Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cell ; 181(3): 557-573.e18, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259484

ABSTRACT

Central nervous system (CNS) macrophages comprise microglia and border-associated macrophages (BAMs) residing in the meninges, the choroid plexus, and the perivascular spaces. Most CNS macrophages emerge during development, with the exception of choroid plexus and dural macrophages, which are replaced by monocytes in adulthood. Whether microglia and BAMs share a developmental program or arise from separate lineages remains unknown. Here, we identified two phenotypically, transcriptionally, and locally distinct brain macrophages throughout development, giving rise to either microglia or BAMs. Two macrophage populations were already present in the yolk sac suggesting an early segregation. Fate-mapping models revealed that BAMs mostly derived from early erythro-myeloid progenitors in the yolk sac. The development of microglia was dependent on TGF-ß, whereas the genesis of BAMs occurred independently of this cytokine. Collectively, our data show that developing parenchymal and non-parenchymal brain macrophages are separate entities in terms of ontogeny, gene signature, and requirement for TGF-ß.


Subject(s)
Brain/cytology , Macrophages/cytology , Microglia/cytology , Animals , Brain/metabolism , Cell Lineage , Mice , Monocytes , Signal Transduction , Transforming Growth Factor beta/metabolism
2.
Immunity ; 48(2): 380-395.e6, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29426702

ABSTRACT

Individual reports suggest that the central nervous system (CNS) contains multiple immune cell types with diverse roles in tissue homeostasis, immune defense, and neurological diseases. It has been challenging to map leukocytes across the entire brain, and in particular in pathology, where phenotypic changes and influx of blood-derived cells prevent a clear distinction between reactive leukocyte populations. Here, we applied high-dimensional single-cell mass and fluorescence cytometry, in parallel with genetic fate mapping systems, to identify, locate, and characterize multiple distinct immune populations within the mammalian CNS. Using this approach, we revealed that microglia, several subsets of border-associated macrophages and dendritic cells coexist in the CNS at steady state and exhibit disease-specific transformations in the immune microenvironment during aging and in models of Alzheimer's disease and multiple sclerosis. Together, these data and the described framework provide a resource for the study of disease mechanisms, potential biomarkers, and therapeutic targets in CNS disease.


Subject(s)
Aging/immunology , Central Nervous System/immunology , Leukocytes/immunology , Macrophages/immunology , Animals , Dendritic Cells/immunology , Mice , Mice, Inbred C57BL , Microglia/immunology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/immunology , Single-Cell Analysis
3.
Immunity ; 47(5): 903-912.e4, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29126797

ABSTRACT

Alveolar macrophages (AMs) derive from fetal liver monocytes, which colonize the lung during embryonic development and give rise to fully mature AMs perinatally. AM differentiation requires granulocyte macrophage colony-stimulating factor (GM-CSF), but whether additional factors are involved in AM regulation is not known. Here we report that AMs, in contrast to most other tissue macrophages, were also dependent on transforming growth factor-ß receptor (TGF-ßR) signaling. Conditional deletion of TGF-ßR in mice at different time points halted the development and differentiation of AMs. In adult mice, TGF-ß was also critical for AM homeostasis. The source of TGF-ß was AMs themselves, indicative of an autocrine loop that promotes AM self-maintenance. Mechanistically, TGF-ßR signaling resulted in upregulation of PPAR-γ, a signature transcription factor essential for the development of AMs. These findings reveal an additional layer of complexity regarding the guidance cues, which govern the genesis, maturation, and survival of AMs.


Subject(s)
Homeostasis , Macrophages, Alveolar/physiology , Transforming Growth Factor beta/physiology , Animals , Cell Differentiation , Embryonic Development , Mice , Mice, Inbred C57BL , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction/physiology , Transcriptome
4.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33653955

ABSTRACT

Pericytes regulate the development of organ-specific characteristics of the brain vasculature such as the blood-brain barrier (BBB) and astrocytic end-feet. Whether pericytes are involved in the control of leukocyte trafficking in the adult central nervous system (CNS), a process tightly regulated by CNS vasculature, remains elusive. Using adult pericyte-deficient mice (Pdgfbret/ret ), we show that pericytes limit leukocyte infiltration into the CNS during homeostasis and autoimmune neuroinflammation. The permissiveness of the vasculature toward leukocyte trafficking in Pdgfbret/ret mice inversely correlates with vessel pericyte coverage. Upon induction of experimental autoimmune encephalomyelitis (EAE), pericyte-deficient mice die of severe atypical EAE, which can be reversed with fingolimod, indicating that the mortality is due to the massive influx of immune cells into the brain. Additionally, administration of anti-VCAM-1 and anti-ICAM-1 antibodies reduces leukocyte infiltration and diminishes the severity of atypical EAE symptoms of Pdgfbret/ret mice, indicating that the proinflammatory endothelium due to absence of pericytes facilitates exaggerated neuroinflammation. Furthermore, we show that the presence of myelin peptide-specific peripheral T cells in Pdgfbret/ret ;2D2tg mice leads to the development of spontaneous neurological symptoms paralleled by the massive influx of leukocytes into the brain. These findings indicate that intrinsic changes within brain vasculature can promote the development of a neuroinflammatory disorder.


Subject(s)
Blood-Brain Barrier/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Homeostasis/immunology , Leukocytes/immunology , Pericytes/immunology , Animals , Blood-Brain Barrier/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Homeostasis/genetics , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Leukocytes/pathology , Mice , Mice, Transgenic , Pericytes/pathology , Proto-Oncogene Proteins c-sis/deficiency , Proto-Oncogene Proteins c-sis/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
5.
Eur J Immunol ; 51(7): 1799-1808, 2021 07.
Article in English | MEDLINE | ID: mdl-33759186

ABSTRACT

Mononuclear phagocytes consisting of monocytes, macrophages, and DCs play a complex role in tumor development by either promoting or restricting tumor growth. Cutaneous squamous cell carcinoma (cSCC) is the second most common nonmelanoma skin cancer arising from transformed epidermal keratinocytes. While present at high numbers, the role of tumor-infiltrating and resident myeloid cells in the formation of cSCC is largely unknown. Using transgenic mice and depleting antibodies to eliminate specific myeloid cell types in the skin, we investigated the involvement of mononuclear phagocytes in the development of UV-induced cSCC in K14-HPV8-E6 transgenic mice. Although resident Langerhans cells were enriched in the tumor, their contribution to tumor formation was negligible. Equally, dermal macrophages were dispensable for the development of cSCC. In contrast, mice lacking circulating monocytes were completely resistant to UV-induced cSCC, indicating that monocytes promote tumor development. Collectively, these results demonstrate a critical role for classical monocytes in the initiation of skin cancer.


Subject(s)
Carcinogenesis/pathology , Epidermis/pathology , Monocytes/pathology , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/radiation effects , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epidermis/radiation effects , Female , Keratinocytes/pathology , Keratinocytes/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/radiation effects , Skin/pathology , Skin/radiation effects
7.
Acta Neuropathol ; 141(6): 901-915, 2021 06.
Article in English | MEDLINE | ID: mdl-33774709

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease characterized by impaired neuromuscular signaling due to autoantibodies targeting the acetylcholine receptor. Although its auto-antigens and effector mechanisms are well defined, the cellular and molecular drivers underpinning MG remain elusive. Here, we employed high-dimensional single-cell mass and spectral cytometry of blood and thymus samples from MG patients in combination with supervised and unsupervised machine-learning tools to gain insight into the immune dysregulation underlying MG. By creating a comprehensive immune map, we identified two dysregulated subsets of inflammatory circulating memory T helper (Th) cells. These signature ThCD103 and ThGM cells populated the diseased thymus, were reduced in the blood of MG patients, and were inversely correlated with disease severity. Both signature Th subsets rebounded in the blood of MG patients after surgical thymus removal, indicative of their role as cellular markers of disease activity. Together, this in-depth analysis of the immune landscape of MG provides valuable insight into disease pathogenesis, suggests novel biomarkers and identifies new potential therapeutic targets for treatment.


Subject(s)
Immunophenotyping/methods , Myasthenia Gravis/immunology , Myasthenia Gravis/pathology , Single-Cell Analysis , T-Lymphocytes/pathology , Adult , Aged , Aged, 80 and over , Autoantibodies , Autoimmunity , B-Lymphocytes/immunology , Biomarkers , Female , Humans , Machine Learning , Male , Middle Aged , Myasthenia Gravis/blood , Receptors, Cholinergic/immunology , T-Lymphocytes/immunology , Thymectomy , Thymus Gland
9.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31940023

ABSTRACT

Macrophages are part of the innate immune system and are present in every organ of the body. They fulfill critical roles in tissue homeostasis and development and are involved in various pathologies. An essential factor for the development, homeostasis, and function of mononuclear phagocytes is the colony stimulating factor-1 receptor (CSF-1R), which has two known ligands: CSF-1 and interleukin-34 (IL-34). While CSF-1 has been extensively studied, the biology and functions of IL-34 are only now beginning to be uncovered. In this review, we discuss recent advances of IL-34 biology in health and disease with a specific focus on mononuclear phagocytes.


Subject(s)
Interleukins/immunology , Animals , Humans , Immunity, Innate/immunology , Macrophages/immunology , Signal Transduction/immunology
10.
Stem Cell Reports ; 15(3): 566-576, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32857979

ABSTRACT

Fatty acid ß-oxidation (FAO), the breakdown of lipids, is a metabolic pathway used by various stem cells. FAO levels are generally high during quiescence and downregulated with proliferation. The endogenous metabolite malonyl-CoA modulates lipid metabolism as a reversible FAO inhibitor and as a substrate for de novo lipogenesis. Here we assessed whether malonyl-CoA can be exploited to steer the behavior of hematopoietic stem/progenitor cells (HSPCs), quiescent stem cells of clinical relevance. Treatment of mouse HSPCs in vitro with malonyl-CoA increases HSPC numbers compared with nontreated controls and ameliorates blood reconstitution capacity when transplanted in vivo, mainly through enhanced lymphoid reconstitution. Similarly, human HSPC numbers also increase upon malonyl-CoA treatment in vitro. These data corroborate that lipid metabolism can be targeted to direct cell fate and stem cell proliferation. Physiological modulation of metabolic pathways, rather than genetic or pharmacological inhibition, provides unique perspectives for stem cell manipulations in health and disease.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lipid Metabolism , Lymphocytes/cytology , Metabolome , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Cells, Cultured , Fatty Acids/metabolism , Gene Expression Regulation , Lipid Metabolism/genetics , Lymphocytes/metabolism , Malonyl Coenzyme A/metabolism , Metabolome/genetics , Mice, Inbred C57BL , Oxidation-Reduction
11.
Sci Immunol ; 4(31)2019 01 25.
Article in English | MEDLINE | ID: mdl-30679199

ABSTRACT

The central nervous system (CNS) is under close surveillance by immune cells, which mediate tissue homeostasis, protection, and repair. Conversely, in neuroinflammation, dysregulated leukocyte invasion into the CNS leads to immunopathology and neurological disability. To invade the brain parenchyma, autoimmune encephalitogenic T helper (TH) cells must encounter their cognate antigens (Ags) presented via local Ag-presenting cells (APCs). The precise identity of the APC that samples, processes, and presents CNS-derived Ags to autoaggressive T cells is unknown. Here, we used a combination of high-dimensional single-cell mapping and conditional MHC class II ablation across all CNS APCs to systematically interrogate each population for its ability to reactivate encephalitogenic TH cells in vivo. We found a population of conventional dendritic cells, but not border-associated macrophages or microglia, to be essential for licensing T cells to initiate neuroinflammation.


Subject(s)
Antigen Presentation , Antigens/immunology , Central Nervous System/immunology , Dendritic Cells/immunology , Myelin Sheath/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Histocompatibility Antigens Class II/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/immunology
12.
Nat Neurosci ; 22(6): 848-850, 2019 06.
Article in English | MEDLINE | ID: mdl-31061495

Subject(s)
Brain , Macrophages , Animals , Mice
SELECTION OF CITATIONS
SEARCH DETAIL