Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Brain Behav Immun ; 99: 363-382, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34343617

RESUMEN

Alzheimer's disease (AD) pathology is characterized by amyloid-ß (Aß) deposition and tau hyper-phosphorylation, accompanied by a progressive cognitive decline. Monocytes have been recently shown to play a major role in modulating Aß pathology, and thereby have been pointed as potential therapeutic targets. However, the main challenge remains in identifying clinically relevant interventions that could modulate monocyte immune functions in absence of undesired off-target effects. Erythropoietin (EPO), a key regulator of erythrocyte production, has been shown to possess immunomodulatory potential and to provide beneficial effects in preclinical models of AD. However, the transition to use recombinant human EPO in clinical trials was hindered by unwanted erythropoietic effects that could lead to thrombosis. Here, we used a recently identified non-erythropoietic analogue of EPO, ARA 290, to evaluate its therapeutic potential in AD therapy. We first evaluated the effects of early systemic ARA 290 administration on AD-like pathology in an early-onset model, represented by young APP/PS1 mice. Our findings indicate that ARA 290 early treatment decelerated Aß pathology progression in APP/PS1 mice while improving cognitive functions. ARA 290 potently increased the levels of total monocytes by specifically stimulating the generation of Ly6CLow patrolling subset, which are implicated in clearing Aß from the cerebral vasculature, and subsequently reducing overall Aß burden in the brain. Moreover, ARA 290 increased the levels of monocyte progenitors in the bone marrow. Using chimeric APP/PS1 mice in which Ly6CLow patrolling subset are selectively depleted, ARA 290 was inefficient in attenuating Aß pathology and ameliorating cognitive functions in young animals. Interestingly, ARA 290 effects were compromised when delivered in a late-onset model, represented by aged APP1/PS1. In aged APP/PS1 mice in which AD-like pathology is at advanced stages, ARA 290 failed to reverse Aß pathology and to increase the levels of circulating monocytes. Our study suggests that ARA 290 early systemic treatment could prevent AD-like progression via modulation of monocyte functions by specifically increasing the ratio of patrolling monocytes.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Precursor de Proteína beta-Amiloide , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Monocitos/patología , Presenilina-1
2.
Proc Natl Acad Sci U S A ; 110(5): 1941-6, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23322736

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia worldwide. The pathogenesis of this neurodegenerative disease, currently without curative treatment, is associated with the accumulation of amyloid ß (Aß) in brain parenchyma and cerebral vasculature. AD patients are unable to clear this toxic peptide, leading to Aß accumulation in their brains and, presumably, the pathology associated with this devastating disease. Compounds that stimulate the immune system to clear Aß may therefore have great therapeutic potential in AD patients. Monophosphoryl lipid A (MPL) is an LPS-derived Toll-like receptor 4 agonist that exhibits unique immunomodulatory properties at doses that are nonpyrogenic. We show here that repeated systemic injections of MPL, but not LPS, significantly improved AD-related pathology in APP(swe)/PS1 mice. MPL treatment led to a significant reduction in Aß load in the brain of these mice, as well as enhanced cognitive function. MPL induced a potent phagocytic response by microglia while triggering a moderate inflammatory reaction. Our data suggest that the Toll-like receptor 4 agonist MPL may be a treatment for AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Encéfalo/efectos de los fármacos , Lípido A/análogos & derivados , Receptor Toll-Like 4/agonistas , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Inmunidad Innata/efectos de los fármacos , Ligandos , Lípido A/administración & dosificación , Lípido A/uso terapéutico , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Microscopía Fluorescente , Fagocitosis/efectos de los fármacos , Presenilina-1/genética , Presenilina-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor Toll-Like 4/metabolismo
3.
Int J Mol Sci ; 15(4): 6453-74, 2014 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-24743889

RESUMEN

Neurons are extremely vulnerable cells that tightly rely on the brain's highly dynamic and complex vascular network that assures an accurate and adequate distribution of nutrients and oxygen. The neurovascular unit (NVU) couples neuronal activity to vascular function, controls brain homeostasis, and maintains an optimal brain microenvironment adequate for neuronal survival by adjusting blood-brain barrier (BBB) parameters based on brain needs. The NVU is a heterogeneous structure constituted by different cell types that includes pericytes. Pericytes are localized at the abluminal side of brain microvessels and contribute to NVU function. Pericytes play essential roles in the development and maturation of the neurovascular system during embryogenesis and stability during adulthood. Initially, pericytes were described as contractile cells involved in controlling neurovascular tone. However, recent reports have shown that pericytes dynamically respond to stress induced by injury upon brain diseases, by chemically and physically communicating with neighboring cells, by their immune properties and by their potential pluripotent nature within the neurovascular niche. As such, in this paper, we would like to review the role of pericytes in NVU remodeling, and their potential as targets for NVU repair strategies and consequently neuroprotection in two pathophysiologically distinct brain disorders: ischemic stroke and Alzheimer's disease (AD).


Asunto(s)
Encefalopatías/metabolismo , Pericitos/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/metabolismo , Encefalopatías/patología , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Neuronas/metabolismo , Pericitos/citología , Transducción de Señal
4.
Cell Rep ; 20(8): 1830-1843, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28834747

RESUMEN

The signals that regulate the fate of circulating monocytes remain unknown. In the present study, we demonstrate that triggering of the NOD2 receptor by muramyl dipeptide (MDP) converts inflammatory Ly6Chigh monocytes into patrolling Ly6Clow monocytes. Administration of MDP to Nr4a1-/- mice, which lack Ly6Clow monocytes, or to Ly6Clow-depleted mice led to the emergence of blood-patrolling monocytes with a profile similar to that of Ly6Clow monocytes, including high expression of CX3CR1 and LFA1. Using intravital microscopy in animal models of inflammatory diseases, we also found that converted Ly6Chigh monocytes patrol the endothelium of blood vessels and that their presence contributes to a reduction in the inflammatory response following MDP injection. Our results demonstrate that NOD2 contributes to the regulation of blood monocytes and suggest that it could be therapeutically targeted to treat inflammatory diseases.


Asunto(s)
Antígenos Ly/inmunología , Monocitos/inmunología , Proteína Adaptadora de Señalización NOD2/inmunología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Adaptadora de Señalización NOD2/genética
5.
Curr Biol ; 26(16): R772-5, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27554659

RESUMEN

Growing evidence highlights the crucial physiological functions of microglia that rely on their phagocytic activities, which can be compromised with age. A new study reports the impaired clearance of myelin debris by microglia in the brain, leading to insoluble lysosomal inclusions and contributing to the immune dysfunction and senescence of these cells.


Asunto(s)
Microglía , Vaina de Mielina , Encéfalo , Humanos , Fagocitos , Fagocitosis
6.
Oncotarget ; 7(42): 67808-67827, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27661129

RESUMEN

Alzheimer's disease (AD) is mainly characterized by the accumulation and aggregation of amyloid-ß (Aß) peptides in brain parenchyma and cerebral microvasculature. Unfortunately, the exact causes of the disease are still unclear. However, blood-brain barrier (BBB) dysfunction and activation of inflammatory pathways are implicated in AD pathogenesis. Importantly, advanced age and high fat diet, two major risk factors associated with AD, were shown to deeply affect BBB function and modulate the immune response. As such, this study evaluated the impact of age and high fat diet on AD progression. For this purpose, 3 (i.e. young) and 12 (i.e. aged) months old APPswe/PS1 mice were fed for 4 months with a high fat diet (i.e. Western diet (WD)) or normal diet. Interestingly, neurobehavioral tests revealed that WD accelerates age-associated cognitive decline without affecting parenchymal Aß. Nonetheless, WD decreases matrix metalloproteinase-9 enzymatic activity and brain-derived neurotrophic factor mRNA and protein levels in brain, suggesting loss of synaptic plasticity. In the periphery, WD promotes systemic inflammation by increasing the levels of blood-circulating monocytes and monocyte chemotactic protein-1 production, which is accompanied by an augmentation of oxidized-low density lipoprotein levels in blood circulation. At the BBB, WD potentiates the age-induced increase of Aß 1-40 accumulation and exacerbates the oxidative stress, specifically in cerebral microvasculature. These effects were accompanied by the dysfunction of pericytes, thus altering BBB functionality without compromising its integrity. Our study provides new insights into the implication of high fat diet in accelerating the cognitive decline in AD.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Quimiocina CCL2/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Estrés Oxidativo , Fragmentos de Péptidos/metabolismo
7.
Oncotarget ; 7(24): 35552-35561, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27248662

RESUMEN

Stroke is associated with neuroinflammation, neuronal loss and blood-brain barrier (BBB) breakdown. Thus far, recombinant tissue-type plasminogen activator (rtPA), the only approved treatment for acute ischemic stroke, increases the risk of intracerebral hemorrhage and is poorly efficient in disaggregating platelet-rich thrombi. Therefore, the development of safer and more efficient therapies is highly awaited. Encouraging neuroprotective effects were reported in mouse models of ischemic stroke following administration of erythropoietin (EPO). However, previous preclinical studies did not investigate the effects of EPO in focal ischemic stroke induced by a platelet-rich thrombus and did not consider the implication of age. Here, we performed middle cerebral artery occlusion by inducing platelet-rich thrombus formation in chimeric 5- (i.e. young) and 20- (i.e. aged) months old C57BL/6 mice, in which hematopoietic stem cells carried the green fluorescent protein (GFP)-tag. Recombinant human EPO (rhEPO) was administered 24 hours post-occlusion and blood-circulating monocyte populations were studied by flow cytometry 3 hours post-rhEPO administration. Twenty-four hours following rhEPO treatment, neuronal loss and BBB integrity were assessed by quantification of Fluoro-Jade B (FJB)-positive cells and extravasated serum immunoglobulins G (IgG), respectively. Neuroinflammation was determined by quantifying infiltration of GFP-positive bone marrow-derived cells (BMDC) and recruitment of microglial cells into brain parenchyma, along with monocyte chemotactic protein-1 (MCP-1) brain protein levels. Here, rhEPO anti-inflammatory properties rescued ischemic injury by reducing neuronal loss and BBB breakdown in young animals, but not in aged littermates. Such age-dependent effects of rhEPO must therefore be taken into consideration in future studies aiming to develop new therapies for ischemic stroke.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Encéfalo/metabolismo , Eritropoyetina/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Factores de Edad , Animales , Barrera Hematoencefálica/patología , Encéfalo/patología , Isquemia Encefálica/complicaciones , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Eritropoyetina/efectos adversos , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Humanos , Inflamación/tratamiento farmacológico , Inflamación/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Arteria Cerebral Media/patología , Fármacos Neuroprotectores/efectos adversos , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Accidente Cerebrovascular/complicaciones , Trombosis/inducido químicamente
8.
Neuropsychopharmacology ; 41(5): 1297-307, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26349911

RESUMEN

Alzheimer's disease (AD) is the leading cause of dementia among elderly population. AD is characterized by the accumulation of beta-amyloid (Aß) peptides, which aggregate over time to form amyloid plaques in the brain. Reducing soluble Aß levels and consequently amyloid plaques constitute an attractive therapeutic avenue to, at least, stabilize AD pathogenesis. The brain possesses several mechanisms involved in controlling cerebral Aß levels, among which are the tissue-plasminogen activator (t-PA)/plasmin system and microglia. However, these mechanisms are impaired and ineffective in AD. Here we show that the systemic chronic administration of recombinant t-PA (Activase rt-PA) attenuates AD-related pathology in APPswe/PS1 transgenic mice by reducing cerebral Aß levels and improving the cognitive function of treated mice. Interestingly, these effects do not appear to be mediated by rt-PA-induced plasmin and matrix metalloproteinases 2/9 activation. We observed that rt-PA essentially mediated a slight transient increase in the frequency of patrolling monocytes in the circulation and stimulated microglia in the brain to adopt a neuroprotective phenotype, both of which contribute to Aß elimination. Our study unraveled a new role of rt-PA in maintaining the phagocytic capacity of microglia without exacerbating the inflammatory response and therefore might constitute an interesting approach to stimulate the key populations of cells involved in Aß clearance from the brain.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Microglía/efectos de los fármacos , Monocitos/efectos de los fármacos , Activador de Tejido Plasminógeno/administración & dosificación , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/genética , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Microglía/fisiología , Monocitos/fisiología , Estrés Oxidativo/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Presenilina-1/genética
9.
Alzheimers Res Ther ; 7(1): 41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25878730

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder affecting older people worldwide. It is a progressive disorder mainly characterized by the presence of amyloid-beta (Aß) plaques and neurofibrillary tangles within the brain parenchyma. It is now well accepted that neuroinflammation constitutes an important feature in AD, wherein the exact role of innate immunity remains unclear. Although innate immune cells are at the forefront to protect the brain in the presence of toxic molecules including Aß, this natural defense mechanism seems insufficient in AD patients. Monocytes are a key component of the innate immune system and they play multiple roles, such as the removal of debris and dead cells via phagocytosis. These cells respond quickly and mobilize toward the inflamed site, where they proliferate and differentiate into macrophages in response to inflammatory signals. Many studies have underlined the ability of circulating and infiltrating monocytes to clear vascular Aß microaggregates and parenchymal Aß deposits respectively, which are very important features of AD. On the other hand, microglia are the resident immune cells of the brain and they play multiple physiological roles, including maintenance of the brain's microenvironment homeostasis. In the injured brain, activated microglia migrate to the inflamed site, where they remove neurotoxic elements by phagocytosis. However, aged resident microglia are less efficient than their circulating sister immune cells in eliminating Aß deposits from the brain parenchyma, thus underlining the importance to further investigate the functions of these innate immune cells in AD. The present review summarizes current knowledge on the role of monocytes and microglia in AD and how these cells can be mobilized to prevent and treat the disease.

10.
Acta Neuropathol Commun ; 1: 75, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24252187

RESUMEN

BACKGROUND: The Blood-brain barrier (BBB) controls brain supply with oxygen and nutrients, and protects the brain from toxic metabolites, such as beta-amyloid (Aß) peptides. The neurovascular unit (NVU) couples vascular and neuronal functions by controlling BBB parameters based on brain needs. As such, NVU/BBB dysfunction, associated to irregularities in cerebral blood flow (CBF), has been proposed to contribute in the pathogenesis of Alzheimer's disease (AD), mainly by impairing cerebral Aß clearance. However, the spatiotemporal contribution of the NVU/BBB in the neurodegenerative cascades remains elusive. RESULTS: By using C57BL/6J mice subjected to right common carotid artery (rCCA) permanent ligation in order to induce mild chronic cerebral hypoperfusion, we show here that cerebral hypoperfusion induced NVU dysfunction by reducing ABCB1 protein expression in brain capillaries. ABCB1 reduction was mainly triggered by an enhanced Glycogen Synthase Kinase 3 (GSK3ß) activation, which decreased ß-catenin nuclear abundance. Moreover, cerebral hypoperfusion triggered early vascular deposition of peripherally applied human Aß1-42 peptides, which has shifted from highly vascular to the parenchyma 6 weeks later, forming small stable Aß deposits. Hypoperfusion induced a deregulation in glucose metabolism, as brain reperfusion, or the administration of a high dose of glucose, diminished GSK3ß activation, recuperated ß-catenin nuclear abundance, reestablished ABCB1 protein expression, and prevented Aß vascular early deposition. These results demonstrate that mild chronic cerebral hypoperfusion creates a metabolically deregulated microenvironment, thus triggering the brain entry and aggregation of peripherally applied human Aß1-42 peptides. CONCLUSION: Our study offers new insights on the initiation of the neurodegenerative cascades observed in AD, which could be valuable in developing adequate treatment strategies.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/fisiopatología , Isquemia Encefálica/fisiopatología , Encéfalo/fisiopatología , Fragmentos de Péptidos/metabolismo , Animales , Encéfalo/irrigación sanguínea , Capilares/fisiopatología , Enfermedades de las Arterias Carótidas , Circulación Cerebrovascular/fisiología , Enfermedad Crónica , Modelos Animales de Enfermedad , Glucosa/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones Endogámicos C57BL
11.
Behav Brain Res ; 234(2): 334-42, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22796601

RESUMEN

3xTg-AD mutant mice are characterized by parenchymal Aß plaques and neurofibrillary tangles resembling those found in patients with Alzheimer's disease. The mutants were compared with non-transgenic controls in sensorimotor and learning tests. 3xTg-AD mutants were deficient in T-maze reversal, object recognition, and passive avoidance learning. In addition, the mutants showed hypoactivity in two open-field tests, fewer fecal boli in an observation jar, and reduced enclosed arm entries and head-dipping in the elevated plus-maze. On the contrary, the mutants did not differ from controls in pain thresholds, nest-building, and various reflexes determined by the SHIRPA primary screen and were even better on the rotorod test of motor coordination.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Trastornos del Movimiento/etiología , Mutación/genética , Adaptación Fisiológica/genética , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Reacción de Prevención , Discriminación en Psicología , Conducta Exploratoria/fisiología , Humanos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos del Movimiento/genética , Nocicepción/fisiología , Umbral del Dolor/fisiología , Presenilina-1/genética , Desempeño Psicomotor/fisiología , Reconocimiento en Psicología/fisiología , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA