Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
JCI Insight ; 7(1)2022 01 11.
Article En | MEDLINE | ID: mdl-34874920

Nonphlogistic migration of macrophages contributes to the clearance of pathogens and apoptotic cells, a critical step for the resolution of inflammation and return to homeostasis. Angiotensin-(1-7) [Ang-(1-7)] is a heptapeptide of the renin-angiotensin system that acts through Mas receptor (MasR). Ang-(1-7) has recently emerged as a novel proresolving mediator, yet Ang-(1-7) resolution mechanisms are not fully determined. Herein, Ang-(1-7) stimulated migration of human and murine monocytes/macrophages in a MasR-, CCR2-, and MEK/ERK1/2-dependent manner. Pleural injection of Ang-(1-7) promoted nonphlogistic mononuclear cell influx alongside increased levels of CCL2, IL-10, and macrophage polarization toward a regulatory phenotype. Ang-(1-7) induction of CCL2 and mononuclear cell migration was also dependent on MasR and MEK/ERK. Of note, MasR was upregulated during the resolution phase of inflammation, and its pharmacological inhibition or genetic deficiency impaired mononuclear cell recruitment during self-resolving models of LPS pleurisy and E. coli peritonitis. Inhibition/absence of MasR was associated with reduced CCL2 levels, impaired phagocytosis of bacteria, efferocytosis, and delayed resolution of inflammation. In summary, we have uncovered a potentially novel proresolving feature of Ang-(1-7), namely the recruitment of mononuclear cells favoring efferocytosis, phagocytosis, and resolution of inflammation. Mechanistically, cell migration was dependent on MasR, CCR2, and the MEK/ERK pathway.


Angiotensin I , Macrophages , Monocytes , Peptide Fragments , Phagocytosis , Proto-Oncogene Mas/metabolism , Angiotensin I/metabolism , Angiotensin I/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Inflammation/metabolism , MAP Kinase Signaling System/physiology , Macrophages/drug effects , Macrophages/physiology , Male , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Monocytes/physiology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peritonitis , Phagocytosis/drug effects , Phagocytosis/physiology , Phenotype , Receptors, CCR2/metabolism
2.
Pharmacol Res ; 159: 105030, 2020 09.
Article En | MEDLINE | ID: mdl-32562817

A complex intracellular signaling governs different cellular responses in inflammation. Extracellular stimuli are sensed, amplified, and transduced through a dynamic cellular network of messengers converting the first signal into a proper response: production of specific mediators, cell activation, survival, or death. Several overlapping pathways are coordinated to ensure specific and timely induction of inflammation to neutralize potential harms to the tissue. Ideally, the inflammatory response must be controlled and self-limited. Resolution of inflammation is an active process that culminates with termination of inflammation and restoration of tissue homeostasis. Comparably to the onset of inflammation, resolution responses are triggered by coordinated intracellular signaling pathways that transduce the message to the nucleus. However, the key messengers and pathways involved in signaling transduction for resolution are still poorly understood in comparison to the inflammatory network. cAMP has long been recognized as an inducer of anti-inflammatory responses and cAMP-dependent pathways have been extensively exploited pharmacologically to treat inflammatory diseases. Recently, cAMP has been pointed out as coordinator of key steps of resolution of inflammation. Here, we summarize the evidence for the role of cAMP at inducing important features of resolution of inflammation.


Cyclic AMP/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Second Messenger Systems , Animals , Apoptosis , Chemotaxis, Leukocyte , Granulocytes/immunology , Granulocytes/metabolism , Granulocytes/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Phagocytosis , Phenotype
3.
Pharmacol Res ; 158: 104842, 2020 08.
Article En | MEDLINE | ID: mdl-32413484

Macrophages are professional phagocytes that display remarkable plasticity, with a range of phenotypes that can be broadly characterized by the M1/M2 dichotomy. Glucocorticoid (GC)-induced leucine zipper (GILZ) is a protein known to mediate anti-inflammatory and some pro-resolving actions, including as neutrophil apoptosis. However, the role of GILZ in key macrophage function is not well understood. Here, we investigated the role of GILZ on macrophage reprogramming and efferocytosis. Using murine bone-marrow-derived macrophages (BMDMs), we found that GILZ was expressed in naive BMDMs and exhibited increased expression in M2-like macrophages (IL4-differentiated). M1-like macrophages (IFN/LPS-differentiated) from GILZ-/- mice showed higher expression of the M1 markers CD86, MHC class II, iNOS, IL-6 and TNF-α, associated with increased levels of phosphorylated STAT1 and lower IL-10 levels, compared to M1-differentiated cells from WT mice. There were no changes in the M2 markers CD206 and arginase-1 in macrophages from GILZ-/- mice differentiated with IL-4, compared to cells from WT animals. Treatment of M1-like macrophages with TAT-GILZ, a cell-permeable GILZ fusion protein, decreased the levels of CD86 and MHC class II in M1-like macrophages without modifying CD206 levels in M2-like macrophages. In line with the in vitro data, increased numbers of M1-like macrophages were found into the pleural cavity of GILZ-/- mice after LPS-injection, compared to WT mice. Moreover, efferocytosis was defective in the context of GILZ deficiency, both in vitro and in vivo. Conversely, treatment of LPS-injected mice with TAT-GILZ promoted inflammation resolution, associated with lower numbers of M1-like macrophages and increased efferocytosis. Collectively, these data indicate that GILZ is a regulator of important macrophage functions, contributing to macrophage reprogramming and efferocytosis, both key steps for the resolution of inflammation.


Apoptosis/drug effects , Glucocorticoids/pharmacology , Transcription Factors/drug effects , Animals , Bone Marrow Cells/drug effects , Cell Migration Assays, Leukocyte , Cell Physiological Phenomena/drug effects , Gene Expression Regulation/drug effects , Inflammation/chemically induced , Inflammation/pathology , Leukocyte Count , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Pleural Cavity/cytology
4.
Cells ; 9(1)2020 01 06.
Article En | MEDLINE | ID: mdl-31935860

Macrophages are central to inflammation resolution, an active process aimed at restoring tissue homeostasis following an inflammatory response. Here, the effects of db-cAMP on macrophage phenotype and function were investigated. Injection of db-cAMP into the pleural cavity of mice induced monocytes recruitment in a manner dependent on PKA and CCR2/CCL2 pathways. Furthermore, db-cAMP promoted reprogramming of bone-marrow-derived macrophages to a M2 phenotype as seen by increased Arg-1/CD206/Ym-1 expression and IL-10 levels (M2 markers). Db-cAMP also showed a synergistic effect with IL-4 in inducing STAT-3 phosphorylation and Arg-1 expression. Importantly, db-cAMP prevented IFN-γ/LPS-induced macrophage polarization to M1-like as shown by increased Arg-1 associated to lower levels of M1 cytokines (TNF-α/IL-6) and p-STAT1. In vivo, db-cAMP reduced the number of M1 macrophages induced by LPS injection without changes in M2 and Mres numbers. Moreover, db-cAMP enhanced efferocytosis of apoptotic neutrophils in a PKA-dependent manner and increased the expression of Annexin A1 and CD36, two molecules associated with efferocytosis. Finally, inhibition of endogenous PKA during LPS-induced pleurisy impaired the physiological resolution of inflammation. Taken together, the results suggest that cAMP is involved in the major functions of macrophages, such as nonphlogistic recruitment, reprogramming and efferocytosis, all key processes for inflammation resolution.


Cellular Reprogramming , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Macrophages/metabolism , Phagocytosis , Animals , Annexin A1/metabolism , Apoptosis/drug effects , Arginase/metabolism , Bucladesine/pharmacology , CD36 Antigens/metabolism , Cell Polarity/drug effects , Cellular Reprogramming/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Inflammation/pathology , Interleukin-4/metabolism , Isoquinolines/pharmacology , Lipopolysaccharides , Macrophages/drug effects , Male , Mice, Inbred BALB C , Models, Biological , Monocytes/drug effects , Monocytes/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Phagocytosis/drug effects , Phenotype , Phosphorylation/drug effects , Pleural Cavity/metabolism , Receptors, CCR2/metabolism , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology , Time Factors
5.
Front Immunol ; 10: 1458, 2019.
Article En | MEDLINE | ID: mdl-31316511

Inflammation resolution is an active process that functions to restore tissue homeostasis. Clearance of apoptotic leukocytes by efferocytosis at inflammatory sites plays an important role in inflammation resolution and induces remarkable macrophage phenotypic and functional changes. Here, we investigated the effects of deletion of either plasminogen (Plg) or the Plg receptor, Plg-RKT, on the resolution of inflammation. In a murine model of pleurisy, the numbers of total mononuclear cells recruited to the pleural cavity were significantly decreased in both Plg-/- and Plg-RKT-/- mice, a response associated with decreased levels of the chemokine CCL2 in pleural exudates. Increased percentages of M1-like macrophages were determined in pleural lavages of Plg-/- and Plg-RKT-/- mice without significant changes in M2-like macrophage percentages. In vitro, Plg and plasmin (Pla) increased CD206/Arginase-1 expression and the levels of IL-10/TGF-ß (M2 markers) while decreasing IFN/LPS-induced M1 markers in murine bone-marrow-derived macrophages (BMDMs) and human macrophages. Furthermore, IL4-induced M2-like polarization was defective in BMDMs from both Plg-/- and Plg-RKT-/- mice. Mechanistically, Plg and Pla induced transient STAT3 phosphorylation, which was decreased in Plg-/- and Plg-RKT-/- BMDMs after IL-4 or IL-10 stimulation. The extents of expression of CD206 and Annexin A1 (important for clearance of apoptotic cells) were reduced in Plg-/- and Plg-RKT-/- macrophage populations, which exhibited decreased phagocytosis of apoptotic neutrophils (efferocytosis) in vivo and in vitro. Taken together, these results suggest that Plg and its receptor, Plg-RKT, regulate macrophage polarization and efferocytosis, as key contributors to the resolution of inflammation.


Macrophages/immunology , Plasminogen/immunology , Pleurisy/immunology , Receptors, Cell Surface/immunology , Animals , Cell Movement , Humans , Male , Mice, Transgenic , Neutrophils/immunology , Phagocytosis , Phenotype , Plasminogen/genetics , Receptors, Cell Surface/genetics
...