purinergic signaling becomes dysregulated and can affect disease-associated phenotypes of glial cells. In this review, we discuss how cell-specific expression patterns of purinergic signaling components change in neurodegeneration and how dysregulated glial purinergic signaling and crosstalk may contribute to disease pathophysiology, thus bearing promising potential for the development of new therapeutical options for neurodegenerative diseases.

6.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33108356

RESUMEN

Microglia maintain homeostasis in the brain. However, with age, they become primed and respond more strongly to inflammatory stimuli. We show here that microglia from aged mice had upregulated mTOR complex 1 signaling controlling translation, as well as protein levels of inflammatory mediators. Genetic ablation of mTOR signaling showed a dual yet contrasting effect on microglia priming: it caused an NF-κB-dependent upregulation of priming genes at the mRNA level; however, mice displayed reduced cytokine protein levels, diminished microglia activation, and milder sickness behavior. The effect on translation was dependent on reduced phosphorylation of 4EBP1, resulting in decreased binding of eIF4E to eIF4G. Similar changes were present in aged human microglia and in damage-associated microglia, indicating that upregulation of mTOR-dependent translation is an essential aspect of microglia priming in aging and neurodegeneration.


Asunto(s)
Envejecimiento/metabolismo , Microglía/enzimología , Biosíntesis de Proteínas , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Envejecimiento/genética , Animales , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Factor 4G Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/metabolismo , Humanos , Ratones , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/metabolismo , Fosforilación/genética , Serina-Treonina Quinasas TOR/genética
7.
Cell Rep Med ; 1(9): 100159, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33377130

RESUMEN

Stroke leads to the degeneration of short-range and long-range axonal connections emanating from peri-infarct tissue, but it also induces novel axonal projections. However, this regeneration is hampered by growth-inhibitory properties of peri-infarct tissue and fibrotic scarring. Here, we tested the effects of epothilone B and epothilone D, FDA-approved microtubule-stabilizing drugs that are powerful modulators of axonal growth and scar formation, on neuroplasticity and motor outcomes in a photothrombotic mouse model of cortical stroke. We find that both drugs, when administered systemically 1 and 15 days after stroke, augment novel peri-infarct projections connecting the peri-infarct motor cortex with neighboring areas. Both drugs also increase the magnitude of long-range motor projections into the brainstem and reduce peri-infarct fibrotic scarring. Finally, epothilone treatment induces an improvement in skilled forelimb motor function. Thus, pharmacological microtubule stabilization represents a promising target for therapeutic intervention with a wide time window to ameliorate structural and functional sequelae after stroke.


Asunto(s)
Axones/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Epotilonas/farmacología , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Sistema Nervioso Central/fisiopatología , Modelos Animales de Enfermedad , Mamíferos , Corteza Motora/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Recuperación de la Función/fisiología
8.
Brain ; 142(11): 3636-3654, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31599329

RESUMEN

Accumulating data support the role of tau pathology in cognitive decline in ageing and Alzheimer's disease, but underlying mechanisms remain ill-defined. Interestingly, ageing and Alzheimer's disease have been associated with an abnormal upregulation of adenosine A2A receptor (A2AR), a fine tuner of synaptic plasticity. However, the link between A2AR signalling and tau pathology has remained largely unexplored. In the present study, we report for the first time a significant upregulation of A2AR in patients suffering from frontotemporal lobar degeneration with the MAPT P301L mutation. To model these alterations, we induced neuronal A2AR upregulation in a tauopathy mouse model (THY-Tau22) using a new conditional strain allowing forebrain overexpression of the receptor. We found that neuronal A2AR upregulation increases tau hyperphosphorylation, potentiating the onset of tau-induced memory deficits. This detrimental effect was linked to a singular microglial signature as revealed by RNA sequencing analysis. In particular, we found that A2AR overexpression in THY-Tau22 mice led to the hippocampal upregulation of C1q complement protein-also observed in patients with frontotemporal lobar degeneration-and correlated with the loss of glutamatergic synapses, likely underlying the observed memory deficits. These data reveal a key impact of overactive neuronal A2AR in the onset of synaptic loss in tauopathies, paving the way for new therapeutic approaches.


Asunto(s)
Complemento C1q/metabolismo , Neuronas/metabolismo , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2A/metabolismo , Sinapsis/patología , Tauopatías/genética , Tauopatías/patología , Animales , Autopsia , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Trastornos de la Memoria/etiología , Trastornos de la Memoria/psicología , Ratones , Ratones Transgénicos , Mutación , Aprendizaje Espacial , Tauopatías/psicología , Proteínas tau/genética
9.
Glia ; 67(10): 1859-1872, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31231866

RESUMEN

Microglia mediated responses to neuronal damage in the form of neuroinflammation is a common thread propagating neuropathology. In this study, we investigated the microglial alterations occurring as a result of sphingosine 1-phosphate (S1P) accumulation in neural cells. We evidenced increased microglial activation in the brains of neural S1P-lyase (SGPL1) ablated mice (SGPL1fl/fl/Nes ) as shown by an activated and deramified morphology and increased activation markers on microglia. In addition, an increase of pro-inflammatory cytokines in sorted and primary cultured microglia generated from SGPL1 deficient mice was noticed. Further, we assessed autophagy, one of the major mechanisms in the brain that keeps inflammation in check. Indeed, microglial inflammation was accompanied by defective microglial autophagy in SGPL1 ablated mice. Rescuing autophagy by treatment with rapamycin was sufficient to decrease interleukin 6 (IL-6) but not tumor necrosis factor (TNF) secretion in cultured microglia. Rapamycin mediated decrease of IL-6 secretion suggests a particular mechanistic target of rapamycin (mTOR)-IL-6 link and appeared to be microglia specific. Using pharmacological inhibitors of the major receptors of S1P expressed in the microglia, we identified S1P receptor 2 (S1PR2) as the mediator of both impaired autophagy and proinflammatory effects. In line with these results, the addition of exogenous S1P to BV2 microglial cells showed similar effects as those observed in the genetic knock out of SGPL1 in the neural cells. In summary, we show a novel role of the S1P-S1PR2 axis in the microglia of mice with neural-targeted SGPL1 ablation and in BV2 microglial cell line exogenously treated with S1P.


Asunto(s)
Aldehído-Liasas/metabolismo , Autofagia/fisiología , Inflamación/metabolismo , Microglía/metabolismo , Aldehído-Liasas/antagonistas & inhibidores , Aldehído-Liasas/genética , Animales , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Inflamación/patología , Interleucina-6/metabolismo , Ratones Transgénicos , Microglía/patología , Receptores de Esfingosina-1-Fosfato/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
EMBO Mol Med ; 11(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30617153

RESUMEN

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


Asunto(s)
Enfermedad de Alzheimer/patología , Astrocitos/patología , Proliferación Celular , Factor de Transcripción STAT3/análisis , Animales , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Noqueados , Factor de Transcripción STAT3/deficiencia
11.
Mol Psychiatry ; 24(1): 108-125, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29934546

RESUMEN

Extracellular aggregates of amyloid ß (Aß) peptides, which are characteristic of Alzheimer's disease (AD), act as an essential trigger for glial cell activation and the release of ATP, leading to the stimulation of purinergic receptors, especially the P2X7 receptor (P2X7R). However, the involvement of P2X7R in the development of AD is still ill-defined regarding the dual properties of this receptor. Particularly, P2X7R activates the NLRP3 inflammasome leading to the release of the pro-inflammatory cytokine, IL-1ß; however, P2X7R also induces cleavage of the amyloid precursor protein generating Aß peptides or the neuroprotective fragment sAPPα. We thus explored in detail the functions of P2X7R in AD transgenic mice. Here, we show that P2X7R deficiency reduced Aß lesions, rescued cognitive deficits and improved synaptic plasticity in AD mice. However, the lack of P2X7R did not significantly affect the release of IL-1ß or the levels of non-amyloidogenic fragment, sAPPα, in AD mice. Instead, our results show that P2X7R plays a critical role in Aß peptide-mediated release of chemokines, particularly CCL3, which is associated with pathogenic CD8+ T cell recruitment. In conclusion, our study highlights a novel detrimental function of P2X7R in chemokine release and supports the notion that P2X7R may be a promising therapeutic target for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
12.
Glia ; 66(10): 2246-2261, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30277599

RESUMEN

Chemokines are important signaling molecules in the immune and nervous system. Using a fluorescence reporter mouse model, we demonstrate that the chemokine CCL17, a ligand of the chemokine receptor CCR4, is produced in the murine brain, particularly in a subset of hippocampal CA1 neurons. We found that basal expression of Ccl17 in hippocampal neurons was strongly enhanced by peripheral challenge with lipopolysaccharide (LPS). LPS-mediated induction of Ccl17 in the hippocampus was dependent on local tumor necrosis factor (TNF) signaling, whereas upregulation of Ccl22 required granulocyte-macrophage colony-stimulating factor (GM-CSF). CCL17 deficiency resulted in a diminished microglia density under homeostatic and inflammatory conditions. Further, microglia from naïve Ccl17-deficient mice possessed a reduced cellular volume and a more polarized process tree as assessed by computer-assisted imaging analysis. Regarding the overall branching, cell surface area, and total tree length, the morphology of microglia from naïve Ccl17-deficient mice resembled that of microglia from wild-type mice after LPS stimulation. In line, electrophysiological recordings indicated that CCL17 downmodulates basal synaptic transmission at CA3-CA1 Schaffer collaterals in acute slices from naïve but not LPS-treated animals. Taken together, our data identify CCL17 as a homeostatic and inducible neuromodulatory chemokine affecting the presence and morphology of microglia and synaptic transmission in the hippocampus.


Asunto(s)
Quimiocina CCL17/metabolismo , Hipocampo/inmunología , Neuroinmunomodulación/fisiología , Neuronas/inmunología , Animales , Quimiocina CCL17/genética , Quimiocina CCL22/metabolismo , Femenino , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/patología , Homeostasis/fisiología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/inmunología , Microglía/patología , Monocitos/inmunología , Monocitos/patología , Neuronas/patología , Receptores CCR4/metabolismo , Transmisión Sináptica/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
13.
Front Mol Neurosci ; 11: 235, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30050407

RESUMEN

Consumption of caffeine, a non-selective adenosine A2A receptor (A2AR) antagonist, reduces the risk of developing Alzheimer's disease (AD) and mitigates both amyloid and Tau lesions in transgenic mouse models of the disease. While short-term treatment with A2AR antagonists have been shown to alleviate cognitive deficits in mouse models of amyloidogenesis, impact of a chronic and long-term treatment on the development of amyloid burden, associated neuroinflammation and memory deficits has never been assessed. In the present study, we have evaluated the effect of a 6-month treatment of APPsw/PS1dE9 mice with the potent and selective A2AR antagonist MSX-3 from 3 to 9-10 months of age. At completion of the treatment, we found that the MSX-3 treatment prevented the development of memory deficits in APP/PS1dE9 mice, without significantly altering hippocampal and cortical gene expressions. Interestingly, MSX-3 treatment led to a significant decrease of Aß1-42 levels in the cortex of APP/PS1dE9 animals, while Aß1-40 increased, thereby strongly affecting the Aß1-42/Aß1-40 ratio. Together, these data support the idea that A2AR blockade is of therapeutic value for AD.

14.
J Alzheimers Dis ; 64(2): 379-392, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29865078

RESUMEN

It is widely accepted that the endocannabinoid system (ECS) is a modulator of neuroinflammation associated with neurodegenerative disorders, including Alzheimer's disease (AD). Thus, expression of the cannabinoid receptor 2 (CB2) is induced in plaque-associated microglia and astrocytes in brain tissues from AD patients and in genetic mouse models expressing pathogenic variants of the amyloid precursor protein (APP). However, the exact mechanism of CB2 signaling in this mouse model remains elusive, because the genetic deletion of CB2 and the pharmacological activation of CB2 both reduced neuroinflammation. Here, we demonstrate that CB2 deletion also improved cognitive and learning deficits in APP/PS1*CB2-/- mice. This was accompanied by reduced neuronal loss and decreased plaque levels and coincided with increased expression of Aß degrading enzymes. Interestingly, plaque-associated microglia in APP/PS1*CB2-/- mice showed a less activated morphology, while plaques were smaller and more condensed than in APP/PS1 mice. Taken together, these results indicate a beneficial effect of CB2-deficiency in APP transgenic mice. CB2 appears to be part of a protective system that may be detrimental when engaged continuously.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Regulación de la Expresión Génica/genética , Placa Amiloide/etiología , Receptor Cannabinoide CB2/deficiencia , Factores de Edad , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Modelos Animales de Enfermedad , Endocannabinoides/metabolismo , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Placa Amiloide/patología , Presenilina-1/genética , Receptor Cannabinoide CB2/genética , Transducción de Señal/fisiología
15.
J Exp Med ; 215(6): 1649-1663, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29724785

RESUMEN

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


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Cognición , Red Nerviosa/fisiopatología , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Receptores Purinérgicos P2Y1/metabolismo , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Adenosina Difosfato/uso terapéutico , Enfermedad de Alzheimer/patología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Hipocampo/patología , Humanos , Memoria/efectos de los fármacos , Ratones Transgénicos , Red Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Placa Amiloide/metabolismo , Antagonistas del Receptor Purinérgico P2Y/farmacología , Transducción de Señal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
16.
Glia ; 66(7): 1464-1480, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29493017

RESUMEN

Microglia, the central nervous system resident innate immune cells, cluster around Aß plaques in Alzheimer's disease (AD). The activation phenotype of these plaque-associated microglial cells, and their differences to microglia distant to Aß plaques, are incompletely understood. We used novel three-dimensional cell analysis software to comprehensively analyze the morphological properties of microglia in the TgCRND8 mouse model of AD in spatial relation to Aß plaques. We found strong morphological changes exclusively in plaque-associated microglia, whereas plaque-distant microglia showed only minor changes. In addition, patch-clamp recordings of microglia in acute cerebral slices of TgCRND8 mice revealed increased K+ currents in plaque-associated but not plaque-distant microglia. Within the subgroup of plaque-associated microglia, two different current profiles were detected. One subset of cells displayed only increased inward currents, while a second subset showed both increased inward and outward currents, implicating that the plaque microenvironment differentially impacts microglial ion channel expression. Using pharmacological channel blockers, multiplex single-cell PCR analysis and RNA fluorescence in situ hybridization, we identified Kir and Kv channel types contributing to the in- and outward K+ conductance in plaque-associated microglia. In summary, we have identified a previously unrecognized level of morphological and electrophysiological heterogeneity of microglia in relation to amyloid plaques, suggesting that microglia may display multiple activation states in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Microglía/patología , Microglía/fisiología , Placa Amiloide/patología , Placa Amiloide/fisiopatología , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Cationes Monovalentes/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Potasio/metabolismo , Canales de Potasio/metabolismo , Técnicas de Cultivo de Tejidos
17.
Lancet Neurol ; 14(4): 388-405, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25792098

RESUMEN

Increasing evidence suggests that Alzheimer's disease pathogenesis is not restricted to the neuronal compartment, but includes strong interactions with immunological mechanisms in the brain. Misfolded and aggregated proteins bind to pattern recognition receptors on microglia and astroglia, and trigger an innate immune response characterised by release of inflammatory mediators, which contribute to disease progression and severity. Genome-wide analysis suggests that several genes that increase the risk for sporadic Alzheimer's disease encode factors that regulate glial clearance of misfolded proteins and the inflammatory reaction. External factors, including systemic inflammation and obesity, are likely to interfere with immunological processes of the brain and further promote disease progression. Modulation of risk factors and targeting of these immune mechanisms could lead to future therapeutic or preventive strategies for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Antiinflamatorios no Esteroideos/uso terapéutico , Lesiones Encefálicas/complicaciones , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Microglía/inmunología , Microglía/patología , Obesidad/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Animales , Astrocitos/inmunología , Astrocitos/patología , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Lesiones Encefálicas/metabolismo , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inmunización , Inflamación/diagnóstico , Inflamación/inmunología , Mediadores de Inflamación/inmunología , Locus Coeruleus/patología , Nootrópicos/administración & dosificación , Obesidad/metabolismo , Fagocitosis , Pliegue de Proteína , Factores de Riesgo , Índice de Severidad de la Enfermedad
18.
Methods Mol Biol ; 1040: 1-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23852592

RESUMEN

Senile plaques are an important histological hallmark of Alzheimer's disease. They mainly consist of the fibrillar peptide ß-amyloid (Aß) and are surrounded by activated microglia and astrocytes. Microglia in the vicinity of senile plaques express high levels of proinflammatory cytokines and neurotoxic substances, which are believed to influence disease progression. One important cytokine in Alzheimer's disease is IL-1ß. Stimulation of cultured primary microglia by synthetic fibrillar Aß causes the release of IL-1ß via activation of the NLRP3 inflammasome.Here we provide protocols for the preparation of primary microglial cultures and synthetic oligomeric and fibrillar forms of Aß.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Péptidos beta-Amiloides/aislamiento & purificación , Animales , Separación Celular/métodos , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-1beta/biosíntesis , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Cultivo Primario de Células/métodos
19.
PLoS One ; 8(4): e60921, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23577177

RESUMEN

Microglial cells closely interact with senile plaques in Alzheimer's disease and acquire the morphological appearance of an activated phenotype. The significance of this microglial phenotype and the impact of microglia for disease progression have remained controversial. To uncover and characterize putative changes in the functionality of microglia during Alzheimer's disease, we directly assessed microglial behavior in two mouse models of Alzheimer's disease. Using in vivo two-photon microscopy and acute brain slice preparations, we found that important microglial functions - directed process motility and phagocytic activity - were strongly impaired in mice with Alzheimer's disease-like pathology compared to age-matched non-transgenic animals. Notably, impairment of microglial function temporally and spatially correlated with Aß plaque deposition, and phagocytic capacity of microglia could be restored by interventionally decreasing amyloid burden by Aß vaccination. These data suggest that major microglial functions progressively decline in Alzheimer's disease with the appearance of Aß plaques, and that this functional impairment is reversible by lowering Aß burden, e.g. by means of Aß vaccination.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Microglía/patología , Placa Amiloide/patología , Multimerización de Proteína , Péptidos beta-Amiloides/genética , Animales , Movimiento Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación , Fagocitosis , Estructura Secundaria de Proteína , Análisis Espacio-Temporal
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA