Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Carbohydr Polym ; 337: 122156, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38710572

ABSTRACT

Seaweeds represent a rich source of sulfated polysaccharides with similarity to heparan sulfate, a facilitator of myriad virus host cell attachment. For this reason, attention has been drawn to their antiviral activity, including the potential for anti-SARS-CoV-2 activity. We have identified and structurally characterized several fucoidan extracts, including those from different species of brown macroalga, and a rhamnan sulfate from a green macroalga species. A high molecular weight fucoidan extracted from Saccharina japonica (FSjRPI-27), and a rhamnan sulfate extracted from Monostroma nitidum (RSMn), showed potent competitive inhibition of spike glycoprotein receptor binding to a heparin-coated SPR chip. This inhibition was also observed in cell-based assays using hACE2 HEK-293 T cells infected by pseudotyped SARS-CoV-2 virus with IC50 values <1 µg/mL. Effectiveness was demonstrated in vivo using hACE2-transgenic mice. Intranasal administration of FSjRPI-27 showed protection when dosed 6 h prior to and at infection, and then every 2 days post-infection, with 100 % survival and no toxicity at 104 plaque-forming units per mouse vs. buffer control. At 5-fold higher virus dose, FSjRPI-27 reduced mortality and yielded reduced viral titers in bronchioalveolar fluid and lung homogenates vs. buffer control. These findings suggest the potential application of seaweed-based sulfated polysaccharides as promising anti-SARS-CoV-2 prophylactics.


Subject(s)
Antiviral Agents , COVID-19 , Mannans , Polysaccharides , SARS-CoV-2 , Seaweed , Polysaccharides/chemistry , Polysaccharides/pharmacology , Animals , Humans , SARS-CoV-2/drug effects , Seaweed/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , HEK293 Cells , Mice , COVID-19/prevention & control , COVID-19/virology , COVID-19 Drug Treatment , Mice, Transgenic , Spike Glycoprotein, Coronavirus/metabolism , Deoxy Sugars/pharmacology , Deoxy Sugars/chemistry , Angiotensin-Converting Enzyme 2/metabolism
2.
Surg Open Sci ; 10: 83-90, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36039075

ABSTRACT

Background: Clinically useful biomaterials are derived from xenogeneic extracellular matrices, but extensive processes often used to remove all residual DNA are detrimental to their proper biological function. We hypothesized that deliberate and repeated injection of DNA extracted from clinically implantable, xenogeneic extracellular matrices might elicit an immune response in a well-established murine model that could ultimately lead to altered extracellular matrix remodeling. Methods: DNA was purified from unprocessed porcine extracellular matrices and processed extracellular matrices before sterilization (aseptic) and after sterilization. Groups of 10 mice were injected with these 3 purified DNAs and 3 controls: (1) DNA from E. coli; (2) DNA from unprocessed porcine extracellular matrices combined with interleukin-12 and methylated bovine serum albumin and emulsified in incomplete Freund's adjuvant; and (3) buffered saline. Immunizations occurred every 2 weeks for a total of 3 injections. Local cytokines and systemic anti-DNA antibodies were quantified 3 and 7 days after final injection. Results: The DNA extracted from unprocessed, aseptic, or sterilized porcine extracellular matrices failed to elicit a rejection response, and only with significant, proinflammatory adjuvant activation could such a response be seen. Without the adjuvants, biomaterial-derived DNA resulted in a mild accommodation cytokine response locally and no systemic anti-DNA antibody expression even at doses approximately 100-fold larger than would be clinically likely via extracellular matrix implantation. Conclusion: The immunological safety of porcine extracellular matrix biomaterials appears not to be related to DNA residues present. Such biomaterials need not be extensively processed, likely leading to detrimental changes in their bioactivity, solely in an effort to remove the mammalian DNA.

3.
JCI Insight ; 7(11)2022 06 08.
Article in English | MEDLINE | ID: mdl-35482422

ABSTRACT

Secondary infections are frequent complications of viral respiratory infections, but the potential consequence of SARS-CoV-2 coinfection with common pulmonary pathogens is poorly understood. We report that coinfection of human ACE2-transgenic mice with sublethal doses of SARS-CoV-2 and Streptococcus pneumoniae results in synergistic lung inflammation and lethality. Mortality was observed regardless of whether SARS-CoV-2 challenge occurred before or after establishment of sublethal pneumococcal infection. Increased bacterial levels following coinfection were associated with alveolar macrophage depletion, and treatment with murine GM-CSF reduced numbers of lung bacteria and pathology and partially protected from death. However, therapeutic targeting of IFNs, an approach that is effective against influenza coinfections, failed to increase survival. Combined vaccination against both SARS-CoV-2 and pneumococci resulted in 100% protection against subsequent coinfection. The results indicate that when seasonal respiratory infections return to prepandemic levels, they could lead to an increased incidence of lethal COVID-19 superinfections, especially among the unvaccinated population.


Subject(s)
COVID-19 , Coinfection , Animals , COVID-19/prevention & control , Mice , Mice, Transgenic , SARS-CoV-2 , Streptococcus pneumoniae , Vaccination
4.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35169077

ABSTRACT

Functional plasticity of innate lymphoid cells (ILCs) and T cells is regulated by host environmental cues, but the influence of pathogen-derived virulence factors has not been described. We now report the interplay between host interferon (IFN)-γ and viral PB1-F2 virulence protein in regulating the functions of ILC2s and T cells that lead to recovery from influenza virus infection of mice. In the absence of IFN-γ, lung ILC2s from mice challenged with the A/California/04/2009 (CA04) H1N1 virus, containing nonfunctional viral PB1-F2, initiated a robust IL-5 response, which also led to improved tissue integrity and increased survival. Conversely, challenge with Puerto Rico/8/1934 (PR8) H1N1 virus expressing fully functional PB1-F2, suppressed IL-5+ ILC2 responses, and induced a dominant IL-13+ CD8 T cell response, regardless of host IFN-γ expression. IFN-γ-deficient mice had increased survival and improved tissue integrity following challenge with lethal doses of CA04, but not PR8 virus, and increased resistance was dependent on the presence of IFN-γR+ ILC2s. Reverse-engineered influenza viruses differing in functional PB1-F2 activity induced ILC2 and T cell phenotypes similar to the PB1-F2 donor strains, demonstrating the potent role of viral PB1-F2 in host resistance. These results show the ability of a pathogen virulence factor together with host IFN-γ to regulate protective pulmonary immunity during influenza infection.


Subject(s)
Lymphocytes/immunology , Orthomyxoviridae/metabolism , Viral Proteins/metabolism , Animals , Female , Immunity, Innate/immunology , Interferon-gamma/metabolism , Interferons/metabolism , Interleukin-5/immunology , Interleukin-5/metabolism , Lung/metabolism , Lymphocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Viral Proteins/physiology , Virulence/genetics , Virulence Factors/genetics , Virus Replication/genetics
5.
Viruses ; 13(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34960631

ABSTRACT

Disease tolerance has emerged as an alternative way, in addition to host resistance, to survive viral-bacterial co-infections. Disease tolerance plays an important role not in reducing pathogen burden, but in maintaining tissue integrity and controlling organ damage. A common co-infection is the synergy observed between influenza virus and Streptococcus pneumoniae that results in superinfection and lethality. Several host cytokines and cells have shown promise in promoting tissue protection and damage control while others induce severe immunopathology leading to high levels of morbidity and mortality. The focus of this review is to describe the host cytokines and innate immune cells that mediate disease tolerance and lead to a return to host homeostasis and ultimately, survival during viral-bacterial co-infection.


Subject(s)
Immunity, Innate , Influenza, Human/immunology , Orthomyxoviridae/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Coinfection , Cytokines/immunology , Homeostasis , Humans , Influenza, Human/microbiology , Influenza, Human/virology , Pneumococcal Infections/microbiology , Superinfection
6.
J Virol ; 95(22): e0059821, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34468174

ABSTRACT

Interleukin-33 (IL-33) is a multifunctional cytokine that mediates type 2-dominated immune responses. In contrast, the role of IL-33 during viral vaccination, which often aims to induce type 1 immunity, has not been fully investigated. Here, we examined the effects of IL-33 on influenza vaccine responses. We found that intranasal coadministration of IL-33 with an inactivated influenza virus vaccine increases vaccine efficacy against influenza virus infection, not only with the homologous strain but also with heterologous strains, including the 2009 H1N1 influenza virus pandemic strain. Cross-protection was dependent on group 2 innate lymphoid cells (ILC2s), as the beneficial effect of IL-33 on vaccine efficacy was abrogated in ILC2-deficient C57BL/6 Il7rCre/+ Rorafl/fl mice. Furthermore, mechanistic studies revealed that IL-33-activated ILC2s potentiate vaccine efficacy by enhancing mucosal humoral immunity, particularly IgA responses, potentially in a Th2 cytokine-dependent manner. Our results demonstrate that IL-33-mediated activation of ILC2s is a critical early event that is important for the induction of mucosal humoral immunity, which in turn is responsible for cross-strain protection against influenza. Thus, we reveal a previously unrecognized role for the IL-33-ILC2 axis in establishing broadly protective and long-lasting humoral mucosal immunity against influenza, knowledge that may help in the development of a universal influenza vaccine. IMPORTANCE Current influenza vaccines, although capable of protecting against predicted viruses/strains included in the vaccine, are inept at providing cross-protection against emerging/novel strains. Thus, we are in critical need of a universal vaccine that can protect against a wide range of influenza viruses. Our novel findings show that a mucosal vaccination strategy involving the activation of lung ILC2s is highly effective in eliciting cross-protective humoral immunity in the lungs. This suggests that the biology of lung ILC2s can be exploited to increase the cross-reactivity of commercially available influenza subunit vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Interleukin-33/immunology , Orthomyxoviridae Infections/immunology , Vaccines, Inactivated/immunology , Animals , Antibodies, Viral/immunology , Cross Protection , Female , Immunity, Humoral , Lymphocytes/cytology , Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Vaccine Efficacy
7.
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
8.
J Clin Virol ; 141: 104879, 2021 08.
Article in English | MEDLINE | ID: mdl-34153860

ABSTRACT

Highly sensitive nucleic acid amplification tests (NAATs) designed to detect SARS-CoV-2 RNA are the standard of care for the diagnosis of COVID-19. However, the accuracy of these methods for the quantitation of active virus rather than non-infectious RNA fragments that can persist for extended periods of time has been unclear. This issue is particularly relevant for congregate care patients who are unable to return to their home residence until fully negative by NAATs. We tested paired samples from individual patients for the presence of virus at both early and later stages of disease. Culture of nasopharyngeal swab samples for 10 days in Vero E6 cells revealed active virus in only 4 out of 14 (28.6%) patients. The ability to isolate viral plaque-forming units (PFU) correlated with viral RNA loads of >6.79 log genomic copies/ml and only occurred in samples collected from patients early after symptom onset and before development of antibody. Culture in Vero E6 cells lacking the STAT1-dependent interferon signaling pathway increased the numbers of viral PFU detected but did not affect the incidence of positive cultures. We conclude that culturable virus is correlated with SARS-CoV-2 NAATs detection only during early symptom onset and with high viral titers/low antibody titers in non-immunosuppressed patients.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Nasopharynx , Polymerase Chain Reaction , RNA, Viral/genetics
9.
PLoS Pathog ; 17(3): e1009405, 2021 03.
Article in English | MEDLINE | ID: mdl-33690728

ABSTRACT

Bacterial co-infections represent a major clinical complication of influenza. Host-derived interferon (IFN) increases susceptibility to bacterial infections following influenza, but the relative roles of type-I versus type-II IFN remain poorly understood. We have used novel mouse models of co-infection in which colonizing pneumococci were inoculated into the upper respiratory tract; subsequent sublethal influenza virus infection caused the bacteria to enter the lungs and mediate lethal disease. Compared to wild-type mice or mice deficient in only one pathway, mice lacking both IFN pathways demonstrated the least amount of lung tissue damage and mortality following pneumococcal-influenza virus superinfection. Therapeutic neutralization of both type-I and type-II IFN pathways similarly provided optimal protection to co-infected wild-type mice. The most effective treatment regimen was staggered neutralization of the type-I IFN pathway early during co-infection combined with later neutralization of type-II IFN, which was consistent with the expression and reported activities of these IFNs during superinfection. These results are the first to directly compare the activities of type-I and type-II IFN during superinfection and provide new insights into potential host-directed targets for treatment of secondary bacterial infections during influenza.


Subject(s)
Coinfection/immunology , Interferons/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Pneumococcal/immunology , Superinfection/immunology , Animals , Disease Susceptibility , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/immunology
10.
FEBS Lett ; 594(17): 2782-2799, 2020 09.
Article in English | MEDLINE | ID: mdl-32484234

ABSTRACT

Intracellular pathogens affect diverse host cellular defence and metabolic pathways. Here, we used infection with Francisella tularensis to identify SON DNA-binding protein as a central determinant of macrophage activities. RNAi knockdown of SON increases survival of human macrophages following F. tularensis infection or inflammasome stimulation. SON is required for macrophage autophagy, interferon response factor 3 expression, type I interferon response and inflammasome-associated readouts. SON knockdown has gene- and stimulus-specific effects on inflammatory gene expression. SON is required for accurate splicing and expression of GBF1, a key mediator of cis-Golgi structure and function. Chemical GBF1 inhibition has similar effects to SON knockdown, suggesting that SON controls macrophage functions at least in part by controlling Golgi-associated processes.


Subject(s)
Autophagy/genetics , DNA-Binding Proteins/genetics , Francisella tularensis/pathogenicity , Golgi Apparatus/immunology , Guanine Nucleotide Exchange Factors/genetics , Host-Pathogen Interactions/genetics , Macrophages/immunology , Minor Histocompatibility Antigens/genetics , Autophagy/drug effects , Cell Death , Cell Differentiation/drug effects , Cell Line , Cell Survival , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/immunology , Francisella tularensis/genetics , Francisella tularensis/immunology , Gene Expression Profiling , Gene Expression Regulation , Golgi Apparatus/metabolism , Golgi Apparatus/microbiology , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/immunology , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Macrophages/metabolism , Macrophages/microbiology , Minor Histocompatibility Antigens/immunology , Pyridines/pharmacology , Quinolines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , THP-1 Cells , Tetradecanoylphorbol Acetate/pharmacology
11.
J Tissue Viability ; 29(1): 42-47, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31866230

ABSTRACT

BACKGROUND: The SIS Wound Matrix (SISWM) has been shown to improve healing of chronic ulcers over standard of care. In this study, we tested the hypothesis that chronic venous ulcers responsive to treatment with SISWM would more closely mimic an acute wound state as opposed to unresponsive ulcers. METHODS: Serum and wound exudate were collected at baseline and then weekly for up to 12 weeks from 12 patients receiving multiple applications of the SISWM. Levels of matrix metalloproteinases (MMP-1, MMP-2, MMP-3, MMP-9, and MMP-12), pro-inflammatory cytokines (IL-1ß, TNF-α, IL-8), and transforming growth factor beta (TGF-ß1) were evaluated. A variety of Th1/Th2 cytokines were also assayed, as were systemic anti-SIS and anti-α-gal antibody titers. RESULTS: Seven of the 12 patients eventually healed their wounds. Results showed significant decreases in MMP-1, MMP-2, MMP-3, MMP-9, TNF-α and IL-8, and significant increases in TGF-ß1 in wounds responding to treatment with the SISWM versus wounds that did not respond to treatment. None of the 12 patients formed a measurable serum antibody response to the SISWM. CONCLUSIONS: These data show that SISWM does not lead to immune system recognition or sensitization to the matrix and that wounds that went on to heal following treatment were characterized by a more acute wound state. The study confirms that the wound environment is important to healing and that turning a wound toward an acute biochemical state is key to the healing process.


Subject(s)
Leg Ulcer/therapy , Matrix Metalloproteinases/administration & dosage , Adolescent , Adult , Exudates and Transudates/immunology , Female , Humans , Leg Ulcer/blood , Male , Matrix Metalloproteinases/immunology , Treatment Outcome , Wound Healing , Young Adult
12.
J Bacteriol ; 202(4)2020 01 29.
Article in English | MEDLINE | ID: mdl-31767779

ABSTRACT

Streptococcus pneumoniae (the pneumococcus) is a naturally competent organism that causes diseases such as pneumonia, otitis media, and bacteremia. The essential bacterial second messenger cyclic di-AMP (c-di-AMP) is an emerging player in the stress responses of many pathogens. In S. pneumoniae, c-di-AMP is produced by a diadenylate cyclase, CdaA, and cleaved by phosphodiesterases Pde1 and Pde2. c-di-AMP binds a transporter of K+ (Trk) family protein, CabP, which subsequently halts K+ uptake via the transporter TrkH. Recently, it was reported that Pde1 and Pde2 are essential for pneumococcal virulence in mouse models of disease. To elucidate c-di-AMP-mediated transcription that may lead to changes in pathogenesis, we compared the transcriptomes of wild-type (WT) and Δpde1 Δpde2 strains by transcriptome sequencing (RNA-Seq) analysis. Notably, we found that many competence-associated genes are significantly upregulated in the Δpde1 Δpde2 strain compared to the WT. These genes play a role in DNA uptake, recombination, and autolysis. Competence is induced by a quorum-sensing mechanism initiated by the secreted factor competence-stimulating peptide (CSP). Surprisingly, the Δpde1 Δpde2 strain exhibited reduced transformation efficiency compared to WT bacteria, which was c-di-AMP dependent. Transformation efficiency was also directly related to the [K+] in the medium, suggesting a link between c-di-AMP function and the pneumococcal competence state. We found that a strain that possesses a V76G variation in CdaA produced less c-di-AMP and was highly susceptible to CSP. Deletion of cabP and trkH restored the growth of these bacteria in medium with CSP. Overall, our study demonstrates a novel role for c-di-AMP in the competence program of S. pneumoniaeIMPORTANCE Genetic competence in bacteria leads to horizontal gene transfer, which can ultimately affect antibiotic resistance, adaptation to stress conditions, and virulence. While the mechanisms of pneumococcal competence signaling cascades have been well characterized, the molecular mechanism behind competence regulation is not fully understood. The bacterial second messenger c-di-AMP has previously been shown to play a role in bacterial physiology and pathogenesis. In this study, we provide compelling evidence for the interplay between c-di-AMP and the pneumococcal competence state. These findings not only attribute a new biological function to this dinucleotide as a regulator of competence, transformation, and survival under stress conditions in pneumococci but also provide new insights into how pneumococcal competence is modulated.


Subject(s)
Dinucleoside Phosphates/physiology , Second Messenger Systems/physiology , Streptococcus pneumoniae/physiology , Bacterial Proteins/physiology , DNA-Binding Proteins/physiology , Glycine/pharmacology , Hydrogen-Ion Concentration , Potassium/metabolism , Sequence Analysis, RNA , Streptococcus pneumoniae/genetics , Transcriptome
13.
Vaccines (Basel) ; 7(4)2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31614565

ABSTRACT

Secondary bacterial pneumonia is responsible for significant morbidity and mortality during seasonal and pandemic influenza. Due to the unpredictability of influenza A virus evolution and the time-consuming process of manufacturing strain-specific influenza vaccines, recent efforts have been focused on developing anti-Streptococcus pneumoniae immunity to prevent influenza-related illness and death. Bacterial vaccination to prevent viral-bacterial synergistic interaction during co-infection is a promising concept that needs further investigation. Here, we show that immunization with pneumococcal surface protein A (PspA) fully protects mice against low-dose, but not high-dose, secondary bacterial challenge using a murine model of influenza A virus-S. pneumoniae co-infection. We further show that immunization with PspA is more broadly protective than the pneumococcal conjugate vaccine (Prevnar). These results demonstrate that PspA is a promising vaccine target that can provide protection against a physiologically relevant dose of S. pneumoniae following influenza infection.

14.
Aging Cell ; 18(6): e13019, 2019 12.
Article in English | MEDLINE | ID: mdl-31429526

ABSTRACT

The effects of aging on innate immunity and the resulting impacts on immunosenescence remain poorly understood. Here, we report that aging induces compartmentalized changes to the development and function of group 2 innate lymphoid cells (ILC2), an ILC subset implicated in pulmonary homeostasis and tissue repair. Aging enhances bone marrow early ILC2 development through Notch signaling, but the newly generated circulating ILC2 are unable to settle in the lungs to replenish the concomitantly declining mature lung ILC2 pool in aged mice. Aged lung ILC2 are transcriptomically heterogeneous and functionally compromised, failing to produce cytokines at homeostasis and during influenza infection. They have reduced expression of Cyp2e1, a cytochrome P450 oxidase required for optimal ILC2 function. Transfer of lung ILC2 from young mice enhances resistance to influenza infection in old mice. These data highlight compartmentalized effects of aging on ILC and indicate that targeting tissue-resident ILCs might unlock therapies to enhance resistance to infections and diseases in the elderly.


Subject(s)
Aging/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cellular Senescence/immunology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
mBio ; 10(4)2019 07 02.
Article in English | MEDLINE | ID: mdl-31266877

ABSTRACT

Fatal outcomes following influenza infection are often associated with secondary bacterial infections. Allergic airway disease (AAD) is known to influence severe complications from respiratory infections, and yet the mechanistic effect of AAD on influenza virus-Streptococcus pneumoniae coinfection has not been investigated previously. We examined the impact of AAD on host susceptibility to viral-bacterial coinfections. We report that AAD improved survival during coinfection when viral-bacterial challenge occurred 1 week after AAD. Counterintuitively, mice with AAD had significantly deceased proinflammatory responses during infection. Specifically, both CD4+ and CD8+ T cell interferon gamma (IFN-γ) responses were suppressed following AAD. Resistance to coinfection was also associated with strong transforming growth factor ß1 (TGF-ß1) expression and increased bacterial clearance. Treatment of AAD mice with IFN-γ or genetic deletion of TGF-ß receptor II expression reversed the protective effects of AAD. Using a novel triple-challenge model system, we show for the first time that AAD can provide protection against influenza virus-S. pneumoniae coinfection through the production of TGF-ß that suppresses the influenza virus-induced IFN-γ response, thereby preserving antibacterial immunity.IMPORTANCE Asthma has become one of the most common chronic diseases and has been identified as a risk factor for developing influenza. However, the impact of asthma on postinfluenza secondary bacterial infection is currently not known. Here, we developed a novel triple-challenge model of allergic airway disease, primary influenza infection, and secondary Streptococcus pneumoniae infection to investigate the impact of asthma on susceptibility to viral-bacterial coinfections. We report for the first time that mice recovering from acute allergic airway disease are highly resistant to influenza-pneumococcal coinfection and that this resistance is due to inhibition of influenza virus-mediated impairment of bacterial clearance. Further characterization of allergic airway disease-associated resistance against postinfluenza secondary bacterial infection may aid in the development of prophylactic and/or therapeutic treatment against coinfection.


Subject(s)
Asthma/complications , Coinfection/immunology , Coinfection/pathology , Disease Susceptibility , Influenza, Human/immunology , Influenza, Human/pathology , Pneumococcal Infections/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Humans , Influenza A virus/growth & development , Interferon-gamma/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Streptococcus pneumoniae/growth & development , Survival Analysis , Transforming Growth Factor beta/metabolism
16.
Respir Res ; 20(1): 99, 2019 May 22.
Article in English | MEDLINE | ID: mdl-31118031

ABSTRACT

BACKGROUND: Recurrent and persistent infections are known to affect airways of patients with Primary Immunodeficiency despite appropriate replacement immunoglobulin serum levels. Interestingly, patients with Chronic Obstructive Pulmonary Disease or with non-CF bronchiectasis also show similar susceptibility to such infections. This may be due to the limited availability of immunoglobulins from the systemic circulation in the conductive airways, resulting in local immunodeficiency. Topical application of nebulized plasma-derived immunoglobulins may represent a means to address this deficiency. In this study, we assessed the feasibility of nebulizing plasma-derived immunoglobulins and delivering them into the airways of rats and non-human primates. METHODS: Distinct human plasma-derived immunoglobulin isotype preparations were nebulized with an investigational eFlow® nebulizer and analyzed in vitro or deposited into animals. Biochemical and immunohistological analysis of nebulized immunoglobulins were then performed. Lastly, efficacy of topically applied human plasma-derived immunoglobulins was assessed in an acute Streptococcus pneumoniae respiratory infection in mice. RESULTS: Characteristics of the resulting aerosols were comparable between preparations, even when using solutions with elevated viscosity. Neither the structural integrity nor the biological function of nebulized immunoglobulins were compromised by the nebulization process. In animal studies, immunoglobulins levels were assessed in plasma, broncho-alveolar lavages (BAL) and on lung sections of rats and non-human primates in samples collected up to 72 h following application. Nebulized immunoglobulins were detectable over 48 h in the BAL samples and up to 72 h on lung sections. Immunoglobulins recovered from BAL fluid up to 24 h after inhalation remained structurally and functionally intact. Importantly, topical application of human plasma-derived immunoglobulin G into the airways of mice offered significant protection against acute pneumococcal pneumonia. CONCLUSION: Taken together our data demonstrate the feasibility of topically applying plasma-derived immunoglobulins into the lungs using a nebulized liquid formulation. Moreover, topically administered human plasma-derived immunoglobulins prevented acute respiratory infection.


Subject(s)
Immunoglobulin A/administration & dosage , Immunoglobulin G/administration & dosage , Immunoglobulin M/administration & dosage , Lung/drug effects , Nebulizers and Vaporizers/trends , Administration, Topical , Animals , Dose-Response Relationship, Drug , Humans , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Lung/metabolism , Macaca fascicularis , Mice, Inbred C57BL , Mice, Transgenic , Primates , Rats , Rats, Sprague-Dawley , Species Specificity
17.
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
18.
J Infect Dis ; 218(11): 1802-1812, 2018 10 20.
Article in English | MEDLINE | ID: mdl-29931113

ABSTRACT

Disease tolerance can preserve host homeostasis and limit the negative impact of infections. We report that vaccinated mice survived pulmonary challenge with the extremely virulent SchuS4 strain of Francisella tularensis for at least 100 days, despite the persistence of large numbers (~104) of organisms. Transfer of 100 of these resident bacteria to naive animals caused 100% lethality, demonstrating that virulence was maintained. Tissue damage in the lung was limited over the course of infection and was associated with increased levels of amphiregulin. Mice depleted of CD4+ cells had reduced amphiregulin and succumbed to infection. In addition, neutralization of interferon-γ or depletion of CD8+ cells resulted in increased pathogen loads, bacteremia, and death of the host. Conversely, depletion of Ly6G+ neutrophils had no effect on survival and actually resulted in reduced bacterial levels. Understanding the interplay between host resistance and disease tolerance will provide new insights into the understanding of chronic infectious diseases.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis , Tularemia/immunology , Vaccines, Attenuated/immunology , Animals , Disease Resistance/immunology , Female , Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Male , Mice , Mice, Inbred BALB C , Respiratory Tract Infections/immunology , Tularemia/prevention & control
19.
J Immunol ; 201(1): 134-144, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29760191

ABSTRACT

Secondary bacterial coinfections following influenza virus pose a serious threat to human health. Therefore, it is of significant clinical relevance to understand the immunological causes of this increased susceptibility. Influenza-induced alterations in alveolar macrophages (AMs) have been shown to be a major underlying cause of the increased susceptibility to bacterial superinfection. However, the mechanisms responsible for this remain under debate, specifically in terms of whether AMs are depleted in response to influenza infection or are maintained postinfection, but with disrupted phagocytic activity. The data presented in this article resolves this issue by showing that either mechanism can differentially occur in individual mouse strains. BALB/c mice exhibited a dramatic IFN-γ-dependent reduction in levels of AMs following infection with influenza A, whereas AM levels in C57BL/6 mice were maintained throughout the course of influenza infection, although the cells displayed an altered phenotype, namely an upregulation in CD11b expression. These strain differences were observed regardless of whether infection was performed with low or high doses of influenza virus. Furthermore, infection with either the H1N1 A/California/04/2009 (CA04) or H1N1 A/PR8/1934 (PR8) virus strain yielded similar results. Regardless of AM viability, both BALB/c and C57BL/6 mice showed a high level of susceptibility to postinfluenza bacterial infection. These findings resolve the apparent inconsistencies in the literature, identify mouse strain-dependent differences in the AM response to influenza infection, and ultimately may facilitate translation of the mouse model to clinical application.


Subject(s)
Coinfection/immunology , Disease Susceptibility/immunology , Influenza A Virus, H1N1 Subtype/immunology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/immunology , Superinfection/immunology , Animals , Cell Line , Chick Embryo , Coinfection/microbiology , Dogs , Female , Humans , Interferon-gamma/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/immunology , Superinfection/microbiology
20.
Viral Immunol ; 31(6): 470-471, 2018.
Article in English | MEDLINE | ID: mdl-29688839

ABSTRACT

Influenza infection is associated with enhanced pathology in individuals with sickle cell disease (SCD). Despite being a high priority group for annual influenza vaccination, little is known about long-term responses to influenza vaccination in this patient population. To model flu vaccination, we inoculated SCD and wild type (WT) littermate mice with the seasonal flu vaccine [containing pandemic H1N1 (pH1N1) antigen], bled the mice before and after vaccination, and intranasally challenged them with a high dose (400 PFU) of pH1N1 12 weeks later. Both WT and SCD mice were fully protected from infection, and anti-influenza immunoglobulin G titers were significantly elevated in both groups after vaccination. It appears that flu vaccination is effective in SCD mice and our data support the clinical practice of regular flu vaccination in SCD patients.


Subject(s)
Anemia, Sickle Cell/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Mass Vaccination/methods , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Disease Models, Animal , Female , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza Vaccines/immunology , Influenza, Human/blood , Influenza, Human/immunology , Mass Vaccination/standards , Mice , Practice Guidelines as Topic , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...