Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 493
Filter
1.
Vet Microbiol ; 264: 109303, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34923246

ABSTRACT

In this study, whether H9N2 influenza A virus (IAV) infection contributed to secondary Klebsiella pneumoniae infection was investigated. From post-infection onwards, clinical symptoms were monitored, examined and recorded daily for 11 days. As a result, no clinical signs were observed in the mice infected with single H9N2 IAV, implying that H9N2 IAV was less pathogenic to mice. Compared to single K. pneumonia infection, K. pneumoniae infection following H9N2 IAV infection exacerbates lung histopathological lesions and apoptosis, resulting in more severe diseases. Lung index of the mice with H9N2 IAV and K. pneumoniae co-infection was significantly higher than those in the other groups. Bacterial loads in the tissues in H9N2 IAV and K. pneumoniae co-infection group were significantly higher than those in the single K. pneumoniae infection group at 7 dpi. It demonstrated that prior H9N2 IAV infection contributed to K. pneumonia proliferation and delayed bacterial clearance in mice. Secondary K. pneumoniae infection influences seroconversion of anti-H9N2 antibody titers and the cytokine profiles. The findings demonstrated that H9N2 IAV infection facilitated secondary K. pneumonia infection, causing severe the diseases in mice.


Subject(s)
Influenza A Virus, H9N2 Subtype , Klebsiella pneumoniae , Orthomyxoviridae Infections , Pneumonia , Animals , Coinfection , Influenza A Virus, H9N2 Subtype/physiology , Klebsiella pneumoniae/physiology , Mice , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/virology , Pneumonia/microbiology , Pneumonia/virology
2.
Int J Mol Sci ; 22(22)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34829979

ABSTRACT

Influenza is a respiratory virus that alone or in combination with secondary bacterial pathogens can contribute to the development of acute pneumonia in persons >65 years of age. Host innate immune antiviral signaling early in response to influenza is essential to inhibit early viral replication and guide the initiation of adaptive immune responses. Using young adult (3 months) and aged adult mice infected with mouse adapted H1N1 or H3N2, the results of our study illustrate dysregulated and/or diminished activation of key signaling pathways in aged lung contribute to increased lung inflammation and morbidity. Specifically, within the first seven days of infection, there were significant changes in genes associated with TLR and RIG-I signaling detected in aged murine lung in response to H1N1 or H3N2. Taken together, the results of our study expand our current understanding of age-associated changes in antiviral signaling in the lung.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/genetics , Pneumonia/genetics , A549 Cells , Animals , DEAD Box Protein 58/genetics , Disease Models, Animal , Gene Expression Regulation, Viral/genetics , Humans , Immunity, Innate/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza, Human/microbiology , Influenza, Human/virology , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/virology , Pneumonia/microbiology , Pneumonia/virology , Toll-Like Receptors/genetics , Virus Replication/genetics
3.
Front Immunol ; 12: 732029, 2021.
Article in English | MEDLINE | ID: mdl-34804016

ABSTRACT

Transient receptor potential (TRP) channels, neuronal stimulations widely known to be associated with thermal responses, pain induction, and osmoregulation, have been shown in recent studies to have underlying mechanisms associated with inflammatory responses. The role of TRP channels on inflammatory milieu during bacterial infections has been widely demonstrated. It may vary among types of channels/pathogens, however, and it is not known how TRP channels function during pneumococcal infections. Streptococcus pneumoniae can cause severe infections such as pneumonia, bacteremia, and meningitis, with systemic inflammatory responses. This study examines the role of TRP channels (TRPV1 and TRPV4) for pneumococcal nasal colonization and subsequent development of invasive pneumococcal disease in a mouse model. Both TRPV1 and TRPV4 channels were shown to be related to regulation of the development of pneumococcal diseases. In particular, the influx of neutrophils (polymorphonuclear cells) in the nasal cavity and the bactericidal activity were significantly suppressed among TRPV4 knockout mice. This may lead to severe pneumococcal pneumonia, resulting in dissemination of the bacteria to various organs and causing high mortality during influenza virus coinfection. Regulating host immune responses by TRP channels could be a novel strategy against pathogenic microorganisms causing strong local/systemic inflammation.


Subject(s)
Nasal Mucosa/metabolism , Pneumococcal Infections/metabolism , Streptococcus pneumoniae/pathogenicity , TRPV Cation Channels/metabolism , Animals , Coinfection , Cytokines/metabolism , Disease Models, Animal , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Influenza A Virus, H3N2 Subtype/pathogenicity , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Nasal Mucosa/virology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/microbiology , Phagocytosis , Pneumococcal Infections/genetics , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Signal Transduction , Streptococcus pneumoniae/immunology , TRPV Cation Channels/genetics , Virulence
4.
mBio ; 12(3): e0326920, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34061598

ABSTRACT

Influenza A virus (IAV) infection predisposes the host to secondary bacterial pneumonia, known as a major cause of morbidity and mortality during influenza virus epidemics. Analysis of interactions between IAV-infected human epithelial cells and Streptococcus pneumoniae revealed that infected cells ectopically exhibited the endoplasmic reticulum chaperone glycoprotein 96 (GP96) on the surface. Importantly, efficient pneumococcal adherence to epithelial cells was imparted by interactions with extracellular GP96 and integrin αV, with the surface expression mediated by GP96 chaperone activity. Furthermore, abrogation of adherence was gained by chemical inhibition or genetic knockout of GP96 as well as addition of RGD peptide, an inhibitor of integrin-ligand interactions. Direct binding of extracellular GP96 and pneumococci was shown to be mediated by pneumococcal oligopeptide permease components. Additionally, IAV infection induced activation of calpains and Snail1, which are responsible for degradation and transcriptional repression of junctional proteins in the host, respectively, indicating increased bacterial translocation across the epithelial barrier. Notably, treatment of IAV-infected mice with the GP96 inhibitor enhanced pneumococcal clearance from lung tissues and ameliorated lung pathology. Taken together, the present findings indicate a viral-bacterial synergy in relation to disease progression and suggest a paradigm for developing novel therapeutic strategies tailored to inhibit pneumococcal colonization in an IAV-infected respiratory tract. IMPORTANCE Secondary bacterial pneumonia following an influenza A virus (IAV) infection is a major cause of morbidity and mortality. Although it is generally accepted that preceding IAV infection leads to increased susceptibility to secondary bacterial infection, details regarding the pathogenic mechanism during the early stage of superinfection remain elusive. Here, we focused on the interaction of IAV-infected cells and Streptococcus pneumoniae, which revealed that human epithelial cells infected with IAV exhibit a cell surface display of GP96, an endoplasmic reticulum chaperon. Notably, extracellular GP96 was shown to impart efficient adherence for secondary infection by S. pneumoniae, and GP96 inhibition ameliorated lung pathology of superinfected mice, indicating it to be a useful target for development of therapeutic strategies for patients with superinfection.


Subject(s)
Influenza A virus/pathogenicity , Influenza, Human/complications , Membrane Glycoproteins/genetics , Pneumonia, Bacterial/virology , Streptococcus pneumoniae/pathogenicity , Symptom Flare Up , A549 Cells , Animals , Bacterial Adhesion , Coinfection/complications , Coinfection/microbiology , Coinfection/virology , Epithelial Cells/microbiology , Female , Humans , Influenza, Human/virology , Lung/microbiology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/virology , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/pathology
5.
Biosci Rep ; 41(3)2021 03 26.
Article in English | MEDLINE | ID: mdl-33604601

ABSTRACT

Experimental experience suggests that microbial agents including probiotics and prebiotics (representative microbial agents) play a critical role in defending against respiratory virus infection. We aim to systematically examine these agents' effect on respiratory viral infection and encourage research into clinical applications. An electronic literature search was conducted from published data with a combination of a microbial agents search component containing synonyms for microbial agents-related terms and a customized search component for respiratory virus infection. Hazard ratio (HR), risk ratio (RR) and standard deviation (SD) were employed as effect estimates. In 45 preclinical studies, the mortality rates decreased in the respiratory viral infection models that included prebiotics or prebiotics as interventions (HR: 0.70; 95% confidence interval (CI): 0.56-0.87; P=0.002). There was a significant decrease in viral load due to improved gut microbiota (SD: -1.22; 95% CI: -1.50 to -0.94; P<0.001). Concentrations of interferon (IFN)-α (SD: 1.05; 95% CI: 0.33-1.77; P=0.004), IFN-γ (SD: 0.83; 95% CI: 0.01-1.65; P=0.05) and interleukin (IL)-12 (SD: 2.42; 95% CI: 0.32-4.52; P=0.02), IL-1ß (SD: 0.01; 95% CI: -0.37 to 0.40; P=0.94) increased, whereas those of TNF-α (SD: -0.58; 95% CI: -1.59 to 0.43; P=0.26) and IL-6 (SD: -0.59; 95% CI: -1.24 to 0.07; P=0.08) decreased. Six clinical studies had lower symptom scores (SD: -0.09; 95% CI: -0.44 to 0.26; P=0.61) and less incidence of infection (RR: 0.80; 95% CI: 0.64-1.01; P=0.06). Our research indicates that probiotics and prebiotics pose a defensive possibility on respiratory viral infection and may encourage the clinical application.


Subject(s)
Common Cold/microbiology , Orthomyxoviridae Infections/microbiology , Pneumonia, Viral/microbiology , Prebiotics/administration & dosage , Probiotics/therapeutic use , Animals , Common Cold/therapy , Gastrointestinal Microbiome , Humans , Interferons/metabolism , Interleukins/metabolism , Mice , Orthomyxoviridae Infections/therapy , Pneumonia, Viral/therapy
6.
Proc Natl Acad Sci U S A ; 117(49): 31386-31397, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229573

ABSTRACT

Influenza A virus (IAV)-related mortality is often due to secondary bacterial infections, primarily by pneumococci. Here, we study how IAV-modulated changes in the lungs affect bacterial replication in the lower respiratory tract (LRT). Bronchoalveolar lavages (BALs) from coinfected mice showed rapid bacterial proliferation 4 to 6 h after pneumococcal challenge. Metabolomic and quantitative proteomic analyses demonstrated capillary leakage with efflux of nutrients and antioxidants into the alveolar space. Pneumococcal adaptation to IAV-induced inflammation and redox imbalance increased the expression of the pneumococcal chaperone/protease HtrA. Presence of HtrA resulted in bacterial growth advantage in the IAV-infected LRT and protection from complement-mediated opsonophagocytosis due to capsular production. Absence of HtrA led to growth arrest in vitro that was partially restored by antioxidants. Pneumococcal ability to grow in the IAV-infected LRT depends on the nutrient-rich milieu with increased levels of antioxidants such as ascorbic acid and its ability to adapt to and cope with oxidative damage and immune clearance.


