Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
2.
Mucosal Immunol ; 15(6): 1309-1320, 2022 06.
Article in English | MEDLINE | ID: mdl-36352099

ABSTRACT

Neonates exhibit increased susceptibility to respiratory viral infections, attributed to inflammation at the developing pulmonary air-blood interface. IFN I are antiviral cytokines critical to control viral replication, but also promote inflammation. Previously, we established a neonatal murine influenza virus (IV) model, which demonstrates increased mortality. Here, we sought to determine the role of IFN I in this increased mortality. We found that three-day-old IFNAR-deficient mice are highly protected from IV-induced mortality. In addition, exposure to IFNß 24 h post IV infection accelerated death in WT neonatal animals but did not impact adult mortality. In contrast, IFN IIIs are protective to neonatal mice. IFNß induced an oxidative stress imbalance specifically in primary neonatal IV-infected pulmonary type II epithelial cells (TIIEC), not in adult TIIECs. Moreover, neonates did not have an infection-induced increase in antioxidants, including a key antioxidant, superoxide dismutase 3, as compared to adults. Importantly, antioxidant treatment rescued IV-infected neonatal mice, but had no impact on adult morbidity. We propose that IFN I exacerbate an oxidative stress imbalance in the neonate because of IFN I-induced pulmonary TIIEC ROS production coupled with developmentally regulated, defective antioxidant production in response to IV infection. This age-specific imbalance contributes to mortality after respiratory infections in this vulnerable population.


Subject(s)
Interferon Type I , Orthomyxoviridae Infections , Oxidative Stress , Animals , Mice , Antioxidants/metabolism , Inflammation , Interferon Type I/metabolism , Interferon-beta , Mice, Inbred C57BL , Orthomyxoviridae Infections/physiopathology , Animals, Newborn
3.
J Virol ; 96(9): e0035222, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35446142

ABSTRACT

Influenza A viruses (IAV) can cause severe disease and death in humans. IAV infection and the accompanying immune response can result in systemic inflammation, leading to intestinal damage and disruption of the intestinal microbiome. Here, we demonstrate that a specific subset of epithelial cells, tuft cells, increase across the small intestine during active respiratory IAV infection. Upon viral clearance, tuft cell numbers return to baseline levels. Intestinal tuft cell increases were not protective against disease, as animals with either increased tuft cells or a lack of tuft cells did not have any change in disease morbidity after infection. Respiratory IAV infection also caused transient increases in type 1 and 2 innate lymphoid cells (ILC1 and ILC2, respectively) in the small intestine. ILC2 increases were significantly blunted in the absence of tuft cells, whereas ILC1s were unaffected. Unlike the intestines, ILCs in the lungs were not altered in the absence of tuft cells. This work establishes that respiratory IAV infection causes dynamic changes to tuft cells and ILCs in the small intestines and that tuft cells are necessary for the infection-induced increase in small intestine ILC2s. These intestinal changes in tuft cell and ILC populations may represent unexplored mechanisms preventing systemic infection and/or contributing to severe disease in humans with preexisting conditions. IMPORTANCE Influenza A virus (IAV) is a respiratory infection in humans that can lead to a wide range of symptoms and disease severity. Respiratory infection can cause systemic inflammation and damage in the intestines. Few studies have explored how inflammation alters the intestinal environment. We found that active infection caused an increase in the epithelial population called tuft cells as well as type 1 and 2 innate lymphoid cells (ILCs) in the small intestine. In the absence of tuft cells, this increase in type 2 ILCs was seriously blunted, whereas type 1 ILCs still increased. These findings indicate that tuft cells are necessary for infection-induced changes in small intestine type 2 ILCs and implicate tuft cells as regulators of the intestinal environment in response to systemic inflammation.


Subject(s)
Enteritis , Influenza A virus , Intestine, Small , Orthomyxoviridae Infections , Animals , Enteritis/immunology , Enteritis/physiopathology , Enteritis/virology , Humans , Immunity, Innate , Influenza A virus/immunology , Intestine, Small/cytology , Intestine, Small/virology , Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology
4.
J Biol Chem ; 297(1): 100885, 2021 07.
Article in English | MEDLINE | ID: mdl-34146545

ABSTRACT

PB1-F2 is a virulence factor of influenza A virus known to increase viral pathogenicity in mammalian hosts. PB1-F2 is an intrinsically disordered protein displaying a propensity to form amyloid-like fibers. However, the correlation between PB1-F2 structures and the resulting inflammatory response is unknown. Here, we used synchrotron-coupled Fourier transform-IR and deep UV microscopies to determine the presence of PB1-F2 fibers in influenza A virus-infected mice. In order to study the correlation between PB1-F2 structure and the inflammatory response, transgenic mice expressing luciferase under the control of an NF-κB promotor, allowing in vivo monitoring of inflammation, were intranasally instilled with monomeric, fibrillated, or truncated forms of recombinant PB1-F2. Our intravital NF-κB imaging, supported by cytokine quantification, clearly shows the proinflammatory effect of PB1-F2 fibers compared with N-terminal region of PB1-F2 unable to fibrillate. It is noteworthy that instillation of monomeric PB1-F2 of H5N1 virus induced a stronger inflammatory response when compared with prefibrillated PB1-F2 of H1N1 virus, suggesting mechanisms of virulence depending on PB1-F2 sequence. Finally, using whole-body plethysmography to measure volume changes in the lungs, we quantified the effects of the different forms of PB1-F2 on respiratory parameters. Thus, we conclude that PB1-F2-induced inflammation and respiratory distress are tightly correlated with sequence polymorphism and oligomerization status of the protein.


Subject(s)
Orthomyxoviridae Infections/metabolism , Protein Multimerization , Respiration , Signal Transduction , Viral Proteins/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Female , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Luciferases/genetics , Luciferases/metabolism , Lung/metabolism , Lung/physiopathology , Lung/virology , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , Polymorphism, Genetic , Promoter Regions, Genetic , Viral Proteins/genetics
5.
PLoS One ; 16(5): e0251473, 2021.
Article in English | MEDLINE | ID: mdl-33979408

ABSTRACT

During influenza A virus (IAV) entry, the hemagglutinin (HA) protein is triggered by endosomal low pH to undergo irreversible structural changes that mediate membrane fusion. HA proteins from different isolates vary in the pH at which they become activated in endosomes or become irreversible inactivated if exposed to extracellular acid. Little is known about extracellular pH in the upper respiratory tracts of mammals, how pH may shift during IAV infection, and its impact on replication of viruses that vary in HA activation pH. Here, we inoculated DBA/2J mice intranasally with A/TN/1-560/2009 (H1N1) (activation pH 5.5) or a mutant containing the destabilizing mutation HA1-Y17H (pH 6.0). We measured the kinetics of extracellular pH during infection using an optical pH-sensitive microsensor probe placed in the naris, nasal sinus, soft palate, and trachea. We also measured intracellular pH of single-cell suspensions of live, primary lung epithelial cells with various wavelength pH-sensitive dyes localized to cell membranes, cytosol, endosomes, secretory vesicles, microtubules, and lysosomes. Infection with either virus decreased extracellular pH and increased intracellular pH. Peak host immune responses were observed at 2 days post infection (DPI) and peak pH changes at 5 DPI. Extracellular and intracellular pH returned to baseline by 7 DPI in mice infected with HA1-Y17H and was restored later in wildtype-infected. Overall, IAV infection altered respiratory tract pH, which in turn modulated replication efficiency. This suggests a virus-host pH feedback loop that may select for IAV strains containing HA proteins of optimal pH stability, which may be approximately pH 5.5 in mice but may differ in other species.


Subject(s)
Immunity/physiology , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/physiopathology , Respiratory System/virology , Animals , Disease Models, Animal , Hydrogen-Ion Concentration , Mice , Respiratory System/physiopathology , Virus Internalization , Virus Replication
6.
Viruses ; 12(9)2020 08 27.
Article in English | MEDLINE | ID: mdl-32867061

ABSTRACT

Chronic inhalation of fungi and fungal components has been linked to the development of respiratory disorders, although their role with respect to the pathogenesis of acute respiratory virus infection remains unclear. Here, we evaluate inflammatory pathology induced by repetitive administration of a filtrate of the ubiquitous fungus, Alternaria alternata, and its impact on susceptibility to infection with influenza A. We showed previously that A. alternata at the nasal mucosae resulted in increased susceptibility to an otherwise sublethal inoculum of influenza A in wild-type mice. Here we demonstrate that A. alternata-induced potentiation of influenza A infection was not dependent on fungal serine protease or ribonuclease activity. Repetitive challenge with A. alternata prior to virus infection resulted proinflammatory cytokines, neutrophil recruitment, and loss of alveolar macrophages to a degree that substantially exceeded that observed in response to influenza A infection alone. Concomitant administration of immunomodulatory Lactobacillus plantarum, a strategy shown previously to limit virus-induced inflammation in the airways, blocked the exaggerated lethal response. These observations promote an improved understanding of severe influenza infection with potential clinical relevance for individuals subjected to continuous exposure to molds and fungi.


Subject(s)
Alternaria , Alternariosis/immunology , Influenza A virus , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/physiopathology , Alternaria/metabolism , Alternariosis/pathology , Alternariosis/physiopathology , Animals , Bacteria/growth & development , Bronchoalveolar Lavage Fluid/microbiology , Cytokines/metabolism , Disease Susceptibility , Female , Inflammation , Lactobacillus plantarum/physiology , Lung/immunology , Male , Mice, Inbred C57BL , Neutrophil Infiltration , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Ribonucleases/metabolism , Serine Proteases/metabolism , Severity of Illness Index
7.
Curr Opin Virol ; 44: 154-161, 2020 10.
Article in English | MEDLINE | ID: mdl-32932215

ABSTRACT

Influenza D is the only type of influenza virus that mainly affects cattle with frequent spillover to other species. Since the initial description of influenza D virus (IDV) in 2011, the virus has been found to circulate among cattle and swine populations worldwide. Research conducted during the past several years has led to an increased understanding of this novel influenza virus with bovines as a reservoir. In this review, we describe the current knowledge of epidemiology and host range of IDV followed by discussion of infection biology and animal model development for IDV. Finally, we review progress towards understanding of the pathogenesis and host response of IDV as well as developing preventive vaccines for IDV.


Subject(s)
Disease Reservoirs/veterinary , Host Specificity , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/prevention & control , Thogotovirus/immunology , Animals , Antibodies, Viral/immunology , Cattle , Disease Models, Animal , Disease Reservoirs/virology , Genome, Viral , Host-Pathogen Interactions/immunology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/physiopathology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Swine , Thogotovirus/genetics , Virus Replication
8.
Life Sci ; 258: 118214, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32768585

ABSTRACT

Large numbers of rodents are often used in the study of disease progression and in the evaluation of its potential treatments. To avoid subjective observation and to minimize home cage interference, we developed a computerized home cage monitoring system (HCMS100) based on a standard cage rack adapted with a single laser beam and a detector mounted on each cage, enabling to monitor mice movements based on laser beam interruptions. This retrofit system provided continuous and uninterrupted monitoring of spontaneous movement of a group of mice in a home cage. Validity was evaluated using disease state induced by LPS modelling bacterial infection and by influenza virus. RESULTS: Spontaneous activity of different number of mice (2-8) per cage showed the expected circadian rhythm with increased activity during the night, and its extent dependent on the number of mice in the cage. Females and males show similar circadian rhythm. Intranasal LPS administration and pulmonary infection with live influenza virus resulted in major reduction of mice activity along disease progression. Increase in activity over time was a good indicator of the recovery process from both LPS exposure and the flu infection. CONCLUSIONS: HCMS100 was shown to be a reliable, inexpensive, easy to use system that requires no changes in the common housing of various experimental animals (mice, hamsters, rats etc.). With minimal intervention, HCMS100 provides a continuous record of group activity with clear pattern of circadian rhythm, allowing long term recording of home cage activity even in restricted access environments.


Subject(s)
Disease Progression , Housing, Animal , Lipopolysaccharides/toxicity , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae , Recovery of Function/physiology , Animals , Circadian Rhythm/physiology , Female , Housing, Animal/trends , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Orthomyxoviridae Infections/psychology , Recovery of Function/drug effects
9.
Gac Med Mex ; 156(4): 273-278, 2020.
Article in English | MEDLINE | ID: mdl-32831336

ABSTRACT

BACKGROUND: Influenza virus infection is often complicated by a bacterial infection, with this coinfection causing severe pneumonia. If not timely treated, the disease can cause death. OBJECTIVE: To demonstrate, in animal models, that coinfection with influenza virus and bacteria that affect the respiratory tract causes multisystemic damage. METHOD: Six groups of mice were formed: a control group, one infected with the influenza virus, two infected with bacteria: Haemophilus influenzae and Streptococcus pneumoniae, respectively; and two co-infected with influenza virus and Haemophilus influenzae or Streptococcus pneumoniae, respectively. RESULTS: Of the six groups of mice, only the group co-infected with influenza virus and Streptococcus pneumoniae showed damage to thoracic and abdominal organs. A decrease in serum cytokine levels was found in all study groups, which was more pronounced in the co-infected mice. CONCLUSIONS: The groups of mice infected with Streptococcus pneumoniae or influenza virus alone showed no damage, which indicates that coexistence of these infections caused the damage in the group of co-infected mice.


ANTECEDENTES: La infección por el virus de la influenza con frecuencia se complica con una infección bacteriana, coinfección que provoca cuadros graves de neumonía, la cual puede ocasionar la muerte si no es tratada en forma oportuna. OBJETIVO: Demostrar en modelos animales que la coinfección por el virus de la influenza y bacterias que afectan el tracto respiratorio ocasiona daño multisistémico. MÉTODO: Se formaron seis grupos de ratones: un grupo control, uno infectado de virus de la influenza, dos infectados de bacterias: Haemophilus influenzae y Streptococcus pneumoniae, respectivamente; y dos coinfectados de virus de la influenza y Haemophilus influenzae y Streptococcus pneumoniae, respectivamente. RESULTADOS: De los seis grupos de ratones, solo en el grupo coinfectado de virus de la influenza y Streptococcus pneumoniae se observó daño en órganos torácicos y abdominales. En todos los grupos se encontró disminución de los niveles séricos de las citocinas, mayor en los ratones coinfectados. CONCLUSIONES: Los grupos de ratones infectados solo de Streptococcus pneumoniae o el virus de la influenza no presentaron daños, lo cual indica que la coexistencia de estas infecciones fue la que ocasionó el daño en el grupo de ratones coinfectados.


Subject(s)
Haemophilus Infections/physiopathology , Orthomyxoviridae Infections/physiopathology , Pneumococcal Infections/physiopathology , Animals , Coinfection/physiopathology , Cytokines/blood , Disease Models, Animal , Haemophilus Infections/microbiology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , Pneumococcal Infections/microbiology , Pneumonia/microbiology , Pneumonia/physiopathology , Pneumonia/virology , Streptococcus pneumoniae/isolation & purification
10.
Gac. méd. Méx ; 156(4): 270-275, Jul.-Aug. 2020. graf
Article in English | LILACS | ID: biblio-1249910

ABSTRACT

Abstract Background: Influenza virus infection is often complicated by a bacterial infection, with this coinfection causing severe pneumonia. If not timely treated, the disease can cause death. Objective: To demonstrate, in animal models, that coinfection with influenza virus and bacteria that affect the respiratory tract causes multisystemic damage. Method: Six groups of mice were formed: a control group, one infected with the influenza virus, two infected with bacteria: Haemophilus influenzae and Streptococcus pneumoniae, respectively; and two co-infected with influenza virus and Haemophilus influenzae or Streptococcus pneumoniae, respectively. Results: Of the six groups of mice, only the group co-infected with influenza virus and Streptococcus pneumoniae showed damage to thoracic and abdominal organs. A decrease in serum cytokine levels was found in all study groups, which was more pronounced in the co-infected mice. Conclusions: The groups of mice infected with Streptococcus pneumoniae or influenza virus alone showed no damage, which indicates that coexistence of these infections caused the damage in the group of co-infected mice.


Resumen Antecedentes: La infección por el virus de la influenza con frecuencia se complica con una infección bacteriana, coinfección que provoca cuadros graves de neumonía, la cual puede ocasionar la muerte si no es tratada en forma oportuna. Objetivo: Demostrar en modelos animales que la coinfección por el virus de la influenza y bacterias que afectan el tracto respiratorio ocasiona daño multisistémico. Método: Se formaron seis grupos de ratones: un grupo control, uno infectado de virus de la influenza, dos infectados de bacterias: Haemophilus influenzae y Streptococcus pneumoniae, respectivamente; y dos coinfectados de virus de la influenza y Haemophilus influenzae y Streptococcus pneumoniae, respectivamente. Resultados: De los seis grupos de ratones, solo en el grupo coinfectado de virus de la influenza y Streptococcus pneumoniae se observó daño en órganos torácicos y abdominales. En todos los grupos se encontró disminución de los niveles séricos de las citocinas, mayor en los ratones coinfectados. Conclusiones: Los grupos de ratones infectados solo de Streptococcus pneumoniae o el virus de la influenza no presentaron daños, lo cual indica que la coexistencia de estas infecciones fue la que ocasionó el daño en el grupo de ratones coinfectados.


Subject(s)
Animals , Male , Rats , Pneumococcal Infections/physiopathology , Orthomyxoviridae Infections/physiopathology , Haemophilus Infections/physiopathology , Pneumococcal Infections/microbiology , Pneumonia/physiopathology , Pneumonia/microbiology , Pneumonia/virology , Streptococcus pneumoniae/isolation & purification , Cytokines/blood , Orthomyxoviridae Infections/virology , Disease Models, Animal , Coinfection/physiopathology , Haemophilus Infections/microbiology , Mice, Inbred BALB C
12.
mBio ; 11(2)2020 03 24.
Article in English | MEDLINE | ID: mdl-32209691

ABSTRACT

People with diabetes are two times more likely to die from influenza than people with no underlying medical condition. The mechanisms underlying this susceptibility are poorly understood. In healthy individuals, small and short-lived postprandial peaks in blood glucose levels occur. In diabetes mellitus, these fluctuations become greater and more frequent. This glycemic variability is associated with oxidative stress and hyperinflammation. However, the contribution of glycemic variability to the pathogenesis of influenza A virus (IAV) has not been explored. Here, we used an in vitro model of the pulmonary epithelial-endothelial barrier and novel murine models to investigate the role of glycemic variability in influenza severity. In vitro, a history of glycemic variability significantly increased influenza-driven cell death and destruction of the epithelial-endothelial barrier. In vivo, influenza virus-infected mice with a history of glycemic variability lost significantly more body weight than mice with constant blood glucose levels. This increased disease severity was associated with markers of oxidative stress and hyperinflammation both in vitro and in vivo Together, these results provide the first indication that glycemic variability may help drive the increased risk of severe influenza in people with diabetes mellitus.IMPORTANCE Every winter, people with diabetes are at increased risk of severe influenza. At present, the mechanisms that cause this increased susceptibility are unclear. Here, we show that the fluctuations in blood glucose levels common in people with diabetes are associated with severe influenza. These data suggest that glycemic stability could become a greater clinical priority for patients with diabetes during outbreaks of influenza.


Subject(s)
Blood Glucose/analysis , Diabetes Mellitus, Type 2/complications , Influenza, Human/physiopathology , Severity of Illness Index , Animals , Biomarkers/blood , Cell Death , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Type 2/blood , Endothelial Cells/immunology , Endothelial Cells/virology , Glycemic Load , Humans , Inflammation , Influenza A virus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/physiopathology , Oxidative Stress
13.
Emerg Microbes Infect ; 9(1): 691-706, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32208814

ABSTRACT

Infection of influenza A virus (IAV) can trigger exaggerated pulmonary inflammation and induce acute lung injury (ALI). Limiting IAV replication and alleviation of pulmonary inflammation are two important therapeutic strategies for influenza virus infection. Recent studies have shown that hypoxia inducible factor-1α (HIF-1α) is an essential factor for the development and repair of ALI; however, the role and the underlying mechanisms of HIF-1α in IAV-induced ALI remain elusive. Here, we demonstrated that lung epithelial cell-specific Hif1α knockout mice infected with IAV developed more lung IAV replication and severe lung inflammation, which led to increased mortality compared to IAV-infected control mice. Moreover, knockdown of HIF1A in A549 cells (human alveolar type II epithelial cell line) promoted IAV replication in vitro. Mechanistically, knockdown of HIF1A reduced glycolysis by regulating transcription of glycolysis-related enzymes, which subsequently activated the AMPKα-ULK1 signalling pathway. Interestingly, AMPKα-ULK1 signalling promoted autophagy and augmented IAV replication. Taken together, deficiency of HIF-1α in lung epithelial cells reduces glycolysis and enhances AMPKα-ULK1-mediated autophagy, which finally facilitates IAV replication. These findings have deepened our understanding of the role of HIF-1α in regulating IAV replication and provided us novel therapeutic targets for combating influenza infection.


Subject(s)
Alveolar Epithelial Cells/physiology , Alveolar Epithelial Cells/virology , Autophagy , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Influenza A virus/physiology , Lung/virology , Orthomyxoviridae Infections/virology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy-Related Protein-1 Homolog/metabolism , Cell Line , Cytokines/biosynthesis , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Orthomyxoviridae Infections/physiopathology , Pneumonia, Viral/physiopathology , Pneumonia, Viral/virology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation , Virus Replication
14.
Nat Protoc ; 15(3): 1041-1065, 2020 03.
Article in English | MEDLINE | ID: mdl-31996843

ABSTRACT

In vivo two-photon imaging is a valuable technique for studies of viral pathogenesis and host responses to infection in vivo. In this protocol, we describe a methodology for analyzing influenza virus-infected lung in vivo by two-photon imaging microscopy. We describe the surgical procedure, how to stabilize the lung, and an approach to analyzing the data. Further, we provide a database of fluorescent dyes, antibodies, and reporter mouse lines that can be used in combination with a reporter influenza virus (Color-flu) for multicolor analysis. Setup of this model typically takes ~30 min and enables the observation of influenza virus-infected lungs for >4 h during the acute phase of the inflammation and at least 1 h in the lethal phase. This imaging system, which we termed two-photon IMPRESS (imaging pathophysiology research system), is broadly applicable to analyses of other respiratory pathogens and reveals disease progression at the cellular level in vivo.


Subject(s)
Influenza A virus/genetics , Luminescent Proteins/metabolism , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , Animals , Dogs , Gene Expression Regulation, Viral , Genes, Reporter , Luminescent Proteins/chemistry , Lung/physiopathology , Lung/virology , Madin Darby Canine Kidney Cells , Mice
15.
J Theor Biol ; 484: 110026, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31574283

ABSTRACT

We present results of a study of the early-time response of the innate immune system to influenza virus infection in an agent-based model (ABM) of epithelial cell layers. We find that the competition between the anti-viral immune response and viral antagonism can lead to viral titers non-monotonic in the initial infection fraction as found in experiments. Our model includes a coarse-grained version of intra-cellular processes and inter-cellular communication via cytokine and virion diffusion. We use ABM to follow the propagation of viral infection in the layer and the increase of the viral load as a function of time for different values of the multiplicity of infection (MOI), the initial number of viruses added per cell. We find that for moderately strong host immune response, the number of infected cells and viral load for a smaller MOI exceeds that for larger MOI, as seen in experiments. We elucidate the mechanism underlying this result as the synergistic action of cytokines secreted by infected cells in controlling viral amplification for larger MOI. We investigate the length and time scales that determine this non-monotonic behavior within the ABM. We study the diffusive spread of virions and cytokines from a single infected cell in an absorbing medium analytically and numerically and deduce the length scale that yields a good estimate of the MOI at which we find non-monotonicity. Detailed computations of the temporal behavior of averaged quantities and spatial measures provide further insights into host-viral interactions and connections to experimental observations.


Subject(s)
Host Microbial Interactions , Influenza A virus , Models, Biological , Animals , Epithelial Cells/virology , Host Microbial Interactions/physiology , Humans , Influenza A virus/physiology , Orthomyxoviridae Infections/physiopathology , Time Factors
16.
Nat Commun ; 10(1): 4107, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511530

ABSTRACT

Influenza is a leading cause of respiratory mortality and morbidity. While inflammation is essential for fighting infection, a balance of anti-viral defense and host tolerance is necessary for recovery. Circadian rhythms have been shown to modulate inflammation. However, the importance of diurnal variability in the timing of influenza infection is not well understood. Here we demonstrate that endogenous rhythms affect survival in influenza infection. Circadian control of influenza infection is mediated by enhanced inflammation as proven by increased cellularity in bronchoalveolar lavage (BAL), pulmonary transcriptomic profile and histology and is not attributable to viral burden. Better survival is associated with a time dependent preponderance of NK and NKT cells and lower proportion of inflammatory monocytes in the lung. Further, using a series of genetic mouse mutants, we elucidate cellular mechanisms underlying circadian gating of influenza infection.


Subject(s)
Circadian Rhythm/physiology , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/physiopathology , Pneumonia/complications , Pneumonia/physiopathology , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/metabolism , Animals , Antigens, Ly , Female , Influenza A virus/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Myeloid Cells/metabolism , Natural Killer T-Cells/immunology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Phenotype , Pneumonia/virology , Time Factors , Transcriptome/genetics , Virus Replication
17.
Chin J Nat Med ; 17(3): 187-197, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30910055

ABSTRACT

Houttuynia cordata polysaccharide (HCP) is extracted from Houttuynia cordata, a key traditional Chinese medicine. The study was to investigate the effects of HCP on intestinal barrier and microbiota in H1N1 virus infected mice. Mice were infected with H1N1 virus and orally administrated HCP at a dosage of 40 mg(kg-1(d-1. H1N1 infection caused pulmonary and intestinal injury and gut microbiota imbalance. HCP significantly suppressed the expression of hypoxia inducible factor-1α and decreased mucosubstances in goblet cells, but restored the level of zonula occludens-1 in intestine. HCP also reversed the composition change of intestinal microbiota caused by H1N1 infection, with significantly reduced relative abundances of Vibrio and Bacillus, the pathogenic bacterial genera. Furthermore, HCP rebalanced the gut microbiota and restored the intestinal homeostasis to some degree. The inhibition of inflammation was associated with the reduced level of Toll-like receptors and interleukin-1ß in intestine, as well as the increased production of interleukin-10. Oral administration of HCP alleviated lung injury and intestinal dysfunction caused by H1N1 infection. HCP may gain systemic treatment by local acting on intestine and microbiota. This study proved the high-value application of HCP.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Houttuynia/chemistry , Orthomyxoviridae Infections/drug therapy , Polysaccharides/therapeutic use , Animals , Cytokines/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/drug therapy , Inflammation/pathology , Influenza A Virus, H1N1 Subtype/pathogenicity , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/physiopathology , Plant Extracts/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Toll-Like Receptors/metabolism , Zonula Occludens-1 Protein/metabolism
18.
J Microbiol ; 57(2): 163-169, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30706345

ABSTRACT

To evaluate the efficacy of a non-adjuvant A/H1N1/2009 influenza A vaccine (GC1115), we demonstrated the immunogenicity and protective efficacy of GC1115 in mouse and ferret models. The immunogenicity of GC1115 was confirmed after intramuscular administration of 1.875, 3.75, 7.5, and 15 µg hemagglutinin antigen (HA) in mice and 7.5, 15, and 30 µg HA in ferrets at 3-week intervals. A single immunization with GC1115 at HA doses > 7.5 µg induced detectable seroconversion in most mice, and all mice given a second dose exhibited high antibody responses in a dose-dependent manner. The mice in the mock (PBS) and 1.875 µg HA immunized groups succumbed by 13 days following A/California/ 04/09 infection, while all mice in groups given more than 3.75 µg HA were protected from lethal challenge with the A/California/04/09 virus. In ferrets, although immunization with even a single dose of 15 or 30 µg of HA induced detectable HI antibodies, all ferrets given two doses of vaccine seroconverted and exhibited HI titers greater than 80 units. Following challenge with A/California/04/09, the mock (PBS) immunized ferrets showed influenza-like clinical symptoms, such as increased numbers of coughs, elevated body temperature, and body weight loss, for 7 days, while GC1115- immunized ferrets showed attenuated clinical symptoms only for short time period (3-4 days). Further, GC1115-immunized ferrets displayed significantly lower viral titers in the upper respiratory tract (nasal cavity) than the mock vaccinated group in a dose-dependent manner. Taken together, this study demonstrates the immunogenicity and protective efficacy of GC1115 as a non-adjuvanted vaccine.


Subject(s)
Immunogenicity, Vaccine/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , Body Temperature , Body Weight , Cough , Disease Models, Animal , Dose-Response Relationship, Immunologic , Ferrets , Hemagglutination Inhibition Tests , Influenza Vaccines/administration & dosage , Injections, Intramuscular , Lung/pathology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/physiopathology , Respiratory System/virology , Survival Rate , Vaccination/methods , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Viral Load
19.
Peptides ; 112: 14-22, 2019 02.
Article in English | MEDLINE | ID: mdl-30447229

ABSTRACT

Influenza A virus is a leading cause of mortality in humans and poses a global health emergency due to its newly adapted and resistant strains. Thus, there is an urgency to develop novel anti-influenza drugs. Peptides are a type of biological molecule having a wide range of inhibitory effects against bacteria, fungi, viruses and cancer cells. The prospects of several peptides and their mechanisms of action have received significant attention. BF-30, a 30 amino acid residue peptide isolated from the venom of the snake, Bungarus fasciatus, is reported to have antibacterial and antitumor activities. Here, we demonstrated that the 50% cytotoxic concentration (CC50) of the peptide to MDCK cells is 67.7 µM. While BF-30 could inhibit the influenza virus strains H1N1, H3N2 and the oseltamivir-resistant strain H1N1, in vitro, with 50% effective concentration (EC50) of 5.2, 7.4 and 18.9 µM, respectively. In animal experiments, mice treated with BF-30 showed 50% survival at a dosage of 4 µM, with an approximately 2 log viral titer decrease in the lung. However, further studies showed that BF-30 worked on only the virus invasion stage, and inhibited the influenza virus infection by causing virion membrane fusion rather than interacting with hemagglutinin or neuraminidase. These results demonstrated that the peptide BF-30 exhibited an effective inhibitory activity against the influenza A virus and could be a promising candidate for influenza virus therapy.


Subject(s)
Cathelicidins/pharmacology , Influenza A virus/drug effects , Membrane Fusion , Orthomyxoviridae Infections/drug therapy , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cathelicidins/therapeutic use , Influenza A virus/physiology , Male , Mice , Orthomyxoviridae Infections/physiopathology , Virion/drug effects , Virion/physiology
20.
Virology ; 526: 155-164, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30390564

ABSTRACT

IFNγ is a key regulator of inflammatory responses but its role in influenza A virus (IAV) pathogenesis is unclear. Our studies show that infection of mice lacking the IFNγ receptor (IFNγR-/-) at a dose which caused severe disease in wild type 129 Sv/Ev (WT) mice resulted in milder clinical symptoms and significantly lower lung virus titers by 6 days post-infection (dpi). Viral spread was reduced in IFNγR-/- lungs at 2 and 4 dpi. Levels of inflammatory cytokines and chemokines were lower in IFNγR-/- mice at 2 dpi and there was less infiltration of monocyte/macrophage lineage cells than in WT mice. There was no difference in CD4+ and CD8+ T cells and alveolar macrophages in the bronchoalveolar lavage fluid (BALF) at 2 and 4 dpi but by 4 dpi IFNγR-/- mice had significantly higher percentages of neutrophils. Our data strongly suggest that IAV can use the inflammatory response to promote viral spread.


Subject(s)
Influenza A virus/pathogenicity , Orthomyxoviridae Infections/physiopathology , Receptors, Interferon/genetics , Signal Transduction , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/metabolism , Lung/metabolism , Lung/pathology , Lung/virology , Macrophages/immunology , Mice , Mice, Transgenic , Neutrophils/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Signal Transduction/genetics , Viral Load , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...