Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Methods Mol Biol ; 2782: 89-95, 2024.
Article En | MEDLINE | ID: mdl-38622394

Communication among neutrophils plays critical roles during various phases of inflammatory responses, with clinical relevance to both acute and chronic inflammatory diseases. Despite its significance, underlying mechanisms are not well understood, due to the lack of an effective in vitro system to properly address this important question. Here we report a robust in vitro method to culture primary murine neutrophils derived from bone marrow, amenable for well-controlled studies of both neutrophil activation and intercellular communication among co-cultured neutrophils. This protocol can generate primary neutrophils with high purity and survival for an extended culture period, suitable for further phenotypic and functional analyses.


Cell Communication , Neutrophils , Animals , Mice , Coculture Techniques , Bone Marrow
2.
Inflamm Res ; 72(8): 1733-1744, 2023 Aug.
Article En | MEDLINE | ID: mdl-37563334

OBJECTIVE: Proper inflammation resolution is crucial to prevent runaway inflammation during sepsis and reduce sepsis-related mortality/morbidity. Previous studies suggest that deleting TRAM, a key TLR4 signaling adaptor, can reprogram the first inflammatory responder cell-neutrophil from an inflammatory state to a resolving state. In this study, we aim to examine the therapeutic potential of TRAM-deficient neutrophils in vivo with recipient mice undergoing experimental sepsis. MATERIAL AND METHODS: Wild-type or Tram-/- mice were intraperitoneally injected with cecal slurry to induce either severe or mild sepsis. Phenotypic examinations of sepsis and neutrophil characteristics were examined in vivo and ex vivo. The propagations of resolution from donor neutrophils to recipient cells such as monocytes, T cells, and endothelial cells were examined through co-culture assays in vitro. The efficacies of Tram-/- neutrophils in reducing inflammation were studied by transfusing either wild-type or Tram-/- neutrophils into septic recipient mice. RESULTS: Tram-/- septic mice had improved survival and attenuated injuries within the lung and kidney tissues as compared to wild-type septic mice. Wild-type septic mice transfused with Tram-/- resolving neutrophils exhibited reduced multi-organ damages and improved cellular homeostasis. In vitro co-culture studies revealed that donor Tram-/- neutrophils can effectively propagate cellular homeostasis to co-cultured neighboring monocytes, neutrophils, T cells as well as endothelial cells. CONCLUSIONS: Neutrophils with TRAM deletion render effective reprogramming into a resolving state beneficial for ameliorating experimental sepsis, with therapeutic potential in propagating cellular and tissue homeostasis as well as treating sepsis.


Neutrophils , Sepsis , Animals , Mice , Endothelial Cells , Inflammation , Mice, Inbred C57BL , Sepsis/genetics , Sepsis/therapy
3.
Antioxid Redox Signal ; 39(16-18): 1027-1038, 2023 12.
Article En | MEDLINE | ID: mdl-37082952

Significance: Innate immune cells adopt distinct memory states during the pathogenesis of acute and chronic inflammatory diseases. Intracellular generations of reactive oxygen species (ROS) play key roles during the programming dynamics of innate immune cells such as monocytes and macrophages. Recent Advances: ROS modulate the adaptation of innate leukocytes to varying intensities and durations of inflammatory signals, facilitate fundamental reprogramming dynamics such as priming, tolerance, and exhaustion, in addition to fundamental processes of proliferation, differentiation, phagocytosis, chemotaxis, as well as expression of pro- and anti-inflammatory mediators. ROS can be generated at distinct subcellular compartments including cellular membrane, mitochondria, and peroxisome. Complex inflammatory signals may finely regulate ROS generation within distinct subcellular compartments, which in turn may differentially facilitate innate memory dynamics. Critical Issues: Complex inflammatory signals with varying strengths and durations may differentially trigger ROS generation at peroxisome, mitochondria, and other subcellular organelles. Peroxisomal or mitochondrial ROS may facilitate the assembly of distinct signaling platforms involved in the programming of memory innate leukocytes. Despite the emerging connection of subcellular ROS with innate immune memory, underlying mechanisms are still not well defined. Future Directions: Recent important discoveries linking subcellular ROS and innate memory as critically reviewed here hold novel translational relevance related to acute and chronic inflammatory diseases. Capitalizing on these novel findings, future systems studies that use next-generation single-cell dynamic analyses in response to complex inflammatory environments are urgently needed to comprehensively decipher the programming dynamics of innate immune memory, finely modulated by subcellular ROS. Antioxid. Redox Signal. 39, 1027-1038.


Mitochondria , Trained Immunity , Reactive Oxygen Species/metabolism , Mitochondria/metabolism , Organelles/metabolism , Signal Transduction , Immunity, Innate
4.
Front Immunol ; 14: 1297329, 2023.
Article En | MEDLINE | ID: mdl-38162637

Monocyte exhaustion characterized by immune-suppressive features can develop during sepsis and contribute to adverse patient outcomes. However, molecular mechanisms responsible for the establishment of immune-suppressive monocytes with reduced expression of immune-enhancing mediators such as CD86 during sepsis are not well understood. In this study, we identified that the TLR4 intracellular adaptor TRAM plays a key role in mediating the sustained reduction of CD86 expression on exhausted monocytes and generating an immune-suppressive monocyte state. TRAM contributes to the prolonged suppression of CD86 through inducing TAX1BP1 as well as SARM1, collectively inhibiting Akt and NFκB. TRAM deficient mice are protected from cecal slurry-induced experimental sepsis and retain immune-competent monocytes with CD86 expression. Our data reveal a key molecular circuitry responsible for monocyte exhaustion and provide a viable target for rejuvenating functional monocytes and treating sepsis.


B7-2 Antigen , Immune System Exhaustion , Monocytes , Receptors, Interleukin , Sepsis , Animals , Humans , Mice , Armadillo Domain Proteins/metabolism , B7-2 Antigen/metabolism , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , NF-kappa B p50 Subunit/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sepsis/metabolism , Sepsis/pathology , Signal Transduction , Mice, Knockout , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism
5.
Cell Death Dis ; 13(4): 345, 2022 04 13.
Article En | MEDLINE | ID: mdl-35418110

Neutrophils are the dominant leukocytes in circulation and the first responders to infection and inflammatory cues. While the roles of neutrophils in driving inflammation have been widely recognized, the contribution of neutrophils in facilitating inflammation resolution is under-studied. Here, through single-cell RNA sequencing analysis, we identified a subpopulation of neutrophils exhibiting pro-resolving characteristics with greater Cd200r and Cd86 expression at the resting state. We further discovered that 4-PBA, a peroxisomal stress-reducing agent, can potently train neutrophils into the resolving state with enhanced expression of CD200R, CD86, as well as soluble pro-resolving mediators Resolvin D1 and SerpinB1. Resolving neutrophils trained by 4-PBA manifest enhanced phagocytosis and bacterial-killing functions. Mechanistically, the generation of resolving neutrophils is mediated by the PPARγ/LMO4/STAT3 signaling circuit modulated by TLR4 adaptor molecule TRAM. We further demonstrated that genetic deletion of TRAM renders the constitutive expansion of resolving neutrophils, with an enhanced signaling circuitry of PPARγ/LMO4/STAT3. These findings may have profound implications for the effective training of resolving neutrophils with therapeutic potential in the treatment of both acute infection as well as chronic inflammatory diseases.


Neutrophils , Serpins , Adaptor Proteins, Signal Transducing/metabolism , Butylamines , Humans , Inflammation/metabolism , LIM Domain Proteins/metabolism , Neutrophils/metabolism , PPAR gamma/metabolism , Phagocytosis , Serpins/metabolism
6.
Handb Exp Pharmacol ; 276: 43-64, 2022.
Article En | MEDLINE | ID: mdl-34486096

Neutrophils, the most abundant leukocytes in circulation and the first responders to infection and inflammation, closely modulate both acute and chronic inflammatory processes. Resting neutrophils constantly patrol vasculature and migrate to tissues when challenges occur. When infection and/or inflammation recede, tissue neutrophils will be subsequently cleaned up by macrophages which collectively contribute to the resolution of inflammation. While most studies focus on the anti-microbial function of neutrophils including phagocytosis, degranulation, and neutrophil extracellular traps (NETs) formation, recent research highlighted additional contributions of neutrophils beyond simply controlling infectious agents. Neutrophils with resolving characteristics may alter the activities of neighboring cells and facilitate inflammation resolution, modulate long-term macrophage and adaptive immune responses, therefore having important impacts on host pathophysiology. The focus of this chapter is to provide an updated assessment of recent progress in the emerging field of neutrophil programming and memory in the context of both acute and chronic diseases.


Extracellular Traps , Neutrophils , Humans , Immunity, Innate , Inflammation , Macrophages , Neutrophils/pathology , Phagocytosis
7.
J Immunol ; 206(12): 2980-2988, 2021 06 15.
Article En | MEDLINE | ID: mdl-34031144

Low-grade inflammatory monocytes critically contribute to the pathogenesis of chronic inflammatory diseases such as atherosclerosis. The elevated expression of coactivating molecule CD40 as well as key adhesion molecule CD11a is a critical signature of inflammatory monocytes from both human patients with coronary artery diseases as well as in animal models of atherosclerosis. In this study, we report that subclinical superlow-dose LPS, a key risk factor for low-grade inflammation and atherosclerosis, can potently trigger the induction of CD40 and CD11a on low-grade inflammatory monocytes. Subclinical endotoxin-derived monocytes demonstrate immune-enhancing effects and suppress the generation of regulatory CD8+CD122+ T cells, which further exacerbate the inflammatory environment conducive for chronic diseases. Mechanistically, subclinical endotoxemia activates TRAM-mediated signaling processes, leading to the activation of MAPK and STAT5, which is responsible for the expression of CD40 and CD11a. We also demonstrate that TRAM-mediated monocyte polarization can be suppressed by IRAK-M. IRAK-M-deficient monocytes have increased expression of TRAM, elevated induction of CD40 and CD11a by subclinical-dose endotoxin, and are more potent in suppressing the CD8 regulatory T cells. Mice with IRAK-M deficiency generate an increased population of inflammatory monocytes and a reduced population of CD8 T regulatory cells. In contrast, mice with TRAM deficiency exhibit a significantly reduced inflammatory monocyte population and an elevated CD8 T regulatory cell population. Together, our data reveal a competing intracellular circuitry involving TRAM and IRAK-M that modulate the polarization of low-grade inflammatory monocytes with an immune-enhancing function.


Cell Adhesion Molecules/immunology , Inflammation/immunology , Interleukin-1 Receptor-Associated Kinases/immunology , Monocytes/immunology , Receptors, Interleukin/immunology , Toll-Like Receptor 4/immunology , Animals , Cells, Cultured , Interleukin-1 Receptor-Associated Kinases/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
J Leukoc Biol ; 109(5): 861-863, 2021 05.
Article En | MEDLINE | ID: mdl-33527505

Discussion on neutrophil CD11d's potential role in facilitating neutrophil survival and macrophage efferocytosis during sepsis.


Endotoxemia , Sepsis , CD18 Antigens , Defense Mechanisms , Humans , Macrophages , Neutrophils , Phagocytosis
9.
Sci Rep ; 10(1): 14397, 2020 09 01.
Article En | MEDLINE | ID: mdl-32873853

Pathogenic inflammation and immune suppression are the cardinal features that underlie the pathogenesis of severe systemic inflammatory syndrome and sepsis. Neutrophil exhaustion may play a key role during the establishment of pathogenic inflammation and immune suppression through elevated expression of inflammatory adhesion molecules such as ICAM1 and CD11b as well as immune-suppressors such as PD-L1. However, the mechanism of neutrophil exhaustion is not well understood. We demonstrated that murine primary neutrophils cultured in vitro with the prolonged lipopolysaccharides (LPS) stimulation can effectively develop an exhaustive phenotype resembling human septic neutrophils with elevated expression of ICAM1, CD11b, PD-L1 as well as enhanced swarming and aggregation. Mechanistically, we observed that TICAM2 is involved in the generation of neutrophil exhaustion, as TICAM2 deficient neutrophils have the decreased expression of ICAM1, CD11b, PD-L1, and the reduced aggregation following the prolonged LPS challenge as compared to wild type (WT) neutrophils. LPS drives neutrophil exhaustion through TICAM2 mediated activation of Src family kinases (SFK) and STAT1, as the application of SFK inhibitor Dasatinib blocks neutrophil exhaustion triggered by the prolonged LPS challenge. Functionally, TICAM2 deficient mice were protected from developing severe systemic inflammation and multi-organ injury following the chemical-induced mucosal damage. Together, our data defined a key role of TICAM2 in facilitating neutrophil exhaustion and that targeting TICAM2 may be a potential approach to treating the severe systemic inflammation.


CD11b Antigen/metabolism , Cell Aggregation/genetics , Integrin beta1/metabolism , Intercellular Adhesion Molecule-1/metabolism , Neutrophils/immunology , Receptors, Interleukin/metabolism , Sepsis/immunology , Signal Transduction/genetics , Animals , Cell Aggregation/drug effects , Cells, Cultured , Dasatinib/pharmacology , Disease Models, Animal , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Phenotype , Phosphorylation/drug effects , Phosphorylation/genetics , Receptors, Interleukin/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
10.
Immunohorizons ; 4(7): 392-401, 2020 07 06.
Article En | MEDLINE | ID: mdl-32631901

Excessive inflammatory reactions mediated by first-responder cells such as neutrophils contribute to the severity of multiorgan failure associated with systemic injury and infection. Systemic subclinical endotoxemia due to mucosal leakage may aggravate neutrophil activation and tissue injury. However, mechanisms responsible for neutrophil inflammatory polarization are not well understood. In this study, we demonstrate that subclinical low-dose endotoxemia can potently polarize neutrophils into an inflammatory state in vivo and in vitro, as reflected in elevated expression of adhesion molecules such as ICAM-1 and CD29, and reduced expression of suppressor molecule CD244. When subjected to a controlled administration of gut-damaging chemical dextran sulfate sodium, mice conditioned with subclinical dose LPS exhibit significantly elevated infiltration of neutrophils into organs such as liver, colon, and spleen, associated with severe multiorgan damage as measured by biochemical as well as histological assays. Subclinical dose LPS is sufficient to induce potent activation of SRC kinase as well as downstream activation of STAT1/STAT5 in neutrophils, contributing to the inflammatory neutrophil polarization. We also demonstrate that the administration of 4-phenylbutyric acid, an agent known to relieve cell stress and enhance peroxisome function, can reduce the activation of SRC kinase and enhance the expression of suppressor molecule CD244 in neutrophils. We show that i.v. injection of 4-phenylbutyric acid conditioned neutrophils can effectively reduce the severity of multiorgan damage in mice challenged with dextran sulfate sodium. Collectively, our data, to our knowledge, reveal novel inflammatory polarization of neutrophils by subclinical endotoxemia conducive for aggravated multiorgan damage as well as potential therapeutic intervention.


Endotoxemia/immunology , Neutrophils/immunology , STAT1 Transcription Factor/immunology , Animals , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Colon/pathology , Endotoxemia/metabolism , Inflammation/immunology , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/pharmacology , Intercellular Adhesion Molecule-1/therapeutic use , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Liver/pathology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , STAT1 Transcription Factor/metabolism , Spleen/pathology
...