Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.558
Filter
Add more filters

Publication year range
1.
Cell ; 184(24): 5902-5915.e17, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34752731

ABSTRACT

Increasing evidence indicates that the brain regulates peripheral immunity, yet whether and how the brain represents the state of the immune system remains unclear. Here, we show that the brain's insular cortex (InsCtx) stores immune-related information. Using activity-dependent cell labeling in mice (FosTRAP), we captured neuronal ensembles in the InsCtx that were active under two different inflammatory conditions (dextran sulfate sodium [DSS]-induced colitis and zymosan-induced peritonitis). Chemogenetic reactivation of these neuronal ensembles was sufficient to broadly retrieve the inflammatory state under which these neurons were captured. Thus, we show that the brain can store and retrieve specific immune responses, extending the classical concept of immunological memory to neuronal representations of inflammatory information.


Subject(s)
Immunity , Insular Cortex/physiology , Neurons/physiology , Animals , Colitis/chemically induced , Colitis/complications , Colitis/immunology , Colon/pathology , Dextran Sulfate , Female , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Peritoneum/pathology , Peritonitis/complications , Peritonitis/immunology , Peritonitis/pathology , Synapses/metabolism , Zymosan
2.
Nat Immunol ; 21(9): 1119-1133, 2020 09.
Article in English | MEDLINE | ID: mdl-32719519

ABSTRACT

The full neutrophil heterogeneity and differentiation landscape remains incompletely characterized. Here, we profiled >25,000 differentiating and mature mouse neutrophils using single-cell RNA sequencing to provide a comprehensive transcriptional landscape of neutrophil maturation, function and fate decision in their steady state and during bacterial infection. Eight neutrophil populations were defined by distinct molecular signatures. The three mature peripheral blood neutrophil subsets arise from distinct maturing bone marrow neutrophil subsets. Driven by both known and uncharacterized transcription factors, neutrophils gradually acquire microbicidal capability as they traverse the transcriptional landscape, representing an evolved mechanism for fine-tuned regulation of an effective but balanced neutrophil response. Bacterial infection reprograms the genetic architecture of neutrophil populations, alters dynamic transitions between subpopulations and primes neutrophils for augmented functionality without affecting overall heterogeneity. In summary, these data establish a reference model and general framework for studying neutrophil-related disease mechanisms, biomarkers and therapeutic targets at single-cell resolution.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli/physiology , Neutrophils/physiology , Peritonitis/immunology , Single-Cell Analysis/methods , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Female , Gene Expression Profiling , Homeostasis , Humans , Mice , Sequence Analysis, RNA
3.
Nat Immunol ; 20(9): 1138-1149, 2019 09.
Article in English | MEDLINE | ID: mdl-31427775

ABSTRACT

Interleukin (IL)-1R3 is the co-receptor in three signaling pathways that involve six cytokines of the IL-1 family (IL-1α, IL-1ß, IL-33, IL-36α, IL-36ß and IL-36γ). In many diseases, multiple cytokines contribute to disease pathogenesis. For example, in asthma, both IL-33 and IL-1 are of major importance, as are IL-36 and IL-1 in psoriasis. We developed a blocking monoclonal antibody (mAb) to human IL-1R3 (MAB-hR3) and demonstrate here that this antibody specifically inhibits signaling via IL-1, IL-33 and IL-36 in vitro. Also, in three distinct in vivo models of disease (crystal-induced peritonitis, allergic airway inflammation and psoriasis), we found that targeting IL-1R3 with a single mAb to mouse IL-1R3 (MAB-mR3) significantly attenuated heterogeneous cytokine-driven inflammation and disease severity. We conclude that in diseases driven by multiple cytokines, a single antagonistic agent such as a mAb to IL-1R3 is a therapeutic option with considerable translational benefit.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors , Peritonitis/immunology , Pneumonia/immunology , Psoriasis/immunology , A549 Cells , Animals , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Humans , Imiquimod/toxicity , Inflammation/pathology , Interleukin-1/immunology , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1beta/immunology , Interleukin-33/immunology , Male , Mice , Mice, Inbred C57BL , Ovalbumin/toxicity , Peritonitis/drug therapy , Peritonitis/pathology , Pneumonia/drug therapy , Pneumonia/pathology , Psoriasis/drug therapy , Psoriasis/pathology , Signal Transduction/immunology , Uric Acid/toxicity
4.
Immunity ; 52(4): 700-715.e6, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294409

ABSTRACT

The omentum is a visceral adipose tissue rich in fat-associated lymphoid clusters (FALCs) that collects peritoneal contaminants and provides a first layer of immunological defense within the abdomen. Here, we investigated the mechanisms that mediate the capture of peritoneal contaminants during peritonitis. Single-cell RNA sequencing and spatial analysis of omental stromal cells revealed that the surface of FALCs were covered by CXCL1+ mesothelial cells, which we termed FALC cover cells. Blockade of CXCL1 inhibited the recruitment and aggregation of neutrophils at FALCs during zymosan-induced peritonitis. Inhibition of protein arginine deiminase 4, an enzyme important for the release of neutrophil extracellular traps, abolished neutrophil aggregation and the capture of peritoneal contaminants by omental FALCs. Analysis of omental samples from patients with acute appendicitis confirmed neutrophil recruitment and bacterial capture at FALCs. Thus, specialized omental mesothelial cells coordinate the recruitment and aggregation of neutrophils to capture peritoneal contaminants.


Subject(s)
Appendicitis/immunology , Lymphocytes/immunology , Neutrophils/immunology , Omentum/immunology , Peritonitis/immunology , Stromal Cells/immunology , Acute Disease , Animals , Appendicitis/genetics , Appendicitis/microbiology , Cell Communication/immunology , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelium/immunology , Epithelium/microbiology , Escherichia coli/growth & development , Escherichia coli/pathogenicity , Extracellular Traps/immunology , Female , Gene Expression , Humans , Lymphocytes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/microbiology , Omentum/microbiology , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/microbiology , Protein-Arginine Deiminase Type 4/genetics , Protein-Arginine Deiminase Type 4/immunology , Sequence Analysis, RNA , Single-Cell Analysis , Stromal Cells/microbiology , Tissue Culture Techniques , Zymosan/administration & dosage
5.
Immunity ; 49(4): 666-677.e6, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30291029

ABSTRACT

Regulatory T (Treg) cell responses and apoptotic cell clearance (efferocytosis) represent critical arms of the inflammation resolution response. We sought to determine whether these processes might be linked through Treg-cell-mediated enhancement of efferocytosis. In zymosan-induced peritonitis and lipopolysaccharide-induced lung injury, Treg cells increased early in resolution, and Treg cell depletion decreased efferocytosis. In advanced atherosclerosis, where defective efferocytosis drives disease progression, Treg cell expansion improved efferocytosis. Mechanistic studies revealed the following sequence: (1) Treg cells secreted interleukin-13 (IL-13), which stimulated IL-10 production in macrophages; (2) autocrine-paracrine signaling by IL-10 induced Vav1 in macrophages; and (3) Vav1 activated Rac1 to promote apoptotic cell engulfment. In summary, Treg cells promote macrophage efferocytosis during inflammation resolution via a transcellular signaling pathway that enhances apoptotic cell internalization. These findings suggest an expanded role of Treg cells in inflammation resolution and provide a mechanistic basis for Treg-cell-enhancement strategies for non-resolving inflammatory diseases.


Subject(s)
Apoptosis/immunology , Inflammation/immunology , Macrophages/immunology , Phagocytosis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line , Cells, Cultured , Humans , Inflammation/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Jurkat Cells , Lipopolysaccharides , Lung Diseases/chemically induced , Lung Diseases/immunology , Lung Diseases/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/chemically induced , Peritonitis/immunology , Peritonitis/metabolism , T-Lymphocytes, Regulatory/metabolism , Zymosan
6.
Proc Natl Acad Sci U S A ; 121(27): e2402143121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38923993

ABSTRACT

The non-neural cholinergic system plays a critical role in regulating immune equilibrium and tissue homeostasis. While the expression of choline acetyltransferase (ChAT), the enzyme catalyzing acetylcholine biosynthesis, has been well documented in lymphocytes, its role in the myeloid compartment is less understood. Here, we identify a significant population of macrophages (Mϕs) expressing ChAT and synthesizing acetylcholine in the resolution phase of acute peritonitis. Using Chat-GFP reporter mice, we observed marked upregulation of ChAT in monocyte-derived small peritoneal Mϕs (SmPMs) in response to Toll-like receptor agonists and bacterial infections. These SmPMs, phenotypically and transcriptionally distinct from tissue-resident large peritoneal macrophages, up-regulated ChAT expression through a MyD88-dependent pathway involving MAPK signaling. Notably, this process was attenuated by the TRIF-dependent TLR signaling pathway, and our tests with a range of neurotransmitters and cytokines failed to induce a similar response. Functionally, Chat deficiency in Mϕs led to significantly decreased peritoneal acetylcholine levels, reduced efferocytosis of apoptotic neutrophils, and a delayed resolution of peritonitis, which were reversible with exogenous ACh supplementation. Intriguingly, despite B lymphocytes being a notable ChAT-expressing population within the peritoneal cavity, Chat deletion in B cells did not significantly alter the resolution process. Collectively, these findings underscore the crucial role of Mϕ-derived acetylcholine in the resolution of inflammation and highlight the importance of the non-neuronal cholinergic system in immune regulation.


Subject(s)
Acetylcholine , Choline O-Acetyltransferase , Macrophages, Peritoneal , Peritonitis , Animals , Choline O-Acetyltransferase/metabolism , Choline O-Acetyltransferase/genetics , Peritonitis/immunology , Peritonitis/metabolism , Mice , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/immunology , Acetylcholine/metabolism , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Mice, Inbred C57BL , Signal Transduction , Inflammation/metabolism , Inflammation/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Toll-Like Receptors/metabolism , Phagocytosis , Macrophages/metabolism , Macrophages/immunology , Mice, Knockout
7.
Immunity ; 46(1): 92-105, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28065837

ABSTRACT

Uncovering mechanisms that control immune responses in the resolution of bacterial infections is critical for the development of new therapeutic strategies that resolve infectious inflammation without unwanted side effects. We found that disruption of the vagal system in mice delayed resolution of Escherichia coli infection. Dissection of the right vagus decreased peritoneal group 3 innate lymphoid cell (ILC3) numbers and altered peritoneal macrophage responses. Vagotomy resulted in an inflammatory peritoneal lipid mediator profile characterized by reduced concentrations of pro-resolving mediators, including the protective immunoresolvent PCTR1, along with elevated inflammation-initiating eicosanoids. We found that acetylcholine upregulated the PCTR biosynthetic pathway in ILC3s. Administration of PCTR1 or ILC3s to vagotomized mice restored tissue resolution tone and host responses to E. coli infections. Together these findings elucidate a host protective mechanism mediated by ILC3-derived pro-resolving circuit, including PCTR1, that is controlled by local neuronal output to regulate tissue resolution tone and myeloid cell responses.


Subject(s)
Docosahexaenoic Acids/immunology , Inflammation Mediators/immunology , Lymphocytes/immunology , Peritonitis/immunology , Vagus Nerve/immunology , Animals , Cell Separation , Disease Models, Animal , Escherichia coli Infections/immunology , Flow Cytometry , Humans , Male , Mice , Vagotomy
8.
Immunity ; 46(2): 261-272, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228282

ABSTRACT

Variants of the CFH gene, encoding complement factor H (CFH), show strong association with age-related macular degeneration (AMD), a major cause of blindness. Here, we used murine models of AMD to examine the contribution of CFH to disease etiology. Cfh deletion protected the mice from the pathogenic subretinal accumulation of mononuclear phagocytes (MP) that characterize AMD and showed accelerated resolution of inflammation. MP persistence arose secondary to binding of CFH to CD11b, which obstructed the homeostatic elimination of MPs from the subretinal space mediated by thrombospsondin-1 (TSP-1) activation of CD47. The AMD-associated CFH(H402) variant markedly increased this inhibitory effect on microglial cells, supporting a causal link to disease etiology. This mechanism is not restricted to the eye, as similar results were observed in a model of acute sterile peritonitis. Pharmacological activation of CD47 accelerated resolution of both subretinal and peritoneal inflammation, with implications for the treatment of chronic inflammatory disease.


Subject(s)
CD47 Antigen/immunology , Complement Factor H/immunology , Inflammation/immunology , Macular Degeneration/immunology , Animals , Complement Factor H/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Inflammation/genetics , Macular Degeneration/genetics , Mice , Mice, Knockout , Peritonitis/genetics , Peritonitis/immunology , Polymorphism, Single Nucleotide
9.
Immunity ; 44(6): 1365-78, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27261276

ABSTRACT

Receptor CD300b is implicated in regulating the immune response to bacterial infection by an unknown mechanism. Here, we identified CD300b as a lipopolysaccharide (LPS)-binding receptor and determined the mechanism underlying CD300b augmentation of septic shock. In vivo depletion and adoptive transfer studies identified CD300b-expressing macrophages as the key cell type augmenting sepsis. We showed that CD300b, and its adaptor DAP12, associated with Toll-like receptor 4 (TLR4) upon LPS binding, thereby enhancing TLR4-adaptor MyD88- and TRIF-dependent signaling that resulted in an elevated pro-inflammatory cytokine storm. LPS engagement of the CD300b-TLR4 complex led to the recruitment and activation of spleen tyrosine kinase (Syk) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K). This resulted in an inhibition of the ERK1/2 protein kinase- and NF-κB transcription factor-mediated signaling pathways, which subsequently led to a reduced interleukin-10 (IL-10) production. Collectively, our data describe a mechanism of TLR4 signaling regulated by CD300b in myeloid cells in response to LPS.


Subject(s)
Interleukin-10/metabolism , Macrophages/immunology , Peritonitis/immunology , Receptors, Immunologic/metabolism , Sepsis/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , HEK293 Cells , Humans , Interleukin-10/genetics , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Receptors, Immunologic/genetics , Signal Transduction , Syk Kinase/metabolism , Toll-Like Receptor 4/metabolism
10.
EMBO J ; 39(2): e102201, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31762063

ABSTRACT

The innate immune sensor NLRP3 assembles an inflammasome complex with NEK7 and ASC to activate caspase-1 and drive the maturation of proinflammatory cytokines IL-1ß and IL-18. NLRP3 inflammasome activity must be tightly controlled, as its over-activation is involved in the pathogenesis of inflammatory diseases. Here, we show that NLRP3 inflammasome activation is suppressed by a centrosomal protein Spata2. Spata2 deficiency enhances NLRP3 inflammasome activity both in the macrophages and in an animal model of peritonitis. Mechanistically, Spata2 recruits the deubiquitinase CYLD to the centrosome for deubiquitination of polo-like kinase 4 (PLK4), the master regulator of centrosome duplication. Deubiquitination of PLK4 facilitates its binding to and phosphorylation of NEK7 at Ser204. NEK7 phosphorylation in turn attenuates NEK7 and NLRP3 interaction, which is required for NLRP3 inflammasome activation. Pharmacological or shRNA-mediated inhibition of PLK4, or mutation of the NEK7 Ser204 phosphorylation site, augments NEK7 interaction with NLRP3 and causes increased NLRP3 inflammasome activation. Our study unravels a novel centrosomal regulatory pathway of inflammasome activation and may provide new therapeutic targets for the treatment of NLRP3-associated inflammatory diseases.


Subject(s)
Centrosome/immunology , Deubiquitinating Enzyme CYLD/metabolism , Inflammasomes/immunology , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/physiology , Animals , Centrosome/metabolism , Cytokines/metabolism , Deubiquitinating Enzyme CYLD/genetics , Disease Models, Animal , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIMA-Related Kinases/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Peritonitis/immunology , Peritonitis/metabolism , Peritonitis/pathology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Ubiquitination
11.
Mol Med ; 30(1): 153, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294573

ABSTRACT

BACKGROUND: Specialized pro-resolving mediators (SPMs) promote resolution of inflammation, clear infections and stimulate tissue regeneration. These include resolvins, protectins, and maresins. During self-resolving acute inflammation, SPMs are produced and have key functions activating endogenous resolution response for returning to homeostasis. Herein, we addressed whether infections initiated with ongoing inflammation alter resolution programs, and if low-dose repetitive SPM regimen re-programs the resolution response. METHODS: Inflammation was initiated with zymosan (1 mg/mouse) followed by E. coli (105 CFU/mouse) infections carried out in murine peritonitis, and exudates collected at 4-72 h. Leukocytes were enumerated using light microscopy, percentages of PMN, monocytes and macrophages were determined using flow cytometry, and resolution indices calculated. Lipid mediators and SPM profiles were established using mass spectrometry-based metabololipidomics. Repetitive dosing with a SPM panel consisting of RvD1, RvD2, RvD5, MaR1 and RvE2 (0.1 ng/mouse each, i.p.) was given to mice, followed by zymosan challenge. Leukocyte composition, resolution indices and RNA-sequencing were carried out for the repetitive SPM treatments. RESULTS: E. coli infections initiated acute inflammation-resolution programs with temporal SPM production in the infectious exudates. Zymosan-induced inflammation prior to E. coli peritonitis shifted exudate resolution indices and delayed E. coli clearance. Lipid mediator metabololipidomics demonstrated that E. coli infection with ongoing zymosan-induced inflammation shifted the time course of exudate SPMs, activating a SPM cluster that included RvD1, RvD5 and MaR1 during the initiation phase of infectious inflammation (0-4 h); RvD5 and MaR1 were present also in the resolution phase (24-48 h). To emulate daily SPM regimens used in humans, a repetitive subthreshold dosing of the SPM panel RvD1, RvD2, RvD5, MaR1 and RvE2 each at 0.1 ng per mouse was administered. This low-dose SPM regimen accelerated exudate PMN clearance following zymosan-induced inflammation, and shortened the resolution interval by > 70%. These low-dose SPMs regulated genes and pathways related to immune response, chemokine clearance and tissue repair, as demonstrated by using RNA-sequencing. CONCLUSIONS: Infections encountered during ongoing inflammation in mice reset the resolution mechanisms of inflammation via SPM clusters. Low-dose SPMs activate innate immune responses and pathways towards the resolution response that can be reprogrammed.


Subject(s)
Escherichia coli Infections , Inflammation , Peritonitis , Animals , Mice , Peritonitis/immunology , Peritonitis/microbiology , Peritonitis/metabolism , Peritonitis/drug therapy , Inflammation/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Zymosan , Inflammation Mediators/metabolism , Escherichia coli , Male , Docosahexaenoic Acids , Disease Models, Animal , Mice, Inbred C57BL
12.
PLoS Pathog ; 18(1): e1010227, 2022 01.
Article in English | MEDLINE | ID: mdl-35041705

ABSTRACT

The blood-clotting protein fibrin(ogen) plays a critical role in host defense against invading pathogens, particularly against peritoneal infection by the Gram-positive microbe Staphylococcus aureus. Here, we tested the hypothesis that direct binding between fibrin(ogen) and S. aureus is a component of the primary host antimicrobial response mechanism and prevention of secondary microbe dissemination from the peritoneal cavity. To establish a model system, we showed that fibrinogen isolated from FibγΔ5 mice, which express a mutant form lacking the final 5 amino acids of the fibrinogen γ chain (termed fibrinogenγΔ5), did not support S. aureus adherence when immobilized and clumping when in suspension. In contrast, purified wildtype fibrinogen supported robust adhesion and clumping that was largely dependent on S. aureus expression of the receptor clumping factor A (ClfA). Following peritoneal infection with S. aureus USA300, FibγΔ5 mice displayed worse survival compared to WT mice coupled to reduced bacterial killing within the peritoneal cavity and increased dissemination of the microbes into circulation and distant organs. The failure of acute bacterial killing, but not enhanced dissemination, was partially recapitulated by mice infected with S. aureus USA300 lacking ClfA. Fibrin polymer formation and coagulation transglutaminase Factor XIII each contributed to killing of the microbes within the peritoneal cavity, but only elimination of polymer formation enhanced systemic dissemination. Host macrophage depletion or selective elimination of the fibrin(ogen) ß2-integrin binding motif both compromised local bacterial killing and enhanced S. aureus systemic dissemination, suggesting fibrin polymer formation in and of itself was not sufficient to retain S. aureus within the peritoneal cavity. Collectively, these findings suggest that following peritoneal infection, the binding of S. aureus to stabilized fibrin matrices promotes a local, macrophage-mediated antimicrobial response essential for prevention of microbe dissemination and downstream host mortality.


Subject(s)
Fibrinogen/immunology , Peritonitis/immunology , Staphylococcal Infections/immunology , Animals , Coagulase/immunology , Coagulase/metabolism , Fibrin/metabolism , Mice , Peritonitis/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism
13.
Cytokine ; 179: 156619, 2024 07.
Article in English | MEDLINE | ID: mdl-38669908

ABSTRACT

Interleukin (IL)-23 is implicated in the pathogenesis of several inflammatory diseases and is usually linked with helper T cell (Th17) biology. However, there is some data linking IL-23 with innate immune biology in such diseases. We therefore examined the effects of IL-23p19 genetic deletion and/or neutralization on in vitro macrophage activation and in an innate immune-driven peritonitis model. We report that endogenous IL-23 was required for maximal macrophage activation by zymosan as determined by pro-inflammatory cytokine production, including a dramatic upregulation of granulocyte-colony stimulating factor (G-CSF). Furthermore, both IL-23p19 genetic deletion and neutralization in zymosan-induced peritonitis (ZIP) led to a specific reduction in the neutrophil numbers, as well as a reduction in the G-CSF levels in exudate fluids. We conclude that endogenous IL-23 can contribute significantly to macrophage activation during an inflammatory response, mostly likely via an autocrine/paracrine mechanism; of note, endogenous IL-23 can directly up-regulate macrophage G-CSF expression, which in turn is likely to contribute to the regulation of IL-23-dependent neutrophil number and function during an inflammatory response, with potential significance for IL-23 targeting particularly in neutrophil-associated inflammatory diseases.


Subject(s)
Inflammation , Interleukin-23 , Myeloid Cells , Neutrophils , Zymosan , Animals , Inflammation/metabolism , Inflammation/immunology , Interleukin-23/metabolism , Mice , Neutrophils/metabolism , Neutrophils/immunology , Myeloid Cells/metabolism , Peritonitis/metabolism , Peritonitis/immunology , Mice, Inbred C57BL , Granulocyte Colony-Stimulating Factor/metabolism , Macrophage Activation , Macrophages/metabolism , Macrophages/immunology , Interleukin-23 Subunit p19/metabolism , Interleukin-23 Subunit p19/genetics , Mice, Knockout
14.
Immunity ; 43(2): 213-5, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26287676

ABSTRACT

Inhibition of the inflammasome might be beneficial for numerous inflammatory pathologies. In this issue of Immunity, de Almeida et al. (2015) report that the PYRIN domain-only protein (POP1) efficiently inhibits inflammasome activation, identifying it as a pan-inflammasome inhibitor.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/immunology , Dendritic Cells/immunology , Inflammasomes/metabolism , Macrophages, Peritoneal/immunology , Monocytes/immunology , Peritonitis/immunology , Ribonucleoproteins/metabolism , Animals , Female , Humans
15.
Immunity ; 43(2): 264-76, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26275995

ABSTRACT

In response to infections and tissue damage, ASC-containing inflammasome protein complexes are assembled that promote caspase-1 activation, IL-1ß and IL-18 processing and release, pyroptosis, and the release of ASC particles. However, excessive or persistent activation of the inflammasome causes inflammatory diseases. Therefore, a well-balanced inflammasome response is crucial for the maintenance of homeostasis. We show that the PYD-only protein POP1 inhibited ASC-dependent inflammasome assembly by preventing inflammasome nucleation, and consequently interfered with caspase-1 activation, IL-1ß and IL-18 release, pyroptosis, and the release of ASC particles. There is no mouse ortholog for POP1, but transgenic expression of human POP1 in monocytes, macrophages, and dendritic cells protected mice from systemic inflammation triggered by molecular PAMPs, inflammasome component NLRP3 mutation, and ASC danger particles. POP1 expression was regulated by TLR and IL-1R signaling, and we propose that POP1 provides a regulatory feedback loop that shuts down excessive inflammatory responses and thereby prevents systemic inflammation.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/immunology , Dendritic Cells/immunology , Inflammasomes/metabolism , Macrophages, Peritoneal/immunology , Monocytes/immunology , Peritonitis/immunology , Ribonucleoproteins/metabolism , Animals , Apoptosis/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 1/metabolism , Cell Line , Female , Gene Expression Regulation/genetics , Homeostasis , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Peritonitis/chemically induced , Protein Multimerization/genetics , RNA, Small Interfering/genetics , Ribonucleoproteins/genetics
16.
Medicina (Kaunas) ; 60(8)2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39202616

ABSTRACT

Background and Objectives: Spontaneous bacterial peritonitis (SBP) is a life-threatening disease that requires early diagnosis and treatment. It is known that a positive culture result for SBP, which is a common reason for admission to the emergency department, is related to the severity and prognosis of the disease. However, as it is not possible to determine the culture result in the early stage of the disease, different methods are required to predict prognosis in the emergency department. This study was conducted to evaluate the success of the SII, SIRI, NLR and PLR in predicting culture results, intensive care needs and mortality in patients with SBP admitted to the emergency department. Materials and Methods: This study was a retrospective, observational study. Patients with SBP who applied to the emergency department were included in this study. Pregnant women, patients with a malignancy, patients with another infection and patients with liver failure were excluded from this study. Data were analyzed in terms of culture results, the need for intensive care and mortality development. Analyses were performed using SPSS version 26. Results are presented with a 95% confidence interval. A p value less than 0.05 was considered statistically significant. Participant data were analyzed using the independent samples t-test or the Mann-Whitney U test based on normality, and ROC analyses were conducted to assess test accuracies and determine cut-off values. Results: A total of 275 patients were included in this study. Although the culture results of 183 patients were positive, 92 were negative. The SII, NLR and PLR were found to be significantly higher in culture-positive patients (p < 0.001, p = 0.013 and p = 0.002, respectively). The SII and NLR were found to be significantly higher in patients with high mortality (p < 0.001 and p = 0.017, respectively). Conclusions: This study showed that the SII, NLR and PLR may be useful in predicting culture positivity and prognosis in SBP patients in the emergency department.


Subject(s)
Emergency Service, Hospital , Lymphocytes , Neutrophils , Peritonitis , Humans , Female , Retrospective Studies , Male , Peritonitis/microbiology , Peritonitis/blood , Peritonitis/immunology , Middle Aged , Prognosis , Adult , Aged , Blood Platelets , Predictive Value of Tests , Bacterial Infections/blood , Bacterial Infections/diagnosis , Bacterial Infections/mortality , ROC Curve , Inflammation/blood
17.
J Immunol ; 207(7): 1755-1762, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34470853

ABSTRACT

Conformation-specific Ags are ideal targets for mAb-based immunotherapy. Here, we demonstrate that the monomeric form of C-reactive protein (mCRP) is a specific therapeutic target for arthritis and nephritis in a murine model. Screening of >1800 anti-mCRP mAb clones identified 3C as a clone recognizing the monomeric, but not polymeric, form of CRP. The anti-mCRP mAb suppressed leukocyte infiltration in thioglycollate-induced peritonitis, attenuated rheumatoid arthritis symptoms in collagen Ab-induced arthritis model mice, and attenuated lupus nephritis symptoms in MRL/Mp-lpr/lpr lupus-prone model mice. These data suggest that the anti-mCRP mAb 3C has therapeutic potential against rheumatoid arthritis and lupus nephritis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , C-Reactive Protein/immunology , Immunotherapy/methods , Lupus Nephritis/immunology , Peritonitis/immunology , Pleura/metabolism , Animals , Antibodies, Monoclonal/metabolism , Arthritis, Rheumatoid/therapy , Disease Models, Animal , Humans , Lupus Nephritis/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Inbred MRL lpr , Peritonitis/therapy , Protein Binding , Protein Conformation , Protein Isoforms , Thoracentesis
18.
J Immunol ; 206(10): 2441-2452, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33941658

ABSTRACT

Intestinal barrier is essential for dietary products and microbiota compartmentalization and therefore gut homeostasis. When this barrier is broken, cecal content overflows into the peritoneal cavity, leading to local and systemic robust inflammatory response, characterizing peritonitis and sepsis. It has been shown that IL-1ß contributes with inflammatory storm during peritonitis and sepsis and its inhibition has beneficial effects to the host. Therefore, we investigated the mechanisms underlying IL-1ß secretion using a widely adopted murine model of experimental peritonitis. The combined injection of sterile cecal content (SCC) and the gut commensal bacteria Bacteroides fragilis leads to IL-1ß-dependent peritonitis, which was mitigated in mice deficient in NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome components. Typically acting as a damage signal, SCC, but not B. fragilis, activates canonical pathway of NLRP3 promoting IL-1ß secretion in vitro and in vivo. Strikingly, absence of fiber in the SCC drastically reduces IL-1ß production, whereas high-fiber SCC conversely increases this response in an NLRP3-dependent manner. In addition, NLRP3 was also required for IL-1ß production induced by purified dietary fiber in primed macrophages. Extending to the in vivo context, IL-1ß-dependent peritonitis was worsened in mice injected with B. fragilis and high-fiber SCC, whereas zero-fiber SCC ameliorates the pathology. Corroborating with the proinflammatory role of dietary fiber, IL-1R-deficient mice were protected from peritonitis induced by B. fragilis and particulate bran. Overall, our study highlights a function, previously unknown, for dietary fibers in fueling peritonitis through NLRP3 activation and IL-1ß secretion outside the gut.


Subject(s)
Bacteroides Infections/immunology , Bacteroides fragilis/immunology , Dietary Fiber/adverse effects , Inflammasomes/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Peritonitis/immunology , Animals , Bacteroides Infections/microbiology , Diet , Dietary Fiber/administration & dosage , Disease Models, Animal , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Peritonitis/microbiology , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Signal Transduction/drug effects , Signal Transduction/immunology
19.
Zhonghua Gan Zang Bing Za Zhi ; 31(2): 138-146, 2023 Feb 20.
Article in Zh | MEDLINE | ID: mdl-37137828

ABSTRACT

Objective: To observe the level and detection of ascites CD100 on the activity of CD4(+) and CD8(+) T lymphocytes in vitro in the peripheral blood of patients with liver cirrhosis combined with spontaneous bacterial peritonitis. Methods: Peripheral blood and ascites were collected from 77 cases of liver cirrhosis (49 patients with liver cirrhosis combined with simple ascites and 28 patients with liver cirrhosis combined with SBP), and peripheral blood was collected from 22 controls. Soluble CD100 (sCD100) in peripheral blood and ascites was detected by an enzyme-linked immunosorbent assay. Flow cytometry was used to detect membrane-bound CD100 (mCD100) on the surface of CD4(+) and CD8(+)T lymphocytes. CD4(+) and CD8(+)T lymphocytes in ascites were sorted. CD4(+)T lymphocyte proliferation, key transcription factor mRNA, and secreted cytokine changes, as well as CD8(+)T lymphocyte proliferation, important toxic molecule mRNA, and secreted cytokine changes, were detected after CD100 stimulation. The killing activity of CD8(+)T cells was detected by direct contact and indirect contact culture systems. Data conforming to normality were compared using one-way ANOVA, a student's t-test, or a paired t-test. Data that did not conform to a normal distribution were compared using either the Krusal-Willis test or the Mann-Whitney test. Results: There was no statistically significant difference in plasma sCD100 level between patients with liver cirrhosis combined simple ascites (1 415 ± 434.1) pg/ml, patients with liver cirrhosis combined with SBP (1 465 ± 386.8) pg/ml, and controls (1 355 ± 428.0) pg/ml (P = 0.655). The ascites sCD100 level was lower in patients with liver cirrhosis combined with SBP than that of patients with simple ascites [(2 409 ± 743.0) pg/ml vs. (2825±664.2) pg/ml, P=0.014]. There was no statistically significant difference in the level of mCD100 in peripheral blood CD4(+) and CD8(+) T lymphocytes among the three groups (P > 0.05). The levels of mCD100 in ascites CD4(+) and CD8(+) T lymphocytes were higher in patients with liver cirrhosis combined with SBP than those in patients with simple ascites (P < 0.05). CD100 stimulation had no significant effect on the proliferation of CD4(+) and CD8(+)T lymphocytes in the ascites of patients with liver cirrhosis combined with SBP (P > 0.05). There were no significant effects on the expression of transcription factors in effector CD4(+)T lymphocytes (T-bet, retinoic acid associated solitary nuclear receptor γt, aromatic hydrocarbon receptor) or secretion of cytokines (interferon-γ, 17, and 22) (P > 0.05). CD100 stimulation had increased the relative expression of perforin, granzyme B, and granlysin mRNA and the levels of secreted interferon-γ and tumor necrosis factor-α, killing activity in ascites CD8+ T lymphocytes of patients with liver cirrhosis combined with SBP (P < 0.05). Conclusion: The active form of CD100 is sCD100 instead of mCD100. There is an imbalance between the expression of sCD100 and mCD100 in the ascites of patients with cirrhosis combined with SBP. sCD100 can enhance the function of CD8(+)T lymphocytes in the ascites of patients with cirrhosis combined with SBP and thus is one of the potential therapeutic targets.


Subject(s)
Antigens, CD , Ascites , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Liver Cirrhosis , Peritonitis , Ascites/immunology , Immunomodulation/immunology , Antigens, CD/blood , Antigens, CD/immunology , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Cirrhosis/immunology , Peritonitis/blood , Peritonitis/complications , Peritonitis/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans
20.
J Biol Chem ; 296: 100410, 2021.
Article in English | MEDLINE | ID: mdl-33581115

ABSTRACT

Trace element selenium (Se) is incorporated as the 21st amino acid, selenocysteine, into selenoproteins through tRNA[Ser]Sec. Selenoproteins act as gatekeepers of redox homeostasis and modulate immune function to effect anti-inflammation and resolution. However, mechanistic underpinnings involving metabolic reprogramming during inflammation and resolution remain poorly understood. Bacterial endotoxin lipopolysaccharide (LPS) activation of murine bone marrow-derived macrophages cultured in the presence or absence of Se (as selenite) was used to examine temporal changes in the proteome and metabolome by multiplexed tandem mass tag-quantitative proteomics, metabolomics, and machine-learning approaches. Kinetic deltagram and clustering analysis indicated that addition of Se led to extensive reprogramming of cellular metabolism upon stimulation with LPS enhancing the pentose phosphate pathway, tricarboxylic acid cycle, and oxidative phosphorylation, to aid in the phenotypic transition toward alternatively activated macrophages, synonymous with resolution of inflammation. Remodeling of metabolic pathways and consequent metabolic adaptation toward proresolving phenotypes began with Se treatment at 0 h and became most prominent around 8 h after LPS stimulation that included succinate dehydrogenase complex, pyruvate kinase, and sedoheptulokinase. Se-dependent modulation of these pathways predisposed bone marrow-derived macrophages to preferentially increase oxidative phosphorylation to efficiently regulate inflammation and its timely resolution. The use of macrophages lacking selenoproteins indicated that all three metabolic nodes were sensitive to selenoproteome expression. Furthermore, inhibition of succinate dehydrogenase complex with dimethylmalonate affected the proresolving effects of Se by increasing the resolution interval in a murine peritonitis model. In summary, our studies provide novel insights into the role of cellular Se via metabolic reprograming to facilitate anti-inflammation and proresolution.


Subject(s)
Selenium/metabolism , Selenoproteins/metabolism , Animals , Disease Susceptibility/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Lipopolysaccharides/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peritonitis/drug therapy , Peritonitis/immunology , Proteome/metabolism , Proteomics , Selenium/pharmacology , Selenoproteins/genetics , Selenoproteins/physiology , Succinate Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL