Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(5): 841-854, 2023 05.
Article in English | MEDLINE | ID: mdl-36928412

ABSTRACT

Regulatory T (Treg) cells are an immunosuppressive population that are required to maintain peripheral tolerance and prevent tissue damage from immunopathology, via anti-inflammatory cytokines, inhibitor receptors and metabolic disruption. Here we show that Treg cells acquire an effector-like state, yet remain stable and functional, when exposed to interferon gamma (IFNγ) during infection with lymphocytic choriomeningitis and influenza A virus. Treg cell-restricted deletion of the IFNγ receptor (encoded by Ifngr1), but not the interleukin 12 (IL12) receptor (encoded by Il12rb2), prevented TH1-like polarization (decreased expression of T-bet, CXC motif chemokine receptor 3 and IFNγ) and promoted TH2-like polarization (increased expression of GATA-3, CCR4 and IL4). TH1-like Treg cells limited CD8+ T cell effector function, proliferation and memory formation during acute and chronic infection. These findings provide fundamental insights into how Treg cells sense inflammatory cues from the environment (such as IFNγ) during viral infection to provide guidance to the effector immune response. This regulatory circuit prevents prolonged immunoinflammatory responses and shapes the quality and quantity of the memory T cell response.


Subject(s)
Interferon-gamma , T-Lymphocytes, Regulatory , Interferon-gamma/metabolism , Cytokines/metabolism , CD8-Positive T-Lymphocytes , Antiviral Agents/metabolism , Th1 Cells
2.
PLoS Pathog ; 20(2): e1011840, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38315735

ABSTRACT

Human metapneumovirus (HMPV) is an important cause of acute lower respiratory infection in children and adults worldwide. There are four genetic subgroups of HMPV and both neutralizing antibodies and T cells contribute to protection. However, little is known about mechanisms of pathogenesis and most published work is based on a few extensively passaged, laboratory-adapted strains of HMPV. In this study, we isolated and characterized a panel of low passage HMPV clinical isolates representing all four genetic subgroups. The clinical isolates exhibited lower levels of in vitro replication compared to a lab-adapted strain. We compared disease phenotypes using a well-established mouse model. Several virulent isolates caused severe weight loss, lung pathology, airway dysfunction, and fatal disease in mice, which was confirmed in three inbred mouse strains. Disease severity did not correlate with lung viral titer, as virulent strains exhibited restricted replication in the lower airway. Virulent HMPV isolates were associated with markedly increased proinflammatory cytokine production and neutrophil influx; however, depletion of neutrophils or genetic ablation of inflammasome components did not reverse disease. Virulent clinical isolates induced markedly increased type I and type III interferon (IFN) secretion in vitro and in vivo. STAT1/2-deficient mice lacking both type I and type III IFN signaling showed reduced disease severity and increased lung viral replication. Inhibition of type I IFN signaling using a blocking antibody or genetic ablation of the type I IFN receptor reduced pathology with minimal effect on viral replication. Conversely, blockade of type III IFN signaling with a neutralizing antibody or genetic ablation of the IFN-lambda receptor had no effect on pathogenesis but restored viral replication. Collectively, these results demonstrate distinct roles for type I and type III IFN in HMPV pathogenesis and immunity.


Subject(s)
Metapneumovirus , Paramyxoviridae Infections , Respiratory Tract Infections , Child , Animals , Mice , Humans , Interferon Lambda , Lung , Respiratory Tract Infections/pathology , Interferons
3.
Am J Pathol ; 194(3): 384-401, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38159723

ABSTRACT

Respiratory tract virus infections cause millions of hospitalizations worldwide each year. Severe infections lead to lung damage that coincides with persistent inflammation and a lengthy repair period. Vaccination and antiviral therapy help to mitigate severe infections before or during the acute stage of disease, but there are currently limited specific treatment options available to individuals experiencing the long-term sequelae of respiratory viral infection. Herein, C57BL/6 mice were infected with influenza A/PR/8/34 as a model for severe viral lung infection and allowed to recover for 21 days. Mice were treated with rapamycin, a well-characterized mammalian target of rapamycin complex 1 (mTORC1) inhibitor, on days 12 to 20 after infection, a time period after viral clearance. Persistent inflammation following severe influenza infection in mice was primarily driven by macrophages and T cells. Uniform manifold approximation and projection analysis of flow cytometry data revealed that lung macrophages had high activation of mTORC1, an energy-sensing kinase involved in inflammatory immune cell effector functions. Rapamycin treatment reduced lung inflammation and the frequency of exudate macrophages, T cells, and B cells in the lung, while not impacting epithelial progenitor cells or adaptive immune memory. These data highlight mTORC1's role in sustaining persistent inflammation following clearance of a viral respiratory pathogen and suggest a possible intervention for post-viral chronic lung inflammation.


Subject(s)
Influenza, Human , Orthomyxoviridae Infections , Pneumonia , Mice , Animals , Humans , Orthomyxoviridae Infections/complications , Mice, Inbred C57BL , Lung , Macrophages , Inflammation/complications , Sirolimus/pharmacology , Mechanistic Target of Rapamycin Complex 1 , TOR Serine-Threonine Kinases , Mammals
4.
Article in English | MEDLINE | ID: mdl-38810239

ABSTRACT

Children are susceptible to influenza infections and can experience severe disease presentation due to a lack of or limited pre-existing immunity. Despite the disproportionate impact influenza has on this population, there is a lack of focus on pediatric influenza research, particularly when it comes to identifying the pathogenesis of long-term outcomes that persist beyond the point of viral clearance. In this study, juvenile outbred male and female mice were infected with influenza and analyzed following viral clearance to determine how sex impacts the persistent inflammatory responses to influenza. It was found that females maintained a broader cytokine response in the lung following clearance of influenza, with innate, type I and type II cytokine signatures in almost all mice. Males, on the other hand, had higher levels of IL-6 and other macrophage-related cytokines, but no evidence of a type I or type II response. The immune landscape was similar in the lungs between males and females post-infection, but males had a higher regulatory T cell to TH1 ratio compared to female mice. Cytokine production positively correlated with the frequency of TH1 cells and exudate macrophages, as well as the number of cells in the bronchoalveolar lavage fluid. Furthermore, female lungs were enriched for metabolites involved in the glycolytic pathway, suggesting glycolysis is higher in female lungs compared to males after viral clearance. These data suggest juvenile female mice have persistent and excessive lung inflammation beyond the point of viral clearance, while juvenile males had a more immunosuppressive phenotype.

5.
J Immunol ; 209(4): 760-771, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35914833

ABSTRACT

Influenza-associated bacterial superinfections have devastating impacts on the lung and can result in increased risk of mortality. New strains of influenza circulate throughout the population yearly, promoting the establishment of immune memory. Nearly all individuals have some degree of influenza memory before adulthood. Due to this, we sought to understand the role of immune memory during bacterial superinfections. An influenza heterotypic immunity model was established using influenza A/Puerto Rico/8/34 and influenza A/X31. We report in this article that influenza-experienced mice are more resistant to secondary bacterial infection with methicillin-resistant Staphylococcus aureus as determined by wasting, bacterial burden, pulmonary inflammation, and lung leak, despite significant ongoing lung remodeling. Multidimensional flow cytometry and lung transcriptomics revealed significant alterations in the lung environment in influenza-experienced mice compared with naive animals. These include changes in the lung monocyte and T cell compartments, characterized by increased expansion of influenza tetramer-specific CD8+ T cells. The protection that was seen in the memory-experienced mouse model is associated with the reduction in inflammatory mechanisms, making the lung less susceptible to damage and subsequent bacterial colonization. These findings provide insight into how influenza heterotypic immunity reshapes the lung environment and the immune response to a rechallenge event, which is highly relevant to the context of human infection.


Subject(s)
Bacterial Infections , Coinfection , Influenza, Human , Methicillin-Resistant Staphylococcus aureus , Orthomyxoviridae Infections , Superinfection , Adult , Animals , CD8-Positive T-Lymphocytes , Humans , Lung , Mice , Mice, Inbred C57BL , Superinfection/microbiology
6.
J Immunol ; 208(7): 1711-1718, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35321882

ABSTRACT

COVID-19 has had an unprecedented global impact on human health. Understanding the Ab memory responses to infection is one tool needed to effectively control the pandemic. Among 173 outpatients who had virologically confirmed SARS-CoV-2 infection, we evaluated serum Ab concentrations, microneutralization activity, and enumerated SARS-CoV-2-specific B cells in convalescent human blood specimens. Serum Ab concentrations were variable, allowing for stratification of the cohort into high and low responders. Neither participant sex, the timing of blood sampling following the onset of illness, nor the number of SARS-CoV-2 spike protein-specific B cells correlated with serum Ab concentration. Serum Ab concentration was positively associated with microneutralization activity and participant age, with participants under the age of 30 showing the lowest Ab level. These data suggest that young adult outpatients did not generate as robust Ab memory, compared with older adults. Body mass index was also positively correlated with serum Ab levels. Multivariate analyses showed that participant age and body mass index were independently associated with Ab levels. These findings have direct implications for public health policy and current vaccine efforts. Knowledge gained regarding Ab memory following infection will inform the need for vaccination in those previously infected and allow for a better approximation of population-wide protective immunity.


Subject(s)
Age Factors , Antibody Formation , Body Mass Index , COVID-19 , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , COVID-19/immunology , Humans , Outpatients , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/immunology
7.
J Infect Dis ; 2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37665976

ABSTRACT

Cell-based quadrivalent inactivated influenza vaccine has been shown to have higher vaccine effectiveness than traditional egg-based quadrivalent inactivated influenza vaccine. This is observed despite similar levels of serum hemagglutinin antibodies induced by each vaccine. Here, we examine peripheral immune activation following egg-based or cell-based influenza vaccination in a clinical trial in children. Peripheral blood mononuclear cells were isolated and RNA sequenced from 81 study participants (41 Fluzone, egg-based and 40 Flucelvax, cell based) pre- and 7 days post- vaccination. Seroconversion was assessed by hemagglutinin inhibition assay. Differential gene expression was determined and pathway analysis was conducted. Cell-based influenza vaccine induced greater interferon stimulated and innate immune gene activation compared with egg-based influenza vaccine. Participants who seroconverted had increased interferon signaling activation versus those who did not seroconvert. These data suggest that cell-based influenza vaccine stimulates immune activation differently from egg-based influenza vaccine, shedding light on reported differences in vaccine effectiveness.

8.
J Immunol ; 205(2): 480-488, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32522833

ABSTRACT

Aspergillus fumigatus is an environmental fungus that can cause invasive pulmonary aspergillosis when spores are inhaled into the respiratory tract and invade airway or lung tissue. Influenza is a common respiratory virus that can cause severe respiratory disease, and postinfluenza invasive pulmonary aspergillosis, which is becoming a well-recognized clinical problem, typically occurs in critically ill patients. Mice challenged with influenza A PR/8/34 H1N1 and subsequently challenged with A. fumigatus had increased fungal burden, viral burden, inflammation, and mortality compared with single infected mice. Neutrophil recruitment in the lung of superinfected mice was decreased; however, mice were not neutropenic, and there was no difference in absolute blood neutrophils between groups. Additionally, CXCL1 and CXCL2 were decreased in lungs of superinfected mice compared with controls. IFN levels were increased in mice that received influenza, and deletion of STAT1 resulted in decreased fungal burden, increased airway and lung neutrophils, and increased CXCL1 compared with wild-type mice, whereas deletion of STAT2 did not change fungal burden or airway neutrophilia compared with wild-type mice. These data demonstrate a mechanism by which influenza A-induced STAT1 signaling inhibits neutrophil recruitment and increases susceptibility to postinfluenza invasive pulmonary aspergillosis.


Subject(s)
Aspergillus fumigatus/physiology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Invasive Pulmonary Aspergillosis/immunology , Lung/immunology , Neutrophils/immunology , Orthomyxoviridae Infections/immunology , Animals , Chemokine CXCL1/metabolism , Colony Count, Microbial , Disease Progression , Humans , Immune Evasion , Influenza, Human/complications , Invasive Pulmonary Aspergillosis/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Orthomyxoviridae Infections/complications , STAT1 Transcription Factor/metabolism , Signal Transduction
9.
Int J Mol Sci ; 23(3)2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35162999

ABSTRACT

Influenza (IAV) neuraminidase (NA) is a glycoprotein required for the viral exit from the cell. NA requires disulfide bonds for proper function. We have recently demonstrated that protein disulfide isomerase (PDI)A3 is required for oxidative folding of IAV hemagglutinin (HA), and viral propagation. However, it not known whether PDIs are required for NA maturation or if these interactions represent a putative target for the treatment of influenza infection. We sought to determine whether PDIA3 is required for disulfide bonds of NA, its activity, and propagation of the virus. Requirement of disulfides for NA oligomerization and activity were determined using biotin switch and redox assays in WT and PDIA3-/- in A549 cells. A PDI specific inhibitor (LOC14) was utilized to determine the requirement of PDIs in NA activity, IAV burden, and inflammatory response in A549 and primary mouse tracheal epithelial cells. Mice were treated with the inhibitor LOC14 and subsequently examined for IAV burden, NA activity, cytokine, and immune response. IAV-NA interacts with PDIA3 and this interaction is required for NA activity. PDIA3 ablation or inhibition decreased NA activity, viral burden, and inflammatory response in lung epithelial cells. LOC14 treatment significantly attenuated the influenza-induced inflammatory response in mice including the overall viral burden. These results provide evidence for PDIA3 inhibition suppressing NA activity, potentially providing a novel platform for host-targeted antiviral therapies.


Subject(s)
Enzyme Inhibitors/administration & dosage , Influenza A Virus, H1N1 Subtype/enzymology , Neuraminidase/metabolism , Orthomyxoviridae Infections/drug therapy , Protein Disulfide-Isomerases/metabolism , Viral Proteins/metabolism , A549 Cells , Animals , Cells, Cultured , Disease Models, Animal , Dogs , Enzyme Inhibitors/pharmacology , Female , Humans , Madin Darby Canine Kidney Cells , Mice , Neuraminidase/chemistry , Orthomyxoviridae Infections/metabolism , Primary Cell Culture , Protein Folding , Trachea/cytology , Trachea/drug effects , Trachea/metabolism , Trachea/virology , Viral Proteins/chemistry
10.
J Immunol ; 203(11): 3000-3012, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31645418

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is a potent negative regulator capable of restraining overactivation of the renin-angiotensin system, which contributes to exuberant inflammation after bacterial infection. However, the mechanism through which ACE2 modulates this inflammatory response is not well understood. Accumulating evidence indicates that infectious insults perturb ACE2 activity, allowing for uncontrolled inflammation. In the current study, we demonstrate that pulmonary ACE2 levels are dynamically varied during bacterial lung infection, and the fluctuation is critical in determining the severity of bacterial pneumonia. Specifically, we found that a pre-existing and persistent deficiency of active ACE2 led to excessive neutrophil accumulation in mouse lungs subjected to bacterial infection, resulting in a hyperinflammatory response and lung damage. In contrast, pre-existing and persistent increased ACE2 activity reduces neutrophil infiltration and compromises host defense, leading to overwhelming bacterial infection. Further, we found that the interruption of pulmonary ACE2 restitution in the model of bacterial lung infection delays the recovery process from neutrophilic lung inflammation. We observed the beneficial effects of recombinant ACE2 when administered to bacterially infected mouse lungs following an initial inflammatory response. In seeking to elucidate the mechanisms involved, we discovered that ACE2 inhibits neutrophil infiltration and lung inflammation by limiting IL-17 signaling by reducing the activity of the STAT3 pathway. The results suggest that the alteration of active ACE2 is not only a consequence of bacterial lung infection but also a critical component of host defense through modulation of the innate immune response to bacterial lung infection by regulating neutrophil influx.


Subject(s)
Inflammation/immunology , Neutrophils/immunology , Peptidyl-Dipeptidase A/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Angiotensin-Converting Enzyme 2 , Animals , Disease Models, Animal , Female , Imidazoles/administration & dosage , Imidazoles/pharmacology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Inflammation/drug therapy , Inflammation/pathology , Leucine/administration & dosage , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbial Sensitivity Tests , Neutrophils/drug effects , Neutrophils/pathology , Peptidyl-Dipeptidase A/deficiency , Peptidyl-Dipeptidase A/genetics , Pseudomonas Infections/drug therapy , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/drug effects
11.
Am J Respir Crit Care Med ; 201(8): 934-945, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31834999

ABSTRACT

Rationale: The role of FSTL-1 (follistatin-like 1) in lung homeostasis is unknown.Objectives: We aimed to define the impact of FSTL-1 attenuation on lung structure and function and to identify FSTL-1-regulated transcriptional pathways in the lung. Further, we aimed to analyze the association of FSTL-1 SNPs with lung disease.Methods: FSTL-1 hypomorphic (FSTL-1 Hypo) mice underwent lung morphometry, pulmonary function testing, and micro-computed tomography. Fstl1 expression was determined in wild-type lung cell populations from three independent research groups. RNA sequencing of wild-type and FSTL-1 Hypo mice identified FSTL-1-regulated gene expression, followed by validation and mechanistic in vitro examination. FSTL1 SNP analysis was performed in the COPDGene (Genetic Epidemiology of Chronic Obstructive Pulmonary Disease) cohort.Measurements and Main Results: FSTL-1 Hypo mice developed spontaneous emphysema, independent of smoke exposure. Fstl1 is highly expressed in the lung by mesenchymal and endothelial cells but not immune cells. RNA sequencing of whole lung identified 33 FSTL-1-regulated genes, including Nr4a1, an orphan nuclear hormone receptor that negatively regulates NF-κB (nuclear factor-κB) signaling. In vitro, recombinant FSTL-1 treatment of macrophages attenuated NF-κB p65 phosphorylation in an Nr4a1-dependent manner. Within the COPDGene cohort, several SNPs in the FSTL1 region corresponded to chronic obstructive pulmonary disease and lung function.Conclusions: This work identifies a novel role for FSTL-1 protecting against emphysema development independent of smoke exposure. This FSTL-1-deficient emphysema implicates regulation of immune tolerance in lung macrophages through Nr4a1. Further study of the mechanisms involving FSTL-1 in lung homeostasis, immune regulation, and NF-κB signaling may provide additional insight into the pathophysiology of emphysema and inflammatory lung diseases.


Subject(s)
Follistatin-Related Proteins/genetics , Lung/diagnostic imaging , Pulmonary Emphysema/genetics , Smoke/adverse effects , Animals , Endothelial Cells/metabolism , Follistatin-Related Proteins/pharmacology , Gene Expression Regulation , Gene Knockdown Techniques , Humans , In Vitro Techniques , Lung/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mutation , Nuclear Receptor Subfamily 4, Group A, Member 1/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phosphorylation/drug effects , Polymorphism, Single Nucleotide , Positron Emission Tomography Computed Tomography , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Emphysema/diagnostic imaging , Pulmonary Emphysema/metabolism , Single Photon Emission Computed Tomography Computed Tomography , Nicotiana , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism , X-Ray Microtomography
12.
Clin Microbiol Rev ; 32(3)2019 06 19.
Article in English | MEDLINE | ID: mdl-31142498

ABSTRACT

Community-acquired pneumonia (CAP) is a leading cause of morbidity and mortality worldwide. Despite broad literature including basic and translational scientific studies, many gaps in our understanding of host-pathogen interactions remain. In this review, pathogen virulence factors that drive lung infection and injury are discussed in relation to their associated host immune pathways. CAP epidemiology is considered, with a focus on Staphylococcus aureus and Streptococcus pneumoniae as primary pathogens. Bacterial factors involved in nasal colonization and subsequent virulence are illuminated. A particular emphasis is placed on bacterial pore-forming toxins, host cell death, and inflammasome activation. Identified host-pathogen interactions are then examined by linking pathogen factors to aberrant host response pathways in the context of acute lung injury in both primary and secondary infection. While much is known regarding bacterial virulence and host immune responses, CAP management is still limited to mostly supportive care. It is likely that improvements in therapy will be derived from combinatorial targeting of both pathogen virulence factors and host immunomodulation.


Subject(s)
Community-Acquired Infections/immunology , Community-Acquired Infections/microbiology , Host-Pathogen Interactions/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Humans , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/physiology , Streptococcus pneumoniae/pathogenicity , Streptococcus pneumoniae/physiology
13.
Clin Sci (Lond) ; 134(13): 1697-1714, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32648583

ABSTRACT

Acute influenza virus infections are a global public health concern accounting for millions of illnesses worldwide ranging from mild to severe with, at time, severe complications. Once an individual is infected, the immune system is triggered in response to the pathogen. This immune response can be beneficial ultimately leading to the clearance of the viral infection and establishment of immune memory mechanisms. However, it can be detrimental by increasing susceptibility to secondary bacterial infections and resulting in permanent changes to the lung architecture, in the form of fibrotic sequelae. Here, we review influenza associated bacterial super-infection, the formation of T-cell memory, and persistent lung injury resulting from influenza infection.


Subject(s)
Coinfection/immunology , Influenza, Human/complications , Lung Diseases/etiology , Pulmonary Alveoli/immunology , Animals , Coinfection/etiology , Humans , Influenza, Human/immunology , Lung Diseases/immunology , T-Lymphocytes/immunology
14.
Immunity ; 35(6): 997-1009, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22195749

ABSTRACT

The interleukin-17 (IL-17) family of cytokines phylogenetically predates the evolution of T cells in jawed vertebrates, suggesting that the ontogeny of the Th17 cell lineage must have arisen to confer an evolutionary advantage to the host over innate sources of IL-17. Utilizing a model of mucosal immunization with the encapsulated bacteria Klebsiella pneumoniae, we found that B cells, which largely recognized polysaccharide capsular antigens, afforded protection to only the vaccine strain. In contrast, memory Th17 cells proliferated in response to conserved outer membrane proteins and conferred protection against several serotypes of K. pneumoniae, including the recently described multidrug resistant New Dehli metallolactamase strain. Notably, this heterologous, clade-specific protection was antibody independent, demonstrating the Th17 cell lineage confers a host advantage by providing heterologous mucosal immunity independent of serotype-specific antibody.


Subject(s)
Immunity, Mucosal/immunology , Klebsiella pneumoniae/immunology , Th17 Cells/immunology , Animals , Antibodies, Bacterial/immunology , Antibodies, Bacterial/metabolism , Bacterial Outer Membrane Proteins/immunology , Cross Protection/immunology , Cytokines/immunology , Cytokines/metabolism , Klebsiella Infections/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/immunology , Mucous Membrane/microbiology , Nasal Mucosa/immunology , Th17 Cells/metabolism
15.
BMC Bioinformatics ; 20(1): 297, 2019 Jun 03.
Article in English | MEDLINE | ID: mdl-31159726

ABSTRACT

BACKGROUND: Host factors of influenza virus replication are often found in key topological positions within protein-protein interaction networks. This work explores how protein states can be manipulated through controllability analysis: the determination of the minimum manipulation needed to drive the cell system to any desired state. Here, we complete a two-part controllability analysis of two protein networks: a host network representing the healthy cell state and an influenza A virus-host network representing the infected cell state. In this context, controllability analyses aim to identify key regulating host factors of the infected cell's progression. This knowledge can be utilized in further biological analysis to understand disease dynamics and isolate proteins for study as drug target candidates. RESULTS: Both topological and controllability analyses provide evidence of wide-reaching network effects stemming from the addition of viral-host protein interactions. Virus interacting and driver host proteins are significant both topologically and in controllability, therefore playing important roles in cell behavior during infection. Functional analysis finds overlap of results with previous siRNA studies of host factors involved in influenza replication, NF-kB pathway and infection relevance, and roles as interferon regulating genes. 24 proteins are identified as holding regulatory roles specific to the infected cell by measures of topology, controllability, and functional role. These proteins are recommended for further study as potential antiviral drug targets. CONCLUSIONS: Seasonal outbreaks of influenza A virus are a major cause of illness and death around the world each year with a constant threat of pandemic infection. This research aims to increase the efficiency of antiviral drug target discovery using existing protein-protein interaction data and network analysis methods. These results are beneficial to future studies of influenza virus, both experimental and computational, and provide evidence that the combination of topology and controllability analyses may be valuable for future efforts in drug target discovery.


Subject(s)
Antiviral Agents/pharmacology , Drug Delivery Systems , Drug Discovery , Host-Pathogen Interactions , Protein Interaction Maps , Humans , Influenza A virus/drug effects , Influenza A virus/metabolism , RNA, Small Interfering/metabolism , Reproducibility of Results , Virus Replication/drug effects
16.
Infect Immun ; 87(5)2019 03.
Article in English | MEDLINE | ID: mdl-30804099

ABSTRACT

Influenza kills 30,000 to 40,000 people each year in the United States and causes 10 times as many hospitalizations. A common complication of influenza is bacterial superinfection, which exacerbates morbidity and mortality from the viral illness. Recently, methicillin-resistant Staphylococcus aureus (MRSA) has emerged as the dominant pathogen found in bacterial superinfection, with Streptococcus pneumoniae a close second. However, clinicians have few tools to treat bacterial superinfection. Current therapy for influenza/bacterial superinfection consists of treating the underlying influenza infection and adding various antibiotics, which are increasingly rendered ineffective by rising bacterial multidrug resistance. Several groups have recently proposed the use of the antiviral cytokine interferon lambda (IFN-λ) as a therapeutic for influenza, as administration of pegylated IFN-λ improves lung function and survival during influenza by reducing the overabundance of neutrophils in the lung. However, our data suggest that therapeutic IFN-λ impairs bacterial clearance during influenza superinfection. Specifically, mice treated with an adenoviral vector to overexpress IFN-λ during influenza infection exhibited increased bacterial burdens upon superinfection with either MRSA or S. pneumoniae Surprisingly, adhesion molecule expression, antimicrobial peptide production, and reactive oxygen species activity were not altered by IFN-λ treatment. However, neutrophil uptake of MRSA and S. pneumoniae was significantly reduced upon IFN-λ treatment during influenza superinfection in vivo Together, these data support the theory that IFN-λ decreases neutrophil motility and function in the influenza-infected lung, which increases the bacterial burden during superinfection. Thus, we believe that caution should be exercised in the possible future use of IFN-λ as therapy for influenza.


Subject(s)
Antiviral Agents/therapeutic use , Influenza, Human/complications , Influenza, Human/drug therapy , Interferons/therapeutic use , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Superinfection/drug therapy , Animals , Disease Models, Animal , Disease Susceptibility , Humans , Male , Mice , Mice, Inbred C57BL , Staphylococcal Infections/pathology , Superinfection/etiology , United States
17.
Infect Immun ; 87(11)2019 11.
Article in English | MEDLINE | ID: mdl-31451621

ABSTRACT

Streptococcus pneumoniae is the most common cause of community-acquired pneumonia worldwide, and interleukin-22 (IL-22) helps contain pneumococcal burden in lungs and extrapulmonary tissues. Administration of IL-22 increases hepatic complement 3 and complement deposition on bacteria and improves phagocytosis by neutrophils. The effects of IL-22 can be tempered by a secreted natural antagonist, known as IL-22 binding protein (IL-22BP), encoded by Il22ra2 To date, the degree to which IL-22BP controls IL-22 in pulmonary infection is not well defined. Here, we show that Il22ra2 inhibits IL-22 during S. pneumoniae lung infection and that Il22ra2 deficiency favors downregulation of oxidative phosphorylation (OXPHOS) genes in an IL-22-dependent manner. Il22ra2-/- mice are more resistant to S. pneumoniae infection, have increased IL-22 in lung tissues, and sustain longer survival upon infection than control mice. Transcriptome sequencing (RNA-seq) analysis of infected Il22ra2-/- mouse lungs revealed downregulation of genes involved in OXPHOS. Downregulation of this metabolic process is necessary for increased glycolysis, a crucial step for transitioning to a proinflammatory phenotype, in particular macrophages and dendritic cells (DCs). Accordingly, we saw that macrophages from Il22ra2-/- mice displayed reduced OXPHOS gene expression upon infection with S. pneumoniae, changes that were IL-22 dependent. Furthermore, we showed that macrophages express IL-22 receptor subunit alpha-1 (IL-22Ra1) during pneumococcal infection and that Il22ra2-/- macrophages rely more on the glycolytic pathway than wild-type (WT) controls. Together, these data indicate that IL-22BP deficiency enhances IL-22 signaling in the lung, thus contributing to resistance to pneumococcal pneumonia by downregulating OXPHOS genes and increasing glycolysis in macrophages.


Subject(s)
Interleukins/metabolism , Pneumonia, Pneumococcal/metabolism , Receptors, Interleukin/metabolism , Animals , Cell Line , Disease Susceptibility , Epithelial Cells/physiology , Gene Expression Regulation , Interleukins/genetics , Leukocyte Common Antigens , Lung/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Oxidation-Reduction , Phosphorylation , Pneumonia, Pneumococcal/immunology , Receptors, Interleukin/genetics , Streptococcus pneumoniae , Interleukin-22
18.
J Pediatr ; 209: 146-153.e1, 2019 06.
Article in English | MEDLINE | ID: mdl-30905425

ABSTRACT

OBJECTIVE: To determine whether treatment for urinary tract infections in children could be individualized using biomarkers for acute pyelonephritis. STUDY DESIGN: We enrolled 61 children with febrile urinary tract infections, collected blood and urine samples, and performed a renal scan within 2 weeks of diagnosis to identify those with pyelonephritis. Renal scans were interpreted centrally by 2 experts. We measured inflammatory proteins in blood and urine using LUMINEX or an enzyme-linked immunosorbent assay. We evaluated serum RNA expression using RNA sequencing in a subset of children. Finally, for children with Escherichia coli isolated from urine cultures, we performed a polymerase chain reaction for 4 previously identified virulence genes. RESULTS: Urinary markers that best differentiated pyelonephritis from cystitis included chemokine (C-X-C motif) ligand (CXCL)1, CXCL9, CXCL12, C-C motif chemokine ligand 2, INF γ, and IL-15. Serum procalcitonin was the best serum marker for pyelonephritis. Genes in the interferon-γ pathway were upregulated in serum of children with pyelonephritis. The presence of E coli virulence genes did not correlate with pyelonephritis. CONCLUSIONS: Immune response to pyelonephritis and cystitis differs quantitatively and qualitatively; this may be useful in differentiating these 2 conditions.


Subject(s)
Bacterial Infections , Cystitis/microbiology , Pyelonephritis/microbiology , Urinary Tract Infections , Acute Disease , Bacterial Infections/blood , Bacterial Infections/urine , Biomarkers/analysis , Child, Preschool , Cystitis/blood , Cystitis/diagnosis , Cystitis/urine , Diagnosis, Differential , Female , Humans , Infant , Male , Pilot Projects , Prospective Studies , Pyelonephritis/blood , Pyelonephritis/chemically induced , Pyelonephritis/urine , Urinary Tract Infections/blood , Urinary Tract Infections/urine
19.
J Immunol ; 198(11): 4383-4393, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28424239

ABSTRACT

The absent in melanoma 2 (AIM2) inflammasome plays an important role in many viral and bacterial infections, but very little is known about its role in RNA virus infection, including influenza A virus (IAV). In this study, we have designed in vivo and in vitro studies to determine the role of AIM2 in infections with lethal doses of IAVs A/PR8/34 and A/California/07/09. In wild-type mice, IAV infection enhanced AIM2 expression, induced dsDNA release, and stimulated caspase-1 activation and release of cleaved IL-1ß in the lung, which was significantly reduced in AIM2-deficient mice. Interestingly, AIM2 deficiency did not affect the transcription of caspase-1 and IL-1ß. In addition, AIM2-deficient mice exhibited attenuated lung injury and significantly improved survival against IAV challenges, but did not alter viral burden in the lung. However, AIM2 deficiency did not seem to affect adaptive immune response against IAV infections. Furthermore, experiments with AIM2-specific small interfering RNA-treated and AIM2-deficient human and mouse lung alveolar macrophages and type II cells indicated a macrophage-specific function of AIM2 in regulation of IAV-stimulated proinflammatory response. Collectively, our results demonstrate that influenza infection activates the AIM2 inflammasome, which plays a critical role in IAV-induced lung injury and mortality. AIM2 might serve as a therapeutic target for combating influenza-associated morbidity and mortality without compromising the host antiviral responses.


Subject(s)
DNA-Binding Proteins/physiology , Inflammasomes/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/mortality , Lung Injury/immunology , Adaptive Immunity , Animals , Caspase 1/genetics , Caspase 1/immunology , Caspase 1/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Humans , Influenza, Human/physiopathology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lung Injury/physiopathology , Lung Injury/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Mice , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , RNA, Small Interfering/genetics , Viral Load/immunology
20.
Respirology ; 24(7): 646-651, 2019 07.
Article in English | MEDLINE | ID: mdl-30761687

ABSTRACT

BACKGROUND AND OBJECTIVE: Chronic lower respiratory diseases (CLRD) increase the risk of type 2 diabetes, which in turn may worsen lung function. Metformin, a common antidiabetic with anti-inflammatory and antioxidant properties, may improve respiratory outcomes. Therefore, we examined the association of metformin use with the risk of mortality from CLRD. METHODS: We analysed data from the National Health and Nutrition Examination Survey during 1988-1994 and 1999-2010 for participants aged 40 years or older who had diabetes and were followed up for mortality through 2011. Information on prescription medicine was collected at baseline and CLRD-related mortality during follow-up was defined using the 10th Revision of the International Classification of Diseases (ICD-10). Cox proportional hazards modelling was used to determine the mortality hazard ratio (HR) associated with metformin use, adjusting for relevant covariates. RESULTS: A total of 5266 participants with a median follow-up of 6.1 years were included. The prevalence of metformin use was 31.9% and 1869 participants died during follow-up, including 72 of CLRD. In the adjusted Cox proportional regression analysis, metformin was associated with a decreased risk of CLRD mortality in the overall population (HR: 0.39, 95% CI: 0.15-0.99) and among participants with baseline CLRD (HR: 0.30, 95% CI: 0.10-0.93), after adjusting for age, gender, race/ethnicity, cigarette smoking, body mass index, current asthma and chronic obstructive pulmonary disease (COPD), insulin and other diabetic medications, and glycohaemoglobin level. We found no association between other antidiabetic medications and CLRD mortality. CONCLUSION: In this sample representative of the U.S. population, metformin was associated with lower CLRD mortality in adults with diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Metformin/therapeutic use , Nutrition Surveys , Respiratory Tract Diseases/mortality , Adult , Aged , Cross-Sectional Studies , Diabetes Mellitus, Type 2/etiology , Female , Follow-Up Studies , Humans , Hypoglycemic Agents/therapeutic use , Male , Middle Aged , Respiratory Tract Diseases/complications , Survival Rate/trends , Time Factors , United States/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL