Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Nat Immunol ; 25(6): 994-1006, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38671323

ABSTRACT

The lung is constantly exposed to the outside world and optimal adaptation of immune responses is crucial for efficient pathogen clearance. However, mechanisms that lead to lung-associated macrophages' functional and developmental adaptation remain elusive. To reveal such mechanisms, we developed a reductionist model of environmental intranasal ß-glucan exposure, allowing for the detailed interrogation of molecular mechanisms of pulmonary macrophage adaptation. Employing single-cell transcriptomics, high-dimensional imaging and flow cytometric characterization paired with in vivo and ex vivo challenge models, we reveal that pulmonary low-grade inflammation results in the development of apolipoprotein E (ApoE)-dependent monocyte-derived alveolar macrophages (ApoE+CD11b+ AMs). ApoE+CD11b+ AMs expressed high levels of CD11b, ApoE, Gpnmb and Ccl6, were glycolytic, highly phagocytic and produced large amounts of interleukin-6 upon restimulation. Functional differences were cell intrinsic, and myeloid cell-specific ApoE ablation inhibited Ly6c+ monocyte to ApoE+CD11b+ AM differentiation dependent on macrophage colony-stimulating factor secretion, promoting ApoE+CD11b+ AM cell death and thus impeding ApoE+CD11b+ AM maintenance. In vivo, ß-glucan-elicited ApoE+CD11b+ AMs limited the bacterial burden of Legionella pneumophilia after infection and improved the disease outcome in vivo and ex vivo in a murine lung fibrosis model. Collectively these data identify ApoE+CD11b+ AMs generated upon environmental cues, under the control of ApoE signaling, as an essential determinant for lung adaptation enhancing tissue resilience.


Subject(s)
Apolipoproteins E , Lectins, C-Type , Macrophages, Alveolar , Mice, Inbred C57BL , beta-Glucans , Animals , Mice , Adaptation, Physiological/immunology , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , CD11b Antigen/metabolism , Cell Differentiation , Lectins, C-Type/metabolism , Lung/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism
3.
Nat Commun ; 12(1): 4434, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290249

ABSTRACT

Dyslipidemia is a main driver of cardiovascular diseases. The ability of macrophages to scavenge excess lipids implicate them as mediators in this process and understanding the mechanisms underlying macrophage lipid metabolism is key to the development of new treatments. Here, we investigated how adipose tissue macrophages regulate post-prandial cholesterol transport. Single-cell RNA sequencing and protected bone marrow chimeras demonstrated that ingestion of lipids led to specific transcriptional activation of a population of resident macrophages expressing Lyve1, Tim4, and ABCA1. Blocking the phosphatidylserine receptor Tim4 inhibited lysosomal activation and the release of post-prandial high density lipoprotein cholesterol following a high fat meal. Both effects were recapitulated by chloroquine, an inhibitor of lysosomal function. Moreover, clodronate-mediated cell-depletion implicated Tim4+ resident adipose tissue macrophages in this process. Thus, these data indicate that Tim4 is a key regulator of post-prandial cholesterol transport and adipose tissue macrophage function and may represent a novel pathway to treat dyslipidemia.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Adipose Tissue/metabolism , Cholesterol/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , Postprandial Period/physiology , Adipose Tissue/cytology , Animals , Cholesterol, HDL/metabolism , Diet, High-Fat , Lipid Metabolism , Lysosomes/metabolism , Macrophages/cytology , Mice , Obesity/metabolism , Obesity/pathology , Transcriptional Activation , Vesicular Transport Proteins/metabolism
4.
Nat Commun ; 12(1): 1770, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741914

ABSTRACT

Inflammation generally leads to recruitment of monocyte-derived macrophages. What regulates the fate of these cells and to what extent they can assume the identity and function of resident macrophages is unclear. Here, we show that macrophages elicited into the peritoneal cavity during mild inflammation persist long-term but are retained in an immature transitory state of differentiation due to the presence of enduring resident macrophages. By contrast, severe inflammation results in ablation of resident macrophages and a protracted phase wherein the cavity is incapable of sustaining a resident phenotype, yet ultimately elicited cells acquire a mature resident identity. These macrophages also have transcriptionally and functionally divergent features that result from inflammation-driven alterations to the peritoneal cavity micro-environment and, to a lesser extent, effects of origin and time-of-residency. Hence, rather than being predetermined, the fate of inflammation-elicited peritoneal macrophages seems to be regulated by the environment.


Subject(s)
Cell Differentiation/genetics , Inflammation/genetics , Macrophages, Peritoneal/metabolism , Macrophages/metabolism , Peritoneal Cavity/pathology , Animals , Cells, Cultured , Cytokines/metabolism , Female , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Inflammation/metabolism , Macrophages/cytology , Macrophages, Peritoneal/cytology , Male , Mice, Congenic , Mice, Inbred C57BL , Peritonitis/genetics , Peritonitis/metabolism
5.
Sci Immunol ; 5(48)2020 06 19.
Article in English | MEDLINE | ID: mdl-32561560

ABSTRACT

Macrophages reside in the body cavities where they maintain serosal homeostasis and provide immune surveillance. Peritoneal macrophages are implicated in the etiology of pathologies including peritonitis, endometriosis, and metastatic cancer; thus, understanding the factors that govern their behavior is vital. Using a combination of fate mapping techniques, we have investigated the impact of sex and age on murine peritoneal macrophage differentiation, turnover, and function. We demonstrate that the sexually dimorphic replenishment of peritoneal macrophages from the bone marrow, which is high in males and very low in females, is driven by changes in the local microenvironment that arise upon sexual maturation. Population and single-cell RNA sequencing revealed marked dimorphisms in gene expression between male and female peritoneal macrophages that was, in part, explained by differences in composition of these populations. By estimating the time of residency of different subsets within the cavity and assessing development of dimorphisms with age and in monocytopenic Ccr2 -/- mice, we demonstrate that key sex-dependent features of peritoneal macrophages are a function of the differential rate of replenishment from the bone marrow, whereas others are reliant on local microenvironment signals. We demonstrate that the dimorphic turnover of peritoneal macrophages contributes to differences in the ability to protect against pneumococcal peritonitis between the sexes. These data highlight the importance of considering both sex and age in susceptibility to inflammatory and infectious diseases.


Subject(s)
Macrophages, Peritoneal/immunology , Sex Characteristics , Animals , Cell Differentiation/immunology , Female , Homeostasis/immunology , Male , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA/genetics , RNA/immunology , Sequence Analysis, RNA , Single-Cell Analysis
6.
Clin Exp Immunol ; 199(1): 9-11, 2020 01.
Article in English | MEDLINE | ID: mdl-31777062

ABSTRACT

The field of mucosal immunology has, for the last 10 years, been largely dominated by advances in our understanding of the commensal microbiota. Developments of novel experimental methodologies and analysis techniques have provided unparalleled insight into the profound impact the microbiota has on the development and function of the immune system. In this cross-journal review series published in Immunology and Clinical and Experimental Immunology, we aim to summarize the current state of research concerning the interplay between the microbiota and mucosal immunity. In addition, the series examines how the increased understanding of the microbiota is changing the nature of immunological research, both in the laboratory and in the clinic.


Subject(s)
Biomedical Research , Immunity, Mucosal , Animals , Humans , Periodicals as Topic
7.
Nat Commun ; 8(1): 620, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28931816

ABSTRACT

CD103+CD11b+ dendritic cells (DCs) are unique to the intestine, but the factors governing their differentiation are unclear. Here we show that transforming growth factor receptor 1 (TGFßR1) has an indispensable, cell intrinsic role in the development of these cells. Deletion of Tgfbr1 results in markedly fewer intestinal CD103+CD11b+ DCs and a reciprocal increase in the CD103-CD11b+ dendritic cell subset. Transcriptional profiling identifies markers that define the CD103+CD11b+ DC lineage, including CD101, TREM1 and Siglec-F, and shows that the absence of CD103+CD11b+ DCs in CD11c-Cre.Tgfbr1 fl/fl mice reflects defective differentiation from CD103-CD11b+ intermediaries, rather than an isolated loss of CD103 expression. The defect in CD103+CD11b+ DCs is accompanied by reduced generation of antigen-specific, inducible FoxP3+ regulatory T cells in vitro and in vivo, and by reduced numbers of endogenous Th17 cells in the intestinal mucosa. Thus, TGFßR1-mediated signalling may explain the tissue-specific development of these unique DCs.Developmental cues for the different dendritic cell (DC) subsets in the intestine are yet to be defined. Here the authors show that TGFßR1 signalling is needed for development of CD103+CD11b+ intestinal DCs from CD103-CD11b+ cells and that they contribute to the generation of Th17 and regulatory T cells.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Antigens, CD/immunology , CD11b Antigen/immunology , Cell Lineage , Colitis/immunology , Dendritic Cells/cytology , Immunity, Mucosal , Integrin alpha Chains/immunology , Intestinal Mucosa/cytology , Intestines/cytology , Intestines/immunology , Lymphopoiesis/genetics , Mice , Mice, Knockout , Receptor, Transforming Growth Factor-beta Type I , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology
8.
Mucosal Immunol ; 10(6): 1387-1399, 2017 11.
Article in English | MEDLINE | ID: mdl-28145440

ABSTRACT

Intestinal macrophages (mφ) form one of the largest populations of mφ in the body and are vital for the maintenance of gut homeostasis. They have several unique properties and are derived from local differentiation of classical Ly6Chi monocytes, but the factors driving this tissue-specific process are not understood. Here we have used global transcriptomic analysis to identify a unique homeostatic signature of mature colonic mφ that is acquired as they differentiate in the mucosa. By comparing the analogous monocyte differentiation process found in the dermis, we identify TGFß as an indispensable part of monocyte differentiation in the intestine and show that it enables mφ to adapt precisely to the requirements of their environment. Importantly, TGFßR signaling on mφ has a crucial role in regulating the accumulation of monocytes in the mucosa, via mechanisms that are distinct from those used by IL10.


Subject(s)
Colon/immunology , Macrophages/immunology , Monocytes/immunology , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antigens, Ly/metabolism , Cell Differentiation , Cells, Cultured , Cellular Microenvironment , Female , Gene Expression Profiling , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Transcriptome
9.
Mucosal Immunol ; 9(2): 492-502, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26349660

ABSTRACT

Dectin-1 is an innate antifungal C-type lectin receptor necessary for protective antifungal immunity. We recently discovered that Dectin-1 is involved in controlling fungal infections of the gastrointestinal (GI) tract, but how this C-type lectin receptor mediates these activities is unknown. Here, we show that Dectin-1 is essential for driving fungal-specific CD4(+) T-cell responses in the GI tract. Loss of Dectin-1 resulted in abrogated dendritic cell responses in the mesenteric lymph nodes (mLNs) and defective T-cell co-stimulation, causing substantial increases in CD4(+) T-cell apoptosis and reductions in the cellularity of GI-associated lymphoid tissues. CD8(+) T-cell responses were unaffected by Dectin-1 deficiency. These functions of Dectin-1 have significant implications for our understanding of intestinal immunity and susceptibility to fungal infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Candida albicans/immunology , Candidiasis/immunology , Gastrointestinal Tract/immunology , Lectins, C-Type/immunology , Adoptive Transfer , Animals , Apoptosis/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , Candidiasis/genetics , Candidiasis/microbiology , Candidiasis/pathology , Cell Survival/immunology , Female , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Gene Expression , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Lymph Nodes/immunology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Lymphocyte Activation , Mesentery/immunology , Mesentery/microbiology , Mesentery/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Mucosal Immunol ; 8(1): 38-48, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24850430

ABSTRACT

Cross-presentation of cellular antigens is crucial for priming CD8(+) T cells, and generating immunity to intracellular pathogens--particularly viruses. It is unclear which intestinal phagocytes perform this function in vivo. To address this, we examined dendritic cells (DCs) from the intestinal lymph of IFABP-tOVA 232-4 mice, which express ovalbumin in small intestinal epithelial cells (IECs). Among lymph DCs (LDCs) only CD103(+) CD11b(-) CD8α(+) DCs cross-present IEC-derived ovalbumin to CD8(+) OT-I T cells. Similarly, in the mesenteric lymph nodes (MLNs), cross-presentation of IEC-ovalbumin was limited to the CD11c(+) MHCII(hi) CD8α(+) migratory DCs, but absent from all other subsets, including the resident CD8α(hi) DCs. Crucially, delivery of purified CD8α(+) LDCs, but not other LDC subsets, into the MLN subcapsular lymphatic sinus induced proliferation of ovalbumin-specific, gut-tropic CD8(+) T cells in vivo. Finally, in 232-4 mice treated with R848, CD8α(+) LDCs were uniquely able to cross-prime interferon γ-producing CD8(+) T cells and drive their migration to the intestine. Our results clearly demonstrate that migrating CD8α(+) intestinal DCs are indispensable for cross-presentation of cellular antigens and, in conditions of inflammation, for the initial differentiation of effector CD8(+) T cells. They may therefore represent an important target for the development of antiviral vaccinations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Ovalbumin/metabolism , Animals , Antigens/immunology , CD8 Antigens/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cross-Priming/drug effects , Cross-Priming/genetics , Imidazoles/administration & dosage , Imidazoles/pharmacology , Interferon-gamma/metabolism , Intestinal Mucosa/immunology , Lymph/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Membrane Glycoproteins/agonists , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Toll-Like Receptor 7/agonists
11.
Mucosal Immunol ; 8(2): 327-39, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25138666

ABSTRACT

The identification of intestinal macrophages (mφs) and dendritic cells (DCs) is a matter of intense debate. Although CD103(+) mononuclear phagocytes (MPs) appear to be genuine DCs, the nature and origins of CD103(-) MPs remain controversial. We show here that intestinal CD103(-)CD11b(+) MPs can be separated clearly into DCs and mφs based on phenotype, gene profile, and kinetics. CD64(-)CD103(-)CD11b(+) MPs are classical DCs, being derived from Flt3 ligand-dependent, DC-committed precursors, not Ly6C(hi) monocytes. Surprisingly, a significant proportion of these CD103(-)CD11b(+) DCs express CCR2 and there is a selective decrease in CD103(-)CD11b(+) DCs in mice lacking this chemokine receptor. CCR2(+)CD103(-) DCs are present in both the murine and human intestine, drive interleukin (IL)-17a production by T cells in vitro, and show constitutive expression of IL-12/IL-23p40. These data highlight the heterogeneity of intestinal DCs and reveal a bona fide population of CCR2(+) DCs that is involved in priming mucosal T helper type 17 (Th17) responses.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/metabolism , Animals , Antigens, CD/metabolism , Dendritic Cells/metabolism , Humans , Immunophenotyping , Integrin alpha Chains/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-12/metabolism , Interleukin-17/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Phenotype , Receptors, CCR2/metabolism , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology
12.
Mucosal Immunol ; 6(3): 498-510, 2013 May.
Article in English | MEDLINE | ID: mdl-22990622

ABSTRACT

Macrophages (mφ) are essential for intestinal homeostasis and the pathology of inflammatory bowel disease (IBD), but it is unclear whether discrete mφ populations carry out these distinct functions or if resident mφ change during inflammation. We show here that most resident mφ in resting mouse colon express very high levels of CX3CR1, are avidly phagocytic and MHCII(hi), but are resistant to Toll-like receptor (TLR) stimulation, produce interleukin 10 constitutively, and express CD163 and CD206. A smaller population of CX3CR1(int) cells is present in resting colon and it expands during experimental colitis. Ly6C(hi)CCR2(+) monocytes can give rise to all mφ subsets in both healthy and inflamed colon and we show that the CX3CR1(int) pool represents a continuum in which newly arrived, recently divided monocytes develop into resident CX3CR1(hi) mφ. This process is arrested during experimental colitis, resulting in the accumulation of TLR-responsive pro-inflammatory mφ. Phenotypic analysis of human intestinal mφ indicates that analogous processes occur in the normal and Crohn's disease ileum. These studies show for the first time that resident and inflammatory mφ in the intestine represent alternative differentiation outcomes of the same precursor and targeting these events could offer routes for therapeutic intervention in IBD.


Subject(s)
Colitis/immunology , Colon/immunology , Inflammatory Bowel Diseases/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Antigens, Ly/metabolism , CX3C Chemokine Receptor 1 , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Colitis/chemically induced , Histocompatibility Antigens Class II/metabolism , Humans , Inflammation/pathology , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL