Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Leukoc Biol ; 105(6): 1183-1194, 2019 06.
Article in English | MEDLINE | ID: mdl-30942918

ABSTRACT

GM-CSF is important in regulating acute, persistent neutrophilic inflammation in certain settings, including lung injury. Ligand binding induces rapid internalization of the GM-CSF receptor (GM-CSFRα) complex, a process essential for signaling. Whereas GM-CSF controls many aspects of neutrophil biology, regulation of GM-CSFRα expression is poorly understood, particularly the role of GM-CSFRα in ligand clearance and whether signaling is sustained despite major down-regulation of GM-CSFRα surface expression. We established a quantitative assay of GM-CSFRα surface expression and used this, together with selective anti-GM-CSFR antibodies, to define GM-CSFRα kinetics in human neutrophils, and in murine blood and alveolar neutrophils in a lung injury model. Despite rapid sustained ligand-induced GM-CSFRα loss from the neutrophil surface, which persisted even following ligand removal, pro-survival effects of GM-CSF required ongoing ligand-receptor interaction. Neutrophils recruited to the lungs following LPS challenge showed initially high mGM-CSFRα expression, which along with mGM-CSFRß declined over 24 hr; this was associated with a transient increase in bronchoalveolar lavage fluid (BALF) mGM-CSF concentration. Treating mice in an LPS challenge model with CAM-3003, an anti-mGM-CSFRα mAb, inhibited inflammatory cell influx into the lung and maintained the level of BALF mGM-CSF. Consistent with neutrophil consumption of GM-CSF, human neutrophils depleted exogenous GM-CSF, independent of protease activity. These data show that loss of membrane GM-CSFRα following GM-CSF exposure does not preclude sustained GM-CSF/GM-CSFRα signaling and that this receptor plays a key role in ligand clearance. Hence neutrophilic activation via GM-CSFR may play an important role in neutrophilic lung inflammation even in the absence of high GM-CSF levels or GM-CSFRα expression.


Subject(s)
Acute Lung Injury/immunology , Gene Expression Regulation/immunology , Neutrophils/immunology , Pulmonary Alveoli/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Adult , Animals , Cell Line, Tumor , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/immunology , Disease Models, Animal , Female , Humans , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutrophils/pathology , Pulmonary Alveoli/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Time Factors
2.
Thorax ; 73(10): 918-925, 2018 10.
Article in English | MEDLINE | ID: mdl-30064991

ABSTRACT

BACKGROUND: Critically ill patients with impaired neutrophil phagocytosis have significantly increased risk of nosocomial infection. Granulocyte-macrophage colony-stimulating factor (GM-CSF) improves phagocytosis by neutrophils ex vivo. This study tested the hypothesis that GM-CSF improves neutrophil phagocytosis in critically ill patients in whom phagocytosis is known to be impaired. METHODS: This was a multicentre, phase IIa randomised, placebo-controlled clinical trial. Using a personalised medicine approach, only critically ill patients with impaired neutrophil phagocytosis were included. Patients were randomised 1:1 to subcutaneous GM-CSF (3 µg/kg/day) or placebo, once daily for 4 days. The primary outcome measure was neutrophil phagocytosis 2 days after initiation of GM-CSF. Secondary outcomes included neutrophil phagocytosis over time, neutrophil functions other than phagocytosis, monocyte HLA-DR expression and safety. RESULTS: Thirty-eight patients were recruited from five intensive care units (17 randomised to GM-CSF). Mean neutrophil phagocytosis at day 2 was 57.2% (SD 13.2%) in the GM-CSF group and 49.8% (13.4%) in the placebo group, p=0.73. The proportion of patients with neutrophil phagocytosis≥50% at day 2, and monocyte HLA-DR, appeared significantly higher in the GM-CSF group. Neutrophil functions other than phagocytosis did not appear significantly different between the groups. The most common adverse event associated with GM-CSF was fever. CONCLUSIONS: GM-CSF did not improve mean neutrophil phagocytosis at day 2, but was safe and appeared to increase the proportion of patients with adequate phagocytosis. The study suggests proof of principle for a pharmacological effect on neutrophil function in a subset of critically ill patients.


Subject(s)
Critical Illness/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Neutrophils/drug effects , Phagocytosis/drug effects , Adult , Aged , Aged, 80 and over , Female , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , HLA-DR Antigens/metabolism , Humans , Male , Middle Aged , Monocytes/metabolism , Neutrophils/physiology , Treatment Outcome
3.
Front Immunol ; 8: 1185, 2017.
Article in English | MEDLINE | ID: mdl-28993776

ABSTRACT

Human cytomegalovirus (HCMV) is a major cause of viral disease in the young and the immune-suppressed. At sites of infection, HCMV recruits the neutrophil, a cell with a key role in orchestrating the initial immune response. Herein, we report a profound survival response in human neutrophils exposed to the clinical HCMV isolate Merlin, but not evident with the attenuated strain AD169, through suppression of apoptosis. The initial survival event, which is independent of viral gene expression and involves activation of the ERK/MAPK and NF-κB pathways, is augmented by HCMV-stimulated release of a secretory cytokine profile that further prolongs neutrophil lifespan. As aberrant neutrophil survival contributes to tissue damage, we predict that this may be relevant to the immune pathology of HCMV, and the presence of this effect in clinical HCMV strains and its absence in attenuated strains implies a beneficial effect to the virus in pathogenesis and/or dissemination. In addition, we show that HCMV-exposed neutrophils release factors that enhance monocyte recruitment and drive monocyte differentiation to a HCMV-permissive phenotype in an IL-6-dependent manner, thus providing an ideal vehicle for viral dissemination. This study increases understanding of HCMV-neutrophil interactions, highlighting the potential role of neutrophil recruitment as a virulence mechanism to promote HCMV pathology in the host and influence the dissemination of HCMV infection. Targeting these mechanisms may lead to new antiviral strategies aimed at limiting host damage and inhibiting viral spread.

4.
J Clin Invest ; 127(9): 3521-3526, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28783043

ABSTRACT

Mutations in the human NBEAL2 gene cause gray platelet syndrome (GPS), a bleeding diathesis characterized by a lack of α granules in platelets. The functions of the NBEAL2 protein have not been explored outside platelet biology, but there are reports of increased frequency of infection and abnormal neutrophil morphology in patients with GPS. We therefore investigated the role of NBEAL2 in immunity by analyzing the phenotype of Nbeal2-deficient mice. We found profound abnormalities in the Nbeal2-deficient immune system, particularly in the function of neutrophils and NK cells. Phenotyping of Nbeal2-deficient neutrophils showed a severe reduction in granule contents across all granule subsets. Despite this, Nbeal2-deficient neutrophils had an enhanced phagocyte respiratory burst relative to Nbeal2-expressing neutrophils. This respiratory burst was associated with increased expression of cytosolic components of the NADPH oxidase complex. Nbeal2-deficient NK cells were also dysfunctional and showed reduced degranulation. These abnormalities were associated with increased susceptibility to both bacterial (Staphylococcus aureus) and viral (murine CMV) infection in vivo. These results define an essential role for NBEAL2 in mammalian immunity.


Subject(s)
Blood Proteins/metabolism , Killer Cells, Natural/metabolism , Mutation , Neutrophils/metabolism , Animals , Blood Platelets/metabolism , Blood Proteins/genetics , Cytosol/metabolism , Gray Platelet Syndrome/genetics , Humans , Immune System , Immunophenotyping , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Phenotype , Staphylococcal Infections/metabolism , Staphylococcus aureus
5.
Eur J Clin Invest ; 47(10): 736-745, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28796316

ABSTRACT

BACKGROUND: Decreases in circulating neutrophils (polymorphonuclear leucocytes, PMNs) have been reported in patients treated with the anti-interleukin-6 receptor (IL-6R) antibody tocilizumab (TCZ); the mechanism for this is unclear. We hypothesize that TCZ reduces circulating neutrophils by affecting margination and/or bone marrow trafficking without affecting neutrophil function or apoptosis. MATERIALS AND METHODS: Eighteen healthy subjects were randomized to single intravenous dose of TCZ 8 mg/kg (n = 12) or placebo (n = 6) on day 0. On day 4, each subject had autologous indium-111-labelled neutrophils re-injected, and their kinetics quantified with longitudinal profiling in a whole body gamma-counter. TCZ-treated subjects were divided into two groups according to the extent of reduction in neutrophil count. RESULTS: Mean day 4 neutrophil counts, as % baseline, were 101·9%, 68·3% and 44·2% in the placebo, TCZ-PMN-'high' and TCZ-PMN-'low' groups, respectively (P < 0·001). Following TCZ, neutrophil function, activation and apoptosis ex vivo were all unaffected. In vivo, there were no differences in early blood recovery or margination to liver/spleen and bone marrow; however, later neutrophil re-distribution to bone marrow was markedly reduced in the TCZ-PMN-low group (peak pelvic count as % day 4 count on: day 5, 188% placebo vs. 127% TCZ-PMN-low, P < 0·001; day 10, 180% placebo vs. 132% TCZ-PMN-low, P < 0·01), with a trend towards higher liver/spleen neutrophil retention. CONCLUSIONS: We have demonstrated for the first time in humans that IL-6R blockade affects neutrophil trafficking to the bone marrow without influencing neutrophil functional capacity.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Apoptosis/drug effects , Cell Movement/drug effects , Neutrophils/drug effects , Adolescent , Adult , Aged , Antibodies, Monoclonal, Humanized/pharmacokinetics , Healthy Volunteers , Humans , Infusions, Intravenous , Kinetics , Male , Middle Aged , Neutrophils/cytology , Neutrophils/physiology , Reference Values , Sensitivity and Specificity , Single-Blind Method , Young Adult
6.
J Exp Med ; 214(4): 1111-1128, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28351984

ABSTRACT

The phagocyte respiratory burst is crucial for innate immunity. The transfer of electrons to oxygen is mediated by a membrane-bound heterodimer, comprising gp91phox and p22phox subunits. Deficiency of either subunit leads to severe immunodeficiency. We describe Eros (essential for reactive oxygen species), a protein encoded by the previously undefined mouse gene bc017643, and show that it is essential for host defense via the phagocyte NAPDH oxidase. Eros is required for expression of the NADPH oxidase components, gp91phox and p22phox Consequently, Eros-deficient mice quickly succumb to infection. Eros also contributes to the formation of neutrophil extracellular traps (NETS) and impacts on the immune response to melanoma metastases. Eros is an ortholog of the plant protein Ycf4, which is necessary for expression of proteins of the photosynthetic photosystem 1 complex, itself also an NADPH oxio-reductase. We thus describe the key role of the previously uncharacterized protein Eros in host defense.


Subject(s)
Membrane Proteins/physiology , Phagocytes/physiology , Reactive Oxygen Species/metabolism , Respiratory Burst/physiology , Animals , Cytochrome b Group/analysis , Cytochrome b Group/physiology , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Immunity, Innate , Macrophages/immunology , Membrane Glycoproteins/analysis , Membrane Glycoproteins/physiology , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/analysis , NADPH Oxidases/physiology , Neutrophils/immunology , Phagocytosis
7.
Am J Respir Cell Mol Biol ; 56(4): 423-431, 2017 04.
Article in English | MEDLINE | ID: mdl-27854516

ABSTRACT

The release of neutrophil extracellular traps (NETs) is a major immune mechanism intended to capture pathogens. These histone- and protease-coated DNA structures are released by neutrophils in response to a variety of stimuli, including respiratory pathogens, and have been identified in the airways of patients with respiratory infection, cystic fibrosis, acute lung injury, primary graft dysfunction, and chronic obstructive pulmonary disease. NET production has been demonstrated in the lungs of mice infected with Staphylococcus aureus, Klebsiella pneumoniae, and Aspergillus fumigatus. Since the discovery of NETs over a decade ago, evidence that "NET evasion" might act as an immune protection strategy among respiratory pathogens, including group A Streptococcus, Bordetella pertussis, and Haemophilus influenzae, has been growing, with the majority of these studies being published in the past 2 years. Evasion strategies fall into three main categories: inhibition of NET release by down-regulating host inflammatory responses; degradation of NETs using pathogen-derived DNases; and resistance to the microbicidal components of NETs, which involves a variety of mechanisms, including encapsulation. Hence, the evasion of NETs appears to be a widespread strategy to allow pathogen proliferation and dissemination, and is currently a topic of intense research interest. This article outlines the evidence supporting the three main strategies of NET evasion-inhibition, degradation, and resistance-with particular reference to common respiratory pathogens.


Subject(s)
Extracellular Traps/immunology , Immune Evasion , Lung/microbiology , Lung/virology , Animals , Humans , Models, Immunological
8.
Thorax ; 71(11): 1030-1038, 2016 11.
Article in English | MEDLINE | ID: mdl-27581620

ABSTRACT

BACKGROUND: The inflamed bronchial mucosal surface is a profoundly hypoxic environment. Neutrophilic airway inflammation and neutrophil-derived proteases have been linked to disease progression in conditions such as COPD and cystic fibrosis, but the effects of hypoxia on potentially harmful neutrophil functional responses such as degranulation are unknown. METHODS AND RESULTS: Following exposure to hypoxia (0.8% oxygen, 3 kPa for 4 h), neutrophils stimulated with inflammatory agonists (granulocyte-macrophage colony stimulating factor or platelet-activating factor and formylated peptide) displayed a markedly augmented (twofold to sixfold) release of azurophilic (neutrophil elastase, myeloperoxidase), specific (lactoferrin) and gelatinase (matrix metalloproteinase-9) granule contents. Neutrophil supernatants derived under hypoxic but not normoxic conditions induced extensive airway epithelial cell detachment and death, which was prevented by coincubation with the antiprotease α-1 antitrypsin; both normoxic and hypoxic supernatants impaired ciliary function. Surprisingly, the hypoxic upregulation of neutrophil degranulation was not dependent on hypoxia-inducible factor (HIF), nor was it fully reversed by inhibition of phospholipase C signalling. Hypoxia augmented the resting and cytokine-stimulated phosphorylation of AKT, and inhibition of phosphoinositide 3-kinase (PI3K)γ (but not other PI3K isoforms) prevented the hypoxic upregulation of neutrophil elastase release. CONCLUSION: Hypoxia augments neutrophil degranulation and confers enhanced potential for damage to respiratory airway epithelial cells in a HIF-independent but PI3Kγ-dependent fashion.


Subject(s)
Cell Degranulation/drug effects , Hypoxia/metabolism , Hypoxia/physiopathology , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Apoptosis , Blotting, Western , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunohistochemistry , Lactoferrin/metabolism , Leukocyte Elastase/metabolism , Matrix Metalloproteinase 9/metabolism , Microscopy, Electron , Peroxidase/metabolism , Platelet Activating Factor/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Formyl Peptide/metabolism , Signal Transduction , Up-Regulation
9.
Am J Respir Crit Care Med ; 194(8): 961-973, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27064380

ABSTRACT

RATIONALE: Acute respiratory distress syndrome is refractory to pharmacological intervention. Inappropriate activation of alveolar neutrophils is believed to underpin this disease's complex pathophysiology, yet these cells have been little studied. OBJECTIVES: To examine the functional and transcriptional profiles of patient blood and alveolar neutrophils compared with healthy volunteer cells, and to define their sensitivity to phosphoinositide 3-kinase inhibition. METHODS: Twenty-three ventilated patients underwent bronchoalveolar lavage. Alveolar and blood neutrophil apoptosis, phagocytosis, and adhesion molecules were quantified by flow cytometry, and oxidase responses were quantified by chemiluminescence. Cytokine and transcriptional profiling were used in multiplex and GeneChip arrays. MEASUREMENTS AND MAIN RESULTS: Patient blood and alveolar neutrophils were distinct from healthy circulating cells, with increased CD11b and reduced CD62L expression, delayed constitutive apoptosis, and primed oxidase responses. Incubating control cells with disease bronchoalveolar lavage recapitulated the aberrant functional phenotype, and this could be reversed by phosphoinositide 3-kinase inhibitors. In contrast, the prosurvival phenotype of patient cells was resistant to phosphoinositide 3-kinase inhibition. RNA transcriptomic analysis revealed modified immune, cytoskeletal, and cell death pathways in patient cells, aligning closely to sepsis and burns datasets but not to phosphoinositide 3-kinase signatures. CONCLUSIONS: Acute respiratory distress syndrome blood and alveolar neutrophils display a distinct primed prosurvival profile and transcriptional signature. The enhanced respiratory burst was phosphoinositide 3-kinase-dependent but delayed apoptosis and the altered transcriptional profile were not. These unexpected findings cast doubt over the utility of phosphoinositide 3-kinase inhibition in acute respiratory distress syndrome and highlight the importance of evaluating novel therapeutic strategies in patient-derived cells.


Subject(s)
Neutrophils/drug effects , Phosphoinositide-3 Kinase Inhibitors , Respiratory Distress Syndrome/immunology , Bronchoalveolar Lavage Fluid/cytology , CD11b Antigen/metabolism , Cytokines/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Humans , L-Selectin/metabolism , Male , Middle Aged , Neutrophils/metabolism , Neutrophils/physiology , Oligonucleotide Array Sequence Analysis , Phenotype , Respiratory Distress Syndrome/drug therapy
10.
J Immunol ; 195(7): 3149-59, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26311901

ABSTRACT

We identified a novel, evolutionarily conserved receptor encoded within the human leukocyte receptor complex and syntenic region of mouse chromosome 7, named T cell-interacting, activating receptor on myeloid cells-1 (TARM1). The transmembrane region of TARM1 contained a conserved arginine residue, consistent with association with a signaling adaptor. TARM1 associated with the ITAM adaptor FcRγ but not with DAP10 or DAP12. In healthy mice, TARM1 is constitutively expressed on the cell surface of mature and immature CD11b(+)Gr-1(+) neutrophils within the bone marrow. Following i.p. LPS treatment or systemic bacterial challenge, TARM1 expression was upregulated by neutrophils and inflammatory monocytes and TARM1(+) cells were rapidly recruited to sites of inflammation. TARM1 expression was also upregulated by bone marrow-derived macrophages and dendritic cells following stimulation with TLR agonists in vitro. Ligation of TARM1 receptor in the presence of TLR ligands, such as LPS, enhanced the secretion of proinflammatory cytokines by macrophages and primary mouse neutrophils, whereas TARM1 stimulation alone had no effect. Finally, an immobilized TARM1-Fc fusion protein suppressed CD4(+) T cell activation and proliferation in vitro. These results suggest that a putative T cell ligand can interact with TARM1 receptor, resulting in bidirectional signaling and raising the T cell activation threshold while costimulating the release of proinflammatory cytokines by macrophages and neutrophils.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Macrophages/immunology , Neutrophils/immunology , Receptors, Immunologic/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Granulocytes/immunology , Granulocytes/metabolism , HEK293 Cells , HLA Antigens/genetics , Humans , Inflammation/immunology , Ligands , Lipopolysaccharides/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Molecular Sequence Data , Neutrophils/metabolism , Protein Transport/immunology , Receptors, Immunologic/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction/immunology
11.
J Infect Dis ; 211(12): 1936-42, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25552371

ABSTRACT

Human cytomegalovirus (HCMV) causes significant morbidity in the immunocompromised host. Following primary infection, the virus establishes latent infection in progenitor cells of the myeloid lineage. These cells exhibit limited viral gene transcription and no evidence of de novo virion production. It is well recognized that differentiation of latently infected myeloid progenitor cells to dendritic or macrophage-like cells permits viral reactivation in vitro. This has been used to support the concept that viral reactivation in HCMV carriers routinely occurs from such terminally differentiated myeloid cells in vivo. However, to date this has not been shown for in vivo-differentiated macrophages. This study is the first to demonstrate that alveolar macrophages from HCMV carriers express immediate early lytic genes and produce infectious virus. This supports the view, until now based on in vitro data, that terminally differentiated myeloid cells in vivo are sites of HCMV reactivation and potential centers of viral dissemination in latently infected individuals with no evidence of virus disease or dissemination.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Macrophages, Alveolar/virology , Virus Activation , Humans
12.
PLoS One ; 9(12): e116055, 2014.
Article in English | MEDLINE | ID: mdl-25541728

ABSTRACT

Human rhinoviruses (HRV) are a major cause of exacerbations of airways disease. Aspects of cell signalling responses to HRV infection remain unclear, particularly with regard to signalling via PI3K, and the PI3K-dependent pathway, autophagy. We investigated the roles of PI3K and autophagy in the responses of epithelial cells to major and minor group HRV infection. The PI3K inhibitor 3-MA, commonly used to inhibit autophagy, markedly reduced HRV-induced cytokine induction. Further investigation of potential targets of 3-MA and comparison of results using this inhibitor to a panel of general and class I-selective PI3K inhibitors showed that several PI3Ks cooperatively regulate responses to HRV. Targeting by siRNA of the autophagy proteins Beclin-1, Atg7, LC3, alone or in combination, or targeting of the autophagy-specific class III PI3K had at most only modest effects on HRV-induced cell signalling as judged by induction of proinflammatory cytokine production. Our data indicate that PI3K and mTOR are involved in induction of proinflammatory cytokines after HRV infection, and that autophagy has little role in the cytokine response to HRV or control of HRV replication.


Subject(s)
Autophagy , Epithelial Cells/microbiology , Phosphoinositide-3 Kinase Inhibitors , Picornaviridae Infections/enzymology , Picornaviridae Infections/physiopathology , Protein Kinase Inhibitors/pharmacology , Rhinovirus/physiology , Cell Line , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Host-Pathogen Interactions , Humans , Phosphatidylinositol 3-Kinases/immunology , Picornaviridae Infections/immunology , Signal Transduction , TOR Serine-Threonine Kinases/immunology
13.
Science ; 342(6160): 866-71, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24136356

ABSTRACT

Genetic mutations cause primary immunodeficiencies (PIDs) that predispose to infections. Here, we describe activated PI3K-δ syndrome (APDS), a PID associated with a dominant gain-of-function mutation in which lysine replaced glutamic acid at residue 1021 (E1021K) in the p110δ protein, the catalytic subunit of phosphoinositide 3-kinase δ (PI3Kδ), encoded by the PIK3CD gene. We found E1021K in 17 patients from seven unrelated families, but not among 3346 healthy subjects. APDS was characterized by recurrent respiratory infections, progressive airway damage, lymphopenia, increased circulating transitional B cells, increased immunoglobulin M, and reduced immunoglobulin G2 levels in serum and impaired vaccine responses. The E1021K mutation enhanced membrane association and kinase activity of p110δ. Patient-derived lymphocytes had increased levels of phosphatidylinositol 3,4,5-trisphosphate and phosphorylated AKT protein and were prone to activation-induced cell death. Selective p110δ inhibitors IC87114 and GS-1101 reduced the activity of the mutant enzyme in vitro, which suggested a therapeutic approach for patients with APDS.


Subject(s)
Genetic Predisposition to Disease , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Phosphatidylinositol 3-Kinases/genetics , Respiratory Tract Infections/genetics , Respiratory Tract Infections/pathology , Class I Phosphatidylinositol 3-Kinases , Humans , Immunologic Deficiency Syndromes/immunology , Lymphocytes/immunology , Mutation , Pedigree , Phosphatidylinositol Phosphates/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Respiratory Tract Infections/immunology
14.
PLoS One ; 7(9): e45933, 2012.
Article in English | MEDLINE | ID: mdl-23029326

ABSTRACT

We have investigated the contribution of individual phosphoinositide 3-kinase (PI3K) Class I isoforms to the regulation of neutrophil survival using (i) a panel of commercially available small molecule isoform-selective PI3K Class I inhibitors, (ii) novel inhibitors, which target single or multiple Class I isoforms (PI3Kα, PI3Kß, PI3Kδ, and PI3Kγ), and (iii) transgenic mice lacking functional PI3K isoforms (p110δ(KO)γ(KO) or p110γ(KO)). Our data suggest that there is considerable functional redundancy amongst Class I PI3Ks (both Class IA and Class IB) with regard to GM-CSF-mediated suppression of neutrophil apoptosis. Hence pharmacological inhibition of any 3 or more PI3K isoforms was required to block the GM-CSF survival response in human neutrophils, with inhibition of individual or any two isoforms having little or no effect. Likewise, isolated blood neutrophils derived from double knockout PI3K p110δ(KO)γ(KO) mice underwent normal time-dependent constitutive apoptosis and displayed identical GM-CSF mediated survival to wild type cells, but were sensitized to pharmacological inhibition of the remaining PI3K isoforms. Surprisingly, the pro-survival neutrophil phenotype observed in patients with an acute exacerbation of chronic obstructive pulmonary disease (COPD) was resilient to inactivation of the PI3K pathway.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Aged , Aged, 80 and over , Animals , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neutrophils/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/metabolism , Signal Transduction/drug effects
15.
Sci Signal ; 4(168): ra23, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21487106

ABSTRACT

Neutrophils are activated by immunoglobulin G (IgG)-containing immune complexes through receptors that recognize the Fc portion of IgG (FcγRs). Here, we used genetic and pharmacological approaches to define a selective role for the ß isoform of phosphoinositide 3-kinase (PI3Kß) in FcγR-dependent activation of mouse neutrophils by immune complexes of IgG and antigen immobilized on a plate surface. At low concentrations of immune complexes, loss of PI3Kß alone substantially inhibited the production of reactive oxygen species (ROS) by neutrophils, whereas at higher doses, similar suppression of ROS production was achieved only by targeting both PI3Kß and PI3Kδ, suggesting that this pathway displays stimulus strength-dependent redundancy. Activation of PI3Kß by immune complexes involved cooperation between FcγRs and BLT1, the receptor for the endogenous proinflammatory lipid leukotriene B4. Coincident activation by a tyrosine kinase-coupled receptor (FcγR) and a heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptor (BLT1) may provide a rationale for the preferential activation of the ß isoform of PI3K. PI3Kß-deficient mice were highly protected in an FcγR-dependent model of autoantibody-induced skin blistering and were partially protected in an FcγR-dependent model of inflammatory arthritis, whereas combined deficiency of PI3Kß and PI3Kδ resulted in near-complete protection in the latter case. These results define PI3Kß as a potential therapeutic target in inflammatory disease.


Subject(s)
Antigen-Antibody Complex/immunology , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Neutrophil Activation/immunology , Neutrophils/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Blotting, Western , CD2 Antigens/genetics , CD2 Antigens/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Enzyme Inhibitors/pharmacology , Female , Flow Cytometry , Gene Rearrangement, B-Lymphocyte/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Joining Region/genetics , Immunoglobulin Joining Region/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Knockout , Mice, Transgenic , Neutrophils/metabolism , Phosphoinositide-3 Kinase Inhibitors , Reactive Oxygen Species/metabolism , Receptors, IgG/metabolism , Receptors, Leukotriene B4/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
17.
Acupunct Med ; 26(3): 193-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18818566

ABSTRACT

We report a significant complication of acupuncture in a 50 year old woman who developed a pneumothorax shortly after receiving acupuncture needling to her scapulothoracic region in a lateral oblique direction. As acupuncture is increasingly being used in pain management, physicians need to be aware of its potential adverse effects. We discuss issues relating to appropriate counselling of patients receiving this form of therapy. The inner Bladder line should be needled obliquely towards the spine.


Subject(s)
Acupuncture Points , Acupuncture Therapy/adverse effects , Pneumothorax/etiology , Adult , Chest Pain/etiology , Dyspnea/etiology , Female , Humans , Middle Aged
19.
Age Ageing ; 34(5): 524-5, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16107463

ABSTRACT

Management of electrolyte abnormalities is challenging in older people as the sensation of thirst, renal function and hormonal modulators of the milieu interior are often impaired. Furthermore, the complex effects of ageing upon these homeostatic mechanisms are often superimposed upon a background of chronic disease, malnutrition and co-existent medications. Hyponatraemia is one of the commonest electrolyte abnormalities, occurring in approximately 7% of healthy elderly persons. Hyponatraemia may only come to light when some other ailment prompts investigations or hospital admission. Drug-induced hyponatraemia is common in older people and is most commonly associated with diuretics and SSRI/SNRI antidepressants, but has also been reported with a wide range of other drugs. We believe this is the first case report of hyponatraemia due to tolterodine.


Subject(s)
Benzhydryl Compounds/adverse effects , Cresols/adverse effects , Hyponatremia/chemically induced , Muscarinic Antagonists/adverse effects , Phenylpropanolamine/adverse effects , Aged , Female , Humans , Tolterodine Tartrate
SELECTION OF CITATIONS
SEARCH DETAIL
...