Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Nat Metab ; 6(4): 697-707, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38413806

ABSTRACT

Post-translational modifications (PTMs) on histones are a key source of regulation on chromatin through impacting cellular processes, including gene expression1. These PTMs often arise from metabolites and are thus impacted by metabolism and environmental cues2-7. One class of metabolically regulated PTMs are histone acylations, which include histone acetylation, butyrylation, crotonylation and propionylation3,8. As these PTMs can be derived from short-chain fatty acids, which are generated by the commensal microbiota in the intestinal lumen9-11, we aimed to define how microbes impact the host intestinal chromatin landscape, mainly in female mice. Here we show that in addition to acetylation, intestinal epithelial cells from the caecum and distal mouse intestine also harbour high levels of butyrylation and propionylation on lysines 9 and 27 of histone H3. We demonstrate that these acylations are regulated by the microbiota and that histone butyrylation is additionally regulated by the metabolite tributyrin. Tributyrin-regulated gene programmes are correlated with histone butyrylation, which is associated with active gene-regulatory elements and levels of gene expression. Together, our study uncovers a regulatory layer of how the microbiota and metabolites influence the intestinal epithelium through chromatin, demonstrating a physiological setting in which histone acylations are dynamically regulated and associated with gene regulation.


Subject(s)
Gastrointestinal Microbiome , Gene Expression Regulation , Histones , Protein Processing, Post-Translational , Animals , Histones/metabolism , Mice , Female , Intestinal Mucosa/metabolism , Acetylation , Intestines/microbiology , Triglycerides/metabolism , Mice, Inbred C57BL
2.
Cell Rep ; 41(11): 111809, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36516747

ABSTRACT

The gut microbiota influences acetylation on host histones by fermenting dietary fiber into butyrate. Although butyrate could promote histone acetylation by inhibiting histone deacetylases, it may also undergo oxidation to acetyl-coenzyme A (CoA), a necessary cofactor for histone acetyltransferases. Here, we find that epithelial cells from germ-free mice harbor a loss of histone H4 acetylation across the genome except at promoter regions. Using stable isotope tracing in vivo with 13C-labeled fiber, we demonstrate that the microbiota supplies carbon for histone acetylation. Subsequent metabolomic profiling revealed hundreds of labeled molecules and supported a microbial contribution to host fatty acid metabolism, which declined in response to colitis and correlated with reduced expression of genes involved in fatty acid oxidation. These results illuminate the flow of carbon from the diet to the host via the microbiota, disruptions to which may affect energy homeostasis in the distal gut and contribute to the development of colitis.


Subject(s)
Colitis , Microbiota , Mice , Animals , Acetylation , Histones/metabolism , Histone Acetyltransferases/metabolism , Isotopes/metabolism , Carbon/metabolism , Butyrates , Fatty Acids
3.
Nat Commun ; 9(1): 477, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29396460

ABSTRACT

Major depressive disorder is associated with abnormalities in the brain and the immune system. Chronic stress in animals showed that epigenetic and inflammatory mechanisms play important roles in mediating resilience and susceptibility to depression. Here, through a high-throughput screening, we identify two phytochemicals, dihydrocaffeic acid (DHCA) and malvidin-3'-O-glucoside (Mal-gluc) that are effective in promoting resilience against stress by modulating brain synaptic plasticity and peripheral inflammation. DHCA/Mal-gluc also significantly reduces depression-like phenotypes in a mouse model of increased systemic inflammation induced by transplantation of hematopoietic progenitor cells from stress-susceptible mice. DHCA reduces pro-inflammatory interleukin 6 (IL-6) generations by inhibiting DNA methylation at the CpG-rich IL-6 sequences introns 1 and 3, while Mal-gluc modulates synaptic plasticity by increasing histone acetylation of the regulatory sequences of the Rac1 gene. Peripheral inflammation and synaptic maladaptation are in line with newly hypothesized clinical intervention targets for depression that are not addressed by currently available antidepressants.


Subject(s)
Anthocyanins/pharmacology , Caffeic Acids/pharmacology , Epigenesis, Genetic , Glucosides/pharmacology , Inflammation/genetics , Neuronal Plasticity/genetics , Stress, Psychological/genetics , Animals , Anthocyanins/administration & dosage , Caffeic Acids/administration & dosage , CpG Islands/drug effects , Depression/drug therapy , Drug Evaluation, Preclinical/methods , Glucosides/administration & dosage , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Leukocyte Common Antigens/genetics , Male , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Neuropeptides/genetics , Neuropeptides/metabolism , Polyphenols/pharmacology , Social Behavior , Stress, Psychological/drug therapy , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
4.
J Exp Med ; 215(1): 319-336, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29263218

ABSTRACT

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia characterized by granulomatous lesions containing pathological CD207+ dendritic cells (DCs) with constitutively activated mitogen-activated protein kinase (MAPK) pathway signaling. Approximately 60% of LCH patients harbor somatic BRAFV600E mutations localizing to CD207+ DCs within lesions. However, the mechanisms driving BRAFV600E+ LCH cell accumulation in lesions remain unknown. Here we show that sustained extracellular signal-related kinase activity induced by BRAFV600E inhibits C-C motif chemokine receptor 7 (CCR7)-mediated DC migration, trapping DCs in tissue lesions. Additionally, BRAFV600E increases expression of BCL2-like protein 1 (BCL2L1) in DCs, resulting in resistance to apoptosis. Pharmacological MAPK inhibition restores migration and apoptosis potential in a mouse LCH model, as well as in primary human LCH cells. We also demonstrate that MEK inhibitor-loaded nanoparticles have the capacity to concentrate drug delivery to phagocytic cells, significantly reducing off-target toxicity. Collectively, our results indicate that MAPK tightly suppresses DC migration and augments DC survival, rendering DCs in LCH lesions trapped and resistant to cell death.


Subject(s)
Cell Movement/physiology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Histiocytosis, Langerhans-Cell/metabolism , Langerhans Cells/metabolism , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins B-raf/metabolism , Animals , Apoptosis/physiology , Histiocytosis, Langerhans-Cell/pathology , Humans , Langerhans Cells/physiology , Mice , Mice, Inbred C57BL , Mutation/physiology , Phagocytosis/physiology
5.
Immunity ; 44(4): 924-38, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27096321

ABSTRACT

Large numbers of melanoma lesions develop resistance to targeted inhibition of mutant BRAF or fail to respond to checkpoint blockade. We explored whether modulation of intratumoral antigen-presenting cells (APCs) could increase responses to these therapies. Using mouse melanoma models, we found that CD103(+) dendritic cells (DCs) were the only APCs transporting intact antigens to the lymph nodes and priming tumor-specific CD8(+) T cells. CD103(+) DCs were required to promote anti-tumoral effects upon blockade of the checkpoint ligand PD-L1; however, PD-L1 inhibition only led to partial responses. Systemic administration of the growth factor FLT3L followed by intratumoral poly I:C injections expanded and activated CD103(+) DC progenitors in the tumor, enhancing responses to BRAF and PD-L1 blockade and protecting mice from tumor rechallenge. Thus, the paucity of activated CD103(+) DCs in tumors limits checkpoint-blockade efficacy and combined FLT3L and poly I:C therapy can enhance tumor responses to checkpoint and BRAF blockade.


Subject(s)
Antigens, CD/metabolism , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Integrin alpha Chains/metabolism , Melanoma, Experimental/immunology , Poly I-C/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/pharmacology , Animals , Antigen Presentation/immunology , Cell Line, Tumor , Dendritic Cells/cytology , Mice, Inbred C57BL , Mice, Knockout
6.
Nat Immunol ; 16(10): 1060-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26343536

ABSTRACT

Treatment with ionizing radiation (IR) can lead to the accumulation of tumor-infiltrating regulatory T cells (Treg cells) and subsequent resistance of tumors to radiotherapy. Here we focused on the contribution of the epidermal mononuclear phagocytes Langerhans cells (LCs) to this phenomenon because of their ability to resist depletion by high-dose IR. We found that LCs resisted apoptosis and rapidly repaired DNA damage after exposure to IR. In particular, we found that the cyclin-dependent kinase inhibitor CDKN1A (p21) was overexpressed in LCs and that Cdkn1a(-/-) LCs underwent apoptosis and accumulated DNA damage following IR treatment. Wild-type LCs upregulated major histocompatibility complex class II molecules, migrated to the draining lymph nodes and induced an increase in Treg cell numbers upon exposure to IR, but Cdkn1a(-/-) LCs did not. Our findings suggest a means for manipulating the resistance of LCs to IR to enhance the response of cutaneous tumors to radiotherapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Langerhans Cells/radiation effects , Radiation, Ionizing , T-Lymphocytes, Regulatory/radiation effects , Animals , Cell Survival/genetics , Cell Survival/radiation effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Flow Cytometry , Mice , Microarray Analysis , Polymerase Chain Reaction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Up-Regulation
8.
Cancer Immunol Res ; 3(3): 296-304, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25600438

ABSTRACT

We sought to define cellular immune mechanisms of synergy between tumor-antigen-targeted monoclonal antibodies and chemotherapy. Established B16 melanoma in mice was treated with cytotoxic doses of cyclophosphamide in combination with an antibody targeting tyrosinase-related protein 1 (αTRP1), a native melanoma differentiation antigen. We find that Fcγ receptors are required for efficacy, showing that antitumor activity of combination therapy is immune mediated. Rag1(-/-) mice deficient in adaptive immunity are able to clear tumors, and thus innate immunity is sufficient for efficacy. Furthermore, previously treated wild-type mice are not significantly protected against tumor reinduction, as compared with mice inoculated with irradiated B16 alone, consistent with a primarily innate immune mechanism of action of chemo-immunotherapy. In contrast, mice deficient in both classical natural killer (NK) lymphocytes and nonclassical innate lymphocytes (ILC) due to deletion of the IL2 receptor common gamma chain IL2γc(-/-)) are refractory to chemo-immunotherapy. Classical NK lymphocytes are not critical for treatment, as depletion of NK1.1⁺ cells does not impair antitumor effect. Depletion of CD90⁺NK1.1⁻ lymphocytes, however, both diminishes therapeutic benefit and decreases accumulation of macrophages within the tumor. Tumor clearance during combination chemo-immunotherapy with monoclonal antibodies against native antigen is mediated by the innate immune system. We highlight a novel potential role for CD90⁺NK1.1⁻ ILCs in chemo-immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cyclophosphamide/therapeutic use , Immunity, Innate , Immunotherapy , Killer Cells, Natural/immunology , Melanoma, Experimental/drug therapy , Adaptive Immunity , Animals , Antigens, Ly/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Oxidoreductases/immunology , Receptors, IgG/immunology , Thy-1 Antigens/metabolism
9.
Proc Natl Acad Sci U S A ; 111(45): 16136-41, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25331895

ABSTRACT

Depression and anxiety disorders are associated with increased release of peripheral cytokines; however, their functional relevance remains unknown. Using a social stress model in mice, we find preexisting individual differences in the sensitivity of the peripheral immune system that predict and promote vulnerability to social stress. Cytokine profiles were obtained 20 min after the first social stress exposure. Of the cytokines regulated by stress, IL-6 was most highly up-regulated only in mice that ultimately developed a susceptible behavioral phenotype following a subsequent chronic stress, and levels remained elevated for at least 1 mo. We confirmed a similar elevation of serum IL-6 in two separate cohorts of patients with treatment-resistant major depressive disorder. Before any physical contact in mice, we observed individual differences in IL-6 levels from ex vivo stimulated leukocytes that predict susceptibility versus resilience to a subsequent stressor. To shift the sensitivity of the peripheral immune system to a pro- or antidepressant state, bone marrow (BM) chimeras were generated by transplanting hematopoietic progenitor cells from stress-susceptible mice releasing high IL-6 or from IL-6 knockout (IL-6(-/-)) mice. Stress-susceptible BM chimeras exhibited increased social avoidance behavior after exposure to either subthreshold repeated social defeat stress (RSDS) or a purely emotional stressor termed witness defeat. IL-6(-/-) BM chimeric and IL-6(-/-) mice, as well as those treated with a systemic IL-6 monoclonal antibody, were resilient to social stress. These data establish that preexisting differences in stress-responsive IL-6 release from BM-derived leukocytes functionally contribute to social stress-induced behavioral abnormalities.


Subject(s)
Anxiety Disorders/immunology , Behavior, Animal , Interleukin-6/immunology , Stress, Psychological/immunology , Allografts , Animals , Anxiety Disorders/genetics , Anxiety Disorders/pathology , Bone Marrow Transplantation , Disease Susceptibility/immunology , Disease Susceptibility/pathology , Interleukin-6/genetics , Mice , Mice, Knockout , Stress, Psychological/genetics , Stress, Psychological/pathology , Time Factors , Transplantation Chimera/genetics , Transplantation Chimera/immunology
10.
Cell ; 158(2): 300-313, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036630

ABSTRACT

Intestinal peristalsis is a dynamic physiologic process influenced by dietary and microbial changes. It is tightly regulated by complex cellular interactions; however, our understanding of these controls is incomplete. A distinct population of macrophages is distributed in the intestinal muscularis externa. We demonstrate that, in the steady state, muscularis macrophages regulate peristaltic activity of the colon. They change the pattern of smooth muscle contractions by secreting bone morphogenetic protein 2 (BMP2), which activates BMP receptor (BMPR) expressed by enteric neurons. Enteric neurons, in turn, secrete colony stimulatory factor 1 (CSF1), a growth factor required for macrophage development. Finally, stimuli from microbial commensals regulate BMP2 expression by macrophages and CSF1 expression by enteric neurons. Our findings identify a plastic, microbiota-driven crosstalk between muscularis macrophages and enteric neurons that controls gastrointestinal motility. PAPERFLICK:


Subject(s)
Gastrointestinal Motility , Gastrointestinal Tract/cytology , Gastrointestinal Tract/microbiology , Macrophages/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiology , In Vitro Techniques , Macrophage Colony-Stimulating Factor , Mice , Neurons/metabolism , Peristalsis , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction
11.
Transplantation ; 98(5): 563-8, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24717220

ABSTRACT

BACKGROUND: Langerhans cells (LCs) are self-renewing epidermal myeloid cells that can migrate and mature into dendritic cells. Recipient LCs that survive cytotoxic therapy given in preparation for allogeneic hematopoietic cell transplantation may prime donor T cells to mediate cutaneous graft-versus-host disease (GVHD). This possible association, however, has not been investigated in the setting of nonmyeloablative allografting. METHODS: We prospectively studied the kinetics of LC-chimerism after sex-mismatched allogeneic hematopoietic cell transplantation with nonmyeloablative (n=23) or myeloablative (n=25) conditioning. Combined XY-FISH and Langerin-staining was used to assess donor LC-chimerism in skin biopsies obtained on days 28, 56, and 84 after transplant. The degree of donor LC-chimerism was correlated with the development of skin GVHD. RESULTS: We observed significantly delayed donor LC-engraftment after nonmyeloablative transplantation compared with other hematopoietic compartments and compared with LC-engraftment after myeloablative conditioning. In most recipients of nonmyeloablative transplants, recipient LCs proliferated in situ, recruitment of donor-LCs was delayed by two months, and full donor LC-chimerism was only reached by day 84 after transplant. Although persistence of host LCs on day-28 after transplant was not predictive for acute or chronic skin GVHD, the recruitment of donor-derived LCs was associated with nonspecific inflammatory infiltrates (P=0.009). CONCLUSIONS: These results show that LCs can self-renew locally but are replaced by circulating precursors even after minimally toxic nonmyeloablative transplant conditioning. Cutaneous inflammation accompanies donor LC-engraftment, but differences in LC conversion-kinetics do not predict clinical or histopathological GVHD.


Subject(s)
Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/methods , Homeostasis , Langerhans Cells/physiology , Transplantation Chimera , Transplantation Conditioning/methods , Adult , Aged , Female , Graft vs Host Disease/prevention & control , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Prospective Studies , Transplantation, Homologous/methods
12.
J Immunol ; 192(7): 3374-82, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24591364

ABSTRACT

Neutrophils are the most abundant cell type in the immune system and play an important role in the innate immune response. Using a diverse range of mouse models with either defective dendritic cell (DC) development or conditional DC depletion, we provide in vivo evidence indicating that conventional DCs play an important role in the regulation of neutrophil homeostasis. Flk2, Flt3L, and Batf3 knockout mice, which have defects in DC development, have increased numbers of liver neutrophils in the steady state. Conversely, neutrophil frequency is reduced in DC-specific PTEN knockout mice, which have an expansion of CD8(+) and CD103(+) DCs. In chimeric CD11c-DTR mice, conventional DC depletion results in a systemic increase of neutrophils in peripheral organs in the absence of histological inflammation or an increase in proinflammatory cytokines. This effect is also present in splenectomized chimeric CD11c-DTR mice and is absent in chimeric mice with 50% normal bone marrow. In chimeric CD11c-DTR mice, diphtheria toxin treatment results in enhanced neutrophil trafficking from the bone marrow into circulation and increased neutrophil recruitment. Moreover, there is an increased expression of chemokines/cytokines involved in neutrophil homeostasis and reduced neutrophil apoptosis. These data underscore the role of the DC pool in regulating the neutrophil compartment in nonlymphoid organs.


Subject(s)
Bone Marrow/immunology , Dendritic Cells/immunology , Homeostasis/immunology , Neutrophils/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Bone Marrow/metabolism , Bone Marrow Transplantation , CD11c Antigen/genetics , CD11c Antigen/immunology , CD11c Antigen/metabolism , Cell Survival/genetics , Cell Survival/immunology , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Female , Flow Cytometry , Heparin-binding EGF-like Growth Factor , Homeostasis/genetics , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Liver/immunology , Liver/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Neutrophils/metabolism , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/immunology , fms-Like Tyrosine Kinase 3/deficiency , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/immunology
13.
J Exp Med ; 211(4): 669-83, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24638167

ABSTRACT

Langerhans cell histiocytosis (LCH) is a clonal disorder with elusive etiology, characterized by the accumulation of CD207(+) dendritic cells (DCs) in inflammatory lesions. Recurrent BRAF-V600E mutations have been reported in LCH. In this study, lesions from 100 patients were genotyped, and 64% carried the BRAF-V600E mutation within infiltrating CD207(+) DCs. BRAF-V600E expression in tissue DCs did not define specific clinical risk groups but was associated with increased risk of recurrence. Strikingly, we found that patients with active, high-risk LCH also carried BRAF-V600E in circulating CD11c(+) and CD14(+) fractions and in bone marrow (BM) CD34(+) hematopoietic cell progenitors, whereas the mutation was restricted to lesional CD207(+) DC in low-risk LCH patients. Importantly, BRAF-V600E expression in DCs was sufficient to drive LCH-like disease in mice. Consistent with our findings in humans, expression of BRAF-V600E in BM DC progenitors recapitulated many features of the human high-risk LCH, whereas BRAF-V600E expression in differentiated DCs more closely resembled low-risk LCH. We therefore propose classification of LCH as a myeloid neoplasia and hypothesize that high-risk LCH arises from somatic mutation of a hematopoietic progenitor, whereas low-risk disease arises from somatic mutation of tissue-restricted precursor DCs.


Subject(s)
Cell Differentiation , Dendritic Cells/metabolism , Genetic Predisposition to Disease , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/pathology , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Animals , Antigens, CD34/metabolism , Antigens, Surface/metabolism , Bone Marrow/pathology , CD11c Antigen/metabolism , Cell Lineage , Child , Child, Preschool , Female , Hematopoietic Stem Cells/metabolism , Histiocytosis, Langerhans-Cell/blood , Histocompatibility Antigens Class II/metabolism , Humans , Infant , Lectins, C-Type/metabolism , Male , Mannose-Binding Lectins/metabolism , Mice , Phenotype , Risk Factors , Treatment Outcome
14.
Nat Immunol ; 15(1): 54-62, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24270517

ABSTRACT

miR-126 is a microRNA expressed predominately by endothelial cells and controls angiogenesis. We found miR-126 was required for the innate response to pathogen-associated nucleic acids and that miR-126-deficient mice had greater susceptibility to infection with pseudotyped HIV. Profiling of miRNA indicated that miR-126 had high and specific expression by plasmacytoid dendritic cells (pDCs). Moreover, miR-126 controlled the survival and function of pDCs and regulated the expression of genes encoding molecules involved in the innate response, including Tlr7, Tlr9 and Nfkb1, as well as Kdr, which encodes the growth factor receptor VEGFR2. Deletion of Kdr in DCs resulted in reduced production of type I interferon, which supports the proposal of a role for VEGFR2 in miR-126 regulation of pDCs. Our studies identify the miR-126-VEGFR2 axis as an important regulator of the innate response that operates through multiscale control of pDCs.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate/immunology , MicroRNAs/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Dendritic Cells/metabolism , Flow Cytometry , HIV Infections/immunology , HIV Infections/virology , Humans , Immunity, Innate/genetics , Immunoblotting , Interferon-alpha/blood , Interferon-alpha/immunology , Interferon-alpha/metabolism , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/immunology , NF-kappa B p50 Subunit/metabolism , Nucleic Acids/immunology , Nucleic Acids/metabolism , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/immunology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , Transcriptome/immunology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Immunity ; 38(4): 818-30, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23562160

ABSTRACT

In comparison to murine dendritic cells (DCs), less is known about the function of human DCs in tissues. Here, we analyzed, by using lung tissues from humans and humanized mice, the role of human CD1c(+) and CD141(+) DCs in determining the type of CD8(+) T cell immunity generated to live-attenuated influenza virus (LAIV) vaccine. We found that both lung DC subsets acquired influenza antigens in vivo and expanded specific cytotoxic CD8(+) T cells in vitro. However, lung-tissue-resident CD1c(+) DCs, but not CD141(+) DCs, were able to drive CD103 expression on CD8(+) T cells and promoted CD8(+) T cell accumulation in lung epithelia in vitro and in vivo. CD1c(+) DCs induction of CD103 expression was dependent on membrane-bound cytokine TGF-ß1. Thus, CD1c(+) and CD141(+) DCs generate CD8(+) T cells with different properties, and CD1c(+) DCs specialize in the regulation of mucosal CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lung/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/metabolism , Antigens, CD1/metabolism , Antigens, Viral/immunology , Cell Differentiation , Cells, Cultured , Cytotoxicity, Immunologic , Glycoproteins/metabolism , Humans , Immunity, Mucosal , Immunologic Memory , Influenza Vaccines/immunology , Integrin alpha Chains/metabolism , Lung/virology , Lymphocyte Activation , Mice , Mice, SCID , Microarray Analysis
17.
Immunity ; 38(4): 792-804, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601688

ABSTRACT

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


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lung/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Adult , Animals , Bone Marrow Transplantation , Cell Proliferation , Cell Survival , Cells, Cultured , Homeostasis , Humans , Interleukin-4/metabolism , Macrophages/transplantation , Mice , Mice, Knockout , Mice, Mutant Strains , Parabiosis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
18.
Nat Med ; 19(4): 429-36, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23502962

ABSTRACT

A role for macrophages in erythropoiesis was suggested several decades ago when erythroblastic islands in the bone marrow, composed of a central macrophage surrounded by developing erythroblasts, were described. However, the in vivo role of macrophages in erythropoiesis under homeostatic conditions or in disease remains unclear. We found that specific depletion of CD169(+) macrophages markedly reduced the number of erythroblasts in the bone marrow but did not result in overt anemia under homeostatic conditions, probably because of concomitant alterations in red blood cell clearance. However, CD169(+) macrophage depletion significantly impaired erythropoietic recovery from hemolytic anemia, acute blood loss and myeloablation. Furthermore, macrophage depletion normalized the erythroid compartment in a JAK2(V617F)-driven mouse model of polycythemia vera, suggesting that erythropoiesis in polycythemia vera remains under the control of macrophages in the bone marrow and splenic microenvironments. These results indicate that CD169(+) macrophages promote late erythroid maturation and that modulation of the macrophage compartment may be a new strategy to treat erythropoietic disorders.


Subject(s)
Erythropoiesis/physiology , Homeostasis/physiology , Macrophages/physiology , Sialic Acid Binding Ig-like Lectin 1/physiology , Stress, Physiological/physiology , Anemia/physiopathology , Anemia, Hemolytic/physiopathology , Animals , Erythroblasts/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Polycythemia Vera/physiopathology , Spleen/physiology , Vascular Cell Adhesion Molecule-1/physiology
19.
Immunity ; 37(6): 1050-1060, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23177320

ABSTRACT

Colony stimulating factor-1 (Csf-1) receptor and its ligand Csf-1 control macrophage development, maintenance, and function. The development of both Langerhans cells (LCs) and microglia is highly dependent on Csf-1 receptor signaling but independent of Csf-1. Here we show that in both mice and humans, interleukin-34 (IL-34), an alternative ligand for Csf-1 receptor, is produced by keratinocytes in the epidermis and by neurons in the brain. Mice lacking IL-34 displayed a marked reduction of LCs and a decrease of microglia, whereas monocytes, dermal, and lymphoid tissue macrophages and DCs were unaffected. We identified IL-34 as a nonredundant cytokine for the development of LCs during embryogenesis as well as for their homeostasis in the adult skin. Whereas inflammation-induced repopulation of LCs appears to be dependent on Csf-1, once inflammation is resolved, LC survival is again IL-34-dependent. In contrast, microglia and their yolk sac precursors develop independently of IL-34 but rely on it for their maintenance in the adult brain.


Subject(s)
Interleukins/physiology , Langerhans Cells/immunology , Microglia/immunology , Stromal Cells/metabolism , Animals , Brain/immunology , Brain/metabolism , Cell Differentiation/genetics , Epidermis/immunology , Epidermis/metabolism , Homeostasis , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interleukins/genetics , Interleukins/immunology , Interleukins/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Langerhans Cells/cytology , Langerhans Cells/metabolism , Mice , Microglia/cytology , Microglia/metabolism , Psoriasis/chemically induced , Psoriasis/immunology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Skin/immunology , Skin/metabolism
20.
J Clin Invest ; 122(11): 4037-47, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23041628

ABSTRACT

CD8+ cytotoxic T cells are critical for viral clearance from the lungs upon influenza virus infection. The contribution of antigen cross-presentation by DCs to the induction of anti-viral cytotoxic T cells remains controversial. Here, we used a recombinant influenza virus expressing a nonstructural 1-GFP (NS1-GFP) reporter gene to visualize the route of antigen presentation by lung DCs upon viral infection in mice. We found that lung CD103+ DCs were the only subset of cells that carried intact GFP protein to the draining LNs. Strikingly, lung migratory CD103+ DCs were not productively infected by influenza virus and thus were able to induce virus-specific CD8+ T cells through the cross-presentation of antigens from virally infected cells. We also observed that CD103+ DC resistance to infection correlates with an increased anti-viral state in these cells that is dependent on the expression of type I IFN receptor. These results show that efficient cross-priming by migratory lung DCs is coupled to the acquisition of an anti-viral status, which is dependent on the type I IFN signaling pathway.


Subject(s)
Antigen Presentation , Antigens, CD , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Influenza A virus/immunology , Integrin alpha Chains , Orthomyxoviridae Infections/prevention & control , Animals , Antigens, Viral/genetics , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line , Dendritic Cells/pathology , Dogs , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Influenza A virus/genetics , Interferon Type I/genetics , Interferon Type I/immunology , Lung/immunology , Lung/pathology , Lung/virology , Mice , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...