Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 23(6): 927-939, 2022 06.
Article in English | MEDLINE | ID: mdl-35624205

ABSTRACT

Hypoxemia is a defining feature of acute respiratory distress syndrome (ARDS), an often-fatal complication of pulmonary or systemic inflammation, yet the resulting tissue hypoxia, and its impact on immune responses, is often neglected. In the present study, we have shown that ARDS patients were hypoxemic and monocytopenic within the first 48 h of ventilation. Monocytopenia was also observed in mouse models of hypoxic acute lung injury, in which hypoxemia drove the suppression of type I interferon signaling in the bone marrow. This impaired monopoiesis resulted in reduced accumulation of monocyte-derived macrophages and enhanced neutrophil-mediated inflammation in the lung. Administration of colony-stimulating factor 1 in mice with hypoxic lung injury rescued the monocytopenia, altered the phenotype of circulating monocytes, increased monocyte-derived macrophages in the lung and limited injury. Thus, tissue hypoxia altered the dynamics of the immune response to the detriment of the host and interventions to address the aberrant response offer new therapeutic strategies for ARDS.


Subject(s)
Lung Injury , Respiratory Distress Syndrome , Animals , Humans , Hypoxia/etiology , Inflammation/complications , Lung , Lung Injury/complications , Mice
3.
Blood ; 139(2): 281-286, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34411229

ABSTRACT

Neutrophils are predominantly glycolytic cells that derive little ATP from oxidative phosphorylation; however, they possess an extensive mitochondrial network and maintain a mitochondrial membrane potential. Although studies have shown neutrophils need their mitochondria to undergo apoptosis and regulate NETosis, the metabolic role of the respiratory chain in these highly glycolytic cells is still unclear. Recent studies have expanded on the role of reactive oxygen species (ROS) released from the mitochondria as intracellular signaling molecules. Our study shows that neutrophils can use their mitochondria to generate ROS and that mitochondrial ROS release is increased in hypoxic conditions. This is needed for the stabilization of a high level of the critical hypoxic response factor and pro-survival protein HIF-1α in hypoxia. Further, we demonstrate that neutrophils use the glycerol 3-phosphate pathway as a way of directly regulating mitochondrial function through glycolysis, specifically to maintain polarized mitochondria and produce ROS. This illustrates an additional pathway by which neutrophils can regulate HIF-1α stability and will therefore be an important consideration when looking for treatments of inflammatory conditions in which HIF-1α activation and neutrophil persistence at the site of inflammation are linked to disease severity.


Subject(s)
Glycerophosphates/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/metabolism , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Cell Hypoxia , Cells, Cultured , Humans , Protein Stability
4.
Am J Respir Crit Care Med ; 207(8): 998-1011, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36724365

ABSTRACT

Rationale: Chronic obstructive pulmonary disease (COPD) is a disease characterized by persistent airway inflammation and disordered macrophage function. The extent to which alterations in macrophage bioenergetics contribute to impaired antioxidant responses and disease pathogenesis has yet to be fully delineated. Objectives: Through the study of COPD alveolar macrophages (AMs) and peripheral monocyte-derived macrophages (MDMs), we sought to establish if intrinsic defects in core metabolic processes drive macrophage dysfunction and redox imbalance. Methods: AMs and MDMs from donors with COPD and healthy donors underwent functional, metabolic, and transcriptional profiling. Measurements and Main Results: We observed that AMs and MDMs from donors with COPD display a critical depletion in glycolytic- and mitochondrial respiration-derived energy reserves and an overreliance on glycolysis as a source for ATP, resulting in reduced energy status. Defects in oxidative metabolism extend to an impaired redox balance associated with defective expression of the NADPH-generating enzyme, ME1 (malic enzyme 1), a known target of the antioxidant transcription factor NRF2 (nuclear factor erythroid 2-related factor 2). Consequently, selective activation of NRF2 resets the COPD transcriptome, resulting in increased generation of TCA cycle intermediaries, improved energetic status, favorable redox balance, and recovery of macrophage function. Conclusions: In COPD, an inherent loss of metabolic plasticity leads to metabolic exhaustion and reduced redox capacity, which can be rescued by activation of the NRF2 pathway. Targeting these defects, via NRF2 augmentation, may therefore present an attractive therapeutic strategy for the treatment of the aberrant airway inflammation described in COPD.


Subject(s)
NF-E2-Related Factor 2 , Pulmonary Disease, Chronic Obstructive , Humans , Macrophages/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Malate Dehydrogenase/metabolism
5.
Am J Respir Crit Care Med ; 207(11): 1515-1524, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36780644

ABSTRACT

Rationale: Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by limited treatment options and high mortality. A better understanding of the molecular drivers of IPF progression is needed. Objectives: To identify and validate molecular determinants of IPF survival. Methods: A staged genome-wide association study was performed using paired genomic and survival data. Stage I cases were drawn from centers across the United States and Europe and stage II cases from Vanderbilt University. Cox proportional hazards regression was used to identify gene variants associated with differential transplantation-free survival (TFS). Stage I variants with nominal significance (P < 5 × 10-5) were advanced for stage II testing and meta-analyzed to identify those reaching genome-wide significance (P < 5 × 10-8). Downstream analyses were performed for genes and proteins associated with variants reaching genome-wide significance. Measurements and Main Results: After quality controls, 1,481 stage I cases and 397 stage II cases were included in the analysis. After filtering, 9,075,629 variants were tested in stage I, with 158 meeting advancement criteria. Four variants associated with TFS with consistent effect direction were identified in stage II, including one in an intron of PCSK6 (proprotein convertase subtilisin/kexin type 6) reaching genome-wide significance (hazard ratio, 4.11 [95% confidence interval, 2.54-6.67]; P = 9.45 × 10-9). PCSK6 protein was highly expressed in IPF lung parenchyma. PCSK6 lung staining intensity, peripheral blood gene expression, and plasma concentration were associated with reduced TFS. Conclusions: We identified four novel variants associated with IPF survival, including one in PCSK6 that reached genome-wide significance. Downstream analyses suggested that PCSK6 protein plays a potentially important role in IPF progression.


Subject(s)
Genome-Wide Association Study , Idiopathic Pulmonary Fibrosis , Humans , Lung , Proportional Hazards Models , Europe , Serine Endopeptidases , Proprotein Convertases
6.
Am J Respir Crit Care Med ; 201(5): 564-574, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31710517

ABSTRACT

Rationale: Idiopathic pulmonary fibrosis (IPF) is a complex lung disease characterized by scarring of the lung that is believed to result from an atypical response to injury of the epithelium. Genome-wide association studies have reported signals of association implicating multiple pathways including host defense, telomere maintenance, signaling, and cell-cell adhesion.Objectives: To improve our understanding of factors that increase IPF susceptibility by identifying previously unreported genetic associations.Methods: We conducted genome-wide analyses across three independent studies and meta-analyzed these results to generate the largest genome-wide association study of IPF to date (2,668 IPF cases and 8,591 controls). We performed replication in two independent studies (1,456 IPF cases and 11,874 controls) and functional analyses (including statistical fine-mapping, investigations into gene expression, and testing for enrichment of IPF susceptibility signals in regulatory regions) to determine putatively causal genes. Polygenic risk scores were used to assess the collective effect of variants not reported as associated with IPF.Measurements and Main Results: We identified and replicated three new genome-wide significant (P < 5 × 10-8) signals of association with IPF susceptibility (associated with altered gene expression of KIF15, MAD1L1, and DEPTOR) and confirmed associations at 11 previously reported loci. Polygenic risk score analyses showed that the combined effect of many thousands of as yet unreported IPF susceptibility variants contribute to IPF susceptibility.Conclusions: The observation that decreased DEPTOR expression associates with increased susceptibility to IPF supports recent studies demonstrating the importance of mTOR signaling in lung fibrosis. New signals of association implicating KIF15 and MAD1L1 suggest a possible role of mitotic spindle-assembly genes in IPF susceptibility.


Subject(s)
Idiopathic Pulmonary Fibrosis/genetics , Aged , Case-Control Studies , Cell Cycle Proteins/genetics , Female , Gene Expression , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kinesins/genetics , Male , Middle Aged , Risk Assessment , Signal Transduction , Spindle Apparatus , TOR Serine-Threonine Kinases/metabolism
7.
Am J Respir Crit Care Med ; 200(2): 235-246, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30849228

ABSTRACT

Rationale: Acute respiratory distress syndrome is defined by the presence of systemic hypoxia and consequent on disordered neutrophilic inflammation. Local mechanisms limiting the duration and magnitude of this neutrophilic response remain poorly understood. Objectives: To test the hypothesis that during acute lung inflammation tissue production of proresolution type 2 cytokines (IL-4 and IL-13) dampens the proinflammatory effects of hypoxia through suppression of HIF-1α (hypoxia-inducible factor-1α)-mediated neutrophil adaptation, resulting in resolution of lung injury. Methods: Neutrophil activation of IL4Ra (IL-4 receptor α) signaling pathways was explored ex vivo in human acute respiratory distress syndrome patient samples, in vitro after the culture of human peripheral blood neutrophils with recombinant IL-4 under conditions of hypoxia, and in vivo through the study of IL4Ra-deficient neutrophils in competitive chimera models and wild-type mice treated with IL-4. Measurements and Main Results: IL-4 was elevated in human BAL from patients with acute respiratory distress syndrome, and its receptor was identified on patient blood neutrophils. Treatment of human neutrophils with IL-4 suppressed HIF-1α-dependent hypoxic survival and limited proinflammatory transcriptional responses. Increased neutrophil apoptosis in hypoxia, also observed with IL-13, required active STAT signaling, and was dependent on expression of the oxygen-sensing prolyl hydroxylase PHD2. In vivo, IL-4Ra-deficient neutrophils had a survival advantage within a hypoxic inflamed niche; in contrast, inflamed lung treatment with IL-4 accelerated resolution through increased neutrophil apoptosis. Conclusions: We describe an important interaction whereby IL4Rα-dependent type 2 cytokine signaling can directly inhibit hypoxic neutrophil survival in tissues and promote resolution of neutrophil-mediated acute lung injury.


Subject(s)
Acute Lung Injury/immunology , Interleukin-4 Receptor alpha Subunit/immunology , Interleukin-4/immunology , Neutrophils/immunology , Receptors, Cell Surface/immunology , Respiratory Distress Syndrome/immunology , Acute Lung Injury/metabolism , Animals , Apoptosis/drug effects , Cell Hypoxia/immunology , Cell Survival/drug effects , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-4/metabolism , Interleukin-4/pharmacology , Interleukin-4 Receptor alpha Subunit/genetics , Interleukin-4 Receptor alpha Subunit/metabolism , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/metabolism , Receptors, Cell Surface/metabolism , Respiratory Distress Syndrome/metabolism , STAT Transcription Factors/metabolism , Signal Transduction
8.
Am J Respir Crit Care Med ; 200(1): 84-97, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30649895

ABSTRACT

Rationale: Antimicrobial resistance challenges therapy of pneumonia. Enhancing macrophage microbicidal responses would combat this problem but is limited by our understanding of how alveolar macrophages (AMs) kill bacteria. Objectives: To define the role and mechanism of AM apoptosis-associated bacterial killing in the lung. Methods: We generated a unique CD68.hMcl-1 transgenic mouse with macrophage-specific overexpression of the human antiapoptotic Mcl-1 protein, a factor upregulated in AMs from patients at increased risk of community-acquired pneumonia, to address the requirement for apoptosis-associated killing. Measurements and Main Results: Wild-type and transgenic macrophages demonstrated comparable ingestion and initial phagolysosomal killing of bacteria. Continued ingestion (for ≥12 h) overwhelmed initial killing, and a second, late-phase microbicidal response killed viable bacteria in wild-type macrophages, but this response was blunted in CD68.hMcl-1 transgenic macrophages. The late phase of bacterial killing required both caspase-induced generation of mitochondrial reactive oxygen species and nitric oxide, the peak generation of which coincided with the late phase of killing. The CD68.hMcl-1 transgene prevented mitochondrial reactive oxygen species but not nitric oxide generation. Apoptosis-associated killing enhanced pulmonary clearance of Streptococcus pneumoniae and Haemophilus influenzae in wild-type mice but not CD68.hMcl-1 transgenic mice. Bacterial clearance was enhanced in vivo in CD68.hMcl-1 transgenic mice by reconstitution of apoptosis with BH3 mimetics or clodronate-encapsulated liposomes. Apoptosis-associated killing was not activated during Staphylococcus aureus lung infection. Conclusions: Mcl-1 upregulation prevents macrophage apoptosis-associated killing and establishes that apoptosis-associated killing is required to allow AMs to clear ingested bacteria. Engagement of macrophage apoptosis should be investigated as a novel, host-based antimicrobial strategy.


Subject(s)
Apoptosis/physiology , Macrophages, Alveolar/physiology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Phagocytosis/genetics , Phagosomes/physiology , Pneumonia, Bacterial , Animals , Apoptosis/drug effects , Bacteria , Biphenyl Compounds/pharmacology , Caspases/metabolism , Clodronic Acid/pharmacology , Disease Models, Animal , Haemophilus influenzae , Humans , Macrophages, Alveolar/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nitric Oxide/metabolism , Nitrophenols/pharmacology , Piperazines/pharmacology , Reactive Oxygen Species/metabolism , Staphylococcus aureus , Streptococcus pneumoniae , Sulfonamides/pharmacology
9.
Blood ; 130(8): 1014-1025, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28637666

ABSTRACT

The lifespan of neutrophils is plastic and highly responsive to factors that regulate cellular survival. Defects in neutrophil number and survival are common to both hematologic disorders and chronic inflammatory diseases. At sites of inflammation, neutrophils respond to multiple signals that activate protein kinase A (PKA) signaling, which positively regulates neutrophil survival. The aim of this study was to define transcriptional responses to PKA activation and to delineate the roles of these factors in neutrophil function and survival. In human neutrophil gene array studies, we show that PKA activation upregulates a significant number of apoptosis-related genes, the most highly regulated of these being NR4A2 and NR4A3 Direct PKA activation by the site-selective PKA agonist pair N6/8-AHA (8-AHA-cAMP and N6-MB-cAMP) and treatment with endogenous activators of PKA, including adenosine and prostaglandin E2, results in a profound delay of neutrophil apoptosis and concomitant upregulation of NR4A2/3 in a PKA-dependent manner. NR4A3 expression is also increased at sites of neutrophilic inflammation in a human model of intradermal inflammation. PKA activation also promotes survival of murine neutrophil progenitor cells, and small interfering RNA to NR4A2 decreases neutrophil production in this model. Antisense knockdown of NR4A2 and NR4A3 homologs in zebrafish larvae significantly reduces the absolute neutrophil number without affecting cellular migration. In summary, we show that NR4A2 and NR4A3 are components of a downstream transcriptional response to PKA activation in the neutrophil, and that they positively regulate neutrophil survival and homeostasis.


Subject(s)
Neutrophils/cytology , Neutrophils/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 3/metabolism , Zebrafish/metabolism , Animals , Cell Count , Cell Proliferation , Cell Survival , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/metabolism , Enzyme Activation , Gene Deletion , Gene Knockdown Techniques , Humans , Inflammation/pathology , Larva/metabolism , Mice , Multigene Family , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Signal Transduction , Transcription, Genetic
10.
Am J Respir Crit Care Med ; 198(6): 739-750, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29547002

ABSTRACT

RATIONALE: Previous studies have identified defects in bacterial phagocytosis by alveolar macrophages (AMs) in patients with chronic obstructive pulmonary disease (COPD), but the mechanisms and clinical consequences remain incompletely defined. OBJECTIVES: To examine the effect of COPD on AM phagocytic responses and identify the mechanisms, clinical consequences, and potential for therapeutic manipulation of these defects. METHODS: We isolated AMs and monocyte-derived macrophages (MDMs) from a cohort of patients with COPD and control subjects within the Medical Research Council COPDMAP consortium and measured phagocytosis of bacteria in relation to opsonic conditions and clinical features. MEASUREMENTS AND MAIN RESULTS: COPD AMs and MDMs have impaired phagocytosis of Streptococcus pneumoniae. COPD AMs have a selective defect in uptake of opsonized bacteria, despite the presence of antipneumococcal antibodies in BAL, not observed in MDMs or healthy donor AMs. AM defects in phagocytosis in COPD are significantly associated with exacerbation frequency, isolation of pathogenic bacteria, and health-related quality-of-life scores. Bacterial binding and initial intracellular killing of opsonized bacteria in COPD AMs was not reduced. COPD AMs have reduced transcriptional responses to opsonized bacteria, such as cellular stress responses that include transcriptional modules involving antioxidant defenses and Nrf2 (nuclear factor erythroid 2-related factor 2)-regulated genes. Agonists of the cytoprotective transcription factor Nrf2 (sulforaphane and compound 7) reverse defects in phagocytosis of S. pneumoniae and nontypeable Haemophilus influenzae by COPD AMs. CONCLUSIONS: Patients with COPD have clinically relevant defects in opsonic phagocytosis by AMs, associated with impaired transcriptional responses to cellular stress, which are reversed by therapeutic targeting with Nrf2 agonists.


Subject(s)
NF-E2-Related Factor 2/antagonists & inhibitors , Phagocytosis/drug effects , Pulmonary Disease, Chronic Obstructive/physiopathology , Adult , Aged , Case-Control Studies , Female , Humans , Isothiocyanates/pharmacology , Macrophages/drug effects , Macrophages/physiology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/physiology , Male , Middle Aged , Phagocytosis/physiology , Streptococcus pneumoniae , Sulfoxides
11.
Thorax ; 73(11): 1081-1084, 2018 11.
Article in English | MEDLINE | ID: mdl-29574419

ABSTRACT

Acute lung injury is a neutrophil-dominant, life-threatening disease without effective therapies and better understanding of the pathophysiological mechanisms involved is an urgent need. Here we show that interleukin (IL)-22 is produced from innate lymphoid cells (ILC) and is responsible for suppression of experimental lung neutrophilic inflammation. Blocking prostaglandin E2 (PGE2) synthesis reduces lung ILCs and IL-22 production, resulting in exacerbation of lung neutrophilic inflammation. In contrast, activation of the PGE2 receptor EP4 prevents acute lung inflammation. We thus demonstrate a mechanism for production of innate IL-22 in the lung during acute injury, highlighting potential therapeutic strategies for control of lung neutrophilic inflammation by targeting the PGE2/ILC/IL-22 axis.


Subject(s)
Dinoprostone/pharmacology , Immunity, Innate/drug effects , Interleukins/biosynthesis , Lymphocytes/metabolism , Pneumonia/prevention & control , Animals , Disease Models, Animal , Lymphocytes/drug effects , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Interleukin-22
12.
Thorax ; 73(2): 134-144, 2018 02.
Article in English | MEDLINE | ID: mdl-28916704

ABSTRACT

BACKGROUND: Cystic fibrosis (CF) lung disease is defined by large numbers of neutrophils and associated damaging products in the airway. Delayed neutrophil apoptosis is described in CF although it is unclear whether this is a primary neutrophil defect or a response to chronic inflammation. Increased levels of neutrophil extracellular traps (NETs) have been measured in CF and we aimed to investigate the causal relationship between these phenomena and their potential to serve as a driver of inflammation. We hypothesised that the delay in apoptosis in CF is a primary defect and preferentially allows CF neutrophils to form NETs, contributing to inflammation. METHODS: Blood neutrophils were isolated from patients with CF, CF pigs and appropriate controls. Neutrophils were also obtained from patients with CF before and after commencing ivacaftor. Apoptosis was assessed by morphology and flow cytometry. NET formation was determined by fluorescent microscopy and DNA release assays. NET interaction with macrophages was examined by measuring cytokine generation with ELISA and qRT-PCR. RESULTS: CF neutrophils live longer due to decreased apoptosis. This was observed in both cystic fibrosis transmembrane conductance regulator (CFTR) null piglets and patients with CF, and furthermore was reversed by ivacaftor (CFTR potentiator) in patients with gating (G551D) mutations. CF neutrophils formed more NETs and this was reversed by cyclin-dependent kinase inhibitor exposure. NETs provided a proinflammatory stimulus to macrophages, which was enhanced in CF. CONCLUSIONS: CF neutrophils have a prosurvival phenotype that is associated with an absence of CFTR function and allows increased NET production, which can in turn induce inflammation. Augmenting neutrophil apoptosis in CF may allow more appropriate neutrophil disposal, decreasing NET formation and thus inflammation.


Subject(s)
Apoptosis/physiology , Cystic Fibrosis/pathology , Extracellular Traps , Neutrophils/physiology , Adult , Animals , Case-Control Studies , Cell Survival , Cystic Fibrosis/blood , Cystic Fibrosis/immunology , Humans , Inflammation , Swine , Time Factors
13.
Br Med Bull ; 128(1): 5-14, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30137312

ABSTRACT

Introduction: It has been known for some time that neutrophils are present in the tumour microenvironment, but only recently have their roles been explored. Sources of data: Comprehensive literature search of neutrophils and cancer (PubMed, Google Scholar and CrossRef) for key articles (systematic reviews, meta-analyses, primary research). References from these articles cross-checked for additional relevant studies. Areas of agreement: Neutrophils are a heterogeneous population with both pro- and antitumour roles, and display plasticity. Several neutrophil subpopulations have been identified, defined by a combination of features (density, maturity, surface markers, morphology and anatomical site). Areas of controversy: Limitations in translating murine tumour models to human pathology and paucity of human data. Consensus in defining human neutrophil subpopulations. Growing points: Neutrophils as therapeutic targets and as possible playmakers in the biological response to newer targeted cancer drugs. Areas timely for developing research: Understanding the metabolic programming of neutrophils in the tumour microenvironment.


Subject(s)
Neoplasms/immunology , Neutrophils/pathology , Tumor Microenvironment/immunology , Humans , Immunity, Cellular/physiology , Neoplasms/pathology , Neutrophils/immunology , Signal Transduction
14.
Am J Respir Crit Care Med ; 196(7): 845-855, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28557543

ABSTRACT

RATIONALE: Chronic obstructive pulmonary disease (COPD) is characterized by impaired clearance of pulmonary bacteria. OBJECTIVES: The effect of COPD on alveolar macrophage (AM) microbicidal responses was investigated. METHODS: AMs were obtained from bronchoalveolar lavage from healthy donors or patients with COPD and challenged with opsonized serotype 14 Streptococcus pneumoniae. Cells were assessed for apoptosis, bactericidal activity, and mitochondrial reactive oxygen species (mROS) production. A transgenic mouse line in which the CD68 promoter ensures macrophage-specific expression of human induced myeloid leukemia cell differentiation protein Mcl-1 (CD68.hMcl-1) was used to model the molecular aspects of COPD. MEASUREMENTS AND MAIN RESULTS: COPD AMs had elevated levels of Mcl-1, an antiapoptotic B-cell lymphoma 2 family member, with selective reduction of delayed intracellular bacterial killing. CD68.hMcl-1 AMs phenocopied the microbicidal defect because transgenic mice demonstrated impaired clearance of pulmonary bacteria and increased neutrophilic inflammation. Murine bone marrow-derived macrophages and human monocyte-derived macrophages generated mROS in response to pneumococci, which colocalized with bacteria and phagolysosomes to enhance bacterial killing. The Mcl-1 transgene increased oxygen consumption rates and mROS expression in mock-infected bone marrow-derived macrophages but reduced caspase-dependent mROS production after pneumococcal challenge. COPD AMs also increased basal mROS expression, but they failed to increase production after pneumococcal challenge, in keeping with reduced intracellular bacterial killing. The defect in COPD AM intracellular killing was associated with a reduced ratio of mROS/superoxide dismutase 2. CONCLUSIONS: Up-regulation of Mcl-1 and chronic adaption to oxidative stress alter mitochondrial metabolism and microbicidal function, reducing the delayed phase of intracellular bacterial clearance in COPD.


Subject(s)
Anti-Infective Agents/pharmacology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Animals , Blotting, Western , Bronchoalveolar Lavage , Disease Models, Animal , Flow Cytometry , Humans , Mice , Mice, Transgenic , Oxidative Stress/drug effects , Pulmonary Disease, Chronic Obstructive/physiopathology
15.
Nat Rev Immunol ; 7(1): 77-82, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17186032

ABSTRACT

Although there is overwhelming pressure from funding agencies and the general public for scientists to bridge basic and translational studies, the fact remains that there are significant hurdles to overcome in order to achieve this goal. The purpose of this Opinion article is to examine the nature of these hurdles and to provide food for thought on the main obstacles that impede this process.


Subject(s)
Biomedical Research/trends , Animals , Biomedical Research/ethics , Biomedical Research/legislation & jurisprudence , Disease Models, Animal , Humans
16.
Blood ; 123(18): 2753-4, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24786455

ABSTRACT

In this issue of Blood, Jun et al, through the study of neutrophils deficient in the glucose-6-phosphate transporter, describe a novel role for the peroxisome proliferator-activated receptor-γ (PPARG) pathway in the regulation of key neutrophil functions and link this to concomitant hypoxia-inducible factor (HIF) 1α stabilization.


Subject(s)
Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/metabolism , Neutrophils/metabolism , Humans
17.
Blood ; 123(3): 366-76, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24196071

ABSTRACT

Neutrophil lifespan and function are regulated by hypoxia via components of the hypoxia inducible factor (HIF)/von Hippel Lindau/hydroxylase pathway, including specific roles for HIF-1α and prolyl hydroxylase-3. HIF-2α has both distinct and overlapping biological roles with HIF-1α and has not previously been studied in the context of neutrophil biology. We investigated the role of HIF-2α in regulating key neutrophil functions. Human and murine peripheral blood neutrophils expressed HIF-2α, with expression up-regulated by acute and chronic inflammatory stimuli and in disease-associated inflammatory neutrophil. HIF2A gain-of-function mutations resulted in a reduction in neutrophil apoptosis both ex vivo, through the study of patient cells, and in vivo in a zebrafish tail injury model. In contrast, HIF-2α-deficient murine inflammatory neutrophils displayed increased sensitivity to nitrosative stress induced apoptosis ex vivo and increased neutrophil apoptosis in vivo, resulting in a reduction in neutrophilic inflammation and reduced tissue injury. Expression of HIF-2α was temporally dissociated from HIF-1α in vivo and predominated in the resolution phase of inflammation. These data support a critical and selective role for HIF-2α in persistence of neutrophilic inflammation and provide a platform to dissect the therapeutic utility of targeting HIF-2α in chronic inflammatory diseases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Inflammation , Neutrophils/metabolism , Animals , Apoptosis , Cell Hypoxia , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Muramidase , Neutrophils/cytology , Phagocytosis , Phenotype , RNA/metabolism , Respiratory Burst , Zebrafish
18.
J Immunol ; 192(4): 1796-805, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24431232

ABSTRACT

The inflammatory response is integral to maintaining health by functioning to resist microbial infection and repair tissue damage. Large numbers of neutrophils are recruited to inflammatory sites to neutralize invading bacteria through phagocytosis and the release of proteases and reactive oxygen species into the extracellular environment. Removal of the original inflammatory stimulus must be accompanied by resolution of the inflammatory response, including neutrophil clearance, to prevent inadvertent tissue damage. Neutrophil apoptosis and its temporary inhibition by survival signals provides a target for anti-inflammatory therapeutics, making it essential to better understand this process. GM-CSF, a neutrophil survival factor, causes a significant increase in mRNA levels for the known anti-apoptotic protein serum and glucocorticoid-regulated kinase 1 (SGK1). We have characterized the expression patterns and regulation of SGK family members in human neutrophils and shown that inhibition of SGK activity completely abrogates the antiapoptotic effect of GM-CSF. Using a transgenic zebrafish model, we have disrupted sgk1 gene function and shown this specifically delays inflammation resolution, without altering neutrophil recruitment to inflammatory sites in vivo. These data suggest SGK1 plays a key role in regulating neutrophil survival signaling and thus may prove a valuable therapeutic target for the treatment of inflammatory disease.


Subject(s)
Apoptosis/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immediate-Early Proteins/metabolism , Inflammation/immunology , Neutrophils/immunology , Protein Serine-Threonine Kinases/metabolism , Animals , Animals, Genetically Modified , Benzoates/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Movement/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/genetics , Morpholinos/genetics , Neutrophils/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/biosynthesis , Zebrafish/genetics
19.
PLoS Pathog ; 8(7): e1002814, 2012.
Article in English | MEDLINE | ID: mdl-22829769

ABSTRACT

Monocytes and T-cells are critical to the host response to acute bacterial infection but monocytes are primarily viewed as amplifying the inflammatory signal. The mechanisms of cell death regulating T-cell numbers at sites of infection are incompletely characterized. T-cell death in cultures of peripheral blood mononuclear cells (PBMC) showed 'classic' features of apoptosis following exposure to pneumococci. Conversely, purified CD3(+) T-cells cultured with pneumococci demonstrated necrosis with membrane permeabilization. The death of purified CD3(+) T-cells was not inhibited by necrostatin, but required the bacterial toxin pneumolysin. Apoptosis of CD3(+) T-cells in PBMC cultures required 'classical' CD14(+) monocytes, which enhanced T-cell activation. CD3(+) T-cell death was enhanced in HIV-seropositive individuals. Monocyte-mediated CD3(+) T-cell apoptotic death was Fas-dependent both in vitro and in vivo. In the early stages of the T-cell dependent host response to pneumococci reduced Fas ligand mediated T-cell apoptosis was associated with decreased bacterial clearance in the lung and increased bacteremia. In summary monocytes converted pathogen-associated necrosis into Fas-dependent apoptosis and regulated levels of activated T-cells at sites of acute bacterial infection. These changes were associated with enhanced bacterial clearance in the lung and reduced levels of invasive pneumococcal disease.


Subject(s)
Apoptosis , Fas Ligand Protein/metabolism , Monocytes/immunology , Pneumococcal Infections/immunology , T-Lymphocytes/physiology , Animals , Bacteremia , Bacterial Proteins , CD3 Complex/biosynthesis , Cells, Cultured , HIV Infections/immunology , HIV-1/immunology , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Lipopolysaccharide Receptors/biosynthesis , Lung/microbiology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/microbiology , Necrosis , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity , Streptolysins , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
20.
J Immunol ; 189(4): 1955-65, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22786772

ABSTRACT

Hypoxia and HIFs (HIF-1α and HIF-2α) modulate innate immune responses in the setting of systemic inflammatory responses and sepsis. The HIF prolyl hydroxylase enzymes PHD1, PHD2 and PHD3 regulate the mammalian adaptive response to hypoxia; however, their significance in the innate immune response has not been elucidated. We demonstrate in this study that deficiency of PHD3 (PHD3(-/-)) specifically shortens the survival of mice subjected to various models of abdominal sepsis because of an overwhelming innate immune response, leading to premature organ dysfunction. By contrast, this phenotype was absent in mice deficient for PHD1 (PHD1(-/-)) or PHD2 (PHD2(+/-)). In vivo, plasma levels of proinflammatory cytokines were enhanced, and recruitment of macrophages to internal organs was increased in septic PHD3-deficient mice. Reciprocal bone marrow transplantation in sublethally irradiated mice revealed that enhanced susceptibility of PHD3-deficient mice to sepsis-related lethality was specifically caused by loss of PHD3 in myeloid cells. Several in vitro assays revealed enhanced cytokine production, migration, phagocytic capacity, and proinflammatory activation of PHD3-deficient macrophages. Increased proinflammatory activity of PHD3-deficient macrophages occurred concomitantly with enhanced HIF-1α protein stabilization and increased NF-κB activity, and interference with the expression of HIF-1α or the canonical NF-κB pathway blunted their proinflammatory phenotype. It is concluded that impairment of PHD3 enzyme function aggravates the clinical course of abdominal sepsis via HIF-1α- and NF-κB-mediated enhancement of the innate immune response.


Subject(s)
Immunity, Innate/immunology , Macrophages/immunology , Procollagen-Proline Dioxygenase/immunology , Sepsis/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Macrophages/metabolism , Mice , Mice, Knockout , NF-kappa B/immunology , NF-kappa B/metabolism , Procollagen-Proline Dioxygenase/metabolism , Real-Time Polymerase Chain Reaction , Sepsis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL