Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cell ; 187(15): 4078-4094.e21, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38897196

ABSTRACT

Reversing CD8+ T cell dysfunction is crucial in treating chronic hepatitis B virus (HBV) infection, yet specific molecular targets remain unclear. Our study analyzed co-signaling receptors during hepatocellular priming and traced the trajectory and fate of dysfunctional HBV-specific CD8+ T cells. Early on, these cells upregulate PD-1, CTLA-4, LAG-3, OX40, 4-1BB, and ICOS. While blocking co-inhibitory receptors had minimal effect, activating 4-1BB and OX40 converted them into antiviral effectors. Prolonged stimulation led to a self-renewing, long-lived, heterogeneous population with a unique transcriptional profile. This includes dysfunctional progenitor/stem-like (TSL) cells and two distinct dysfunctional tissue-resident memory (TRM) populations. While 4-1BB expression is ubiquitously maintained, OX40 expression is limited to TSL. In chronic settings, only 4-1BB stimulation conferred antiviral activity. In HBeAg+ chronic patients, 4-1BB activation showed the highest potential to rejuvenate dysfunctional CD8+ T cells. Targeting all dysfunctional T cells, rather than only stem-like precursors, holds promise for treating chronic HBV infection.


Subject(s)
CD8-Positive T-Lymphocytes , Hepatitis B virus , Hepatitis B, Chronic , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , Hepatitis B, Chronic/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Signal Transduction , Animals , Receptors, OX40/metabolism , Mice , Programmed Cell Death 1 Receptor/metabolism , Antigens, CD/metabolism
2.
Cell ; 183(5): 1282-1297.e18, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33098771

ABSTRACT

Classically considered short-lived and purely defensive leukocytes, neutrophils are unique in their fast and moldable response to stimulation. This plastic behavior may underlie variable and even antagonistic functions during inflammation or cancer, yet the full spectrum of neutrophil properties as they enter healthy tissues remains unexplored. Using a new model to track neutrophil fates, we found short but variable lifetimes across multiple tissues. Through analysis of the receptor, transcriptional, and chromatin accessibility landscapes, we identify varying neutrophil states and assign non-canonical functions, including vascular repair and hematopoietic homeostasis. Accordingly, depletion of neutrophils compromised angiogenesis during early age, genotoxic injury, and viral infection, and impaired hematopoietic recovery after irradiation. Neutrophils acquired these properties in target tissues, a process that, in the lungs, occurred in CXCL12-rich areas and relied on CXCR4. Our results reveal that tissues co-opt neutrophils en route for elimination to induce programs that support their physiological demands.


Subject(s)
Cell Lineage , Neutrophils/metabolism , Organ Specificity , Animals , Chromatin/metabolism , Female , Hematopoiesis , Intestines/blood supply , Lung/blood supply , Male , Mice, Inbred C57BL , Neovascularization, Physiologic , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, CXCR4/metabolism , Single-Cell Analysis , Transcription, Genetic , Transcriptome/genetics
3.
Nat Immunol ; 23(10): 1470-1483, 2022 10.
Article in English | MEDLINE | ID: mdl-36138183

ABSTRACT

Traditionally viewed as poorly plastic, neutrophils are now recognized as functionally diverse; however, the extent and determinants of neutrophil heterogeneity in humans remain unclear. We performed a comprehensive immunophenotypic and transcriptome analysis, at a bulk and single-cell level, of neutrophils from healthy donors and patients undergoing stress myelopoiesis upon exposure to growth factors, transplantation of hematopoietic stem cells (HSC-T), development of pancreatic cancer and viral infection. We uncover an extreme diversity of human neutrophils in vivo, reflecting the rates of cell mobilization, differentiation and exposure to environmental signals. Integrated control of developmental and inducible transcriptional programs linked flexible granulopoietic outputs with elicitation of stimulus-specific functional responses. In this context, we detected an acute interferon (IFN) response in the blood of patients receiving HSC-T that was mirrored by marked upregulation of IFN-stimulated genes in neutrophils but not in monocytes. Systematic characterization of human neutrophil plasticity may uncover clinically relevant biomarkers and support the development of diagnostic and therapeutic tools.


Subject(s)
Myelopoiesis , Neutrophils , Biomarkers/metabolism , Humans , Interferons/genetics , Interferons/metabolism , Neutrophils/metabolism , Plastics/metabolism
4.
Nat Immunol ; 20(7): 783-792, 2019 07.
Article in English | MEDLINE | ID: mdl-31213714

ABSTRACT

Adaptation is the ability of cells, tissues and organisms to rapidly and reversibly modify their properties to maximize fitness in a changing environment. The activity of immune-system components unfolds in the remarkably heterogeneous milieus to which they are exposed in different tissues, during homeostasis or during various acute or chronic pathological states. Therefore, adaptation is essential for immune cells to tune their responses to a large variety of contexts and conditions. The adaptation of immune cells reflects the integration of multiple inputs acting simultaneously or in a temporal sequence, which eventually leads to transcriptional reprogramming and to various functional consequences, some of which extend beyond the duration of the stimulus. A range of adaptive responses have been observed in both adaptive immune cells and innate immune cells; these are referred to with terms such as 'plasticity', 'priming', 'training', 'exhaustion' and 'tolerance', among others, all of which can be useful for defining a certain immunological process or outcome but whose underlying molecular frameworks are often incompletely understood. Here we review and analyze mechanisms of adaptation and memory in immunity with the aim of providing basic concepts that rationalize the properties and molecular bases of these essential processes.


Subject(s)
Adaptation, Physiological , Immunity , Immunologic Memory , Adaptive Immunity , Animals , Gene Expression Regulation , Histones/metabolism , Humans , Hypersensitivity/immunology , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Immune Tolerance , Immunity, Innate , Organ Specificity/immunology , Phenotype , Signal Transduction
5.
Immunity ; 55(6): 973-975, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35704999

ABSTRACT

In this issue of Immunity, Eisenstein, Hiliard, et al., uncover a novel mechanism of some widely used non-steroidal anti-inflammatory drugs (NSAIDs): activation of the antioxidant transcription factor NRF2 in myeloid immune cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Inflammation , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Humans
6.
Nat Immunol ; 24(5): 746-748, 2023 05.
Article in English | MEDLINE | ID: mdl-37095374
7.
Immunity ; 54(8): 1665-1682.e14, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34129840

ABSTRACT

Tight control of inflammatory gene expression by antagonistic environmental cues is key to ensure immune protection while preventing tissue damage. Prostaglandin E2 (PGE2) modulates macrophage activation during homeostasis and disease, but the underlying mechanisms remain incompletely characterized. Here we dissected the genomic properties of lipopolysaccharide (LPS)-induced genes whose expression is antagonized by PGE2. The latter molecule targeted a set of inflammatory gene enhancers that, already in unstimulated macrophages, displayed poorly permissive chromatin organization and were marked by the transcription factor myocyte enhancer factor 2A (MEF2A). Deletion of MEF2A phenocopied PGE2 treatment and abolished type I interferon (IFN I) induction upon exposure to innate immune stimuli. Mechanistically, PGE2 interfered with LPS-mediated activation of ERK5, a known transcriptional partner of MEF2. This study highlights principles of plasticity and adaptation in cells exposed to a complex environment and uncovers a transcriptional circuit for IFN I induction with relevance for infectious diseases or cancer.


Subject(s)
Dinoprostone/immunology , Interferon Type I/immunology , Macrophage Activation/immunology , Macrophages/immunology , Animals , Cell Line , Cells, Cultured , Gene Expression Regulation/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Interferon Type I/biosynthesis , Lipopolysaccharides , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 7/metabolism
8.
Nat Immunol ; 18(5): 530-540, 2017 05.
Article in English | MEDLINE | ID: mdl-28288101

ABSTRACT

Stimulation of macrophages with interferon-γ (IFN-γ) and interleukin 4 (IL-4) triggers distinct and opposing activation programs. During mixed infections or cancer, macrophages are often exposed to both cytokines, but how these two programs influence each other remains unclear. We found that IFN-γ and IL-4 mutually inhibited the epigenomic and transcriptional changes induced by each cytokine alone. Computational and functional analyses revealed the genomic bases for gene-specific cross-repression. For instance, while binding motifs for the transcription factors STAT1 and IRF1 were associated with robust and IL-4-resistant responses to IFN-γ, their coexistence with binding sites for auxiliary transcription factors such as AP-1 generated vulnerability to IL-4-mediated inhibition. These data provide a core mechanistic framework for the integration of signals that control macrophage activation in complex environmental conditions.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Macrophages/physiology , Proto-Oncogene Proteins c-myc/metabolism , Transcriptional Activation , Animals , Cell Line , Gene Expression Regulation , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred Strains , Proto-Oncogene Proteins c-myc/genetics , RNA, Small Interfering/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism
9.
Nature ; 623(7986): 415-422, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37914939

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with high resistance to therapies1. Inflammatory and immunomodulatory signals co-exist in the pancreatic tumour microenvironment, leading to dysregulated repair and cytotoxic responses. Tumour-associated macrophages (TAMs) have key roles in PDAC2, but their diversity has prevented therapeutic exploitation. Here we combined single-cell and spatial genomics with functional experiments to unravel macrophage functions in pancreatic cancer. We uncovered an inflammatory loop between tumour cells and interleukin-1ß (IL-1ß)-expressing TAMs, a subset of macrophages elicited by a local synergy between prostaglandin E2 (PGE2) and tumour necrosis factor (TNF). Physical proximity with IL-1ß+ TAMs was associated with inflammatory reprogramming and acquisition of pathogenic properties by a subset of PDAC cells. This occurrence was an early event in pancreatic tumorigenesis and led to persistent transcriptional changes associated with disease progression and poor outcomes for patients. Blocking PGE2 or IL-1ß activity elicited TAM reprogramming and antagonized tumour cell-intrinsic and -extrinsic inflammation, leading to PDAC control in vivo. Targeting the PGE2-IL-1ß axis may enable preventive or therapeutic strategies for reprogramming of immune dynamics in pancreatic cancer.


Subject(s)
Inflammation , Interleukin-1beta , Pancreatic Neoplasms , Tumor-Associated Macrophages , Humans , Carcinogenesis , Carcinoma, Pancreatic Ductal/complications , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Dinoprostone/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Tumor Necrosis Factors/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
10.
Cell ; 152(1-2): 157-71, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332752

ABSTRACT

According to current models, once the cell has reached terminal differentiation, the enhancer repertoire is completely established and maintained by cooperatively acting lineage-specific transcription factors (TFs). TFs activated by extracellular stimuli operate within this predetermined repertoire, landing close to where master regulators are constitutively bound. Here, we describe latent enhancers, defined as regions of the genome that in terminally differentiated cells are unbound by TFs and lack the histone marks characteristic of enhancers but acquire these features in response to stimulation. Macrophage stimulation caused sequential binding of stimulus-activated and lineage-determining TFs to these regions, enabling deposition of enhancer marks. Once unveiled, many of these enhancers did not return to a latent state when stimulation ceased; instead, they persisted and mediated a faster and stronger response upon restimulation. We suggest that stimulus-specific expansion of the cis-regulatory repertoire provides an epigenomic memory of the exposure to environmental agents.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Macrophages/metabolism , Animals , Cell Differentiation , Epigenomics , Histone Code , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism
11.
Cell ; 147(4): 868-80, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-22078883

ABSTRACT

The transport of Toll-like Receptors (TLRs) to various organelles has emerged as an essential means by which innate immunity is regulated. While most of our knowledge is restricted to regulators that promote the transport of newly synthesized receptors, the regulators that control TLR transport after microbial detection remain unknown. Here, we report that the plasma membrane localized Pattern Recognition Receptor (PRR) CD14 is required for the microbe-induced endocytosis of TLR4. In dendritic cells, this CD14-dependent endocytosis pathway is upregulated upon exposure to inflammatory mediators. We identify the tyrosine kinase Syk and its downstream effector PLCγ2 as important regulators of TLR4 endocytosis and signaling. These data establish that upon microbial detection, an upstream PRR (CD14) controls the trafficking and signaling functions of a downstream PRR (TLR4). This innate immune trafficking cascade illustrates how pathogen detection systems operate to induce both membrane transport and signal transduction.


Subject(s)
Endocytosis , Lipopolysaccharide Receptors/metabolism , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Dendritic Cells/immunology , Endosomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Phospholipase C gamma/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Syk Kinase
12.
Nature ; 574(7777): 200-205, 2019 10.
Article in English | MEDLINE | ID: mdl-31582858

ABSTRACT

The responses of CD8+ T cells to hepatotropic viruses such as hepatitis B range from dysfunction to differentiation into effector cells, but the mechanisms that underlie these distinct outcomes remain poorly understood. Here we show that priming by Kupffer cells, which are not natural targets of hepatitis B, leads to differentiation of CD8+ T cells into effector cells that form dense, extravascular clusters of immotile cells scattered throughout the liver. By contrast, priming by hepatocytes, which are natural targets of hepatitis B, leads to local activation and proliferation of CD8+ T cells but not to differentiation into effector cells; these cells form loose, intravascular clusters of motile cells that coalesce around portal tracts. Transcriptomic and chromatin accessibility analyses reveal unique features of these dysfunctional CD8+ T cells, with limited overlap with those of exhausted or tolerant T cells; accordingly, CD8+ T cells primed by hepatocytes cannot be rescued by treatment with anti-PD-L1, but instead respond to IL-2. These findings suggest immunotherapeutic strategies against chronic hepatitis B infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Hepatitis B virus/immunology , Hepatocytes/immunology , Hepatocytes/virology , Animals , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chromatin/metabolism , Female , Hepatitis B/drug therapy , Hepatitis B/immunology , Hepatitis B/virology , Humans , Immune Tolerance , Interleukin-2/immunology , Interleukin-2/therapeutic use , Kupffer Cells/immunology , Lymphocyte Activation , Male , Mice , Transcriptome/genetics
13.
Immunol Rev ; 300(1): 220-236, 2021 03.
Article in English | MEDLINE | ID: mdl-33565148

ABSTRACT

Most, if not all, aspects of carcinogenesis are influenced by the tumor microenvironment (TME), a complex architecture of cells, matrix components, soluble signals, and their dynamic interactions in the context of physical traits of the tissue. Expanding application of technologies for high-dimensional analyses with single-cell resolution has begun to decipher the contributions of the immune system to cancer progression and its implications for therapy. In this review, we will discuss the multifaceted roles of tumor-associated macrophages and neutrophils, focusing on factors that subvert tissue immune homeostasis and offer therapeutic opportunities for TME reprogramming. By performing a critical analysis of available datasets, we elaborate on diversification mechanisms and unifying principles of myeloid cell heterogeneity in human tumors.


Subject(s)
Neoplasms , Tumor Microenvironment , Carcinogenesis , Humans , Myeloid Cells , Neoplasms/therapy , Neutrophils
14.
Genes Dev ; 29(4): 394-408, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25637355

ABSTRACT

The transcription factor (TF) interferon regulatory factor 8 (IRF8) controls both developmental and inflammatory stimulus-inducible genes in macrophages, but the mechanisms underlying these two different functions are largely unknown. One possibility is that these different roles are linked to the ability of IRF8 to bind alternative DNA sequences. We found that IRF8 is recruited to distinct sets of DNA consensus sequences before and after lipopolysaccharide (LPS) stimulation. In resting cells, IRF8 was mainly bound to composite sites together with the master regulator of myeloid development PU.1. Basal IRF8-PU.1 binding maintained the expression of a broad panel of genes essential for macrophage functions (such as microbial recognition and response to purines) and contributed to basal expression of many LPS-inducible genes. After LPS stimulation, increased expression of IRF8, other IRFs, and AP-1 family TFs enabled IRF8 binding to thousands of additional regions containing low-affinity multimerized IRF sites and composite IRF-AP-1 sites, which were not premarked by PU.1 and did not contribute to the basal IRF8 cistrome. While constitutively expressed IRF8-dependent genes contained only sites mediating basal IRF8/PU.1 recruitment, inducible IRF8-dependent genes contained variable combinations of constitutive and inducible sites. Overall, these data show at the genome scale how the same TF can be linked to constitutive and inducible gene regulation via distinct combinations of alternative DNA-binding sites.


Subject(s)
Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Macrophages/metabolism , Animals , Cells, Cultured , Interferon Regulatory Factors/genetics , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Protein Binding
15.
Immunity ; 36(4): 572-85, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22483804

ABSTRACT

Histone methyltransferases catalyze site-specific deposition of methyl groups, enabling recruitment of transcriptional regulators. In mammals, trimethylation of lysine 4 in histone H3, a modification localized at the transcription start sites of active genes, is catalyzed by six enzymes (SET1a and SET1b, MLL1-MLL4) whose specific functions are largely unknown. By using a genomic approach, we found that in macrophages, MLL4 (also known as Wbp7) was required for the expression of Pigp, an essential component of the GPI-GlcNAc transferase, the enzyme catalyzing the first step of glycosylphosphatidylinositol (GPI) anchor synthesis. Impaired Pigp expression in Wbp7(-/-) macrophages abolished GPI anchor-dependent loading of proteins on the cell membrane. Consistently, loss of GPI-anchored CD14, the coreceptor for lipopolysaccharide (LPS) and other bacterial molecules, markedly attenuated LPS-triggered intracellular signals and gene expression changes. These data link a histone-modifying enzyme to a biosynthetic pathway and indicate a specialized biological role for Wbp7 in macrophage function and antimicrobial response.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Glycosylphosphatidylinositols/biosynthesis , Hexosyltransferases/biosynthesis , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharides/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/biosynthesis , Myeloid-Lymphoid Leukemia Protein/genetics , Signal Transduction
16.
Semin Immunol ; 28(2): 83-93, 2016 04.
Article in English | MEDLINE | ID: mdl-27084194

ABSTRACT

In addition to performing well-defined effector functions, neutrophils are now recognized as versatile and sophisticated cells with critical immunoregulatory roles. These include the release of a variety of proinflammatory or immunosuppressive cytokines, as well as the expression of genes with regulatory functions. Neutrophils share broad transcriptional features with monocytes, in keeping with the close developmental relation between the two cell types. However, neutrophil-specific gene expression patterns conferring cell type-specific responses to bacterial, viral or fungal components have been identified. Accumulating evidence suggest that these differences reflect the peculiar epigenomic and regulatory landscapes of neutrophils and monocytes, in turn controlled by the specific lineage-determining transcription factors shaping their identity. In this review, we will describe current knowledge on how neutrophil identity and function are controlled at the molecular level, focusing on transcriptional and chromatin regulation of neutrophil development and activation in response to inflammatory stimuli.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation , Immunity, Innate , Neutrophils/cytology , Neutrophils/physiology , Animals , Cytokines/genetics , Cytokines/metabolism , DNA Methylation , Humans , Myelopoiesis/genetics
17.
Hepatology ; 65(5): 1708-1719, 2017 05.
Article in English | MEDLINE | ID: mdl-27859418

ABSTRACT

The ST18 gene has been proposed to act either as a tumor suppressor or as an oncogene in different human cancers, but direct evidence for its role in tumorigenesis has been lacking thus far. Here, we demonstrate that ST18 is critical for tumor progression and maintenance in a mouse model of liver cancer, based on oncogenic transformation and adoptive transfer of primary precursor cells (hepatoblasts). ST18 messenger RNA (mRNA) and protein were detectable neither in normal liver nor in cultured hepatoblasts, but were readily expressed after subcutaneous engraftment and tumor growth. ST18 expression in liver cells was induced by inflammatory cues, including acute or chronic inflammation in vivo, as well as coculture with macrophages in vitro. Knocking down the ST18 mRNA in transplanted hepatoblasts delayed tumor progression. Induction of ST18 knockdown in pre-established tumors caused rapid tumor involution associated with pervasive morphological changes, proliferative arrest, and apoptosis in tumor cells, as well as depletion of tumor-associated macrophages, vascular ectasia, and hemorrhage. Reciprocally, systemic depletion of macrophages in recipient animals had very similar phenotypic consequences, impairing either tumor development or maintenance, and suppressing ST18 expression in hepatoblasts. Finally, RNA sequencing of ST18-depleted tumors before involution revealed down-regulation of inflammatory response genes, pointing to the suppression of nuclear factor kappa B-dependent transcription. CONCLUSION: ST18 expression in epithelial cells is induced by tumor-associated macrophages, contributing to the reciprocal feed-forward loop between both cell types in liver tumorigenesis. Our findings warrant the exploration of means to interfere with ST18-dependent epithelium-macrophage interactions in a therapeutic setting. (Hepatology 2017;65:1708-1719).


Subject(s)
Carcinoma, Hepatocellular/etiology , Liver Neoplasms, Experimental/etiology , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms, Experimental/metabolism , Mice, Inbred C57BL
18.
Trends Immunol ; 36(4): 229-39, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25770924

ABSTRACT

Infiltration by immune cells is a hallmark of most forms of malignancy. In this context, tumor-associated macrophages (TAMs) represent key regulators of the complex interplay between the immune system and cancer. We discuss evidence indicating that in many settings TAMs fuel, rather than limit, tumor progression, and negatively impact on responses to therapy. We discuss how the unique functional properties of TAMs are shaped by tumor-derived signals, placing TAM development in the context of the broader understanding of the cellular and molecular mechanisms controlling macrophage origin, differentiation, and maintenance in tissues. Finally, we provide examples of how a molecular understanding of the relationships between TAMs and the tumor microenvironment may lead to improved cancer therapies.


Subject(s)
Macrophages/immunology , Neoplasms/immunology , Neoplasms/therapy , Humans , Neoplasms/pathology , Tumor Microenvironment/drug effects
19.
J Immunol ; 190(5): 1921-5, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355741

ABSTRACT

To identify the molecular basis of IL-10 expression in human phagocytes, we evaluated the chromatin modification status at their IL-10 genomic locus. We analyzed posttranslational modifications of histones associated with genes that are active, repressed, or poised for transcriptional activation, including H3K4me3, H4Ac, H3K27Ac, and H3K4me1 marks. Differently from autologous IL-10-producing monocytes, none of the marks under evaluation was detected at the IL-10 locus of resting or activated neutrophils from healthy subjects or melanoma patients. By contrast, increased H3K4me3, H4Ac, H3K4me1, and H3K27Ac levels were detected at syntenic regions of the IL-10 locus in mouse neutrophils. Altogether, data demonstrate that human neutrophils, differently from either monocytes or mouse neutrophils, cannot switch on the IL-10 gene because its locus is in an inactive state, likely reflecting a neutrophil-specific developmental outcome. Implicitly, data also definitively settle a currently unsolved issue on the capacity of human neutrophils to produce IL-10.


Subject(s)
Chromatin/genetics , Histones/genetics , Interleukin-10/genetics , Melanoma/genetics , Neutrophils/metabolism , Protein Processing, Post-Translational , Skin Neoplasms/genetics , Animals , Cells, Cultured , Chromatin/chemistry , Chromatin/immunology , Chromatin Immunoprecipitation , Enhancer Elements, Genetic , Gene Expression Regulation , Genetic Loci , Histones/immunology , Humans , Interleukin-10/immunology , Melanoma/immunology , Melanoma/pathology , Methylation , Mice , Monocytes/immunology , Monocytes/metabolism , Neutrophils/immunology , Protein Conformation , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Species Specificity , Synteny
20.
Nature ; 460(7252): 264-8, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19525933

ABSTRACT

Toll-like receptors (TLRs) are the best characterized pattern recognition receptors. Individual TLRs recruit diverse combinations of adaptor proteins, triggering signal transduction pathways and leading to the activation of various transcription factors, including nuclear factor kappaB, activation protein 1 and interferon regulatory factors. Interleukin-2 is one of the molecules produced by mouse dendritic cells after stimulation by different pattern recognition receptor agonists. By analogy with the events after T-cell receptor engagement leading to interleukin-2 production, it is therefore plausible that the stimulation of TLRs on dendritic cells may lead to activation of the Ca(2+)/calcineurin and NFAT (nuclear factor of activated T cells) pathway. Here we show that mouse dendritic cell stimulation with lipopolysaccharide (LPS) induces Src-family kinase and phospholipase Cgamma2 activation, influx of extracellular Ca(2+) and calcineurin-dependent nuclear NFAT translocation. The initiation of this pathway is independent of TLR4 engagement, and dependent exclusively on CD14. We also show that LPS-induced NFAT activation via CD14 is necessary to cause the apoptotic death of terminally differentiated dendritic cells, an event that is essential for maintaining self-tolerance and preventing autoimmunity. Consequently, blocking this pathway in vivo causes prolonged dendritic cell survival and an increase in T-cell priming capability. Our findings reveal novel aspects of molecular signalling triggered by LPS in dendritic cells, and identify a new role for CD14: the regulation of the dendritic cell life cycle through NFAT activation. Given the involvement of CD14 in disease, including sepsis and chronic heart failure, the discovery of signal transduction pathways activated exclusively via CD14 is an important step towards the development of potential treatments involving interference with CD14 functions.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/immunology , NFATC Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Calcium/metabolism , Calcium Signaling/drug effects , Cell Differentiation , Cell Survival/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Phospholipase C gamma/metabolism , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL