Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 24(5): 767-779, 2023 05.
Article in English | MEDLINE | ID: mdl-37095375

ABSTRACT

Sepsis arises from diverse and incompletely understood dysregulated host response processes following infection that leads to life-threatening organ dysfunction. Here we showed that neutrophils and emergency granulopoiesis drove a maladaptive response during sepsis. We generated a whole-blood single-cell multiomic atlas (272,993 cells, n = 39 individuals) of the sepsis immune response that identified populations of immunosuppressive mature and immature neutrophils. In co-culture, CD66b+ sepsis neutrophils inhibited proliferation and activation of CD4+ T cells. Single-cell multiomic mapping of circulating hematopoietic stem and progenitor cells (HSPCs) (29,366 cells, n = 27) indicated altered granulopoiesis in patients with sepsis. These features were enriched in a patient subset with poor outcome and a specific sepsis response signature that displayed higher frequencies of IL1R2+ immature neutrophils, epigenetic and transcriptomic signatures of emergency granulopoiesis in HSPCs and STAT3-mediated gene regulation across different infectious etiologies and syndromes. Our findings offer potential therapeutic targets and opportunities for stratified medicine in severe infection.


Subject(s)
Neutrophils , Sepsis , Humans , Hematopoiesis , Hematopoietic Stem Cells , Gene Expression Regulation
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 ; 22(9): 1093-1106, 2021 09.
Article in English | MEDLINE | ID: mdl-34282331

ABSTRACT

Neutrophils display distinct gene expression patters depending on their developmental stage, activation state and tissue microenvironment. To determine the transcription factor networks that shape these responses in a mouse model, we integrated transcriptional and chromatin analyses of neutrophils during acute inflammation. We showed active chromatin remodeling at two transition stages: bone marrow-to-blood and blood-to-tissue. Analysis of differentially accessible regions revealed distinct sets of putative transcription factors associated with control of neutrophil inflammatory responses. Using ex vivo and in vivo approaches, we confirmed that RUNX1 and KLF6 modulate neutrophil maturation, whereas RELB, IRF5 and JUNB drive neutrophil effector responses and RFX2 and RELB promote survival. Interfering with neutrophil activation by targeting one of these factors, JUNB, reduced pathological inflammation in a mouse model of myocardial infarction. Therefore, our study represents a blueprint for transcriptional control of neutrophil responses in acute inflammation and opens possibilities for stage-specific therapeutic modulation of neutrophil function in disease.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Inflammation/immunology , Neutrophils/immunology , Transcriptional Activation/genetics , Animals , CHO Cells , Cell Line , Core Binding Factor Alpha 2 Subunit/metabolism , Cricetulus , Female , Interferon Regulatory Factors/metabolism , Kruppel-Like Factor 6/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Regulatory Factor X Transcription Factors/metabolism , Transcription Factor RelB/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics
4.
Immunity ; 55(12): 2217-2219, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36516813

ABSTRACT

Inflammatory insults affect platelet production, but it is yet unknown what mechanisms can drive rapid adaptations in thrombopoiesis. In this issue of Immunity, Petzold et al. (2022) propose that neutrophils "pluck" on megakaryocytes in the bone marrow to tune platelet release.


Subject(s)
Blood Platelets , Neutrophils , Thrombopoiesis , Megakaryocytes , Bone Marrow
5.
Trends Immunol ; 42(9): 795-806, 2021 09.
Article in English | MEDLINE | ID: mdl-34373208

ABSTRACT

The tissue microenvironment is a major driver in imprinting tissue-specific macrophage functions in various mammalian tissues. As monocytes are recruited into the gastrointestinal (GI) tract at steady state and inflammation, they rapidly adopt a tissue-specific and distinct transcriptome. However, the GI tract varies significantly along its length, yet most studies of intestinal macrophages do not directly compare the phenotype and function of these macrophages in the small and large intestine, thus leading to disparities in data interpretations. This review highlights differences along the GI tract that are likely to influence macrophage function, with a specific focus on diet and microbiota. This analysis may fuel further investigation regarding the interplay between the intestinal immune system and GI tissue microenvironments, ideally providing unique therapeutic targets to modulate specific intestinal macrophage populations and/or functions.


Subject(s)
Gastrointestinal Tract , Microbiota , Animals , Intestines , Macrophages , Monocytes
6.
Nat Immunol ; 12(3): 231-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21240265

ABSTRACT

Polymorphisms in the gene encoding the transcription factor IRF5 that lead to higher mRNA expression are associated with many autoimmune diseases. Here we show that IRF5 expression in macrophages was reversibly induced by inflammatory stimuli and contributed to the plasticity of macrophage polarization. High expression of IRF5 was characteristic of M1 macrophages, in which it directly activated transcription of the genes encoding interleukin 12 subunit p40 (IL-12p40), IL-12p35 and IL-23p19 and repressed the gene encoding IL-10. Consequently, those macrophages set up the environment for a potent T helper type 1 (T(H)1)-T(H)17 response. Global gene expression analysis demonstrated that exogenous IRF5 upregulated or downregulated expression of established phenotypic markers of M1 or M2 macrophages, respectively. Our data suggest a critical role for IRF5 in M1 macrophage polarization and define a previously unknown function for IRF5 as a transcriptional repressor.


Subject(s)
Interferon Regulatory Factors/immunology , Macrophages/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Flow Cytometry , Humans , Immunoblotting , Interferon Regulatory Factors/genetics , Mice , Mice, Knockout , Microarray Analysis
7.
Nat Immunol ; 13(1): 95-102, 2011 Nov 20.
Article in English | MEDLINE | ID: mdl-22101729

ABSTRACT

The unique DNA-binding properties of distinct NF-κB dimers influence the selective regulation of NF-κB target genes. To more thoroughly investigate these dimer-specific differences, we combined protein-binding microarrays and surface plasmon resonance to evaluate DNA sites recognized by eight different NF-κB dimers. We observed three distinct binding-specificity classes and clarified mechanisms by which dimers might regulate distinct sets of genes. We identified many new nontraditional NF-κB binding site (κB site) sequences and highlight the plasticity of NF-κB dimers in recognizing κB sites with a single consensus half-site. This study provides a database that can be used in efforts to identify NF-κB target sites and uncover gene regulatory circuitry.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cluster Analysis , DNA/chemistry , DNA/genetics , Databases, Genetic , Humans , Macrophages/metabolism , Mice , NF-kappa B/genetics , Protein Array Analysis , Protein Binding , Protein Multimerization
8.
J Immunol ; 206(7): 1515-1527, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33608456

ABSTRACT

The PI3K pathway plays a key role in B cell activation and is important for the differentiation of Ab producing plasma cells (PCs). Although much is known about the molecular mechanisms that modulate PI3K signaling in B cells, the transcriptional regulation of PI3K expression is poorly understood. In this study, we identify the zinc finger protein Zbtb18 as a transcriptional repressor that directly binds enhancer/promoter regions of genes encoding class I PI3K regulatory subunits, subsequently limiting their expression, dampening PI3K signaling and suppressing PC responses. Following activation, dividing B cells progressively downregulated Zbtb18, allowing gradual amplification of PI3K signals and enhanced development of PCs. Human Zbtb18 displayed similar expression patterns and function in human B cells, acting to inhibit development of PCs. Furthermore, a number of Zbtb18 mutants identified in cancer patients showed loss of suppressor activity, which was also accompanied by impaired regulation of PI3K genes. Taken together, our study identifies Zbtb18 as a repressor of PC differentiation and reveals its previously unappreciated function as a transcription modulator of the PI3K signaling pathway.


