Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 31(3): 265-279, 2024 03.
Article in English | MEDLINE | ID: mdl-38383888

ABSTRACT

PirB is an inhibitory cell surface receptor particularly prominent on myeloid cells. PirB curtails the phenotypes of activated macrophages during inflammation or tumorigenesis, but its functions in macrophage homeostasis are obscure. To elucidate PirB-related functions in macrophages at steady-state, we generated and compared single-cell RNA-sequencing (scRNAseq) datasets obtained from myeloid cell subsets of wild type (WT) and PirB-deficient knockout (PirB KO) mice. To facilitate this analysis, we developed a novel approach to clustering parameter optimization called "Cluster Similarity Scoring and Distinction Index" (CaSSiDI). We demonstrate that CaSSiDI is an adaptable computational framework that facilitates tandem analysis of two scRNAseq datasets by optimizing clustering parameters. We further show that CaSSiDI offers more advantages than a standard Seurat analysis because it allows direct comparison of two or more independently clustered datasets, thereby alleviating the need for batch-correction while identifying the most similar and different clusters. Using CaSSiDI, we found that PirB is a novel regulator of Cebpb expression that controls the generation of Ly6Clo patrolling monocytes and the expansion properties of peritoneal macrophages. PirB's effect on Cebpb is tissue-specific since it was not observed in splenic red pulp macrophages (RPMs). However, CaSSiDI revealed a segregation of the WT RPM population into a CD68loIrf8+ "neuronal-primed" subset and an CD68hiFtl1+ "iron-loaded" subset. Our results establish the utility of CaSSiDI for single-cell assay analyses and the determination of optimal clustering parameters. Our application of CaSSiDI in this study has revealed previously unknown roles for PirB in myeloid cell populations. In particular, we have discovered homeostatic functions for PirB that are related to Cebpb expression in distinct macrophage subsets.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Macrophages , Receptors, Immunologic , Single-Cell Analysis , Animals , Mice , Macrophages/metabolism , Monocytes/metabolism , Myeloid Cells/metabolism , Receptors, Cell Surface , Receptors, Immunologic/metabolism , Single-Cell Analysis/methods , CCAAT-Enhancer-Binding Protein-beta/metabolism
2.
Nat Cancer ; 4(10): 1437-1454, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37640929

ABSTRACT

Cholinergic nerves are involved in tumor progression and dissemination. In contrast to other visceral tissues, cholinergic innervation in the hepatic parenchyma is poorly detected. It remains unclear whether there is any form of cholinergic regulation of liver cancer. Here, we show that cholinergic T cells curtail the development of liver cancer by supporting antitumor immune responses. In a mouse multihit model of hepatocellular carcinoma (HCC), we observed activation of the adaptive immune response and induction of two populations of CD4+ T cells expressing choline acetyltransferase (ChAT), including regulatory T cells and dysfunctional PD-1+ T cells. Tumor antigens drove the clonal expansion of these cholinergic T cells in HCC. Genetic ablation of Chat in T cells led to an increased prevalence of preneoplastic cells and exacerbated liver cancer due to compromised antitumor immunity. Mechanistically, the cholinergic activity intrinsic in T cells constrained Ca2+-NFAT signaling induced by T cell antigen receptor engagement. Without this cholinergic modulation, hyperactivated CD25+ T regulatory cells and dysregulated PD-1+ T cells impaired HCC immunosurveillance. Our results unveil a previously unappreciated role for cholinergic T cells in liver cancer immunobiology.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Monitoring, Immunologic , T-Lymphocytes, Regulatory/pathology
3.
Cell Death Differ ; 30(2): 407-416, 2023 02.
Article in English | MEDLINE | ID: mdl-36528755

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a mouse model of multiple sclerosis (MS) in which Th17 cells have a crucial but unclear function. Here we show that choline acetyltransferase (ChAT), which synthesizes acetylcholine (ACh), is a critical driver of pathogenicity in EAE. Mice with ChAT-deficient Th17 cells resist disease progression and show reduced brain-infiltrating immune cells. ChAT expression in Th17 cells is linked to strong TCR signaling, expression of the transcription factor Bhlhe40, and increased Il2, Il17, Il22, and Il23r mRNA levels. ChAT expression in Th17 cells is independent of IL21r signaling but dampened by TGFß, implicating ChAT in controlling the dichotomous nature of Th17 cells. Our study establishes a cholinergic program in which ACh signaling primes chronic activation of Th17 cells, and thereby constitutes a pathogenic determinant of EAE. Our work may point to novel targets for therapeutic immunomodulation in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Th17 Cells , Virulence , Cholinergic Agents , Multiple Sclerosis/genetics , Acetylcholine/metabolism , Mice, Inbred C57BL , Cell Differentiation
4.
Sci Immunol ; 7(67): eabf7777, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34995099

ABSTRACT

Resident macrophages orchestrate homeostatic, inflammatory, and reparative activities. It is appreciated that different tissues instruct specialized macrophage functions. However, individual tissues contain heterogeneous subpopulations, and how these subpopulations are related is unclear. We asked whether common transcriptional and functional elements could reveal an underlying framework across tissues. Using single-cell RNA sequencing and random forest modeling, we observed that four genes could predict three macrophage subsets that were present in murine heart, liver, lung, kidney, and brain. Parabiotic and genetic fate mapping studies revealed that these core markers predicted three unique life cycles across 17 tissues. TLF+ (expressing TIMD4 and/or LYVE1 and/or FOLR2) macrophages were maintained through self-renewal with minimal monocyte input; CCR2+ (TIMD4−LYVE1−FOLR2−) macrophages were almost entirely replaced by monocytes, and MHC-IIhi macrophages (TIMD4−LYVE1−FOLR2−CCR2−), while receiving modest monocyte contribution, were not continually replaced. Rather, monocyte-derived macrophages contributed to the resident macrophage population until they reached a defined upper limit after which they did not outcompete pre-existing resident macrophages. Developmentally, TLF+ macrophages were first to emerge in the yolk sac and early fetal organs. Fate mapping studies in the mouse and human single-cell RNA sequencing indicated that TLF+ macrophages originated from both yolk sac and fetal monocyte precursors. Furthermore, TLF+ macrophages were the most transcriptionally conserved subset across mouse tissues and between mice and humans, despite organ- and species-specific transcriptional differences. Here, we define the existence of three murine macrophage subpopulations based on common life cycle properties and core gene signatures and provide a common starting point to understand tissue macrophage heterogeneity.


Subject(s)
Folate Receptor 2/immunology , Histocompatibility Antigens Class II/immunology , Macrophages/immunology , Membrane Proteins/immunology , Receptors, CCR2/immunology , Vesicular Transport Proteins/immunology , Animals , Life Cycle Stages/immunology , Macrophage Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, CCR2/deficiency
5.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Article in English | MEDLINE | ID: mdl-34210797

ABSTRACT

While modulatory effects of gut microbes on neurological phenotypes have been reported, the mechanisms remain largely unknown. Here, we demonstrate that indole, a tryptophan metabolite produced by tryptophanase-expressing gut microbes, elicits neurogenic effects in the adult mouse hippocampus. Neurogenesis is reduced in germ-free (GF) mice and in GF mice monocolonized with a single-gene tnaA knockout (KO) mutant Escherichia coli unable to produce indole. External administration of systemic indole increases adult neurogenesis in the dentate gyrus in these mouse models and in specific pathogen-free (SPF) control mice. Indole-treated mice display elevated synaptic markers postsynaptic density protein 95 and synaptophysin, suggesting synaptic maturation effects in vivo. By contrast, neurogenesis is not induced by indole in aryl hydrocarbon receptor KO (AhR-/-) mice or in ex vivo neurospheres derived from them. Neural progenitor cells exposed to indole exit the cell cycle, terminally differentiate, and mature into neurons that display longer and more branched neurites. These effects are not observed with kynurenine, another AhR ligand. The indole-AhR-mediated signaling pathway elevated the expression of ß-catenin, Neurog2, and VEGF-α genes, thus identifying a molecular pathway connecting gut microbiota composition and their metabolic function to neurogenesis in the adult hippocampus. Our data have implications for the understanding of mechanisms of brain aging and for potential next-generation therapeutic opportunities.


Subject(s)
Aging/metabolism , Gastrointestinal Microbiome , Neurogenesis , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/metabolism , Animals , Indoles/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neural Stem Cells/metabolism
7.
Nat Commun ; 10(1): 2678, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31213601

ABSTRACT

Myeloid cells contribute to tumor progression, but how the constellation of receptors they express regulates their functions within the tumor microenvironment (TME) is unclear. We demonstrate that Fcmr (Toso), the putative receptor for soluble IgM, modulates myeloid cell responses to cancer. In a syngeneic melanoma model, Fcmr ablation in myeloid cells suppressed tumor growth and extended mouse survival. Fcmr deficiency increased myeloid cell population density in this malignancy and enhanced anti-tumor immunity. Single-cell RNA sequencing of Fcmr-deficient tumor-associated mononuclear phagocytes revealed a unique subset with enhanced antigen processing/presenting properties. Conversely, Fcmr activity negatively regulated the activation and migratory capacity of myeloid cells in vivo, and T cell activation by bone marrow-derived dendritic cells in vitro. Therapeutic targeting of Fcmr during oncogenesis decreased tumor growth when used as a single agent or in combination with anti-PD-1. Thus, Fcmr regulates myeloid cell activation within the TME and may be a potential therapeutic target.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carrier Proteins/metabolism , Melanoma, Experimental/immunology , Membrane Proteins/metabolism , Monocytes/immunology , Skin Neoplasms/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line, Tumor/transplantation , Cell Movement/drug effects , Cell Movement/immunology , Female , Lymphocyte Activation/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/mortality , Melanoma, Experimental/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Analysis , T-Lymphocytes/immunology , Tumor Microenvironment/immunology
8.
Curr Opin Immunol ; 48: 61-67, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28865357

ABSTRACT

The recent successes of immune check point targeting therapies in treating cancer patients has driven a resurgence of interest in targeting these pathways in chronically infected patients. While still in early stages, basic and clinical data suggest that blockade of CTLA-4 and PD-1 can be beneficial in the treatment of chronic HIV, HBV, and HCV infection, as well as other chronic maladies. Furthermore, novel inhibitory receptors such as Tim-3, LAG-3, and TIGIT are the potential next wave of check points that can be manipulated for the treatment of chronic infection. Blockade of these pathways influences more than simply T cell responses, and may provide new therapeutic options for chronically infected patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CTLA-4 Antigen/immunology , Costimulatory and Inhibitory T-Cell Receptors/immunology , HIV-1/immunology , Hepacivirus/immunology , Hepatitis B virus/immunology , Immunotherapy/methods , Programmed Cell Death 1 Receptor/immunology , Virus Diseases/therapy , Animals , Antigens, CD/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Molecular Targeted Therapy , Receptors, Immunologic/metabolism , Virus Diseases/immunology , Lymphocyte Activation Gene 3 Protein
9.
Cell Stem Cell ; 19(2): 205-216, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27184401

ABSTRACT

The E3 ubiquitin ligase Mule is often overexpressed in human colorectal cancers, but its role in gut tumorigenesis is unknown. Here, we show in vivo that Mule controls murine intestinal stem and progenitor cell proliferation by modulating Wnt signaling via c-Myc. Mule also regulates protein levels of the receptor tyrosine kinase EphB3 by targeting it for proteasomal and lysosomal degradation. In the intestine, EphB/ephrinB interactions position cells along the crypt-villus axis and compartmentalize incipient colorectal tumors. Our study thus unveils an important new avenue by which Mule acts as an intestinal tumor suppressor by regulation of the intestinal stem cell niche.


Subject(s)
Ephrin-B3/metabolism , Intestines/cytology , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Stem Cell Niche , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway , Adenomatous Polyposis Coli/pathology , Alleles , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Endocytosis , HEK293 Cells , Humans , Mice, Knockout , Models, Biological , Mutation/genetics , Paneth Cells/pathology , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/deficiency
10.
Immunity ; 44(3): 527-541, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26982363

ABSTRACT

Lymphopoiesis requires the activation of lineage-specific genes embedded in naive, inaccessible chromatin or in primed, accessible chromatin. The mechanisms responsible for de novo gain of chromatin accessibility, known as "pioneer" function, remain poorly defined. Here, we showed that the EBF1 C-terminal domain (CTD) is required for the regulation of a specific gene set involved in B cell fate decision and differentiation, independently of activation and repression functions. Using genome-wide analysis of DNaseI hypersensitivity and DNA methylation in multipotent Ebf1(-/-) progenitors and derivative EBF1wt- or EBF1ΔC-expressing cells, we found that the CTD promoted chromatin accessibility and DNA demethylation in previously naive chromatin. The CTD allowed EBF1 to bind at inaccessible genomic regions that offer limited co-occupancy by other transcription factors, whereas the CTD was dispensable for EBF1 binding at regions that are occupied by multiple transcription factors. Thus, the CTD enables EBF1 to confer permissive lineage-specific changes in progenitor chromatin landscape.


Subject(s)
B-Lymphocytes/physiology , Chromatin/metabolism , Lymphoid Progenitor Cells/physiology , Trans-Activators/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , DNA Methylation/genetics , Gene Regulatory Networks/genetics , Lymphopoiesis , Mice , Mice, Knockout , Mice, Transgenic , Protein Structure, Tertiary/genetics , Trans-Activators/genetics
11.
Immunity ; 43(1): 9-11, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26200008

ABSTRACT

Passenger mutations specific to particular mouse strains can distort experimental outcomes. In this issue of Immunity, Vanden Berghe et al. (2015) demonstrate that passenger mutations are frequent in most genetically engineered congenic mice and persist even after extensive backcrossing.


Subject(s)
Genetic Variation/genetics , Genome/genetics , Mice, Inbred C57BL/genetics , Animals
12.
Nat Immunol ; 14(8): 867-75, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23812095

ABSTRACT

The transcription factors EBF1 and Pax5 have been linked to activation of the B cell lineage program and irreversible loss of alternative lineage potential (commitment), respectively. Here we conditionally deleted Ebf1 in committed pro-B cells after transfer into alymphoid mice. We found that those cells converted into innate lymphoid cells (ILCs) and T cells with variable-diversity-joining (VDJ) rearrangements of loci encoding both B cell and T cell antigen receptors. As intermediates in lineage conversion, Ebf1-deficient CD19(+) cells expressing Pax5 and transcriptional regulators of the ILC and T cell fates were detectable. In particular, genes encoding the transcription factors Id2 and TCF-1 were bound and repressed by EBF1. Thus, both EBF1 and Pax5 are required for B lineage commitment by repressing distinct and common determinants of alternative cell fates.


Subject(s)
B-Lymphocytes/immunology , Trans-Activators/immunology , Adoptive Transfer , Animals , B-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage , DNA/chemistry , DNA/genetics , Gene Expression Regulation , Lymphopoiesis/immunology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Trans-Activators/genetics , V(D)J Recombination/genetics , V(D)J Recombination/immunology
13.
Dev Cell ; 23(4): 866-71, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23079603

ABSTRACT

Satb1 and Satb2 have been recently described as regulators of embryonic stem (ES) cell pluripotency and as silencing factors in X chromosome inactivation. The influence of the pluripotency machinery on X chromosome inactivation and the lack of an X chromosome inactivation defect in Satb1(-/-) and Satb2(-/-) mice raise the question of whether or not Satb proteins are directly and/or redundantly involved in this process. Here, we analyzed X chromosome inactivation in fibroblastic cells that were derived from female Satb1(-/-)Satb2(-/-) embryos. By fluorescence in situ hybridization to visualize Xist RNA and by immunohistochemistry to detect H3K27me3 histone modifications, we found that female Satb1(-/-)Satb2(-/-) fibroblastic cells contain proper Barr bodies. Moreover, we did not detect an upregulation of X-linked genes, suggesting that Satb proteins are dispensable for X chromosome inactivation in mice.


Subject(s)
Matrix Attachment Region Binding Proteins/deficiency , Transcription Factors/deficiency , X Chromosome Inactivation , Animals , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Mice , Transcription Factors/metabolism , X Chromosome Inactivation/genetics
14.
Genes Dev ; 26(7): 668-82, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22431510

ABSTRACT

The transcription factor Ebf1 is an important determinant of early B lymphopoiesis. To gain insight into the functions of Ebf1 at distinct stages of differentiation, we conditionally inactivated Ebf1. We found that Ebf1 is required for the proliferation, survival, and signaling of pro-B cells and peripheral B-cell subsets, including B1 cells and marginal zone B cells. The proliferation defect of Ebf1-deficient pro-B cells and the impaired expression of multiple cell cycle regulators are overcome by transformation with v-Abl. The survival defect of transformed Ebf1(fl/fl) pro-B cells can be rescued by the forced expression of the Ebf1 targets c-Myb or Bcl-x(L). In mature B cells, Ebf1 deficiency interferes with signaling via the B-cell-activating factor receptor (BAFF-R)- and B-cell receptor (BCR)-dependent Akt pathways. Moreover, Ebf1 is required for germinal center formation and class switch recombination. Genome-wide analyses of Ebf1-mediated gene expression and chromatin binding indicate that Ebf1 regulates both common and distinct sets of genes in early and late stage B cells. By regulating important components of transcription factor and signaling networks, Ebf1 appears to be involved in the coordination of cell proliferation, survival, and differentiation at multiple stages of B lymphopoiesis.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation , Cell Proliferation , Signal Transduction , Trans-Activators/metabolism , Animals , B-Lymphocytes/immunology , Cell Survival , Gene Expression Regulation , Genome-Wide Association Study , Immunoglobulin Heavy Chains/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Trans-Activators/genetics , Trans-Activators/immunology , Transcription, Genetic
15.
Genes Dev ; 23(22): 2625-38, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19933152

ABSTRACT

Satb1 and the closely related Satb2 proteins regulate gene expression and higher-order chromatin structure of multigene clusters in vivo. In examining the role of Satb proteins in murine embryonic stem (ES) cells, we find that Satb1(-/-) cells display an impaired differentiation potential and augmented expression of the pluripotency determinants Nanog, Klf4, and Tbx3. Metastable states of self-renewal and differentiation competence have been attributed to heterogeneity of ES cells in the expression of Nanog. Satb1(-/-) cultures have a higher proportion of Nanog(high) cells, and an increased potential to reprogram human B lymphocytes in cell fusion experiments. Moreover, Satb1-deficient ES cells show an increased expression of Satb2, and we find that forced Satb2 expression in wild-type ES cells antagonizes differentiation-associated silencing of Nanog and enhances the induction of NANOG in cell fusions with human B lymphocytes. An antagonistic function of Satb1 and Satb2 is also supported by the almost normal differentiation potential of Satb1(-/-)Satb2(-/-) ES cells. Taken together with the finding that both Satb1 and Satb2 bind the Nanog locus in vivo, our data suggest that the balance of Satb1 and Satb2 contributes to the plasticity of Nanog expression and ES cell pluripotency.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line , Humans , Kruppel-Like Factor 4 , Mice , Nanog Homeobox Protein
16.
J Immunol ; 181(8): 5213-7, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832674

ABSTRACT

T cell development, originally thought to be completed in the thymus, has recently been shown to continue for several weeks in the lymphoid periphery. The forces that drive this peripheral maturation are unclear. The use of mice transgenic for GFP driven by the RAG2 promoter has enabled the ready identification and analysis of recent thymic emigrants. Here, we show that recent thymic emigrant maturation is a progressive process and is promoted by T cell exit from the thymus. Further, we show that this maturation occurs within secondary lymphoid organs and does not require extensive lymphocyte recirculation.


Subject(s)
Cell Movement/immunology , Thymus Gland/immunology , Animals , Cell Movement/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Thymus Gland/cytology , Transgenes/genetics , Transgenes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...