Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 34: 93-119, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26735697

ABSTRACT

The dendritic cells (DCs) of the immune system function in innate and adaptive responses by directing activity of various effector cells rather than serving as effectors themselves. DCs and closely related myeloid lineages share expression of many surface receptors, presenting a challenge in distinguishing their unique in vivo functions. Recent work has taken advantage of unique transcriptional programs to identify and manipulate murine DCs in vivo. This work has assigned several nonredundant in vivo functions to distinct DC lineages, consisting of plasmacytoid DCs and several subsets of classical DCs that promote different immune effector modules in response to pathogens. In parallel, a correspondence between human and murine DC subsets has emerged, underlying structural similarities for the DC lineages between these species. Recent work has begun to unravel the transcriptional circuitry that controls the development and diversification of DCs from common progenitors in the bone marrow.


Subject(s)
Bone Marrow Cells/physiology , Dendritic Cells/physiology , Gene Expression Regulation , Immunity, Cellular , Animals , Cell Differentiation , Cell Lineage , Gene Expression Profiling , Gene Regulatory Networks , Humans , Immunity, Cellular/genetics , Mice , Transcriptional Activation
2.
Nat Immunol ; 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39375550

ABSTRACT

Autoactivation of lineage-determining transcription factors mediates bistable expression, generating distinct cell phenotypes essential for complex body plans. Classical type 1 dendritic cell (cDC1) and type 2 dendritic cell (cDC2) subsets provide nonredundant functions for defense against distinct immune challenges. Interferon regulatory factor 8 (IRF8), the cDC1 lineage-determining transcription factor, undergoes autoactivation in cDC1 progenitors to establish cDC1 identity, yet its expression is downregulated during cDC2 differentiation by an unknown mechanism. This study reveals that the Irf8 +32-kb enhancer, responsible for IRF8 autoactivation, is naturally suboptimized with low-affinity IRF8 binding sites. Introducing multiple high-affinity IRF8 sites into the Irf8 +32-kb enhancer causes a gain-of-function effect, leading to erroneous IRF8 autoactivation in specified cDC2 progenitors, redirecting them toward cDC1 and a novel hybrid DC subset with mixed-lineage phenotypes. Further, this also causes a loss-of-function effect, reducing Irf8 expression in cDC1s. These developmental alterations critically impair both cDC1-dependent and cDC2-dependent arms of immunity. Collectively, our findings underscore the significance of enhancer suboptimization in the developmental segregation of cDCs required for normal immune function.

3.
Nat Immunol ; 23(11): 1536-1550, 2022 11.
Article in English | MEDLINE | ID: mdl-36271147

ABSTRACT

CD40 signaling in classical type 1 dendritic cells (cDC1s) is required for CD8 T cell-mediated tumor rejection, but the underlying mechanisms are incompletely understood. Here, we identified CD40-induced genes in cDC1s, including Cd70, Tnfsf9, Ptgs2 and Bcl2l1, and examined their contributions to anti-tumor immunity. cDC1-specific inactivation of CD70 and COX-2, and global CD27 inactivation, only partially impaired tumor rejection or tumor-specific CD8 T cell expansion. Loss of 4-1BB, alone or in Cd27-/- mice, did not further impair anti-tumor immunity. However, cDC1-specific CD40 inactivation reduced cDC1 mitochondrial transmembrane potential and increased caspase activation in tumor-draining lymph nodes, reducing migratory cDC1 numbers in vivo. Similar impairments occurred during in vitro antigen presentation by Cd40-/- cDC1s to CD8+ T cells, which were reversed by re-expression of Bcl2l1. Thus, CD40 signaling in cDC1s not only induces costimulatory ligands for CD8+ T cells but also induces Bcl2l1 that sustains cDC1 survival during priming of anti-tumor responses.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , CD40 Antigens/genetics , Antigen Presentation , Dendritic Cells , Mice, Inbred C57BL
4.
Annu Rev Immunol ; 28: 389-411, 2010.
Article in English | MEDLINE | ID: mdl-20307212

ABSTRACT

B and T lymphocyte associated (BTLA) is an Ig domain superfamily protein with cytoplasmic immunoreceptor tyrosine-based inhibitory motifs. Its ligand, herpesvirus entry mediator (HVEM), is a tumor necrosis factor receptor superfamily member. The unique interaction between BTLA and HVEM allows for a system of bidirectional signaling that must be appropriately regulated to balance the outcome of the immune response. HVEM engagement of BTLA produces inhibitory signals through SH2 domain-containing protein tyrosine phosphatase 1 (Shp-1) and Shp-2 association, whereas BTLA engagement of HVEM produces proinflammatory signals via activation of NF-kappaB. The BTLA-HVEM interaction is intriguing and quite complex given that HVEM has four other ligands that also influence immune responses, the conventional TNF ligand LIGHT and lymphotoxin alpha, as well as herpes simplex virus glycoprotein D and the glycosylphosphatidylinositol-linked Ig domain protein CD160. BTLA-HVEM interactions have been shown to regulate responses in several pathogen and autoimmune settings, but our understanding of this complex system of interactions is certainly incomplete. Recent findings of spontaneous inflammation in BTLA-deficient mice may provide an important clue.


Subject(s)
Glycoproteins/immunology , Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor, Member 14/immunology , Animals , Glycoproteins/metabolism , Humans , Immunoglobulins/immunology , Ligands , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Signal Transduction
5.
Nat Immunol ; 20(9): 1174-1185, 2019 09.
Article in English | MEDLINE | ID: mdl-31406377

ABSTRACT

Classical type 1 dendritic cells (cDC1s) are required for antiviral and antitumor immunity, which necessitates an understanding of their development. Development of the cDC1 progenitor requires an E-protein-dependent enhancer located 41 kilobases downstream of the transcription start site of the transcription factor Irf8 (+41-kb Irf8 enhancer), but its maturation instead requires the Batf3-dependent +32-kb Irf8 enhancer. To understand this switch, we performed single-cell RNA sequencing of the common dendritic cell progenitor (CDP) and identified a cluster of cells that expressed transcription factors that influence cDC1 development, such as Nfil3, Id2 and Zeb2. Genetic epistasis among these factors revealed that Nfil3 expression is required for the transition from Zeb2hi and Id2lo CDPs to Zeb2lo and Id2hi CDPs, which represent the earliest committed cDC1 progenitors. This genetic circuit blocks E-protein activity to exclude plasmacytoid dendritic cell potential and explains the switch in Irf8 enhancer usage during cDC1 development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Dendritic Cells/cytology , Enhancer Elements, Genetic/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interferon Regulatory Factors/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/metabolism , Stem Cells/cytology
6.
Nat Immunol ; 20(9): 1161-1173, 2019 09.
Article in English | MEDLINE | ID: mdl-31406378

ABSTRACT

Induction of the transcription factor Irf8 in the common dendritic cell progenitor (CDP) is required for classical type 1 dendritic cell (cDC1) fate specification, but the mechanisms controlling this induction are unclear. In the present study Irf8 enhancers were identified via chromatin profiling of dendritic cells and CRISPR/Cas9 genome editing was used to assess their roles in Irf8 regulation. An enhancer 32 kilobases (kb) downstream of the Irf8 transcriptional start site (+32-kb Irf8) that was active in mature cDC1s was required for the development of this lineage, but not for its specification. Instead, a +41-kb Irf8 enhancer, previously thought to be active only in plasmacytoid dendritic cells, was found to also be transiently accessible in cDC1 progenitors, and deleting this enhancer prevented the induction of Irf8 in CDPs and abolished cDC1 specification. Thus, cryptic activation of the +41-kb Irf8 enhancer in dendritic cell progenitors is responsible for cDC1 fate specification.


Subject(s)
Dendritic Cells/cytology , Enhancer Elements, Genetic/genetics , Interferon Regulatory Factors/metabolism , Macrophages/cytology , Monocytes/cytology , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Lineage , Dendritic Cells/immunology , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Stem Cells/cytology , Tumor Cells, Cultured
7.
Immunity ; 55(6): 1032-1050.e14, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35704993

ABSTRACT

Conventional dendritic cells (cDCs), cDC1 and cDC2, act both to initiate immunity and maintain self-tolerance. The tryptophan metabolic enzyme indoleamine 2,3-dioxygenase 1 (IDO1) is used by cDCs in maintaining tolerance, but its role in different subsets remains unclear. At homeostasis, only mature CCR7+ cDC1 expressed IDO1 that was dependent on IRF8. Lipopolysaccharide treatment induced maturation and IDO1-dependent tolerogenic activity in isolated immature cDC1, but not isolated cDC2. However, both human and mouse cDC2 could induce IDO1 and acquire tolerogenic function when co-cultured with mature cDC1 through the action of cDC1-derived l-kynurenine. Accordingly, cDC1-specific inactivation of IDO1 in vivo exacerbated disease in experimental autoimmune encephalomyelitis. This study identifies a previously unrecognized metabolic communication in which IDO1-expressing cDC1 cells extend their immunoregulatory capacity to the cDC2 subset through their production of tryptophan metabolite l-kynurenine. This metabolic axis represents a potential therapeutic target in treating autoimmune demyelinating diseases.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase , Kynurenine , Animals , Dendritic Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Mice , Signal Transduction , Tryptophan/metabolism
8.
Genes Dev ; 37(7-8): 291-302, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36990511

ABSTRACT

Individual elements within a superenhancer can act in a cooperative or temporal manner, but the underlying mechanisms remain obscure. We recently identified an Irf8 superenhancer, within which different elements act at distinct stages of type 1 classical dendritic cell (cDC1) development. The +41-kb Irf8 enhancer is required for pre-cDC1 specification, while the +32-kb Irf8 enhancer acts to support subsequent cDC1 maturation. Here, we found that compound heterozygous Δ32/Δ41 mice, lacking the +32- and +41-kb enhancers on different chromosomes, show normal pre-cDC1 specification but, surprisingly, completely lack mature cDC1 development, suggesting cis dependence of the +32-kb enhancer on the +41-kb enhancer. Transcription of the +32-kb Irf8 enhancer-associated long noncoding RNA (lncRNA) Gm39266 is also dependent on the +41-kb enhancer. However, cDC1 development in mice remained intact when Gm39266 transcripts were eliminated by CRISPR/Cas9-mediated deletion of lncRNA promoters and when transcription across the +32-kb enhancer was blocked by premature polyadenylation. We showed that chromatin accessibility and BATF3 binding at the +32-kb enhancer were dependent on a functional +41-kb enhancer located in cis Thus, the +41-kb Irf8 enhancer controls the subsequent activation of the +32-kb Irf8 enhancer in a manner that is independent of associated lncRNA transcription.


Subject(s)
RNA, Long Noncoding , Animals , Mice , Enhancer Elements, Genetic , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Promoter Regions, Genetic
9.
Immunity ; 54(7): 1417-1432.e7, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34004142

ABSTRACT

The transcriptional repressor ZEB2 regulates development of many cell fates among somatic, neural, and hematopoietic lineages, but the basis for its requirement in these diverse lineages is unclear. Here, we identified a 400-basepair (bp) region located 165 kilobases (kb) upstream of the Zeb2 transcriptional start site (TSS) that binds the E proteins at several E-box motifs and was active in hematopoietic lineages. Germline deletion of this 400-bp region (Zeb2Δ-165mice) specifically prevented Zeb2 expression in hematopoietic stem cell (HSC)-derived lineages. Zeb2Δ-165 mice lacked development of plasmacytoid dendritic cells (pDCs), monocytes, and B cells. All macrophages in Zeb2Δ-165 mice were exclusively of embryonic origin. Using single-cell chromatin profiling, we identified a second Zeb2 enhancer located at +164-kb that was selectively active in embryonically derived lineages, but not HSC-derived ones. Thus, Zeb2 expression in adult, but not embryonic, hematopoiesis is selectively controlled by the -165-kb Zeb2 enhancer.


Subject(s)
Enhancer Elements, Genetic/genetics , Hematopoiesis/genetics , Transcription, Genetic/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Dendritic Cells/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Monocytes/physiology
10.
Nat Immunol ; 18(5): 563-572, 2017 05.
Article in English | MEDLINE | ID: mdl-28346410

ABSTRACT

Variable strengths of signaling via the T cell antigen receptor (TCR) can produce divergent outcomes, but the mechanism of this remains obscure. The abundance of the transcription factor IRF4 increases with TCR signal strength, but how this would induce distinct types of responses is unclear. We compared the expression of genes in the TH2 subset of helper T cells to enhancer occupancy by the BATF-IRF4 transcription factor complex at varying strengths of TCR stimulation. Genes dependent on BATF-IRF4 clustered into groups with distinct TCR sensitivities. Enhancers exhibited a spectrum of occupancy by the BATF-IRF4 ternary complex that correlated with the sensitivity of gene expression to TCR signal strength. DNA sequences immediately flanking the previously defined AICE motif controlled the affinity of BATF-IRF4 for direct binding to DNA. Analysis by the chromatin immunoprecipitation-exonuclease (ChIP-exo) method allowed the identification of a previously unknown high-affinity AICE2 motif at a human single-nucleotide polymorphism (SNP) of the gene encoding the immunomodulatory receptor CTLA-4 that was associated with resistance to autoimmunity. Thus, the affinity of different enhancers for the BATF-IRF4 complex might underlie divergent signaling outcomes in response to various strengths of TCR signaling.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , CTLA-4 Antigen/genetics , Enhancer Elements, Genetic/genetics , Interferon Regulatory Factors/metabolism , Multiprotein Complexes/metabolism , Receptors, Antigen, T-Cell/metabolism , Th2 Cells/physiology , Animals , Autoimmunity/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Genetic Predisposition to Disease , Humans , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Knockout , Polymorphism, Single Nucleotide , Protein Binding/genetics , Signal Transduction/genetics
11.
Immunity ; 53(4): 759-774.e9, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32795402

ABSTRACT

Development and function of conventional dendritic cell (cDC) subsets, cDC1 and cDC2, depend on transcription factors (TFs) IRF8 and IRF4, respectively. Since IRF8 and IRF4 can each interact with TF BATF3 at AP1-IRF composite elements (AICEs) and with TF PU.1 at Ets-IRF composite elements (EICEs), it is unclear how these factors exert divergent actions. Here, we determined the basis for distinct effects of IRF8 and IRF4 in cDC development. Genes expressed commonly by cDC1 and cDC2 used EICE-dependent enhancers that were redundantly activated by low amounts of either IRF4 or IRF8. By contrast, cDC1-specific genes relied on AICE-dependent enhancers, which required high IRF concentrations, but were activated by either IRF4 or IRF8. IRF8 was specifically required only by a minority of cDC1-specific genes, such as Xcr1, which could distinguish between IRF8 and IRF4 DNA-binding domains. Thus, these results explain how BATF3-dependent Irf8 autoactivation underlies emergence of the cDC1-specific transcriptional program.


Subject(s)
Dendritic Cells/metabolism , Enhancer Elements, Genetic/genetics , Interferon Regulatory Factors/genetics , Animals , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Receptors, Chemokine/genetics , Transcription, Genetic/genetics
12.
Cell ; 156(6): 1223-1234, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24630724

ABSTRACT

Splenic red pulp macrophages (RPM) degrade senescent erythrocytes and recycle heme-associated iron. The transcription factor SPI-C is selectively expressed by RPM and is required for their development, but the physiologic stimulus inducing Spic is unknown. Here, we report that Spic also regulated the development of F4/80(+)VCAM1(+) bone marrow macrophages (BMM) and that Spic expression in BMM and RPM development was induced by heme, a metabolite of erythrocyte degradation. Pathologic hemolysis induced loss of RPM and BMM due to excess heme but induced Spic in monocytes to generate new RPM and BMM. Spic expression in monocytes was constitutively inhibited by the transcriptional repressor BACH1. Heme induced proteasome-dependent BACH1 degradation and rapid Spic derepression. Furthermore, cysteine-proline dipeptide motifs in BACH1 that mediate heme-dependent degradation were necessary for Spic induction by heme. These findings are the first example of metabolite-driven differentiation of a tissue-resident macrophage subset and provide new insights into iron homeostasis.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Heme/metabolism , Iron/metabolism , Monocytes/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , DNA-Binding Proteins/genetics , Female , Macrophages/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Myeloid Cells/metabolism , Spleen/cytology , Spleen/metabolism
13.
Nature ; 607(7917): 142-148, 2022 07.
Article in English | MEDLINE | ID: mdl-35732734

ABSTRACT

The divergence of the common dendritic cell progenitor1-3 (CDP) into the conventional type 1 and type 2 dendritic cell (cDC1 and cDC2, respectively) lineages4,5 is poorly understood. Some transcription factors act in the commitment of already specified progenitors-such as BATF3, which stabilizes Irf8 autoactivation at the +32 kb Irf8 enhancer4,6-but the mechanisms controlling the initial divergence of CDPs remain unknown. Here we report the transcriptional basis of CDP divergence and describe the first requirements for pre-cDC2 specification. Genetic epistasis analysis7 suggested that Nfil3 acts upstream of Id2, Batf3 and Zeb2 in cDC1 development but did not reveal its mechanism or targets. Analysis of newly generated NFIL3 reporter mice showed extremely transient NFIL3 expression during cDC1 specification. CUT&RUN and chromatin immunoprecipitation followed by sequencing identified endogenous NFIL3 binding in the -165 kb Zeb2 enhancer8 at three sites that also bind the CCAAT-enhancer-binding proteins C/EBPα and C/EBPß. In vivo mutational analysis using CRISPR-Cas9 targeting showed that these NFIL3-C/EBP sites are functionally redundant, with C/EBPs supporting and NFIL3 repressing Zeb2 expression at these sites. A triple mutation of all three NFIL3-C/EBP sites ablated Zeb2 expression in myeloid, but not lymphoid progenitors, causing the complete loss of pre-cDC2 specification and mature cDC2 development in vivo. These mice did not generate T helper 2 (TH2) cell responses against Heligmosomoides polygyrus infection, consistent with cDC2 supporting TH2 responses to helminths9-11. Thus, CDP divergence into cDC1 or cDC2 is controlled by competition between NFIL3 and C/EBPs at the -165 kb Zeb2 enhancer.


Subject(s)
Cell Differentiation , Dendritic Cells , Enhancer Elements, Genetic , Mutation , Zinc Finger E-box Binding Homeobox 2 , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/genetics , Dendritic Cells/classification , Dendritic Cells/cytology , Dendritic Cells/pathology , Enhancer Elements, Genetic/genetics , Epistasis, Genetic , Inhibitor of Differentiation Protein 2 , Lymphocytes/cytology , Mice , Myeloid Cells/cytology , Nematospiroides dubius/immunology , Repressor Proteins , Th2 Cells/cytology , Th2 Cells/immunology , Zinc Finger E-box Binding Homeobox 2/genetics
14.
Nat Immunol ; 16(7): 708-17, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26054719

ABSTRACT

The transcription factors Batf3 and IRF8 are required for the development of CD8α(+) conventional dendritic cells (cDCs), but the basis for their actions has remained unclear. Here we identified two progenitor cells positive for the transcription factor Zbtb46 that separately generated CD8α(+) cDCs and CD4(+) cDCs and arose directly from the common DC progenitor (CDP). Irf8 expression in CDPs required prior autoactivation of Irf8 that was dependent on the transcription factor PU.1. Specification of the clonogenic progenitor of CD8α(+) cDCs (the pre-CD8 DC) required IRF8 but not Batf3. However, after specification of pre-CD8 DCs, autoactivation of Irf8 became Batf3 dependent at a CD8α(+) cDC-specific enhancer with multiple transcription factor AP1-IRF composite elements (AICEs) within the Irf8 superenhancer. CDPs from Batf3(-/-) mice that were specified toward development into pre-CD8 DCs failed to complete their development into CD8α(+) cDCs due to decay of Irf8 autoactivation and diverted to the CD4(+) cDC lineage.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Dendritic Cells/immunology , Interferon Regulatory Factors/immunology , Repressor Proteins/immunology , Stem Cells/immunology , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD24 Antigen/immunology , CD24 Antigen/metabolism , CD8 Antigens/immunology , CD8 Antigens/metabolism , Cells, Cultured , Clone Cells/immunology , Clone Cells/metabolism , Dendritic Cells/metabolism , Flow Cytometry , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Protein Binding , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Homology, Nucleic Acid , Stem Cells/metabolism , Transcriptome/genetics , Transcriptome/immunology
15.
Proc Natl Acad Sci U S A ; 121(15): e2315659121, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38564635

ABSTRACT

Monocytes comprise two major subsets, Ly6Chi classical monocytes and Ly6Clo nonclassical monocytes. Notch2 signaling in Ly6Chi monocytes triggers transition to Ly6Clo monocytes, which require Nr4a1, Bcl6, Irf2, and Cebpb. By comparison, less is known about transcriptional requirements for Ly6Chi monocytes. We find transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) is highly expressed in Ly6Chi monocytes, but down-regulated in Ly6Clo monocytes. A few previous studies described the requirement of C/EBPα in the development of neutrophils and eosinophils. However, the role of C/EBPα for in vivo monocyte development has not been understood. We deleted the Cebpa +37 kb enhancer in mice, eliminating hematopoietic expression of C/EBPα, reproducing the expected neutrophil defect. Surprisingly, we also found a severe and selective loss of Ly6Chi monocytes, while preserving Ly6Clo monocytes. We find that BM progenitors from Cebpa +37-/- mice rapidly progress through the monocyte progenitor stage to develop directly into Ly6Clo monocytes even in the absence of Notch2 signaling. These results identify a previously unrecognized role for C/EBPα in maintaining Ly6Chi monocyte identity.


Subject(s)
Gene Expression Regulation , Monocytes , Animals , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Transcription Factors/metabolism
16.
Nature ; 584(7822): 624-629, 2020 08.
Article in English | MEDLINE | ID: mdl-32788723

ABSTRACT

Conventional type 1 dendritic cells (cDC1)1 are thought to perform antigen cross-presentation, which is required to prime CD8+ T cells2,3, whereas cDC2 are specialized for priming CD4+ T cells4,5. CD4+ T cells are also considered to help CD8+ T cell responses through a variety of mechanisms6-11, including a process whereby CD4+ T cells 'license' cDC1 for CD8+ T cell priming12. However, this model has not been directly tested in vivo or in the setting of help-dependent tumour rejection. Here we generated an Xcr1Cre mouse strain to evaluate the cellular interactions that mediate tumour rejection in a model requiring CD4+ and CD8+ T cells. As expected, tumour rejection required cDC1 and CD8+ T cell priming required the expression of major histocompatibility class I molecules by cDC1. Unexpectedly, early priming of CD4+ T cells against tumour-derived antigens also required cDC1, and this was not simply because they transport antigens to lymph nodes for processing by cDC2, as selective deletion of major histocompatibility class II molecules in cDC1 also prevented early CD4+ T cell priming. Furthermore, deletion of either major histocompatibility class II or CD40 in cDC1 impaired tumour rejection, consistent with a role for cognate CD4+ T cell interactions and CD40 signalling in cDC1 licensing. Finally, CD40 signalling in cDC1 was critical not only for CD8+ T cell priming, but also for initial CD4+ T cell activation. Thus, in the setting of tumour-derived antigens, cDC1 function as an autonomous platform capable of antigen processing and priming for both CD4+ and CD8+ T cells and of the direct orchestration of their cross-talk that is required for optimal anti-tumour immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cross-Priming , Dendritic Cells/immunology , Neoplasms/immunology , Animals , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/cytology , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Histocompatibility Antigens Class II/immunology , Mice , Signal Transduction
17.
Proc Natl Acad Sci U S A ; 120(35): e2220853120, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37607223

ABSTRACT

Ly6Clo monocytes are a myeloid subset that specializes in the surveillance of vascular endothelium. Ly6Clo monocytes have been shown to derive from Ly6Chi monocytes. NOTCH2 signaling has been implicated as a trigger for Ly6Clo monocyte development, but the basis for this effect is unclear. Here, we examined the impact of NOTCH2 signaling of myeloid progenitors on the development of Ly6Clo monocytes in vitro. NOTCH2 signaling induced by delta-like ligand 1 (DLL1) efficiently induced the transition of Ly6Chi TREML4- monocytes into Ly6Clo TREML4+ monocytes. We further identified two additional transcriptional requirements for development of Ly6Clo monocytes. Deletion of BCL6 from myeloid progenitors abrogated development of Ly6Clo monocytes. IRF2 was also required for Ly6Clo monocyte development in a cell-intrinsic manner. DLL1-induced in vitro transition into Ly6Clo TREML4+ monocytes required IRF2 but unexpectedly could occur in the absence of NUR77 or BCL6. These results imply a transcriptional hierarchy for these factors in controlling Ly6Clo monocyte development.


Subject(s)
Endothelium, Vascular , Monocytes , Hematopoiesis , Signal Transduction
18.
Proc Natl Acad Sci U S A ; 120(13): e2219956120, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36940342

ABSTRACT

The events that initiate autoimmune diabetes in nonobese diabetic (NOD) mice remain poorly understood. CD4+ and CD8+ T cells are both required to develop disease, but their relative roles in initiating disease are unclear. To test whether CD4+ T cell infiltration into islets requires damage to ß cells induced by autoreactive CD8+ T cells, we inactivated Wdfy4 in nonobese diabetic (NOD) mice (NOD.Wdfy4-/--) using CRISPR/Cas9 targeting to eliminate cross-presentation by type 1 conventional dendritic cells (cDC1s). Similar to C57BL/6 Wdfy4-/- mice, cDC1 in NOD.Wdfy4-/- mice are unable to cross-present cell-associated antigens to prime CD8+ T cells, while cDC1 from heterozygous NOD.Wdfy4+/- mice cross-present normally. Further, NOD.Wdfy4-/- mice fail to develop diabetes while heterozygous NOD.Wdfy4+/- mice develop diabetes similarly to wild-type NOD mice. NOD.Wdfy4-/- mice remain capable of processing and presenting major histocompatibility complex class II (MHC-II)-restricted autoantigens and can activate ß cell-specific CD4+ T cells in lymph nodes. However, disease in these mice does not progress beyond peri-islet inflammation. These results indicate that the priming of autoreactive CD8+ T cells in NOD mice requires cross-presentation by cDC1. Further, autoreactive CD8+ T cells appear to be required not only to develop diabetes, but to recruit autoreactive CD4+ T cells into islets of NOD mice, perhaps in response to progressive ß cell damage.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Mice , Animals , Mice, Inbred NOD , CD8-Positive T-Lymphocytes , Mice, Inbred C57BL , Histocompatibility Antigens Class II
19.
Nat Immunol ; 14(9): 937-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913046

ABSTRACT

Defense against attaching-and-effacing bacteria requires the sequential generation of interleukin 23 (IL-23) and IL-22 to induce protective mucosal responses. Although CD4(+) and NKp46(+) innate lymphoid cells (ILCs) are the critical source of IL-22 during infection, the precise source of IL-23 is unclear. We used genetic techniques to deplete mice of specific subsets of classical dendritic cells (cDCs) and analyzed immunity to the attaching-and-effacing pathogen Citrobacter rodentium. We found that the signaling receptor Notch2 controlled the terminal stage of cDC differentiation. Notch2-dependent intestinal CD11b(+) cDCs were an obligate source of IL-23 required for survival after infection with C. rodentium, but CD103(+) cDCs dependent on the transcription factor Batf3 were not. Our results demonstrate a nonredundant function for CD11b(+) cDCs in the response to pathogens in vivo.


Subject(s)
Citrobacter rodentium/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Receptor, Notch2/metabolism , Animals , Antigens, CD/metabolism , CD11b Antigen/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells/cytology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/mortality , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Interleukin-23/metabolism , Intestinal Mucosa/microbiology , Lectins, C-Type/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Mice , Mice, Transgenic , Minor Histocompatibility Antigens , Receptor, Notch2/deficiency , Receptors, Cell Surface/metabolism , Signal Transduction , Spleen/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Wound Healing/genetics , Wound Healing/immunology
SELECTION OF CITATIONS
SEARCH DETAIL