Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
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.
Nat Immunol ; 22(6): 699-710, 2021 06.
Article in English | MEDLINE | ID: mdl-34040226

ABSTRACT

It is increasingly recognized that immune development within mucosal tissues is under the control of environmental factors during early life. However, the cellular mechanisms that underlie such temporally and regionally restrictive governance of these processes are unclear. Here, we uncover an extrathymic pathway of immune development within the colon that is controlled by embryonic but not bone marrow-derived macrophages, which determines the ability of these organs to receive invariant natural killer T (iNKT) cells and allow them to establish local residency. Consequently, early-life perturbations of fetal-derived macrophages result in persistent decreases of mucosal iNKT cells and is associated with later-life susceptibility or resistance to iNKT cell-associated mucosal disorders. These studies uncover a host developmental program orchestrated by ontogenically distinct macrophages that is regulated by microbiota, and they reveal an important postnatal function of macrophages that emerge in fetal life.


Subject(s)
Colitis/immunology , Intestinal Mucosa/immunology , Listeriosis/immunology , Macrophages/immunology , Mucosal-Associated Invariant T Cells/immunology , Animals , Cell Proliferation/genetics , Colitis/microbiology , Colitis/pathology , Colon/cytology , Colon/embryology , Colon/immunology , Colon/pathology , Cytokines/metabolism , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/immunology , Disease Models, Animal , Embryo, Mammalian , Female , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Developmental/immunology , Germ-Free Life , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/pathology , Listeriosis/microbiology , Listeriosis/pathology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology
3.
Nat Immunol ; 16(5): 505-16, 2015 May.
Article in English | MEDLINE | ID: mdl-25751747

ABSTRACT

A cytosolic role for the histone methyltransferase Ezh2 in regulating lymphocyte activation has been suggested, but the molecular mechanisms underpinning this extranuclear function have remained unclear. Here we found that Ezh2 regulated the integrin signaling and adhesion dynamics of neutrophils and dendritic cells (DCs). Ezh2 deficiency impaired the integrin-dependent transendothelial migration of innate leukocytes and restricted disease progression in an animal model of multiple sclerosis. Direct methylation of talin, a key regulatory molecule in cell migration, by Ezh2 disrupted the binding of talin to F-actin and thereby promoted the turnover of adhesion structures. This regulatory effect was abolished by targeted disruption of the interactions of Ezh2 with the cytoskeletal-reorganization effector Vav1. Our studies reveal an unforeseen extranuclear function for Ezh2 in regulating adhesion dynamics, with implications for leukocyte migration, immune responses and potentially pathogenic processes.


Subject(s)
Cell Nucleus/metabolism , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Neutrophils/immunology , Polycomb Repressive Complex 2/metabolism , Talin/metabolism , Actins/metabolism , Animals , Cell Adhesion/genetics , Cell Movement , Cells, Cultured , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein , Humans , Lymphocyte Activation/genetics , Methylation , Mice , Mice, Knockout , Polycomb Repressive Complex 2/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-vav/metabolism , Talin/genetics , Transendothelial and Transepithelial Migration/genetics
4.
Immunity ; 49(2): 326-341.e7, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30054204

ABSTRACT

The maintenance of appropriate arterial tone is critically important for normal physiological arterial function. However, the cellular and molecular mechanisms remain poorly defined. Here, we have shown that in the mouse aorta, resident macrophages prevented arterial stiffness and collagen deposition in the steady state. Using phenotyping, transcriptional profiling, and targeted deletion of Csf1r, we have demonstrated that these macrophages-which are a feature of blood vessels invested with smooth muscle cells (SMCs) in both mouse and human tissues-expressed the hyaluronan (HA) receptor LYVE-l. Furthermore, we have shown they possessed the unique ability to modulate collagen expression in SMCs by matrix metalloproteinase MMP-9-dependent proteolysis through engagement of LYVE-1 with the HA pericellular matrix of SMCs. Our study has unveiled a hitherto unknown homeostatic contribution of arterial LYVE-1+ macrophages through the control of collagen production by SMCs and has identified a function of LYVE-1 in leukocytes.


Subject(s)
Collagen/metabolism , Glycoproteins/metabolism , Hyaluronan Receptors/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Vascular Stiffness/physiology , Animals , Aorta/physiology , Female , Glycoproteins/genetics , Humans , Hyaluronic Acid/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
5.
Immunity ; 47(1): 183-198.e6, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723550

ABSTRACT

Tissue macrophages arise during embryogenesis from yolk-sac (YS) progenitors that give rise to primitive YS macrophages. Until recently, it has been impossible to isolate or derive sufficient numbers of YS-derived macrophages for further study, but data now suggest that induced pluripotent stem cells (iPSCs) can be driven to undergo a process reminiscent of YS-hematopoiesis in vitro. We asked whether iPSC-derived primitive macrophages (iMacs) can terminally differentiate into specialized macrophages with the help of growth factors and organ-specific cues. Co-culturing human or murine iMacs with iPSC-derived neurons promoted differentiation into microglia-like cells in vitro. Furthermore, murine iMacs differentiated in vivo into microglia after injection into the brain and into functional alveolar macrophages after engraftment in the lung. Finally, iPSCs from a patient with familial Mediterranean fever differentiated into iMacs with pro-inflammatory characteristics, mimicking the disease phenotype. Altogether, iMacs constitute a source of tissue-resident macrophage precursors that can be used for biological, pathophysiological, and therapeutic studies.


Subject(s)
Cell Culture Techniques/methods , Hematopoiesis , Macrophages/physiology , Neurons/physiology , Pluripotent Stem Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Embryo, Mammalian , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis
6.
Immunity ; 42(4): 665-78, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902481

ABSTRACT

Although classified as hematopoietic cells, tissue-resident macrophages (MFs) arise from embryonic precursors that seed the tissues prior to birth to generate a self-renewing population, which is maintained independently of adult hematopoiesis. Here we reveal the identity of these embryonic precursors using an in utero MF-depletion strategy and fate-mapping of yolk sac (YS) and fetal liver (FL) hematopoiesis. We show that YS MFs are the main precursors of microglia, while most other MFs derive from fetal monocytes (MOs). Both YS MFs and fetal MOs arise from erythro-myeloid progenitors (EMPs) generated in the YS. In the YS, EMPs gave rise to MFs without monocytic intermediates, while EMP seeding the FL upon the establishment of blood circulation acquired c-Myb expression and gave rise to fetal MOs that then seeded embryonic tissues and differentiated into MFs. Thus, adult tissue-resident MFs established from hematopoietic stem cell-independent embryonic precursors arise from two distinct developmental programs.


Subject(s)
Aging/immunology , Macrophages/immunology , Monocytes/immunology , Myeloid Progenitor Cells/immunology , Proto-Oncogene Proteins c-myb/immunology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage/immunology , Cell Tracking , Embryo, Mammalian , Female , Fetus , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Kidney/cytology , Kidney/immunology , Liver/cytology , Liver/immunology , Lung/cytology , Lung/immunology , Macrophages/cytology , Mice , Microglia/cytology , Microglia/immunology , Monocytes/cytology , Myeloid Progenitor Cells/cytology , Pregnancy , Primary Cell Culture , Proto-Oncogene Proteins c-myb/metabolism , Skin/cytology , Skin/immunology , Yolk Sac/cytology , Yolk Sac/immunology
8.
Nature ; 546(7660): 662-666, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28614294

ABSTRACT

During gestation the developing human fetus is exposed to a diverse range of potentially immune-stimulatory molecules including semi-allogeneic antigens from maternal cells, substances from ingested amniotic fluid, food antigens, and microbes. Yet the capacity of the fetal immune system, including antigen-presenting cells, to detect and respond to such stimuli remains unclear. In particular, dendritic cells, which are crucial for effective immunity and tolerance, remain poorly characterized in the developing fetus. Here we show that subsets of antigen-presenting cells can be identified in fetal tissues and are related to adult populations of antigen-presenting cells. Similar to adult dendritic cells, fetal dendritic cells migrate to lymph nodes and respond to toll-like receptor ligation; however, they differ markedly in their response to allogeneic antigens, strongly promoting regulatory T-cell induction and inhibiting T-cell tumour-necrosis factor-α production through arginase-2 activity. Our results reveal a previously unappreciated role of dendritic cells within the developing fetus and indicate that they mediate homeostatic immune-suppressive responses during gestation.


Subject(s)
Arginase/metabolism , Dendritic Cells/enzymology , Dendritic Cells/immunology , Fetus/immunology , Immune Tolerance , T-Lymphocytes/immunology , Adult , Cell Movement , Cell Proliferation , Cytokines/biosynthesis , Cytokines/immunology , Fetus/cytology , Fetus/enzymology , Humans , Lymph Nodes/cytology , Lymph Nodes/immunology , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptors/immunology
9.
J Environ Manage ; 331: 117234, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36646040

ABSTRACT

Indigenous Australians used fire in spinifex deserts for millennia. These practices mostly ceased following European colonisation, but many contemporary Indigenous groups seek to restore 'right-way fire' practices, to meet inter-related social, economic, cultural and biodiversity objectives. However, measuring and reporting on the fire pattern outcomes of management is challenging, because the spatio-temporal patterns of right-way fire are not clearly defined, and because spatio-temporal variability in rainfall makes fire occurrence highly variable in these desert environments. We present an approach for measuring and reporting on fire management outcomes to account for spatio-temporal rainfall variability. The purpose is to support Indigenous groups to assess performance against their management targets, and lay the groundwork for developing an accredited method for valuing combined social, cultural and biodiversity outcomes. We reviewed fire management plans of desert Indigenous groups to identify spatial fire pattern indicators for right-way fire in spinifex deserts. We integrated annual rainfall surfaces with time-since fire mapping (using Landsat imagery) to create a new spatial dataset of accumulated rainfall-since-last-fire, that better represents post-fire vegetation recovery as categorised by local Indigenous people. The fire pattern indicators were merged into a single score using an environmental accounting approach. To strengthen interpretation, we developed an approach for identifying a control area with matching vegetation and fire history, up to the point of management. We applied these methods to a 125,000 ha case study area: Durba Hills, managed by the Martu people of Western Australia. Using a 20-year time series, we show that since right-way fire management at Durba Hills was re-introduced (2009), the fire pattern indicators have improved compared to those in the matched control area, and the composite result is closer to the fine-scaled mosaic of right-way fire pattern targets. Our approach could be used by Indigenous groups to track performance, and inform annual fire management planning. As the indicators are standardised for rainfall variation, results from multiple sites can be aggregated to track changes in performance at larger scales. Finally, our approach could be adapted for other fire-prone areas, both in Australia and internationally with high spatio-temporal rainfall variability, to improve management planning and evaluation.


Subject(s)
Biodiversity , Ecosystem , Humans , Australia , Poaceae , Time Factors
10.
Immunity ; 38(4): 792-804, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601688

ABSTRACT

Despite accumulating evidence suggesting local self-maintenance of tissue macrophages in the steady state, the dogma remains that tissue macrophages derive from monocytes. Using parabiosis and fate-mapping approaches, we confirmed that monocytes do not show significant contribution to tissue macrophages in the steady state. Similarly, we found that after depletion of lung macrophages, the majority of repopulation occurred by stochastic cellular proliferation in situ in a macrophage colony-stimulating factor (M-Csf)- and granulocyte macrophage (GM)-CSF-dependent manner but independently of interleukin-4. We also found that after bone marrow transplantation, host macrophages retained the capacity to expand when the development of donor macrophages was compromised. Expansion of host macrophages was functional and prevented the development of alveolar proteinosis in mice transplanted with GM-Csf-receptor-deficient progenitors. Collectively, these results indicate that tissue-resident macrophages and circulating monocytes should be classified as mononuclear phagocyte lineages that are independently maintained in the steady state.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lung/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Adult , Animals , Bone Marrow Transplantation , Cell Proliferation , Cell Survival , Cells, Cultured , Homeostasis , Humans , Interleukin-4/metabolism , Macrophages/transplantation , Mice , Mice, Knockout , Mice, Mutant Strains , Parabiosis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
11.
Immunity ; 38(5): 970-83, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706669

ABSTRACT

Mouse and human dendritic cells (DCs) are composed of functionally specialized subsets, but precise interspecies correlation is currently incomplete. Here, we showed that murine lung and gut lamina propria CD11b+ DC populations were comprised of two subsets: FLT3- and IRF4-dependent CD24(+)CD64(-) DCs and contaminating CSF-1R-dependent CD24(-)CD64(+) macrophages. Functionally, loss of CD24(+)CD11b(+) DCs abrogated CD4+ T cell-mediated interleukin-17 (IL-17) production in steady state and after Aspergillus fumigatus challenge. Human CD1c+ DCs, the equivalent of murine CD24(+)CD11b(+) DCs, also expressed IRF4, secreted IL-23, and promoted T helper 17 cell responses. Our data revealed heterogeneity in the mouse CD11b+ DC compartment and identifed mucosal tissues IRF4-expressing DCs specialized in instructing IL-17 responses in both mouse and human. The demonstration of mouse and human DC subsets specialized in driving IL-17 responses highlights the conservation of key immune functions across species and will facilitate the translation of mouse in vivo findings to advance DC-based clinical therapies.


Subject(s)
Aspergillus fumigatus/immunology , Dendritic Cells/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-17/metabolism , Th17 Cells/metabolism , Animals , CD11b Antigen/metabolism , CD24 Antigen/metabolism , Cell Differentiation/immunology , Dendritic Cells/immunology , Humans , Interleukin-17/biosynthesis , Interleukin-23/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Macrophages/metabolism , Mice , Receptors, IgG/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , fms-Like Tyrosine Kinase 3/metabolism
13.
Proc Natl Acad Sci U S A ; 116(43): 21685-21693, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31591213

ABSTRACT

The human dendritic cell (DC) lineage has recently been unraveled by high-dimensional mapping, revealing the existence of a discrete new population of blood circulating DC precursors (pre-DCs). Whether this new DC population possesses specific functional features as compared to the other blood DC subset upon pathogen encounter remained to be evaluated. A unique feature of pre-DCs among blood DCs is their constitutive expression of the viral adhesion receptor Siglec-1. Here, we show that pre-DCs, but not other blood DC subsets, are susceptible to infection by HIV-1 in a Siglec-1-dependent manner. Siglec-1 mediates pre-DC infection of CCR5- and CXCR4-tropic strains. Infection of pre-DCs is further enhanced in the presence of HIV-2/SIVmac Vpx, indicating that Siglec-1 does not counteract restriction factors such as SAMHD1. Instead, Siglec-1 promotes attachment and fusion of viral particles. HIV-1-infected pre-DCs produce new infectious viral particles that accumulate in intracellular compartments reminiscent of the virus-containing compartment of macrophages. Pre-DC activation by toll-like receptor (TLR) ligands induces an antiviral state that inhibits HIV-1 fusion and infection, but Siglec-1 remains functional and mediates replication-independent transfer of HIV-1 to activated primary T lymphocytes. Altogether, Siglec-1-mediated susceptibility to HIV-1 infection of pre-DCs constitutes a unique functional feature that might represent a preferential relationship of this emerging cell type with viruses.


Subject(s)
Dendritic Cells/virology , HIV Infections/transmission , Sialic Acid Binding Ig-like Lectin 1/metabolism , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Dendritic Cells/cytology , HIV Infections/pathology , HIV Infections/virology , HIV-1/immunology , Humans , Sialic Acid Binding Ig-like Lectin 1/biosynthesis , Signal Transduction/immunology , Virus Attachment
14.
Immunity ; 37(1): 60-73, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22795876

ABSTRACT

Dendritic cell (DC)-mediated cross-presentation of exogenous antigens acquired in the periphery is critical for the initiation of CD8(+) T cell responses. Several DC subsets are described in human tissues but migratory cross-presenting DCs have not been isolated, despite their potential importance in immunity to pathogens, vaccines, and tumors and tolerance to self. Here, we identified a CD141(hi) DC present in human interstitial dermis, liver, and lung that was distinct from the majority of CD1c(+) and CD14(+) tissue DCs and superior at cross-presenting soluble antigens. Cutaneous CD141(hi) DCs were closely related to blood CD141(+) DCs, and migratory counterparts were found among skin-draining lymph node DCs. Comparative transcriptomic analysis with mouse showed tissue DC subsets to be conserved between species and permitted close alignment of human and mouse DC subsets. These studies inform the rational design of targeted immunotherapies and facilitate translation of mouse functional DC biology to the human setting.


Subject(s)
Antigens, CD/metabolism , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Animals , Antigens/immunology , Cell Movement/immunology , Chemokine CXCL10/biosynthesis , Gene Expression Profiling , Humans , Immunophenotyping , Langerhans Cells/immunology , Langerhans Cells/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Skin/immunology , Transcriptome , Tumor Necrosis Factor-alpha/biosynthesis
15.
Blood ; 119(25): 6063-71, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22547585

ABSTRACT

The ontogenic relationship between the common dendritic cell (DC) progenitor (CDP), the committed conventional DC precursor (pre-cDC), and cDC subpopulations in lymphoid and nonlymphoid tissues has been largely unraveled. In contrast, the sequential steps of plasmacytoid DC (pDC) development are less defined, and it is unknown at which developmental stage and location final commitment to the pDC lineage occurs. Here we show that CCR9(-) pDCs from murine BM which enter the circulation and peripheral tissues have a common DC precursor function in vivo in the steady state, in contrast to CCR9(+) pDCs which are terminally differentiated. On adoptive transfer, the fate of CCR9(-) pDC-like precursors is governed by the tissues they enter. In the BM and liver, most transferred CCR9(-) pDC-like precursors differentiate into CCR9(+) pDCs, whereas in peripheral lymphoid organs, lung, and intestine, they additionally give rise to cDCs. CCR9(-) pDC-like precursors which are distinct from pre-cDCs can be generated from the CDP. Thus, CCR9(-) pDC-like cells are novel CDP-derived circulating DC precursors with pDC and cDC potential. Their final differentiation into functionally distinct pDCs and cDCs depends on tissue-specific factors allowing adaptation to local requirements under homeostatic conditions.


Subject(s)
Cell Differentiation , Dendritic Cells/physiology , Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cells, Cultured , Dendritic Cells/metabolism , Female , Gene Expression Profiling , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Lymphoid Tissue/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/cytology , Mucous Membrane/metabolism , Mucous Membrane/physiology , Organ Specificity/genetics , Receptors, CCR/metabolism , Specific Pathogen-Free Organisms , Stem Cells/metabolism
16.
Biochimie ; 216: 181-193, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37748748

ABSTRACT

Malassezia globosa is abundant and prevalent on sebaceous areas of the human skin. Genome annotation reveals that M. globosa possesses a repertoire of secreted hydrolytic enzymes relevant for lipid and protein metabolism. However, the functional significance of these enzymes is uncertain and presence of these genes in the genome does not always translate to expression at the cutaneous surface. In this study we utilized targeted RNA sequencing from samples isolated directly from the skin to quantify gene expression of M. globosa secreted proteases, lipases, phospholipases and sphingomyelinases. Our findings indicate that the expression of these enzymes is dynamically regulated by the environment in which the fungus resides, as different growth phases of the planktonic culture of M. globosa show distinct expression levels. Furthermore, we observed significant differences in the expression of these enzymes in culture compared to healthy sebaceous skin sites. By examining the in situ gene expression of M. globosa's secreted hydrolases, we identified a predicted aspartyl protease, MGL_3331, which is highly expressed on both healthy and disease-affected dermatological sites. However, molecular modeling and biochemical studies revealed that this protein has a non-canonical active site motif and lacks measurable proteolytic activity. This pseudoprotease MGL_3331 elicits a heightened IgE-reactivity in blood plasma isolated from patients with atopic dermatitis compared to healthy individuals and invokes a pro-inflammatory response in peripheral blood mononuclear cells. Overall, our study highlights the importance of studying fungal proteins expressed in physiologically relevant environments and underscores the notion that secreted inactive enzymes may have important functions in influencing host immunity.


Subject(s)
Allergens , Malassezia , Humans , Allergens/metabolism , Malassezia/genetics , Malassezia/metabolism , Leukocytes, Mononuclear/metabolism , Skin/metabolism , Lipase/metabolism
17.
Cell Rep ; 42(2): 112046, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36708514

ABSTRACT

The diversity of mononuclear phagocyte (MNP) subpopulations across tissues is one of the key physiological characteristics of the immune system. Here, we focus on understanding the metabolic variability of MNPs through metabolic network analysis applied to three large-scale transcriptional datasets: we introduce (1) an ImmGen MNP open-source dataset of 337 samples across 26 tissues; (2) a myeloid subset of ImmGen Phase I dataset (202 MNP samples); and (3) a myeloid mouse single-cell RNA sequencing (scRNA-seq) dataset (51,364 cells) assembled based on Tabula Muris Senis. To analyze such large-scale datasets, we develop a network-based computational approach, genes and metabolites (GAM) clustering, for unbiased identification of the key metabolic subnetworks based on transcriptional profiles. We define 9 metabolic subnetworks that encapsulate the metabolic differences within MNP from 38 different tissues. Obtained modules reveal that cholesterol synthesis appears particularly active within the migratory dendritic cells, while glutathione synthesis is essential for cysteinyl leukotriene production by peritoneal and lung macrophages.


