Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Front Cell Neurosci ; 17: 1178504, 2023.
Article En | MEDLINE | ID: mdl-37435046

Microglia are the resident macrophages of the central nervous system (CNS) and play a key role in CNS development, homeostasis, and disease. Good in vitro models are indispensable to study their cellular biology, and although much progress has been made, in vitro cultures of primary microglia still only partially recapitulate the transcriptome of in vivo microglia. In this study, we explored a combination of in silico and in vitro methodologies to gain insight into cues that are involved in the induction or maintenance of the ex vivo microglia reference transcriptome. First, we used the in silico tool NicheNet to investigate which (CNS-derived) cues could underlie the differences between the transcriptomes of ex vivo and in vitro microglia. Modeling on basis of gene products that were found to be upregulated in vitro, predicted that high mobility group box 2 (HMGB2)- and interleukin (IL)-1ß-associated signaling pathways were driving their expression. Modeling on basis of gene products that were found to be downregulated in vitro, did not lead to predictions on the involvement of specific signaling pathways. This is consistent with the idea that in vivo microenvironmental cues that determine microglial identity are for most part of inhibitory nature. In a second approach, primary microglia were exposed to conditioned medium from different CNS cell types. Conditioned medium from spheres composed of microglia, oligodendrocytes, and radial glia, increased the mRNA expression levels of the microglia signature gene P2RY12. NicheNet analyses of ligands expressed by oligodendrocytes and radial glia predicted transforming growth factor beta 3 (TGF-ß3) and LAMA2 as drivers of microglia signature gene expression. In a third approach, we exposed microglia to TGF-ß3 and laminin. In vitro exposure to TGF-ß3 increased the mRNA expression levels of the microglia signature gene TREM2. Microglia cultured on laminin-coated substrates were characterized by reduced mRNA expression levels of extracellular matrix-associated genes MMP3 and MMP7, and by increased mRNA expression levels of the microglia signature genes GPR34 and P2RY13. Together, our results suggest to explore inhibition of HMGB2- and IL-1ß-associated pathways in in vitro microglia. In addition, exposure to TGF-ß3 and cultivation on laminin-coated substrates are suggested as potential improvements to current in vitro microglia culture protocols.

2.
Front Immunol ; 13: 967951, 2022.
Article En | MEDLINE | ID: mdl-36203578

TLR-induced signaling initiates inflammatory responses in cells of the innate immune system. These responses are amongst others characterized by the secretion of high levels of pro-inflammatory cytokines, which are tightly regulated and adapted to the microenvironment. Purinergic receptors are powerful modulators of TLR-induced responses, and we here characterized the effects of P2Y6 receptor (P2RY6)-mediated signaling on TLR responses of rhesus macaque primary bone marrow-derived macrophages (BMDM) and microglia, using the selective P2RY6 antagonist MRS2578. We demonstrate that P2RY6-mediated signaling enhances the levels of TLR-induced pro-inflammatory cytokines in microglia in particular. TLR1, 2, 4, 5 and 8-induced responses were all enhanced in microglia, whereas such effects were much less pronounced in BMDM from the same donors. Transcriptome analysis revealed that the overall contribution of P2RY6-mediated signaling to TLR-induced responses in microglia leads to an amplification of pro-inflammatory responses. Detailed target gene analysis predicts that P2RY6-mediated signaling regulates the expression of these genes via modulation of the activity of transcription factors NFAT, IRF and NF-κB. Interestingly, we found that the expression levels of heat shock proteins were strongly induced by inhibition of P2RY6-mediated signaling, both under homeostatic conditions as well as after TLR engagement. Together, our results shed new lights on the specific pro-inflammatory contribution of P2RY6-mediated signaling in neuroinflammation, which might open novel avenues to control brain inflammatory responses.


Microglia , NF-kappa B , Animals , Cytokines/metabolism , Heat-Shock Proteins/metabolism , Macaca mulatta , NF-kappa B/metabolism , Receptors, Purinergic P2 , Toll-Like Receptor 1/metabolism
3.
Glia ; 70(4): 728-747, 2022 04.
Article En | MEDLINE | ID: mdl-34961968

Microglia are increasingly being recognized as druggable targets in neurodegenerative disorders, and good in vitro models are crucial to address cell biological questions. Major challenges are to recapitulate the complex microglial morphology and their in vivo transcriptome. We have therefore exposed primary microglia from adult rhesus macaques to a variety of different culture conditions including exposure to soluble factors as M-CSF, IL-34, and TGF-ß as well as serum replacement approaches, and compared their morphologies and transcriptomes to those of mature, homeostatic in vivo microglia. This enabled us to develop a new, partially serum-free, monoculture protocol, that yields high numbers of ramified cells. We also demonstrate that exposure of adult microglia to M-CSF or IL-34 induces similar transcriptomes, and that exposure to TGF-ß has much less pronounced effects than it does on rodent microglia. However, regardless of culture conditions, the transcriptomes of in vitro and in vivo microglia remained substantially different. Analysis of differentially expressed genes inspired us to perform 3D-spherical coculture experiments of microglia with oligodendrocytes and radial glia. In such spheres, microglia signature genes were strongly induced, even in the absence of neurons and astrocytes. These data reveal a novel role for oligodendrocyte and radial glia-derived cues in the maintenance of microglial identity, providing new anchor points to study microglia in health and disease.


Ependymoglial Cells , Microglia , Animals , Cues , Gene Expression Profiling , Macaca mulatta , Oligodendroglia , Transcriptome
4.
Front Immunol ; 12: 771453, 2021.
Article En | MEDLINE | ID: mdl-34880868

Interleukin (IL)-4 is a cytokine that affects both adaptive and innate immune responses. In the central nervous system, microglia express IL-4 receptors and it has been described that IL-4-exposed microglia acquire anti-inflammatory properties. We here demonstrate that IL-4 exposure induces changes in the cell surface protein expression profile of primary rhesus macaque microglia and enhances their potential to induce proliferation of T cells with a regulatory signature. Moreover, we show that Toll like receptor (TLR)-induced cytokine production is broadly impaired in IL-4-exposed microglia at the transcriptional level. IL-4 type 2 receptor-mediated signaling is shown to be crucial for the inhibition of microglial innate immune responses. TLR-induced nuclear translocalization of NF-κB appeared intact, and we found no evidence for epigenetic modulation of target genes. By contrast, nuclear extracts from IL-4-exposed microglia contained significantly less NF-κB capable of binding to its DNA consensus site. Further identification of the molecular mechanisms that underlie the inhibition of TLR-induced responses in IL-4-exposed microglia may aid the design of strategies that aim to modulate innate immune responses in the brain, for example in gliomas.


Cytokines/immunology , Microglia/immunology , NF-kappa B/immunology , Toll-Like Receptors/immunology , Animals , Cell Proliferation , Cells, Cultured , Female , Histone Deacetylases/genetics , Lipopolysaccharides/pharmacology , Macaca mulatta , Male , T-Lymphocytes/immunology , Transcription, Genetic
5.
Viruses ; 13(8)2021 08 23.
Article En | MEDLINE | ID: mdl-34452537

The post-acute phase of SARS-CoV-2 infection was investigated in rhesus (Macaca mulatta) and cynomolgus macaques (Macaca fascicularis). During the acute phase of infection, SARS-CoV-2 was shed via the nose and throat, and viral RNA was occasionally detected in feces. This phase coincided with a transient change in systemic immune activation. Even after the alleged resolution of the infection, computed tomography (CT) and positron emission tomography (PET)-CT revealed pulmonary lesions and activated tracheobronchial lymph nodes in all animals. Post-mortem histological examination of the lung tissue revealed mostly marginal or resolving minimal lesions that were indicative of SARS-CoV-2 infection. Evidence for SARS-CoV-2-induced histopathology was also found in extrapulmonary tissue samples, such as conjunctiva, cervical, and mesenteric lymph nodes. However, 5-6 weeks after SARS-CoV-2 exposure, upon necropsy, viral RNA was still detectable in a wide range of tissue samples in 50% of the macaques and included amongst others the heart, the respiratory tract and surrounding lymph nodes, salivary gland, and conjunctiva. Subgenomic messenger RNA was detected in the lungs and tracheobronchial lymph nodes, indicative of ongoing virus replication during the post-acute phase. These results could be relevant for understanding the long-term consequences of COVID-19 in humans.


COVID-19/pathology , COVID-19/virology , Lung/pathology , SARS-CoV-2/physiology , Animals , Antibodies, Viral/blood , COVID-19/immunology , Cytokines/blood , Disease Models, Animal , Humans , Lung/virology , Lymph Nodes/pathology , Lymph Nodes/physiopathology , Macaca fascicularis , Macaca mulatta , RNA, Messenger/analysis , RNA, Viral/analysis , Respiratory System/pathology , Respiratory System/virology , SARS-CoV-2/immunology , Virus Replication
6.
J Immunol ; 202(8): 2421-2430, 2019 04 15.
Article En | MEDLINE | ID: mdl-30804043

TLR-induced signaling potently activates cells of the innate immune system and is subject to regulation at different levels. Inflammatory conditions are associated with increased levels of extracellular adenosine, which can modulate TLR-induced production of cytokines through adenosine receptor-mediated signaling. There are four adenosine receptor subtypes that induce different signaling cascades. In this study, we demonstrate a pivotal contribution of adenosine A3 receptor (A3R)-mediated signaling to the TLR4-induced expression of IL-12 in different types of human myeloid APC. In dendritic cells, IL-12 and CCL2 responses as evoked by TLR2, 3, 4, 5, and 8, as well as IL-12 responses evoked by whole pathogens, were all reduced when A3R-mediated signaling was blocked. As a result, concomitant production of IFN-γ and IL-17 by T cells was significantly inhibited. We further show that selective inhibition of A3R-mediated signaling reduced TLR-induced phosphorylation of the transcription factor STAT1 at tyrosine 701. Next-generation sequencing revealed that A3R-mediated signaling controls the expression of metallothioneins, known inhibitors of STAT1 phosphorylation. Together our results reveal a novel regulatory layer of innate immune responses, with a central role for metallothioneins and autocrine/paracrine signaling via A3Rs.


Antigen-Presenting Cells/immunology , Chemokine CCL2/immunology , Interleukin-12/immunology , Myeloid Cells/immunology , Receptor, Adenosine A3/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Antigen-Presenting Cells/cytology , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Myeloid Cells/cytology , THP-1 Cells
7.
Glia ; 64(12): 2231-2246, 2016 12.
Article En | MEDLINE | ID: mdl-27641912

Under stressful conditions nucleotides are released from dying cells into the extracellular space, where they can bind to purinergic P2X and P2Y receptors. High concentrations of extracellular ATP in particular induce P2X7-mediated signaling, which leads to inflammasome activation. This in turn leads to the processing and secretion of pro-inflammatory cytokines, like interleukin (IL)-1ß. During neurodegenerative diseases, innate immune responses are shaped by microglia and we have previously identified microglia-specific features of inflammasome-mediated responses. Here, we compared ATP-induced IL-1ß secretion in primary rhesus macaque microglia and bone marrow-derived macrophages (BMDM). We assessed the full expression profile of P2 receptors and characterized the induction and modulation of IL-1ß secretion by extracellular nucleotides. Microglia secreted significantly lower levels of IL-1ß in response to ATP when compared to BMDM. We demonstrate that this is not due to differences in sensitivity, kinetics or expression of ATP-processing enzymes, but rather to differences in purinergic receptor expression levels and usage. Using a combined approach of purinergic receptor agonists and antagonists, we demonstrate that ATP-induced IL-1ß secretion in BMDM was fully dependent on P2X7 signaling, whereas in microglia multiple purinergic receptors were involved, including P2X7 and P2X4. These cell type-specific features of conserved innate immune responses may reflect adaptations to the vulnerable CNS microenvironment. GLIA 2016;64:2231-2246.


Adenosine Triphosphate/pharmacology , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Microglia/drug effects , Microglia/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Interleukin-6/metabolism , Lipopolysaccharide Receptors/metabolism , Macaca mulatta , Male , Polysaccharides/pharmacology , Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , RNA, Messenger/metabolism , Receptors, Purinergic P2X/genetics , Receptors, Purinergic P2X/metabolism
8.
J Neuroinflammation ; 13(1): 138, 2016 06 06.
Article En | MEDLINE | ID: mdl-27266875

BACKGROUND: Interleukin (IL)-1ß is a pro-inflammatory cytokine that plays a role in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), the animal model for MS. Yet, detailed studies on IL-1ß expression in different stages of MS lesion development and a comparison of IL-1ß expression in MS and EAE are lacking. METHODS: Here, we performed an extensive characterization of IL-1ß expression in brain tissue of MS patients, which included different MS lesion types, and in brain tissue of rhesus macaques with EAE. RESULTS: In rhesus EAE brain tissue, we observed prominent IL-1ß staining in MHC class II(+) cells within perivascular infiltrates and at the edges of large demyelinating lesions. Surprisingly, staining was localized to resident microglia or differentiated macrophages rather than to infiltrating monocytes, suggesting that IL-1ß expression is induced within the central nervous system (CNS). By contrast, IL-1ß staining in MS brain tissue was much less pronounced. Staining was found in the parenchyma of active and chronic active MS lesions and in nodules of MHC class II(+) microglia in otherwise normal appearing white matter. IL-1ß expression was detected in a minority of the nodules only, which could not be distinguished by the expression of pro- and anti-inflammatory markers. These nodules were exclusively found in MS, and it remains to be determined whether IL-1ß(+) nodules are destined to progress into active lesions or whether they merely reflect a transient response to cellular stress. CONCLUSIONS: Although the exact localization and relative intensity of IL-1ß expression in EAE and MS is different, the staining pattern in both neuroinflammatory disorders is most consistent with the idea that the expression of IL-1ß during lesion development is induced in the tissue rather than in the periphery.


Central Nervous System/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-1beta/genetics , Multiple Sclerosis/pathology , Adult , Aged , Aged, 80 and over , Animals , Calcium-Binding Proteins , Calgranulin B/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Humans , Interleukin-1beta/metabolism , Macaca mulatta , Male , Microfilament Proteins , Microglia/metabolism , Microglia/pathology , Middle Aged , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism
9.
J Neurosci ; 35(2): 678-87, 2015 Jan 14.
Article En | MEDLINE | ID: mdl-25589762

Inflammasomes are multiprotein complexes that link pathogen recognition and cellular stress to the processing of the proinflammatory cytokine interleukin-1ß (IL-1ß). Whereas inflammasome-mediated activation is heavily studied in hematopoietic macrophages and dendritic cells, much less is known about microglia, resident tissue macrophages of the brain that originate from a distinct progenitor. To directly compare inflammasome-mediated activation in different types of macrophages, we isolated primary microglia and hematopoietic macrophages from adult, healthy rhesus macaques. We analyzed the expression profile of NOD (nucleotide-binding oligomerization domain)-like receptors, adaptor proteins, and caspases and characterized inflammasome activation and regulation in detail. We here demonstrate that primary microglia can respond to the same innate stimuli as hematopoietic macrophages. However, microglial responses are more persistent due to lack of negative regulation on pro-IL-1ß expression. In addition, we show that while caspase 1, 4, and 5 activation is pivotal for inflammasome-induced IL-1ß secretion by hematopoietic macrophages, microglial secretion of IL-1ß is only partially dependent on these inflammatory caspases. These results identify key cell type-specific differences that may aid the development of strategies to modulate innate immune responses in the brain.


Caspases/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Microglia/metabolism , Animals , Caspases/genetics , Cells, Cultured , Female , Interleukin-1beta/genetics , Kinetics , Macaca mulatta , Macrophages/metabolism , Male , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Reaction Time
10.
Glia ; 60(1): 43-52, 2012 Jan.
Article En | MEDLINE | ID: mdl-21964955

Statins inhibit the endogenous intracellular mevalonate pathway and exposure to statins affects innate and adaptive immune responses. Different statins are currently under evaluation as (co)therapy in neuro-inflammatory diseases like multiple sclerosis. However, there are important discrepancies in the reported effects of statins on innate immune responses in different cell types. Studies to characterize such responses in clinically relevant primary cells are currently lacking. In this study, we investigated the effect of statins on Toll-like receptor (TLR)-induced responses of microglia, the resident macrophages of the central nervous system (CNS). Exposure of primary microglia from adult rhesus monkeys to different statins strongly amplified pro-inflammatory cytokine protein and mRNA levels in response to myeloid differentiation primary response gene 88-dependent TLR activation in particular. Rather than affecting nuclear facor-κB activation levels, statin exposure affected stress-activated protein/Jun-amino-terminal and p38 kinase signaling pathways. Mechanistic studies using specific pathway inhibitors and rescue experiments show that statin-induced inhibition of cholesterol biosynthesis, rather than inhibition of isoprenylation, was mainly responsible for the amplified TLR responses. Additionally, microglia were more sensitive to statin-mediated effects than bone marrow-derived macrophages of the same donor. This correlated to lower intrinsic microglial expression levels of 3-hydroxy-3-methylglutaryl coenzyme A reductase, the enzyme targeted by statins. Amplification of TLR-induced responses in microglia by statin exposure might contribute to the generation of a more pro-inflammatory CNS microenvironment which can be of relevance for the pathogenesis of neuroinflammatory disorders.


Anticholesteremic Agents/pharmacology , Cholesterol/biosynthesis , Heptanoic Acids/pharmacology , Microglia/drug effects , Pyrroles/pharmacology , Toll-Like Receptor 2/metabolism , Animals , Atorvastatin , Bone Marrow , Brain/cytology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Hydroxymethylglutaryl CoA Reductases/metabolism , Macaca mulatta , Macrophages/drug effects , Microglia/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , RNA, Messenger , Signal Transduction/drug effects , Signal Transduction/immunology , Toll-Like Receptor 2/immunology
11.
J Immunol ; 182(12): 7603-12, 2009 Jun 15.
Article En | MEDLINE | ID: mdl-19494284

Microglia activation is a prominent feature in many neuroinflammatory disorders. Unrestrained activation can generate a chronic inflammatory environment that might lead to neurodegeneration and autoimmunity. Extracellular adenosine modulates cellular activation through adenosine receptor (ADORA)-mediated signaling. There are four ADORA subtypes that can either increase (A(2A) and A(2B) receptors) or decrease (A(1) and A(3) receptors) intracellular cyclic AMP levels. The expression pattern of the subtypes thus orchestrates the cellular response to extracellular adenosine. We have investigated the expression of ADORA subtypes in unstimulated and TLR-activated primary rhesus monkey microglia. Activation induced an up-regulation of A(2A) and a down-regulation of A(3) receptor (A(3)R) levels. The altered ADORA-expression pattern sensitized microglia to A(2A) receptor (A(2A)R)-mediated inhibition of subsequent TLR-induced cytokine responses. By using combinations of subtype-specific agonists and antagonists, we revealed that in unstimulated microglia, A(2A)R-mediated inhibitory signaling was effectively counteracted by A(3)R-mediated signaling. In activated microglia, the decrease in A(3)R-mediated signaling sensitized them to A(2A)R-mediated inhibitory signaling. We report a differential, activation state-specific expression of ADORA in microglia and uncover a role for A(3)R as dynamically regulated suppressors of A(2A)R-mediated inhibition of TLR-induced responses. This would suggest exploration of combinations of A(2A)R agonists and A(3)R antagonists to dampen microglial activation during chronic neuroinflammatory conditions.


Microglia/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A3/metabolism , Toll-Like Receptors/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Interleukin-12/biosynthesis , Interleukin-12/immunology , Lipopolysaccharides/pharmacology , Macaca mulatta , Microglia/drug effects , Microglia/immunology , NF-kappa B/metabolism , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A3/genetics , Signal Transduction , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
12.
Glia ; 55(15): 1589-600, 2007 Nov 15.
Article En | MEDLINE | ID: mdl-17823968

Activated microglia are found in a variety of neuroinflammatory disorders where they have attributed roles as effector as well as antigen-presenting cells (APC). Critical determinants for the multifaceted role of microglia are the differentiation potential of microglia and their mode of activation. In this study, we have investigated the effects of M-CSF and GM-CSF-mediated differentiation of adult primate microglia on their cellular phenotype, antigen presentation, and phagocytic function as well as on Toll-like receptor (TLR)-mediated responses. We show that although cell morphology and expression levels of activation markers were markedly different, differentiation with either factor yielded microglia that phenotypically and functionally resemble macrophages. Both M-CSF and GM-CSF-differentiated microglia were responsive to TLR1/2, 2, 3, 4, 5, 6/2, and 8-mediated activation, but not to TLR7 or 9-mediated activation. Intriguingly, M-CSF-differentiated microglia expressed higher levels of TLR8-encoding mRNA and protein, and produced larger amounts of proinflammatory cytokines in response to TLR8-mediated activation as compared to GM-CSF-differentiated microglia. While differentiation of adult microglia by growth factors that can be produced endogenously in the central nervous system is thus unlikely to change their APC function, it can alter their innate responses to infectious stimuli such as ssRNA viruses. Resident primate microglia may thereby help shape rather than initiate adaptive immune responses.


Antigen-Presenting Cells/physiology , Microglia/physiology , Toll-Like Receptor 8/physiology , Animals , Antigen-Presenting Cells/immunology , Bone Marrow Cells/drug effects , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Proliferation , Cell Separation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Lymphocyte Culture Test, Mixed , Macaca mulatta , Macrophage Activation/physiology , Macrophage Colony-Stimulating Factor/pharmacology , Male , Microglia/immunology , Phagocytosis/drug effects , Phagocytosis/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 8/biosynthesis , Toll-Like Receptor 8/genetics
...