Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Biol Chem ; 287(3): 2032-44, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22130662

RESUMEN

Allelic variation in the apolipoprotein E (APOE) gene is the major risk factor of sporadic Alzheimer disease. ApoE is the primary cholesterol carrier in the brain. Previously, we demonstrated that intracellular degradation of ß-amyloid (Aß) peptides by microglia is dramatically enhanced in the presence of apoE. However, the molecular mechanisms subserving this effect remain unknown. This study reports a mechanistic link between apoE-regulated cholesterol homeostasis and Aß degradation. We demonstrate that promoting intracellular Aß degradation by microglia is a common feature of HDL apolipoproteins, including apoE and apoA-I. This effect was not dependent on the direct interaction of apoE and Aß. Regulation of Aß degradation was achieved by solely manipulating cellular cholesterol levels. The expression and the activity of Aß degrading enzymes, however, were not regulated by cholesterol. We observed that reducing cellular cholesterol levels by apoE resulted in faster delivery of Aß to lysosomes and enhanced degradation. Moreover, apoE facilitated the recycling of Rab7, a small GTPase responsible for recruiting the motor complex to late endosomes/lysosomes. These data indicate that faster endocytic trafficking of Aß-containing vesicles in the presence of apoE resulted from efficient recycling of Rab7 from lysosomes to early endosomes. Thus, apoE-induced intracellular Aß degradation is mediated by the cholesterol efflux function of apoE, which lowers cellular cholesterol levels and subsequently facilitates the intracellular trafficking of Aß to lysosomes for degradation. These findings demonstrate a direct role of cholesterol in the intracellular Aß degradation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Colesterol/metabolismo , Lisosomas/metabolismo , Microglía/metabolismo , Proteolisis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Animales , Apolipoproteínas E/genética , Células Cultivadas , Colesterol/genética , Humanos , Lisosomas/genética , Ratones , Ratones Noqueados , Transporte de Proteínas/fisiología , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
2.
bioRxiv ; 2023 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-37577582

RESUMEN

Background: Genetic study of late-onset Alzheimer's disease (AD) reveals that a rare Arginine-to-Histamine mutation at amino acid residue 47 (R47H) in Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) results in increased disease risk. TREM2 plays critical roles in regulating microglial response to amyloid plaques in AD, leading to their clustering and activation surrounding the plaques. We previously showed that increasing human TREM2 gene dosage exerts neuroprotective effects against AD-related deficits in amyloid depositing mouse models of AD. However, the in vivo effects of the R47H mutation on human TREM2-mediated microglial reprogramming and neuroprotection remains poorly understood. Method: Here we created a BAC transgenic mouse model expressing human TREM2 with the R47H mutation in its cognate genomic context (BAC-TREM2-R47H). Importantly, the BAC used in this study was engineered to delete critical exons of other TREM-like genes on the BAC to prevent confounding effects of overexpressing multiple TREM-like genes. We crossed BAC-TREM2- R47H mice with 5xFAD [1], an amyloid depositing mouse model of AD, to evaluate amyloid pathologies and microglial phenotypes, transcriptomics and in situ expression of key TREM2 -dosage dependent genes. We also compared the key findings in 5xFAD/BAC-TREM2-R47H to those observed in 5xFAD/BAC-TREM2 mice. Result: Both BAC-TREM2 and BAC-TREM2-R47H showed proper expression of three splicing isoforms of TREM2 that are normally found in human. In 5xFAD background, elevated TREM2-R47H gene dosages significantly reduced the plaque burden, especially the filamentous type. The results were consistent with enhanced phagocytosis and altered NLRP3 inflammasome activation in BAC- TREM2-R47H microglia in vitro. However, unlike TREM2 overexpression, elevated TREM2- R47H in 5xFAD failed to ameliorate cognitive and transcriptomic deficits. In situ analysis of key TREM2 -dosage dependent genes and microglial morphology uncovered that TREM2-R47H showed a loss-of-function phenotype in reprogramming of plaque-associated microglial reactivity and gene expression in 5xFAD. Conclusion: Our study demonstrated that the AD-risk variant has a previously unknown, mixture of partial and full loss of TREM2 functions in modulating microglial response in AD mouse brains. Together, our new BAC-TREM2-R47H model and prior BAC-TREM2 mice are invaluable resource to facilitate the therapeutic discovery that target human TREM2 and its R47H variant to ameliorate AD and other neurodegenerative disorders.

3.
J Neurosci ; 29(13): 4252-62, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19339619

RESUMEN

Alzheimer's disease is characterized by the progressive deposition of beta-amyloid (Abeta) within the brain parenchyma and its subsequent accumulation into senile plaques. Pathogenesis of the disease is associated with perturbations in Abeta homeostasis and the inefficient clearance of these soluble and insoluble peptides from the brain. Microglia have been reported to mediate the clearance of fibrillar Abeta (fAbeta) through receptor-mediated phagocytosis; however, their participation in clearance of soluble Abeta peptides (sAbeta) is largely unknown. We report that microglia internalize sAbeta from the extracellular milieu through a nonsaturable, fluid phase macropinocytic mechanism that is distinct from phagocytosis and receptor-mediated endocytosis both in vitro and in vivo. The uptake of sAbeta is dependent on both actin and tubulin dynamics and does not involve clathrin assembly, coated vesicles or membrane cholesterol. Upon internalization, fluorescently labeled sAbeta colocalizes to pinocytic vesicles. Microglia rapidly traffic these soluble peptides into late endolysosomal compartments where they are subject to degradation. Additionally, we demonstrate that the uptake of sAbeta and fAbeta occurs largely through distinct mechanisms and upon internalization are segregated into separate subcellular vesicular compartments. Significantly, we found that upon proteolytic degradation of fluorescently labeled sAbeta, the fluorescent chromophore is retained by the microglial cell. These studies identify an important mechanism through which microglial cells participate in the maintenance of Abeta homeostasis, through their capacity to constitutively clear sAbeta peptides from the brain.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Microglía/fisiología , Fragmentos de Péptidos/metabolismo , Pinocitosis/fisiología , Péptidos beta-Amiloides/administración & dosificación , Animales , Animales Recién Nacidos , Encéfalo/citología , Células Cultivadas , Técnicas de Cocultivo , Citocalasina D/farmacología , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Citometría de Flujo/métodos , Proteínas Fluorescentes Verdes/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Nocodazol/farmacología , Fragmentos de Péptidos/administración & dosificación , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Pinocitosis/genética , Transporte de Proteínas/efectos de los fármacos , Análisis Espectral , Moduladores de Tubulina/farmacología
4.
J Neural Transm (Vienna) ; 117(8): 949-60, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20552234

RESUMEN

Alzheimer's disease (AD), the most prominent cause of senile dementia, is clinically characterized by the extracellular deposition of beta-amyloid (Abeta) and the intracellular neurofibrillary tangles. It has been well accepted that AD pathogenesis arises from perturbation in the homeostasis of Abeta in the brain. Abeta is normally produced at high levels in the brain and cleared in an equivalent rate. Thus, even a moderate decrease in the clearance leads to the accumulation of Abeta and subsequent amyloid deposition. Microglia are the tissue macrophages in the central nervous system (CNS) and have been shown to play major roles in internalization and degradation of Abeta. Abeta exists in the brain both in soluble and in fibrillar forms. Microglia interact with these two forms of Abeta in different ways. They take up soluble forms of Abeta through macropinocytosis and LDL receptor-related proteins (LRPs) mediated pathway. Fibrillar forms of Abeta interact with the cell surface innate immune receptor complex, initiating intracellular signaling cascades that stimulate phagocytosis. Inflammatory responses influence the activation status of microglia and subsequently regulate their ability to take up and degrade Abeta. ApoE and its receptors have been shown to play critical roles in these processes. In this review, we will explore the mechanisms that microglia utilize to clear Abeta and the effectors that modulate the processes.


Asunto(s)
Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Encéfalo/citología , Microglía/metabolismo , Apolipoproteínas E/fisiología , Humanos , Inflamación/metabolismo
5.
Neuron ; 106(1): 4-6, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32272066

RESUMEN

Forward genetic screen, typically performed in invertebrates or mammalian cell lines, has been instrumental in discovering genes essential for neural function. In this issue of Neuron, Wertz et al. (2020) demonstrate the first viral-mediated, genome-wide screen to identify neuroprotective genes in wild-type and Huntington's disease (HD) mouse brains.


Asunto(s)
Enfermedad de Huntington , Animales , Muerte Celular , Supervivencia Celular , Ratones , Neuronas , Neuroprotección
6.
Sci Rep ; 10(1): 20295, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33219289

RESUMEN

In Huntington's disease (HD), the mutant Huntingtin (mHTT) is postulated to mediate template-based aggregation that can propagate across cells. It has been difficult to quantitatively detect such pathological seeding activities in patient biosamples, e.g. cerebrospinal fluids (CSF), and study their correlation with the disease manifestation. Here we developed a cell line expressing a domain-engineered mHTT-exon 1 reporter, which showed remarkably high sensitivity and specificity in detecting mHTT seeding species in HD patient biosamples. We showed that the seeding-competent mHTT species in HD CSF are significantly elevated upon disease onset and with the progression of neuropathological grades. Mechanistically, we showed that mHTT seeding activities in patient CSF could be ameliorated by the overexpression of chaperone DNAJB6 and by antibodies against the polyproline domain of mHTT. Together, our study developed a selective and scalable cell-based tool to investigate mHTT seeding activities in HD CSF, and demonstrated that the CSF mHTT seeding species are significantly associated with certain disease states. This seeding activity can be ameliorated by targeting specific domain or proteostatic pathway of mHTT, providing novel insights into such pathological activities.


Asunto(s)
Líquido Cefalorraquídeo/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/patología , Chaperonas Moleculares/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Agregación Patológica de Proteínas/patología , Adulto , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Línea Celular , Exones/genética , Femenino , Genes Reporteros/genética , Proteínas del Choque Térmico HSP40/genética , Humanos , Proteína Huntingtina/líquido cefalorraquídeo , Proteína Huntingtina/genética , Enfermedad de Huntington/líquido cefalorraquídeo , Enfermedad de Huntington/genética , Microscopía Intravital , Masculino , Persona de Mediana Edad , Chaperonas Moleculares/genética , Mutación , Proteínas del Tejido Nervioso/genética , Agregación Patológica de Proteínas/líquido cefalorraquídeo , Agregación Patológica de Proteínas/genética , Dominios Proteicos/genética , Ingeniería de Proteínas , Pliegue de Proteína
7.
Neuron ; 97(5): 1032-1048.e5, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29518357

RESUMEN

Variants of TREM2 are associated with Alzheimer's disease (AD). To study whether increasing TREM2 gene dosage could modify the disease pathogenesis, we developed BAC transgenic mice expressing human TREM2 (BAC-TREM2) in microglia. We found that elevated TREM2 expression reduced amyloid burden in the 5xFAD mouse model. Transcriptomic profiling demonstrated that increasing TREM2 levels conferred a rescuing effect, which includes dampening the expression of multiple disease-associated microglial genes and augmenting downregulated neuronal genes. Interestingly, 5xFAD/BAC-TREM2 mice showed further upregulation of several reactive microglial genes linked to phagocytosis and negative regulation of immune cell activation. Moreover, these mice showed enhanced process ramification and phagocytic marker expression in plaque-associated microglia and reduced neuritic dystrophy. Finally, elevated TREM2 gene dosage led to improved memory performance in AD models. In summary, our study shows that a genomic transgene-driven increase in TREM2 expression reprograms microglia responsivity and ameliorates neuropathological and behavioral deficits in AD mouse models.


Asunto(s)
Enfermedad de Alzheimer/genética , Modelos Animales de Enfermedad , Dosificación de Gen/genética , Glicoproteínas de Membrana/genética , Microglía/fisiología , Fenotipo , Receptores Inmunológicos/genética , Enfermedad de Alzheimer/patología , Animales , Animales Recién Nacidos , Células Cultivadas , Técnicas de Reprogramación Celular/métodos , Femenino , Humanos , Masculino , Glicoproteínas de Membrana/biosíntesis , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Técnicas de Cultivo de Órganos , Receptores Inmunológicos/biosíntesis
8.
Neuron ; 85(4): 726-41, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25661181

RESUMEN

The nucleus is a critical subcellular compartment for the pathogenesis of polyglutamine disorders, including Huntington's disease (HD). Recent studies suggest the first 17-amino-acid domain (N17) of mutant huntingtin (mHTT) mediates its nuclear exclusion in cultured cells. Here, we test whether N17 could be a molecular determinant of nuclear mHTT pathogenesis in vivo. BAC transgenic mice expressing mHTT lacking the N17 domain (BACHD-ΔN17) show dramatically accelerated mHTT pathology exclusively in the nucleus, which is associated with HD-like transcriptionopathy. Interestingly, BACHD-ΔN17 mice manifest more overt disease-like phenotypes than the original BACHD mice, including body weight loss, movement deficits, robust striatal neuron loss, and neuroinflammation. Mechanistically, N17 is necessary for nuclear exclusion of small mHTT fragments that are part of nuclear pathology in HD. Together, our study suggests that N17 modifies nuclear pathogenesis and disease severity in HD mice by regulating subcellular localization of known nuclear pathogenic mHTT species.


Asunto(s)
Nucléolo Celular/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Expansión de Repetición de Trinucleótido/genética , Factores de Edad , Animales , Encéfalo/metabolismo , Encéfalo/patología , Nucléolo Celular/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Células HEK293/ultraestructura , Humanos , Proteína Huntingtina , Enfermedad de Huntington/complicaciones , Locomoción/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Tamaño de los Órganos/genética , Fenotipo , Estructura Terciaria de Proteína/genética , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología
9.
FEBS J ; 280(18): 4382-94, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23829302

RESUMEN

This year (2013) marks the 20th anniversary of identification of the causal genetic mutation for Huntington's disease (HD), a landmark discovery that heralded study of the biological underpinnings of this most common dominantly inherited neurodegenerative disorder. Among the variety of model organisms used to study HD pathogenesis, the mouse model is by far the most commonly used mammalian genetic model. Much of our current knowledge regarding mutant huntingtin (mHtt)-induced disease pathogenesis in mammalian models has been obtained by studying transgenic mouse models expressing mHtt N-terminal fragments, full-length murine or human mHtt. In this review, we focus on recent progress in using novel HD mouse models with targeted mHtt expression in specific brain cell types. These models help to address the role of distinct neuronal and non-neuronal cell types in eliciting cell-autonomous or non-cell-autonomous disease processes in HD. We also describe several mHtt transgenic mouse models with targeted mutations in Htt cis-domains to address specific pathogenic hypotheses, ranging from mHtt proteolysis to post-translational modifications. These novel mouse genetic studies, through direct manipulations of the causal HD gene, utilize a reductionist approach to systematically unravel the cellular and molecular pathways that are targeted by mHtt in disease pathogenesis, and to potentially identify novel targets for therapy.


Asunto(s)
Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Proteínas Nucleares/genética , Animales , Caspasa 6/genética , Caspasa 6/metabolismo , Corteza Cerebral/patología , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/metabolismo , Neuronas/patología , Proteínas Nucleares/metabolismo , Ingeniería de Proteínas , Estructura Terciaria de Proteína , Transducción de Señal
10.
Science ; 335(6075): 1503-6, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22323736

RESUMEN

Alzheimer's disease (AD) is associated with impaired clearance of ß-amyloid (Aß) from the brain, a process normally facilitated by apolipoprotein E (apoE). ApoE expression is transcriptionally induced through the action of the nuclear receptors peroxisome proliferator-activated receptor gamma and liver X receptors in coordination with retinoid X receptors (RXRs). Oral administration of the RXR agonist bexarotene to a mouse model of AD resulted in enhanced clearance of soluble Aß within hours in an apoE-dependent manner. Aß plaque area was reduced more than 50% within just 72 hours. Furthermore, bexarotene stimulated the rapid reversal of cognitive, social, and olfactory deficits and improved neural circuit function. Thus, RXR activation stimulates physiological Aß clearance mechanisms, resulting in the rapid reversal of a broad range of Aß-induced deficits.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Tetrahidronaftalenos/farmacología , Tetrahidronaftalenos/uso terapéutico , Amiloidosis/tratamiento farmacológico , Amiloidosis/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Conducta Animal/efectos de los fármacos , Bexaroteno , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Terapia Molecular Dirigida , Odorantes , Vías Olfatorias/efectos de los fármacos , Vías Olfatorias/fisiología , Receptores Nucleares Huérfanos/metabolismo , PPAR gamma/metabolismo , Fagocitosis , Placa Amiloide/tratamiento farmacológico , Receptores X Retinoide/agonistas , Receptores X Retinoide/metabolismo
11.
J Neuroimmune Pharmacol ; 3(2): 130-40, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18247125

RESUMEN

Removal of apoptotic cells is an essential process for normal development and tissue maintenance. Importantly, apoptotic cells stimulate their phagocytosis by macrophages while actively suppressing inflammatory responses. Growth arrest specific gene 6 (Gas6) is involved in this process, bridging phosphatidylserine residues on the surface of apoptotic cells to the Axl/Mer family of tyrosine kinases which stimulate phagocytosis. Animals with mutations or loss of these receptors exhibit phenotypes reflective of impaired phagocytosis and a hyperactive immune response. We report that Gas6 induces phagocytosis in microglia through a novel non-classical phagocytic mechanism. Gas6 stimulates a type-II-related phagocytic response, but requires Vav phosphorylation and Rac activation, distinguishing it from the classical type II mechanism. Importantly, Gas6 suppressed lipopolysaccharide-induced expression of the inflammatory molecules IL-1beta and iNOS. Gas6 inhibited iNOS expression through suppression of promoter activity. The present data provide direct evidence for the role of Gas6 receptors in mediating an anti-inflammatory response to ligands found on apoptotic cells with the simultaneous stimulation of phagocytosis. These data provide a mechanistic explanation for the phenotype observed in animals lacking Axl/Mer receptors.


Asunto(s)
Inflamación/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Microglía/efectos de los fármacos , Proteínas Oncogénicas/fisiología , Fagocitosis/efectos de los fármacos , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Encéfalo/metabolismo , Línea Celular/efectos de los fármacos , Línea Celular/enzimología , Línea Celular/fisiología , Citoesqueleto/fisiología , Citoesqueleto/ultraestructura , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Lipopolisacáridos/toxicidad , Ratones , Microglía/enzimología , Microglía/fisiología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Fagocitosis/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-vav/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Transcripción Genética/efectos de los fármacos , Tirosina Quinasa c-Mer , Proteínas de Unión al GTP rac/metabolismo , Tirosina Quinasa del Receptor Axl
12.
Neuron ; 58(5): 681-93, 2008 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-18549781

RESUMEN

Apolipoprotein E is associated with age-related risk for Alzheimer's disease and plays critical roles in Abeta homeostasis. We report that ApoE plays a role in facilitating the proteolytic clearance of soluble Abeta from the brain. The endolytic degradation of Abeta peptides within microglia by neprilysin and related enzymes is dramatically enhanced by ApoE. Similarly, Abeta degradation extracellularly by insulin-degrading enzyme is facilitated by ApoE. The capacity of ApoE to promote Abeta degradation is dependent upon the ApoE isoform and its lipidation status. The enhanced expression of lipidated ApoE, through the activation of liver X receptors, stimulates Abeta degradation. Indeed, aged Tg2576 mice treated with the LXR agonist GW3965 exhibited a dramatic reduction in brain Abeta load. GW3965 treatment also reversed contextual memory deficits. These data demonstrate a mechanism through which ApoE facilitates the clearance of Abeta from the brain and suggest that LXR agonists may represent a novel therapy for AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/farmacología , Microglía/efectos de los fármacos , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Envejecimiento , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Benzoatos/farmacología , Bencilaminas/farmacología , Encéfalo/patología , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Receptores X del Hígado , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Receptores Nucleares Huérfanos , Fragmentos de Péptidos/metabolismo , Placa Amiloide/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Tiempo
13.
J Immunol ; 178(12): 7794-804, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17548617

RESUMEN

Dendritic cells (DC) are essential for the initiation of primary adaptive immune responses, and their functionality is strongly down-modulated by IL-10. Both innate and adaptive immune signals trigger the up-regulation of antiapoptotic Bcl-2 family members to facilitate the survival of DCs after maturation. However, whether IL-10 alters the expression of apoptotic-related genes in maturing DCs has not been determined. In this study, we demonstrate that spontaneous apoptosis rapidly occurred in myeloid DCs exposed to exogenous IL-10 upon maturation. Microarray analysis indicates that IL-10 suppressed the induction of three antiapoptotic genes, bcl-2, bcl-x, and bfl-1, which was coincident with the increased sensitivity of mature DCs to spontaneous apoptosis. IL-10 markedly inhibited the accumulation of steady state Bcl-2 message and protein in myeloid DCs activated through TLRs or TNFR family members, whereas exogenous IL-10 affected Bcl-x(L) expression in a moderate manner. In contrast, bcl-2 expression of plasmacytoid DCs was less sensitive to the effects of IL-10. We further show that autocrine IL-10 significantly limited the longevity of myeloid DCs and altered the expression kinetics of Bcl-2 but not Bcl-x(L) in maturing DCs. We conclude that the degree of IL-10 exposure and/or the level of endogenous IL-10 production upon myeloid DC maturation play a critical role in determining DC longevity. This regulatory mechanism of IL-10 is associated with the dynamic control of antiapoptotic Bcl-2 proteins.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Células Dendríticas/efectos de los fármacos , Interleucina-10/farmacología , Células Mieloides/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/análisis , Proteínas Reguladoras de la Apoptosis/genética , Antígeno CD11c/análisis , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Expresión Génica/efectos de los fármacos , Humanos , Activación de Linfocitos , Células Mieloides/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-bcl-2 , Linfocitos T Colaboradores-Inductores/inmunología , Proteína bcl-X/análisis , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
14.
J Biol Chem ; 281(30): 20842-20850, 2006 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-16728400

RESUMEN

Microglial interaction with extracellular beta-amyloid fibrils (fAbeta) is mediated through an ensemble of cell surface receptors, including the B-class scavenger receptor CD36, the alpha(6)beta(1)-integrin, and the integrin-associated protein/CD47. The binding of fAbeta to this receptor complex has been shown to drive a tyrosine kinase-based signaling cascade leading to production of reactive oxygen species and stimulation of phagocytic activity; however, little is known about the intracellular signaling cascades governing the microglial response to fAbeta. This study reports a direct mechanistic link between the fAbeta cell surface receptor complex and downstream signaling events responsible for NADPH oxidase activation and phagosome formation. The Vav guanine nucleotide exchange factor is tyrosine-phosphorylated in response to fAbeta peptides as a result of the engagement of the microglia fAbeta cell surface receptor complex. Co-immunoprecipitation studies demonstrate an Abeta-dependent association between Vav and both Lyn and Syk kinases. The downstream target of Vav, the small GTPase Rac1, is GTP-loaded in an Abeta-dependent manner. Rac1 is both an essential component of the NADPH oxidase and a critical regulator of microglial phagocytosis. The direct role of Vav in fAbeta-stimulated intracellular signaling cascades was established using primary microglia obtained from Vav(-/-) mice. Stimulation of Vav(-/-) microglia with fAbeta failed to generate NADPH oxidase-derived reactive oxygen species and displayed a dramatically attenuated phagocytic response. These findings directly link Vav phosphorylation to the Abeta-receptor complex and demonstrate that Vav activity is required for fAbeta-stimulated intracellular signaling events upstream of reactive oxygen species production and phagosome formation.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Microglía/metabolismo , Monocitos/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Estallido Respiratorio , Transducción de Señal , Animales , Antígenos CD36/biosíntesis , Antígeno CD47/biosíntesis , Humanos , Integrina alfa6beta1/metabolismo , Ratones , Ratones Transgénicos , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA