Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.146
Filter
Add more filters

Publication year range
1.
Cell ; 176(4): 757-774.e23, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30712866

ABSTRACT

ROCK-Myosin II drives fast rounded-amoeboid migration in cancer cells during metastatic dissemination. Analysis of human melanoma biopsies revealed that amoeboid melanoma cells with high Myosin II activity are predominant in the invasive fronts of primary tumors in proximity to CD206+CD163+ tumor-associated macrophages and vessels. Proteomic analysis shows that ROCK-Myosin II activity in amoeboid cancer cells controls an immunomodulatory secretome, enabling the recruitment of monocytes and their differentiation into tumor-promoting macrophages. Both amoeboid cancer cells and their associated macrophages support an abnormal vasculature, which ultimately facilitates tumor progression. Mechanistically, amoeboid cancer cells perpetuate their behavior via ROCK-Myosin II-driven IL-1α secretion and NF-κB activation. Using an array of tumor models, we show that high Myosin II activity in tumor cells reprograms the innate immune microenvironment to support tumor growth. We describe an unexpected role for Myosin II dynamics in cancer cells controlling myeloid function via secreted factors.


Subject(s)
Cell Movement/physiology , Myosin Type II/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement/immunology , Cytoskeletal Proteins , Female , Humans , Interleukin-1alpha/metabolism , Male , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Middle Aged , NF-kappa B/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Phosphorylation , Proteomics , Receptor Cross-Talk/physiology , Signal Transduction , Tumor Microenvironment/immunology
2.
Nat Immunol ; 21(1): 54-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31819256

ABSTRACT

Ptpn6 is a cytoplasmic phosphatase that functions to prevent autoimmune and interleukin-1 (IL-1) receptor-dependent, caspase-1-independent inflammatory disease. Conditional deletion of Ptpn6 in neutrophils (Ptpn6∆PMN) is sufficient to initiate IL-1 receptor-dependent cutaneous inflammatory disease, but the source of IL-1 and the mechanisms behind IL-1 release remain unclear. Here, we investigate the mechanisms controlling IL-1α/ß release from neutrophils by inhibiting caspase-8-dependent apoptosis and Ripk1-Ripk3-Mlkl-regulated necroptosis. Loss of Ripk1 accelerated disease onset, whereas combined deletion of caspase-8 and either Ripk3 or Mlkl strongly protected Ptpn6∆PMN mice. Ptpn6∆PMN neutrophils displayed increased p38 mitogen-activated protein kinase-dependent Ripk1-independent IL-1 and tumor necrosis factor production, and were prone to cell death. Together, these data emphasize dual functions for Ptpn6 in the negative regulation of p38 mitogen-activated protein kinase activation to control tumor necrosis factor and IL-1α/ß expression, and in maintaining Ripk1 function to prevent caspase-8- and Ripk3-Mlkl-dependent cell death and concomitant IL-1α/ß release.


Subject(s)
Apoptosis/immunology , Caspase 8/immunology , Neutrophils/immunology , Protein Kinases/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Caspase 8/genetics , Cells, Cultured , Gene Deletion , Inflammation/immunology , Interleukin-1/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, Interleukin-1 Type I/immunology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Nat Immunol ; 19(4): 366-374, 2018 04.
Article in English | MEDLINE | ID: mdl-29556001

ABSTRACT

Tumors actively manipulate the immune response through the production of factors that attract immune cells and subsequently alter their ability to recognize and effectively remove the tumor. While this mechanism for evading the immune system is an important aspect of tumor survival, the factors that serve as primary growth factors for the tumor are less understood. Here we demonstrate a previously unknown mechanism by which breast-cancer cells manipulate tumor-infiltrating myeloid cells to maintain their survival. Tumor-derived interleukin 1α (IL-1α), acting on infiltrating myeloid cells, induced the expression of a critical tumor survival factor, the cytokine TSLP. TSLP promoted the survival of the tumor cells through induction of the expression of the anti-apoptotic molecule Bcl-2. TSLP signaling was also required for metastasis to the lungs. These studies define a novel IL-1α-TSLP-mediated crosstalk between tumor-infiltrating myeloid cells and tumor cells in the control of metastatic breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cytokines/immunology , Interleukin-1alpha/immunology , Myeloid Cells/immunology , Tumor Escape/immunology , Animals , Breast Neoplasms/immunology , Disease Progression , Female , Humans , Mice , Thymic Stromal Lymphopoietin
4.
Immunity ; 54(7): 1447-1462.e5, 2021 07 13.
Article in English | MEDLINE | ID: mdl-33979579

ABSTRACT

Two sets of innate immune proteins detect pathogens. Pattern recognition receptors (PRRs) bind microbial products, whereas guard proteins detect virulence factor activities by the surveillance of homeostatic processes within cells. While PRRs are well known for their roles in many types of infections, the role of guard proteins in most infectious contexts remains less understood. Here, we demonstrated that inhibition of protein synthesis during viral infection is sensed as a virulence strategy and initiates pyroptosis in human keratinocytes. We identified the BCL-2 family members MCL-1 and BCL-xL as sensors of translation shutdown. Virus- or chemical-induced translation inhibition resulted in MCL-1 depletion and inactivation of BCL-xL, leading to mitochondrial damage, caspase-3-dependent cleavage of gasdermin E, and release of interleukin-1α (IL-1α). Blocking this pathway enhanced virus replication in an organoid model of human skin. Thus, MCL-1 and BCL-xL can act as guard proteins within barrier epithelia and contribute to antiviral defense.


Subject(s)
Apoptosis/immunology , Epithelial Cells/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , Pyroptosis/immunology , Receptors, Estrogen/immunology , Viruses/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Caspase 3/immunology , Cell Line , Chlorocebus aethiops , HEK293 Cells , Humans , Interleukin-1alpha/immunology , Mice , Mitochondria/immunology , NIH 3T3 Cells , Vero Cells , Virus Replication/immunology , bcl-X Protein/immunology
5.
Nature ; 623(7989): 1044-1052, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37993709

ABSTRACT

All nucleated cells express major histocompatibility complex I and interferon-γ (IFNγ) receptor1, but an epithelial cell-specific function of IFNγ signalling or antigen presentation by means of major histocompatibility complex I has not been explored. We show here that on sensing IFNγ, colonic epithelial cells productively present pathogen and self-derived antigens to cognate intra-epithelial T cells, which are critically located at the epithelial barrier. Antigen presentation by the epithelial cells confers extracellular ATPase expression in cognate intra-epithelial T cells, which limits the accumulation of extracellular adenosine triphosphate and consequent activation of the NLRP3 inflammasome in tissue macrophages. By contrast, antigen presentation by the tissue macrophages alongside inflammasome-associated interleukin-1α and interleukin-1ß production promotes a pathogenic transformation of CD4+ T cells into granulocyte-macrophage colony-stimulating-factor (GM-CSF)-producing T cells in vivo, which promotes colitis and colorectal cancer. Taken together, our study unravels critical checkpoints requiring IFNγ sensing and antigen presentation by epithelial cells that control the development of pathogenic CD4+ T cell responses in vivo.


Subject(s)
Antigen Presentation , Colon , Epithelial Cells , Interferon-gamma , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Colitis/immunology , Colitis/pathology , Colitis/prevention & control , Colon/cytology , Colon/immunology , Colon/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Epithelial Cells/immunology , Epithelial Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Macrophages/immunology , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
6.
Nat Immunol ; 17(8): 906-13, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27434011

ABSTRACT

Inflammation occurs after disruption of tissue homeostasis by cell stress, injury or infection and ultimately involves the recruitment and retention of cells of hematopoietic origin, which arrive at the affected sites to resolve damage and initiate repair. Interleukin 1α (IL-1α) and IL-1ß are equally potent inflammatory cytokines that activate the inflammatory process, and their deregulated signaling causes devastating diseases manifested by severe acute or chronic inflammation. Although much attention has been given to understanding the biogenesis of IL-1ß, the biogenesis of IL-1α and its distinctive role in the inflammatory process remain poorly defined. In this review we examine key aspects of IL-1α biology and regulation and discuss its emerging importance in the initiation and maintenance of inflammation that underlie the pathology of many human diseases.


Subject(s)
Inflammation/physiopathology , Interleukin-1alpha/physiology , Alarmins/metabolism , Animals , Cell Membrane/metabolism , Gene Expression Regulation , Granuloma/etiology , Granuloma/metabolism , Humans , Inflammation/metabolism , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/genetics , Macrophages/physiology , Mice , Mice, Inbred BALB C , Models, Biological , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/physiopathology , Protein Binding , Protein Biosynthesis , Protein Processing, Post-Translational , Receptors, Interleukin-1/physiology , Signal Transduction
7.
Immunity ; 50(4): 1024-1026, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995493

ABSTRACT

Hemostasis and immunity were long considered entirely separate entities. In this issue of Immunity, Burzynski et al. (2019) find that thrombin, the key enzyme within the coagulation cascade, activates IL-1α, a central pleiotropic pro-inflammatory cytokine, to promote wound healing and platelet production following ectoderm injury.


Subject(s)
Interleukin-1alpha , Thrombin , Blood Coagulation , Immune System , Immunity, Innate
8.
Immunity ; 50(4): 1033-1042.e6, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30926232

ABSTRACT

Ancient organisms have a combined coagulation and immune system, and although links between inflammation and hemostasis exist in mammals, they are indirect and slower to act. Here we investigated direct links between mammalian immune and coagulation systems by examining cytokine proproteins for potential thrombin protease consensus sites. We found that interleukin (IL)-1α is directly activated by thrombin. Thrombin cleaved pro-IL-1α at a site perfectly conserved across disparate species, indicating functional importance. Surface pro-IL-1α on macrophages and activated platelets was cleaved and activated by thrombin, while tissue factor, a potent thrombin activator, colocalized with pro-IL-1α in the epidermis. Mice bearing a mutation in the IL-1α thrombin cleavage site (R114Q) exhibited defects in efficient wound healing and rapid thrombopoiesis after acute platelet loss. Thrombin-cleaved IL-1α was detected in humans during sepsis, pointing to the relevance of this pathway for normal physiology and the pathogenesis of inflammatory and thrombotic diseases.


Subject(s)
Blood Coagulation/physiology , Immune System/immunology , Interleukin-1alpha/physiology , Thrombin/physiology , Adaptive Immunity , Amino Acid Sequence , Animals , Blood Platelets/metabolism , Humans , Immunity, Innate , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Keratinocytes/metabolism , Macrophages/metabolism , Mammals/immunology , Mice , Protein Precursors/metabolism , Selection, Genetic , Sepsis/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Thrombopoiesis/immunology , Wound Healing/immunology
9.
Immunity ; 51(6): 983-996.e6, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31836429

ABSTRACT

Excessive activation of the coagulation system leads to life-threatening disseminated intravascular coagulation (DIC). Here, we examined the mechanisms underlying the activation of coagulation by lipopolysaccharide (LPS), the major cell-wall component of Gram-negative bacteria. We found that caspase-11, a cytosolic LPS receptor, activated the coagulation cascade. Caspase-11 enhanced the activation of tissue factor (TF), an initiator of coagulation, through triggering the formation of gasdermin D (GSDMD) pores and subsequent phosphatidylserine exposure, in a manner independent of cell death. GSDMD pores mediated calcium influx, which induced phosphatidylserine exposure through transmembrane protein 16F, a calcium-dependent phospholipid scramblase. Deletion of Casp11, ablation of Gsdmd, or neutralization of phosphatidylserine or TF prevented LPS-induced DIC. In septic patients, plasma concentrations of interleukin (IL)-1α and IL-1ß, biomarkers of GSDMD activation, correlated with phosphatidylserine exposure in peripheral leukocytes and DIC scores. Our findings mechanistically link immune recognition of LPS to coagulation, with implications for the treatment of DIC.


Subject(s)
Caspases, Initiator/metabolism , Disseminated Intravascular Coagulation/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/metabolism , Phosphate-Binding Proteins/metabolism , Phosphatidylserines/metabolism , Thromboplastin/metabolism , Animals , Blood Coagulation/physiology , Caspases, Initiator/genetics , Cell Line, Tumor , Endotoxemia/pathology , Enzyme Activation , HT29 Cells , HeLa Cells , Humans , Interleukin-1alpha/blood , Interleukin-1beta/blood , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphate-Binding Proteins/genetics , Pyroptosis/physiology , Signal Transduction/physiology
10.
Nat Immunol ; 15(11): 1064-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25240383

ABSTRACT

It remains largely unclear how antigen-presenting cells (APCs) encounter effector or memory T cells efficiently in the periphery. Here we used a mouse contact hypersensitivity (CHS) model to show that upon epicutaneous antigen challenge, dendritic cells (DCs) formed clusters with effector T cells in dermal perivascular areas to promote in situ proliferation and activation of skin T cells in a manner dependent on antigen and the integrin LFA-1. We found that DCs accumulated in perivascular areas and that DC clustering was abrogated by depletion of macrophages. Treatment with interleukin 1α (IL-1α) induced production of the chemokine CXCL2 by dermal macrophages, and DC clustering was suppressed by blockade of either the receptor for IL-1 (IL-1R) or the receptor for CXCL2 (CXCR2). Our findings suggest that the dermal leukocyte cluster is an essential structure for elicitating acquired cutaneous immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Skin/immunology , Animals , CD11c Antigen/genetics , Cell Proliferation , Chemokine CXCL2/biosynthesis , Female , Immunologic Memory/immunology , Interleukin-1alpha/pharmacology , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Neutrophils/immunology , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Skin/pathology
11.
Blood ; 143(26): 2791-2803, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38598839

ABSTRACT

ABSTRACT: Thrombotic thrombocytopenic purpura (TTP), a rare but fatal disease if untreated, is due to alteration in von Willebrand factor cleavage resulting in capillary microthrombus formation and ischemic organ damage. Interleukin-1 (IL-1) has been shown to drive sterile inflammation after ischemia and could play an essential contribution to postischemic organ damage in TTP. Our objectives were to evaluate IL-1 involvement during TTP and to test the efficacy of the recombinant IL-1 receptor antagonist, anakinra, in a murine TTP model. We retrospectively measured plasma IL-1 concentrations in patients with TTP and controls. Patients with TTP exhibited elevated plasma IL-1α and -1ß concentrations, which correlated with disease course and survival. In a mouse model of TTP, we administered anakinra (IL-1 inhibitor) or placebo for 5 days and evaluated the efficacy of this treatment. Anakinra significantly reduced mortality of mice (P < .001). Anakinra significantly decreased TTP-induced cardiac damage as assessed by blood troponin concentrations, evaluation of left ventricular function by echocardiography, [18F]fluorodeoxyglucose positron emission tomography of myocardial glucose metabolism, and cardiac histology. Anakinra also significantly reduced brain TTP-induced damage evaluated through blood PS100b concentrations, nuclear imaging, and histology. We finally showed that IL-1α and -1ß trigger endothelial degranulation in vitro, leading to the release of von Willebrand factor. In conclusion, anakinra significantly reduced TTP mortality in a preclinical model of the disease by inhibiting both endothelial degranulation and postischemic inflammation, supporting further evaluations in humans.


Subject(s)
Disease Models, Animal , Interleukin 1 Receptor Antagonist Protein , Purpura, Thrombotic Thrombocytopenic , Animals , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Mice , Humans , Male , Female , Purpura, Thrombotic Thrombocytopenic/drug therapy , Purpura, Thrombotic Thrombocytopenic/pathology , Purpura, Thrombotic Thrombocytopenic/mortality , Interleukin-1beta/metabolism , Interleukin-1beta/blood , Interleukin-1alpha/metabolism , Retrospective Studies , ADAMTS13 Protein/metabolism , Middle Aged , Adult , Mice, Inbred C57BL , von Willebrand Factor/metabolism , von Willebrand Factor/antagonists & inhibitors
12.
Immunity ; 46(4): 635-648, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28410990

ABSTRACT

Mice carrying a hypomorphic point mutation in the Ptpn6 gene (Ptpn6spin mice) develop an inflammatory skin disease that resembles neutrophilic dermatosis in humans. Here, we demonstrated that interleukin-1α (IL-1α) signaling through IL-1R and MyD88 in both stromal and immune cells drive inflammation in Ptpn6spin mice. We further identified SYK as a critical kinase that phosphorylates MyD88, promoted MyD88-dependent signaling and mediates dermatosis in Ptpn6spin mice. Our studies further demonstrated that SHP1 encoded by Ptpn6 binds and suppresses SYK activation to inhibit MyD88 phosphorylation. Downstream of SHP1 and SYK-dependent counterregulation of MyD88 tyrosine phosphorylation, we have demonstrated that the scaffolding function of receptor interacting protein kinase 1 (RIPK1) and tumor growth factor-ß activated kinase 1 (TAK1)-mediating signaling were required to spur inflammatory disease. Overall, these studies identify SHP1 and SYK crosstalk as a critical regulator of MyD88 post-translational modifications and IL-1-driven inflammation.


Subject(s)
Inflammation/immunology , Interleukin-1alpha/immunology , Myeloid Differentiation Factor 88/immunology , Skin Diseases/immunology , Syk Kinase/immunology , Animals , Flow Cytometry , HEK293 Cells , Humans , Immunoblotting , Inflammation/genetics , Inflammation/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Mice, Knockout , Models, Immunological , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Skin Diseases/genetics , Skin Diseases/metabolism , Syk Kinase/genetics , Syk Kinase/metabolism
13.
PLoS Biol ; 21(5): e3002104, 2023 05.
Article in English | MEDLINE | ID: mdl-37141182

ABSTRACT

Tumors protect themselves from immune clearance by promoting extramedullary hematopoiesis. A new study in PLOS Biology provides insights into the mechanisms underlying this process, which may hold the key to disrupting generation of the immunosuppressive tumor microenvironment.


Subject(s)
Hematologic Diseases , Hematopoiesis, Extramedullary , Neoplasms , Humans , Leukemia Inhibitory Factor , Interleukin-1alpha , Hematopoiesis , Tumor Microenvironment
14.
PLoS Biol ; 21(5): e3001746, 2023 05.
Article in English | MEDLINE | ID: mdl-37134077

ABSTRACT

Extramedullary hematopoiesis (EMH) expands hematopoietic capacity outside of the bone marrow in response to inflammatory conditions, including infections and cancer. Because of its inducible nature, EMH offers a unique opportunity to study the interaction between hematopoietic stem and progenitor cells (HSPCs) and their niche. In cancer patients, the spleen frequently serves as an EMH organ and provides myeloid cells that may worsen pathology. Here, we examined the relationship between HSPCs and their splenic niche in EMH in a mouse breast cancer model. We identify tumor produced IL-1α and leukemia inhibitory factor (LIF) acting on splenic HSPCs and splenic niche cells, respectively. IL-1α induced TNFα expression in splenic HSPCs, which then activated splenic niche activity, while LIF induced proliferation of splenic niche cells. IL-1α and LIF display cooperative effects in activating EMH and are both up-regulated in some human cancers. Together, these data expand avenues for developing niche-directed therapies and further exploring EMH accompanying inflammatory pathologies like cancer.


Subject(s)
Hematologic Diseases , Hematopoiesis, Extramedullary , Neoplasms , Humans , Animals , Mice , Hematopoiesis, Extramedullary/physiology , Leukemia Inhibitory Factor/pharmacology , Interleukin-1alpha/pharmacology , Hematopoiesis
15.
Nat Immunol ; 14(10): 1045-53, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995233

ABSTRACT

Chronic inflammation is a fundamental aspect of metabolic disorders such as obesity, diabetes and cardiovascular disease. Cholesterol crystals are metabolic signals that trigger sterile inflammation in atherosclerosis, presumably by activating inflammasomes for IL-1ß production. We found here that atherogenesis was mediated by IL-1α and we identified fatty acids as potent inducers of IL-1α-driven vascular inflammation. Fatty acids selectively stimulated the release of IL-1α but not of IL-1ß by uncoupling mitochondrial respiration. Fatty acid-induced mitochondrial uncoupling abrogated IL-1ß secretion, which deviated the cholesterol crystal-elicited response toward selective production of IL-1α. Our findings delineate a previously unknown pathway for vascular immunopathology that links the cellular response to metabolic stress with innate inflammation, and suggest that IL-1α, not IL-1ß, should be targeted in patients with cardiovascular disease.


Subject(s)
Atherosclerosis/metabolism , Fatty Acids/metabolism , Inflammasomes/metabolism , Interleukin-1alpha/metabolism , Mitochondria/metabolism , Vasculitis/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Calcium Signaling , Dietary Fats/metabolism , Fatty Acids/pharmacology , Female , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Ion Channels/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Oleic Acid/pharmacology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Uncoupling Protein 2 , Vasculitis/pathology
16.
Immunity ; 45(6): 1299-1310, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28002730

ABSTRACT

Particulate pollution is thought to function as an adjuvant that can induce allergic responses. However, the exact cell types and immunological factors that initiate the lung-specific immune responses are unclear. We found that upon intratracheal instillation, particulates such as aluminum salts and silica killed alveolar macrophages (AMs), which then released interleukin-1α (IL-1α) and caused inducible bronchus-associated lymphoid tissue (iBALT) formation in the lung. IL-1α release continued for up to 2 weeks after particulate exposure, and type-2 allergic immune responses were induced by the inhalation of antigen during IL-1α release and iBALT formation, even long after particulate instillation. Recombinant IL-1α was sufficient to induce iBALTs, which coincided with subsequent immunoglobulin E responses, and IL-1-receptor-deficient mice failed to induce iBALT formation. Therefore, the AM-IL-1α-iBALT axis might be a therapeutic target for particulate-induced allergic inflammation.


Subject(s)
Bronchi/immunology , Interleukin-1alpha/immunology , Lymphoid Tissue/immunology , Macrophages, Alveolar/pathology , Particulate Matter/toxicity , Aluminum Compounds/toxicity , Animals , Female , Mice , Mice, Inbred C57BL , Silicon Dioxide/toxicity
17.
EMBO J ; 39(1): e101533, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31701553

ABSTRACT

How cytokine-driven changes in chromatin topology are converted into gene regulatory circuits during inflammation still remains unclear. Here, we show that interleukin (IL)-1α induces acute and widespread changes in chromatin accessibility via the TAK1 kinase and NF-κB at regions that are highly enriched for inflammatory disease-relevant SNPs. Two enhancers in the extended chemokine locus on human chromosome 4 regulate the IL-1α-inducible IL8 and CXCL1-3 genes. Both enhancers engage in dynamic spatial interactions with gene promoters in an IL-1α/TAK1-inducible manner. Microdeletions of p65-binding sites in either of the two enhancers impair NF-κB recruitment, suppress activation and biallelic transcription of the IL8/CXCL2 genes, and reshuffle higher-order chromatin interactions as judged by i4C interactome profiles. Notably, these findings support a dominant role of the IL8 "master" enhancer in the regulation of sustained IL-1α signaling, as well as for IL-8 and IL-6 secretion. CRISPR-guided transactivation of the IL8 locus or cross-TAD regulation by TNFα-responsive enhancers in a different model locus supports the existence of complex enhancer hierarchies in response to cytokine stimulation that prime and orchestrate proinflammatory chromatin responses downstream of NF-κB.


Subject(s)
Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Interleukin-1alpha/pharmacology , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Binding Sites , Cells, Cultured , Chemokines/metabolism , Chromatin/chemistry , Chromatin/genetics , HeLa Cells , Humans , MAP Kinase Kinase Kinases/genetics , NF-kappa B/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
18.
N Engl J Med ; 384(1): 31-41, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33200890

ABSTRACT

BACKGROUND: Interleukin-1 has been implicated as a mediator of recurrent pericarditis. The efficacy and safety of rilonacept, an interleukin-1α and interleukin-1ß cytokine trap, were studied previously in a phase 2 trial involving patients with recurrent pericarditis. METHODS: We conducted a phase 3 multicenter, double-blind, event-driven, randomized-withdrawal trial of rilonacept in patients with acute symptoms of recurrent pericarditis (as assessed on a patient-reported scale) and systemic inflammation (as shown by an elevated C-reactive protein [CRP] level). Patients presenting with pericarditis recurrence while receiving standard therapy were enrolled in a 12-week run-in period, during which rilonacept was initiated and background medications were discontinued. Patients who had a clinical response (i.e., met prespecified response criteria) were randomly assigned in a 1:1 ratio to receive continued rilonacept monotherapy or placebo, administered subcutaneously once weekly. The primary efficacy end point, assessed with a Cox proportional-hazards model, was the time to the first pericarditis recurrence. Safety was also assessed. RESULTS: A total of 86 patients with pericarditis pain and an elevated CRP level were enrolled in the run-in period. During the run-in period, the median time to resolution or near-resolution of pain was 5 days, and the median time to normalization of the CRP level was 7 days. A total of 61 patients underwent randomization. During the randomized-withdrawal period, there were too few recurrence events in the rilonacept group to allow for the median time to the first adjudicated recurrence to be calculated; the median time to the first adjudicated recurrence in the placebo group was 8.6 weeks (95% confidence interval [CI], 4.0 to 11.7; hazard ratio in a Cox proportional-hazards model, 0.04; 95% CI, 0.01 to 0.18; P<0.001 by the log-rank test). During this period, 2 of 30 patients (7%) in the rilonacept group had a pericarditis recurrence, as compared with 23 of 31 patients (74%) in the placebo group. In the run-in period, 4 patients had adverse events leading to the discontinuation of rilonacept therapy. The most common adverse events with rilonacept were injection-site reactions and upper respiratory tract infections. CONCLUSIONS: Among patients with recurrent pericarditis, rilonacept led to rapid resolution of recurrent pericarditis episodes and to a significantly lower risk of pericarditis recurrence than placebo. (Funded by Kiniksa Pharmaceuticals; RHAPSODY ClinicalTrials.gov number, NCT03737110.).


Subject(s)
Pericarditis/drug therapy , Receptors, Interleukin-1 Type I/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Adolescent , Adult , Aged , Double-Blind Method , Female , Humans , Injections, Subcutaneous/adverse effects , Interleukin-1alpha , Interleukin-1beta , Male , Middle Aged , Proportional Hazards Models , Recombinant Fusion Proteins/adverse effects , Recurrence , Respiratory Tract Infections/etiology , Young Adult
19.
Blood ; 139(1): 44-58, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34525198

ABSTRACT

Aging is associated with impaired hematopoietic and immune function caused in part by decreased fitness in the hematopoietic stem cell (HSC) population and an increased myeloid differentiation bias. The reasons for this aging-associated HSC impairment are incompletely understood. Here we demonstrate that older specific pathogen free (SPF) wild-type (WT) mice in contrast to young SPF mice produce more interleukin-1a and interleukin-1b (IL-1a/b) in steady-state bone marrow (BM), with most of the IL-1a/b being derived from myeloid BM cells. Furthermore, blood from steady-state older SPF WT mice contains higher levels of microbe-associated molecular patterns, specifically TLR4 and TLR8 ligands. In addition, BM myeloid cells from older mice produce more IL-1b in vitro, and older mice show higher and more durable IL-1a/b responses upon stimulation with lipopolysaccharide in vivo. To test whether HSC aging is driven by IL-1a/b, we evaluated HSCs from IL-1 receptor 1 (IL-1R1) knockout (KO) mice. Indeed, older HSCs from IL-1R1KO mice show significantly mitigated aging-associated inflammatory signatures. Moreover, HSCs from older IL-1R1KO and from germ-free mice maintain unbiased lymphomyeloid hematopoietic differentiation upon transplantation, thus resembling this functionality of young HSCs. Importantly, in vivo antibiotic suppression of microbiota or pharmacologic blockade of IL-1 signaling in older WT mice was similarly sufficient to reverse myeloid-biased output of their HSC populations. Collectively, our data define the microbiome/IL-1/IL-1R1 axis as a key, self-sustaining and also therapeutically partially reversible driver of HSC inflammaging.


Subject(s)
Hematopoietic Stem Cells/metabolism , Inflammation/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Microbiota , Aging , Animals , Cellular Senescence , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/microbiology , Inflammation/microbiology , Mice , Mice, Knockout
20.
Microb Pathog ; 192: 106671, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38729381

ABSTRACT

This work evaluated aspects of the immune response of BALB/c mice infected with Corynebacterium pseudotuberculosis (T1 and C57). The fifteen BALB/c mice were euthanized after 70 days of infection and morphologically evaluated, also analyzing the innate and adaptive immune responses. The C57 strain induced more pronounced morphological changes than the T1 strain. There was an increase in CD4+ and CD8+ T cells identified during infection with the C57 strain. Cytokines of the inflammatory profile IL-1α and IL-6 and regulatory IL-13 and IL-10 presented significant differences. Cytokines IL-2, IL-4, INF-γ, IL-22, IL-21, and IL-27 did not differ significantly between groups. The obtained results contribute to a better understanding of the type of response and the immunological mechanisms involved during infection with different strains of C. pseudotuberculosis.


Subject(s)
CD8-Positive T-Lymphocytes , Corynebacterium Infections , Corynebacterium pseudotuberculosis , Cytokines , Mice, Inbred BALB C , Animals , Corynebacterium pseudotuberculosis/immunology , Corynebacterium Infections/immunology , Corynebacterium Infections/microbiology , Mice , Cytokines/metabolism , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukin-10 , Adaptive Immunity , Immunity, Innate , Interleukin-6 , Disease Models, Animal , Mice, Inbred C57BL , Interleukin-1alpha/metabolism , Interleukin-1alpha/immunology , Interferon-gamma/metabolism , Interleukin-4/metabolism , Interleukins , Interleukin-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL