Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
J Immunol ; 211(2): 274-286, 2023 07 15.
Article En | MEDLINE | ID: mdl-37272871

Cytokines that signal via STAT1 and STAT3 transcription factors instruct decisions affecting tissue homeostasis, antimicrobial host defense, and inflammation-induced tissue injury. To understand the coordination of these activities, we applied RNA sequencing, chromatin immunoprecipitation sequencing, and assay for transposase-accessible chromatin with high-throughput sequencing to identify the transcriptional output of STAT1 and STAT3 in peritoneal tissues from mice during acute resolving inflammation and inflammation primed to drive fibrosis. Bioinformatics focused on the transcriptional signature of the immunomodulatory cytokine IL-6 in both settings and examined how profibrotic IFN-γ-secreting CD4+ T cells altered the interpretation of STAT1 and STAT3 cytokine cues. In resolving inflammation, STAT1 and STAT3 cooperated to drive stromal gene expression affecting antimicrobial immunity and tissue homeostasis. The introduction of IFN-γ-secreting CD4+ T cells altered this transcriptional program and channeled STAT1 and STAT3 to a previously latent IFN-γ activation site motif in Alu-like elements. STAT1 and STAT3 binding to this conserved sequence revealed evidence of reciprocal cross-regulation and gene signatures relevant to pathophysiology. Thus, we propose that effector T cells retune the transcriptional output of IL-6 by shaping a regulatory interplay between STAT1 and STAT3 in inflammation.


Interleukin-6 , Th1 Cells , Animals , Mice , Cytokines/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Retroelements , STAT Transcription Factors/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Th1 Cells/metabolism
2.
Methods Mol Biol ; 2691: 81-95, 2023.
Article En | MEDLINE | ID: mdl-37355539

Antimicrobial host defense is dependent on the rapid recruitment of inflammatory cells to the site of infection, the elimination of invading pathogens, and the efficient resolution of inflammation that minimizes damage to the host. The peritoneal cavity provides an accessible and physiologically relevant system where the delicate balance of these processes may be studied. Here, we describe murine models of peritoneal inflammation that enable studies of competent antimicrobial immunity and inflammation-associated tissue damage as a consequence of recurrent bacterial challenge. The inflammatory hallmarks of these models reflect the clinical and molecular features of peritonitis seen in renal failure patients on peritoneal dialysis. The development of these models relies on the preparation of a cell-free supernatant derived from an isolate of Staphylococcus epidermidis (termed SES). Intraperitoneal administration of SES induces a Toll-like receptor 2-driven acute inflammatory response that is characterized by an initial transient influx of neutrophils that are replaced by a more sustained recruitment of mononuclear cells and lymphocytes. Adaptation of this model using a repeated administration of SES allows investigations into the development of adaptive immunity and the hallmarks associated with tissue remodelling and fibrosis. These models are therefore clinically relevant and provide exciting opportunities to study innate and adaptive immunity and the response of the stromal tissue compartment to bacterial infection and the ensuing inflammatory reaction.


Peritoneal Dialysis , Peritonitis , Humans , Mice , Animals , Peritoneum , Inflammation , Peritoneal Cavity , Peritoneal Dialysis/adverse effects
3.
Life Sci Alliance ; 6(2)2023 02.
Article En | MEDLINE | ID: mdl-36622345

Neutrophils are vital in defence against pathogens, but excessive neutrophil activity can lead to tissue damage and promote acute respiratory distress syndrome. COVID-19 is associated with systemic expansion of immature neutrophils, but the functional consequences of this shift to immaturity are not understood. We used flow cytometry to investigate activity and phenotypic diversity of circulating neutrophils in acute and convalescent COVID-19 patients. First, we demonstrate hyperactivation of immature CD10- subpopulations in severe disease, with elevated markers of secondary granule release. Partially activated immature neutrophils were detectable 12 wk post-hospitalisation, indicating long term myeloid dysregulation in convalescent COVID-19 patients. Second, we demonstrate that neutrophils from moderately ill patients down-regulate the chemokine receptor CXCR2, whereas neutrophils from severely ill individuals fail to do so, suggesting an altered ability for organ trafficking and a potential mechanism for induction of disease tolerance. CD10- and CXCR2hi neutrophil subpopulations were enriched in severe disease and may represent prognostic biomarkers for the identification of individuals at high risk of progressing to severe COVID-19.


COVID-19 , Neutrophils , Receptors, Interleukin-8B , Humans , COVID-19/immunology , Flow Cytometry , Neutrophils/immunology , Receptors, Interleukin-8B/metabolism
4.
J Leukoc Biol ; 111(6): 1235-1242, 2022 06.
Article En | MEDLINE | ID: mdl-34755385

Peptidylarginine deiminase 4 (PAD4) is a key regulator of inflammation but its function in infections remains incompletely understood. We investigate PAD4 in the context of malaria and demonstrate a role in regulation of immune cell trafficking and chemokine production. PAD4 regulates liver immunopathology by promoting neutrophil trafficking in a Plasmodium chabaudi mouse malaria model. In human macrophages, PAD4 regulates expression of CXCL chemokines in response to stimulation with TLR ligands and P. falciparum. Using patient samples, we show that CXCL1 may be a biomarker for severe malaria. PAD4 inhibition promotes disease tolerance and may represent a therapeutic avenue in malaria.


Malaria , Neutrophils , Animals , Chemotactic Factors , Disease Models, Animal , Humans , Malaria/metabolism , Mice , Protein-Arginine Deiminase Type 4
5.
Sci Signal ; 14(694)2021 08 03.
Article En | MEDLINE | ID: mdl-34344832

Noncanonical inflammasome activation by cytosolic lipopolysaccharide (LPS) is a critical component of the host response to Gram-negative bacteria. Cytosolic LPS recognition in macrophages is preceded by a Toll-like receptor (TLR) priming signal required to induce transcription of inflammasome components and facilitate the metabolic reprograming that fuels the inflammatory response. Using a genome-scale arrayed siRNA screen to find inflammasome regulators in mouse macrophages, we identified the mitochondrial enzyme nucleoside diphosphate kinase D (NDPK-D) as a regulator of both noncanonical and canonical inflammasomes. NDPK-D was required for both mitochondrial DNA synthesis and cardiolipin exposure on the mitochondrial surface in response to inflammasome priming signals mediated by TLRs, and macrophages deficient in NDPK-D had multiple defects in LPS-induced inflammasome activation. In addition, NDPK-D was required for the recruitment of TNF receptor-associated factor 6 (TRAF6) to mitochondria, which was critical for reactive oxygen species (ROS) production and the metabolic reprogramming that supported the TLR-induced gene program. NDPK-D knockout mice were protected from LPS-induced shock, consistent with decreased ROS production and attenuated glycolytic commitment during priming. Our findings suggest that, in response to microbial challenge, NDPK-D-dependent TRAF6 mitochondrial recruitment triggers an energetic fitness checkpoint required to engage and maintain the transcriptional program necessary for inflammasome activation.


Inflammasomes , Nucleoside Diphosphate Kinase D , Animals , Inflammasomes/genetics , Inflammasomes/metabolism , Lipopolysaccharides/metabolism , Macrophages/metabolism , Mice , Mitochondria/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleoside Diphosphate Kinase D/metabolism , Reactive Oxygen Species/metabolism
6.
Nat Commun ; 11(1): 698, 2020 02 04.
Article En | MEDLINE | ID: mdl-32019928

Profound metabolic changes are characteristic of macrophages during classical activation and have been implicated in this phenotype. Here we demonstrate that nitric oxide (NO) produced by murine macrophages is responsible for TCA cycle alterations and citrate accumulation associated with polarization. 13C tracing and mitochondrial respiration experiments map NO-mediated suppression of metabolism to mitochondrial aconitase (ACO2). Moreover, we find that inflammatory macrophages reroute pyruvate away from pyruvate dehydrogenase (PDH) in an NO-dependent and hypoxia-inducible factor 1α (Hif1α)-independent manner, thereby promoting glutamine-based anaplerosis. Ultimately, NO accumulation leads to suppression and loss of mitochondrial electron transport chain (ETC) complexes. Our data reveal that macrophages metabolic rewiring, in vitro and in vivo, is dependent on NO targeting specific pathways, resulting in reduced production of inflammatory mediators. Our findings require modification to current models of macrophage biology and demonstrate that reprogramming of metabolism should be considered a result rather than a mediator of inflammatory polarization.


Aconitate Hydratase/metabolism , Macrophages/enzymology , Nitric Oxide/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Aconitate Hydratase/genetics , Animals , Citric Acid/metabolism , Citric Acid Cycle , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/enzymology , Mitochondria/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvic Acid/metabolism
7.
J Leukoc Biol ; 105(1): 37-48, 2019 01.
Article En | MEDLINE | ID: mdl-30247792

Phagocytes are cells of the immune system that play important roles in phagocytosis, respiratory burst and degranulation-key components of innate immunity and response to infection. This diverse group of cells includes monocytes, macrophages, dendritic cells, neutrophils, eosinophils, and basophils-heterogeneous cell populations possessing cell and tissue-specific functions of which cellular metabolism comprises a critical underpinning. Core functions of phagocytic cells are diverse and sensitive to alterations in environmental- and tissue-specific nutrients and growth factors. As phagocytic cells adapt to these extracellular cues, cellular processes are altered and may contribute to pathogenesis. The considerable degree of functional heterogeneity among monocyte, neutrophil, and other phagocytic cell populations necessitates diverse metabolism. As we review our current understanding of metabolism in phagocytic cells, gaps are focused on to highlight the need for additional studies that hopefully enable improved cell-based strategies for counteracting cancer and other diseases.


Adaptation, Physiological , Phagocytes/metabolism , Animals , Humans , Macrophages/metabolism , Metabolic Networks and Pathways , Neoplasms/metabolism , Neoplasms/pathology , Neutrophils/metabolism
8.
Nat Commun ; 9(1): 5099, 2018 11 30.
Article En | MEDLINE | ID: mdl-30504842

Neutrophils are a vital component of immune protection, yet in cancer they may promote tumour progression, partly by generating reactive oxygen species (ROS) that disrupts lymphocyte functions. Metabolically, neutrophils are often discounted as purely glycolytic. Here we show that immature, c-Kit+ neutrophils subsets can engage in oxidative mitochondrial metabolism. With limited glucose supply, oxidative neutrophils use mitochondrial fatty acid oxidation to support NADPH oxidase-dependent ROS production. In 4T1 tumour-bearing mice, mitochondrial fitness is enhanced in splenic neutrophils and is driven by c-Kit signalling. Concordantly, tumour-elicited oxidative neutrophils are able to maintain ROS production and T cell suppression when glucose utilisation is restricted. Consistent with these findings, peripheral blood neutrophils from patients with cancer also display increased immaturity, mitochondrial content and oxidative phosphorylation. Together, our data suggest that the glucose-restricted tumour microenvironment induces metabolically adapted, oxidative neutrophils to maintain local immune suppression.


Mitochondria/metabolism , Neutrophils/physiology , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cells, Cultured , Flow Cytometry , Immunoblotting , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Confocal , Neutrophils/metabolism , Oxidation-Reduction , Oxidative Phosphorylation , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
9.
Can J Public Health ; 109(4): 451-458, 2018 08.
Article En | MEDLINE | ID: mdl-30276636

OBJECTIVE: For patients who belonged to physician rosters at a family medicine practice in the core of Calgary, Canada, we compared primary care utilization for those who were stably housed and those experiencing homelessness. METHODS: This retrospective chart review accessed electronic medical record data for rostered patients who visited their family physician between July 1, 2015 and August 31, 2016. We assessed the association between homelessness status (defined as having been sheltered in overnight shelters and/or emergency/provisional housing during the study period) and the rate of visits to primary care (defined as the count of visits associated with a patient accounting for the length of the patient's relationship with their family physician) using multivariate negative binomial regression. RESULTS: We analyzed 1013 patients belonging to three family physician rosters, of whom 112 experienced homelessness during the study period (11.1%). The mean number of visits for patients who experienced homelessness was 9.6 (SD 10.5), compared to 4.2 (SD 3.6) visits for stably housed patients (p < 0.0001). The rate of accessing primary care for patients experiencing homelessness was 2.02 times greater than the rate for stably housed individuals (rate ratio [RR] 2.02, 95% confidence interval [95% CI] 1.74-2.35; p < 0.0001). CONCLUSION: In the context of an inner-city primary care clinic in Calgary, Canada, homelessness status is associated with an increased rate of visits to primary care. This work has implications for public health and health systems decision-makers involved in developing equitable health policy, as well as for frontline care providers who serve this vulnerable population.


Ill-Housed Persons/statistics & numerical data , Patient Acceptance of Health Care/statistics & numerical data , Primary Health Care/statistics & numerical data , Adult , Canada , Female , Humans , Male , Middle Aged , Models, Theoretical , Retrospective Studies
10.
Nat Commun ; 8(1): 2074, 2017 12 12.
Article En | MEDLINE | ID: mdl-29234000

The importance of metabolism in macrophage function has been reported, but the in vivo relevance of the in vitro observations is still unclear. Here we show that macrophage metabolites are defined in a specific tissue context, and these metabolites are crucially linked to tissue-resident macrophage functions. We find the peritoneum to be rich in glutamate, a glutaminolysis-fuel that is exploited by peritoneal-resident macrophages to maintain respiratory burst during phagocytosis via enhancing mitochondrial complex-II metabolism. This niche-supported, inducible mitochondrial function is dependent on protein kinase C activity, and is required to fine-tune the cytokine responses that control inflammation. In addition, we find that peritoneal-resident macrophage mitochondria are recruited to phagosomes and produce mitochondrially derived reactive oxygen species, which are necessary for microbial killing. We propose that tissue-resident macrophages are metabolically poised in situ to protect and exploit their tissue-niche by utilising locally available fuels to implement specific metabolic programmes upon microbial sensing.


Macrophages, Peritoneal/metabolism , Peritoneum/immunology , Phagocytosis/immunology , Respiratory Burst/immunology , Stem Cell Niche/immunology , Animals , Electron Transport Complex II/metabolism , Female , Glutamic Acid/metabolism , Inflammation/immunology , Inflammation/microbiology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Peritoneum/cytology , Peritoneum/microbiology , Phagosomes/immunology , Phagosomes/metabolism , Primary Cell Culture , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism
11.
Immunity ; 40(1): 40-50, 2014 Jan 16.
Article En | MEDLINE | ID: mdl-24412616

Fibrosis in response to tissue damage or persistent inflammation is a pathological hallmark of many chronic degenerative diseases. By using a model of acute peritoneal inflammation, we have examined how repeated inflammatory activation promotes fibrotic tissue injury. In this context, fibrosis was strictly dependent on interleukin-6 (IL-6). Repeat inflammation induced IL-6-mediated T helper 1 (Th1) cell effector commitment and the emergence of STAT1 (signal transducer and activator of transcription-1) activity within the peritoneal membrane. Fibrosis was not observed in mice lacking interferon-γ (IFN-γ), STAT1, or RAG-1. Here, IFN-γ and STAT1 signaling disrupted the turnover of extracellular matrix by metalloproteases. Whereas IL-6-deficient mice resisted fibrosis, transfer of polarized Th1 cells or inhibition of MMP activity reversed this outcome. Thus, IL-6 causes compromised tissue repair by shifting acute inflammation into a more chronic profibrotic state through induction of Th1 cell responses as a consequence of recurrent inflammation.


Interleukin-6/metabolism , Peritoneum/pathology , Peritonitis/genetics , Peritonitis/pathology , Th1 Cells/immunology , Acute Disease , Adoptive Transfer , Animals , Cells, Cultured , Chronic Disease , Disease Models, Animal , Extracellular Matrix/immunology , Feedback, Physiological , Fibrosis , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Th1 Cells/transplantation
...