Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters

Publication year range
1.
J Neurosci ; 43(24): 4541-4557, 2023 06 14.
Article in English | MEDLINE | ID: mdl-37208174

ABSTRACT

Vascular endothelial cells play an important role in maintaining brain health, but their contribution to Alzheimer's disease (AD) is obscured by limited understanding of the cellular heterogeneity in normal aged brain and in disease. To address this, we performed single nucleus RNAseq on tissue from 32 human AD and non-AD donors (19 female, 13 male) each with five cortical regions: entorhinal cortex, inferior temporal gyrus, prefrontal cortex, visual association cortex, and primary visual cortex. Analysis of 51,586 endothelial cells revealed unique gene expression patterns across the five regions in non-AD donors. Alzheimer's brain endothelial cells were characterized by upregulated protein folding genes and distinct transcriptomic differences in response to amyloid ß plaques and cerebral amyloid angiopathy. This dataset demonstrates previously unrecognized regional heterogeneity in the endothelial cell transcriptome in both aged non-AD and AD brain.SIGNIFICANCE STATEMENT In this work, we show that vascular endothelial cells collected from five different brain regions display surprising variability in gene expression. In the presence of Alzheimer's disease pathology, endothelial cell gene expression is dramatically altered with clear differences in regional and temporal changes. These findings help explain why certain brain regions appear to differ in susceptibility to disease-related vascular remodeling events that may impact blood flow.


Subject(s)
Alzheimer Disease , Cerebral Amyloid Angiopathy , Male , Female , Humans , Aged , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Endothelial Cells/metabolism , Brain/metabolism , Cerebral Amyloid Angiopathy/genetics , Plaque, Amyloid/pathology , Solitary Nucleus/metabolism , Entorhinal Cortex/metabolism
2.
Acta Neuropathol ; 147(1): 65, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38557897

ABSTRACT

Human microglia are critically involved in Alzheimer's disease (AD) progression, as shown by genetic and molecular studies. However, their role in tau pathology progression in human brain has not been well described. Here, we characterized 32 human donors along progression of AD pathology, both in time-from early to late pathology-and in space-from entorhinal cortex (EC), inferior temporal gyrus (ITG), prefrontal cortex (PFC) to visual cortex (V2 and V1)-with biochemistry, immunohistochemistry, and single nuclei-RNA-sequencing, profiling a total of 337,512 brain myeloid cells, including microglia. While the majority of microglia are similar across brain regions, we identified a specific subset unique to EC which may contribute to the early tau pathology present in this region. We calculated conversion of microglia subtypes to diseased states and compared conversion patterns to those from AD animal models. Targeting genes implicated in this conversion, or their upstream/downstream pathways, could halt gene programs initiated by early tau progression. We used expression patterns of early tau progression to identify genes whose expression is reversed along spreading of spatial tau pathology (EC > ITG > PFC > V2 > V1) and identified their potential involvement in microglia subtype conversion to a diseased state. This study provides a data resource that builds on our knowledge of myeloid cell contribution to AD by defining the heterogeneity of microglia and brain macrophages during both temporal and regional pathology aspects of AD progression at an unprecedented resolution.


Subject(s)
Alzheimer Disease , Animals , Humans , Alzheimer Disease/pathology , tau Proteins/genetics , tau Proteins/metabolism , Transcriptome , Brain/pathology , Myeloid Cells/pathology , Microglia/pathology , Amyloid beta-Peptides/metabolism
3.
Alzheimers Dement ; 20(1): 74-90, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37461318

ABSTRACT

INTRODUCTION: Omics studies have revealed that various brain cell types undergo profound molecular changes in Alzheimer's disease (AD) but the spatial relationships with plaques and tangles and APOE-linked differences remain unclear. METHODS: We performed laser capture microdissection of amyloid beta (Aß) plaques, the 50 µm halo around them, tangles with the 50 µm halo around them, and areas distant (> 50 µm) from plaques and tangles in the temporal cortex of AD and control donors, followed by RNA-sequencing. RESULTS: Aß plaques exhibited upregulated microglial (neuroinflammation/phagocytosis) and downregulated neuronal (neurotransmission/energy metabolism) genes, whereas tangles had mostly downregulated neuronal genes. Aß plaques had more differentially expressed genes than tangles. We identified a gradient Aß plaque > peri-plaque > tangle > distant for these changes. AD APOE ε4 homozygotes had greater changes than APOE ε3 across locations, especially within Aß plaques. DISCUSSION: Transcriptomic changes in AD consist primarily of neuroinflammation and neuronal dysfunction, are spatially associated mainly with Aß plaques, and are exacerbated by the APOE ε4 allele.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Neurofibrillary Tangles , Apolipoprotein E4/genetics , Neuroinflammatory Diseases , Brain/metabolism , Transcriptome , Plaque, Amyloid/metabolism , Gene Expression Profiling
4.
Glia ; 70(5): 935-960, 2022 05.
Article in English | MEDLINE | ID: mdl-35092321

ABSTRACT

A key pathological process in Parkinson's disease (PD) is the transneuronal spreading of α-synuclein. Alpha-synuclein (α-syn) is a presynaptic protein that, in PD, forms pathological inclusions. Other hallmarks of PD include neurodegeneration and microgliosis in susceptible brain regions. Whether it is primarily transneuronal spreading of α-syn particles, inclusion formation, or other mechanisms, such as inflammation, that cause neurodegeneration in PD is unclear. We used a model of spreading of α-syn induced by striatal injection of α-syn preformed fibrils into the mouse striatum to address this question. We performed quantitative analysis for α-syn inclusions, neurodegeneration, and microgliosis in different brain regions, and generated gene expression profiles of the ventral midbrain, at two different timepoints after disease induction. We observed significant neurodegeneration and microgliosis in brain regions not only with, but also without α-syn inclusions. We also observed prominent microgliosis in injured brain regions that did not correlate with neurodegeneration nor with inclusion load. Using longitudinal gene expression profiling, we observed early gene expression changes, linked to neuroinflammation, that preceded neurodegeneration, indicating an active role of microglia in this process. Altered gene pathways overlapped with those typical of PD. Our observations indicate that α-syn inclusion formation is not the major driver in the early phases of PD-like neurodegeneration, but that microglia, activated by diffusible, oligomeric α-syn, may play a key role in this process. Our findings uncover new features of α-syn induced pathologies, in particular microgliosis, and point to the necessity for a broader view of the process of α-syn spreading.


Subject(s)
Parkinson Disease , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Mice , Microglia/metabolism , Neuroinflammatory Diseases , Parkinson Disease/genetics , alpha-Synuclein/genetics
5.
Acta Neuropathol ; 141(5): 681-696, 2021 05.
Article in English | MEDLINE | ID: mdl-33609158

ABSTRACT

Alzheimer's disease (AD) is the most prevalent form of dementia and is characterized by abnormal extracellular aggregates of amyloid-ß and intraneuronal hyperphosphorylated tau tangles and neuropil threads. Microglia, the tissue-resident macrophages of the central nervous system (CNS), are important for CNS homeostasis and implicated in AD pathology. In amyloid mouse models, a phagocytic/activated microglia phenotype has been identified. How increasing levels of amyloid-ß and tau pathology affect human microglia transcriptional profiles is unknown. Here, we performed snRNAseq on 482,472 nuclei from non-demented control brains and AD brains containing only amyloid-ß plaques or both amyloid-ß plaques and tau pathology. Within the microglia population, distinct expression profiles were identified of which two were AD pathology-associated. The phagocytic/activated AD1-microglia population abundance strongly correlated with tissue amyloid-ß load and localized to amyloid-ß plaques. The AD2-microglia abundance strongly correlated with tissue phospho-tau load and these microglia were more abundant in samples with overt tau pathology. This full characterization of human disease-associated microglia phenotypes provides new insights in the pathophysiological role of microglia in AD and offers new targets for microglia-state-specific therapeutic strategies.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Microglia/pathology , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Brain/metabolism , Brain/pathology , Female , Humans , Male
6.
EMBO Rep ; 19(11)2018 11.
Article in English | MEDLINE | ID: mdl-30206190

ABSTRACT

Microglia are specialized parenchymal-resident phagocytes of the central nervous system (CNS) that actively support, defend and modulate the neural environment. Dysfunctional microglial responses are thought to worsen CNS diseases; nevertheless, their impact during neuroinflammatory processes remains largely obscure. Here, using a combination of single-cell RNA sequencing and multicolour flow cytometry, we comprehensively profile microglia in the brain of lipopolysaccharide (LPS)-injected mice. By excluding the contribution of other immune CNS-resident and peripheral cells, we show that microglia isolated from LPS-injected mice display a global downregulation of their homeostatic signature together with an upregulation of inflammatory genes. Notably, we identify distinct microglial activated profiles under inflammatory conditions, which greatly differ from neurodegenerative disease-associated profiles. These results provide insights into microglial heterogeneity and establish a resource for the identification of specific phenotypes in CNS disorders, such as neuroinflammatory and neurodegenerative diseases.


Subject(s)
Inflammation/pathology , Microglia/metabolism , Single-Cell Analysis/methods , Animals , CD11b Antigen/metabolism , Encephalitis/genetics , Encephalitis/metabolism , Encephalitis/pathology , Female , Flow Cytometry/methods , Gene Expression Regulation , Homeostasis , Inflammation/genetics , Inflammation/metabolism , Leukocyte Common Antigens/metabolism , Lipopolysaccharides/toxicity , Male , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Neurodegenerative Diseases/pathology , Sequence Analysis, RNA/methods
7.
J Neurochem ; 151(1): 11-27, 2019 10.
Article in English | MEDLINE | ID: mdl-31361031

ABSTRACT

Adenosine receptor subtypes, first described 40 years ago, are known to regulate diverse biological functions and have a role in various conditions, such as cerebral and cardiac ischemia, immune and inflammatory disorders and cancer. In the brain, they limit potentially dangerous over excitation, but also regulate mechanisms essential in sleep and psychiatric disorders. In this review, we discuss the role of adenosine receptors in mood and anxiety disorders. Activation of A2A receptors is associated with increased depression-like symptoms, while increased A1 receptors signaling elicits rapid antidepressant effects. Indeed, several lines of evidence demonstrate that the therapeutic effects of different non-pharmacological treatments of depression, like sleep deprivation and electroconvulsive therapy are mediated by A1 receptor up-regulation or activation. In addition, A1 receptors may also play a role in the antidepressant effects of transcranial direct current stimulation and deep brain stimulation. As a potential downstream mechanism, which facilitates the antidepressant effects of A1 receptors, we propose a crosstalk between adenosinergic and glutamatergic systems mediated via synaptic plasticity protein Homer1a and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. Moreover, adenosine receptors are also involved in the control of circadian rhythms, sleep homeostasis and some neuro-immunological mechanisms, all of them implicated in mood regulation. Antagonists of adenosine receptors such as caffeine have general anxiogenic effects. In particular, A2A receptors appear to have an important role in the pathophysiology of anxiety disorders. Taken together, the results discussed here indicate that the adenosinergic system is involved in both the etiology and the treatment of mood and anxiety disorders.


Subject(s)
Anxiety Disorders/metabolism , Mood Disorders/metabolism , Receptors, Purinergic P1/metabolism , Animals , Humans
8.
EMBO J ; 34(12): 1612-29, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25896511

ABSTRACT

Microglia are tissue macrophages of the central nervous system (CNS) that control tissue homeostasis. Microglia dysregulation is thought to be causal for a group of neuropsychiatric, neurodegenerative and neuroinflammatory diseases, called "microgliopathies". However, how the intracellular stimulation machinery in microglia is controlled is poorly understood. Here, we identified the ubiquitin-specific protease (Usp) 18 in white matter microglia that essentially contributes to microglial quiescence. We further found that microglial Usp18 negatively regulates the activation of Stat1 and concomitant induction of interferon-induced genes, thereby terminating IFN signaling. The Usp18-mediated control was independent from its catalytic activity but instead required the interaction with Ifnar2. Additionally, the absence of Ifnar1 restored microglial activation, indicating a tonic IFN signal which needs to be negatively controlled by Usp18 under non-diseased conditions. These results identify Usp18 as a critical negative regulator of microglia activation and demonstrate a protective role of Usp18 for microglia function by regulating the Ifnar pathway. The findings establish Usp18 as a new molecule preventing destructive microgliopathy.


Subject(s)
Brain/metabolism , Endopeptidases/deficiency , Interferons/metabolism , Microglia/metabolism , Models, Neurological , Signal Transduction/physiology , Animals , Blotting, Western , Cloning, Molecular , DNA Primers/genetics , Endopeptidases/genetics , Endopeptidases/metabolism , Histological Techniques , Mice , Mice, Knockout , Microarray Analysis , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Statistics, Nonparametric , Ubiquitin Thiolesterase
9.
Glia ; 66(4): 708-724, 2018 04.
Article in English | MEDLINE | ID: mdl-29226424

ABSTRACT

Brain inflammation is a critical factor involved in neurodegeneration. Recently, the prostaglandin E2 (PGE2 ) downstream members were suggested to modulate neuroinflammatory responses accompanying neurodegenerative diseases. In this study, we investigated the protective effects of prostaglandin E2 receptor 2 (EP2 ) during TLR3 and TLR4-driven inflammatory response using in vitro primary microglia and ex vivo organotypic hippocampal slice cultures (OHSCs). Depletion of microglia from OHSCs differentially affected TLR3 and TLR4 receptor expression. Poly(I:C) induced the production of prostaglandin E2 in OHSCs by increasing cyclooxygenase (COX-2) and microsomal prostaglandin E synthase (mPGES)-1. Besides, stimulation of OHSCs and microglia with Poly(I:C) upregulated EP2 receptor expression. Co-stimulation of OHSCs and microglia with the EP2 agonist butaprost reduced inflammatory mediators induced by LPS and Poly(I:C). In Poly(I:C) challenged OHSCs, butaprost almost restored microglia ramified morphology and reduced Iba1 immunoreactivity. Importantly, microglia depletion prevented the induction of inflammatory mediators following Poly(I:C) or LPS challenge in OHSCs. Activation of EP2 receptor reversed the Poly(I:C)/LPS-induced phosphorylation of the mitogen activated protein kinases (MAPKs) ERK, p38 MAPK and c-Jun N-terminal kinase (JNK) in microglia. Collectively, these data identify an anti-inflammatory function for EP2 signaling in diverse innate immune responses, through a mechanism that involves the mitogen-activated protein kinases pathway.


Subject(s)
Hippocampus/immunology , Inflammation/metabolism , Microglia/immunology , Mitogen-Activated Protein Kinases/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/immunology , Cerebral Cortex/pathology , Hippocampus/drug effects , Hippocampus/pathology , Immunity, Innate/physiology , Immunologic Factors/pharmacology , Inflammation/drug therapy , Inflammation/pathology , Lipopolysaccharides , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/pathology , Poly I-C , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Prostaglandin E, EP2 Subtype/agonists , Tissue Culture Techniques , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism
10.
Genet Res (Camb) ; 99: e1, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28132660

ABSTRACT

Glutamate is the most important excitatory neurotransmitter in the brain. The N-methyl-D-aspartate (NMDA) receptor is a glutamate-gated ionotropic cation channel that is composed of several subunits and modulated by a glycine binding site. Many forms of synaptic plasticity depend on the influx of calcium ions through NMDA receptors, and NMDA receptor dysfunction has been linked to a number of neuropsychiatric disorders, including schizophrenia. Whole-exome sequencing was performed in a family with a strong history of psychotic disorders over three generations. We used an iterative strategy to obtain condense and meaningful variants. In this highly affected family, we found a frameshift mutation (rs10666583) in the GRIN3B gene, which codes for the GluN3B subunit of the NMDA receptor in all family members with a psychotic disorder, but not in the healthy relatives. Matsuno et al., also reported this null variant as a risk factor for schizophrenia in 2015. In a broader sample of 22 patients with psychosis, the allele frequency of the rs10666583 mutation variant was increased compared to those of healthy population samples and unaffected relatives. Compared to the 1000 Genomes Project population, we found a significant increase of this variant with a large effect size among patients. The amino acid shift degrades the S1/S2 glycine binding domain of the dominant modulatory GluN3B subunit of the NMDA receptor, which subsequently affects the permeability of the channel pore to calcium ions. A decreased glycine affinity for the GluN3B subunit might cause impaired functional capability of the NMDA receptor and could be an important risk factor for the pathogenesis of psychotic disorders.


Subject(s)
Exome/genetics , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing/methods , Mutation, Missense/genetics , Psychotic Disorders/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/genetics , Adult , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Pedigree , Risk Factors , Young Adult
11.
Brain Behav Immun ; 62: 362-381, 2017 May.
Article in English | MEDLINE | ID: mdl-28088641

ABSTRACT

Delirium is a frequent outcome for aged and demented patients that suffer a systemic inflammatory insult. Animal models that reconstruct these etiological processes have potential to provide a better understanding of the pathophysiology of delirium. Therefore, we systematically reviewed animal studies in which systemic inflammation was superimposed on aged or diseased animal models. In total, 77 studies were identified. Aged animals were challenged with a bacterial endotoxin in 29 studies, 25 studies superimposed surgery on aged animals, and in 6 studies a bacterial infection, Escherichia coli (E. coli), was used. Diseased animals were challenged with a bacterial endotoxin in 15 studies, two studies examined effects of the cytokine IL-1ß, and one study used polyinosinic:polycytidilic acid (poly I:C). This systematic review analyzed the impact of systemic inflammation on the production of inflammatory and neurotoxic mediators in peripheral blood, cerebrospinal fluid (CSF), and on the central nervous system (CNS). Moreover, concomitant behavioral and cognitive symptoms were also evaluated. Finally, outcomes of behavioral and cognitive tests from animal studies were compared to features and symptoms present in delirious patients.


Subject(s)
Behavior, Animal/physiology , Delirium/psychology , Inflammation/psychology , Animals , Delirium/immunology , Disease Models, Animal , Inflammation/immunology , Mice , Rats
12.
Glia ; 64(10): 1772-87, 2016 10.
Article in English | MEDLINE | ID: mdl-27219534

ABSTRACT

Based on promising preclinical evidence, microglial P2X7 has increasingly being recognized as a target for therapeutic intervention in neurological and psychiatric diseases. However, despite this knowledge no P2X7-related drug has yet entered clinical trials with respect to CNS diseases. We here discuss the current literature on P2X7 being a drug target and identify unsolved issues and still open questions that have hampered the development of P2X7 dependent therapeutic approaches for CNS diseases. It is concluded here that the lack of brain penetrating P2X7 antagonists is a major obstacle in the field and that central P2X7 is a yet untested clinical drug target. In the CNS, microglial P2X7 activation causes neuroinflammation, which in turn plays a role in various CNS disorders. This has resulted in a surge of brain penetrant P2X7 antagonists. P2X7 is a viable, clinically untested CNS drug target. GLIA 2016;64:1772-1787.


Subject(s)
Central Nervous System Diseases/pathology , Microglia/metabolism , Purinergic Agents/therapeutic use , Receptors, Purinergic P2X7/metabolism , Animals , Central Nervous System Diseases/drug therapy , Humans , Microglia/drug effects , Purinergic Agents/pharmacology
13.
Glia ; 64(1): 76-89, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26295445

ABSTRACT

Recently, we have demonstrated that ramified microglia are neuroprotective in N-methyl-D-aspartate (NMDA)-induced excitotoxicity in organotypic hippocampal slice cultures (OHSCs). The present study aimed to elucidate the underlying neuron-glia communication mechanism. It is shown here that pretreatment of OHSC with high concentrations of adenosine 5'-triphosphate (ATP) reduced NMDA-induced neuronal death only in presence of microglia. Specific agonists and antagonists identified the P2X7 receptor as neuroprotective receptor which was confirmed by absence of ATP-dependent neuroprotection in P2X7-deficient OHSC. Microglia replenished chimeric OHSC consisting of wild-type tissue replenished with P2X7-deficient microglia confirmed the involvement of microglial P2X7 receptor in neuroprotection. Stimulation of P2X7 in primary microglia induced tumor necrosis factor α (TNFα) release and blocking TNFα by a neutralizing antibody in OHSC abolished neuroprotection by ATP. OHSC from TNFα-deficient mice show increased exicitoxicity and activation of P2X7 did not rescue neuronal survival in the absence of TNFα. The neuroprotective effect of valproic acid (VPA) was strictly dependent on the presence of microglia and was mediated by upregulation of P2X7 in the cells. The present study demonstrates that microglia-mediated neuroprotection depends on ATP-activated purine receptor P2X7 and induction of TNFα release. This neuroprotective pathway was strengthened by VPA elucidating a novel mechanism for the neuroprotective function of VPA.


Subject(s)
Microglia/physiology , Neurons/physiology , Neuroprotective Agents/pharmacology , Receptors, Purinergic P2X7/metabolism , Tumor Necrosis Factor-alpha/metabolism , Valproic Acid/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiology , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/pathology , N-Methylaspartate/metabolism , N-Methylaspartate/toxicity , Neurons/drug effects , Neurons/pathology , RNA, Messenger/metabolism , Receptors, Purinergic P2X7/genetics , Tissue Culture Techniques , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects
14.
Glia ; 64(8): 1285-97, 2016 08.
Article in English | MEDLINE | ID: mdl-27145902

ABSTRACT

Recent data suggest that ramified microglia fulfil various tasks in the brain. However, to investigate this unique cell type cultured primary microglia are only a poor model. We here describe a method to deplete and repopulate organotypic hippocampal slice cultures (OHSC) with ramified microglia isolated from adult mouse brain creating microglia-replenished OHSC (Mrep-OHSC). Replenished microglia integrate into the tissue and ramify to a degree indistinguishable from their counterparts in the mouse brain. Moreover, wild-type slices replenished with microglia from TNFα-deficient animals provide similar results as OHSC prepared from microglia-specific TNFα-knockout mice (CX3CR1(cre) /TNFα(fl/fl) ). Furthermore, this study demonstrates that replenished microglia in OHSC maintain original functions and properties acquired in vivo. Microglia from ERCC1(Δ/ko) mice, a mouse model of accelerated aging, maintain enhanced Mac2 expression and their activated phenotype after replenishment to wild-type OHSC tissue. Thus, the present study demonstrates that Mrep-OHSC are a unique tool to construct chimeric brain slices allowing studying the function of different phenotypes of in vivo like microglia in a tissue culture setting. GLIA 2016 GLIA 2016;64:1285-1297.


Subject(s)
Hippocampus/physiology , Microglia/physiology , Tissue Culture Techniques , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Galectin 3/metabolism , Hippocampus/cytology , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microscopy, Confocal , Neuroprotection/physiology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
15.
Glia ; 64(10): 1788-94, 2016 10.
Article in English | MEDLINE | ID: mdl-27246804

ABSTRACT

Minocycline, a second generation broad-spectrum antibiotic, has been frequently postulated to be a "microglia inhibitor." A considerable number of publications have used minocycline as a tool and concluded, after achieving a pharmacological effect, that the effect must be due to "inhibition" of microglia. It is, however, unclear how this "inhibition" is achieved at the molecular and cellular levels. Here, we weigh the evidence whether minocycline is indeed a bona fide microglia inhibitor and discuss how data generated with minocycline should be interpreted. GLIA 2016;64:1788-1794.


Subject(s)
Anti-Bacterial Agents/pharmacology , Microglia/drug effects , Minocycline/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Databases, Factual/statistics & numerical data , Humans , Microglia/physiology , Minocycline/therapeutic use
16.
EMBO J ; 36(5): 565-567, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28213481
17.
Biol Chem ; 397(3): 207-14, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26641965

ABSTRACT

Homer1a is upregulated by several different antidepressant measures, including non-pharmacological treatments, like sleep deprivation (SD) and electroconvulsive therapy (ECT) and antidepressant drugs, such as imipramine, fluoxetine and ketamine. Homer1a induction might thus be a crucial joint mechanism for antidepressant therapy in general. However, the upstream signaling pathways that regulate or induce Homer1a expression are still not well understood. The main focus of the present review is to offer an overview of the current knowledge about the potential role of Homer1a in depression and the signaling pathways responsible for Homer1a regulation. It is suggested here that a detailed characterization of the signaling mechanisms leading to Homer1a expression might provide novel therapeutic targets for antidepressant drug development.


Subject(s)
Carrier Proteins/genetics , Depression/metabolism , Gene Expression Regulation , Signal Transduction , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Carrier Proteins/metabolism , Depression/drug therapy , Depression/genetics , Depressive Disorder/drug therapy , Depressive Disorder/genetics , Depressive Disorder/metabolism , Gene Expression Regulation/drug effects , Homer Scaffolding Proteins , Humans , Receptor, Adenosine A1/metabolism , Signal Transduction/drug effects , ras Proteins/metabolism
18.
Brain Behav Immun ; 55: 126-137, 2016 07.
Article in English | MEDLINE | ID: mdl-26576722

ABSTRACT

Microglia are suggested to be involved in several neuropsychiatric diseases. Indeed changes in microglia morphology have been reported in different mouse models of depression. A crucial regulatory system for microglia function is the well-defined CX3C axis. Thus, we aimed to clarify the role of microglia and CX3CR1 in depressive behavior by subjecting CX3CR1-deficient mice to a particular chronic despair model (CDM) paradigm known to exhibit face validity to major depressive disorder. In wild-type mice we observed the development of chronic depressive-like behavior after 5days of repetitive swim stress. 3D-reconstructions of Iba-1-labeled microglia in the dentate molecular layer revealed that behavioral effects were associated with changes in microglia morphology towards a state of hyper-ramification. Chronic treatment with the anti-depressant venlafaxine ameliorated depression-like behavior and restored microglia morphology. In contrast, CX3CR1 deficient mice showed a clear resistance to either (i) stress-induced depressive-like behavior, (ii) changes in microglia morphology and (iii) antidepressant treatment. Our data point towards a role of hyper-ramified microglia in the etiology of chronic depression. The lack of effects in CX3CR1 deficient mice suggests that microglia hyper-ramification is controlled by neuron-microglia signaling via the CX3C axis. However, it remains to be elucidated how hyper-ramified microglia contribute to depressive-like behavior.


Subject(s)
Behavior, Animal/physiology , Chemokine CX3CL1/deficiency , Depressive Disorder, Major/physiopathology , Microglia/pathology , Stress, Psychological/complications , Animals , Depressive Disorder, Major/etiology , Disease Models, Animal , Mice , Mice, Inbred C57BL
19.
Glia ; 63(4): 611-25, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25471735

ABSTRACT

The brain's immune privilege has been also attributed to the lack of dendritic cells (DC) within its parenchyma and the adjacent meninges, an assumption, which implies maintenance of antigens rather than their presentation in lymphoid organs. Using mice transcribing the green fluorescent protein under the promoter of the DC marker CD11c (itgax), we identified a juxtavascular population of cells expressing this DC marker and demonstrated their origin from bone marrow and local microglia. We now phenotypically compared this population with CD11c/CD45 double-positive cells from lung, liver, and spleen in healthy mice using seven-color flow cytometry. We identified unique, site-specific expression patterns of F4/80, CD80, CD86, CX3CR1, CCR2, FLT3, CD103, and MHC-II. Furthermore, we observed the two known CD45-positive populations (CD45(high) and CD45(int) ) in the brain, whereas liver, lung, and spleen exhibited a homogeneous CD45(high) population. CD11c-positive microglia lacked MHC-II expression and CD45(high) /CD11c-positive cells from the brain have a lower percentage of MHC-II-positive cells. To test whether phenotypical differences are fixed by origin or specifically develop due to environmental factors, we transplanted brain and spleen mononuclear cells on organotypic slice cultures from brain (OHSC) and spleen (OSSC). We demonstrate that adaption and ramification of MHC-II-positive splenocytes is paralleled by down-regulation of MHC-II, whereas brain-derived mononuclear cells neither ramified nor up-regulated MHC-II in OSSCs. Thus, brain-derived mononuclear cells maintain their MHC-II-negative phenotype within the environment of an immune organ. Intraparenchymal CD11c-positive cells share immunophenotypical characteristics of DCs from other organs but remain unique for their low MHC-II expression.


Subject(s)
Brain/cytology , CD11c Antigen/metabolism , Dendritic Cells/metabolism , Liver/cytology , Lung/cytology , Microglia/metabolism , Spleen/cytology , Animals , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CX3C Chemokine Receptor 1 , Flow Cytometry , Genes, MHC Class II/genetics , Green Fluorescent Proteins , Integrin alpha Chains/metabolism , Leukocyte Common Antigens/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, CCR2/metabolism , Receptors, Chemokine/metabolism , fms-Like Tyrosine Kinase 3/metabolism
20.
EMBO J ; 30(9): 1864-73, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21441897

ABSTRACT

Up-regulation of P2X4 receptors in spinal cord microglia is crucial for tactile allodynia, an untreatable pathological pain reaction occurring after peripheral nerve injury. How nerve injury in the periphery leads to this microglia reaction in the dorsal horn of the spinal cord is not yet understood. It is shown here that CCL21 was rapidly expressed in injured small-sized primary sensory neurons and transported to their central terminals in the dorsal horn. Intrathecal administration of a CCL21-blocking antibody diminished tactile allodynia development in wild-type animals. Mice deficient for CCL21 did not develop any signs of tactile allodynia and failed to up-regulate microglial P2X4 receptor expression. Microglia P2X4 expression was enhanced by CCL21 application in vitro and in vivo. A single intrathecal injection of CCL21 to nerve-injured CCL21-deficient mice induced long-lasting allodynia that was undistinguishable from the wild-type response. This effect of CCL21 injection was strictly dependent on P2X4 receptor function. Since neuronal CCL21 is the earliest yet identified factor in the cascade leading to tactile allodynia, these findings may lead to a preventive therapy in neuropathic pain.


Subject(s)
Chemokine CCL21/metabolism , Hyperalgesia/metabolism , Microglia/metabolism , Neuralgia/metabolism , Neurons/metabolism , Receptors, Purinergic P2X4/metabolism , Spinal Nerves/injuries , Analysis of Variance , Animals , Blotting, Western , DNA Primers/genetics , Gene Expression Regulation/physiology , Hyperalgesia/pathology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neuralgia/genetics , Pain Measurement , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL