Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Transl Med ; 16(751): eadj9672, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38865481

ABSTRACT

Cancer treatment with anti-PD-1 immunotherapy can cause central nervous system immune-related adverse events (CNS-irAEs). The role of microglia in anti-PD-1 immunotherapy-induced CNS-irAEs is unclear. We found that anti-PD-1 treatment of mice caused morphological signs of activation and major histocompatibility complex (MHC) class II up-regulation on microglia. Functionally, anti-PD-1 treatment induced neurocognitive deficits in mice, independent of T cells, B cells, and natural killer cells. Instead, we found that microglia mediated these CNS-irAEs. Single-cell RNA sequencing revealed major transcriptional changes in microglia upon anti-PD-1 treatment. The anti-PD-1 effects were mediated by anti-PD-1 antibodies interacting directly with microglia and were not secondary to peripheral T cell activation. Using a proteomics approach, we identified spleen tyrosine kinase (Syk) as a potential target in activated microglia upon anti-PD-1 treatment. Syk inhibition reduced microglia activation and improved neurocognitive function without impairing anti-melanoma effects. Moreover, we analyzed CNS tissue from a patient cohort that had received anti-PD-1 treatment. Imaging mass cytometry revealed that anti-PD-1 treatment of patients was associated with increased surface marker expression indicative of microglia activation. In summary, we identified a disease-promoting role for microglia in CNS-irAEs driven by Syk and provide an inhibitor-based approach to interfere with this complication after anti-PD-1 immunotherapy.


Subject(s)
Central Nervous System , Immunotherapy , Microglia , Programmed Cell Death 1 Receptor , Animals , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Immunotherapy/adverse effects , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Humans , Central Nervous System/pathology , Central Nervous System/drug effects , Mice, Inbred C57BL , Syk Kinase/metabolism , Mice
2.
Nat Commun ; 14(1): 2721, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37169749

ABSTRACT

While the precise processes underlying a sex bias in the development of central nervous system (CNS) disorders are unknown, there is growing evidence that an early life immune activation can contribute to the disease pathogenesis. When we mimicked an early systemic viral infection or applied murine cytomegalovirus (MCMV) systemically in neonatal female and male mice, only male adolescent mice presented behavioral deficits, including reduced social behavior and cognition. This was paralleled by an increased amount of infiltrating T cells in the brain parenchyma, enhanced interferon-γ (IFNγ) signaling, and epigenetic reprogramming of microglial cells. These microglial cells showed increased phagocytic activity, which resulted in abnormal loss of excitatory synapses within the hippocampal brain region. None of these alterations were seen in female adolescent mice. Our findings underscore the early postnatal period's susceptibility to cause sex-dependent long-term CNS deficiencies following infections.


Subject(s)
Central Nervous System Diseases , Microglia , Animals , Female , Male , Mice , Microglia/pathology , Brain , Central Nervous System Diseases/pathology , Interferon-gamma/genetics , Epigenesis, Genetic
3.
STAR Protoc ; 3(1): 101186, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35243376

ABSTRACT

Most of the protocols to analyze metabolic features of cell populations from different tissues rely on in vitro cell culture conditions. Here, we present a flow-cytometry-based protocol for measuring the respiratory chain function in permeabilized mouse microglia ex vivo. We describe microglial cell isolation, followed by analyzing complex I and II using flow cytometry. This optimized protocol requires a low input of permeabilized cells and can be applied to other ex vivo isolated cells or cells derived from cell cultures. For complete details on the use and execution of this protocol, please refer to Erny et al. (2021).


Subject(s)
Cell Culture Techniques , Microglia , Animals , Cell Separation/methods , Electron Transport , Flow Cytometry/methods , Mice
4.
Nat Neurosci ; 25(3): 295-305, 2022 03.
Article in English | MEDLINE | ID: mdl-35241804

ABSTRACT

Microglial function declines during aging. The interaction of microglia with the gut microbiota has been well characterized during development and adulthood but not in aging. Here, we compared microglial transcriptomes from young-adult and aged mice housed under germ-free and specific pathogen-free conditions and found that the microbiota influenced aging associated-changes in microglial gene expression. The absence of gut microbiota diminished oxidative stress and ameliorated mitochondrial dysfunction in microglia from the brains of aged mice. Unbiased metabolomic analyses of serum and brain tissue revealed the accumulation of N6-carboxymethyllysine (CML) in the microglia of the aging brain. CML mediated a burst of reactive oxygen species and impeded mitochondrial activity and ATP reservoirs in microglia. We validated the age-dependent rise in CML levels in the sera and brains of humans. Finally, a microbiota-dependent increase in intestinal permeability in aged mice mediated the elevated levels of CML. This study adds insight into how specific features of microglia from aged mice are regulated by the gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Microglia , Animals , Lysine/analogs & derivatives , Lysine/metabolism , Mice , Microglia/metabolism , Oxidative Stress
5.
Front Cell Neurosci ; 15: 698126, 2021.
Article in English | MEDLINE | ID: mdl-34295223

ABSTRACT

The immune system is crucial for defending against various invaders, such as pathogens, cancer cells or misfolded proteins. With increasing age, the diminishing immune response, known as immunosenescence, becomes evident. Concomitantly, some diseases like infections, autoimmune diseases, chronic inflammatory diseases and cancer, accumulate with age. Different cell types are part of the innate immunity response and produce soluble factors, cytokines, chemokines, and type I interferons. Improper maturation of innate immune cells or their dysfunction have been linked to numerous age-related diseases. In parallel to the occurrence of the many functional facets of the immune response, a symbiotic microbiota had been acquired. For the relevant and situation-dependent function of the immune system the microbiome plays an essential role because it fine-tunes the immune system and its responses during life. Nevertheless, how the age-related alterations in the microbiota are reflected in the innate immune system, is still poorly understood. With this review, we provide an up-to-date overview on our present understanding of the gut microbiota effects on innate immunity, with a particular emphasis on aging-associated changes in the gut microbiota and the implications for the brain innate immune response.

6.
Sci Rep ; 11(1): 13462, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34188106

ABSTRACT

CD33/Sialic acid-binding Ig-like lectin 3 (SIGLEC3) is an innate immune receptor expressed on myeloid cells and mediates inhibitory signaling via tyrosine phosphatases. Variants of CD33 are associated with Alzheimer's disease (AD) suggesting that modulation of CD33 signaling might be beneficial in AD. Hence, there is an urgent need for reliable cellular CD33 reporter systems. Therefore, we generated a CD33 reporter cell line expressing a fusion protein consisting of the extracellular domain of either human full-length CD33 (CD33M) or the AD-protective variant CD33ΔE2 (D2-CD33/CD33m) linked to TYRO protein tyrosine kinase binding protein (TYROBP/DAP12) to investigate possible ligands and antibodies for modulation of CD33 signaling. Application of the CD33-specific antibodies P67.6 and 1c7/1 to the CD33M-DAP12 reporter cells resulted in increased phosphorylation of the kinase SYK, which is downstream of DAP12. CD33M-DAP12 but not CD33ΔE2-DAP12 expressing reporter cells showed increased intracellular calcium levels upon treatment with CD33 antibody P67.6 and partially for 1c7/1. Furthermore, stimulation of human induced pluripotent stem cell-derived microglia with the CD33 antibodies P67.6 or 1c7/1 directly counteracted the triggering receptor expressed on myeloid cells 2 (TREM2)-induced phosphorylation of SYK and decreased the phagocytic uptake of bacterial particles. Thus, the developed reporter system confirmed CD33 pathway activation by CD33 antibody clones P67.6 and 1c7/1. In addition, data showed that phosphorylation of SYK by TREM2 activation and phagocytosis of bacterial particles can be directly antagonized by CD33 signaling.


Subject(s)
Alzheimer Disease/immunology , Antibodies/immunology , Induced Pluripotent Stem Cells/immunology , Microglia/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Alzheimer Disease/genetics , Cell Line , Humans , Sialic Acid Binding Ig-like Lectin 3/genetics
7.
Nat Aging ; 1(12): 1127-1136, 2021 12.
Article in English | MEDLINE | ID: mdl-37117525

ABSTRACT

Understanding the physiological origins of age-related cognitive decline is of critical importance given the rising age of the world's population1. Previous work in animal models has established a strong link between cognitive performance and the microbiota2-5, and it is known that the microbiome undergoes profound remodeling in older adults6. Despite growing evidence for the association between age-related cognitive decline and changes in the gut microbiome, the mechanisms underlying such interactions between the brain and the gut are poorly understood. Here, using fecal microbiota transplantation (FMT), we demonstrate that age-related remodeling of the gut microbiota leads to decline in cognitive function in mice and that this impairment can be rescued by transplantation of microbiota from young animals. Moreover, using a metabolomic approach, we found elevated concentrations of δ-valerobetaine, a gut microbiota-derived metabolite, in the blood and brain of aged mice and older adults. We then demonstrated that δ-valerobetaine is deleterious to learning and memory processes in mice. At the neuronal level, we showed that δ-valerobetaine modulates inhibitory synaptic transmission and neuronal network activity. Finally, we identified specific bacterial taxa that significantly correlate with δ-valerobetaine levels in the brain. Based on our findings, we propose that δ-valerobetaine contributes to microbiota-driven brain aging and that the associated mechanisms represent a promising target for countering age-related cognitive decline.


Subject(s)
Cognitive Dysfunction , Gastrointestinal Microbiome , Microbiota , Animals , Mice , Microbiota/physiology , Gastrointestinal Microbiome/physiology , Cognition/physiology , Cognitive Dysfunction/metabolism , Brain/metabolism
8.
Cell Metab ; 33(11): 2260-2276.e7, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731656

ABSTRACT

As tissue macrophages of the central nervous system (CNS), microglia constitute the pivotal immune cells of this organ. Microglial features are strongly dependent on environmental cues such as commensal microbiota. Gut bacteria are known to continuously modulate microglia maturation and function by the production of short-chain fatty acids (SCFAs). However, the precise mechanism of this crosstalk is unknown. Here we determined that the immature phenotype of microglia from germ-free (GF) mice is epigenetically imprinted by H3K4me3 and H3K9ac on metabolic genes associated with substantial functional alterations including increased mitochondrial mass and specific respiratory chain dysfunctions. We identified acetate as the essential microbiome-derived SCFA driving microglia maturation and regulating the homeostatic metabolic state, and further showed that it is able to modulate microglial phagocytosis and disease progression during neurodegeneration. These findings indicate that acetate is an essential bacteria-derived molecule driving metabolic pathways and functions of microglia during health and perturbation.


Subject(s)
Microbiota , Acetates/pharmacology , Animals , Brain/metabolism , Fatty Acids, Volatile/metabolism , Immune System/metabolism , Mice , Microbiota/physiology
9.
Brain Pathol ; 30(6): 1159-1177, 2020 11.
Article in English | MEDLINE | ID: mdl-33073887

ABSTRACT

The innate immune system in the central nervous system (CNS) is mainly represented by specialized tissue-resident macrophages, called microglia. In the past years, various species-, host- and tissue-specific as well as environmental factors were recognized that essentially affect microglial properties and functions in the healthy and diseased brain. Host microbiota are mostly residing in the gut and contribute to microglial activation states, for example, via short-chain fatty acids (SCFAs) or aryl hydrocarbon receptor (AhR) ligands. Thereby, the gut microorganisms are deemed to influence numerous CNS diseases mediated by microglia. In this review, we summarize recent findings of the interaction between the host microbiota and the CNS in health and disease, where we specifically highlight the resident gut microbiota as a crucial environmental factor for microglial function as what we coin "the microbiota-microglia axis."


Subject(s)
Central Nervous System Diseases/pathology , Microbiota/physiology , Microglia/pathology , Animals , Gastrointestinal Microbiome/physiology , Humans
10.
Acta Neuropathol Commun ; 8(1): 119, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32727612

ABSTRACT

It was recently revealed that gut microbiota promote amyloid-beta (Aß) burden in mouse models of Alzheimer's disease (AD). However, the underlying mechanisms when using either germ-free (GF) housing conditions or treatments with antibiotics (ABX) remained unknown. In this study, we show that GF and ABX-treated 5x familial AD (5xFAD) mice developed attenuated hippocampal Aß pathology and associated neuronal loss, and thereby delayed disease-related memory deficits. While Aß production remained unaffected in both GF and ABX-treated 5xFAD mice, we noticed in GF 5xFAD mice enhanced microglial Aß uptake at early stages of the disease compared to ABX-treated 5xFAD mice. Furthermore, RNA-sequencing of hippocampal microglia from SPF, GF and ABX-treated 5xFAD mice revealed distinct microbiota-dependent gene expression profiles associated with phagocytosis and altered microglial activation states. Taken together, we observed that constitutive or induced microbiota modulation in 5xFAD mice differentially controls microglial Aß clearance mechanisms preventing neurodegeneration and cognitive deficits.


Subject(s)
Alzheimer Disease/pathology , Gastrointestinal Microbiome/physiology , Hippocampus/pathology , Microglia/metabolism , Alzheimer Disease/microbiology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Humans , Male , Mice
11.
Nat Neurosci ; 23(5): 676-689, 2020 05.
Article in English | MEDLINE | ID: mdl-32284604

ABSTRACT

While CNS microglia have been extensively studied, relatively little is known about macrophages populating the peripheral nervous system. Here we performed ontogenic, transcriptomic and spatial characterization of sciatic nerve macrophages (snMacs). Using multiple fate-mapping systems, we show that snMacs do not derive from the early embryonic precursors colonizing the CNS, but originate primarily from late embryonic precursors and become replaced by bone-marrow-derived macrophages over time. Using single-cell transcriptomics, we identified a tissue-specific core signature of snMacs and two spatially separated snMacs: Relmα+Mgl1+ snMacs in the epineurium and Relmα-Mgl1- snMacs in the endoneurium. Globally, snMacs lack most of the core signature genes of microglia, with only the endoneurial subset expressing a restricted number of these genes. In response to nerve injury, the two resident snMac populations respond differently. Moreover, and unlike in the CNS, monocyte-derived macrophages that develop during injury can engraft efficiently in the pool of resident peripheral nervous system macrophages.


Subject(s)
Macrophages/cytology , Macrophages/physiology , Sciatic Nerve/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Nerve Crush , Transcriptome
12.
Nat Cell Biol ; 22(7): 828-841, 2020 07.
Article in English | MEDLINE | ID: mdl-32541879

ABSTRACT

Mutations in chromatin-modifying complexes and metabolic enzymes commonly underlie complex human developmental syndromes affecting multiple organs. A major challenge is to determine how disease-causing genetic lesions cause deregulation of homeostasis in unique cell types. Here we show that neural-specific depletion of three members of the non-specific lethal (NSL) chromatin complex-Mof, Kansl2 or Kansl3-unexpectedly leads to severe vascular defects and brain haemorrhaging. Deregulation of the epigenetic landscape induced by the loss of the NSL complex in neural cells causes widespread metabolic defects, including an accumulation of free long-chain fatty acids (LCFAs). Free LCFAs induce a Toll-like receptor 4 (TLR4)-NFκB-dependent pro-inflammatory signalling cascade in neighbouring vascular pericytes that is rescued by TLR4 inhibition. Pericytes display functional changes in response to LCFA-induced activation that result in vascular breakdown. Our work establishes that neurovascular function is determined by the neural metabolic environment.


Subject(s)
Cell Nucleus/pathology , Chromatin/metabolism , Histone Acetyltransferases/physiology , Inflammation/pathology , Neovascularization, Pathologic/pathology , Neurons/pathology , Pericytes/pathology , Animals , Brain/cytology , Brain/metabolism , Cell Nucleus/metabolism , Chromatin/genetics , Fatty Acids/metabolism , Female , Fetus/cytology , Fetus/metabolism , Humans , Inflammation/metabolism , Male , Metabolome , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pericytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL