Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Res ; 119(1): 236-251, 2023 03 17.
Article in English | MEDLINE | ID: mdl-35134856

ABSTRACT

AIMS: Acute myocardial infarction rapidly increases blood neutrophils (<2 h). Release from bone marrow, in response to chemokine elevation, has been considered their source, but chemokine levels peak up to 24 h after injury, and after neutrophil elevation. This suggests that additional non-chemokine-dependent processes may be involved. Endothelial cell (EC) activation promotes the rapid (<30 min) release of extracellular vesicles (EVs), which have emerged as an important means of cell-cell signalling and are thus a potential mechanism for communicating with remote tissues. METHODS AND RESULTS: Here, we show that injury to the myocardium rapidly mobilizes neutrophils from the spleen to peripheral blood and induces their transcriptional activation prior to arrival at the injured tissue. Time course analysis of plasma-EV composition revealed a rapid and selective increase in EVs bearing VCAM-1. These EVs, which were also enriched for miRNA-126, accumulated preferentially in the spleen where they induced local inflammatory gene and chemokine protein expression, and mobilized splenic-neutrophils to peripheral blood. Using CRISPR/Cas9 genome editing, we generated VCAM-1-deficient EC-EVs and showed that its deletion removed the ability of EC-EVs to provoke the mobilization of neutrophils. Furthermore, inhibition of miRNA-126 in vivo reduced myocardial infarction size in a mouse model. CONCLUSIONS: Our findings show a novel EV-dependent mechanism for the rapid mobilization of neutrophils to peripheral blood from a splenic reserve and establish a proof of concept for functional manipulation of EV-communications through genetic alteration of parent cells.


Subject(s)
Extracellular Vesicles , MicroRNAs , Myocardial Infarction , Mice , Animals , Neutrophils/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Extracellular Vesicles/metabolism , Myocardial Infarction/metabolism , Endothelial Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
2.
Nat Commun ; 12(1): 6702, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795257

ABSTRACT

Interferon regulating factor 5 (IRF5) is a multifunctional regulator of immune responses, and has a key pathogenic function in gut inflammation, but how IRF5 is modulated is still unclear. Having performed a kinase inhibitor library screening in macrophages, here we identify protein-tyrosine kinase 2-beta (PTK2B/PYK2) as a putative IRF5 kinase. PYK2-deficient macrophages display impaired endogenous IRF5 activation, leading to reduction of inflammatory gene expression. Meanwhile, a PYK2 inhibitor, defactinib, has a similar effect on IRF5 activation in vitro, and induces a transcriptomic signature in macrophages similar to that caused by IRF5 deficiency. Finally, defactinib reduces pro-inflammatory cytokines in human colon biopsies from patients with ulcerative colitis, as well as in a mouse colitis model. Our results thus implicate a function of PYK2 in regulating the inflammatory response in the gut via the IRF5 innate sensing pathway, thereby opening opportunities for related therapeutic interventions for inflammatory bowel diseases and other inflammatory conditions.


Subject(s)
Benzamides/pharmacology , Focal Adhesion Kinase 2/metabolism , Inflammation/prevention & control , Interferon Regulatory Factors/metabolism , Pyrazines/pharmacology , Sulfonamides/pharmacology , Animals , Cells, Cultured , Colitis/genetics , Colitis/metabolism , Colitis/prevention & control , Cytokines/genetics , Cytokines/metabolism , Focal Adhesion Kinase 2/genetics , Gene Expression Profiling/methods , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Interferon Regulatory Factors/genetics , Intestines/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Phosphorylation/drug effects , RAW 264.7 Cells
3.
Circulation ; 144(12): 961-982, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34255973

ABSTRACT

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


Subject(s)
Atherosclerosis/immunology , Diabetes Mellitus, Experimental/immunology , Hyperglycemia/immunology , Immunity, Cellular/immunology , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Animals , Atherosclerosis/pathology , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Endarterectomy, Carotid , Humans , Hyperglycemia/pathology , Leukocytes, Mononuclear/pathology , Macrophages/pathology , Mice , Mice, 129 Strain , Mice, Transgenic
4.
Sci Immunol ; 5(47)2020 05 22.
Article in English | MEDLINE | ID: mdl-32444476

ABSTRACT

Mononuclear phagocytes (MNPs) are vital for maintaining intestinal homeostasis but, in response to acute microbial stimulation, can also trigger immunopathology, accelerating recruitment of Ly6Chi monocytes to the gut. The regulators that control monocyte tissue adaptation in the gut remain poorly understood. Interferon regulatory factor 5 (IRF5) is a transcription factor previously shown to play a key role in maintaining the inflammatory phenotype of macrophages. Here, we investigate the impact of IRF5 on the MNP system and physiology of the gut at homeostasis and during inflammation. We demonstrate that IRF5 deficiency has a limited impact on colon physiology at steady state but ameliorates immunopathology during Helicobacter hepaticus-induced colitis. Inhibition of IRF5 activity in MNPs phenocopies global IRF5 deficiency. Using a combination of bone marrow chimera and single-cell RNA-sequencing approaches, we examined the intrinsic role of IRF5 in controlling colonic MNP development. We demonstrate that IRF5 promotes differentiation of Ly6Chi monocytes into CD11c+ macrophages and controls the production of antimicrobial and inflammatory mediators by these cells. Thus, we identify IRF5 as a key transcriptional regulator of the colonic MNP system during intestinal inflammation.


Subject(s)
CD11 Antigens/immunology , Inflammation/immunology , Interferon Regulatory Factors/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Helicobacter hepaticus/immunology , Inflammation/pathology , Interferon Regulatory Factors/deficiency , Macrophages/pathology , Mice , Mice, Knockout , Monocytes/pathology , Phenotype
5.
Methods Mol Biol ; 1745: 113-124, 2018.
Article in English | MEDLINE | ID: mdl-29476466

ABSTRACT

Flow cytometry is extensively used for the immune-profiling of leukocytes in tissue during homeostasis and inflammation. The multiparametric power of using fluorescently conjugated antibodies for specific surface and activation markers provides a comprehensive profile of immune cells. This chapter describes the identification and characterization of myeloid populations using flow cytometric analysis in an acute model of resolving inflammation. This model allows the examination of heterogenic populations across different systemic and tissue locations. We describe tissue processing, antibody staining, and analysis, which include a newly described viSNE tool to generate two-dimensional clustering within myeloid populations. We also reference the use of transgenic reporter mice on specific myeloid cells that provides enhanced specificity and profiling when defining myeloid heterogeneity.


Subject(s)
Flow Cytometry/methods , Immunophenotyping/methods , Myeloid Cells/metabolism , Animals , Biomarkers , Data Analysis , Genes, Reporter , Macrophages/metabolism , Mice , Mice, Transgenic , Monocytes/metabolism , Neutrophils/metabolism , Staining and Labeling
6.
JCI Insight ; 2(17)2017 09 07.
Article in English | MEDLINE | ID: mdl-28878126

ABSTRACT

Transcriptionally activated monocytes are recruited to the heart after acute myocardial infarction (AMI). After AMI in mice and humans, the number of extracellular vesicles (EVs) increased acutely. In humans, EV number correlated closely with the extent of myocardial injury. We hypothesized that EVs mediate splenic monocyte mobilization and program transcription following AMI. Some plasma EVs bear endothelial cell (EC) integrins, and both proinflammatory stimulation of ECs and AMI significantly increased VCAM-1-positive EV release. Injected EC-EVs localized to the spleen and interacted with, and mobilized, splenic monocytes in otherwise naive, healthy animals. Analysis of human plasma EV-associated miRNA showed 12 markedly enriched miRNAs after AMI; functional enrichment analyses identified 1,869 putative mRNA targets, which regulate relevant cellular functions (e.g., proliferation and cell movement). Furthermore, gene ontology termed positive chemotaxis as the most enriched pathway for the miRNA-mRNA targets. Among the identified EV miRNAs, EC-associated miRNA-126-3p and -5p were highly regulated after AMI. miRNA-126-3p and -5p regulate cell adhesion- and chemotaxis-associated genes, including the negative regulator of cell motility, plexin-B2. EC-EV exposure significantly downregulated plexin-B2 mRNA in monocytes and upregulated motility integrin ITGB2. These findings identify EVs as a possible novel signaling pathway by linking ischemic myocardium with monocyte mobilization and transcriptional activation following AMI.


Subject(s)
Extracellular Vesicles/metabolism , Monocytes/metabolism , Myocardial Infarction/pathology , Spleen/pathology , Animals , CD18 Antigens/genetics , Cell Adhesion/genetics , Chemotaxis, Leukocyte/genetics , Down-Regulation , Endothelial Cells/metabolism , Female , Gene Expression , Gene Ontology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Nerve Tissue Proteins/genetics , RAW 264.7 Cells , RNA, Messenger/metabolism , Up-Regulation
7.
Transl Res ; 167(1): 167-82, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26207886

ABSTRACT

Interferon regulatory factor 5 (IRF5) has been demonstrated as a key transcription factor of the immune system, playing important roles in modulating inflammatory immune responses in numerous cell types including dendritic cells, macrophages, and B cells. As well as driving the expression of type I interferon in antiviral responses, IRF5 is also crucial for driving macrophages toward a proinflammatory phenotype by regulating cytokine and chemokine expression and modulating B-cell maturity and antibody production. This review highlights the functional importance of IRF5 in a disease setting, by discussing polymorphic mutations at the human Irf5 locus that lead to susceptibility to systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease. In concordance with this, we also discuss lessons in IRF5 functionality learned from murine in vivo models of autoimmune disease and inflammation and hypothesize that modulation of IRF5 activity and expression could provide potential therapeutic benefits in the clinic.


Subject(s)
Autoimmune Diseases/metabolism , Autoimmunity/physiology , Disease Models, Animal , Interferon Regulatory Factors/physiology , Animals , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...