Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Euro Surveill ; 29(3)2024 Jan.
Article in English | MEDLINE | ID: mdl-38240057

ABSTRACT

Under International Health Regulations from 2005, a human infection caused by a novel influenza A virus variant is considered an event that has potential for high public health impact and is immediately notifiable to the World Health Organisation. We here describe the clinical, epidemiological and virological features of a confirmed human case of swine influenza A(H1N2)v in England detected through community respiratory virus surveillance. Swabbing and contact tracing helped refine public health risk assessment, following this unusual and unexpected finding.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Orthomyxoviridae Infections , Swine Diseases , Animals , Humans , Swine , Influenza A Virus, H1N2 Subtype , Influenza A Virus, H1N1 Subtype/genetics , Swine Diseases/diagnosis , Swine Diseases/epidemiology , Influenza, Human/diagnosis , Influenza, Human/epidemiology , England/epidemiology
2.
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
4.
Cell Metab ; 33(2): 411-423.e4, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33306983

ABSTRACT

Neutrophils can function and survive in injured and infected tissues, where oxygen and metabolic substrates are limited. Using radioactive flux assays and LC-MS tracing with U-13C glucose, glutamine, and pyruvate, we observe that neutrophils require the generation of intracellular glycogen stores by gluconeogenesis and glycogenesis for effective survival and bacterial killing. These metabolic adaptations are dynamic, with net increases in glycogen stores observed following LPS challenge or altitude-induced hypoxia. Neutrophils from patients with chronic obstructive pulmonary disease have reduced glycogen cycling, resulting in impaired function. Metabolic specialization of neutrophils may therefore underpin disease pathology and allow selective therapeutic targeting.


Subject(s)
Glucose/immunology , Neutrophils/immunology , Adult , Aged , Animals , Cells, Cultured , Female , Gluconeogenesis , Humans , Male , Mice , Mice, Knockout , Middle Aged , Young Adult
5.
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
6.
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
7.
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
8.
PLoS Pathog ; 13(7): e1006526, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28742152

ABSTRACT

Enterococcus faecalis is an opportunistic pathogen frequently isolated in clinical settings. This organism is intrinsically resistant to several clinically relevant antibiotics and can transfer resistance to other pathogens. Although E. faecalis has emerged as a major nosocomial pathogen, the mechanisms underlying the virulence of this organism remain elusive. We studied the regulation of daughter cell separation during growth and explored the impact of this process on pathogenesis. We demonstrate that the activity of the AtlA peptidoglycan hydrolase, an enzyme dedicated to septum cleavage, is controlled by several mechanisms, including glycosylation and recognition of the peptidoglycan substrate. We show that the long cell chains of E. faecalis mutants are more susceptible to phagocytosis and are no longer able to cause lethality in the zebrafish model of infection. Altogether, this work indicates that control of cell separation during division underpins the pathogenesis of E. faecalis infections and represents a novel enterococcal virulence factor. We propose that inhibition of septum cleavage during division represents an attractive therapeutic strategy to control infections.


Subject(s)
Cell Wall/metabolism , Enterococcus faecalis/cytology , Enterococcus faecalis/pathogenicity , Gram-Positive Bacterial Infections/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Division , Cell Wall/genetics , Enterococcus faecalis/enzymology , Enterococcus faecalis/genetics , Humans , N-Acetylmuramoyl-L-alanine Amidase/genetics , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Virulence , Zebrafish/microbiology
9.
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
10.
PLoS One ; 11(9): e0163139, 2016.
Article in English | MEDLINE | ID: mdl-27680884

ABSTRACT

Pulmonary inflammation and bacterial colonization are central to the pathogenesis of chronic obstructive pulmonary disease (COPD). Defects in macrophage phagocytosis of both bacteria and apoptotic cells contribute to the COPD phenotype. Small molecule inhibitors with anti-inflammatory activity against p38 mitogen activated protein kinases (MAPKs), phosphatidyl-inositol-3 kinase (PI3K) and Rho kinase (ROCK) are being investigated as novel therapeutics in COPD. Concerns exist, however, about off-target effects. We investigated the effect of p38 MAPK inhibitors (VX745 and SCIO469), specific inhibitors of PI3K α (NVS-P13K-2), δ (NVS-P13K-3) or γ (NVS-P13K-5) and a ROCK inhibitor PF4950834 on macrophage phagocytosis, early intracellular killing of bacteria and efferocytosis of apoptotic neutrophils. Alveolar macrophages (AM) obtained from broncho-alveolar lavage (BAL) or monocyte-derived macrophages (MDM) from COPD patients (GOLD stage II/III) enrolled from a well characterized clinical cohort (MRC COPD-MAP consortium) or from healthy ex-smoker controls were studied. Both COPD AM and MDM exhibited lower levels of bacterial phagocytosis (using Streptococcus pneumoniae and non-typeable Haemophilus influenzae) and efferocytosis than healthy controls. None of the inhibitors altered bacterial internalization or early intracellular bacterial killing in AM or MDM. Conversely PF4950834, but not other inhibitors, enhanced efferocytosis in COPD AM and MDM. These results suggest none of these inhibitors are likely to exacerbate phagocytosis-related defects in COPD, while confirming ROCK inhibitors can enhance efferocytosis in COPD.

11.
Br J Pharmacol ; 173(21): 3099-3109, 2016 11.
Article in English | MEDLINE | ID: mdl-27460634

ABSTRACT

BACKGROUND AND PURPOSE: PGE2 inhibits cytokine generation from human lung macrophages. However, the EP receptor that mediates this beneficial anti-inflammatory effect of PGE2 has not been defined. The aim of this study was to identify the EP receptor by which PGE2 inhibits cytokine generation from human lung macrophages. This was determined by using recently developed EP receptor ligands. EXPERIMENTAL APPROACH: The effects of PGE2 and EP-selective agonists on LPS-induced generation of TNF-α and IL-6 from macrophages were evaluated. The effects of EP2 -selective (PF-04852946, PF-04418948) and EP4 -selective (L-161,982, CJ-042794) receptor antagonists on PGE2 responses were studied. The expression of EP receptor subtypes by human lung macrophages was determined by RT-PCR. KEY RESULTS: PGE2 inhibited LPS-induced and Streptococcus pneumoniae-induced cytokine generation from human lung macrophages. Analysis of mRNA levels indicated that macrophages expressed EP2 and EP4 receptors. L-902,688 (EP4 receptor-selective agonist) was considerably more potent than butaprost (EP2 receptor-selective agonist) as an inhibitor of TNF-α generation from macrophages. EP2 receptor-selective antagonists had marginal effects on the PGE2 inhibition of TNF-α generation, whereas EP4 receptor-selective antagonists caused rightward shifts in the PGE2 concentration-response curves. CONCLUSIONS AND IMPLICATIONS: These studies demonstrate that the EP4 receptor is the principal receptor that mediates the anti-inflammatory effects of PGE2 on human lung macrophages. This suggests that EP4 receptor agonists could be effective anti-inflammatory agents in human lung disease.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dinoprostone/pharmacology , Macrophages, Alveolar/drug effects , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Anti-Bacterial Agents/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Dinoprostone/chemistry , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Microbial Sensitivity Tests , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/genetics , Streptococcus pneumoniae/drug effects
12.
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
13.
mBio ; 5(5): e01710-14, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25293758

ABSTRACT

Intracellular killing of Streptococcus pneumoniae is complemented by induction of macrophage apoptosis. Here, we show that the toxin pneumolysin (PLY) contributes both to lysosomal/phagolysosomal membrane permeabilization (LMP), an upstream event programing susceptibility to apoptosis, and to apoptosis execution via a mitochondrial pathway, through distinct mechanisms. PLY is necessary but not sufficient for the maximal induction of LMP and apoptosis. PLY's ability to induce both LMP and apoptosis is independent of its ability to form cytolytic pores and requires only the first three domains of PLY. LMP involves TLR (Toll-like receptor) but not NLRP3/ASC (nucleotide-binding oligomerization domain [Nod]-like receptor family, pyrin domain-containing protein 3/apoptosis-associated speck-like protein containing a caspase recruitment domain) signaling and is part of a PLY-dependent but phagocytosis-independent host response that includes the production of cytokines, including interleukin-1 beta (IL-1ß). LMP involves progressive and selective permeability to 40-kDa but not to 250-kDa fluorescein isothiocyanate (FITC)-labeled dextran, as PLY accumulates in the cytoplasm. In contrast, the PLY-dependent execution of apoptosis requires phagocytosis and is part of a host response to intracellular bacteria that also includes NO generation. In cells challenged with PLY-deficient bacteria, reconstitution of LMP using the lysomotrophic detergent LeuLeuOMe favored cell necrosis whereas PLY reconstituted apoptosis. The results suggest that PLY contributes to macrophage activation and cytokine production but also engages LMP. Following bacterial phagocytosis, PLY triggers apoptosis and prevents macrophage necrosis as a component of a broad-based antimicrobial strategy. This illustrates how a key virulence factor can become the focus of a multilayered and coordinated innate response by macrophages, optimizing pathogen clearance and limiting inflammation. Importance: Streptococcus pneumoniae, the commonest cause of bacterial pneumonia, expresses the toxin pneumolysin, which can make holes in cell surfaces, causing tissue damage. Macrophages, resident immune cells essential for responses to bacteria in tissues, activate a program of cell suicide called apoptosis, maximizing bacterial clearance and limiting harmful inflammation. We examined pneumolysin's role in activating this response. We demonstrate that pneumolysin did not directly form holes in cells to trigger apoptosis and show that pneumolysin has two distinct roles which require only part of the molecule. Pneumolysin and other bacterial factors released by bacteria that have not been eaten by macrophages activate macrophages to release inflammatory factors but also make the cell compartment containing ingested bacteria leaky. Once inside the cell, pneumolysin ensures that the bacteria activate macrophage apoptosis, rather than necrosis, enhancing bacterial killing and limiting inflammation. This dual response to pneumolysin is critical for an effective immune response to S. pneumoniae.


Subject(s)
Apoptosis , Intracellular Membranes/drug effects , Lysosomes/drug effects , Macrophages/drug effects , Permeability/drug effects , Streptococcus pneumoniae/immunology , Streptolysins/toxicity , Animals , Bacterial Proteins/toxicity , Cells, Cultured , Humans , Mice , Streptococcus pneumoniae/metabolism
14.
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
15.
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
16.
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
17.
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
18.
J Immunol ; 180(5): 3502-11, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292577

ABSTRACT

Neutrophils undergo rapid constitutive apoptosis that is accelerated following bacterial ingestion as part of effective immunity, but is also accelerated by bacterial exotoxins as a mechanism of immune evasion. The paradigm of pathogen-driven neutrophil apoptosis is exemplified by the Pseudomonas aeruginosa toxic metabolite, pyocyanin. We previously showed pyocyanin dramatically accelerates neutrophil apoptosis both in vitro and in vivo, impairs host defenses, and favors bacterial persistence. In this study, we investigated the mechanisms of pyocyanin-induced neutrophil apoptosis. Pyocyanin induced early lysosomal dysfunction, shown by altered lysosomal pH, within 15 min of exposure. Lysosomal disruption was followed by mitochondrial membrane permeabilization, caspase activation, and destabilization of Mcl-1. Pharmacological inhibitors of a lysosomal protease, cathepsin D (CTSD), abrogated pyocyanin-induced apoptosis, and translocation of CTSD to the cytosol followed pyocyanin treatment and lysosomal disruption. A stable analog of cAMP (dibutyryl cAMP) impeded the translocation of CTSD and prevented the destabilization of Mcl-1 by pyocyanin. Thus, pyocyanin activated a coordinated series of events dependent upon lysosomal dysfunction and protease release, the first description of a bacterial toxin using a lysosomal cell death pathway. This may be a pathological pathway of cell death to which neutrophils are particularly susceptible, and could be therapeutically targeted to limit neutrophil death and preserve host responses.


Subject(s)
Apoptosis/immunology , Bacterial Toxins/pharmacology , Lysosomes/immunology , Neutrophils/immunology , Neutrophils/microbiology , Pyocyanine/pharmacology , Signal Transduction/immunology , Caspase 3/physiology , Cathepsin D/antagonists & inhibitors , Cathepsin D/metabolism , Cell Death/immunology , Cell Survival/immunology , Cells, Cultured , Humans , Hydrogen-Ion Concentration , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Intracellular Fluid/microbiology , Lysosomes/enzymology , Lysosomes/microbiology , Neutrophils/enzymology , Neutrophils/pathology , Oxidative Stress/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...