Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
ACS Infect Dis ; 1(9): 399-452, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26448476

ABSTRACT

While differences in the rate of virus fusion and budding from the host cell membrane have been correlated with pathogenicity, no systematic study of the contribution of differences in viral envelope composition has previously been attempted. Using rigorous virus purification, marked differences between virions and host were observed. Over 125 phospholipid species have been quantitated for three strains of influenza (HKx31- H3N2, PR8- H1N1, and VN1203- H5N1) grown in eggs. The glycerophospholipid composition of purified virions differs from that of the host or that of typical mammalian cells. Phosphatidylcholine is the major component in most mammalian cell membranes, while in purified virions phosphatidylethanolamine dominates. Due to its effects on membrane curvature, it is likely that the variations in its content are important to viral processing during infection. This integrated method of virion isolation with systematic analysis of glycerophospholipids provides a tool for the assessment of species specific biomarkers of viral pathogenicity.

2.
PLoS Pathog ; 11(2): e1004642, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25668410

ABSTRACT

The recent emergence of a novel H7N9 influenza A virus (IAV) causing severe human infections in China raises concerns about a possible pandemic. The lack of pre-existing neutralizing antibodies in the broader population highlights the potential protective role of IAV-specific CD8(+) cytotoxic T lymphocyte (CTL) memory specific for epitopes conserved between H7N9 and previously encountered IAVs. In the present study, the heterosubtypic immunity generated by prior H9N2 or H1N1 infections significantly, but variably, reduced morbidity and mortality, pulmonary virus load and time to clearance in mice challenged with the H7N9 virus. In all cases, the recall of established CTL memory was characterized by earlier, greater airway infiltration of effectors targeting the conserved or cross-reactive H7N9 IAV peptides; though, depending on the priming IAV, each case was accompanied by distinct CTL epitope immunodominance hierarchies for the prominent K(b)PB(1703, D(b)PA(224), and D(b)NP(366) epitopes. While the presence of conserved, variable, or cross-reactive epitopes between the priming H9N2 and H1N1 and the challenge H7N9 IAVs clearly influenced any change in the immunodominance hierarchy, the changing patterns were not tied solely to epitope conservation. Furthermore, the total size of the IAV-specific memory CTL pool after priming was a better predictor of favorable outcomes than the extent of epitope conservation or secondary CTL expansion. Modifying the size of the memory CTL pool significantly altered its subsequent protective efficacy on disease severity or virus clearance, confirming the important role of heterologous priming. These findings establish that both the protective efficacy of heterosubtypic immunity and CTL immunodominance hierarchies are reflective of the immunological history of the host, a finding that has implications for understanding human CTL responses and the rational design of CTL-mediated vaccines.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunity, Heterologous/immunology , Immunodominant Epitopes/immunology , Immunologic Memory/immunology , Influenza A Virus, H7N9 Subtype/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cross Reactions/immunology , Disease Models, Animal , Female , Flow Cytometry , Male , Mice , Mice, Inbred C57BL
3.
J Virol ; 88(3): 1636-51, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24257598

ABSTRACT

One question that continues to challenge influenza A research is why some strains of virus are so devastating compared to their more mild counterparts. We approached this question from an immunological perspective, investigating the CD8(+) T cell response in a mouse model system comparing high- and low-pathological influenza virus infections. Our findings reveal that the early (day 0 to 5) viral titer was not the determining factor in the outcome of disease. Instead, increased numbers of antigen-specific CD8(+) T cells and elevated effector function on a per-cell basis were found in the low-pathological infection and correlated with reduced illness and later-time-point (day 6 to 10) viral titer. High-pathological infection was associated with increased PD-1 expression on influenza virus-specific CD8(+) T cells, and blockade of PD-L1 in vivo led to reduced virus titers and increased CD8(+) T cell numbers in high- but not low-pathological infection, though T cell functionality was not restored. These data show that high-pathological acute influenza virus infection is associated with a dysregulated CD8(+) T cell response, which is likely caused by the more highly inflamed airway microenvironment during the early days of infection. Therapeutic approaches specifically aimed at modulating innate airway inflammation may therefore promote efficient CD8(+) T cell activity. We show that during a severe influenza virus infection, one type of immune cell, the CD8 T cell, is less abundant and less functional than in a more mild infection. This dysregulated T cell phenotype correlates with a lower rate of virus clearance in the severe infection and is partially regulated by the expression of a suppressive coreceptor called PD-1. Treatment with an antibody that blocks PD-1 improves T cell functionality and increases virus clearance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza A virus/physiology , Influenza, Human/immunology , Programmed Cell Death 1 Receptor/genetics , Up-Regulation , Animals , CD8-Positive T-Lymphocytes/virology , Female , Humans , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza, Human/pathology , Influenza, Human/virology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Species Specificity , Virulence
4.
J Gen Virol ; 95(Pt 2): 350-362, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24243730

ABSTRACT

Type I alveolar epithelial cells are a replicative niche for influenza in vivo, yet their response to infection is not fully understood. To better characterize their cellular responses, we have created an immortalized murine lung epithelial type I cell line (LET1). These cells support spreading influenza virus infection in the absence of exogenous protease and thus permit simultaneous analysis of viral replication dynamics and host cell responses. LET1 cells can be productively infected with human, swine and mouse-adapted strains of influenza virus and exhibit expression of an antiviral transcriptional programme and robust cytokine secretion. We characterized influenza virus replication dynamics and host responses of lung type I epithelial cells and identified the capacity of epithelial cell-derived type I IFN to regulate specific modules of antiviral effectors to establish an effective antiviral state. Together, our results indicate that the type I epithelial cell can play a major role in restricting influenza virus infection without contribution from the haematopoietic compartment.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/virology , Immunity, Innate , Influenza A virus/immunology , Influenza A virus/physiology , Virus Replication , Animals , Cell Line , Interferon Type I/immunology , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL
5.
Nat Immunol ; 14(12): 1266-76, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24141387

ABSTRACT

Highly pathogenic avian influenza viruses pose a continuing global threat. Current vaccines will not protect against newly evolved pandemic viruses. The creation of 'universal' vaccines has been unsuccessful because the immunological mechanisms that promote heterosubtypic immunity are incompletely defined. We found here that rapamycin, an immunosuppressive drug that inhibits the kinase mTOR, promoted cross-strain protection against lethal infection with influenza virus of various subtypes when administered during immunization with influenza virus subtype H3N2. Rapamycin reduced the formation of germinal centers and inhibited class switching in B cells, which yielded a unique repertoire of antibodies that mediated heterosubtypic protection. Our data established a requirement for the mTORC1 complex in B cell class switching and demonstrated that rapamycin skewed the antibody response away from high-affinity variant epitopes and targeted more conserved elements of hemagglutinin. Our findings have implications for the design of a vaccine against influenza virus.


Subject(s)
Adaptive Immunity/immunology , Antibody Formation/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , TOR Serine-Threonine Kinases/immunology , Animals , Antibodies, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line , Female , Flow Cytometry , Host-Pathogen Interactions/immunology , Immunoglobulin Class Switching/drug effects , Immunoglobulin Class Switching/immunology , Immunoglobulin M/immunology , Immunosuppressive Agents/pharmacology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/physiology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Orthomyxoviridae/classification , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Sirolimus/pharmacology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
6.
Am J Physiol Lung Cell Mol Physiol ; 304(7): L481-8, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23355384

ABSTRACT

During influenza virus infection, it is unclear how much alveolar cell loss can be tolerated before the host succumbs to the disease. We sought to define relevant correlates of disease severity in the mouse influenza model, hypothesizing that a susceptibility threshold exists for alveolar epithelial cell loss. We compared lung pathology, virus spread, alveolar epithelial cell depletion, arterial blood oxygenation, physiological responses measured by unrestrained plethysmography, and oxygen consumption and carbon dioxide production by gas analysis in mice at intervals after infection with virus strains and doses that cause mild (x31) or severe (PR/8) influenza. Both mild and severe infections showed similar degrees of lung damage and virus dissemination until day 6 after inoculation but diverged in survival outcomes from day 9. Day 6 PR/8-infected mice had normal respiratory and gas exchange functions with 10% type I cell loss. However, day 10 PR/8-infected mice had 40% type I cell loss with a concomitant drastic decreases in tidal and minute volumes, Vo(2), Vco(2), and arterial blood oxygenation, compared with a maximum 3% type I cell loss for x31 on day 10 when they recovered body weight and respiratory functions. Alterations in breaths per minute, expiratory time, and metabolic rate were observed in both infections. A threshold for maintenance of proper respiratory function appears to be crossed once 10% of alveolar type I cells are lost. These data indicate that lethality in influenza virus infection is a matter of degree rather than quality.


Subject(s)
Epithelial Cells/metabolism , Influenza A virus , Orthomyxoviridae Infections/metabolism , Oxygen Consumption , Pulmonary Alveoli/metabolism , Pulmonary Gas Exchange , Animals , Disease Models, Animal , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Mice , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/physiopathology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology , Pulmonary Alveoli/virology , Respiratory Mechanics
7.
J Neurosci ; 32(5): 1545-59, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22302798

ABSTRACT

The A/VN/1203/04 strain of the H5N1 influenza virus is capable of infecting the CNS of mice and inducing a number of neurodegenerative pathologies. Here, we examined the effects of H5N1 on several pathological aspects affected in parkinsonism, including loss of the phenotype of dopaminergic neurons located in the substantia nigra pars compacta (SNpc), expression of monoamines and indolamines in brain, alterations in SNpc microglia number and morphology, and expression of cytokines, chemokines, and growth factors. We find that H5N1 induces a transient loss of the dopaminergic phenotype in SNpc and now report that this loss recovers by 90 d after infection. A similar pattern of loss and recovery was seen in monoamine levels of the basal ganglia. The inflammatory response in lung and different regions of the brain known to be targets of the H5N1 virus (brainstem, substantia nigra, striatum, and cortex) were examined at 3, 10, 21, 60, and 90 d after infection. In each of these brain regions, we found a significant increase in the number of activated microglia that lasted at least 90 d. We also quantified expression of IL-1α, IL-1ß, IL-2, IL-6, IL-9, IL-10, IL-12(p70), IL-13, TNF-α, IFN-γ, granulocyte-macrophage colony-stimulating factor, granulocyte colony-stimulating factor, macrophage colony-stimulating factor, eotaxin, interferon-inducible protein 10, cytokine-induced neutrophil chemoattractant, monocyte chemotactic protein-1, macrophage inflammatory protein (MIP) 1α, MIP-1ß, and VEGF, and found that the pattern and levels of expression are dependent on both brain region and time after infection. We conclude that H5N1 infection in mice induces a long-lasting inflammatory response in brain and may play a contributing factor in the development of pathologies in neurodegenerative disorders.


Subject(s)
Central Nervous System Viral Diseases/pathology , Inflammation Mediators/adverse effects , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/pathology , Animals , Brain/metabolism , Brain/pathology , Brain/virology , Central Nervous System Viral Diseases/metabolism , Central Nervous System Viral Diseases/virology , Chick Embryo , Female , Humans , Inflammation/pathology , Inflammation/virology , Inflammation Mediators/metabolism , Influenza, Human/metabolism , Influenza, Human/virology , Mice , Mice, Inbred C57BL
8.
PLoS Pathog ; 7(12): e1002377, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22144888

ABSTRACT

Following respiratory syncytial virus infection of adult CB6F1 hybrid mice, a predictable CD8+ T cell epitope hierarchy is established with a strongly dominant response to a K(d)-restricted peptide (SYIGSINNI) from the M2 protein. The response to K(d)M2(82-90) is ∼5-fold higher than the response to a subdominant epitope from the M protein (NAITNAKII, D(b)M(187-195)). After infection of neonatal mice, a distinctly different epitope hierarchy emerges with codominant responses to K(d)M2(82-90) and D(b)M(187-195). Adoptive transfer of naïve CD8+ T cells from adults into congenic neonates prior to infection indicates that intrinsic CD8+ T cell factors contribute to age-related differences in hierarchy. Epitope-specific precursor frequency differs between adults and neonates and influences, but does not predict the hierarchy following infection. Additionally, dominance of K(d)M2(82-90)-specific cells does not correlate with TdT activity. Epitope-specific Vß repertoire usage is more restricted and functional avidity is lower in neonatal mice. The neonatal pattern of codominance changes after infection at 10 days of age, and rapidly shifts to the adult pattern of extreme K(d)M2(82-90)-dominance. Thus, the functional properties of T cells are selectively modified by developmental factors in an epitope-specific and age-dependent manner.


Subject(s)
Aging/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Viral Proteins/immunology , Adoptive Transfer , Animals , Animals, Newborn , Female , Male , Mice , Mice, Inbred BALB C
9.
Proc Natl Acad Sci U S A ; 108(35): 14602-7, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21873213

ABSTRACT

It is currently thought that T cells with specificity for self-peptide/MHC (pMHC) ligands are deleted during thymic development, thereby preventing autoimmunity. In the case of CD4(+) T cells, what is unclear is the extent to which self-peptide/MHC class II (pMHCII)-specific T cells are deleted or become Foxp3(+) regulatory T cells. We addressed this issue by characterizing a natural polyclonal pMHCII-specific CD4(+) T-cell population in mice that either lacked or expressed the relevant antigen in a ubiquitous pattern. Mice expressing the antigen contained one-third the number of pMHCII-specific T cells as mice lacking the antigen, and the remaining cells exhibited low TCR avidity. In mice lacking the antigen, the pMHCII-specific T-cell population was dominated by phenotypically naive Foxp3(-) cells, but also contained a subset of Foxp3(+) regulatory cells. Both Foxp3(-) and Foxp3(+) pMHCII-specific T-cell numbers were reduced in mice expressing the antigen, but the Foxp3(+) subset was more resistant to changes in number and TCR repertoire. Therefore, thymic selection of self-pMHCII-specific CD4(+) T cells results in incomplete deletion within the normal polyclonal repertoire, especially among regulatory T cells.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/analysis , Histocompatibility Antigens Class II/immunology , Thymus Gland/immunology , Animals , Clonal Deletion , Immune Tolerance , Mice , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes, Regulatory/immunology
11.
J Clin Invest ; 121(1): 288-95, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21135507

ABSTRACT

Characterizing the TCRα and TCRß chains expressed by T cells responding to a given pathogen or underlying autoimmunity helps in the development of vaccines and immunotherapies, respectively. However, our understanding of complementary TCRα and TCRß chain utilization is very limited for pathogen- and autoantigen-induced immunity. To address this problem, we have developed a multiplex nested RT-PCR method for the simultaneous amplification of transcripts encoding the TCRα and TCRß chains from single cells. This multiplex method circumvented the lack of antibodies specific for variable regions of mouse TCRα chains and the need for prior knowledge of variable region usage in the TCRß chain, resulting in a comprehensive, unbiased TCR repertoire analysis with paired coexpression of TCRα and TCRß chains with single-cell resolution. Using CD8+ CTLs specific for an influenza epitope recovered directly from the pneumonic lungs of mice, this technique determined that 25% of such effectors expressed a dominant, nonproductively rearranged Tcra transcript. T cells with these out-of-frame Tcra mRNAs also expressed an alternate, in-frame Tcra, whereas approximately 10% of T cells had 2 productive Tcra transcripts. The proportion of cells with biallelic transcription increased over the course of a response, a finding that has implications for immune memory and autoimmunity. This technique may have broad applications in mouse models of human disease.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Alleles , Amino Acid Sequence , Animals , Antigens, Viral/immunology , Complementarity Determining Regions , Epitopes/immunology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Orthomyxoviridae/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transcription, Genetic
12.
J Virol ; 84(16): 8042-50, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20519385

ABSTRACT

The fitness of oseltamivir-resistant highly pathogenic H5N1 influenza viruses has important clinical implications. We generated recombinant human A/Vietnam/1203/04 (VN; clade 1) and A/Turkey/15/06 (TK; clade 2.2) influenza viruses containing the H274Y neuraminidase (NA) mutation, which confers resistance to NA inhibitors, and compared the fitness levels of the wild-type (WT) and resistant virus pairs in ferrets. The VN-H274Y and VN-WT viruses replicated to similar titers in the upper respiratory tract (URT) and caused comparable disease signs, and none of the animals survived. On days 1 to 3 postinoculation, disease signs caused by oseltamivir-resistant TK-H274Y virus were milder than those caused by TK-WT virus, and all animals survived. We then studied fitness by using a novel approach. We coinoculated ferrets with different ratios of oseltamivir-resistant and -sensitive H5N1 viruses and measured the proportion of clones in day-6 nasal washes that contained the H274Y NA mutation. Although the proportion of VN-H274Y clones increased consistently, that of TK-H274Y virus decreased. Mutations within NA catalytic (R292K) and framework (E119A/K, I222L, H274L, and N294S) sites or near the NA enzyme active site (V116I, I117T/V, Q136H, K150N, and A250T) emerged spontaneously (without drug pressure) in both pairs of viruses. The NA substitutions I254V and E276A could exert a compensatory effect on the fitness of VN-H274Y and TK-H274Y viruses. NA enzymatic function was reduced in both drug-resistant H5N1 viruses. These results show that the H274Y NA mutation affects the fitness of two H5N1 influenza viruses differently. Our novel method of assessing viral fitness accounts for both virus-host interactions and virus-virus interactions within the host.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral , Influenza A Virus, H5N1 Subtype/pathogenicity , Mutation, Missense , Neuraminidase/genetics , Oseltamivir/pharmacology , Viral Proteins/genetics , Virus Replication , Amino Acid Substitution/genetics , Animals , Disease Models, Animal , Ferrets , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/genetics , Male , Neuraminidase/physiology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Recombination, Genetic , Respiratory System/virology , Survival Analysis , Viral Proteins/physiology , Virulence
13.
Antimicrob Agents Chemother ; 53(7): 3088-96, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19349520

ABSTRACT

While the neuraminidase (NA) inhibitor oseltamivir is currently our first line of defense against a pandemic threat, there is little information about whether in vitro testing can predict the in vivo effectiveness of antiviral treatment. Using a panel of five H5N1 influenza viruses (H5 clades 1 and 2), we determined that four viruses were susceptible to the drug in vitro (mean 50% inhibitory concentration [IC(50)], 0.1 to 4.9 nM), and A/Turkey/65-1242/06 virus was slightly less susceptible (mean IC(50), 10.8 nM). Two avian viruses showed significantly greater NA enzymatic activity (V(max)) than the human viruses, and the five viruses varied in their affinity for the NA substrate MUNANA (K(m), 64 to 300 muM) and for oseltamivir carboxylate (K(i), 0.1 to 7.9 nM). The protection of mice provided by a standard oseltamivir regimen (20 mg/kg/day for 5 days) also varied among the viruses used. We observed (i) complete protection against the less virulent A/chicken/Jogjakarta/BBVET/IX/04 virus; (ii) moderate protection (60 to 80% survival) against three viruses, two of which are neurotropic; and (iii) no protection against A/Turkey/65-1242/06 virus, which induced high pulmonary expression of proinflammatory mediators (interleukin-1alpha [IL-1alpha], IL-6, alpha interferon, and monocyte chemotactic protein 1) and contained a minor subpopulation of drug-resistant clones (I117V and E119A NA mutations). We found no correlation between in vitro susceptibility and in vivo protection (Spearman rank correlation coefficient rho = -0.1; P > 0.05). Therefore, the in vivo efficacy of oseltamivir against highly pathogenic H5N1 influenza viruses cannot be reliably predicted by susceptibility testing, and more prognostic ways to evaluate anti-influenza compounds must be developed. Multiple viral and host factors modulate the effectiveness of NA inhibitor regimens against such viruses and new, more consistently effective treatment options, including combination therapies, are needed.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H5N1 Subtype/pathogenicity , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Animals , Brain/virology , Drug Resistance, Viral/genetics , Female , Lung/virology , Mice , Mice, Inbred BALB C , Virus Replication/drug effects
14.
J Infect Dis ; 197(9): 1315-23, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18422444

ABSTRACT

BACKGROUND: Current oseltamivir prophylactic regimens may not be as effective against highly pathogenic H5N1 influenza viruses as they are against less pathogenic strains. An optimal regimen is urgently needed. METHODS: Ferrets were given the neuraminidase inhibitor oseltamivir orally for 10 days (5 or 10 mg/kg once daily or 2.5 or 5 mg/kg twice daily). Prophylaxis was initiated 1 day before infection, and oseltamivir was given 4 h before the ferrets were inoculated with a lethal dose of A/Vietnam/1203/04 (H5N1) influenza virus. RESULTS: At a dose of 5 mg/kg once daily, oseltamivir prevented death but not clinical signs of infection in ferrets; severe pathology was observed in the lungs, brain, and liver. At 10 mg/kg once daily, oseltamivir reduced clinical symptoms and systemic virus replication, but pathology was observed in the internal organs. The best results were obtained at a dose of 2.5 or 5 mg/kg given twice daily. Both regimens resulted in 100% survival and the absence of clinical symptoms, systemic virus spread, and organ pathology. Serum antibody titers were comparable across regimens and were sufficient to protect against rechallenge. CONCLUSIONS: An increased dose of oseltamivir or twice-daily administration effectively protects ferrets against morbidity and mortality caused by H5N1 infection and does not interfere with the development of protective antibodies against subsequent H5N1 infection.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H5N1 Subtype/drug effects , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/prevention & control , Oseltamivir/therapeutic use , Administration, Oral , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Ferrets , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/pathology , Oseltamivir/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...