Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 5651, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-37024553

RESUMEN

Generalized status epilepticus (SE) triggers a robust neuroinflammatory response involving reactive astrocytosis, activation of brain-resident microglia, and brain infiltration of CCR2+ monocytes. Multiple lines of evidence indicate that quenching SE-induced neuroinflammation can alleviate the adverse consequences of SE, including neuronal damage and cognitive impairments. Our recent findings show that blocking monocyte brain entry after SE, via global Ccr2 KO, rescues several SE-induced adverse effects including blood-brain barrier (BBB) erosion, microgliosis and neuronal damage while enhancing weight regain. The goals of the present study were to determine if CCR2 antagonism with a small molecule after SE replicates the effects of the CCR2 knockout. Male Ccr2+/rfp heterozygous mice were subject to intraperitoneal injection of kainic acid, scored for seizure severity, weight recovery, and nest building capability. Surviving mice were randomized into CCR2 antagonist and vehicle groups. The CCR2 antagonist, or vehicle, was administered 24- and 48-h post-SE via oral gavage, and mice were sacrificed three days post-SE. Mice subject to the CCR2 antagonist displayed faster weight recovery between one- and three-days post-SE and modestly enhanced ability to build a nest on the third day after SE when compared to vehicle-treated controls. CCR2 antagonism limited monocyte recruitment to the hippocampus and reduced numbers of Iba1+ macrophages. The mRNA levels of inflammatory mediators were depressed by 47%, and glial markers were reduced by 30% in mice treated with the CCR2 antagonist compared to controls. Astrocytosis was reduced in four brain regions. Neuroprotection was observed in the hippocampus, and erosion of the BBB was lessened in mice subject to the antagonist. Our findings provide proof-of-concept that brief CCR2 antagonism beginning one day after SE can alleviate multiple adverse SE-induced effects, including functional impairment, and identify circulating CCR2+ monocytes as a viable therapeutic target.


Asunto(s)
Gliosis , Estado Epiléptico , Ratones , Masculino , Animales , Gliosis/tratamiento farmacológico , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Monocitos/fisiología , Macrófagos , Convulsiones , Inflamación , Receptores de Quimiocina , Receptores CCR2/genética , Ratones Endogámicos C57BL
2.
Neuropharmacology ; 224: 109356, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36460083

RESUMEN

Cognitive comorbidities can substantially reduce quality of life in people with epilepsy. Inflammation is a component of all chronic diseases including epilepsy, as well as acute events like status epilepticus (SE). Neuroinflammation is the consequence of several broad signaling cascades including cyclooxygenase-2 (COX-2)-associated pathways. Activation of the EP2 receptor for prostaglandin E2 appears responsible for blood-brain barrier leakage and much of the inflammatory reaction, neuronal injury and cognitive deficit that follows seizure-provoked COX-2 induction in brain. Here we show that brief exposure of mice to TG11-77, a potent, selective, orally available and brain permeant EP2 antagonist, eliminates the profound cognitive deficit in Y-maze performance after SE and reduces delayed mortality and microgliosis, with a minimum effective i.p. dose (as free base) of 8.8 mg/kg. All in vitro studies required to submit an investigational new drug (IND) application for TG11-77 have been completed, and non-GLP dose range-finding toxicology in the rat identified no overt, organ or histopathology signs of toxicity after 7 days of oral administration at 1000 mg/kg/day. Plasma exposure in the rat was dose-linear between 15 and 1000 mg/kg dosing. TG11-77 thus appears poised to continue development towards the initial clinical test of the hypothesis that EP2 receptor modulation after SE can provide the first preventive treatment for one of the chief comorbidities of epilepsy.


Asunto(s)
Epilepsia , Estado Epiléptico , Ratas , Ratones , Animales , Ciclooxigenasa 2/metabolismo , Calidad de Vida , Subtipo EP2 de Receptores de Prostaglandina E , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/metabolismo , Inflamación , Cognición
3.
Front Immunol ; 13: 1056417, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36618392

RESUMEN

Introduction: Progranulin (PGRN) is a secreted glycoprotein, the expression of which is linked to several neurodegenerative diseases. Although its specific function is still unclear, several studies have linked it with lysosomal functions and immune system regulation. Here, we have explored the role of PGRN in peripheral and central immune system homeostasis by investigating the consequences of PGRN deficiency on adaptive and innate immune cell populations. Methods: First, we used gene co-expression network analysis of published data to test the hypothesis that Grn has a critical role in regulating the activation status of immune cell populations in both central and peripheral compartments. To investigate the extent to which PGRN-deficiency resulted in immune dysregulation, we performed deep immunophenotyping by flow cytometry of 19-24-month old male and female Grn-deficient mice (PGRN KO) and littermate Grn-sufficient controls (WT). Results: Male PGRN KO mice exhibited a lower abundance of microglial cells with higher MHC-II expression, increased CD44 expression on monocytes in the brain, and more CNS-associated CD8+ T cells compared to WT mice. Furthermore, we observed an increase in CD44 on CD8+ T cells in the peripheral blood. Female PGRN KO mice also had fewer microglia compared to WT mice, and we also observed reduced expression of MHC-II on brain monocytes. Additionally, we found an increase in Ly-6Chigh monocyte frequency and decreased CD44 expression on CD8+ and CD4+ T cells in PGRN KO female blood. Given that Gpnmb, which encodes for the lysosomal protein Glycoprotein non-metastatic melanoma protein B, has been reported to be upregulated in PGRN KO mice, we investigated changes in GPNMB protein expression associated with PGRN deficits and found that GPNMB is modulated in myeloid cells in a sex-specific manner. Discussion: Our data suggest that PGRN and GPNMB jointly regulate the peripheral and the central immune system in a sex-specific manner; thus, understanding their associated mechanisms could pave the way for developing new neuroprotective strategies to modulate central and peripheral inflammation to lower risk for neurodegenerative diseases and possibly delay or halt progression.


Asunto(s)
Linfocitos T CD8-positivos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Femenino , Animales , Ratones , Progranulinas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Granulinas , Ratones Noqueados , Sistema Inmunológico
4.
J Neurosci ; 41(5): 1105-1117, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33293358

RESUMEN

A multidimensional inflammatory response ensues after status epilepticus (SE), driven partly by cyclooxygenase-2-mediated activation of prostaglandin EP2 receptors. The inflammatory response is typified by astrocytosis, microgliosis, erosion of the blood-brain barrier (BBB), formation of inflammatory cytokines, and brain infiltration of blood-borne monocytes. Our previous studies have shown that inhibition of monocyte brain invasion or systemic administration of an EP2 receptor antagonist relieves multiple deleterious consequences of SE. Here we identify those effects of EP2 antagonism that are reproduced by conditional ablation of EP2 receptors in immune myeloid cells and show that systemic EP2 antagonism blocks monocyte brain entry in male mice. The induction of hippocampal IL-6 after pilocarpine SE was nearly abolished in EP2 conditional KO mice. Serum albumin levels in the cortex, a measure of BBB breakdown, were significantly higher after SE in EP2-sufficient mice but not in EP2 conditional KOs. EP2 deficiency in innate immune cells accelerated the recovery from sickness behaviors following SE. Surprisingly, neurodegeneration was not alleviated in myeloid conditional KOs. Systemic EP2 antagonism prevented monocyte brain infiltration and provided broader rescue of SE-induced effects than myeloid EP2 ablation, including neuroprotection and broader suppression of inflammatory mediators. Reporter expression indicated that the cellular target of CD11b-driven Cre was circulating myeloid cells but, unexpectedly, not microglia. These findings indicate that activation of EP2 receptors on immune myeloid cells drives substantial deficits in behavior and disrupts the BBB after SE. The benefits of systemic EP2 antagonism can be attributed, in part, to blocking brain recruitment of blood-borne monocytes.SIGNIFICANCE STATEMENT Unabated seizures reduce quality of life, promote the development of epilepsy, and can be fatal. We previously identified activation of prostaglandin EP2 receptors as a driver of undesirable consequences of seizures. However, the relevant EP2-expressing cell types remain unclear. Here we identify peripheral innate immune cells as a driver of the EP2-related negative consequences of seizures. Removal of EP2 from peripheral immune cells was beneficial, abolishing production of a key inflammatory cytokine, accelerating weight regain, and limiting behavioral deficits. These findings provide evidence that EP2 engagement on peripheral immune and brain endothelia contributes to the deleterious effects of SE, and will assist in the development of beneficial therapies to enhance quality of life in individuals who suffer prolonged seizures.


Asunto(s)
Inmunidad Innata/fisiología , Células Mieloides/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/biosíntesis , Estado Epiléptico/metabolismo , Animales , Citometría de Flujo/métodos , Hipocampo/citología , Hipocampo/inmunología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/inmunología , Subtipo EP2 de Receptores de Prostaglandina E/genética , Subtipo EP2 de Receptores de Prostaglandina E/inmunología , Estado Epiléptico/genética , Estado Epiléptico/inmunología
5.
Methods Mol Biol ; 2034: 293-301, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31392694

RESUMEN

During insults and disease blood-borne monocytes can invade brain and spinal cord, contributing to the neuroimmune response together with brain-resident microglia. The specific function of brain-infiltrating monocytes has been difficult to ascertain because of shared marker expression and morphology of these two immune cell types. Here we describe our method of repopulating the brain with circulating monocytes after microglia ablation to investigate the physiology of brain-invading monocytes, which engraft under these conditions.


Asunto(s)
Encefalopatías , Encéfalo , Movimiento Celular/inmunología , Microglía , Monocitos , Neuroinmunomodulación , Animales , Encéfalo/inmunología , Encéfalo/patología , Encefalopatías/inmunología , Encefalopatías/patología , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/patología , Ratones , Microglía/inmunología , Microglía/patología , Monocitos/inmunología , Monocitos/patología , Médula Espinal/inmunología , Médula Espinal/patología , Enfermedades de la Columna Vertebral/inmunología , Enfermedades de la Columna Vertebral/patología
6.
J Alzheimers Dis ; 70(4): 1259-1274, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31322556

RESUMEN

Alzheimer's disease (AD) pathology consists of extracellular deposits of amyloid-ß peptides (Aß) and intracellular neurofibrillary tangles. These pathological alterations are accompanied by a neuroinflammatory response consisting of increased expression of inflammatory mediators. An anti-inflammatory strategy designed to prevent or delay the development of AD would benefit from knowing when neuroinflammation appears in the transgenic models during prodromal disease stages relative to Aß pathology. We investigated the expression patterns of inflammatory mediators in the brain of 5xFAD mice in comparison to development of Aß deposition. Expression changes in inflammatory mediators and glial markers are more robust in female mice starting at three months of age, in contrast to males in which there is no clear trend through five months. Female and male 5xFAD mice also displayed an age-dependent increase in cortical Aß deposition congruent with neuroinflammation. Thus, in the 5xFAD mouse model of AD, administration of an anti-inflammatory agent would be most efficacious when administered before three months of age.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Mediadores de Inflamación/metabolismo , Síntomas Prodrómicos , Caracteres Sexuales , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovillos Neurofibrilares/genética , Ovillos Neurofibrilares/metabolismo
7.
Expert Opin Ther Targets ; 23(1): 1-13, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30484341

RESUMEN

Introduction:A robust neuroinflammatory response is a prevalent feature of multiple neurological disorders, including epilepsy and acute status epilepticus. One component of this neuroinflammatory reaction is the induction of cyclooxygenase-2 (COX-2), synthesis of several prostaglandins and endocannabinoid metabolites, and subsequent activation of prostaglandin and related receptors. Neuroinflammation mediated by COX-2 and its downstream effectors has received considerable attention as a potential target class to ameliorate the deleterious consequences of neurological injury. Areas covered: Here we describe the roles of COX-2 as a major inflammatory mediator. In addition, we discuss the receptors for prostanoids PGE2, prostaglandin D2, and PGF2α as potential therapeutic targets for inflammation-driven diseases. The consequences of prostanoid receptor activation after seizure activity are discussed with an emphasis on the utilization of small molecules to modulate prostanoid receptor activity. Expert opinion: Limited clinical trial experience is supportive but not definitive for a role of the COX signaling cascade in epileptogenesis. The cardiotoxicity associated with chronic coxib use, and the expectation that COX-2 inhibition will influence the levels of endocannabinoids, leukotrienes, and lipoxins as well as the prostaglandins and their endocannabinoid metabolite analogs, is shifting attention toward downstream synthases and receptors that mediate inflammation in the brain.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Epilepsia/fisiopatología , Terapia Molecular Dirigida , Animales , Encéfalo/fisiopatología , Ciclooxigenasa 2/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Inhibidores de la Ciclooxigenasa 2/farmacología , Epilepsia/tratamiento farmacológico , Humanos , Inflamación/fisiopatología , Prostaglandinas/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Proc Natl Acad Sci U S A ; 113(38): E5665-74, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27601660

RESUMEN

The generalized seizures of status epilepticus (SE) trigger a series of molecular and cellular events that produce cognitive deficits and can culminate in the development of epilepsy. Known early events include opening of the blood-brain barrier (BBB) and astrocytosis accompanied by activation of brain microglia. Whereas circulating monocytes do not infiltrate the healthy CNS, monocytes can enter the brain in response to injury and contribute to the immune response. We examined the cellular components of innate immune inflammation in the days following SE by discriminating microglia vs. brain-infiltrating monocytes. Chemokine receptor 2 (CCR2(+)) monocytes invade the hippocampus between 1 and 3 d after SE. In contrast, only an occasional CD3(+) T lymphocyte was encountered 3 d after SE. The initial cellular sources of the chemokine CCL2, a ligand for CCR2, included perivascular macrophages and microglia. The induction of the proinflammatory cytokine IL-1ß was greater in FACS-isolated microglia than in brain-invading monocytes. However, Ccr2 knockout mice displayed greatly reduced monocyte recruitment into brain and reduced levels of the proinflammatory cytokine IL-1ß in hippocampus after SE, which was explained by higher expression of the cytokine in circulating and brain monocytes in wild-type mice. Importantly, preventing monocyte recruitment accelerated weight regain, reduced BBB degradation, and attenuated neuronal damage. Our findings identify brain-infiltrating monocytes as a myeloid-cell subclass that contributes to neuroinflammation and morbidity after SE. Inhibiting brain invasion of CCR2(+) monocytes could represent a viable method for alleviating the deleterious consequences of SE.


Asunto(s)
Quimiocina CCL2/genética , Interleucina-1beta/metabolismo , Monocitos/patología , Receptores CCR2/genética , Estado Epiléptico/inmunología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Quimiocina CCL2/metabolismo , Encefalitis/inmunología , Encefalitis/metabolismo , Encefalitis/patología , Gliosis/inmunología , Gliosis/metabolismo , Gliosis/patología , Inmunidad Innata/genética , Interleucina-1beta/genética , Ratones , Ratones Noqueados , Neuronas/inmunología , Neuronas/patología , Receptores CCR2/metabolismo , Convulsiones/genética , Convulsiones/inmunología , Convulsiones/metabolismo , Convulsiones/patología , Estado Epiléptico/metabolismo , Estado Epiléptico/patología
9.
J Exp Med ; 212(11): 1803-9, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26458770

RESUMEN

Immune cells of myeloid lineage are encountered in the Alzheimer's disease (AD) brain, where they cluster around amyloid-ß plaques. However, assigning functional roles to myeloid cell subtypes has been problematic, and the potential for peripheral myeloid cells to alleviate AD pathology remains unclear. Therefore, we asked whether replacement of brain-resident myeloid cells with peripheral monocytes alters amyloid deposition in two mouse models of cerebral ß-amyloidosis (APP23 and APPPS1). Interestingly, early after repopulation, infiltrating monocytes neither clustered around plaques nor showed Trem2 expression. However, with increasing time in the brain, infiltrating monocytes became plaque associated and also Trem2 positive. Strikingly, however, monocyte repopulation for up to 6 mo did not modify amyloid load in either model, independent of the stage of pathology at the time of repopulation. Our results argue against a long-term role of peripheral monocytes that is sufficiently distinct from microglial function to modify cerebral ß-amyloidosis. Therefore, myeloid replacement by itself is not likely to be effective as a therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Células Mieloides/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Glicoproteínas de Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Monocitos/fisiología , Receptores Inmunológicos/análisis
10.
Annu Rev Pharmacol Toxicol ; 55: 229-47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25196047

RESUMEN

Epilepsy is a prevalent neurological disorder afflicting nearly 50 million people worldwide. The disorder is characterized clinically by recurrent spontaneous seizures attributed to abnormal synchrony of brain neurons. Despite advances in the treatment of epilepsy, nearly one-third of patients are resistant to current therapies, and the underlying mechanisms whereby a healthy brain becomes epileptic remain unresolved. Therefore, researchers have a major impetus to identify and exploit new drug targets. Here we distinguish between epileptic effectors, or proteins that set the seizure threshold, and epileptogenic mediators, which control the expression or functional state of the effector proteins. Under this framework, we then discuss attempts to regulate the mediators to control epilepsy. Further insights into the complex processes that render the brain susceptible to seizures and the identification of novel mediators of these processes will lead the way to the development of drugs to modify disease outcome and, potentially, to prevent epileptogenesis.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Encéfalo/efectos de los fármacos , Descubrimiento de Drogas/métodos , Epilepsia/tratamiento farmacológico , Epilepsia/prevención & control , Terapia Molecular Dirigida/métodos , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Ondas Encefálicas/efectos de los fármacos , Epilepsia/metabolismo , Epilepsia/fisiopatología , Humanos , Transducción de Señal/efectos de los fármacos
11.
Adv Exp Med Biol ; 813: 109-22, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25012371

RESUMEN

The effect of seizures on neuronal death and the role of seizure-induced neuronal death in acquired epileptogenesis have been debated for decades. Isolated brief seizures probably do not kill neurons; however, severe and repetitive seizures (i.e., status epilepticus) certainly do. Because status epilepticus both kills neurons and also leads to chronic epilepsy, neuronal death has been proposed to be an integral part of acquired epileptogenesis. Several studies, particularly in the immature brain, have suggested that neuronal death is not necessary for acquired epileptogenesis; however, the lack of neuronal death is difficult if not impossible to prove, and more recent studies have challenged this concept. Novel mechanisms of cell death, beyond the traditional concepts of necrosis and apoptosis, include autophagy, phagoptosis, necroptosis, and pyroptosis. The traditional proposal for why neuronal death may be necessary for epileptogenesis is based on the recapitulation of development hypothesis, where a loss of synaptic input from the dying neurons is considered a critical signal to induce axonal sprouting and synaptic-circuit reorganization. We propose a second hypothesis - the neuronal death pathway hypothesis, which states that the biochemical pathways causing programmed neurodegeneration, rather than neuronal death per se, are responsible for or contribute to epileptogenesis. The reprogramming of neuronal death pathways - if true - is proposed to derive from necroptosis or pyroptosis. The proposed new hypothesis may inform on why neuronal death seems closely linked to epileptogenesis, but may not always be.


Asunto(s)
Muerte Celular , Epilepsia/patología , Neuronas/patología , Humanos
12.
Neurobiol Dis ; 62: 273-85, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24141019

RESUMEN

Massive neuronal loss is a key pathological hallmark of Alzheimer's disease (AD). However, the mechanisms are still unclear. Here we demonstrate that neuroinflammation, cell autonomous to microglia, is capable of inducing neuronal cell cycle events (CCEs), which are toxic for terminally differentiated neurons. First, oligomeric amyloid-beta peptide (AßO)-mediated microglial activation induced neuronal CCEs via the tumor-necrosis factor-α (TNFα) and the c-Jun Kinase (JNK) signaling pathway. Second, adoptive transfer of CD11b+ microglia from AD transgenic mice (R1.40) induced neuronal cyclin D1 expression via TNFα signaling pathway. Third, genetic deficiency of TNFα in R1.40 mice (R1.40-Tnfα(-/-)) failed to induce neuronal CCEs. Finally, the mitotically active neurons spatially co-exist with F4/80+ activated microglia in the human AD brain and that a portion of these neurons are apoptotic. Together our data suggest a cell-autonomous role of microglia, and identify TNFα as the responsible cytokine, in promoting neuronal CCEs in the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Ciclo Celular , Microglía/metabolismo , Neuronas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Células Cultivadas , Lóbulo Frontal/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Lóbulo Temporal/metabolismo
13.
Proc Natl Acad Sci U S A ; 109(44): 18150-5, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23071306

RESUMEN

Under most physiological circumstances, monocytes are excluded from parenchymal CNS tissues. When widespread monocyte entry occurs, their numbers decrease shortly after engraftment in the presence of microglia. However, some disease processes lead to focal and selective loss, or dysfunction, of microglia, and microglial senescence typifies the aged brain. In this regard, the long-term engraftment of monocytes in the microglia-depleted brain remains unknown. Here, we report a model in which a niche for myeloid cells was created through microglia depletion. We show that microglia-depleted brain regions of CD11b-HSVTK transgenic mice are repopulated with new Iba-1-positive cells within 2 wk. The engrafted cells expressed high levels of CD45 and CCR2 and appeared in a wave-like pattern frequently associated with blood vessels, suggesting the engrafted cells were peripheral monocytes. Although two times more numerous and morphologically distinct from resident microglia up to 27 wk after initial engraftment, the overall distribution of the engrafted cells was remarkably similar to that of microglia. Two-photon in vivo imaging revealed that the engrafted myeloid cells extended their processes toward an ATP source and displayed intracellular calcium transients. Moreover, the engrafted cells migrated toward areas of kainic acid-induced neuronal death. These data provide evidence that circulating monocytes have the potential to occupy the adult CNS myeloid niche normally inhabited by microglia and identify a strong homeostatic drive to maintain the myeloid component in the mature brain.


Asunto(s)
Sistema Nervioso Central/citología , Homeostasis , Microglía/citología , Adenosina Trifosfato/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Ratones , Microglía/metabolismo , Timidina Quinasa/genética
14.
Neurobiol Aging ; 33(1): 197.e21-32, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20696495

RESUMEN

Considerable evidence points to important roles for inflammation in Alzheimer's disease (AD) pathophysiology. Epidemiological studies have suggested that long-term nonsteroidal anti-inflammatory drug (NSAID) therapy reduces the risk for Alzheimer's disease; however, the mechanism remains unknown. We report that a 9-month treatment of aged R1.40 mice resulted in 90% decrease in plaque burden and a similar reduction in microglial activation. Ibuprofen treatment reduced levels of lipid peroxidation, tyrosine nitration, and protein oxidation, demonstrating a dramatic effect on oxidative damage in vivo. Fibrillar ß-amyloid (Aß) stimulation has previously been demonstrated to induce the assembly and activation of the microglial nicotinamide adenine dinucleotide phosphate (NADPH) oxidase leading to superoxide production through a tyrosine kinase-based signaling cascade. Ibuprofen treatment of microglia or monocytes with racemic or S-ibuprofen inhibited Aß-stimulated Vav tyrosine phosphorylation, NADPH oxidase assembly, and superoxide production. Interestingly, Aß-stimulated Vav phosphorylation was not inhibited by COX inhibitors. These findings suggest that ibuprofen acts independently of cyclooxygenase COX inhibition to disrupt signaling cascades leading to microglial NADPH oxidase (NOX2) activation, preventing oxidative damage and enhancing plaque clearance in the brain.


Asunto(s)
Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Ibuprofeno/farmacología , NADPH Oxidasas/antagonistas & inhibidores , Péptidos beta-Amiloides , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Ibuprofeno/uso terapéutico , Masculino , Ratones , Ratones Transgénicos , Microglía/enzimología , Microglía/metabolismo , Microglía/patología , Monocitos/metabolismo , NADPH Oxidasas/fisiología , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Placa Amiloide , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-vav , Transducción de Señal/fisiología
15.
Acta Neuropathol ; 123(1): 31-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22101366

RESUMEN

The deposition of the ß-amyloid (Aß) peptide in senile plaques and cerebral Aß-amyloid angiopathy can be seeded in ß-amyloid precursor protein (APP)-transgenic mice by the intracerebral infusion of brain extracts containing aggregated Aß. Previous studies of seeded ß-amyloid induction have used relatively short incubation periods to dissociate seeded ß-amyloid induction from endogenous ß-amyloid deposition of the host, thus precluding the analysis of the impact of age and extended incubation periods on the instigation and spread of Aß lesions in brain. In the present study using R1.40 APP-transgenic mice (which do not develop endogenous Aß deposition up to 15 months of age) we show that: (1) seeding at 9 months of age does not induce more Aß deposition than seeding at 3 months of age, provided that the incubation period (6 months) is the same; and (2) very long-term (12 months) incubation after a focal application of the seed results in the emergence of Aß deposits throughout the forebrain. These findings indicate that the presence of Aß seeds, and not the age of the host per se, is critical to the initiation of Aß aggregation in the brain, and that Aß deposition, actuated in one brain area, eventually spreads throughout the brain.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Factores de Edad , Péptidos beta-Amiloides/farmacología , Animales , Modelos Animales de Enfermedad , Hipocampo/patología , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos
16.
Am J Pathol ; 177(5): 2549-62, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20864679

RESUMEN

Microglia, the primary immune effector cells in the brain, continually monitor the tissue parenchyma for pathological alterations and become activated in Alzheimer's disease. Loss of signaling between neurons and microglia via deletion of the microglial receptor, CX3CR1, worsens phenotypes in various models of neurodegenerative diseases. In contrast, CX3CR1 deficiency ameliorates pathology in murine stroke models. To examine the role of CX3CR1 in Alzheimer's disease-related ß-amyloid pathology, we generated APPPS1 and R1.40 transgenic mouse models of Alzheimer's disease deficient for CX3CR1. Surprisingly, CX3CR1 deficiency resulted in a gene dose-dependent reduction in ß-amyloid deposition in both the APPPS1 and R1.40 mouse models of AD. Immunohistochemical analysis revealed reduced staining for CD68, a marker of microglial activation. Furthermore, quantitative immunohistochemical analysis revealed reduced numbers of microglia surrounding ß-amyloid deposits in the CX3CR1-deficient APPPS1 animals. The reduced ß-amyloid pathology correlated with reduced levels of TNFα and CCL2 mRNAs, but elevated IL1ß mRNA levels, suggesting an altered neuroinflammatory milieu. Finally, to account for these seemingly disparate results, both in vitro and in vivo studies provided evidence that CX3CL1/CX3CR1 signaling alters the phagocytic capacity of microglia, including the uptake of Aß fibrils. Taken together, these results demonstrate that loss of neuron-microglial fractalkine signaling leads to reduced ß-amyloid deposition in mouse models of AD that is potentially mediated by altered activation and phagocytic capability of CX3CR1-deficient microglia.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Animales de Enfermedad , Microglía/metabolismo , Receptores de Quimiocina/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Receptor 1 de Quimiocinas CX3C , Movimiento Celular , Proliferación Celular , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Fagocitosis , Receptores de Quimiocina/genética
17.
Invest Ophthalmol Vis Sci ; 51(6): 3264-72, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20071672

RESUMEN

PURPOSE: To determine the molecular basis and the pathologic consequences of a chemically induced mutation in a mouse model of photoreceptor degeneration, nmf240. METHODS: Mice from a G3 N-ethyl-N-nitrosourea mutagenesis program were screened by indirect ophthalmoscopy for abnormal fundi. A chromosomal position for the recessive nmf240 mutation was determined by a genome-wide linkage analysis by use of simple sequence length polymorphic markers in an F2 intercross. The critical region was refined, and candidate genes were screened by direct sequencing. The nmf240 phenotype was characterized by histologic analysis of the retina, brain, and male reproductive organs and by electroretinogram (ERG)-based studies of the retina and retinal pigment epithelium (RPE). RESULTS: Clinically, homozygous nmf240 mutants exhibit a grainy retina that progresses to panretinal patches of depigmentation. The mutation was localized to a region on chromosome 16 containing Clcn2, a gene associated with retinal degeneration. Sequencing identified a missense C-T mutation at nucleotide 1063 in Clcn2 that converts a glutamine to a stop codon. Mice homozygous for the Clcn2(nmf240) mutation experience a severe loss of photoreceptor cells at 14 days of age that is preceded by an elongation of RPE apical microvilli. Homozygous mutants also experience leukoencephalopathy in multiple brain areas and male sterility. Despite a normal retinal histology in nmf240 heterozygotes, the ERG light peak, generated by the RPE, is reduced. CONCLUSIONS: The nmf240 phenotype closely resembles that reported for Clcn2 knockout mice. The observation that heterozygous nmf240 mice present with a reduced ERG light peak component suggests that CLCN2 is necessary for the generation of this response component.


Asunto(s)
Azoospermia/genética , Canales de Cloruro/genética , Codón sin Sentido , Leucoencefalopatías/genética , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/genética , Epitelio Pigmentado de la Retina/patología , Animales , Azoospermia/patología , Western Blotting , Encéfalo/patología , Canales de Cloruro CLC-2 , Electrorretinografía , Etilnitrosourea/toxicidad , Femenino , Estudio de Asociación del Genoma Completo , Leucoencefalopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Mutagénesis/efectos de los fármacos , Degeneración Retiniana/patología
18.
J Clin Invest ; 119(12): 3692-702, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19907078

RESUMEN

Ectopic cell cycle events (CCEs) mark vulnerable neuronal populations in human Alzheimer disease (AD) and are observed early in disease progression. In transgenic mouse models of AD, CCEs are found before the onset of beta-amyloid peptide (Abeta) deposition to form senile plaques, a hallmark of AD. Here, we have demonstrated that alterations in brain microglia occur coincidently with the appearance of CCEs in the R1.40 transgenic mouse model of AD. Furthermore, promotion of inflammation with LPS at young ages in R1.40 mice induced the early appearance of neuronal CCEs, whereas treatment with 2 different nonsteroidal antiinflammatory drugs (NSAIDs) blocked neuronal CCEs and alterations in brain microglia without altering amyloid precursor protein (APP) processing and steady-state Abeta levels. In addition, NSAID treatment of older R1.40 animals prevented new neuronal CCEs, although it failed to reverse existing ones. Retrospective human epidemiological studies have identified long-term use of NSAIDs as protective against AD. Prospective clinical trials, however, have failed to demonstrate a similar benefit. Our use of CCEs as an outcome measure offers fresh insight into this discrepancy and provides important information for future clinical trials, as it suggests that NSAID use in human AD may need to be initiated as early as possible to prevent disease progression.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Antiinflamatorios no Esteroideos/farmacología , Neuronas/efectos de los fármacos , Neuronas/patología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Ciclo Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Ibuprofeno/farmacología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/patología , Mutación , Naproxeno/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
J Neurosci ; 29(11): 3565-78, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19295161

RESUMEN

The amyloid-beta (Abeta) precursor protein (APP), a transmembrane protein that undergoes proteolytic cleavage into defined fragments, has been implicated in axonal transport. The proposed role of APP as a vesicle receptor for the microtubule motor kinesin-1 has relevance for the pathogenesis of Alzheimer's disease. Nevertheless, this function, which relies on the transport to the cell periphery of full-length APP rather than its cleavage fragments, remains controversial. Other proposed functions of APP, such as regulating transcription, neurogenesis, cell movement, or neurite growth also rely on APP's presence as a full-length protein at the cell surface, implying that APP cleavage occurs after its transport to the cell periphery. To test this hypothesis, we mapped the localization of various APP epitopes in neurons in culture and in the mouse brain. Surprisingly, epitopes from the N-terminal, C-terminal, and central (Abeta) domains of APP each showed a distinct distribution throughout the cell and rarely colocalized. Within neurites, these epitopes were localized to distinct transport vesicles that associated with different sets of microtubules and, occasionally, actin filaments. C-terminal APP fragments were preferentially transported into neurites as phosphorylated forms, entered the lamellipodium and filopodia of growth cones, and concentrated in regions of growth cone turning and advancement (unlike the N-terminal and Abeta fragments). We conclude that, under normal conditions, the proteolytic cleavage of APP primarily occurs before its sorting into axonal transport vesicles and the cleaved fragments segregate into separate vesicle populations that reach different destinations, and thus have different functions.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Neuritas/química , Neuritas/metabolismo , Fragmentos de Péptidos/metabolismo , Vesículas Sinápticas/química , Vesículas Sinápticas/metabolismo , Precursor de Proteína beta-Amiloide/fisiología , Animales , Transporte Axonal/fisiología , Células Cultivadas , Humanos , Hidrólisis , Líquido Intracelular/química , Líquido Intracelular/metabolismo , Líquido Intracelular/fisiología , Ratones , Ratones Transgénicos , Neuritas/fisiología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/fisiología , Transporte de Proteínas/fisiología , Vesículas Sinápticas/fisiología
20.
J Neurosci ; 28(43): 10786-93, 2008 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-18945886

RESUMEN

Neurons subject to degeneration in Alzheimer's disease (AD) exhibit evidence of re-entry into a mitotic cell cycle even before the development of substantial AD brain pathology. In efforts to identify the initiating factors underlying these cell cycle events (CCEs), we have characterized the appearance of the neuronal CCEs in the genomic-based R1.40 transgenic mouse model of AD. Notably, R1.40 mice exhibit neuronal CCEs in a reproducible temporal and spatial pattern that recapitulates the neuronal vulnerability seen in human AD. Neuronal CCEs first appear at 6 months in the frontal cortex layers II/III. This is 6-8 months before detectable amyloid beta (Abeta) deposition, suggesting that specific amyloid precursor protein (APP) processing products are responsible for the induction of neuronal CCEs. Furthermore, a reduction in the levels of Abeta (achieved by shifting the genetic background from C57BL/6 to the DBA/2 mouse strain) dramatically delays the appearance of neuronal CCEs. More significantly, elimination of beta-secretase activity blocks the appearance of CCEs, providing direct genetic evidence that the amyloidogenic processing of APP is required for the induction of CCEs. Finally, in vitro preparations of oligomeric, but not monomeric, Abeta induce DNA synthesis in dissociated cortical neurons, and this response is blocked by antioligomer specific antibodies. Together, our data suggest that low molecular weight aggregates of Abeta induce neuronal cell cycle re-entry in mouse models of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Ciclo Celular/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Factores de Edad , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/deficiencia , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/deficiencia , Bromodesoxiuridina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...