Subject(s)
Phagocytes , Single-Cell Analysis , Animals , Mice
18.
Eur J Immunol ; 41(8): 2165-75, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21630253

ABSTRACT

The dendritic cells (DCs) present in lymphoid and non-lymphoid organs are generated from progenitors with myeloid-restricted potential. However, in the thymus a major subset of DCs expressing CD8α and langerin (CD207) appears to stand apart from all other DCs in that it is thought to derive from progenitors with lymphoid potential. Using mice expressing a fluorescent reporter and a diphtheria toxin receptor under the control of the cd207 gene, we demonstrated that CD207(+) CD8α(+) thymic DCs do not share a common origin with T cells but originate from intrathymic precursors that express markers that are normally present on all (CD11c(+) and MHCII molecules) or on some (CD207, CD135, CD8α, CX3CR1) DC subsets. Those intrathymic myeloid-type precursors correspond to CD44(+) CD25(-) double-negative 1c (DN1c) cells and are continuously renewed from bone marrow-derived canonical DC precursors. In conclusion, our results demonstrate that the earliest intrathymic precursors of CD8α(+) thymic DCs correspond to myeloid-type DN1c cells and support the view that under physiological conditions myeloid-restricted progenitors generate the whole constellation of DCs present in the body including the thymus.


Subject(s)
CD8 Antigens/immunology , Dendritic Cells/immunology , Myeloid Cells/immunology , Thymus Gland/immunology , Animals , Antigens, Surface/genetics , Antigens, Surface/immunology , Antigens, Surface/metabolism , CD8 Antigens/metabolism , Cell Differentiation/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Male , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Myeloid Cells/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Time Factors
19.
Life Sci Alliance ; 3(1)2020 01.
Article in English | MEDLINE | ID: mdl-31818882

ABSTRACT

Acquired immune responses are initiated by activation of CD4+ helper T (Th) cells via recognition of antigens presented by conventional dendritic cells (cDCs). DCs instruct Th-cell polarization program into specific effector Th subset, which will dictate the type of immune responses. Hence, it is important to unravel how differentiation and/or activation of DC are linked with Th-cell-intrinsic mechanism that directs differentiation toward a specific effector Th subset. Here, we show that loss of Runx/Cbfß transcription factors complexes during DC development leads to loss of CD103+CD11b+ cDC2s and alters characteristics of CD103-CD11b+ cDCs in the intestine, which was accompanied with impaired differentiation of Rorγt+ Th17 cells and type 3 Rorγt+ regulatory T cells. We also show that a Runx-binding enhancer in the Rorc gene is essential for T cells to integrate cDC-derived signals to induce Rorγt expression. These findings reveal that Runx/Cbfß complexes play crucial and complementary roles in cDCs and Th cells to shape converging type 3 immune responses.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , Dendritic Cells/metabolism , Intestinal Mucosa/cytology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Adaptive Immunity , Animals , Cell Differentiation/immunology , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor beta Subunit/genetics , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
20.
Methods Mol Biol ; 2034: 217-230, 2019.
Article in English | MEDLINE | ID: mdl-31392688

ABSTRACT

Microglia are the main population of macrophage residing in the central nervous system (CNS). Depletion experiments gave important insights into the physiology and function of microglia in healthy and diseased CNS. Ablation of microglia can be achieved by application of pharmacological or genetic tools. Here, we describe two approaches to ablate microglia: an efficient genetic model that utilizes DTRMG mouse line that has diphtheria toxin receptor (DTR) expression regulated by the promoter activity of the fractalkine receptor (CX3CR1) gene, and a pharmacological model that utilizes the blocking of macrophage colony-stimulating factor 1 receptor (CSF-1R) with a blocking antibody. Both the administration of systemic diphtheria toxin or anti-CSF-1R blocking antibody result in highly efficient and reversible depletion of microglia population in the CNS, which can be easily assessed by flow cytometry.


Subject(s)
Antibodies, Blocking/pharmacology , CX3C Chemokine Receptor 1/genetics , Diphtheria Toxin/pharmacology , Gene Expression , Heparin-binding EGF-like Growth Factor , Microglia , Promoter Regions, Genetic , Receptor, Macrophage Colony-Stimulating Factor , Animals , Heparin-binding EGF-like Growth Factor/biosynthesis , Heparin-binding EGF-like Growth Factor/genetics , Mice , Mice, Transgenic , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL