Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Nat Immunol ; 23(3): 458-468, 2022 03.
Article in English | MEDLINE | ID: mdl-35210623

ABSTRACT

Alveolar macrophages (AMs) are lung tissue-resident macrophages that can be expanded in culture, but it is unknown to what extent culture affects their in vivo identity. Here we show that mouse long-term ex vivo expanded AMs (exAMs) maintained a core AM gene expression program, but showed culture adaptations related to adhesion, metabolism and proliferation. Upon transplantation into the lung, exAMs reacquired full transcriptional and epigenetic AM identity, even after several months in culture and could self-maintain long-term in the alveolar niche. Changes in open chromatin regions observed in culture were fully reversible in transplanted exAMs and resulted in a gene expression profile indistinguishable from resident AMs. Our results indicate that long-term proliferation of AMs in culture did not compromise cellular identity in vivo. The robustness of exAM identity provides new opportunities for mechanistic analysis and highlights the therapeutic potential of exAMs.


Subject(s)
Lung , Macrophages, Alveolar , Animals , Chromatin/metabolism , Epigenesis, Genetic , Epigenomics , Lung/metabolism , Macrophages, Alveolar/metabolism , Mice
2.
Nat Immunol ; 20(3): 374, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30733606

ABSTRACT

In the version of this article initially published, the Supplementary Data file was an incorrect version. The correct version is now provided. The error has been corrected in the HTML and PDF version of the article.

3.
Cell ; 167(5): 1264-1280.e18, 2016 11 17.
Article in English | MEDLINE | ID: mdl-28084216

ABSTRACT

Granulomas are immune cell aggregates formed in response to persistent inflammatory stimuli. Granuloma macrophage subsets are diverse and carry varying copy numbers of their genomic information. The molecular programs that control the differentiation of such macrophage populations in response to a chronic stimulus, though critical for disease outcome, have not been defined. Here, we delineate a macrophage differentiation pathway by which a persistent Toll-like receptor (TLR) 2 signal instructs polyploid macrophage fate by inducing replication stress and activating the DNA damage response. Polyploid granuloma-resident macrophages formed via modified cell divisions and mitotic defects and not, as previously thought, by cell-to-cell fusion. TLR2 signaling promoted macrophage polyploidy and suppressed genomic instability by regulating Myc and ATR. We propose that, in the presence of persistent inflammatory stimuli, pathways previously linked to oncogene-initiated carcinogenesis instruct a long-lived granuloma-resident macrophage differentiation program that regulates granulomatous tissue remodeling.


Subject(s)
DNA Damage , Granuloma/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Differentiation , Cell Proliferation , Humans , Inflammation/immunology , Lipoproteins/immunology , Mice , Mice, Inbred C57BL , Mitosis , Proto-Oncogene Proteins c-myc/metabolism , Toll-Like Receptor 2
4.
Nat Immunol ; 19(5): 497-507, 2018 05.
Article in English | MEDLINE | ID: mdl-29662170

ABSTRACT

The transcription factor c-Maf induces the anti-inflammatory cytokine IL-10 in CD4+ T cells in vitro. However, the global effects of c-Maf on diverse immune responses in vivo are unknown. Here we found that c-Maf regulated IL-10 production in CD4+ T cells in disease models involving the TH1 subset of helper T cells (malaria), TH2 cells (allergy) and TH17 cells (autoimmunity) in vivo. Although mice with c-Maf deficiency targeted to T cells showed greater pathology in TH1 and TH2 responses, TH17 cell-mediated pathology was reduced in this context, with an accompanying decrease in TH17 cells and increase in Foxp3+ regulatory T cells. Bivariate genomic footprinting elucidated the c-Maf transcription-factor network, including enhanced activity of NFAT; this led to the identification and validation of c-Maf as a negative regulator of IL-2. The decreased expression of the gene encoding the transcription factor RORγt (Rorc) that resulted from c-Maf deficiency was dependent on IL-2, which explained the in vivo observations. Thus, c-Maf is a positive and negative regulator of the expression of cytokine-encoding genes, with context-specific effects that allow each immune response to occur in a controlled yet effective manner.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Interleukin-2/biosynthesis , Proto-Oncogene Proteins c-maf/immunology , Animals , Interleukin-2/immunology , Mice
9.
Immunity ; 42(1): 10-2, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607453

ABSTRACT

Liver Kupffer cells (KCs) are self-maintained tissue-resident macrophages. In this issue of Immunity, Blériot et al. demonstrate that bacterial infection leads to KC necroptosis and quantitative replacement by monocyte-derived macrophages that contribute to antibacterial immunity and restoration of tissue integrity.


Subject(s)
Kupffer Cells/physiology , Listeria monocytogenes/immunology , Listeriosis/immunology , Liver/pathology , Monocytes/immunology , Animals
10.
Cell ; 138(2): 300-13, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19632180

ABSTRACT

While hematopoietic stem cell (HSC) self-renewal is well studied, it remains unknown whether distinct control mechanisms enable HSC divisions that generate progeny cells with specific lineage bias. Here, we report that the monocytic transcription factor MafB specifically restricts the ability of M-CSF to instruct myeloid commitment divisions in HSCs. MafB deficiency specifically enhanced sensitivity to M-CSF and caused activation of the myeloid master-regulator PU.1 in HSCs in vivo. Single-cell analysis revealed that reduced MafB levels enabled M-CSF to instruct divisions producing asymmetric daughter pairs with one PU.1(+) cell. As a consequence, MafB(-/-) HSCs showed a PU.1 and M-CSF receptor-dependent competitive repopulation advantage specifically in the myelomonocytic, but not T lymphoid or erythroid, compartment. Lineage-biased repopulation advantage was progressive, maintained long term, and serially transplantable. Together, this indicates that an integrated transcription factor/cytokine circuit can control the rate of specific HSC commitment divisions without compromising other lineages or self-renewal.


Subject(s)
Cell Lineage , Hematopoietic Stem Cells/cytology , Macrophage Colony-Stimulating Factor/metabolism , MafB Transcription Factor/metabolism , Myeloid Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Trans-Activators/metabolism
11.
EMBO J ; 38(19): e101233, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31414712

ABSTRACT

Tissues in multicellular organisms are populated by resident macrophages, which perform both generic and tissue-specific functions. The latter are induced by signals from the microenvironment and rely on unique tissue-specific molecular programs requiring the combinatorial action of tissue-specific and broadly expressed transcriptional regulators. Here, we identify the transcription factors Bhlhe40 and Bhlhe41 as novel regulators of alveolar macrophages (AMs)-a population that provides the first line of immune defense and executes homeostatic functions in lung alveoli. In the absence of these factors, AMs exhibited decreased proliferation that resulted in a severe disadvantage of knockout AMs in a competitive setting. Gene expression analyses revealed a broad cell-intrinsic footprint of Bhlhe40/Bhlhe41 deficiency manifested by a downregulation of AM signature genes and induction of signature genes of other macrophage lineages. Genome-wide characterization of Bhlhe40 DNA binding suggested that these transcription factors directly repress the expression of lineage-inappropriate genes in AMs. Taken together, these results identify Bhlhe40 and Bhlhe41 as key regulators of AM self-renewal and guardians of their identity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Profiling/methods , Homeodomain Proteins/genetics , Macrophages, Alveolar/cytology , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Cell Survival , Down-Regulation , Gene Knockdown Techniques , Histones/metabolism , Homeodomain Proteins/metabolism , Macrophages, Alveolar/metabolism , Mice , Organ Specificity , Phenotype , Sequence Analysis, RNA
12.
Nat Immunol ; 17(6): 609-11, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27196510

Subject(s)
Eosinophils , Mast Cells , Humans
13.
EMBO J ; 36(16): 2353-2372, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28701484

ABSTRACT

Mature differentiated macrophages can self-maintain by local proliferation in tissues and can be extensively expanded in culture under specific conditions, but the mechanisms of this phenomenon remain only partially defined. Here, we show that SIRT1, an evolutionary conserved regulator of life span, positively affects macrophage self-renewal ability in vitro and in vivo Overexpression of SIRT1 during bone marrow-derived macrophage differentiation increased their proliferative capacity. Conversely, decrease of SIRT1 expression by shRNA inactivation, CRISPR/Cas9 mediated deletion and pharmacological inhibition restricted macrophage self-renewal in culture. Furthermore, pharmacological SIRT1 inhibition in vivo reduced steady state and cytokine-induced proliferation of alveolar and peritoneal macrophages. Mechanistically, SIRT1 inhibition negatively regulated G1/S transition, cell cycle progression and a network of self-renewal genes. This included inhibition of E2F1 and Myc and concomitant activation of FoxO1, SIRT1 targets mediating cell cycle progression and stress response, respectively. Our findings indicate that SIRT1 is a key regulator of macrophage self-renewal that integrates cell cycle and longevity pathways. This suggests that macrophage self-renewal might be a relevant parameter of ageing.


Subject(s)
Cell Proliferation , Cell Self Renewal , Macrophages/physiology , Sirtuin 1/metabolism , Animals , Cell Cycle , Gene Expression , Gene Knockdown Techniques , Gene Knockout Techniques , Mice , Sirtuin 1/genetics
14.
EMBO J ; 34(15): 2042-58, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26139534

ABSTRACT

T cells infiltrating neoplasms express surface molecules typical of chronically virus-stimulated T cells, often termed "exhausted" T cells. We compared the transcriptome of "exhausted" CD8 T cells infiltrating autochthonous melanomas to those of naïve and acutely stimulated CD8 T cells. Despite strong similarities between transcriptional signatures of tumor- and virus-induced exhausted CD8 T cells, notable differences appeared. Among transcriptional regulators, Nr4a2 and Maf were highly overexpressed in tumor-exhausted T cells and significantly upregulated in CD8 T cells from human melanoma metastases. Transduction of murine tumor-specific CD8 T cells to express Maf partially reproduced the transcriptional program associated with tumor-induced exhaustion. Upon adoptive transfer, the transduced cells showed normal homeostasis but failed to accumulate in tumor-bearing hosts and developed defective anti-tumor effector responses. We further identified TGFß and IL-6 as main inducers of Maf expression in CD8 T cells and showed that Maf-deleted tumor-specific CD8 T cells were much more potent to restrain tumor growth in vivo. Therefore, the melanoma microenvironment contributes to skewing of CD8 T cell differentiation programs, in part by TGFß/IL-6-mediated induction of Maf.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Melanoma/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Tumor Microenvironment/physiology , Animals , CD8-Positive T-Lymphocytes/metabolism , DNA Primers/genetics , Flow Cytometry , Gene Expression Profiling , Homeodomain Proteins/genetics , Interleukin-6/metabolism , Luciferases , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-maf/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/metabolism
15.
Nature ; 497(7448): 239-43, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23575636

ABSTRACT

Under stress conditions such as infection or inflammation the body rapidly needs to generate new blood cells that are adapted to the challenge. Haematopoietic cytokines are known to increase output of specific mature cells by affecting survival, expansion and differentiation of lineage-committed progenitors, but it has been debated whether long-term haematopoietic stem cells (HSCs) are susceptible to direct lineage-specifying effects of cytokines. Although genetic changes in transcription factor balance can sensitize HSCs to cytokine instruction, the initiation of HSC commitment is generally thought to be triggered by stochastic fluctuation in cell-intrinsic regulators such as lineage-specific transcription factors, leaving cytokines to ensure survival and proliferation of the progeny cells. Here we show that macrophage colony-stimulating factor (M-CSF, also called CSF1), a myeloid cytokine released during infection and inflammation, can directly induce the myeloid master regulator PU.1 and instruct myeloid cell-fate change in mouse HSCs, independently of selective survival or proliferation. Video imaging and single-cell gene expression analysis revealed that stimulation of highly purified HSCs with M-CSF in culture resulted in activation of the PU.1 promoter and an increased number of PU.1(+) cells with myeloid gene signature and differentiation potential. In vivo, high systemic levels of M-CSF directly stimulated M-CSF-receptor-dependent activation of endogenous PU.1 protein in single HSCs and induced a PU.1-dependent myeloid differentiation preference. Our data demonstrate that lineage-specific cytokines can act directly on HSCs in vitro and in vivo to instruct a change of cell identity. This fundamentally changes the current view of how HSCs respond to environmental challenge and implicates stress-induced cytokines as direct instructors of HSC fate.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Myeloid Cells/cytology , Myeloid Cells/drug effects , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Single-Cell Analysis , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/metabolism
16.
Proc Natl Acad Sci U S A ; 113(44): 12514-12519, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27729526

ABSTRACT

Applying clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9)-mediated mutagenesis to primary mouse immune cells, we used high-fidelity single guide RNAs (sgRNAs) designed with an sgRNA design tool (CrispRGold) to target genes in primary B cells, T cells, and macrophages isolated from a Cas9 transgenic mouse line. Using this system, we achieved an average knockout efficiency of 80% in B cells. On this basis, we established a robust small-scale CRISPR-mediated screen in these cells and identified genes essential for B-cell activation and plasma cell differentiation. This screening system does not require deep sequencing and may serve as a precedent for the application of CRISPR/Cas9 to primary mouse cells.


Subject(s)
B-Lymphocytes/metabolism , CRISPR-Cas Systems , Gene Editing/methods , Macrophages/metabolism , Mutagenesis , T-Lymphocytes/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Lymphocyte Activation/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Plasma Cells/metabolism , Reproducibility of Results
17.
Cell Immunol ; 330: 120-125, 2018 08.
Article in English | MEDLINE | ID: mdl-29650243

ABSTRACT

Macrophages are innate immune cells present in essentially every organ of the body with dedicated tissue specific functions. We will present in this review the unique properties and functions of macrophage populations residing in the testis, an immune-privileged organ. Testicular macrophages (tMΦ) could be seen as guardians of fertility due to their immunosuppressive functions protecting spermatogenesis from auto immune-attack. They exhibit testis specific functions with essential roles in normal testis homeostasis and fetal testicular development. Recently, two distinct testicular macrophage populations have been characterized based on different localization, morphology, gene expression profiles, developmental origin and postnatal development. We will discuss the importance of these two testicular macrophage populations for organ specific functions such as testosterone production and spermatogenesis, as well as their role in establishing immuno-privilege highlighting the contributions of macrophages to male fertility.


Subject(s)
Fertility/immunology , Macrophages/immunology , Testis/immunology , Testosterone/immunology , Animals , Humans , Immunity/immunology , Macrophages/metabolism , Male , Models, Immunological , Spermatogenesis/immunology , Testis/cytology , Testis/metabolism , Testosterone/metabolism
18.
Immunol Rev ; 262(1): 56-73, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25319327

ABSTRACT

Macrophages are cellular components of the innate immune system that reside in virtually all tissues and contribute to immunity, repair, and homeostasis. The traditional view that all tissue-resident macrophages derive from the bone marrow through circulating monocyte intermediates has dramatically shifted recently with the observation that macrophages from embryonic progenitors can persist into adulthood and self-maintain by local proliferation. In several tissues, however, monocytes also contribute to the resident macrophage population, on which the local environment can impose tissue-specific macrophage functions. These observations have raised important questions: What determines resident macrophage identity and function, ontogeny or environment? How is macrophage proliferation regulated? In this review, we summarize the current knowledge about the identity, proliferation, and turnover of tissue-resident macrophages and how they differ from freshly recruited short-lived monocyte-derived cells. We examine whether macrophage proliferation can be qualified as self-renewal of mature differentiated cells and whether the concepts and molecular pathways are comparable to self-renewal mechanisms in stem cells. Finally, we discuss how improved understanding of macrophage identity and self-renewal could be exploited for therapeutic intervention of macrophage-mediated pathologies by selectively targeting freshly recruited or resident macrophages.


Subject(s)
Macrophages/cytology , Macrophages/metabolism , Phenotype , Animals , Cell Differentiation , Cell Proliferation , Humans , Macrophages/immunology , Organ Specificity
19.
Methods Mol Biol ; 2713: 231-251, 2024.
Article in English | MEDLINE | ID: mdl-37639127

ABSTRACT

Alveolar macrophages (AM) are resident macrophages of the lung and play important roles in the maintenance of tissue homeostasis as well as host defense. Here, we describe how they can be harvested from murine lungs, expanded in vitro, and transduced with lentiviral vectors.


Subject(s)
Macrophages, Alveolar , Macrophages , Animals , Mice , Thorax
20.
Exp Hematol ; 121: 6-11, 2023 05.
Article in English | MEDLINE | ID: mdl-36764598

ABSTRACT

Immunologic memory is a feature typically ascribed to the adaptive arm of the immune system. However, recent studies have demonstrated that hematopoietic stem cells (HSCs) and innate immune cells such as monocytes and macrophages can gain epigenetic signatures to enhance their response in the context of reinfection. This suggests the presence of long-term memory, a phenomenon referred to as trained immunity. Trained immunity in HSCs can occur via changes in the epigenetic landscape and enhanced chromatin accessibility in lineage-specific genes, as well as through metabolic alterations. These changes can lead to a skewing in lineage bias, particularly enhanced myelopoiesis and the generation of epigenetically modified innate immune cells that provide better protection against pathogens on secondary infection. Here, we summarize recent advancements in trained immunity and epigenetic memory formation in HSCs and self-renewing alveolar macrophages, which was the focus of the Spring 2022 International Society for Experimental Hematology (ISEH) webinar.


Subject(s)
Immunity, Innate , Trained Immunity , Immunity, Innate/genetics , Epigenetic Memory , Macrophages , Immunologic Memory/genetics
SELECTION OF CITATIONS
SEARCH DETAIL