Subject(s)
B-Lymphocytes/immunology , Neoplasms/immunology , Phosphatidylinositol 3-Kinases/metabolism , Plasma Cells/immunology , Repressor Proteins/metabolism , Animals , Cell Differentiation , Gene Expression Regulation , Humans , Immunity, Humoral , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Repressor Proteins/genetics , Signal Transduction
9.
Circulation ; 144(12): 961-982, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34255973

ABSTRACT

BACKGROUND: Cardiovascular risk in diabetes remains elevated despite glucose-lowering therapies. We hypothesized that hyperglycemia induces trained immunity in macrophages, promoting persistent proatherogenic characteristics. METHODS: Bone marrow-derived macrophages from control mice and mice with diabetes were grown in physiological glucose (5 mmol/L) and subjected to RNA sequencing (n=6), assay for transposase accessible chromatin sequencing (n=6), and chromatin immunoprecipitation sequencing (n=6) for determination of hyperglycemia-induced trained immunity. Bone marrow transplantation from mice with (n=9) or without (n=6) diabetes into (normoglycemic) Ldlr-/- mice was used to assess its functional significance in vivo. Evidence of hyperglycemia-induced trained immunity was sought in human peripheral blood mononuclear cells from patients with diabetes (n=8) compared with control subjects (n=16) and in human atherosclerotic plaque macrophages excised by laser capture microdissection. RESULTS: In macrophages, high extracellular glucose promoted proinflammatory gene expression and proatherogenic functional characteristics through glycolysis-dependent mechanisms. Bone marrow-derived macrophages from diabetic mice retained these characteristics, even when cultured in physiological glucose, indicating hyperglycemia-induced trained immunity. Bone marrow transplantation from diabetic mice into (normoglycemic) Ldlr-/- mice increased aortic root atherosclerosis, confirming a disease-relevant and persistent form of trained innate immunity. Integrated assay for transposase accessible chromatin, chromatin immunoprecipitation, and RNA sequencing analyses of hematopoietic stem cells and bone marrow-derived macrophages revealed a proinflammatory priming effect in diabetes. The pattern of open chromatin implicated transcription factor Runt-related transcription factor 1 (Runx1). Similarly, transcriptomes of atherosclerotic plaque macrophages and peripheral leukocytes in patients with type 2 diabetes were enriched for Runx1 targets, consistent with a potential role in human disease. Pharmacological inhibition of Runx1 in vitro inhibited the trained phenotype. CONCLUSIONS: Hyperglycemia-induced trained immunity may explain why targeting elevated glucose is ineffective in reducing macrovascular risk in diabetes and suggests new targets for disease prevention and therapy.


Subject(s)
Atherosclerosis/immunology , Diabetes Mellitus, Experimental/immunology , Hyperglycemia/immunology , Immunity, Cellular/immunology , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Animals , Atherosclerosis/pathology , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Endarterectomy, Carotid , Humans , Hyperglycemia/pathology , Leukocytes, Mononuclear/pathology , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Transgenic
10.
Rheumatology (Oxford) ; 61(3): 913-925, 2022 03 02.
Article in English | MEDLINE | ID: mdl-34559213

ABSTRACT

Despite extensive research, there is still no treatment that would lead to remission in all patients with rheumatoid arthritis as our understanding of the affected site, the synovium, is still incomplete. Recently, single-cell technologies helped to decipher the cellular heterogeneity of the synovium; however, certain synovial cell populations, such as endothelial cells or peripheral neurons, remain to be profiled on a single-cell level. Furthermore, associations between certain cellular states and inflammation were found; whether these cells cause the inflammation remains to be answered. Similarly, cellular zonation and interactions between individual effectors in the synovium are yet to be fully determined. A deeper understanding of cell signalling and interactions in the synovium is crucial for a better design of therapeutics with the goal of complete remission in all patients.


Subject(s)
Arthritis, Rheumatoid/pathology , Synovial Membrane/cytology , B-Lymphocytes/physiology , Cell Communication/physiology , Endothelial Cells/physiology , Fibroblasts/physiology , Genetic Heterogeneity , Granulocytes/physiology , Humans , Macrophages/physiology , Peripheral Nervous System/cytology , Phagocytes/physiology , Signal Transduction/physiology , Single-Cell Analysis , T-Lymphocytes/physiology , Transcriptome
11.
Proc Natl Acad Sci U S A ; 116(24): 11926-11935, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31147458

ABSTRACT

Caspase-8 is a key integrator of cell survival and cell death decisions during infection and inflammation. Following engagement of tumor necrosis factor superfamily receptors or certain Toll-like receptors (TLRs), caspase-8 initiates cell-extrinsic apoptosis while inhibiting RIPK3-dependent programmed necrosis. In addition, caspase-8 has an important, albeit less well understood, role in cell-intrinsic inflammatory gene expression. Macrophages lacking caspase-8 or the adaptor FADD have defective inflammatory cytokine expression and inflammasome priming in response to bacterial infection or TLR stimulation. How caspase-8 regulates cytokine gene expression, and whether caspase-8-mediated gene regulation has a physiological role during infection, remain poorly defined. Here we demonstrate that both caspase-8 enzymatic activity and scaffolding functions contribute to inflammatory cytokine gene expression. Caspase-8 enzymatic activity was necessary for maximal expression of Il1b and Il12b, but caspase-8 deficient cells exhibited a further decrease in expression of these genes. Furthermore, the ability of TLR stimuli to induce optimal IκB kinase phosphorylation and nuclear translocation of the nuclear factor kappa light chain enhancer of activated B cells family member c-Rel required caspase activity. Interestingly, overexpression of c-Rel was sufficient to restore expression of IL-12 and IL-1ß in caspase-8-deficient cells. Moreover, Ripk3-/-Casp8-/- mice were unable to control infection by the intracellular parasite Toxoplasma gondii, which corresponded to defects in monocyte recruitment to the peritoneal cavity, and exogenous IL-12 restored monocyte recruitment and protection of caspase-8-deficient mice during acute toxoplasmosis. These findings provide insight into how caspase-8 controls inflammatory gene expression and identify a critical role for caspase-8 in host defense against eukaryotic pathogens.


Subject(s)
Caspase 8/metabolism , Cytokines/metabolism , Inflammation/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Toxoplasma/pathogenicity , Toxoplasmosis/metabolism , Animals , Apoptosis/physiology , Cell Line , Inflammasomes/metabolism , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Signal Transduction/physiology
12.
Circulation ; 136(12): 1140-1154, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28698173

ABSTRACT

BACKGROUND: Myeloid cells are central to atherosclerotic lesion development and vulnerable plaque formation. Impaired ability of arterial phagocytes to uptake apoptotic cells (efferocytosis) promotes lesion growth and establishment of a necrotic core. The transcription factor interferon regulatory factor (IRF)-5 is an important modulator of myeloid function and programming. We sought to investigate whether IRF5 affects the formation and phenotype of atherosclerotic lesions. METHODS: We investigated the role of IRF5 in atherosclerosis in 2 complementary models. First, atherosclerotic lesion development in hyperlipidemic apolipoprotein E-deficient (ApoE-/-) mice and ApoE-/- mice with a genetic deletion of IRF5 (ApoE-/-Irf5-/-) was compared and then lesion development was assessed in a model of shear stress-modulated vulnerable plaque formation. RESULTS: Both lesion and necrotic core size were significantly reduced in ApoE-/-Irf5-/- mice compared with IRF5-competent ApoE-/- mice. Necrotic core size was also reduced in the model of shear stress-modulated vulnerable plaque formation. A significant loss of CD11c+ macrophages was evident in ApoE-/-Irf5-/- mice in the aorta, draining lymph nodes, and bone marrow cell cultures, indicating that IRF5 maintains CD11c+ macrophages in atherosclerosis. Moreover, we revealed that the CD11c gene is a direct target of IRF5 in macrophages. In the absence of IRF5, CD11c- macrophages displayed a significant increase in expression of the efferocytosis-regulating integrin-ß3 and its ligand milk fat globule-epidermal growth factor 8 protein and enhanced efferocytosis in vitro and in situ. CONCLUSIONS: IRF5 is detrimental in atherosclerosis by promoting the maintenance of proinflammatory CD11c+ macrophages within lesions and controlling the expansion of the necrotic core by impairing efferocytosis.


Subject(s)
Atherosclerosis/pathology , Interferon Regulatory Factors/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , CD11c Antigen/genetics , CD11c Antigen/metabolism , Cells, Cultured , Immunohistochemistry , Integrin beta3/metabolism , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Lymph Nodes/cytology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Phagocytosis , Shear Strength
13.
Proc Natl Acad Sci U S A ; 112(35): 11001-6, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26283380

ABSTRACT

Whereas the importance of macrophages in chronic inflammatory diseases is well recognized, there is an increasing awareness that neutrophils may also play an important role. In addition to the well-documented heterogeneity of macrophage phenotypes and functions, neutrophils also show remarkable phenotypic diversity among tissues. Understanding the molecular pathways that control this heterogeneity should provide abundant scope for the generation of more specific and effective therapeutics. We have shown that the transcription factor IFN regulatory factor 5 (IRF5) polarizes macrophages toward an inflammatory phenotype. IRF5 is also expressed in other myeloid cells, including neutrophils, where it was linked to neutrophil function. In this study we explored the role of IRF5 in models of acute inflammation, including antigen-induced inflammatory arthritis and lung injury, both involving an extensive influx of neutrophils. Mice lacking IRF5 accumulate far fewer neutrophils at the site of inflammation due to the reduced levels of chemokines important for neutrophil recruitment, such as the chemokine (C-X-C motif) ligand 1. Furthermore we found that neutrophils express little IRF5 in the joints and that their migratory properties are not affected by the IRF5 deficiency. These studies extend prior ones suggesting that inhibiting IRF5 might be useful for chronic macrophage-induced inflammation and suggest that IRF5 blockade would ameliorate more acute forms of inflammation, including lung injury.


Subject(s)
Inflammation/physiopathology , Interferon Regulatory Factors/physiology , Acute Disease , Animals , Chemokines/physiology , Chronic Disease , Inflammation/pathology , Macrophages/pathology , Mice , Synovial Membrane/pathology
14.
J Biol Chem ; 288(16): 11546-54, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23508954

ABSTRACT

NF-κB is a key regulator of immune gene expression in metazoans. It is currently unclear what changes occurred in NF-κB during animal evolution and what features remained conserved. To address this question, we compared the biochemical and functional properties of NF-κB proteins derived from human and the starlet sea anemone (Nematostella vectensis) in 1) a high-throughput assay of in vitro preferences for DNA sequences, 2) ChIP analysis of in vivo recruitment to the promoters of target genes, 3) a LUMIER-assisted examination of interactions with cofactors, and 4) a transactivation assay. We observed a remarkable evolutionary conservation of the DNA binding preferences of the animal NF-κB orthologs. We also show that NF-κB dimerization properties, nuclear localization signals, and binding to cytosolic IκBs are conserved. Surprisingly, the Bcl3-type nuclear IκB proteins functionally pair up only with NF-κB derived from their own species. The basis of the differential NF-κB recognition by IκB subfamilies is discussed.


Subject(s)
Biological Evolution , NF-kappa B/genetics , NF-kappa B/metabolism , Sea Anemones/genetics , Sea Anemones/metabolism , Animals , B-Cell Lymphoma 3 Protein , Humans , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Protein Multimerization/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Species Specificity , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Immunol ; 189(10): 4852-8, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23066157

ABSTRACT

Double-stranded RNA-induced antiviral gene expression in mammalian cells requires activation of IFN regulatory factor 3 (IRF3). In this study, we show that the IL-17R adaptor protein Act1/CIKS is involved in this process. Small interfering RNA-mediated knockdown of Act1 in primary human skin fibroblasts specifically attenuates expression of IFN-ß and IFN-stimulated antiviral genes induced by a synthetic viral mimic, polyinosinic-polycytidylic acid. Ectopic expression of Act1 potentiates the IRF3-driven expression of a synthetic reporter construct as well as the induction of antiviral genes. We demonstrate that this effect is dependent on the ability of Act1 to functionally and physically interact with IκB kinase ε (IKKε), a known IRF3 kinase, and IRF3: 1) Act1 binds IKKε and IRF3; 2) Act1-induced IRF3 activation can be blocked specifically by coexpression of a catalytically inactive mutant of IKKε; and 3) mutants of IRF3, either lacking the C terminus or mutated at the key phosphorylation sites, important for its activation by IKKε, do not support Act1-dependent IRF3 activation. We also show that a zebrafish Act1 protein is able to trigger antiviral gene expression in human cells, which suggests an evolutionarily conserved function of vertebrate Act1 in the host defense against viruses. On the whole, our study demonstrates that Act1 is a component of antiviral signaling.


Subject(s)
Fibroblasts/immunology , RNA Virus Infections/immunology , RNA Viruses/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/immunology , Adaptor Proteins, Signal Transducing , Animals , Cell Line, Tumor , Evolution, Molecular , Fibroblasts/metabolism , Fibroblasts/virology , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Mutation , RNA Virus Infections/genetics , RNA Virus Infections/metabolism , RNA Viruses/genetics , RNA Viruses/metabolism , RNA, Small Interfering , Signal Transduction/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Zebrafish/genetics , Zebrafish/immunology , Zebrafish/virology , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology , Zebrafish Proteins/metabolism
16.
Eur J Immunol ; 42(4): 1030-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22531926

ABSTRACT

Cell survival transcription factor FOXO3 has been recently implicated in moderating pro-inflammatory cytokine production by dendritic cells (DCs), but the molecular mechanisms are unclear. It was suggested that FOXO3 could antagonize NF-κB activity, while IKK-ß was demonstrated to inactivate FOXO3, suggesting a cross-talk between the two pathways. Therefore, FOXO3 activity must be tightly regulated to allow for an appropriate inflammatory response. Here, we show that in human monocyte-derived DCs (MDDCs), FOXO3 is able to antagonize signaling intermediates downstream of the Toll-like receptor (TLR) 4, such as NF-κB and interferon regulatory factors (IRFs), resulting in inhibition of interferon (IFN)-ß expression. We also demonstrate that the activity of FOXO3 itself is regulated by IKK-ε, a kinase involved in IFN-ß production, which phosphorylates and inactivates FOXO3 in response to TLR4 agonists. Thus, we identify FOXO3 as a new IKK-ε-controlled check-point of IRF activation and regulation of IFN-ß expression, providing new insight into the role of FOXO3 in immune response control.


Subject(s)
Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , I-kappa B Kinase/immunology , Interferon-beta/immunology , Monocytes/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/genetics , HEK293 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Interferon-beta/biosynthesis , Interferon-beta/genetics , Monocytes/cytology , Monocytes/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphorylation/genetics , Phosphorylation/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
17.
Am J Pathol ; 181(3): 1099-111, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22809957

ABSTRACT

The molecular mechanisms that drive expression of the alarmin interleukin-33 (IL-33) in endothelial cells are unknown. Because nuclear IL-33 is a marker of endothelial cell quiescence (corroborated in this study by coexpression of cyclin-dependent kinase inhibitor p27(Kip1)), we hypothesized that Notch signaling might be involved in regulating IL-33 expression. Activation of Notch1 by immobilized Notch ligands was sufficient to induce nuclear IL-33 expression in cultured endothelial cells. Conversely, IL-33 expression was inhibited by the γ-secretase inhibitor DAPT or by inhibiting the function of Dll4, Jagged1, Notch1, or the canonical Notch transcription factor RBP-Jκ. Insensitivity to cycloheximide indicated that IL-33 was a direct target of Notch signaling, well in line with the identification of several conserved RBP-Jκ binding sites in the IL33 gene. The in vivo expression of Dll4 but not of Jagged1 was well correlated with expression of IL-33 in quiescent vessels, and subcutaneous injection of DAPT in healthy skin reduced IL-33 expression, indicating that Notch signaling was involved. On the other hand, loss of IL-33 during angiogenesis occurred despite sustained Dll4 and Notch1 expression, suggesting that other signals may override the IL-33-driving signal in this context. Taken together, our data demonstrate that endothelial nuclear IL-33 is induced by Notch and that Dll4 may be the dominant ligand responsible for this signaling in vivo.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Interleukins/metabolism , Receptor, Notch1/metabolism , Adaptor Proteins, Signal Transducing , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Binding Sites , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dipeptides/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Female , Genetic Loci/genetics , Genome, Human/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-33 , Interleukins/genetics , Jagged-1 Protein , Male , Membrane Proteins/metabolism , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Protein Binding/drug effects , Rats , Rats, Wistar , Receptor, Notch1/antagonists & inhibitors , Serrate-Jagged Proteins , Signal Transduction/drug effects , Signal Transduction/genetics , Wound Healing/drug effects
18.
Mediators Inflamm ; 2013: 245804, 2013.
Article in English | MEDLINE | ID: mdl-24453413

ABSTRACT

Macrophages are an integral part of the innate immune system and key players in pathogen clearance and tissue remodelling. Both functions are accomplished by a pivotal network of different macrophage subtypes, including proinflammatory M1 and anti-inflammatory M2 macrophages. Previously, our laboratory identified the transcription factor interferon regulatory factor 5 (IRF5) as the master regulator of the M1 macrophage polarisation. IRF5 was found to be highly expressed in human M1 compared to M2 macrophages. Furthermore, IRF5 dictates the expression of proinflammatory genes such as IL12b and IL23a whilst repressing anti-inflammatory genes like IL10. Here we show that murine bone marrow derived macrophages differentiated in vitro with GM-CSF are also characterised by high levels of IRF5 mRNA and protein and express proinflammatory cytokines upon LPS stimulation. These macrophages display characteristic expression of M1-marker MHC II but lack the M2-marker CD206. Significantly, we develop intracellular staining of IRF5- expressing macrophages and utilise it to recapitulate the in vitro results in an in vivo model of antigen-induced arthritis, emphasising their physiological relevance. Thus, we establish the species-invariant role of IRF5 in controlling the inflammatory macrophage phenotype both in vitro and in in vivo.


Subject(s)
Inflammation/etiology , Interferon Regulatory Factors/physiology , Macrophages/physiology , Animals , Arthritis, Experimental/etiology , Biomarkers , Cytokines/genetics , Disease Models, Animal , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interferon Regulatory Factors/analysis , Interferon Regulatory Factors/genetics , Lectins, C-Type/analysis , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Mannose Receptor , Mannose-Binding Lectins/analysis , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Receptors, Cell Surface/analysis
19.
Methods Mol Biol ; 2594: 69-86, 2023.
Article in English | MEDLINE | ID: mdl-36264489

ABSTRACT

Hoxb8 cells are immortalized myeloid progenitors that maintain their multipotent potential and can be differentiated into neutrophils. Genetic modification of Hoxb8 cells can be used as a model system for the functional analysis of regulators of neutrophil maturation and effector functions, such as transcription factors. Here we describe the generation of transcription factor (TF) knockout Hoxb8 cell lines in vitro with the lentivirus (lenti)CRISPR-Cas 9 technique. After their differentiation into neutrophils, the study of their maturation profile, morphology, and effector functions, including NETosis, phagocytosis, and ROS production, is described.


Subject(s)
Homeodomain Proteins , Neutrophils , Neutrophils/metabolism , Homeodomain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Reactive Oxygen Species/metabolism , Cell Differentiation/genetics
20.
J Leukoc Biol ; 114(6): 585-594, 2023 11 24.
Article in English | MEDLINE | ID: mdl-37480361

ABSTRACT

Neutrophils are innate immune cells that are key to protecting the host against infection and maintaining body homeostasis. However, if dysregulated, they can contribute to disease, such as in cancer or chronic autoinflammatory disorders. Recent studies have highlighted the heterogeneity in the neutrophil compartment and identified the presence of immature neutrophils and their precursors in these pathologies. Therefore, understanding neutrophil maturity and the mechanisms through which they contribute to disease is critical. Neutrophils were first characterized morphologically by Ehrlich in 1879 using microscopy, and since then, different technologies have been used to assess neutrophil maturity. The advances in the imaging field, including state-of-the-art microscopy and machine learning algorithms for image analysis, reinforce the use of neutrophil nuclear morphology as a fundamental marker of maturity, applicable for objective classification in clinical diagnostics. New emerging approaches, such as the capture of changes in chromatin topology, will provide mechanistic links between the nuclear shape, chromatin organization, and transcriptional regulation during neutrophil maturation.


Subject(s)
Chromatin , Neutrophils , Gene Expression Regulation
SELECTION OF CITATIONS
SEARCH DETAIL