Subject(s)
Antioxidants/metabolism , Capillaries/pathology , Influenza, Human/microbiology , Pneumococcal Infections/microbiology , Respiratory System/microbiology , Respiratory System/virology , Streptococcus pneumoniae/growth & development , Animals , Bacterial Proteins/metabolism , Glucose/metabolism , Humans , Inflammation/complications , Inflammation/pathology , Mice, Inbred C57BL , Models, Biological , Molecular Chaperones/metabolism , Orthomyxoviridae Infections/microbiology , Oxidation-Reduction , Oxidative Stress , Phagocytosis , Respiratory System/pathology
7.
Nat Commun ; 11(1): 2537, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32439901

ABSTRACT

Infection with influenza can be aggravated by bacterial co-infections, which often results in disease exacerbation. The effects of influenza infection on the upper respiratory tract (URT) microbiome are largely unknown. Here, we report a longitudinal study to assess the temporal dynamics of the URT microbiomes of uninfected and influenza virus-infected humans and ferrets. Uninfected human patients and ferret URT microbiomes have stable healthy ecostate communities both within and between individuals. In contrast, infected patients and ferrets exhibit large changes in bacterial community composition over time and between individuals. The unhealthy ecostates of infected individuals progress towards the healthy ecostate, coinciding with viral clearance and recovery. Pseudomonadales associate statistically with the disturbed microbiomes of infected individuals. The dynamic and resilient microbiome during influenza virus infection in multiple hosts provides a compelling rationale for the maintenance of the microbiome homeostasis as a potential therapeutic target to prevent IAV associated bacterial co-infections.


Subject(s)
Influenza A virus/physiology , Influenza, Human/microbiology , Microbiota , Nasopharynx/microbiology , Adolescent , Adult , Aged , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Biodiversity , Child , Child, Preschool , Dysbiosis/microbiology , Dysbiosis/virology , Female , Ferrets , Humans , Infant , Influenza, Human/virology , Longitudinal Studies , Male , Microbiota/genetics , Middle Aged , Nasopharynx/virology , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/virology , Young Adult
8.
Genome Biol ; 21(1): 99, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32345342

ABSTRACT

BACKGROUND: Influenza is a severe respiratory illness that continually threatens global health. It has been widely known that gut microbiota modulates the host response to protect against influenza infection, but mechanistic details remain largely unknown. Here, we took advantage of the phenomenon of lethal dose 50 (LD50) and metagenomic sequencing analysis to identify specific anti-influenza gut microbes and analyze the underlying mechanism. RESULTS: Transferring fecal microbes from mice that survive virulent influenza H7N9 infection into antibiotic-treated mice confers resistance to infection. Some gut microbes exhibit differential features to lethal influenza infection depending on the infection outcome. Bifidobacterium pseudolongum and Bifidobacterium animalis levels are significantly elevated in surviving mice when compared to dead or mock-infected mice. Oral administration of B. animalis alone or the combination of both significantly reduces the severity of H7N9 infection in both antibiotic-treated and germ-free mice. Functional metagenomic analysis suggests that B. animalis mediates the anti-influenza effect via several specific metabolic molecules. In vivo tests confirm valine and coenzyme A produce an anti-influenza effect. CONCLUSIONS: These findings show that the severity of influenza infection is closely related to the heterogeneous responses of the gut microbiota. We demonstrate the anti-influenza effect of B. animalis, and also find that the gut population of endogenous B. animalis can expand to enhance host influenza resistance when lethal influenza infection occurs, representing a novel interaction between host and gut microbiota. Further, our data suggest the potential utility of Bifidobacterium in the prevention and as a prognostic predictor of influenza.


Subject(s)
Bifidobacterium animalis , Gastrointestinal Microbiome , Orthomyxoviridae Infections/prevention & control , Animals , Bifidobacterium/isolation & purification , Bifidobacterium animalis/isolation & purification , Bifidobacterium animalis/physiology , Coenzyme A/therapeutic use , Feces/microbiology , Influenza A Virus, H7N9 Subtype , Lethal Dose 50 , Lung/pathology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/pathology , Valine/therapeutic use
9.
Microbes Infect ; 22(8): 312-321, 2020 09.
Article in English | MEDLINE | ID: mdl-31958572

ABSTRACT

Pneumococcal conjugate vaccination (PCV) may prevent influenza-related pneumonia, including Streptococcus pneumoniae pneumonia. To investigate PCV efficacy against secondary pneumococcal pneumonia following influenza, PCV was administered intramuscularly 2 and 5 weeks before S. pneumoniae serotype-3 colonization of murine nasopharynges followed by intranasal challenge with a sublethal dose of influenza A virus. Bacterial and viral loads, including innate immune responses were compared across conditions. PCV vaccination improved the survival of mice with secondary pneumococcal pneumonia and significantly reduced the pulmonary bacterial burden. Increased monocyte/macrophage influx into the lungs, alleviated loss of alveolar macrophages and decreased neutrophil influx into the lungs occurred in PCV-treated mice irrespective of pneumococcal colonization. Higher monocyte chemoattractant protein 1 levels and lower levels of CXCL1, interferon-γ, interleukin-17A, and IL-10, were detected in PCV-treated mice. Additionally, PCV treatment activated the macrophage intracellular killing of S. pneumoniae. Collectively, PCV potentially modulates the host's innate immunity and specific antibodies induction. Macrophage-related innate immunity should be further explored to elucidate the efficacy and mechanisms of PCV versus influenza-related life-threatening diseases.


Subject(s)
Coinfection/immunology , Immunity, Innate , Macrophages/immunology , Orthomyxoviridae Infections/immunology , Pneumococcal Vaccines/immunology , Pneumonia, Pneumococcal/immunology , Animals , Antibodies, Bacterial/blood , B7-2 Antigen/metabolism , Bacterial Load , Coinfection/microbiology , Coinfection/mortality , Coinfection/virology , Cytokines/metabolism , Disease Models, Animal , Influenza A virus , Lung/immunology , Lung/microbiology , Lung/virology , Macrophages/microbiology , Mice , Neutrophils/immunology , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Phagocytosis , Pneumococcal Vaccines/administration & dosage , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/mortality , Pneumonia, Pneumococcal/virology , Streptococcus pneumoniae , Survival Rate , Vaccination , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
10.
Vet Microbiol ; 240: 108542, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31902499

ABSTRACT

Influenza A virus (IAV) and bacteria co-infection can influence the host clinical conditions. Both H9N2 IAV and Pseudomonas aeruginosa (P. aeruginosa) are potential pathogens of respiratory diseases in mink. In this study, to clarify the effects of H9N2 IAV and P. aeruginosa co-infections on hemorrhagic pneumonia in mink, we carried out to establish the mink models of the two-pathogen co-infections in different orders. Compared with the single infections with H9N2 IAV or P. aeruginosa, the mink co-infected with H9N2 IAV and P. aeruginosa showed severe respiratory diseases, and exacerbated histopathological lesions and more obvious apoptosis in the lung tissues. H9N2 IAV shedding and viral loads in the lungs of the mink co-infected with H9N2 IAV and P. aeruginosa were higher than those in the mink with single H9N2 IAV infection. Furthermore, the clearance of P. aeruginosa in the co-infected mink lungs was delayed. In addition, the anti-H9N2 antibody titers in mink with P. aeruginosa co-infection following H9N2 IAV infection were significantly higher than those of the other groups. This implied that H9N2 IAV and P. aeruginosa co-infection contributed to the development of hemorrhagic pneumonia in mink, and that P. aeruginosa should play a major role in the disease. The exact interaction mechanism among H9N2 IAV, P. aeruginosa and the host needs to be further investigated.


Subject(s)
Coinfection/veterinary , Hemorrhage/veterinary , Influenza A Virus, H9N2 Subtype/pathogenicity , Orthomyxoviridae Infections/veterinary , Pneumonia/veterinary , Pseudomonas Infections/veterinary , Pseudomonas aeruginosa/pathogenicity , Animals , Antibodies, Viral/blood , Coinfection/microbiology , Coinfection/virology , Hemorrhage/microbiology , Hemorrhage/virology , Humans , Lung/pathology , Lung/virology , Mink/microbiology , Mink/virology , Orthomyxoviridae Infections/microbiology , Pneumonia/microbiology , Pneumonia/virology , Pseudomonas Infections/virology , Virus Replication , Virus Shedding
11.
J Biol Regul Homeost Agents ; 33(5): 1437-1449, 2019.
Article in English | MEDLINE | ID: mdl-31637902

ABSTRACT

Influenza has frequently been epidemic in recent years. However, the mechanisms of severe pneumonia with postinfluenza Streptococcus pneumoniae (SP) secondary infection have not been fully understood. In this study, we explored the mechanisms of pneumonia in postinfluenza A virus (IAV) infection via a mouse model. Mice were intranasally inoculated with SP three days after IAV inoculation. We then collected samples at three time points to dynamically observe the pathological progression. In IAV infection alone, lymphocyte infiltration and widened alveolar intervals were observed. In the blood, levels of the CD19+, CD19+CD21+ and CD19+CD79ß+B lymphocyte subpopulations were reduced, and IFN-γ and IL-10 were elevated. Slight atrophy was seen in the spleen, which was due to splenic B lymphocyteinitiated apoptosis through the mitochondrial pathway. When SP infection occurred after IAV infection, the pulmonary inflammation was significantly aggravated; a fair number of lymphocytes and neutrophils infiltrated simultaneously with exfoliated bronchial epithelial cells, vascular endothelial cells, widened alveolar septum and hemorrhaging. Increasing edema fluid and bacteria accumulated in the alveolar cavity. Decreased CD19+, CD19+CD21+ and CD19+CD79ß+B lymphocyte subpopulations and increased interferon gamma (IFN-γ) or interleukin 10 (IL-10) were more prominent compared to those with viral infection alone. Spleen atrophy resulting from coinfection was more obvious because of massive splenic B lymphocyte apoptosis through the mitochondrial pathway compared to viral infection alone. This study shows that although inflammation caused by SP infection alone was temporary, preceding IAV infection provided favorable conditions for SP colonization and multiplication by destroying lung structure and suppressing humoral immunity. Synergistic IAV-SP coinfection is likely to facilitate more SP colonization and promote B lymphocyte-suppression and reduction. Eventually, the pneumonia worsened.


Subject(s)
B-Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Pneumococcal Infections/immunology , Pneumonia, Bacterial/immunology , Animals , Apoptosis , B-Lymphocytes/cytology , Coinfection/microbiology , Coinfection/virology , Endothelial Cells , Influenza A virus , Lung , Mice , Orthomyxoviridae Infections/microbiology , Pneumococcal Infections/virology , Streptococcus pneumoniae
12.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31138613

ABSTRACT

Streptococcus suis is an important zoonotic pathogen which can infect humans and pigs worldwide, posing a potential risk to global public health. Suilysin, a pore-forming cholesterol-dependent cytolysin, is considered to play an important role in the pathogenesis of S. suis infections. It is known that infection with influenza A viruses may favor susceptibility to secondary bacterial infection, resulting in more severe disease and increased mortality. However, the molecular mechanisms underlying these coinfections are incompletely understood. Applying highly differentiated primary porcine respiratory epithelial cells grown under air-liquid interface (ALI) conditions, we analyzed the contribution of swine influenza viruses (SIV) to the virulence of S. suis, with a special focus on its cytolytic toxin, suilysin. We found that during secondary bacterial infection, suilysin of S. suis contributed to the damage of well-differentiated respiratory epithelial cells in the early stage of infection, whereas the cytotoxic effects induced by SIV became prominent at later stages of infection. Prior infection by SIV enhanced the adherence to and colonization of porcine airway epithelial cells by a wild-type (wt) S. suis strain and a suilysin-negative S. suis mutant in a sialic acid-dependent manner. A striking difference was observed with respect to bacterial invasion. After bacterial monoinfection, only the wt S. suis strain showed an invasive phenotype, whereas the mutant remained adherent. When the epithelial cells were preinfected with SIV, the suilysin-negative mutant also showed an invasion capacity. Therefore, we propose that coinfection with SIV may compensate for the lack of suilysin in the adherence and invasion process of suilysin-negative S. suis.


Subject(s)
Bacterial Adhesion/physiology , Coinfection/microbiology , Hemolysin Proteins/physiology , Lung/microbiology , Orthomyxoviridae Infections/microbiology , Streptococcus suis/pathogenicity , Animals , Cells, Cultured , Dogs , Epithelial Cells/microbiology , Swine
13.
mBio ; 10(3)2019 05 07.
Article in English | MEDLINE | ID: mdl-31064834

ABSTRACT

We previously reported that the Toll-like receptor 4 (TLR4) antagonist Eritoran blocks acute lung injury (ALI) therapeutically in mouse and cotton rat models of influenza. However, secondary (2°) bacterial infection following influenza virus infection is associated with excess morbidity and mortality. Wild-type (WT) mice infected with mouse-adapted influenza A/Puerto Rico/8/34 virus (PR8) and, 7 days later, with Streptococcus pneumoniae serotype 3 (Sp3) exhibited significantly enhanced lung pathology and lethality that was reversed by Eritoran therapy after PR8 infection but before Sp3 infection. Cotton rats infected with nonadapted pH1N1 influenza virus and then superinfected with methicillin-resistant Staphylococcus aureus also exhibited increased lung pathology and serum high-mobility-group box 1 (HMGB1) levels, both of which were blunted by Eritoran therapy. In mice, PR8 infection suppressed Sp3-induced CXCL1 and CXCL2 mRNA, reducing neutrophil infiltration and increasing the bacterial burden, all of which were reversed by Eritoran treatment. While beta interferon (IFN-ß)-deficient (IFN-ß-/-) mice are highly susceptible to PR8, they exhibited delayed death upon Sp3 superinfection, indicating that while IFN-ß was protective against influenza, it negatively impacted the host response to Sp3 IFN-ß-treated WT macrophages selectively suppressed Sp3-induced CXCL1/CXCL2 transcriptionally, as evidenced by reduced recruitment of RNA polymerase II to the CXCL1 promoter. Thus, influenza establishes a "trained" state of immunosuppression toward 2° bacterial infection, in part through the potent induction of IFN-ß and its downstream transcriptional regulation of chemokines, an effect reversed by Eritoran.IMPORTANCE Enhanced susceptibility to 2° bacterial infections following infection with influenza virus is a global health concern that accounts for many hospitalizations and deaths, particularly during pandemics. The complexity of the impaired host immune response during 2° bacterial infection has been widely studied. Both type I IFN and neutrophil dysfunction through decreased chemokine production have been implicated as mechanisms underlying enhanced susceptibility to 2° bacterial infections. Our findings support the conclusion that selective suppression of CXCL1/CXCL2 represents an IFN-ß-mediated "training" of the macrophage transcriptional response to TLR2 agonists and that blocking of TLR4 therapeutically with Eritoran after influenza virus infection reverses this suppression by blunting influenza-induced IFN-ß.


Subject(s)
Coinfection/microbiology , Lung/microbiology , Orthomyxoviridae Infections/microbiology , Superinfection , Acute Lung Injury/microbiology , Acute Lung Injury/virology , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Disaccharides/administration & dosage , Disease Susceptibility , Female , Immunocompromised Host , Influenza A virus , Interferon-beta/immunology , Male , Methicillin-Resistant Staphylococcus aureus , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/complications , Sigmodontinae , Streptococcus pneumoniae/immunology , Sugar Phosphates/administration & dosage , Toll-Like Receptor 4/immunology
14.
Vet Microbiol ; 231: 246-253, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30955818

ABSTRACT

Bovine respiratory disease (BRD) is economically significant, and influenza D virus (IDV) is commonly identified in cattle with BRD. Mannheimia haemolytica (MHA) is an opportunistic bacterial contributor to BRD; surveillance data suggest that MHA and IDV co-infection occurs in cattle. The objective of this study was to evaluate the synergistic pathogenesis in cattle co-infected with IDV and MHA. Sixteen dairy calves were randomly assigned to four groups of four calves. The IDV + MHA + group received D/bovine/C00046 N/Mississippi/2014 (D/46 N) intranasally at 0 days post-inoculation (DPI) and Mannheimia haemolytica D153 (MHA D153) intratracheally at 5 DPI. The IDV + MHA- group received only D/46 N at 0 DPI; the IDV-MHA + group received only MHA D153 at 5 DPI; and the IDV-MHA- group received neither agent. Clinical scores were calculated twice daily. At 10 DPI, IDV + MHA+, IDV-MHA+, and IDV-MHA- calves were euthanized and evaluated for pathologic lesions. The IDV + groups seroconverted to IDV by 10 DPI. Clinical scores were higher in IDV + groups than IDV- groups on 2-5 DPI (p = 0.001). After MHA challenge on 5 DPI, clinical scores (6-10 DPI) were slightly lower in IDV+MHA+ group than IDV-MHA+ group (p < 0.05) but not significantly different between MHA+ groups and MHA- groups. The average gross pathology score was higher for IDV-MHA+ group than groups IDV-MHA- and IDV+MHA+; however, no significant differences were identified among groups. Under the conditions of this study, infection with IDV before MHA enhance neither clinical disease nor lung pathology, relative to calves infected with MHA alone.


Subject(s)
Cattle Diseases/pathology , Coinfection/veterinary , Orthomyxoviridae Infections/veterinary , Pasteurellaceae Infections/veterinary , Respiratory Tract Infections/veterinary , Animals , Cattle , Cattle Diseases/microbiology , Cattle Diseases/virology , Coinfection/microbiology , Coinfection/pathology , Coinfection/virology , Lung/microbiology , Lung/pathology , Lung/virology , Male , Mannheimia haemolytica/pathogenicity , Orthomyxoviridae Infections/microbiology , Pasteurellaceae Infections/virology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Seroconversion , Thogotovirus/pathogenicity
15.
EMBO J ; 38(1)2019 01 03.
Article in English | MEDLINE | ID: mdl-30322895

ABSTRACT

The transcription factor, T-bet, regulates type 1 inflammatory responses against a range of infections. Here, we demonstrate a previously unaddressed role of T-bet, to influenza virus and bacterial superinfection. Interestingly, we found that T-bet deficiency did not adversely affect the efficacy of viral clearance or recovery compared to wild-type hosts. Instead, increased infiltration of neutrophils and production of Th17 cytokines (IL-17 and IL-22), in lungs of influenza virus-infected T-bet-/- mice, were correlated with survival advantage against subsequent infection by Streptococcus pneumoniae Neutralization of IL-17, but not IL-22, in T-bet-/- mice increased pulmonary bacterial load, concomitant with decreased neutrophil infiltration and reduced survival of T-bet-/- mice. IL-17 production by CD8+, CD4+ and γδ T cell types was identified to contribute to this protection against bacterial superinfection. We further showed that neutrophil depletion in T-bet-/- lungs increased pulmonary bacterial burden. These results thus indicate that despite the loss of T-bet, immune defences required for influenza viral clearance are fully functional, which in turn enhances protective type 17 immune responses against lethal bacterial superinfections.


Subject(s)
Orthomyxoviridae Infections/mortality , Superinfection/mortality , T-Box Domain Proteins/genetics , Animals , Coinfection , Dogs , Female , Gene Deletion , Influenza A Virus, H1N1 Subtype/pathogenicity , Interleukin-17/metabolism , Interleukins/metabolism , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/genetics , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/microbiology , Pneumococcal Infections/complications , Pneumococcal Infections/genetics , Pneumococcal Infections/mortality , Pneumococcal Infections/virology , Streptococcus pneumoniae/pathogenicity , Superinfection/genetics , Superinfection/microbiology , Superinfection/virology , Interleukin-22
16.
Front Immunol ; 9: 2589, 2018.
Article in English | MEDLINE | ID: mdl-30473701

ABSTRACT

Influenza virus infections particularly when followed by bacterial superinfections (BSI) result in significant morbidities and mortalities especially during influenza pandemics. Type I interferons (IFNs) regulate both anti-influenza immunity and host susceptibility to subsequent BSIs. These type I IFNs consisting of, among others, 14 IFN-α's and a single IFN-ß, are recognized by and signal through the heterodimeric type I IFN receptor (IFNAR) comprised of IFNAR1 and IFNAR2. However, the individual receptor subunits can bind IFN-ß or IFN-α's independently of each other and induce distinct signaling. The role of type I IFN signaling in regulating host susceptibility to both viral infections and BSI has been only examined with respect to IFNAR1 deficiency. Here, we demonstrate that despite some redundancies, IFNAR1 and IFNAR2 have distinct roles in regulating both anti-influenza A virus (IAV) immunity and in shaping host susceptibility to subsequent BSI caused by S. aureus. We found IFNAR2 to be critical for anti-viral immunity. In contrast to Ifnar1-/- mice, IAV-infected Ifnar2-/- mice displayed both increased and accelerated morbidity and mortality compared to WT mice. Furthermore, unlike IFNAR1, IFNAR2 was sufficient to generate protection from lethal IAV infection when stimulated with IFN-ß. With regards to BSI, unlike what we found previously in Ifnar1-/- mice, Ifnar2-/- mice were not susceptible to BSI induced on day 3 post-IAV, even though absence of IFNAR2 resulted in increased viral burden and an increased inflammatory environment. The Ifnar2-/- mice similar to what we previously found in Ifnar1-/- mice were less susceptible than WT mice to BSI induced on day 7 post-IAV, indicating that signaling through a complete receptor increases BSI susceptibility late during clinical IAV infection. Thus, our results support a role for IFNAR2 in induction of anti-IAV immune responses that are involved in altering host susceptibility to BSI and are essential for decreasing the morbidity and mortality associated with IAV infection. These results begin to elucidate some of the mechanisms involved in how the individual IFNAR subunits shape the anti-viral immune response. Moreover, our results highlight the importance of examining the contributions of entire receptors, as individual subunits can induce distinct outcomes as shown here.


Subject(s)
Orthomyxoviridae Infections/immunology , Receptor, Interferon alpha-beta/immunology , Staphylococcal Infections/immunology , Superinfection/immunology , Animals , Disease Susceptibility/immunology , Female , Influenza A virus/immunology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/microbiology , Staphylococcus aureus/immunology , Superinfection/microbiology , Vaccination/methods
17.
Methods Mol Biol ; 1836: 375-386, 2018.
Article in English | MEDLINE | ID: mdl-30151583

ABSTRACT

Investigation of pathogen-host interactions on a molecular level requires sophisticated in vitro infection procedures, especially in the presence of different pathogens.Super-infections of influenza viruses (IV) and bacteria, with increasing incidence of Staphylococcus aureus (S. aureus) cases, are a long-known phenomenon and represent a major complication in IV-infected patients. Although several in vivo studies have improved our knowledge about pathogenesis and immune responses of super-infections that result in increased morbidity and mortality, the consequences of the direct interplay of viruses and bacteria on a molecular level in affected cells that may contribute to the deadly synergism of these pathogens are so far poorly characterized. Here we describe different infection schemes to study IV and S. aureus coinfections of distinct cell populations in vitro. Depending on the focus of interest, regulation of cell responses such as signalling mechanisms or pro- and anti-inflammatory cytokine expression, or consequences for the viral or bacterial life cycle, can be analyzed. The described infection procedures could be used as guidelines and adapted to super-infection settings of other viral and bacterial pathogens.


Subject(s)
Coinfection , Host-Pathogen Interactions , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae/physiology , Staphylococcal Infections/virology , Staphylococcus aureus/physiology , Biomarkers , Blotting, Western , Cell Line , Fluorescent Antibody Technique , Humans , Orthomyxoviridae Infections/metabolism , Staphylococcal Infections/metabolism
18.
Methods Mol Biol ; 1725: 91-99, 2018.
Article in English | MEDLINE | ID: mdl-29322411

ABSTRACT

The lung is constantly exposed to both environmental and microbial challenge. As a "contained" organ, it also constitutes an excellent "self-contained" tissue to examine inflammatory responses and cellular infiltration into a diseased organ. Influenza A virus (IAV) causes both mild and severe inflammation that is strain specific following infection of the lung epithelium that spreads to other cells of the lung environment. Here, we describe a method of intranasal inoculation of the lung with IAV that can be used as a preclinical model of infection. Mice can be monitored for clinical signs of infection and tissue and lung fluid collected for further analysis to dissect the immunological consequences of IAV infection. Importantly, this method can be modified to introduce other pathogens, therapies and environmental stimuli to examine immune responses in the lung.


Subject(s)
Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Influenza A virus/pathogenicity , Lung/immunology , Orthomyxoviridae Infections/complications , Pneumonia/etiology , Administration, Intranasal , Animals , Bronchoalveolar Lavage Fluid/microbiology , Cytokines/metabolism , Female , Lung/metabolism , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/microbiology , Pneumonia/pathology
19.
Microbiome ; 6(1): 9, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321057

ABSTRACT

BACKGROUND: Microbiota integrity is essential for a growing number of physiological processes. Consequently, disruption of microbiota homeostasis correlates with a variety of pathological states. Importantly, commensal microbiota provide a shield against invading bacterial pathogens, probably by direct competition. The impact of viral infections on host microbiota composition and dynamics is poorly understood. Influenza A viruses (IAV) are common respiratory pathogens causing acute infections. Here, we show dynamic changes in respiratory and intestinal microbiota over the course of a sublethal IAV infection in a mouse model. RESULTS: Using a combination of 16S rRNA gene-specific next generation sequencing and qPCR as well as culturing of bacterial organ content, we found body site-specific and transient microbiota responses. In the lower respiratory tract, we observed only minor qualitative changes in microbiota composition. No quantitative impact on bacterial colonization after IAV infection was detectable, despite a robust antimicrobial host response and increased sensitivity to bacterial super infection. In contrast, in the intestine, IAV induced robust depletion of bacterial content, disruption of mucus layer integrity, and higher levels of antimicrobial peptides in Paneth cells. As a functional consequence of IAV-mediated microbiota depletion, we demonstrated that the small intestine is rendered more susceptible to bacterial pathogen invasion, in a Salmonella typhimurium super infection model. CONCLUSION: We show for the first time the consequences of IAV infection for lower respiratory tract and intestinal microbiobiota in a qualitative and quantitative fashion. The discrepancy of relative 16S rRNA gene next-generation sequencing (NGS) and normalized 16S rRNA gene-specific qPCR stresses the importance of combining qualitative and quantitative approaches to correctly analyze composition of organ associated microbial communities. The transiently induced dysbiosis underlines the overall stability of microbial communities to effects of acute infection. However, during a short-time window, specific ecological niches might lose their microbiota shield and remain vulnerable to bacterial invasion.


Subject(s)
Bacteria/classification , Orthomyxoviridae Infections/microbiology , Paneth Cells/microbiology , RNA, Ribosomal, 16S/genetics , Animals , Bacteria/genetics , Bacteria/isolation & purification , DNA, Bacterial/genetics , DNA, Ribosomal/genetics , Disease Models, Animal , Dysbiosis/microbiology , Female , Gastrointestinal Microbiome , Influenza A virus/pathogenicity , Mice , Sequence Analysis, DNA
20.
PLoS One ; 12(9): e0185387, 2017.
Article in English | MEDLINE | ID: mdl-28945819

ABSTRACT

Pneumocystis carinii f. sp. suis (PCS) nucleic acid and antibody profiles on two Austrian-farrow-to-finish farms were investigated. Furthermore, associations with other respiratory pathogens were evaluated. Respiratory specimen and sera from pigs of five age classes between the 1st week and the 3rd month of life as well as samples from sows were analyzed. On Farm A, PCS infection occurred early in life. The suckling piglets were already infected in the 1st week of life and the pigs remained positive until the 3rd month of life. On Farm B, pigs were infected later, between 3 and 4 months of age. The maximum PCS nucleic acid load on Farm A was 8.3 log10 genome copies/mL BALF, whereas on Farm B the PCS burden was significantly lower, with 4.0 log10 genome copies/mL BALF. Anti-PCS antibodies were detected in sows, as maternal antibodies in suckling piglets and as an immunological reaction to infection. On both farms, PCS infection was accompanied by several co-infections. On Farm A, there were concurrent infections with PRRSV, a virulent strain of Haemophilus parasuis, and Mycoplasma hyopneumoniae. On Farm B, PCS was accompanied by infections with swine influenza virus, Mycoplasma hyopneumoniae, and a non-virulent strain of Haemophilus parasuis. The results clearly show that the PCS profiles can vary between farms. Younger pigs may be more susceptible as they had higher PCS burdens. It is possible that PCS may contribute to a respiratory disease in pigs and further investigation of its potential role is warranted.


Subject(s)
Pneumocystis carinii/pathogenicity , Pneumonia, Pneumocystis/veterinary , Swine Diseases/microbiology , Age Factors , Animals , Animals, Newborn , Antibodies, Fungal/analysis , Antibodies, Fungal/blood , Austria , Coinfection/immunology , Coinfection/microbiology , Coinfection/veterinary , Cross-Sectional Studies , DNA, Bacterial/analysis , DNA, Bacterial/blood , DNA, Bacterial/genetics , DNA, Fungal/analysis , DNA, Fungal/blood , DNA, Fungal/genetics , DNA, Viral/analysis , DNA, Viral/blood , DNA, Viral/genetics , Female , Haemophilus Infections/immunology , Haemophilus Infections/microbiology , Haemophilus Infections/veterinary , Haemophilus parasuis/genetics , Haemophilus parasuis/isolation & purification , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/veterinary , Pneumocystis carinii/genetics , Pneumocystis carinii/immunology , Pneumonia of Swine, Mycoplasmal/immunology , Pneumonia of Swine, Mycoplasmal/microbiology , Pneumonia, Pneumocystis/immunology , Pneumonia, Pneumocystis/microbiology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/microbiology , Porcine Reproductive and Respiratory Syndrome/virology , Sus scrofa , Swine , Swine Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...