Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(2): 113764, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38358885

RESUMO

Over half of patients with brain tumors experience debilitating and often progressive cognitive decline after radiotherapy treatment. Microglia, the resident macrophages in the brain, have been implicated in this decline. In response to various insults, microglia can develop innate immune memory (IIM), which can either enhance (priming or training) or repress (tolerance) the response to subsequent inflammatory challenges. Here, we investigate whether radiation affects the IIM of microglia by irradiating the brains of rats and later exposing them to a secondary inflammatory stimulus. Comparative transcriptomic profiling and protein validation of microglia isolated from irradiated rats show a stronger immune response to a secondary inflammatory insult, demonstrating that radiation can lead to long-lasting molecular reprogramming of microglia. Transcriptomic analysis of postmortem normal-appearing non-tumor brain tissue of patients with glioblastoma indicates that radiation-induced microglial priming is likely conserved in humans. Targeting microglial priming or avoiding further inflammatory insults could decrease radiotherapy-induced neurotoxicity.


Assuntos
Encéfalo , Microglia , Humanos , Ratos , Animais , Microglia/metabolismo , Imunidade Inata
2.
Science ; 373(6550)2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34210853

RESUMO

The mechanisms by which macrophages regulate energy storage remain poorly understood. We identify in a genetic screen a platelet-derived growth factor (PDGF)/vascular endothelial growth factor (VEGF)-family ortholog, Pvf3, that is produced by macrophages and is required for lipid storage in fat-body cells of Drosophila larvae. Genetic and pharmacological experiments indicate that the mouse Pvf3 ortholog PDGFcc, produced by adipose tissue-resident macrophages, controls lipid storage in adipocytes in a leptin receptor- and C-C chemokine receptor type 2-independent manner. PDGFcc production is regulated by diet and acts in a paracrine manner to control lipid storage in adipose tissues of newborn and adult mice. At the organismal level upon PDGFcc blockade, excess lipids are redirected toward thermogenesis in brown fat. These data identify a macrophage-dependent mechanism, conducive to the design of pharmacological interventions, that controls energy storage in metazoans.


Assuntos
Adipócitos/imunologia , Dieta Hiperlipídica , Proteínas de Drosophila/metabolismo , Metabolismo Energético , Linfocinas/metabolismo , Macrófagos/imunologia , Obesidade/imunologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Termogênese , Tecido Adiposo Marrom/imunologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Hemócitos/imunologia , Fígado/imunologia , Linfocinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Crescimento Derivado de Plaquetas/genética , Receptores CCR2/genética , Receptores CCR2/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Sci Adv ; 7(25)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34134993

RESUMO

Mechanisms by which noncoding genetic variation influences gene expression remain only partially understood but are considered to be major determinants of phenotypic diversity and disease risk. Here, we evaluated effects of >50 million single-nucleotide polymorphisms and short insertions/deletions provided by five inbred strains of mice on the responses of macrophages to interleukin-4 (IL-4), a cytokine that plays pleiotropic roles in immunity and tissue homeostasis. Of >600 genes induced >2-fold by IL-4 across the five strains, only 26 genes reached this threshold in all strains. By applying deep learning and motif mutation analyses to epigenetic data for macrophages from each strain, we identified the dominant combinations of lineage-determining and signal-dependent transcription factors driving IL-4 enhancer activation. These studies further revealed mechanisms by which noncoding genetic variation influences absolute levels of enhancer activity and their dynamic responses to IL-4, thereby contributing to strain-differential patterns of gene expression and phenotypic diversity.


Assuntos
Interleucina-4 , Macrófagos , Animais , Elementos Facilitadores Genéticos , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/metabolismo
4.
Science ; 366(6469): 1134-1139, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31727856

RESUMO

Noncoding genetic variation is a major driver of phenotypic diversity, but functional interpretation is challenging. To better understand common genetic variation associated with brain diseases, we defined noncoding regulatory regions for major cell types of the human brain. Whereas psychiatric disorders were primarily associated with variants in transcriptional enhancers and promoters in neurons, sporadic Alzheimer's disease (AD) variants were largely confined to microglia enhancers. Interactome maps connecting disease-risk variants in cell-type-specific enhancers to promoters revealed an extended microglia gene network in AD. Deletion of a microglia-specific enhancer harboring AD-risk variants ablated BIN1 expression in microglia, but not in neurons or astrocytes. These findings revise and expand the list of genes likely to be influenced by noncoding variants in AD and suggest the probable cell types in which they function.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/genética , Encéfalo/metabolismo , Elementos Facilitadores Genéticos/genética , Variação Genética , Microglia/metabolismo , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Proteínas Supressoras de Tumor/genética , Células Cultivadas , Cromatina/metabolismo , Redes Reguladoras de Genes , Estudo de Associação Genômica Ampla , Humanos , Deleção de Sequência
5.
Cell ; 173(7): 1796-1809.e17, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29779944

RESUMO

Non-coding genetic variation is a major driver of phenotypic diversity and allows the investigation of mechanisms that control gene expression. Here, we systematically investigated the effects of >50 million variations from five strains of mice on mRNA, nascent transcription, transcription start sites, and transcription factor binding in resting and activated macrophages. We observed substantial differences associated with distinct molecular pathways. Evaluating genetic variation provided evidence for roles of ∼100 TFs in shaping lineage-determining factor binding. Unexpectedly, a substantial fraction of strain-specific factor binding could not be explained by local mutations. Integration of genomic features with chromatin interaction data provided evidence for hundreds of connected cis-regulatory domains associated with differences in transcription factor binding and gene expression. This system and the >250 datasets establish a substantial new resource for investigation of how genetic variation affects cellular phenotypes.


Assuntos
Variação Genética , Macrófagos/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Células da Medula Óssea/citologia , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Análise por Conglomerados , Elementos Facilitadores Genéticos/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética
6.
J Clin Invest ; 127(9): 3220-3229, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28758903

RESUMO

Microglia are the main resident macrophage population of the CNS and perform numerous functions required for CNS development, homeostasis, immunity, and repair. Many lines of evidence also indicate that dysregulation of microglia contributes to the pathogenesis of neurodegenerative and behavioral diseases. These observations provide a compelling argument to more clearly define the mechanisms that control microglia identity and function in health and disease. In this Review, we present a conceptual framework for how different classes of transcription factors interact to select and activate regulatory elements that control microglia development and their responses to internal and external signals. We then describe functions of specific transcription factors in normal and pathological contexts and conclude with a consideration of open questions to be addressed in the future.


Assuntos
Sistema Nervoso Central/citologia , Microglia/fisiologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Anti-Inflamatórios , Colesterol/metabolismo , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Variação Genética , Homeostase , Humanos , Sistema Imunitário , Inflamação , Metabolismo dos Lipídeos , Macrófagos/citologia , Camundongos , Fenótipo , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Peixe-Zebra
7.
Science ; 356(6344)2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28546318

RESUMO

Microglia play essential roles in central nervous system (CNS) homeostasis and influence diverse aspects of neuronal function. However, the transcriptional mechanisms that specify human microglia phenotypes are largely unknown. We examined the transcriptomes and epigenetic landscapes of human microglia isolated from surgically resected brain tissue ex vivo and after transition to an in vitro environment. Transfer to a tissue culture environment resulted in rapid and extensive down-regulation of microglia-specific genes that were induced in primitive mouse macrophages after migration into the fetal brain. Substantial subsets of these genes exhibited altered expression in neurodegenerative and behavioral diseases and were associated with noncoding risk variants. These findings reveal an environment-dependent transcriptional network specifying microglia-specific programs of gene expression and facilitate efforts to understand the roles of microglia in human brain diseases.


Assuntos
Meio Ambiente , Redes Reguladoras de Genes/fisiologia , Microglia/citologia , Microglia/fisiologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatologia , Células Cultivadas , Epilepsia/genética , Epilepsia/fisiopatologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
Neurobiol Aging ; 55: 115-122, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28434692

RESUMO

Alzheimer's disease (AD) is strongly associated with microglia-induced neuroinflammation. Particularly, Aß plaque-associated microglia take on an "activated" morphology. However, the function and phenotype of these Aß plaque-associated microglia are not well understood. We show hyperreactivity of Aß plaque-associated microglia upon systemic inflammation in transgenic AD mouse models (i.e., 5XFAD and APP23). Gene expression profiling of Aß plaque-associated microglia (major histocompatibility complex II+ microglia) isolated from 5XFAD mice revealed a proinflammatory phenotype. The upregulated genes involved in the biological processes (gene ontology terms) included: "immune response to external stimulus" such as Axl, Cd63, Egr2, and Lgals3, "cell motility", such as Ccl3, Ccl4, Cxcr4, and Sdc3, "cell differentiation", and "system development", such as St14, Trpm1, and Spp1. In human AD tissue with similar Braak stages, expression of phagocytic markers and AD-associated genes, including HLA-DRA, APOE, AXL, TREM2, and TYROBP, was higher in laser-captured early-onset AD (EOAD) plaques than in late-onset AD plaques. Interestingly, the nonplaque parenchyma of both EOAD and late-onset AD brains, the expression of above-mentioned markers were similarly low. Here, we provide evidence that Aß plaque-associated microglia are hyperreactive in their immune response and phagocytosis in the transgenic AD mice as well as in EOAD brain tissue. We suggest that Aß plaque-associated microglia are the primary source of neuroinflammation related to AD pathology.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Microglia/imunologia , Placa Amiloide/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Animais , Apolipoproteínas E , Encéfalo/imunologia , Diferenciação Celular/genética , Movimento Celular/genética , Movimento Celular/imunologia , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Inflamação/genética , Inflamação/imunologia , Masculino , Glicoproteínas de Membrana , Camundongos Transgênicos , Pessoa de Meia-Idade , Fagocitose/genética , Fagocitose/imunologia , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases , Receptores Imunológicos , Receptor Tirosina Quinase Axl
9.
Glia ; 65(3): 460-473, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28063173

RESUMO

The glial stress protein alpha B-crystallin (HSPB5) is an endogenous agonist for Toll-like receptor 2 in CD14+ cells. Following systemic administration, HSPB5 acts as a potent inhibitor of neuroinflammation in animal models and reduces lesion development in multiple sclerosis patients. Here, we show that systemically administered HSPB5 rapidly crosses the blood-brain barrier, implicating microglia as additional targets for HSPB5 along with peripheral monocytes and macrophages. To compare key players in the HSPB5-induced protective response of human macrophages and microglia, we applied weighted gene co-expression network analysis on transcript expression data obtained 1 and 4 h after activation. This approach identified networks of genes that are co-expressed in all datasets, thus reducing the complexity of the nonsynchronous waves of transcripts that appear after activation by HSPB5. In both cell types, HSPB5 activates a network of highly connected genes that appear to be functionally equivalent and consistent with the therapeutic effects of HSPB5 in vivo, since both networks include factors that suppress apoptosis, the production of proinflammatory factors, and the development of adaptive immunity. Yet, hub genes at the core of the network in either cell type were strikingly different. They prominently feature the well-known tolerance-promoting programmed-death ligand 1 as a key player in the macrophage response to HSPB5, and the immune-regulatory enzyme cyclooxygenase-2 (COX-2) in that of microglia. This latter finding indicates that despite its reputation as a potential target for nonsteroidal anti-inflammatory drugs, microglial COX-2 plays a central role in the therapeutic effects of HSPB5 during neuroinflammation. GLIA 2017;65:460-473.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Cadeia B de alfa-Cristalina/farmacologia , Animais , Encéfalo/citologia , Células Cultivadas , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tecido Parenquimatoso/citologia , Tecido Parenquimatoso/efeitos dos fármacos , RNA Mensageiro/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Cadeia B de alfa-Cristalina/metabolismo
10.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 395-405, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27838490

RESUMO

Aging is the key risk factor for Alzheimer's disease (AD). In addition, the amyloid-beta (Aß) peptide is considered a critical neurotoxic agent in AD pathology. However, the connection between these factors is unclear. We aimed to provide an extensive characterization of the gene expression profiles of the amyloidosis APP23 model for AD and control mice and to evaluate the effect of aging on these profiles. We also correlated our findings to changes in soluble Aß-levels and other pathological and symptomatic features of the model. We observed a clear biphasic expression profile. The first phase displayed a maturation profile, which resembled features found in young carriers of familial AD mutations. The second phase reflected aging processes and showed similarities to the progression of human AD pathology. During this phase, the model displayed a clear upregulation of microglial activation and lysosomal pathways and downregulation of neuron differentiation and axon guidance pathways. Interestingly, the changes in expression were all correlated to aging in general, but appeared more extensive/accelerated in APP23 mice.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Citoesqueleto/patologia , Microglia/patologia , Transcriptoma , Envelhecimento , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Citoesqueleto/genética , Modelos Animais de Doenças , Redes Reguladoras de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Mutação
11.
Glia ; 65(1): 138-149, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27757989

RESUMO

Microglia are brain resident macrophages important for brain development, connectivity, homeostasis and disease. However, it is still largely unclear how microglia functions and their identity are regulated at the molecular level. Although recent transcriptomic studies have identified genes specifically expressed in microglia, the function of most of these genes in microglia is still unknown. Here, we performed RNA sequencing on microglia acutely isolated from healthy and neurodegenerative zebrafish brains. We found that a large fraction of the mouse microglial signature is conserved in the zebrafish, corroborating the use of zebrafish to help understand microglial genetics in mammals in addition to studying basic microglia biology. Second, our transcriptome analysis of microglia following neuronal ablation suggested primarily a proliferative response of microglia, which we confirmed by immunohistochemistry and in vivo imaging. Together with the recent improvements in genome editing technology in zebrafish, these data offer opportunities to facilitate functional genetic research on microglia in vivo in the healthy as well as in the diseased brain. GLIA 2016;65:138-149.


Assuntos
Microglia/citologia , Microglia/metabolismo , Transcriptoma/genética , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Morte Celular , Perfilação da Expressão Gênica/métodos , Imuno-Histoquímica/métodos , Macrófagos/citologia , Macrófagos/metabolismo , Análise de Sequência de RNA/métodos , Peixe-Zebra
12.
J Immunol ; 197(4): 1074-88, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27412414

RESUMO

EBV is the major infectious environmental risk factor for multiple sclerosis (MS), but the underlying mechanisms remain obscure. Patient studies do not allow manipulation in vivo. We used the experimental autoimmune encephalomyelitis (EAE) models in the common marmoset and rhesus monkey to model the association of EBV and MS. We report that B cells infected with EBV-related lymphocryptovirus (LCV) are requisite APCs for MHC-E-restricted autoaggressive effector memory CTLs specific for the immunodominant epitope 40-48 of myelin oligodendrocyte glycoprotein (MOG). These T cells drive the EAE pathogenesis to irreversible neurologic deficit. The aim of this study was to determine why LCV infection is important for this pathogenic role of B cells. Transcriptome comparison of LCV-infected B cells and CD20(+) spleen cells from rhesus monkeys shows increased expression of genes encoding elements of the Ag cross-presentation machinery (i.e., of proteasome maturation protein and immunoproteasome subunits) and enhanced expression of MHC-E and of costimulatory molecules (CD70 and CD80, but not CD86). It was also shown that altered expression of endolysosomal proteases (cathepsins) mitigates the fast endolysosomal degradation of the MOG40-48 core epitope. Finally, LCV infection also induced expression of LC3-II(+) cytosolic structures resembling autophagosomes, which seem to form an intracellular compartment where the MOG40-48 epitope is protected against proteolytic degradation by the endolysosomal serine protease cathepsin G. In conclusion, LCV infection induces a variety of changes in B cells that underlies the conversion of destructive processing of the immunodominant MOG40-48 epitope into productive processing and cross-presentation to strongly autoaggressive CTLs.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada/imunologia , Encefalomielite Autoimune Experimental/virologia , Infecções por Herpesviridae/imunologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Linfócitos B/virologia , Western Blotting , Callithrix , Separação Celular , Encefalomielite Autoimune Experimental/imunologia , Epitopos de Linfócito T/imunologia , Imunofluorescência , Lymphocryptovirus , Ativação Linfocitária/imunologia , Macaca mulatta , Reação em Cadeia da Polimerase , Infecções Tumorais por Vírus/imunologia
13.
Glia ; 64(4): 635-49, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26683584

RESUMO

Microglia, innate immune cells of the CNS, sense infection and damage through overlapping receptor sets. Toll-like receptor (TLR) 4 recognizes bacterial lipopolysaccharide (LPS) and multiple injury-associated factors. We show that its co-receptor CD14 serves three non-redundant functions in microglia. First, it confers an up to 100-fold higher LPS sensitivity compared to peripheral macrophages to enable efficient proinflammatory cytokine induction. Second, CD14 prevents excessive responses to massive LPS challenges via an interferon ß-mediated feedback. Third, CD14 is mandatory for microglial reactions to tissue damage-associated signals. In mice, these functions are essential for balanced CNS responses to bacterial infection, traumatic and ischemic injuries, since CD14 deficiency causes either hypo- or hyperinflammation, insufficient or exaggerated immune cell recruitment or worsened stroke outcomes. While CD14 orchestrates functions of TLR4 and related immune receptors, it is itself regulated by TLR and non-TLR systems to thereby fine-tune microglial damage-sensing capacity upon infectious and non-infectious CNS challenges.


Assuntos
Lesões Encefálicas/imunologia , Isquemia Encefálica/imunologia , Infecções por Escherichia coli/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Microglia/imunologia , Acidente Vascular Cerebral/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/patologia , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Isquemia Encefálica/patologia , Células Cultivadas , Modelos Animais de Doenças , Escherichia coli , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/patologia , Retroalimentação Fisiológica/fisiologia , Infarto da Artéria Cerebral Média , Interferon beta/metabolismo , Receptores de Lipopolissacarídeos/genética , Lipopolissacarídeos/toxicidade , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroimunomodulação , Acidente Vascular Cerebral/patologia , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
14.
Glia ; 64(2): 197-213, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26040959

RESUMO

Genome-wide expression profiling technology has resulted in detailed transcriptome data for a wide range of tissues, conditions and diseases. In neuroscience, expression datasets were mostly generated using whole brain tissue samples, resulting in data from a mixture of cell types, including glial cells and neurons. Over the past few years, a rapidly increasing number of expression profiling studies using isolated microglial cell populations have been reported. In these studies, the microglia transcriptome was compared to other cell types, such as other brain cells and peripheral tissue macrophages, and related to aging and neurodegenerative conditions. A commonality found in many of these studies was that microglia possess distinct gene expression signatures. This repertoire of selectively-expressed microglial genes highlight functions beyond immune responses, such as synaptic modulation and neurotrophic support, and open up avenues to explore as-yet-unexpected roles. These data provide improved understanding of disease pathology, and complement not only the aforementioned whole brain tissue transcriptome studies, but also genome- and epigenome-wide association studies. In this review, insights obtained from isolated microglia transcriptome studies are presented, and compared to studies using other genome-wide approaches. The relation of microglia to other tissue macrophages and glial cell populations, as well as the role of microglia in the aging brain and in neurodegenerative conditions, will be discussed. Many more of these types of studies are expected in the near future, hopefully leading to the identification of novel genes and targets for neurodegenerative conditions.


Assuntos
Perfilação da Expressão Gênica/métodos , Genômica/métodos , Microglia/metabolismo , Animais , Epigênese Genética , Humanos , Macrófagos/metabolismo , Doenças Neurodegenerativas/metabolismo
15.
Aging Cell ; 14(6): 1003-13, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26238505

RESUMO

Microglia are a proliferative population of resident brain macrophages that under physiological conditions self-renew independent of hematopoiesis. Microglia are innate immune cells actively surveying the brain and are the earliest responders to injury. During aging, microglia elicit an enhanced innate immune response also referred to as 'priming'. To date, it remains unknown whether telomere shortening affects the proliferative capacity and induces priming of microglia. We addressed this issue using early (first-generation G1 mTerc(-/-) )- and late-generation (third-generation G3 and G4 mTerc(-/-) ) telomerase-deficient mice, which carry a homozygous deletion for the telomerase RNA component gene (mTerc). Late-generation mTerc(-/-) microglia show telomere shortening and decreased proliferation efficiency. Under physiological conditions, gene expression and functionality of G3 mTerc(-/-) microglia are comparable with microglia derived from G1 mTerc(-/-) mice despite changes in morphology. However, after intraperitoneal injection of bacterial lipopolysaccharide (LPS), G3 mTerc(-/-) microglia mice show an enhanced pro-inflammatory response. Nevertheless, this enhanced inflammatory response was not accompanied by an increased expression of genes known to be associated with age-associated microglia priming. The increased inflammatory response in microglia correlates closely with increased peripheral inflammation, a loss of blood-brain barrier integrity, and infiltration of immune cells in the brain parenchyma in this mouse model of telomere shortening.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Encéfalo/fisiopatologia , Inflamação/imunologia , Microglia/imunologia , Encurtamento do Telômero/genética , Envelhecimento/imunologia , Animais , Encéfalo/citologia , Proliferação de Células/genética , Modelos Animais de Doenças , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Telomerase/genética , Telômero/genética
16.
PLoS One ; 10(2): e0116644, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25658639

RESUMO

Malignant glioma belong to the most aggressive neoplasms in humans with no successful treatment available. Patients suffering from glioblastoma multiforme (GBM), the highest-grade glioma, have an average survival time of only around one year after diagnosis. Both microglia and peripheral macrophages/monocytes accumulate within and around glioma, but fail to exert effective anti-tumor activity and even support tumor growth. Here we use microarray analysis to compare the expression profiles of glioma-associated microglia/macrophages and naive control cells. Samples were generated from CD11b+ MACS-isolated cells from naïve and GL261-implanted C57BL/6 mouse brains. Around 1000 genes were more than 2-fold up- or downregulated in glioma-associated microglia/macrophages when compared to control cells. A comparison with published data sets of M1, M2a,b,c-polarized macrophages revealed a gene expression pattern that has only partial overlap with any of the M1 or M2 gene expression patterns. Samples for the qRT-PCR validation of selected M1 and M2a,b,c-specific genes were generated from two different glioma mouse models and isolated by flow cytometry to distinguish between resident microglia and invading macrophages. We confirmed in both models the unique glioma-associated microglia/macrophage phenotype including a mixture of M1 and M2a,b,c-specific genes. To validate the expression of these genes in human we MACS-isolated CD11b+ microglia/macrophages from GBM, lower grade brain tumors and control specimens. Apart from the M1/M2 gene analysis, we demonstrate that the expression of Gpnmb and Spp1 is highly upregulated in both murine and human glioma-associated microglia/macrophages. High expression of these genes has been associated with poor prognosis in human GBM, as indicated by patient survival data linked to gene expression data. We also show that microglia/macrophages are the predominant source of these transcripts in murine and human GBM. Our findings provide new potential targets for future anti-glioma therapy.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Macrófagos/fisiologia , Glicoproteínas de Membrana/genética , Microglia/fisiologia , Osteopontina/genética , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Humanos , Macrófagos/citologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Microglia/citologia , Transplante de Neoplasias , Osteopontina/metabolismo , Prognóstico
17.
Acta Neuropathol ; 128(2): 215-29, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24997049

RESUMO

Activated microglia and macrophages play a key role in driving demyelination during multiple sclerosis (MS), but the factors responsible for their activation remain poorly understood. Here, we present evidence for a dual-trigger role of IFN-γ and alpha B-crystallin (HSPB5) in this context. In MS-affected brain tissue, accumulation of the molecular chaperone HSPB5 by stressed oligodendrocytes is a frequent event. We have shown before that this triggers a TLR2-mediated protective response in surrounding microglia, the molecular signature of which is widespread in normal-appearing brain tissue during MS. Here, we show that IFN-γ, which can be released by infiltrated T cells, changes the protective response of microglia and macrophages to HSPB5 into a robust pro-inflammatory classical response. Exposure of cultured microglia and macrophages to IFN-γ abrogated subsequent IL-10 induction by HSPB5, and strongly promoted HSPB5-triggered release of TNF-α, IL-6, IL-12, IL-1ß and reactive oxygen and nitrogen species. In addition, high levels of CXCL9, CXCL10, CXL11, several guanylate-binding proteins and the ubiquitin-like protein FAT10 were induced by combined activation with IFN-γ and HSPB5. As immunohistochemical markers for microglia and macrophages exposed to both IFN-γ and HSPB5, these latter factors were found to be selectively expressed in inflammatory infiltrates in areas of demyelination during MS. In contrast, they were absent from activated microglia in normal-appearing brain tissue. Together, our data suggest that inflammatory demyelination during MS is selectively associated with IFN-γ-induced re-programming of an otherwise protective response of microglia and macrophages to the endogenous TLR2 agonist HSPB5.


Assuntos
Interferon gama/metabolismo , Macrófagos/fisiologia , Microglia/fisiologia , Esclerose Múltipla/imunologia , Cadeia B de alfa-Cristalina/metabolismo , Encéfalo/imunologia , Encéfalo/patologia , Células Cultivadas , Quimiocina CXCL10/metabolismo , Quimiocina CXCL11/metabolismo , Quimiocina CXCL9/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Macrófagos/patologia , Microglia/patologia , Esclerose Múltipla/patologia , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitinas/metabolismo
18.
Biomaterials ; 34(3): 831-40, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23117214

RESUMO

As an extracellular protein, the small heat-shock protein alpha B-crystallin (HSPB5) has anti-inflammatory effects in several mouse models of inflammation. Here, we show that these effects are associated with the ability of HSPB5 to activate an immune-regulatory response in macrophages via endosomal/phagosomal CD14 and Toll-like receptors 1 and 2. Humans, however, possess natural antibodies against HSPB5 that block receptor binding. To protect it from these antibodies, we encapsulated HSPB5 in porous PLGA microparticles. We document here size, morphology, protein loading and release characteristics of such microparticles. Apart from effectively protecting HSPB5 from neutralization, PLGA microparticles also strongly promoted macrophage targeting of HSPB via phagocytosis. As a result, HSPB5 in porous PLGA microparticles was more than 100-fold more effective in activating macrophages than free soluble protein. Yet, the immune-regulatory nature of the macrophage response, as documented here by microarray transcript profiling, remained the same. In mice developing cigarette smoke-induced COPD, HSPB5-loaded PLGA microparticles were selectively taken up by alveolar macrophages upon intratracheal administration, and significantly suppressed lung infiltration by lymphocytes and neutrophils. In contrast, 30-fold higher doses of free soluble HSPB5 remained ineffective. Our data indicate that porous HSPB5-PLGA microparticles hold considerable promise as an anti-inflammatory biomaterial for humans.


Assuntos
Anti-Inflamatórios/administração & dosagem , Pulmão/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Pneumonia/complicações , Pneumonia/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/complicações , Cadeia B de alfa-Cristalina/administração & dosagem , Animais , Anti-Inflamatórios/imunologia , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Portadores de Fármacos/química , Proteínas de Choque Térmico Pequenas/administração & dosagem , Proteínas de Choque Térmico Pequenas/imunologia , Proteínas de Choque Térmico Pequenas/uso terapêutico , Humanos , Ácido Láctico/química , Receptores de Lipopolissacarídeos/imunologia , Pulmão/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia/imunologia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Receptor 1 Toll-Like/imunologia , Receptor 2 Toll-Like/imunologia , Cadeia B de alfa-Cristalina/imunologia , Cadeia B de alfa-Cristalina/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA