Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
iScience ; 27(6): 110117, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38947521

ABSTRACT

Dysregulated host immune responses contribute to disease severity and worsened prognosis in COVID-19 infection and the underlying mechanisms are not fully understood. In this study, we observed that IL-33, a damage-associated molecular pattern molecule, is significantly increased in COVID-19 patients and in SARS-CoV-2-infected mice. Using IL-33-/- mice, we demonstrated that IL-33 deficiency resulted in significant decreases in bodyweight loss, tissue viral burdens, and lung pathology. These improved outcomes in IL-33-/- mice also correlated with a reduction in innate immune cell infiltrates, i.e., neutrophils, macrophages, natural killer cells, and activated T cells in inflamed lungs. Lung RNA-seq results revealed that IL-33 signaling enhances activation of inflammatory pathways, including interferon signaling, pathogen phagocytosis, macrophage activation, and cytokine/chemokine signals. Overall, these findings demonstrate that the alarmin IL-33 plays a pathogenic role in SARS-CoV-2 infection and provides new insights that will inform the development of effective therapeutic strategies for COVID-19.

2.
PLoS Pathog ; 20(5): e1012020, 2024 May.
Article in English | MEDLINE | ID: mdl-38743761

ABSTRACT

Scrub typhus is an acute febrile disease due to Orientia tsutsugamushi (Ot) infection and can be life-threatening with organ failure, hemorrhage, and fatality. Yet, little is known as to how the host reacts to Ot bacteria at early stages of infection; no reports have addressed the functional roles of type I versus type II interferon (IFN) responses in scrub typhus. In this study, we used comprehensive intradermal (i.d.) inoculation models and two clinically predominant Ot strains (Karp and Gilliam) to uncover early immune events. Karp infection induced sequential expression of Ifnb and Ifng in inflamed skin and draining lymph nodes at days 1 and 3 post-infection. Using double Ifnar1-/-Ifngr1-/- and Stat1-/- mice, we found that deficiency in IFN/STAT1 signaling resulted in lethal infection with profound pathology and skin eschar lesions, which resembled to human scrub typhus. Further analyses demonstrated that deficiency in IFN-γ, but not IFN-I, resulted in impaired NK cell and macrophage activation and uncontrolled bacterial growth and dissemination, leading to metabolic dysregulation, excessive inflammatory cell infiltration, and exacerbated tissue damage. NK cells were found to be the major cellular source of innate IFN-γ, contributing to the initial Ot control in the draining lymph nodes. In vitro studies with dendritic cell cultures revealed a superior antibacterial effect offered by IFN-γ than IFN-ß. Comparative in vivo studies with Karp- and Gilliam-infection revealed a crucial role of IFN-γ signaling in protection against progression of eschar lesions and Ot infection lethality. Additionally, our i.d. mouse models of lethal infection with eschar lesions are promising tools for immunological study and vaccine development for scrub typhus.


Subject(s)
Interferon-gamma , Mice, Knockout , Orientia tsutsugamushi , Scrub Typhus , Signal Transduction , Animals , Scrub Typhus/immunology , Scrub Typhus/microbiology , Orientia tsutsugamushi/immunology , Mice , Interferon-gamma/metabolism , Interferon-gamma/immunology , Mice, Inbred C57BL , Disease Models, Animal , Skin/microbiology , Skin/pathology , Skin/immunology , STAT1 Transcription Factor/metabolism
3.
Am J Respir Cell Mol Biol ; 71(1): 110-120, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38574279

ABSTRACT

Immune activation is essential for lung control of viral and bacterial infection, but an overwhelming inflammatory response often leads to the onset of acute respiratory distress syndrome. IL-10 plays a crucial role in regulating the balance between antimicrobial immunity and immunopathology. In the present study, we investigated the role of IL-10 in acute lung injury induced by influenza A virus and methicillin-resistant Staphylococcus aureus coinfection. This unique coinfection model resembles patients with acute pneumonia undergoing appropriate antibiotic therapies. Using global IL-10 and IL-10 receptor gene-deficient mice, as well as in vivo neutralizing antibodies, we show that IL-10 deficiency promotes IFN-γ-dominant cytokine responses and triggers acute animal death. Interestingly, this extreme susceptibility is fully preventable by IFN-γ neutralization during coinfection. Further studies using mice with Il10ra deletion in selective myeloid subsets reveal that IL-10 primarily acts on mononuclear phagocytes to prevent IFN-γ/TNF-α hyperproduction and acute mortality. Importantly, this antiinflammatory IL-10 signaling is independent of its inhibitory effect on antiviral and antibacterial defense. Collectively, our results demonstrate a key mechanism of IL-10 in preventing hypercytokinemia and acute respiratory distress syndrome pathogenesis by counteracting the IFN-γ response.


Subject(s)
Acute Lung Injury , Disease Models, Animal , Interferon-gamma , Interleukin-10 , Superinfection , Animals , Interleukin-10/metabolism , Interleukin-10/immunology , Acute Lung Injury/virology , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Acute Lung Injury/microbiology , Interferon-gamma/metabolism , Superinfection/immunology , Superinfection/virology , Mice , Mice, Inbred C57BL , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Coinfection/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/virology , Staphylococcal Infections/immunology , Mice, Knockout , Influenza A virus/immunology , Lung/virology , Lung/pathology , Lung/immunology , Lung/metabolism
4.
Front Immunol ; 14: 1272920, 2023.
Article in English | MEDLINE | ID: mdl-37771584

ABSTRACT

Introduction: A frequent sequela of influenza A virus (IAV) infection is secondary bacterial pneumonia. Therefore, it is clinically important to understand the genetic predisposition to IAV and bacterial coinfection. Methods: BALB/c and C57BL/6 (B6) mice were infected with high or low-pathogenic IAV and Streptococcus pneumoniae (SPn). The contribution of cellular and molecular immune factors to the resistance/susceptibility of BALB/c and B6 mice were dissected in nonlethal and lethal IAV/SPn coinfection models. Results: Low-virulent IAV X31 (H3N2) rendered B6 mice extremely susceptible to SPn superinfection, while BALB/c mice remained unaffected. X31 infection alone barely induces IFN-γresponse in two strains of mice; however, SPn superinfection significantly enhances IFN-γ production in the susceptible B6 mice. As a result, IFN-γ signaling inhibits neutrophil recruitment and bacterial clearance, leading to lethal X31/SPn coinfection in B6 mice. Conversely, the diminished IFN-γ and competent neutrophil responses enable BALB/c mice highly resistant to X31/SPn coinfection. Discussion: The results establish that type 1 immune predisposition plays a key role in lethal susceptibility of B6 mice to pneumococcal pneumonia after mild IAV infection.


Subject(s)
Coinfection , Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Pneumonia, Pneumococcal , Superinfection , Animals , Mice , Humans , Influenza A Virus, H3N2 Subtype , Mice, Inbred C57BL , Streptococcus pneumoniae
5.
Front Immunol ; 13: 1011132, 2022.
Article in English | MEDLINE | ID: mdl-36203588

ABSTRACT

Acute Respiratory Distress Syndrome (ARDS) is an inflammatory disease that is associated with high mortality but no specific treatment. Our understanding of initial events that trigger ARDS pathogenesis is limited. We have developed a mouse model of inflammatory lung injury by influenza and methicillin-resistant Staphylococcus aureus (MRSA) coinfection plus daily antibiotic therapy. Using this pneumonic ARDS model, here we show that IFN-γ receptor signaling drives inflammatory cytokine storm and lung tissue damage. By single-cell RNA sequencing (scRNA-seq) analysis, we demonstrate that IFN-γ signaling induces a transcriptional shift in airway immune cells, particularly by upregulating macrophage and monocyte expression of genes associated with inflammatory diseases. Further evidence from conditional knockout mouse models reveals that IFN-γ receptor signaling in myeloid cells, particularly CD11c+ mononuclear phagocytes, directly promotes TNF-α hyperproduction and inflammatory lung damage. Collectively, the findings from this study, ranging from cell-intrinsic gene expression to overall disease outcome, demonstrate that influenza-induced IFN-γ triggers myeloid cell hyperresponsiveness to MRSA, thereby leading to excessive inflammatory response and lethal lung damage during coinfection.


Subject(s)
Coinfection , Influenza, Human , Lung Injury , Methicillin-Resistant Staphylococcus aureus , Respiratory Distress Syndrome , Animals , Anti-Bacterial Agents/pharmacology , Humans , Interferon-gamma/genetics , Lung Injury/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells , Transcriptome , Tumor Necrosis Factor-alpha/genetics
6.
Immunohorizons ; 6(10): 716-721, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36220188

ABSTRACT

Multiple organ damage is common in patients with severe COVID-19, even though the underlying pathogenic mechanisms remain unclear. Acute viral infection typically activates type I IFN (IFN-I) signaling. The antiviral role of IFN-I is well characterized in vitro. However, our understanding of how IFN-I regulates host immune response to SARS-CoV-2 infection in vivo is incomplete. Using a human ACE2-transgenic mouse model, we show in the present study that IFN-I receptor signaling is essential for protection against the acute lethality of SARS-CoV-2 in mice. Interestingly, although IFN-I signaling limits viral replication in the lung, the primary infection site, it is dispensable for efficient viral clearance at the adaptive phase of SARS-CoV-2 infection. Conversely, we found that in the absence of IFN-I receptor signaling, the extreme animal lethality is consistent with heightened infectious virus and prominent pathological manifestations in the brain. Taken together, our results in this study demonstrate that IFN-I receptor signaling is required for restricting virus neuroinvasion, thereby mitigating COVID-19 severity.


Subject(s)
COVID-19 , SARS-CoV-2 , Angiotensin-Converting Enzyme 2 , Animals , Antiviral Agents , Humans , Mice , Mice, Transgenic
7.
J Immunol ; 209(1): 128-135, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35705254

ABSTRACT

Postinfluenza bacterial pneumonia is a significant cause of hospitalization and death in humans. The mechanisms underlying this viral and bacterial synergy remain incompletely understood. Recent evidence indicates that influenza-induced IFNs, particularly type I IFN (IFN-I) and IFN-γ, suppress antibacterial defenses. In this study, we have investigated the relative importance and interplay of IFN-I and IFN-γ pathways in influenza-induced susceptibility to Streptococcus pneumoniae infection. Using gene-deficient mouse models, as well as in vivo blocking Abs, we show that both IFN-I and IFN-γ signaling pathways contribute to the initial suppression of antibacterial immunity; however, IFN-γ plays a dominant role in the disease deterioration, in association with increased TNF-α production and alveolar macrophage (AM) depletion. We have previously shown that IFN-γ impairs AM antibacterial function and thereby acute bacterial clearance. The findings in this study indicate that IFN-γ signaling also impairs AM viability and αß T cell recruitment during the progression of influenza/S. pneumoniae coinfection. Macrophages insensitive to IFN-γ mice express a dominant-negative mutant IFN-γR in mononuclear phagocytes. Interestingly, macrophages insensitive to IFN-γ mice exhibited significantly improved recovery and survival from coinfection, despite delayed bacterial clearance. Importantly, we demonstrate that IFN-I receptor signaling is essential for preventing IFN-γ hyperproduction and animal death during the progression of postinfluenza pneumococcal pneumonia.


Subject(s)
Coinfection , Influenza, Human , Interferon Type I/metabolism , Orthomyxoviridae Infections , Pneumococcal Infections , Pneumonia, Pneumococcal , Animals , Anti-Bacterial Agents , Humans , Interferon-gamma , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Viruses ; 14(1)2022 01 14.
Article in English | MEDLINE | ID: mdl-35062354

ABSTRACT

The susceptibility to respiratory syncytial virus (RSV) infection in early life has been associated with a deficient T-helper cell type 1 (Th1) response. Conversely, healthy adults generally do not exhibit severe illness from RSV infection. In the current study, we investigated whether Th1 cytokine IFN-γ is essential for protection against RSV and RSV-associated comorbidities in adult mice. We found that, distinct from influenza virus, prior RSV infection does not induce significant IFN-γ production and susceptibility to secondary Streptococcus pneumoniae infection in adult wild-type (WT) mice. In ovalbumin (OVA)-induced asthmatic mice, RSV super-infection increases airway neutrophil recruitment and inflammatory lung damage but has no significant effect on OVA-induced eosinophilia. Compared with WT controls, RSV infection of asthmatic Ifng-/- mice results in increased airway eosinophil accumulation. However, a comparable increase in eosinophilia was detected in house dust mite (HDM)-induced asthmatic Ifng-/- mice in the absence of RSV infection. Furthermore, neither WT nor Ifng-/- mice exhibit apparent eosinophil infiltration during RSV infection alone. Together, these findings indicate that, despite its critical role in limiting eosinophilic inflammation during asthma, IFN-γ is not essential for protection against RSV-induced exacerbation of asthmatic inflammation in adult mice.


Subject(s)
Asthma/pathology , Inflammation/immunology , Interferon-gamma/immunology , Lung/immunology , Lung/pathology , Respiratory Syncytial Virus Infections/immunology , Animals , Asthma/chemically induced , Asthma/immunology , Bronchoalveolar Lavage Fluid , Coinfection/immunology , Coinfection/microbiology , Coinfection/prevention & control , Comorbidity , Female , Inflammation/prevention & control , Interferon-gamma/genetics , Lung/microbiology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Respiratory Syncytial Virus Infections/prevention & control , Th1 Cells , Th2 Cells
9.
J Immunol ; 207(5): 1371-1376, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34380647

ABSTRACT

Inflammatory cytokine storm is a known cause for acute respiratory distress syndrome. In this study, we have investigated the role of IFN-γ in lethal lung inflammation using a mouse model of postinfluenza methicillin-resistant Staphylococcus aureus (MRSA) pneumonia. To mimic the clinical scenario, animals were treated with antibiotics for effective bacterial control following MRSA superinfection. However, antibiotic therapy alone is not sufficient to improve survival of wild-type animals in this lethal acute respiratory distress syndrome model. In contrast, antibiotics induce effective protection in mice deficient in IFN-γ response. Mechanistically, we show that rather than inhibiting bacterial clearance, IFN-γ promotes proinflammatory cytokine response to cause lethal lung damage. Neutralization of IFN-γ after influenza prevents hyperproduction of TNF-α, and thereby protects against inflammatory lung damage and animal mortality. Taken together, the current study demonstrates that influenza-induced IFN-γ drives a stepwise propagation of inflammatory cytokine response, which ultimately results in fatal lung damage during secondary MRSA pneumonia, despite of antibiotic therapy.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Inflammation/immunology , Influenza A virus/physiology , Influenza, Human/immunology , Interferon-gamma/metabolism , Lung/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Staphylococcal/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Animals , Cells, Cultured , Humans , Influenza, Human/complications , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/complications , Pneumonia, Staphylococcal/complications , Staphylococcal Infections/complications , Superinfection , Tumor Necrosis Factor-alpha
10.
J Immunol ; 205(6): 1601-1607, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32796026

ABSTRACT

Secondary Streptococcus pneumoniae infection is a significant cause of morbidity and mortality during influenza epidemics and pandemics. Multiple pathogenic mechanisms, such as lung epithelial damage and dysregulation of neutrophils and alveolar macrophages (AMs), have been suggested to contribute to the severity of disease. However, the fundamental reasons for influenza-induced susceptibility to secondary bacterial pneumonia remain unclear. In this study, we revisited these controversies over key pathogenic mechanisms in a lethal model of secondary bacterial pneumonia with an S. pneumoniae strain that is innocuous to mice in the absence of influenza infection. Using a series of in vivo models, we demonstrate that rather than a systemic suppression of immune responses or neutrophil function, influenza infection activates IFN-γR signaling and abrogates AM-dependent bacteria clearance and thereby causes extreme susceptibility to pneumococcal infection. Importantly, using mice carrying conditional knockout of Ifngr1 gene in different myeloid cell subsets, we demonstrate that influenza-induced IFN-γR signaling in AMs impairs their antibacterial function, thereby enabling otherwise noninvasive S. pneumoniae to cause deadly pneumonia.


Subject(s)
Influenza A virus/physiology , Influenza, Human/immunology , Macrophages, Alveolar/physiology , Orthomyxoviridae Infections/immunology , Pneumonia, Pneumococcal/immunology , Receptors, Interferon/metabolism , Streptococcus pneumoniae/physiology , Animals , Coinfection , Disease Models, Animal , Disease Susceptibility , Humans , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interferon/genetics , Signal Transduction , Interferon gamma Receptor
11.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31383747

ABSTRACT

Postinfluenza methicillin-resistant Staphylococcus aureus (MRSA) infection can quickly develop into severe, necrotizing pneumonia, causing over 50% mortality despite antibiotic treatments. In this study, we investigated the efficacy of antibiotic therapies and the impact of S. aureus alpha-toxin in a model of lethal influenza virus and MRSA coinfection. We demonstrate that antibiotics primarily attenuate alpha-toxin-induced acute lethality, even though both alpha-toxin-dependent and -independent mechanisms significantly contribute to animal mortality after coinfection. Furthermore, we found that the protein synthesis-suppressing antibiotic linezolid has an advantageous therapeutic effect on alpha-toxin-induced lung damage, as measured by protein leak and lactate dehydrogenase (LDH) activity. Importantly, using a Panton-Valentine leucocidin (PVL)-negative MRSA isolate from patient sputum, we show that linezolid therapy significantly improves animal survival from postinfluenza MRSA pneumonia compared with vancomycin treatment. Rather than improved viral or bacterial control, this advantageous therapeutic effect is associated with a significantly attenuated proinflammatory cytokine response and acute lung damage in linezolid-treated mice. Together, our findings not only establish a critical role of alpha-toxin in the extreme mortality of secondary MRSA pneumonia after influenza but also provide support for the possibility that linezolid could be a more effective treatment than vancomycin to improve disease outcomes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Linezolid/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Orthomyxoviridae Infections/complications , Pneumonia, Staphylococcal/drug therapy , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Female , Gene Expression , Gentamicins/pharmacology , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , L-Lactate Dehydrogenase/metabolism , Lung/microbiology , Lung/pathology , Male , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Plasmids/chemistry , Plasmids/metabolism , Pneumonia, Staphylococcal/complications , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/mortality , Survival Analysis , Vancomycin/pharmacology
12.
Trends Microbiol ; 27(5): 383-384, 2019 05.
Article in English | MEDLINE | ID: mdl-30871857

ABSTRACT

Bacterial lung infections are frequent causes of mortality following influenza infection, but the fundamental mechanisms remain largely unknown. A new study by Martínez-Colón et al. (PLoS Pathog. 2019;15:e1007560) now suggests that influenza-induced immune suppression of Staphylococcus aureus is mediated by TLR9 signaling.


Subject(s)
Coinfection , Influenza, Human , Methicillin-Resistant Staphylococcus aureus , Orthomyxoviridae Infections , Staphylococcal Infections , Humans , Methicillin , Staphylococcus aureus , Toll-Like Receptor 9
13.
J Immunol ; 202(7): 2027-2034, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30745458

ABSTRACT

Methicillin-resistant Staphylococcus aureus has emerged as a significant contributor to morbidity and mortality associated with influenza infection. In this study, we show in a mouse model that preceding influenza infection promotes S. aureus resistance to killing by antibiotics. This resistance coincides with influenza-induced accumulation of inflammatory monocytes in the lung. CCR type 2 (CCR2) is responsible for pulmonary monocyte recruitment after influenza infection. We found that antibiotic-treated Ccr2-deficient (Ccr2-/-) mice exhibit significantly improved bacterial control and survival from influenza and methicillin-resistant S. aureus coinfection, despite a delay in viral clearance. Mechanistically, our results from in vivo studies indicate that influenza-induced monocytes serve as reservoirs for intracellular S. aureus survival, thereby promoting bacterial resistance to antibiotic treatment. Blocking CCR2 with a small molecular inhibitor (PF-04178903), in conjunction with antibiotic treatment, enhanced lung bacterial clearance and significantly improved animal survival. Collectively, our study demonstrates that inflammatory monocytes constitute an important and hitherto underappreciated mechanism of the conflicting immune requirements for viral and bacterial clearance by hosts, which subsequently leads to exacerbated outcomes of influenza and S. aureus coinfection.


Subject(s)
Coinfection/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Monocytes/immunology , Monocytes/microbiology , Orthomyxoviridae Infections/complications , Animals , Drug Resistance, Bacterial/immunology , Female , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Receptors, CCR2/immunology
14.
J Immunol ; 200(4): 1425-1433, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29311363

ABSTRACT

Influenza and bacterial coinfection is a significant cause of hospitalization and death in humans during influenza epidemics and pandemics. However, the fundamental protective and pathogenic mechanisms involved in this complex virus-host-bacterium interaction remain incompletely understood. In this study, we have developed mild to lethal influenza and Streptococcus pneumoniae coinfection models for comparative analyses of disease pathogenesis. Specifically, wild-type and IL-1R type 1-deficient (Il1r1-/- ) mice were infected with influenza virus and then superchallenged with noninvasive S. pneumoniae serotype 14 (Spn14) or S. pneumoniae serotype 19A (Spn19A). The coinfections were followed by comparative analyses of inflammatory responses and animal protection. We found that resident alveolar macrophages are efficient in the clearance of both pneumococcal serotypes in the absence of influenza infection; in contrast, they are essential for airway control of Spn14 infection but not Spn19A infection. In agreement, TNF-α and neutrophils play a compensatory protective role in secondary bacterial infection associated with Spn19A; however, the essential requirement for alveolar macrophage-mediated clearance significantly enhances the virulence of Spn14 during postinfluenza pneumococcal infection. Furthermore, we show that, although IL-1 signaling is not required for host defense against pneumococcal infection alone, it is essential for sustaining antibacterial immunity during postinfluenza pneumococcal infection, as evidenced by significantly aggravated bacterial burden and animal mortality in Il1r1-/- mice. Mechanistically, we show that through preventing alveolar macrophage depletion, inflammatory cytokine IL-1 signaling is critically involved in host resistance to influenza and pneumococcal coinfection.


Subject(s)
Coinfection/immunology , Interleukin-1/immunology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/immunology , Pneumococcal Infections/immunology , Animals , Humans , Influenza A Virus, H3N2 Subtype , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology
15.
J Exp Med ; 213(9): 1851-64, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27526712

ABSTRACT

Clinical post-influenza Staphylococcus aureus pneumonia is characterized by extensive lung inflammation associated with severe morbidity and mortality even after appropriate antibiotic treatment. In this study, we show that antibiotics rescue nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2)-deficient mice but fail to fully protect WT animals from influenza and S. aureus coinfection. Further experiments indicate that the inefficacy of antibiotics against coinfection is attributable to oxidative stress-associated inflammatory lung injury. However, Nox2-induced lung damage during coinfection was not associated with aggravated inflammatory cytokine response or cell infiltration but rather caused by reduced survival of myeloid cells. Specifically, oxidative stress increased necrotic death of inflammatory cells, thereby resulting in lethal damage to surrounding tissue. Collectively, our results demonstrate that influenza infection disrupts the delicate balance between Nox2-dependent antibacterial immunity and inflammation. This disruption leads to not only increased susceptibility to S. aureus infection, but also extensive lung damage. Importantly, we show that combination treatment of antibiotic and NADPH oxidase inhibitor significantly improved animal survival from coinfection. These findings suggest that treatment strategies that target both bacteria and oxidative stress will significantly benefit patients with influenza-complicated S. aureus pneumonia.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Membrane Glycoproteins/physiology , NADPH Oxidases/physiology , Orthomyxoviridae Infections/complications , Oxidative Stress , Pneumonia, Staphylococcal/drug therapy , Animals , Cell Survival , Female , Inflammation/etiology , Male , Methicillin-Resistant Staphylococcus aureus , Mice , Mice, Inbred C57BL , Myeloid Cells/physiology , NADPH Oxidase 2 , Reactive Oxygen Species/metabolism
16.
J Immunol ; 196(10): 4196-203, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27053759

ABSTRACT

The incidence of methicillin-resistant Staphylococcus aureus (MRSA) pneumonia in otherwise healthy individuals is increasing. To investigate the mechanism underlying the epidemiological success of predominant community-associated (CA)-MRSA strains, we examined their fitness traits during the initial interaction between bacteria and the host occurring in the lower airway. Using a mouse respiratory infection model, we show that clinical isolates often responsible for CA infections are highly resistant to clearance from healthy airways, whereas S. aureus strains not as prevalent or traditionally associated with hospital-associated infections are relatively susceptible. Mechanistically, the competitive fitness of S. aureus is a result of both agr-dependent and -independent resistance to innate bacterial killing. Furthermore, we show that rather than evasion from neutrophil-dependent bactericidal process, the observed S. aureus fitness in the lower airways is due to its intrinsic resistance to resident alveolar macrophage-mediated intracellular killing. Importantly, we demonstrate that the virulence determinants responsible for bacterial persistence in immune-competent mice are dispensable in mice with predisposing conditions such as influenza infection. Taken together, these novel findings of the improved competence of predominant CA-MRSA strains to survive innate killing in healthy hosts, particularly at the very beginning stage of infection, provide a unique insight into their epidemiological success.


Subject(s)
Macrophages, Alveolar/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Neutrophils/immunology , Pneumonia, Bacterial/microbiology , Respiratory System/microbiology , Staphylococcal Infections/microbiology , Animals , Bronchoalveolar Lavage Fluid/microbiology , Cell Line , Coinfection/immunology , DNA, Bacterial/genetics , Female , Genes, Bacterial , Kaplan-Meier Estimate , Male , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/microbiology , Phagocytosis , Pneumonia, Bacterial/immunology , Respiratory System/immunology , Specific Pathogen-Free Organisms , Staphylococcal Infections/immunology , Virulence , Virulence Factors/genetics
17.
J Infect Dis ; 212(3): 445-52, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25649173

ABSTRACT

BACKGROUND: Secondary bacterial infections following influenza represent a major cause of mortality in the human population, which, in turn, has led to a call for stockpiling of bacterial vaccines for pandemic preparedness. METHODS: To investigate the efficacy of bacterial vaccination for protection against secondary pneumococcal infection, mice were immunized with pneumococcal capsular polysaccharide conjugate vaccine, and then sequentially coinfected 5 weeks later with PR8 influenza virus and A66.1 Streptococcus pneumoniae. RESULTS: In the absence of influenza virus exposure, vaccination with polysaccharide conjugate vaccine was highly effective, as indicated by 100% survival from lethal pneumococcal pneumonia and 10 000-fold greater efficiency in clearance of bacteria from the lung compared to unvaccinated mice. Enhanced clearance after vaccination was dependent upon Fc receptor (FcR) expression. However, following influenza, <40% of vaccinated mice survived bacterial coinfection and FcR-dependent clearance of antibody-opsonized bacteria reduced bacterial levels in the lungs only 5-10 fold. No differences in lung myeloid cell numbers or in FcR cell surface expression were observed following influenza. CONCLUSIONS: The results show that induction of antibacterial humoral immunity is only partially effective in protection against secondary bacterial infections that occur following influenza, and suggest that additional therapeutic strategies to overcome defective antibacterial immunity should be explored.


Subject(s)
Orthomyxoviridae Infections/prevention & control , Pneumococcal Vaccines/immunology , Pneumococcal Vaccines/pharmacology , Pneumonia, Pneumococcal/prevention & control , Streptococcus pneumoniae/immunology , Animals , Antigens, CD/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Pneumococcal/immunology , Survival Analysis
18.
PLoS Pathog ; 10(12): e1004560, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25500584

ABSTRACT

Suppressor of cytokine signaling (SOCS) proteins are inducible feedback inhibitors of cytokine signaling. SOCS1-/- mice die within three weeks postnatally due to IFN-γ-induced hyperinflammation. Since it is well established that IFN-γ is dispensable for protection against influenza infection, we generated SOCS1-/-IFN-γ-/- mice to determine whether SOCS1 regulates antiviral immunity in vivo. Here we show that SOCS1-/-IFN-γ-/- mice exhibited significantly enhanced resistance to influenza infection, as evidenced by improved viral clearance, attenuated acute lung damage, and consequently increased survival rates compared to either IFN-γ-/- or WT animals. Enhanced viral clearance in SOCS1-/-IFN-γ-/- mice coincided with a rapid onset of adaptive immune responses during acute infection, while their reduced lung injury was associated with decreased inflammatory cell infiltration at the resolution phase of infection. We further determined the contribution of SOCS1-deficient T cells to antiviral immunity. Anti-CD4 antibody treatment of SOCS1-/-IFN-γ-/- mice had no significant effect on their enhanced resistance to influenza infection, while CD8+ splenocytes from SOCS1-/-IFN-γ-/- mice were sufficient to rescue RAG1-/- animals from an otherwise lethal infection. Surprisingly, despite their markedly reduced viral burdens, RAG1-/- mice reconstituted with SOCS1-/-IFN-γ-/- adaptive immune cells failed to ameliorate influenza-induced lung injury. In conclusion, in the absence of IFN-γ, the cytoplasmic protein SOCS1 not only inhibits adaptive antiviral immune responses but also exacerbates inflammatory lung damage. Importantly, these detrimental effects of SOCS1 are conveyed through discrete cell populations. Specifically, while SOCS1 expression in adaptive immune cells is sufficient to inhibit antiviral immunity, SOCS1 in innate/stromal cells is responsible for aggravated lung injury.


Subject(s)
Lung Injury/physiopathology , Lung Injury/virology , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , Orthomyxoviridae/physiology , Suppressor of Cytokine Signaling Proteins/physiology , Adaptive Immunity/physiology , Animals , Cytokines/physiology , Disease Models, Animal , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/physiology , Lung Diseases/physiopathology , Lung Diseases/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/deficiency , Suppressor of Cytokine Signaling Proteins/genetics
19.
J Immunol ; 192(7): 3301-7, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24563256

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) has emerged as a leading contributor to mortality during recent influenza pandemics. The mechanism for this influenza-induced susceptibility to secondary S. aureus infection is poorly understood. In this study, we show that innate antibacterial immunity was significantly suppressed during the recovery stage of influenza infection, even though MRSA superinfection had no significant effect on viral burdens. Compared with mice infected with bacteria alone, postinfluenza MRSA-infected mice exhibited impaired bacterial clearance, which was not due to defective phagocyte recruitment, but rather coincided with reduced intracellular reactive oxygen species levels in alveolar macrophages and neutrophils. NADPH oxidase is responsible for reactive oxygen species production during phagocytic bacterial killing, a process also known as oxidative burst. We found that gp91(phox)-containing NADPH oxidase activity in macrophages and neutrophils was essential for optimal bacterial clearance during respiratory MRSA infections. In contrast to wild-type animals, gp91(phox-/-) mice exhibited similar defects in MRSA clearance before and after influenza infection. Using gp91(phox+/-) mosaic mice, we further demonstrate that influenza infection inhibits a cell-intrinsic contribution of NADPH oxidase to phagocyte bactericidal activity. Taken together, our results establish that influenza infection suppresses NADPH oxidase-dependent bacterial clearance and leads to susceptibility to secondary MRSA infection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , NADPH Oxidases/immunology , Orthomyxoviridae Infections/immunology , Phagocytes/immunology , Staphylococcal Infections/immunology , Animals , Cells , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Female , Flow Cytometry , Host-Pathogen Interactions/immunology , Influenza A Virus, H1N1 Subtype/physiology , Lung/immunology , Lung/microbiology , Lung/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Methicillin-Resistant Staphylococcus aureus/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Phagocytes/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology
20.
J Immunol ; 191(5): 2047-52, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23964104

ABSTRACT

Secondary pulmonary infections by encapsulated bacteria including Streptococcus pneumoniae and Staphylococcus aureus following influenza represent a common and challenging clinical problem. The reasons for this polymicrobial synergy are still not completely understood, hampering development of effective prophylactic and therapeutic interventions. Although it has been commonly thought that viral-induced epithelial cell damage allows bacterial invasiveness, recent studies by several groups have now implicated dysfunctional innate immune defenses following influenza as the primary culprit for enhanced susceptibility to secondary bacterial infections. Understanding the immunological imbalances that are responsible for virus/bacteria synergy will ultimately allow the design of effective, broad-spectrum therapeutic approaches for prevention of enhanced susceptibility to these pathogens.


Subject(s)
Bacterial Infections/complications , Bacterial Infections/immunology , Coinfection/immunology , Influenza, Human/complications , Influenza, Human/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...