Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Proc Natl Acad Sci U S A ; 119(31): e2201146119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35878041

ABSTRACT

Aberrant immune responses, including hyperresponsiveness to Toll-like receptor (TLR) ligands, underlie acute respiratory distress syndrome (ARDS). Type I interferons confer antiviral activities and could also regulate the inflammatory response, whereas little is known about their actions to resolve aberrant inflammation. Here we report that interferon-ß (IFN-ß) exerts partially overlapping, but also cooperative actions with aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 to counter TLR9-generated cues to regulate neutrophil apoptosis and phagocytosis in human neutrophils. In mice, TLR9 activation impairs bacterial clearance, prolongs Escherichia coli-evoked lung injury, and suppresses production of IFN-ß and the proresolving lipid mediators 15-epi-LXA4 and resolvin D1 (RvD1) in the lung. Neutralization of endogenous IFN-ß delays pulmonary clearance of E. coli and aggravates mucosal injury. Conversely, treatment of mice with IFN-ß accelerates clearance of bacteria, restores neutrophil phagocytosis, promotes neutrophil apoptosis and efferocytosis, and accelerates resolution of airway inflammation with concomitant increases in 15-epi-LXA4 and RvD1 production in the lungs. Pharmacological blockade of the lipoxin receptor ALX/FPR2 partially prevents IFN-ß-mediated resolution. These findings point to a pivotal role of IFN-ß in orchestrating timely resolution of neutrophil and TLR9 activation-driven airway inflammation and uncover an IFN-ß-initiated resolution program, activation of an ALX/FPR2-centered, proresolving lipids-mediated circuit, for ARDS.


Subject(s)
Interferon-beta , Lipoxins , Respiratory Distress Syndrome , Animals , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Escherichia coli , Escherichia coli Infections/immunology , Humans , Inflammation/drug therapy , Interferon-beta/immunology , Interferon-beta/pharmacology , Lipoxins/pharmacology , Mice , Receptors, Formyl Peptide/antagonists & inhibitors , Respiratory Distress Syndrome/drug therapy , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Transcriptional Activation/drug effects
2.
Proc Natl Acad Sci U S A ; 117(14): 7971-7980, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32205444

ABSTRACT

Timely resolution of bacterial infections critically depends on phagocytosis of invading pathogens by polymorphonuclear neutrophil granulocytes (PMNs), followed by PMN apoptosis and efferocytosis. Here we report that bacterial DNA (CpG DNA) and mitochondrial DNA impair phagocytosis and attenuate phagocytosis-induced apoptosis in human PMNs through Toll-like receptor 9 (TLR9)-mediated release of neutrophil elastase and proteinase 3 and subsequent down-regulation of the complement receptor C5aR. Consistently, CpG DNA delays pulmonary clearance of Escherichia coli in mice and suppresses PMN apoptosis, efferocytosis, and generation of proresolving lipid mediators, thereby prolonging lung inflammation evoked by E. coli Genetic deletion of TLR9 renders mice unresponsive to CpG DNA. We also show that aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 (17-epi-RvD1) through the receptor ALX/FPR2 antagonize cues from CpG DNA, preserve C5aR expression, restore impaired phagocytosis, and redirect human PMNs to apoptosis. Treatment of mice with 15-epi-LXA4 or 17-epi-RvD1 at the peak of inflammation accelerates clearance of bacteria, blunts PMN accumulation, and promotes PMN apoptosis and efferocytosis, thereby facilitating resolution of E. coli-evoked lung injury. Collectively, these results uncover a TLR9-mediated endogenous mechanism that impairs PMN phagocytosis and prolongs inflammation, and demonstrate both endogenous and therapeutic potential for 15-epi-LXA4 and 17-epi-RvD1 to restore impaired bacterial clearance and facilitate resolution of acute lung inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Escherichia coli Infections/immunology , Neutrophils/immunology , Phagocytosis/immunology , Pneumonia/immunology , Toll-Like Receptor 9/metabolism , Adult , Aged , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Cells, Cultured , CpG Islands/immunology , DNA, Bacterial/immunology , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Healthy Volunteers , Humans , Lipoxins/pharmacology , Lipoxins/therapeutic use , Lung/microbiology , Lung/pathology , Male , Mice , Middle Aged , Neutrophils/metabolism , Phagocytosis/drug effects , Pneumonia/drug therapy , Pneumonia/microbiology , Pneumonia/pathology , Primary Cell Culture , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/immunology , Receptors, Lipoxin/metabolism
3.
Biol Chem ; 396(11): 1181-97, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26040008

ABSTRACT

The prototypic acute-phase reactant C-reactive protein (CRP) has long been recognized as a useful marker and gauge of inflammation. CRP also plays an important role in host defense against invading pathogens as well as in inflammation. CRP consists of five identical subunits arranged as a cyclic pentamer. CRP exists in at least two conformationally distinct forms, i.e. native pentameric CRP (pCRP) and modified/monomeric CRP (mCRP). These isoforms bind to distinct receptors and lipid rafts, and exhibit distinct functional properties. Dissociation of pCRP into its subunits occurs within the inflammatory microenvironment and newly formed mCRP may then contribute to localizing the inflammatory response. Accumulating evidence indicates that pCRP possesses both pro- and anti-inflammatory actions in a context-dependent manner, whereas mCRP exerts potent pro-inflammatory actions on endothelial cells, endothelial progenitor cells, leukocytes and platelets, and thus may amplify inflammation. Here, we review recent advances that may explain how conformational changes in CRP contribute to shaping the inflammatory response and discuss CRP isomers as potential therapeutic targets to dampen inflammation.


Subject(s)
C-Reactive Protein/chemistry , C-Reactive Protein/metabolism , Inflammation/metabolism , Animals , Humans , Protein Conformation
4.
Crit Care Med ; 43(6): e179-89, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25855902

ABSTRACT

OBJECTIVE: Bacterial DNA (CpG DNA) persists in tissues and blood under pathological conditions that are associated with enhanced intravascular coagulation. Toll-like receptor 9 recognizes CpG DNA and elicits innate and adoptive immunity, yet the impact of CpG DNA on coagulation has not been studied. In this study, we investigated the effects of CpG DNA on the expression and activity of tissue factor, a key initiator of coagulation and tissue factor pathway inhibitor in human coronary artery endothelial cells and on coagulation in mice. DESIGN: Controlled in vitro and in vivo studies. SETTING: University research laboratory. SUBJECTS: Cultured human coronary artery endothelial cell, wild-type mice, and TLR9-deficient mice. INTERVENTIONS: Human coronary artery endothelial cell was challenged with CpG DNA, and tissue factor and tissue factor pathway inhibitor expression and activity were assessed. In mice, the effects of CpG DNA on bleeding time and plasma levels of thrombin-antithrombin complexes and tissue factor were measured. MEASUREMENTS AND MAIN RESULTS: We found that CpG DNA, but not eukaryotic DNA, evoked marked nuclear factor-κB-mediated increases in tissue factor expression at both messenger RNA and protein levels, as well as in tissue factor activity. Conversely, CpG DNA significantly reduced tissue factor pathway inhibitor transcription, secretion, and activity. Inhibition of Toll-like receptor 9 with a telomere-derived Toll-like receptor 9 inhibitory oligonucleotide or transient Toll-like receptor 9 knockdown with small interfering RNA attenuated human coronary artery endothelial cell responses to CpG DNA. In wild-type mice, CpG DNA shortened the bleeding time parallel with dramatic increases in plasma thrombin-antithrombin complex and tissue factor levels. Pretreatment with inhibitory oligonucleotide or anti-tissue factor antibody or genetic deletion of TLR9 prevented these changes, whereas depleting monocytes with clodronate resulted in a modest partial inhibition. CONCLUSIONS: Our findings demonstrate that bacterial DNA through Toll-like receptor 9 shifted the balance of tissue factor and tissue factor pathway inhibitor toward procoagulant phenotype in human coronary artery endothelial cells and activated blood coagulation in mice. Our study identifies Toll-like receptor 9 inhibitory oligonucleotides as potential therapeutic agents for the prevention of coagulation in pathologies where bacterial DNA may abundantly be present.


Subject(s)
Blood Coagulation/physiology , DNA, Bacterial/metabolism , Endothelial Cells/drug effects , Toll-Like Receptor 9/metabolism , Animals , Coronary Vessels/metabolism , Gene Expression , Humans , Lipoproteins , Mice , NF-kappa B/metabolism , RNA, Messenger , RNA, Small Interfering , Thromboplastin , Tissue Culture Techniques
5.
Proc Natl Acad Sci U S A ; 109(37): 14983-8, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22927428

ABSTRACT

Inappropriate neutrophil activation contributes to the pathogenesis of acute lung injury (ALI). Apoptosis is essential for removal of neutrophils from inflamed tissues and timely resolution of inflammation. Resolvin E1 (RvE1) is an endogenous lipid mediator derived from the ω-3 polyunsaturated fatty acid eicosapentaenoic acid that displays proresolving actions. Because the balance of prosurvival and proapoptosis signals determines the fate of neutrophils, we investigated the impact of RvE1 on neutrophil apoptosis and the outcome of neutrophil-mediated pulmonary inflammation in mice. Culture of human neutrophils with RvE1 accelerated apoptosis evoked by phagocytosis of opsonized Escherichia coli or yeast. RvE1 through the leukotriene B(4) receptor BLT1 enhanced NADPH oxidase-derived reactive oxygen species generation and subsequent activation of caspase-8 and caspase-3. RvE1 also attenuated ERK and Akt-mediated apoptosis-suppressing signals from myeloperoxidase, serum amyloid A, and bacterial DNA, shifting the balance of pro- and anti-survival signals toward apoptosis via induction of mitochondrial dysfunction. In mice, RvE1 treatment enhanced the resolution of established neutrophil-mediated pulmonary injury evoked by intratracheal instillation or i.p. administration of live E. coli or intratracheal instillation of carrageenan plus myeloperoxidase via facilitating neutrophil apoptosis and their removal by macrophages. The actions of RvE1 were prevented by the pan-caspase inhibitor zVAD-fmk. These results identify a mechanism, promotion of phagocytosis-induced neutrophil apoptosis and mitigation of potent anti-apoptosis signals, by which RvE1 could enhance resolution of acute lung inflammation.


Subject(s)
Apoptosis/drug effects , Eicosapentaenoic Acid/analogs & derivatives , Neutrophils/drug effects , Phagocytosis/drug effects , Pneumonia/drug therapy , Amino Acid Chloromethyl Ketones , Analysis of Variance , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cells, Cultured , Eicosapentaenoic Acid/antagonists & inhibitors , Eicosapentaenoic Acid/pharmacology , Escherichia coli , Humans , Mice , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Receptors, Leukotriene B4/metabolism , Statistics, Nonparametric , Yeasts
6.
FASEB J ; 25(9): 3186-96, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21670067

ABSTRACT

C-reactive protein (CRP) has been implicated in the regulation of inflammation underlying coronary artery disease; however, little is known about the molecular mechanisms responsible for the expression of its pro- or anti-inflammatory activities. Here, we have identified the intrasubunit disulfide bond as a conserved switch that controls the structure and functions of CRP. Conformational rearrangement in human pentameric CRP to monomeric CRP (mCRP) is the prerequisite for this switch to be activated by reducing agents, including thioredoxin. Immunohistochemical analysis revealed 36-79% colocalization of thioredoxin and mCRP in human advanced coronary atherosclerotic lesions. Nonreduced mCRP was largely inert in activating human coronary artery endothelial cells (HCAECs), whereas reduced or cysteine-mutated mCRP evoked marked release of IL-8 and monocyte chemoattractant protein-1 from HCAECs, with ~50% increase at a concentration of 1 µg/ml. Reduced mCRP was ~4 to 40-fold more potent than mCRP in up-regulating adhesion molecule expression, promoting U937 monocyte adhesion to HCAECs, and inducing cytokine release from rabbit arteries ex vivo and in mice. These actions were primarily due to unlocking the lipid raft interaction motif. Therefore, expression of proinflammatory properties of CRP on endothelial cells requires sequential conformational changes, i.e., loss of pentameric symmetry followed by reduction of the intrasubunit disulfide bond.


Subject(s)
C-Reactive Protein/metabolism , C-Reactive Protein/pharmacology , Endothelial Cells/drug effects , Animals , Aorta/drug effects , Atherosclerosis/metabolism , Atherosclerosis/pathology , C-Reactive Protein/chemistry , Cells, Cultured , Cholesterol, LDL/metabolism , Complement C1q/metabolism , Coronary Vessels/cytology , Gene Expression Regulation , Humans , Inflammation/metabolism , Male , Mice , Oxidation-Reduction , Protein Binding , Protein Conformation , Rabbits
7.
J Immunol ; 182(7): 4386-94, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299739

ABSTRACT

TLR9 detects bacterial DNA (CpG DNA) and elicits both innate and adoptive immunity. Recent evidence indicates that TLR9 is expressed in more diverse cell types than initially thought. In this study, we report that HUVECs constitutively express TLR9 and selectively recognize unmethylated CpG motifs in bacterial DNA and synthetic immune stimulatory CpG oligodeoxynucleotides. HUVECs respond to CpG DNA with rapid phosphorylation of IkappaB-alpha and NF-kappaB-mediated gene transcription and surface expression of the adhesion molecules ICAM-1 and E-selectin independent of MAPK signaling. The telomere-derived TLR9 inhibitory oligonucleotide 5'-TTT AGG GTT AGG GTT AGG G-3', agents that block endosomal acidification such as chloroquine and bafilomycin A, and NF-kappaB inhibitors abrogated CpG DNA-induced signaling. HUVEC activation by CpG DNA led to markedly enhanced neutrophil adhesion under nonstatic conditions that was further enhanced when neutrophils were stimulated with CpG DNA. The adhesive interactions were blocked by Abs against CD18 and, to a lesser degree, by anti-E-selectin and anti-L-selectin Abs. Our findings demonstrate that bacterial DNA promotes beta(2) integrin and E-selectin-mediated HUVEC-neutrophil adherence, and indicate the ability of CpG DNA to initiate and/or maintain the inflammatory response.


Subject(s)
DNA, Bacterial/immunology , Endothelial Cells/immunology , Gene Expression Regulation/immunology , Neutrophils/immunology , Signal Transduction/physiology , Toll-Like Receptor 9/biosynthesis , Blotting, Western , Cell Adhesion/physiology , DNA, Bacterial/metabolism , E-Selectin/immunology , E-Selectin/metabolism , Endothelial Cells/metabolism , Gene Expression , Humans , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Neutrophils/metabolism , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/metabolism , Toll-Like Receptor 9/genetics , Umbilical Veins
8.
Circ Res ; 103(4): 352-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18617697

ABSTRACT

Polymorphonuclear neutrophil granulocytes have a central role in innate immunity and their programmed cell death and removal are critical for efficient resolution of acute inflammation. Myeloperoxidase (MPO), a heme protein abundantly expressed in neutrophils, is generally associated with killing of bacteria and oxidative tissue injury. Because MPO also binds to neutrophils, we investigated whether MPO could affect the lifespan of neutrophils. Here, we report that MPO independent of its catalytic activity through signaling via the adhesion molecule CD11b/CD18 rescued human neutrophils from constitutive apoptosis and prolonged their life span. MPO evoked a transient concurrent activation of extracellular signal-regulated kinase and Akt, leading to phosphorylation of Bad at both Ser112 and Ser136, prevention of mitochondrial dysfunction, and subsequent activation of caspase-3. Consistently, pharmacological inhibition of extracellular signal-regulated kinase, Akt, or caspase-3 reversed the antiapoptosis action of MPO. Acute increases in plasma MPO delayed murine neutrophil apoptosis assayed ex vivo. In a mouse model of self-resolving inflammation, MPO also prolonged the duration of carrageenan-induced acute lung injury, as evidenced by enhanced alveolar permeability and accumulation of neutrophils parallel with suppression of neutrophil apoptosis. Our results indicate that MPO functions as a survival signal for neutrophils and thereby contribute to prolongation of inflammation.


Subject(s)
Apoptosis/physiology , CD11b Antigen/metabolism , CD18 Antigens/metabolism , Inflammation/metabolism , Neutrophils/metabolism , Peroxidase/metabolism , Animals , Carrageenan , Caspase 3/metabolism , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Inflammation/pathology , Inflammation/physiopathology , Male , Mice , Mice, Inbred BALB C , Mitochondria/physiology , Neutrophils/cytology , Neutrophils/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology
9.
Am J Respir Crit Care Med ; 180(4): 311-9, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19483113

ABSTRACT

RATIONALE: Apoptosis is essential for removal of neutrophils from inflamed tissues and efficient resolution of inflammation. Myeloperoxidase (MPO), abundantly expressed in neutrophils, not only generates cytotoxic oxidants but also signals through the beta(2) integrin Mac-1 to rescue neutrophils from constitutive apoptosis, thereby prolonging inflammation. OBJECTIVES: Because aspirin-triggered 15-epi-lipoxin A(4) (15-epi-LXA(4)) modulates Mac-1 expression, we investigated the impact of 15-epi-LXA(4) on MPO suppression of neutrophil apoptosis and MPO-mediated neutrophil-dependent acute lung injury. METHODS: Human neutrophils were cultured with MPO with or without 15-epi-LXA(4) to investigate development of apoptosis. Acute lung injury was produced by intratracheal injection of carrageenan plus MPO or intraperitoneal injection of live Escherichia coli in mice, and the animals were treated with 15-epi-LXA(4) at the peak of inflammation. MEASUREMENTS AND MAIN RESULTS: 15-Epi-LXA(4) through down-regulation of Mac-1 expression promoted apoptosis of human neutrophils by attenuating MPO-induced activation of extracellular signal-regulated kinase and Akt-mediated phosphorylation of Bad and by reducing expression of the antiapoptotic protein Mcl-1, thereby aggravating mitochondrial dysfunction. The proapoptotic effect of 15-epi-LXA(4) was dominant over MPO-mediated effects even when it was added at 4 hours post MPO. In mice, treatment with 15-epi-LXA(4) accelerated the resolution of established carrageenan plus MPO-evoked as well as E. coli-induced neutrophil-dependent pulmonary inflammation through redirecting neutrophils to caspase-mediated cell death and facilitating their removal by macrophages. CONCLUSIONS: These results demonstrate that aspirin-triggered 15-epi-LXA(4) enhances resolution of inflammation by overriding the powerful antiapoptosis signal from MPO, thereby demonstrating a hitherto unrecognized mechanism by which aspirin promotes resolution of inflammation.


Subject(s)
Acute Lung Injury/immunology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Lipoxins/pharmacology , Peroxidase/antagonists & inhibitors , Signal Transduction/drug effects , Acute Lung Injury/pathology , Animals , Apoptosis/drug effects , Aspirin/pharmacology , Bronchoalveolar Lavage Fluid/immunology , CD18 Antigens/blood , Caspase 3/blood , Female , Humans , Interleukin-6/blood , Leukocyte Count , Lipoxins/blood , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , Neutrophils/immunology
10.
ScientificWorldJournal ; 10: 1731-48, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20842319

ABSTRACT

Neutrophil granulocytes play a central role in host defense to infection and tissue injury. Their timely removal is essential for resolution of inflammation. Increasing evidence identified neutrophil apoptosis as an important control point in the development and resolution of inflammation. Delayed apoptosis and/or impaired clearance of neutrophils aggravate and prolong tissue injury. This review will focus on outside-in signals that provide survival cues for neutrophils, the hierarchy of pro- and antiapoptotic signals, and molecular targets in the antiapoptotic signaling network that can be exploited by endogenously produced bioactive lipids, such as lipoxins or pharmacological inhibitors, including cyclin-dependent kinase inhibitors, to redirect neutrophils to apoptosis in vivo, thus promoting resolution of inflammation.


Subject(s)
Apoptosis/immunology , Inflammation/immunology , Neutrophils/immunology , Signal Transduction/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Humans , Inflammation/prevention & control , Leukocyte Count , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Roscovitine , Signal Transduction/drug effects
11.
J Cell Biochem ; 108(5): 1039-46, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19760640

ABSTRACT

Neutrophils are essential for host defense and their programmed cell death and removal are critical for the optimal expression as well as for efficient resolution of inflammation. Delayed neutrophil apoptosis or impaired clearance of apoptotic neutrophils by macrophages contributes to the progression of chronic inflammation. Under most conditions, neutrophils are exposed to multiple factors and their fate would ultimately depend on the balance between pro-survival and pro-apoptotic signals. Life or death decisions are tightly controlled by a complex network of intracellular signaling pathways. Accumulating data indicate that receptors, such as the formyl peptide receptor 2/lipoxin receptor or beta(2)-integrins can generate contrasting cues in neutrophils in a ligand-specific manner and suggest a hierarchy among these signals. In this article, we review recent advances on how pro-apoptosis and pro-survival signals interact to determine the fate of neutrophils and the inflammatory response, and highlight novel pharmacological strategies that could be used to enhance the resolution of inflammation by redirecting neutrophils to apoptosis.


Subject(s)
Apoptosis , Inflammation/pathology , Neutrophils/pathology , Animals , Apoptosis/physiology , CD18 Antigens/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Humans , Inflammation/physiopathology , Ligands , Mice , Neutrophils/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Signal Transduction/physiology
12.
J Leukoc Biol ; 84(3): 600-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18495783

ABSTRACT

Neutrophils have a central role in innate immunity, and their programmed cell death and removal are critical to the optimal expression as well as to efficient resolution of inflammation. Human neutrophils express the pleiotropic receptor formyl peptide receptor-like 1/lipoxin A4 (LXA(4)) receptor that binds a variety of ligands, including the acute-phase reactant serum amyloid A (SAA), the anti-inflammatory lipids LXA(4) and aspirin-triggered 15-epi-LXA(4) (ATL), and the glucocorticoid-inducible protein annexin 1. In addition to regulation of neutrophil activation and recruitment, these ligands have a profound influence on neutrophil survival and apoptosis with contrasting actions, mediating aggravation or resolution of the inflammatory response. Thus, annexin 1 accelerates, whereas SAA rescues human neutrophils from constitutive apoptosis by preventing mitochondrial dysfunction and subsequent activation of caspase-3. Furthermore, ATL overcomes the antiapoptosis signal from SAA and redirects neutrophils to caspase-mediated cell death. We review recent developments about the molecular basis of these actions and suggest a novel mechanism by which aspirin promotes resolution of acute inflammation and tissue injury.


Subject(s)
Apoptosis , Neutrophils/pathology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Annexin A1/metabolism , Apoptosis Regulatory Proteins/physiology , Aspirin/pharmacology , Humans , Lipoxins/metabolism , Neutrophil Activation/drug effects , Neutrophil Infiltration/immunology , Neutrophils/metabolism , Serum Amyloid A Protein/metabolism
13.
Nat Commun ; 10(1): 3471, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31375662

ABSTRACT

The uptake of apoptotic polymorphonuclear cells (PMN) by macrophages is critical for timely resolution of inflammation. High-burden uptake of apoptotic cells is associated with loss of phagocytosis in resolution phase macrophages. Here, using a transcriptomic analysis of macrophage subsets, we show that non-phagocytic resolution phase macrophages express a distinct IFN-ß-related gene signature in mice. We also report elevated levels of IFN-ß in peritoneal and broncho-alveolar exudates in mice during the resolution of peritonitis and pneumonia, respectively. Elimination of endogenous IFN-ß impairs, whereas treatment with exogenous IFN-ß enhances, bacterial clearance, PMN apoptosis, efferocytosis and macrophage reprogramming. STAT3 signalling in response to IFN-ß promotes apoptosis of human PMNs. Finally, uptake of apoptotic cells promotes loss of phagocytic capacity in macrophages alongside decreased surface expression of efferocytic receptors in vivo. Collectively, these results identify IFN-ß produced by resolution phase macrophages as an effector cytokine in resolving bacterial inflammation.


Subject(s)
Interferon-beta/metabolism , Macrophages/immunology , Peritonitis/immunology , Pneumonia, Bacterial/immunology , Adult , Aged , Animals , Apoptosis/immunology , Disease Models, Animal , Escherichia coli/immunology , Female , Gene Expression Profiling , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Jurkat Cells , Macrophages/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Neutrophils , Pneumonia, Bacterial/microbiology , Primary Cell Culture , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism
14.
Arch Immunol Ther Exp (Warsz) ; 56(1): 41-53, 2008.
Article in English | MEDLINE | ID: mdl-18250968

ABSTRACT

Neutrophils are essential for host defense and detect the presence of invading microorganisms through recognition of pathogen-associated molecular patterns. Among these receptors are Toll-like receptors (TLRs). Neutrophils express all known TLRs except for TLR3. TLR9, localized intracellularly, is to date the best characterized sensor for bacterial DNA, containing short sequences of unmethylated CpG motifs, though TLR9-independent intracellular DNA recognition mechanism(s) may also exist. Bacterial DNA has profound impact on neutrophil functions; it promotes neutrophil trafficking in vivo, induces chemokine expression, regulates expression of adhesion molecules, enhances phagocyte activity, and rescues neutrophils from constitutive apoptosis. TLR9 stimulation results in alterations in cellular redox balance, peroxynitrite formation, activation of the mitogen-activated protein kinase, PI3-kinase, and Jun N-terminal kinase pathways and/or nuclear factor kappaB and AP-1. These features identify an important role for bacterial DNA and TLR9 signaling in the regulation of neutrophil functions that are critical for optimal expression as well as for resolution of the inflammatory response.


Subject(s)
DNA, Bacterial , Neutrophil Activation , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/metabolism , Toll-Like Receptor 9/metabolism , Animals , Apoptosis/immunology , Bacterial Infections/immunology , Bacterial Infections/metabolism , Cell Survival/immunology , DNA, Bacterial/immunology , DNA, Bacterial/metabolism , Gene Expression Regulation , Humans , Mice , Neutrophil Activation/immunology , Neutrophil Infiltration/immunology , Neutrophils/pathology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
15.
Eur J Pharmacol ; 833: 339-348, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29935171

ABSTRACT

The formyl peptide receptors (FPRs) are G protein coupled receptors that recognize a broad range of structurally distinct pathogen and danger-associated molecular patterns and mediate host defense to infection and tissue injury. It became evident that the cellular distribution and biological functions of FPRs extend beyond myeloid cells and governing their activation and trafficking. In recent years, significant progress has been made to position FPRs at check points that control the resolution of inflammation, tissue repair and return to homeostasis. Accumulating data indicate a role for FPRs in an ever-increasing range of human diseases, including atherosclerosis, chronic obstructive pulmonary disease, asthma, autoimmune diseases and cancer, in which dysregulated or defective resolution are increasingly recognized as critical component of the pathogenesis. This review summarizes recent advances on how FPRs recognize distinct ligands and integrate opposing cues to govern various responses and will discuss how this knowledge could be harnessed for developing novel therapeutic strategies to counter inflammation that underlies many human diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Homeostasis/drug effects , Host-Pathogen Interactions/drug effects , Inflammation/drug therapy , Receptors, Formyl Peptide/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/therapeutic use , Homeostasis/physiology , Host-Pathogen Interactions/physiology , Humans , Inflammation/etiology , Inflammation/pathology , Ligands , Receptors, Formyl Peptide/metabolism
16.
Cell Signal ; 18(12): 2302-13, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16806822

ABSTRACT

Neutrophils die rapidly via apoptosis and their survival is contingent upon rescue from constitutive programmed cell death by signals from the microenvironment. In these experiments, we investigated whether prevention of K(+) efflux could affect the apoptotic machinery in human neutrophils. Disruption of the natural K(+) electrochemical gradient suppressed neutrophil apoptosis (assessed by annexin V binding, nuclear DNA content and nucleosomal DNA fragmentation) and prolonged cell survival within 24-48 h of culture. High extracellular K(+) (10-100 mM) did not activate extracellular signal-regulated kinase (ERK) and Akt, nor affected phosphorylation of p38 MAPK associated with constitutive apoptosis. Consistently, pharmacological blockade of ERK kinase or phosphatidylinositol 3-kinase (PI 3-kinase) did not affect the anti-apoptotic action of KCl. Inhibition of K(+) efflux effectively reduced, though never completely inhibited, decreases in mitochondrial transmembrane potential (DeltaPsi(m)) that preceded development of apoptotic morphology. Changes in DeltaPsi(m) resulted in attenuation of cytochrome c release from mitochondria into the cytosol and decreases in caspase-3 activity. Culture of neutrophils in medium containing 80 mM KCl with the pan-caspase inhibitor Z-VAD-FMK resulted in slightly greater suppression of apoptosis than KCl alone. High extracellular KCl also attenuated translocation of apoptosis-inducing factor (AIF) and endonuclease G (EndoG) from mitochondria to nuclei. The DNase inhibitor, aurintricarboxylic acid (ATA) partially inhibited nucleosomal DNA fragmentation, and the effects of ATA and 80 mM KCl were not additive. These results show that prevention of K(+) efflux promotes neutrophil survival by suppressing apoptosis through preventing mitochondrial dysfunction and release of the pro-apoptotic proteins cytochrome c, AIF and EndoG independent of ERK, PI 3-kinase and p38 MAPK. Thus, K(+) released locally from damaged cells may function as a survival signal for neutrophils.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Endodeoxyribonucleases/metabolism , Mitochondria/physiology , Neutrophils/metabolism , Potassium/metabolism , Adult , Apoptosis/drug effects , Apoptosis Inducing Factor/antagonists & inhibitors , Apoptosis Inducing Factor/metabolism , Apoptosis Regulatory Proteins/antagonists & inhibitors , Caspase 3/metabolism , Caspase Inhibitors , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endodeoxyribonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Flow Cytometry , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Middle Aged , Mitochondria/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Potassium Chloride/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Leukoc Biol ; 102(6): 1389-1400, 2017 12.
Article in English | MEDLINE | ID: mdl-28928268

ABSTRACT

Accumulating evidence indicates development of local extracellular acidosis in inflamed tissues in response to infection and tissue injury. Activation of infiltrating neutrophils contributes to a transient decrease in pH, which, in turn, triggers innate immunity. In this study, we investigated the impact of extracellular acidosis on neutrophil apoptosis, a critical determinant of the outcome of the inflammatory response and analyzed the underlying signaling pathways. Culture of human isolated neutrophils in mildly acidotic conditions (pH 6.5-7.0) resulted in activation of NF-κB; intracellular accumulation of cAMP; and phosphorylation of Akt, ERK, and p38 MAPK; and preservation of Mcl-1 expression. Consequently, extracellular acidosis prevented disruption of mitochondrial transmembrane potential and translocation of cytochrome c and apoptosis-inducing factor from the mitochondria to cytoplasm and nuclei, respectively and inhibited caspase-3 activity. Pharmacological inhibition of ERK, PI3K, NF-κB, or PKA partially reversed survival cues by extracellular acidosis and redirected neutrophils to apoptosis. Conversely, dibutyryl cAMP (100-500 µM) delayed apoptosis of neutrophils cultured at pH 7.4. Extracellular acidosis-generated survival cues were additive to the potent prosurvival signals from bacterial DNA, LPS, modified C-reactive protein, and serum amyloid A. Acidosis increased CpG DNA uptake by neutrophils and augmented phosphorylation of ERK and Akt, leading to preservation of Mcl-1 expression. Our results identified extracellular acidosis as a survival signal for neutrophils by suppressing the constitutive apoptotic machinery and suggest that transient decreases in local pH can enhance neutrophil responses to inflammatory stimuli, thereby contributing to amplification or prolongation of the inflammatory response.


Subject(s)
Acidosis/metabolism , Acidosis/pathology , Apoptosis , Inflammation Mediators/metabolism , Neutrophils/metabolism , Signal Transduction , Apoptosis/drug effects , Cell Survival/drug effects , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Space/metabolism , Humans , Hydrogen-Ion Concentration , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/metabolism , Neutrophils/drug effects , Oligodeoxyribonucleotides/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects
18.
Chest ; 128(4): 2910-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16236968

ABSTRACT

BACKGROUND: Cardiopulmonary bypass (CPB), a procedure often used during cardiac surgery, is associated with an inflammatory process that leads to lung injury. We hypothesized that inhaled nitric oxide (INO), which has anti-inflammatory properties, possesses the ability to modulate lung cell apoptosis and prevent CPB-induced inflammation. METHODS: Twenty male pigs were randomly classified into four groups: sham, sham plus INO, CPB, and CPB plus INO. INO (20 ppm) was administered for 24 h after anesthesia. CPB was performed 90 min into INO treatment. BAL fluid and blood were collected at time 0 (before CPB), at 4 h after beginning CPB, and 24 h after beginning CPB (T24). RESULTS: At T(24), BAL interleukin (IL)-8 levels and neutrophil percentages were elevated significantly in the CPB group. At T(24), INO reduced IL-8 concentrations and attenuated the increase of neutrophil percentage in the CPB-plus-INO group. Nitrite-plus-nitrate (NOx) concentrations were decreased significantly in groups without INO. Moreover, animals treated with INO showed higher rates of pulmonary apoptosis compared to their respective control groups except for the sham-plus-INO group, in which they were diminished. CONCLUSION: These results demonstrate that NOx production is reduced after CPB, and that INO acts as an anti-inflammatory agent by decreasing neutrophil numbers and their major chemoattractant, IL-8. INO also increases cell apoptosis in the lungs during inflammatory conditions, which may explain, in part, how it resolves pulmonary inflammation.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Inflammation/drug therapy , Nitric Oxide/therapeutic use , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Inflammation/etiology , Instillation, Drug , Male , Nitric Oxide/administration & dosage , Postoperative Complications/drug therapy , Swine
19.
PLoS One ; 9(1): e87006, 2014.
Article in English | MEDLINE | ID: mdl-24466313

ABSTRACT

Neutrophils detect bacterial constituents, including bacterial DNA (CpG DNA), which elicits innate immunity and prolongs the functional life span of neutrophils through suppression of apoptosis. Both the anti-apoptotic protein Mcl-1 and activation of NF-κB have been implicated in neutrophil survival, but there is no evidence that these are linked in neutrophils. We hypothesized that CpG DNA could simultaneously activate these pathways. High purity CpG DNA (0.4-3.2 µg/ml) extended the life span of human neutrophils in vitro by delaying apoptosis through altering the rate of Mcl-1 turnover. CpG DNA slightly decreased Mcl-1 protein level in the presence of cyclohexmide and the proteasome inhibitor MG132 had little effect on Mcl-1 expression in CpG DNA-treated neutrophils. In contrast, CpG DNA evoked rapid increases in DNA binding by NF-κB/p65 and Mcl-1 mRNA. NF-κB inhibitors and the telomere-derived TLR9 inhibitory oligonucleotide 5'-TTT AGG GTT AGG GTT AGG G-3' markedly reduced Mcl-1 protein levels and subsequently abrogated suppression of apoptosis by CpG DNA. Furthermore, CpG DNA attenuated the decreases in Mcl-1 in both cell lysate and nucleus of neutrophils undergoing spontaneous apoptosis and increased Mcl-1 translocation to the mitochondria, leading to preservation of mitochondrial transmembrane potential. These results demonstrate that CpG DNA through toll-like receptor 9 links two survival signaling pathways by delaying apoptosis through induction of NF-κB-mediated Mcl-1 gene transcription and promoting Mcl-1 translocation to the mitochondria.


Subject(s)
Apoptosis/immunology , Gene Expression Regulation/immunology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neutrophils/immunology , Signal Transduction/immunology , Toll-Like Receptor 9/metabolism , DNA Primers/genetics , DNA, Bacterial/immunology , Escherichia coli/genetics , Humans , NF-kappa B/metabolism , Statistics, Nonparametric
20.
Front Immunol ; 4: 60, 2013.
Article in English | MEDLINE | ID: mdl-23508943

ABSTRACT

Precise control of the neutrophil death program provides a balance between their defense functions and safe clearance, whereas impaired regulation of neutrophil death is thought to contribute to a wide range of inflammatory pathologies. Apoptosis is essential for neutrophil functional shutdown, removal of emigrated neutrophils, and timely resolution of inflammation. Neutrophils receive survival and pro-apoptosis cues from the inflammatory microenvironment and integrate these signals through surface receptors and common downstream mechanisms. Among these receptors are the leukocyte-specific membrane receptors ß2 integrins that are best known for regulating adhesion and phagocytosis. Accumulating evidence indicate that outside-in signaling through the ß2 integrin Mac-1 can generate contrasting cues in neutrophils, leading to promotion of their survival or apoptosis. Binding of Mac-1 to its ligands ICAM-1, fibrinogen, or the azurophilic granule enzyme myeloperoxidase suppresses apoptosis, whereas Mac-1-mediated phagocytosis of bacteria evokes apoptotic cell death. Mac-1 signaling is also target for the anti-inflammatory, pro-resolving mediators, including lipoxin A4, aspirin-triggered lipoxin A4, and resolvin E1. This review focuses on molecular mechanisms underlying Mac-1 regulation of neutrophil apoptosis and highlights recent advances how hierarchy of survival and pro-apoptosis signals can be harnessed to facilitate neutrophil apoptosis and the resolution of inflammation.

SELECTION OF CITATIONS
SEARCH DETAIL