Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
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
2.
Am J Respir Crit Care Med ; 197(12): 1604-1615, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29365279

ABSTRACT

RATIONALE: People living with HIV are at significantly increased risk of invasive pneumococcal disease, despite long-term antiretroviral therapy (ART). The mechanism explaining this observation remains undefined. OBJECTIVES: To determine if apoptosis-associated microbicidal mechanisms, required to clear intracellular pneumococci that survive initial phagolysosomal killing, are perturbed. METHODS: Alveolar macrophages (AM) were obtained by BAL from healthy donors or HIV-1-seropositive donors on long-term ART with undetectable plasma viral load. Monocyte-derived macrophages (MDM) were obtained from healthy donors and infected with HIV-1BaL or treated with gp120. Macrophages were challenged with opsonized serotype 2 Streptococcus pneumoniae and assessed for apoptosis, bactericidal activity, protein expression, and mitochondrial reactive oxygen species (mROS). AM phenotyping, ultrasensitive HIV-1 RNA quantification, and gp120 measurement were also performed in BAL. MEASUREMENTS AND MAIN RESULTS: HIV-1BaL infection impaired apoptosis, induction of mROS, and pneumococcal killing by MDM. Apoptosis-associated pneumococcal killing was also reduced in AM from ART-treated HIV-1-seropositive donors. BAL fluid from these individuals demonstrated persistent lung CD8+ T lymphocytosis, and gp120 or HIV-1 RNA was also detected. Despite this, transcriptional activity in AM freshly isolated from people living with HIV was broadly similar to healthy volunteers. Instead, gp120 phenocopied the defect in pneumococcal killing in healthy MDM through post-translational modification of Mcl-1, preventing apoptosis induction, caspase activation, and increased mROS generation. Moreover, gp120 also inhibited mROS-dependent pneumococcal killing in MDM. CONCLUSIONS: Despite ART, HIV-1, via gp120, drives persisting innate immune defects in AM microbicidal mechanisms, enhancing susceptibility to pneumococcal disease.


Subject(s)
Anti-Retroviral Agents/adverse effects , Anti-Retroviral Agents/therapeutic use , Disease Resistance/immunology , HIV Infections/drug therapy , HIV Infections/immunology , Lung Diseases/immunology , Macrophages, Alveolar/immunology , Pneumococcal Infections/immunology , Adult , Female , HIV Envelope Protein gp120/blood , Humans , Lung Diseases/microbiology , Lung Diseases/physiopathology , Male , Middle Aged , Pneumococcal Infections/physiopathology
3.
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
4.
Cell Microbiol ; 18(1): 80-96, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26248337

ABSTRACT

Macrophages are critical effectors of the early innate response to bacteria in tissues. Phagocytosis and killing of bacteria are interrelated functions essential for bacterial clearance but the rate-limiting step when macrophages are challenged with large numbers of the major medical pathogen Staphylococcus aureus is unknown. We show that macrophages have a finite capacity for intracellular killing and fail to match sustained phagocytosis with sustained microbial killing when exposed to large inocula of S. aureus (Newman, SH1000 and USA300 strains). S. aureus ingestion by macrophages is associated with a rapid decline in bacterial viability immediately after phagocytosis. However, not all bacteria are killed in the phagolysosome, and we demonstrate reduced acidification of the phagolysosome, associated with failure of phagolysosomal maturation and reduced activation of cathepsin D. This results in accumulation of viable intracellular bacteria in macrophages. We show macrophages fail to engage apoptosis-associated bacterial killing. Ultittop mately macrophages with viable bacteria undergo cell lysis, and viable bacteria are released and can be internalized by other macrophages. We show that cycles of lysis and reuptake maintain a pool of viable intracellular bacteria over time when killing is overwhelmed and demonstrate intracellular persistence in alveolar macrophages in the lungs in a murine model.


Subject(s)
Macrophages/immunology , Macrophages/microbiology , Microbial Viability , Phagocytosis , Pneumonia, Staphylococcal/pathology , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology , Animals , Cell Survival , Disease Models, Animal , Mice
5.
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
6.
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
7.
PLoS Pathog ; 7(1): e1001262, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21298030

ABSTRACT

The bactericidal function of macrophages against pneumococci is enhanced by their apoptotic demise, which is controlled by the anti-apoptotic protein Mcl-1. Here, we show that lysosomal membrane permeabilization (LMP) and cytosolic translocation of activated cathepsin D occur prior to activation of a mitochondrial pathway of macrophage apoptosis. Pharmacological inhibition or knockout of cathepsin D during pneumococcal infection blocked macrophage apoptosis. As a result of cathepsin D activation, Mcl-1 interacted with its ubiquitin ligase Mule and expression declined. Inhibition of cathepsin D had no effect on early bacterial killing but inhibited the late phase of apoptosis-associated killing of pneumococci in vitro. Mice bearing a cathepsin D(-/-) hematopoietic system demonstrated reduced macrophage apoptosis in vivo, with decreased clearance of pneumococci and enhanced recruitment of neutrophils to control pulmonary infection. These findings establish an unexpected role for a cathepsin D-mediated lysosomal pathway of apoptosis in pulmonary host defense and underscore the importance of apoptosis-associated microbial killing to macrophage function.


Subject(s)
Apoptosis , Cathepsin D/metabolism , Macrophages/enzymology , Streptococcus pneumoniae/physiology , Animals , Bone Marrow Cells , Bone Marrow Transplantation , Cathepsin D/deficiency , Cathepsin D/genetics , Cell Line, Tumor , Cytosol/enzymology , Cytosol/microbiology , Female , Host-Pathogen Interactions , Humans , Intracellular Membranes/enzymology , Intracellular Membranes/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagosomes/enzymology , Phagosomes/microbiology , Streptococcus pneumoniae/pathogenicity
8.
Mol Cell Proteomics ; 10(6): M111.008193, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21474794

ABSTRACT

Macrophages are central effectors of innate immune responses to bacteria. We have investigated how activation of the abundant macrophage lysosomal protease, cathepsin D, regulates the macrophage proteome during killing of Streptococcus pneumoniae. Using the cathepsin D inhibitor pepstatin A, we demonstrate that cathepsin D differentially regulates multiple targets out of 679 proteins identified and quantified by eight-plex isobaric tag for relative and absolute quantitation. Our statistical analysis identified 18 differentially expressed proteins that passed all paired t-tests (α = 0.05). This dataset was enriched for proteins regulating the mitochondrial pathway of apoptosis or inhibiting competing death programs. Five proteins were selected for further analysis. Western blotting, followed by pharmacological inhibition or genetic manipulation of cathepsin D, verified cathepsin D-dependent regulation of these proteins, after exposure to S. pneumoniae. Superoxide dismutase-2 up-regulation was temporally related to increased reactive oxygen species generation. Gelsolin, a known regulator of mitochondrial outer membrane permeabilization, was down-regulated in association with cytochrome c release from mitochondria. Eukaryotic elongation factor (eEF2), a regulator of protein translation, was also down-regulated by cathepsin D. Using absence of the negative regulator of eEF2, eEF2 kinase, we confirm that eEF2 function is required to maintain expression of the anti-apoptotic protein Mcl-1, delaying macrophage apoptosis and confirm using a murine model that maintaining eEF2 function is associated with impaired macrophage apoptosis-associated killing of Streptococcus pneumoniae. These findings demonstrate that cathepsin D regulates multiple proteins controlling the mitochondrial pathway of macrophage apoptosis or competing death processes, facilitating intracellular bacterial killing.


Subject(s)
Cathepsin D/antagonists & inhibitors , Macrophages/physiology , Proteome/metabolism , Streptococcus pneumoniae/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Colony Count, Microbial , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Chaperone BiP , Enzyme Assays , Female , Gelsolin/genetics , Gelsolin/metabolism , Gene Expression Regulation , Heat-Shock Proteins/metabolism , Humans , Lung/microbiology , Macrophages/immunology , Macrophages/microbiology , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membrane Transport Proteins/metabolism , Oxidative Stress , Pepstatins/pharmacology , Protease Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , S100 Calcium Binding Protein A6 , S100 Proteins/metabolism , Streptococcus pneumoniae/immunology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
9.
Front Cell Infect Microbiol ; 13: 1090550, 2023.
Article in English | MEDLINE | ID: mdl-37033482

ABSTRACT

Introduction: Despite epidemiological associations between community acquired pneumonia (CAP) and myocardial infarction, mechanisms that modify cardiovascular disease during CAP are not well defined. In particular, largely due to a lack of relevant experimental models, the effect of pneumonia on atherosclerotic plaques is unclear. We describe the development of a murine model of the commonest cause of CAP, Streptococcus pneumoniae pneumonia, on a background of established atherosclerosis. We go on to use our model to investigate the effects of pneumococcal pneumonia on atherosclerosis. Methods: C57BL/6J and ApoE-/- mice were fed a high fat diet to promote atherosclerotic plaque formation. Mice were then infected with a range of S. pneumoniae serotypes (1, 4 or 14) with the aim of establishing a model to study atherosclerotic plaque evolution after pneumonia and bacteremia. Laser capture microdissection of plaque macrophages enabled transcriptomic analysis. Results: Intratracheal instillation of S. pneumoniae in mice fed a cholate containing diet resulted in low survival rates following infection, suggestive of increased susceptibility to severe infection. Optimization steps resulted in a final model of male ApoE-/- mice fed a Western diet then infected by intranasal instillation of serotype 4 (TIGR4) S. pneumoniae followed by antibiotic administration. This protocol resulted in high rates of bacteremia (88.9%) and survival (88.5%). Pneumonia resulted in increased aortic sinus plaque macrophage content 2 weeks post pneumonia but not at 8 weeks, and no difference in plaque burden or other plaque vulnerability markers were found at either time point. Microarray and qPCR analysis of plaque macrophages identified downregulation of two E3 ubiquitin ligases, Huwe1 and Itch, following pneumonia. Treatment with atorvastatin failed to alter plaque macrophage content or other plaque features. Discussion: Without antibiotics, ApoE-/- mice fed a high fat diet were highly susceptible to mortality following S. pneumoniae infection. The major infection associated change in plaque morphology was an early increase in plaque macrophages. Our results also hint at a role for the ubiquitin proteasome system in the response to pneumococcal infection in the plaque microenvironment.


Subject(s)
Atherosclerosis , Bacteremia , Plaque, Atherosclerotic , Pneumonia, Pneumococcal , Male , Mice , Animals , Streptococcus pneumoniae , Mice, Inbred C57BL , Macrophages , Apolipoproteins E/genetics , Ubiquitins , Mice, Knockout , Disease Models, Animal
10.
Infect Immun ; 80(3): 1140-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22158745

ABSTRACT

The success of Streptococcus pneumoniae (the pneumococcus) as a pulmonary pathogen is related to its restriction of innate immune responses by respiratory epithelial cells. The mechanisms used to overcome this restriction are incompletely elucidated. Pulmonary chemokine expression involves complex cellular and molecular networks, involving the pulmonary epithelium, but the specific cellular interactions and the cytokines that control them are incompletely defined. We show that serotype 2 or 4 pneumococci induce only modest levels of CXCL8 expression from epithelial cell lines, even in the absence of a polysaccharide capsule. In contrast, coculture of A549 cells with the macrophage-like THP-1 cell line, differentiated with vitamin D, or monocyte-derived macrophages enhanced CXCL8 release. Supernatants from the THP-1 cell line prime A549 cells to release CXCL8 at levels similar to cocultures. Interleukin-1Ra (IL-1Ra) inhibits CXCL8 release from cocultures and reduces the activity of macrophage-conditioned media, but inhibition of tumor necrosis factor alpha (TNF-α) had only a minimal effect on CXCL8 release. Release of IL-1ß but not TNF-α was upregulated in cocultures. IL-1 type 1 receptor knockout C57BL/6 and BALB/c mice confirmed the importance of IL-1 signaling in CXC chemokine expression and neutrophil recruitment in vivo. In fulminant disease, increased IL-1 signaling resulted in increased neutrophils in the airway and more invasive disease. These results demonstrate that IL-1 is an important component of the cellular network involving macrophages and epithelial cells, which facilitates CXC chemokine expression and aids neutrophil recruitment during pneumococcal pneumonia. They also highlight a potential clinical role for anti-IL-1 treatment to limit excessive neutrophilic inflammation in the lung.


Subject(s)
Epithelial Cells/immunology , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Lung/immunology , Macrophages/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Animals , Cell Line , Coculture Techniques , Culture Media, Conditioned , Epithelial Cells/microbiology , Female , Humans , Interleukin-1beta/immunology , Interleukin-8/immunology , Lung/microbiology , Lung/pathology , Macrophages/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Pneumococcal Infections/microbiology , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/immunology , Streptococcus pneumoniae/pathogenicity
11.
Thorax ; 67(11): 985-92, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22735687

ABSTRACT

BACKGROUND: Therapeutic strategies to modulate the host response to bacterial pneumonia are needed to improve outcomes during community-acquired pneumonia. This study used mice with impaired Fas signalling to examine susceptibility to pneumococcal pneumonia and decoy receptor 3 analogue (DcR3-a) to correct factors associated with increased susceptibility. METHODS: Wild-type mice and those with varying degrees of impairment of Fas (lpr) or Fas ligand signalling (gld) were challenged with Streptococcus pneumoniae and microbiological and immunological outcomes measured in the presence or absence of DcR3-a. RESULTS: During established pneumonia, neutrophils became the predominant cell in the airway and gld mice were less able to clear bacteria from the lungs, demonstrating localised impairment of pulmonary neutrophil function in comparison to lpr or wild-type mice. T-cells from gld mice had enhanced activation and reduced apoptosis in comparison to wild-type and lpr mice during established pneumonia. Treatment with DcR3-a reduced T-cell activation and corrected the defect in pulmonary bacterial clearance in gld mice. CONCLUSIONS: The results suggest that imbalance in tumour necrosis factor superfamily signalling and excessive T-cell activation can impair bacterial clearance in the lung but that DcR3-a treatment can reduce T-cell activation, restore optimal pulmonary neutrophil function and enhance bacterial clearance during S pneumoniae infection.


Subject(s)
Fas Ligand Protein/metabolism , Neutrophils/immunology , Phagocytes/immunology , Pneumonia, Pneumococcal/immunology , Receptors, Tumor Necrosis Factor, Member 6b/pharmacology , Animals , Disease Models, Animal , Fas Ligand Protein/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neutrophils/drug effects , Phagocytes/drug effects , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/therapy , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/prevention & control , Signal Transduction/drug effects , Streptococcus pneumoniae/immunology
12.
Thorax ; 67(9): 796-803, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22496351

ABSTRACT

BACKGROUND: The death receptor ligand tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) shows considerable clinical promise as a therapeutic agent. TRAIL induces leukocyte apoptosis, reducing acute inflammatory responses in the lung. It is not known whether TRAIL modifies chronic lung injury or whether TRAIL has a role in human idiopathic pulmonary fibrosis (IPF). We therefore explored the capacity of TRAIL to modify chronic inflammatory lung injury and studied TRAIL expression in patients with IPF. METHODS: TRAIL(-/-) and wild-type mice were instilled with bleomycin and inflammation assessed at various time points by bronchoalveolar lavage and histology. Collagen deposition was measured by tissue hydroxyproline content. TRAIL expression in human IPF lung samples was assessed by immunohistochemistry and peripheral blood TRAIL measured by ELISA. RESULTS: TRAIL(-/-) mice had an exaggerated delayed inflammatory response to bleomycin, with increased neutrophil numbers (mean 3.19±0.8 wild type vs 11.5±5.4×10(4) TRAIL(-/-), p<0.0001), reduced neutrophil apoptosis (5.42±1.6% wild type vs 2.47±0.5% TRAIL(-/-), p=0.0003) and increased collagen (3.45±0.2 wild type vs 5.8±1.3 mg TRAIL(-/-), p=0.005). Immunohistochemical analysis showed induction of TRAIL in bleomycin-treated wild-type mice. Patients with IPF demonstrated lower levels of TRAIL expression than in control lung biopsies and their serum levels of TRAIL were significantly lower compared with matched controls (38.1±9.6 controls vs 32.3±7.2 pg/ml patients with IPF, p=0.002). CONCLUSION: These data suggest TRAIL may exert beneficial, anti-inflammatory actions in chronic pulmonary inflammation in murine models and that these mechanisms may be compromised in human IPF.


Subject(s)
Lung Injury/metabolism , Pulmonary Fibrosis/metabolism , TNF-Related Apoptosis-Inducing Ligand/deficiency , Animals , Biomarkers/metabolism , Bleomycin , Bronchoalveolar Lavage , Case-Control Studies , Collagen/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hydroxyproline/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Respiratory Function Tests
13.
Cell Tissue Res ; 350(3): 455-64, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22986921

ABSTRACT

Despite being initially identified in mice, little is known about the sites of production of members of the BPI fold (BPIF) containing (PLUNC) family of putative innate defence proteins in this species. These proteins have largely been considered to be specificaly expressed in the respiratory tract, and we have recently shown that they exhibit differential expression in the epithelium of the proximal airways. In this study, we have used species-specific antibodies to systematically localize two members of this protein family; BPIFA1 (PLUNC/SPLUNC1) and BPIFB1 (LPLUNC1) in adult mice. In general, these proteins exhibit distinct and only partially overlapping localization. BPIFA1 is highly expressed in the respiratory epithelium and Bowman's glands of the nasal passages, whereas BPIFB1 is present in small subset of goblet cells in the nasal passage and pharynx. BPIFB1 is also present in the serous glands in the proximal tongue where is co-localised with the salivary gland specific family member, BPIFA2E (parotid secretory protein) and also in glands of the soft palate. Both proteins exhibit limited expression outside of these regions. These results are consistent with the localization of the proteins seen in man. Knowledge of the complex expression patterns of BPIF proteins in these regions will allow the use of tractable mouse models of disease to dissect their function.


Subject(s)
Carrier Proteins/metabolism , Glycoproteins/metabolism , Mouth/cytology , Mouth/metabolism , Nasal Cavity/cytology , Nasal Cavity/metabolism , Phosphoproteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cell Differentiation/physiology , Glycoproteins/genetics , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phosphoproteins/genetics
14.
J Immunol ; 185(5): 2968-79, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20656927

ABSTRACT

Peripheral blood monocytes represent the rapid response component of mononuclear phagocyte host defense, generating vigorous but finite antibacterial responses. We investigated the fate of highly purified primary human monocytes following phagocytosis of different bacteria. Exposure to high bacterial loads resulted in rapid loss of cell viability and decreased functional competence. Cell death typically involved classical apoptosis. Exposure to high numbers of Escherichia coli and Klebsiella pneumoniae induced nonapoptotic death with loss of cell membrane integrity, marked disruption of phagolysosomes, and caspase-1 activation, while a subset of cells also released caspase-1-regulated extracellular traps. Classical apoptosis increased if extracellular bacterial replication was reduced and decreased if intracellular ATP levels were reduced during these infections. Both classical apoptosis and the alternative forms of cell death allowed monocytes, whose functional competence was exhausted, to downregulate reactive oxygen species and proinflammatory cytokine responses. In contrast, sustained stimulation of glycolytic metabolism and mitochondrial oxidative phosphorylation, with associated hypoxia inducible factor-1alpha upregulation, maintained intracellular ATP levels and prolonged monocyte functional longevity, as assessed by maintenance of phagocytosis, reactive oxygen species production, and proinflammatory cytokine generation. Monocyte innate responses to bacteria are short-lived and are limited by an intrinsic program of apoptosis, a response that is subverted by overwhelming infection with E. coli and K. pneumoniae or bacterial stimulation of cell metabolism. In this regard, the fate of monocytes following bacterial challenge more closely resembles neutrophils than macrophages.


Subject(s)
Escherichia coli Infections/immunology , Immunity, Innate , Klebsiella Infections/immunology , Meningococcal Infections/immunology , Monocytes/immunology , Monocytes/pathology , Neisseriaceae Infections/immunology , Cell Death/immunology , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , DNA/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Humans , Klebsiella Infections/microbiology , Klebsiella Infections/pathology , Meningococcal Infections/microbiology , Meningococcal Infections/pathology , Monocytes/metabolism , Monocytes/microbiology , Neisseriaceae Infections/microbiology , Neisseriaceae Infections/pathology
15.
Front Immunol ; 13: 956991, 2022.
Article in English | MEDLINE | ID: mdl-35967296

ABSTRACT

Dysregulated neutrophilic inflammation can be highly destructive in chronic inflammatory diseases due to prolonged neutrophil lifespan and continual release of histotoxic mediators in inflamed tissues. Therapeutic induction of neutrophil apoptosis, an immunologically silent form of cell death, may be beneficial in these diseases, provided that the apoptotic neutrophils are efficiently cleared from the tissue. Previous research in our group identified ErbB inhibitors as able to induce neutrophil apoptosis and reduce neutrophilic inflammation both in vitro and in vivo. Here, we extend that work using a clinical ErbB inhibitor, neratinib, which has the potential to be repurposed in inflammatory diseases. We show that neratinib reduces neutrophilic migration o an inflammatory site in zebrafish larvae. Neratinib upregulates efferocytosis and reduces the number of persisting neutrophil corpses in mouse models of acute, but not chronic, lung injury, suggesting that the drug may have therapeutic benefits in acute inflammatory settings. Phosphoproteomic analysis of human neutrophils shows that neratinib modifies the phosphorylation of proteins regulating apoptosis, migration, and efferocytosis. This work identifies a potential mechanism for neratinib in treating acute lung inflammation by upregulating the clearance of dead neutrophils and, through examination of the neutrophil phosphoproteome, provides important insights into the mechanisms by which this may be occurring.


Subject(s)
Neutrophils , Zebrafish , Animals , Apoptosis/physiology , ErbB Receptors/metabolism , Humans , Inflammation , Macrophages/metabolism , Mice , Protein Kinase Inhibitors , Proteome/metabolism , Quinolines
16.
Article in English | MEDLINE | ID: mdl-32984077

ABSTRACT

Exposure to respiratory pathogens is a leading cause of exacerbations of airway diseases such as asthma and chronic obstructive pulmonary disease (COPD). Pellino-1 is an E3 ubiquitin ligase known to regulate virally-induced inflammation. We wished to determine the role of Pellino-1 in the host response to respiratory viruses in health and disease. Pellino-1 expression was examined in bronchial sections from patients with GOLD stage two COPD and healthy controls. Primary bronchial epithelial cells (PBECs) in which Pellino-1 expression had been knocked down were extracellularly challenged with the TLR3 agonist poly(I:C). C57BL/6 Peli1-/- mice and wild type littermates were subjected to intranasal infection with clinically-relevant respiratory viruses: rhinovirus (RV1B) and influenza A. We found that Pellino-1 is expressed in the airways of normal subjects and those with COPD, and that Pellino-1 regulates TLR3 signaling and responses to airways viruses. In particular we observed that knockout of Pellino-1 in the murine lung resulted in increased production of proinflammatory cytokines IL-6 and TNFα upon viral infection, accompanied by enhanced recruitment of immune cells to the airways, without any change in viral replication. Pellino-1 therefore regulates inflammatory airway responses without altering replication of respiratory viruses.


Subject(s)
Picornaviridae Infections , Pulmonary Disease, Chronic Obstructive , Virus Diseases , Animals , Humans , Mice , Mice, Inbred C57BL , Nuclear Proteins , Rhinovirus , Ubiquitin-Protein Ligases/genetics
17.
ACS Nano ; 14(7): 8287-8298, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32515944

ABSTRACT

Mononuclear phagocytes such as monocytes, tissue-specific macrophages, and dendritic cells are primary actors in both innate and adaptive immunity. These professional phagocytes can be parasitized by intracellular bacteria, turning them from housekeepers to hiding places and favoring chronic and/or disseminated infection. One of the most infamous is the bacteria that cause tuberculosis (TB), which is the most pandemic and one of the deadliest diseases, with one-third of the world's population infected and an average of 1.8 million deaths/year worldwide. Here we demonstrate the effective targeting and intracellular delivery of antibiotics to infected macrophages both in vitro and in vivo, using pH-sensitive nanoscopic polymersomes made of PMPC-PDPA block copolymer. Polymersomes showed the ability to significantly enhance the efficacy of the antibiotics killing Mycobacterium bovis, Mycobacterium tuberculosis, and another established intracellular pathogen, Staphylococcus aureus. Moreover, they demonstrated to easily access TB-like granuloma tissues-one of the harshest environments to penetrate-in zebrafish models. We thus successfully exploited this targeting for the effective eradication of several intracellular bacteria, including M. tuberculosis, the etiological agent of human TB.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Humans , Macrophages , Monocytes , Tuberculosis/drug therapy , Zebrafish
18.
Am J Respir Crit Care Med ; 177(1): 35-43, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17916805

ABSTRACT

RATIONALE: Cystic fibrosis lung disease is characterized by accumulation of apoptotic neutrophils, indicating impaired clearance of dying cells. Pseudomonas aeruginosa, the principal microbial pathogen in cystic fibrosis, manipulates apoptosis induction via production of toxic metabolites. Whether these metabolites, particularly pyocyanin, can also modulate apoptotic cell engulfment is unknown. OBJECTIVES: To assess the effects of pyocyanin on apoptotic cell engulfment by macrophages in vitro and in vivo and to investigate potential mechanisms of the observed effects. METHODS: Human monocyte-derived macrophages were treated with pyocyanin before challenge with apoptotic neutrophils, apoptotic Jurkat cells, or latex beads, and phagocytosis was assessed by light microscopy and flow cytometry. Effects of pyocyanin production on apoptotic cell clearance in vivo were assessed in a murine model, comparing infection by wild-type or pyocyanin-deficient P. aeruginosa. Oxidant production was investigated using fluorescent probes and pharmacologic inhibition and Rho GTPase signaling by immunoblotting and inhibitor studies. MEASUREMENTS AND MAIN RESULTS: Pyocyanin treatment impaired macrophage engulfment of apoptotic cells in vitro, without inducing significant macrophage apoptosis, whereas latex bead uptake was preserved. Macrophage ingestion of apoptotic cells was reduced and late apoptotic/necrotic cells were increased in mice infected with pyocyanin-producing P. aeruginosa compared with the pyocyanin-deficient strain. Inhibition of apoptotic cell uptake involved intracellular generation of reactive oxygen species (ROS) and effects on Rho GTPase signaling. Antioxidants or blockade of Rho signaling substantially restored apoptotic cell engulfment. CONCLUSIONS: These studies demonstrate that P. aeruginosa can manipulate the inflammatory microenvironment through inhibition of apoptotic cell engulfment, and suggest potential strategies to limit pulmonary inflammation in cystic fibrosis.


Subject(s)
Apoptosis/drug effects , Cystic Fibrosis/immunology , Macrophages, Alveolar/drug effects , Phagocytosis/drug effects , Pneumonia, Bacterial/immunology , Pseudomonas aeruginosa/metabolism , Pyocyanine/pharmacology , Animals , Humans , In Situ Nick-End Labeling , Jurkat Cells , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Microspheres , Monomeric GTP-Binding Proteins/metabolism , Phagocytosis/immunology , Pseudomonas aeruginosa/immunology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
19.
Am J Respir Crit Care Med ; 177(8): 887-95, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18202350

ABSTRACT

RATIONALE: The role of NADPH oxidase activation in pneumonia is complex because reactive oxygen species contribute to both microbial killing and regulation of the acute pulmonary infiltrate. The relative importance of each role remains poorly defined in community-acquired pneumonia. OBJECTIVES: We evaluated the contribution of NADPH oxidase-derived reactive oxygen species to the pathogenesis of pneumococcal pneumonia, addressing both the contribution to microbial killing and regulation of the inflammatory response. METHODS: Mice deficient in the gp91(phox) component of the phagocyte NADPH oxidase were studied after pneumococcal challenge. MEASUREMENTS AND MAIN RESULTS: gp91(phox)(-/-) mice demonstrated no defect in microbial clearance as compared with wild-type C57BL/6 mice. A significant increase in bacterial clearance from the lungs of gp91(phox)(-/-) mice was associated with increased numbers of neutrophils in the lung, lower rates of neutrophil apoptosis, and enhanced activation. Marked alterations in pulmonary cytokine/chemokine expression were also noted in the lungs of gp91(phox)(-/-) mice, characterized by elevated levels of tumor necrosis factor-alpha, KC, macrophage inflammatory protein-2, monocyte chemotactic protein-1, and IL-6. The greater numbers of neutrophils in gp91(phox)(-/-) mice were not associated with increased lung injury. Levels of neutrophil elastase in bronchoalveolar lavage were not decreased in gp91(phox)(-/-) mice. CONCLUSIONS: During pneumococcal pneumonia, NADPH oxidase-derived reactive oxygen species are redundant for host defense but limit neutrophil recruitment and survival. Decreased NADPH oxidase-dependent reactive oxygen species production is well tolerated and improves disease outcome during pneumococcal pneumonia by removing neutrophils from the tight constraints of reactive oxygen species-mediated regulation.


Subject(s)
Membrane Glycoproteins/deficiency , NADPH Oxidases/immunology , Neutrophils/immunology , Pneumonia, Pneumococcal/immunology , Animals , Disease Models, Animal , Female , Inflammation , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/deficiency , Neutrophil Infiltration , Neutrophils/microbiology , Pneumonia, Pneumococcal/physiopathology , Reactive Oxygen Species/immunology
20.
Curr Mol Med ; 8(6): 497-509, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18781957

ABSTRACT

The cholesterol-dependent cytolysins are pore-forming toxins. Pneumolysin is the cytolysin produced by Streptococcus pneumoniae and is a key virulence factor. The protein contains 471 amino acids and four structural domains. Binding to cholesterol is followed by oligomerization and membrane pore formation. Pneumolysin also activates the classical pathway of complement. Mutational analysis of the toxin and knowledge of sequence variation in outbreak strains suggests that additional activities of biologic importance exist. Pneumolysin activates a large number of genes, some by epigenetic modification, in eukaryotic cells and multiple signal transduction pathways. Cytolytic effects contribute to lung injury and neuronal damage while pro-inflammatory effects compound tissue damage. Nevertheless pneumolysin is a focal point of the immune response to pneumococci. Toll-like receptor 4-mediated recognition, osmosensing and T-cell responses to pneumolysin have been identified. In some animal models mutants that lack pneumolysin are associated with impaired bacterial clearance. Pneumolysin, which itself may induce apoptosis in neurones and other cells can activate host-mediated apoptosis in macrophages enhancing clearance. Disease pathogenesis, which has traditionally focused on the harmful effects of the toxin, increasingly recognises that a precarious balance between limited host responses to pneumolysin and either excessive immune responses or toxin-mediated subversion of host immunity exists.


Subject(s)
Host-Pathogen Interactions , Streptolysins/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Death , Cell Membrane/metabolism , Cholesterol/metabolism , Complement Activation , Eukaryotic Cells , Humans , Immune System/metabolism , Immune System/microbiology , Models, Biological , Nervous System/metabolism , Nervous System/microbiology , Protein Binding , Respiratory System/metabolism , Respiratory System/microbiology , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/pathogenicity , Streptolysins/chemistry , Streptolysins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL