Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Science ; 363(6425)2019 01 25.
Article in English | MEDLINE | ID: mdl-30679343

ABSTRACT

The innate immune cell compartment is highly diverse in the healthy central nervous system (CNS), including parenchymal and non-parenchymal macrophages. However, this complexity is increased in inflammatory settings by the recruitment of circulating myeloid cells. It is unclear which disease-specific myeloid subsets exist and what their transcriptional profiles and dynamics during CNS pathology are. Combining deep single-cell transcriptome analysis, fate mapping, in vivo imaging, clonal analysis, and transgenic mouse lines, we comprehensively characterized unappreciated myeloid subsets in several CNS compartments during neuroinflammation. During inflammation, CNS macrophage subsets undergo self-renewal, and random proliferation shifts toward clonal expansion. Last, functional studies demonstrated that endogenous CNS tissue macrophages are redundant for antigen presentation. Our results highlight myeloid cell diversity and provide insights into the brain's innate immune system.


Subject(s)
Central Nervous System/immunology , Immunity, Innate , Inflammation/immunology , Macrophages/cytology , Myeloid Cells/cytology , Animals , Antigen Presentation , Brain/immunology , Dendritic Cells/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , Histocompatibility Antigens Class II/immunology , Homeostasis , Macrophages/immunology , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/cytology , Myeloid Cells/immunology , Sequence Analysis, RNA , Single-Cell Analysis , T-Lymphocytes/immunology
2.
Front Immunol ; 9: 836, 2018.
Article in English | MEDLINE | ID: mdl-29770132

ABSTRACT

gp130 cytokines are differentially involved in regulating the T helper (H) 17-driven pathogenesis of experimental autoimmune encephalomyelitis (EAE), the animal model of human multiple sclerosis. Interleukin (IL)-6 directly promotes the development of TH17 cells through the gp130/IL-6R complex. By contrast, IL-27 has been shown to suppress a TH17 immune response by gp130/IL-27R-alpha (α) receptor ligation. The IL-27-dependent regulation of a TH17 development could be mediated on the level of CD4 T cells. However, because IL-27 also suppresses the secretion of the TH17-driving cytokines IL-6 and IL-12/23p40 in accessory cells, TH17 immune responses may also be controlled by IL-27 on the level of macrophages and/or neutrophils. To analyze these opposing effects of gp130 engagement on the pathogenesis of EAE, we immunized CD4+ T cell-specific gp130-deficient (CD4creposgp130loxP/loxP) and macrophage/neutrophil-specific gp130-deficient (LysMcreposgp130loxP/loxP) mice with the myelin-oligodendrocyte-glycoprotein peptide MOG35-55. Whereas inflammatory immune responses, TH17 differentiation, and pathology in CD4creposgp130loxP/loxP mice were mitigated, disease progression was eventually enhanced in LysMcreposgp130loxP/loxP mice. Exacerbated disease in MOG35-55-immunized LysMcreposgp130loxP/loxP mice was associated with an elevated development of TH17 cells and increased infiltration of the central nervous system with leukocytes indicating a suppressive role of macrophage/neutrophil-gp130. To further prove IL-6 to be responsible for the control of inflammation during EAE through gp130 on macrophages/neutrophils, we immunized LysMcreposIL-6RloxP/loxP mice. In contrast to LysMcreposgp130loxP/loxP mice, neuropathology in MOG35-55-immunized macrophage/neutrophil-specific IL-6R-deficient mice was not enhanced indicating that the alleviation of EAE through macrophage/neutrophil-gp130 is mediated independently of IL-6. Together, this different pathology in macrophage/neutrophil- and CD4 T cell-specific gp130-deficient mice suggests that gp130 cytokines modulate TH17 inflammation differentially by targeting distinct cell types.


Subject(s)
Cytokine Receptor gp130/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Macrophages/immunology , Neutrophils/immunology , Th17 Cells/immunology , Animals , Cell Differentiation , Central Nervous System/immunology , Cytokine Receptor gp130/deficiency , Cytokines/immunology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation , Mice , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Th1 Cells/immunology
3.
Immunity ; 44(4): 901-12, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27096319

ABSTRACT

Sickness behavior and cognitive dysfunction occur frequently by unknown mechanisms in virus-infected individuals with malignancies treated with type I interferons (IFNs) and in patients with autoimmune disorders. We found that during sickness behavior, single-stranded RNA viruses, double-stranded RNA ligands, and IFNs shared pathways involving engagement of melanoma differentiation-associated protein 5 (MDA5), retinoic acid-inducible gene 1 (RIG-I), and mitochondrial antiviral signaling protein (MAVS), and subsequently induced IFN responses specifically in brain endothelia and epithelia of mice. Behavioral alterations were specifically dependent on brain endothelial and epithelial IFN receptor chain 1 (IFNAR). Using gene profiling, we identified that the endothelia-derived chemokine ligand CXCL10 mediated behavioral changes through impairment of synaptic plasticity. These results identified brain endothelial and epithelial cells as natural gatekeepers for virus-induced sickness behavior, demonstrated tissue specific IFNAR engagement, and established the CXCL10-CXCR3 axis as target for the treatment of behavioral changes during virus infection and type I IFN therapy.


Subject(s)
Brain/cytology , Chemokine CXCL10/immunology , Cognition Disorders/genetics , Endothelial Cells/immunology , Epithelial Cells/immunology , Illness Behavior/physiology , Receptor, Interferon alpha-beta/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/immunology , Cell Communication/immunology , Cells, Cultured , Cognition Disorders/psychology , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Endothelium/cytology , Endothelium/immunology , Epithelium/immunology , Interferon Type I/therapeutic use , Interferon-Induced Helicase, IFIH1 , Male , Mice , RNA, Double-Stranded/genetics , Receptor, Interferon alpha-beta/immunology , Receptors, CXCR3/immunology , Signal Transduction/immunology , Virus Diseases/immunology
4.
Semin Immunopathol ; 37(6): 591-605, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26251238

ABSTRACT

Specialized populations of tissue-resident myeloid cells inhabit every organ of the body. While many of these populations appear similar morphologically and phenotypically, they exhibit great functional diversity. The central nervous system (CNS), as an immune privileged organ, possesses a unique tissue-resident macrophage population, the microglia, as well as numerous myeloid cell subsets at its boarders and barriers in CNS-adjoining tissues, namely the meninges, the perivascular space, and the choroid plexus. Recent research has added much to our knowledge about microglia, whereas the populations of CNS-surrounding phagocytes are just starting to be appreciated. As guardians of CNS homeostasis, these myeloid cells perform immune surveillance and immune modulatory tasks in health and disease. As such, microglia and CNS-surrounding antigen-presenting cells have been shown to be crucially involved not only in the initiation and progression but also resolution of multiple sclerosis (MS). MS and its rodent model, experimental autoimmune encephalomyelitis, are autoimmune inflammatory demyelinating CNS pathologies. While some crucial aspects of the disease pathogenesis have been solved, much of the complex involvement and interplay of the innate immune compartment remains yet to be clarified. Here, we will discuss the current understanding of the scope of phenotypes and functions of myeloid cells involved in CNS neuroinflammation.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Microglia/immunology , Multiple Sclerosis/immunology , Myeloid Cells/immunology , Animals , Antigen-Presenting Cells/immunology , Central Nervous System/cytology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Mice , Microglia/cytology , Multiple Sclerosis/pathology , Myeloid Cells/cytology
5.
J Neurosurg Spine ; 23(5): 602-606, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26185898

ABSTRACT

Malignant meningiomas are a rare but aggressive subset of intracranial meningiomas leading to a very limited life expectancy. The occurrence of spinal metastases in these tumors is an even rarer event. The described patient had an intracranial malignant meningioma and developed a symptomatic osteolytic contrast-enhancing lesion in the left C-1 lateral mass suspicious for metastasis. The authors performed a minimally invasive posterior resection of the lesion with vertebroplasty of C-1. Histopathology verified metastasis of the malignant meningioma. The surgical procedure resulted in prompt and permanent pain reduction until the patient died 18 months later. Given the very limited life expectancy in this case, the authors did not consider occipitocervical fusion because of their desire to preserve the range of motion of the head. Therefore, they suggest minimally invasive tumor resection and vertebroplasty in selected palliative tumor patients.

6.
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
7.
Nat Neurosci ; 16(11): 1618-26, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24077561

ABSTRACT

Microglia are brain macrophages and, as such, key immune-competent cells that can respond to environmental changes. Understanding the mechanisms of microglia-specific responses during pathologies is hence vital for reducing disease burden. The definition of microglial functions has so far been hampered by the lack of genetic in vivo approaches that allow discrimination of microglia from closely related peripheral macrophage populations in the body. Here we introduce a mouse experimental system that specifically targets microglia to examine the role of a mitogen-associated protein kinase kinase kinase (MAP3K), transforming growth factor (TGF)-ß-activated kinase 1 (TAK1), during autoimmune inflammation. Conditional depletion of TAK1 in microglia only, not in neuroectodermal cells, suppressed disease, significantly reduced CNS inflammation and diminished axonal and myelin damage by cell-autonomous inhibition of the NF-κB, JNK and ERK1/2 pathways. Thus, we found TAK1 to be pivotal in CNS autoimmunity, and we present a tool for future investigations of microglial function in the CNS.


Subject(s)
Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/immunology , Gene Targeting , MAP Kinase Kinase Kinases/metabolism , Microglia/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/physiology , Brain/metabolism , CD11c Antigen/genetics , CX3C Chemokine Receptor 1 , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Estrogen Antagonists/pharmacology , Gene Expression Regulation/drug effects , Luminescent Proteins/genetics , Lymph Nodes/pathology , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Nerve Tissue Proteins/metabolism , RNA, Untranslated/genetics , Receptors, Chemokine/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Tamoxifen/pharmacology
8.
Magn Reson Imaging ; 31(7): 1156-62, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23773622

ABSTRACT

Previous studies provide evidence that atherosclerosis develops in vascular regions exposed to low wall shear stress (WSS) and high oscillatory shear index (OSI). 4D flow MRI was analyzed in 70 stroke patients with complex plaques (≥4 mm thickness, ulcerated or superimposed thrombi) and in 12 young healthy volunteers. The segmental distribution of peak systolic WSSsystole and OSI was quantified in analysis planes positioned directly at the location of 140 complex plaques found in the 70 patients. In addition, WSSsystole and OSI were evaluated in 8 standard analysis planes distributed along the aorta. Complex plaques were predominantly found at the inner curvature of the aortic arch and of the descending aorta. High OSI was co-located with the segments mostly affected by complex plaque while WSSsystole demonstrated a homogenous distribution. In standard analysis planes, patients demonstrated significantly (p<0.01) altered distribution of wall parameters compared to volunteers (reduced WSSsystole in 91% of aortic wall segments, increased OSI in 48% of segments). OSI distribution was asymmetric with higher values at the inner curvature of the aorta. While WSS and OSI showed expected changes in patients compared to healthy controls, their distribution pattern at complex plaques indicated a more complex and heterogeneous relationship than previously anticipated.


Subject(s)
Aorta, Thoracic/pathology , Aorta/pathology , Atherosclerosis/pathology , Endothelium, Vascular/pathology , Magnetic Resonance Imaging/methods , Plaque, Atherosclerotic/pathology , Stroke/physiopathology , Adult , Aged , Aorta, Thoracic/chemistry , Cohort Studies , Female , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Stress, Mechanical , Young Adult
9.
Immunity ; 38(3): 404-6, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23521879

ABSTRACT

The pathogenic mechanisms driving Sjögren's syndrome (SS) are unclear. In this issue of Immunity, Okuma et al. (2013) demonstrate that tissue-specific dysfunction, namely deficiency of the transcriptional regulator IκB-ζ in epithelial cells, rather than hematopoietic cell pathology, is sufficient to elicit SS-like inflammation.

10.
J Leukoc Biol ; 92(3): 479-88, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22661236

ABSTRACT

The precise mechanisms underlying the effects of IFN-I in CNS autoimmunity remain poorly understood despite the long-standing use of these cytokines as first-line disease-modifying drugs in the treatment of RRMS, a chronic demyelinating CNS autoimmune disease. Systemic use of IFN-I results in pleiotropic immunomodulation linking the innate and adaptive immune responses. Recent research has demonstrated that in the setting of CNS autoimmunity, IFNs-I have multiple effects on myeloid cell subsets, such as circulating monocytes, granulocytes, DCs, and tissue macrophages, such as microglia. These diverse effects include changes in cell activation, maturation, antigen presentation, and cytokine production, thus influencing T cell differentiation and expansion, as well as the regulation of executive functions, such as apoptosis and phagocytosis. Moreover, current data suggest that the engagement of the IFNAR on myeloid cells changes the activation status of the inflammasome in a cell type-specific manner. Whereas most reports support primarily immune-suppressive effects of IFN-I on myeloid cells, endogenously produced, exogenously induced, and peripherally administered IFNs-I exert complex differential spatial effects during CNS autoimmune inflammation. Clearly characterizing the molecular and cellular basis of these effects promises to yield viable targets for a more directed, localized, cell type-specific IFN-I-based therapeutic approach. This kind of approach would allow for replacing the current treatment strategy in MS of broadly and unselectively altering all immune responses, regardless of their beneficial or detrimental nature.


Subject(s)
Autoimmunity/immunology , Demyelinating Autoimmune Diseases, CNS/immunology , Interferon Type I/immunology , Myeloid Cells/immunology , Antigen Presentation/immunology , Demyelinating Autoimmune Diseases, CNS/metabolism , Humans , Interferon Type I/metabolism , Lymphocyte Activation/immunology , Myeloid Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...