ϟ

Jeffery K. Taubenberger

Here are all the papers by Jeffery K. Taubenberger that you can download and read on OA.mg.
Jeffery K. Taubenberger’s last known institution is . Download Jeffery K. Taubenberger PDFs here.

Claim this Profile →
DOI: 10.1086/591708
2008
Cited 1,446 times
Predominant Role of Bacterial Pneumonia as a Cause of Death in Pandemic Influenza: Implications for Pandemic Influenza Preparedness
Despite the availability of published data on 4 pandemics that have occurred over the past 120 years, there is little modern information on the causes of death associated with influenza pandemics.We examined relevant information from the most recent influenza pandemic that occurred during the era prior to the use of antibiotics, the 1918-1919 "Spanish flu" pandemic. We examined lung tissue sections obtained during 58 autopsies and reviewed pathologic and bacteriologic data from 109 published autopsy series that described 8398 individual autopsy investigations.The postmortem samples we examined from people who died of influenza during 1918-1919 uniformly exhibited severe changes indicative of bacterial pneumonia. Bacteriologic and histopathologic results from published autopsy series clearly and consistently implicated secondary bacterial pneumonia caused by common upper respiratory-tract bacteria in most influenza fatalities.The majority of deaths in the 1918-1919 influenza pandemic likely resulted directly from secondary bacterial pneumonia caused by common upper respiratory-tract bacteria. Less substantial data from the subsequent 1957 and 1968 pandemics are consistent with these findings. If severe pandemic influenza is largely a problem of viral-bacterial copathogenesis, pandemic planning needs to go beyond addressing the viral cause alone (e.g., influenza vaccines and antiviral drugs). Prevention, diagnosis, prophylaxis, and treatment of secondary bacterial pneumonia, as well as stockpiling of antibiotics and bacterial vaccines, should also be high priorities for pandemic planning.
DOI: 10.3201/eid1209.05-0979
2006
Cited 1,249 times
1918 Influenza: the Mother of All Pandemics
The "Spanish" influenza pandemic of 1918-1919, which caused ≈50 million deaths worldwide, remains an ominous warning to public health.Many questions about its origins, its unusual epidemiologic features, and the basis of its pathogenicity remain unanswered.The public health implications of the pandemic therefore remain in doubt even as we now grapple with the feared emergence of a pandemic caused by H5N1 or other virus.However, new information about the 1918 virus is emerging, for example, sequencing of the entire genome from archival autopsy tissues.But, the viral genome alone is unlikely to provide answers to some critical questions.Understanding the 1918 pandemic and its implications for future pandemics requires careful experimentation and in-depth historical analysis.
DOI: 10.1126/science.1119392
2005
Cited 1,181 times
Characterization of the Reconstructed 1918 Spanish Influenza Pandemic Virus
The pandemic influenza virus of 1918-1919 killed an estimated 20 to 50 million people worldwide. With the recent availability of the complete 1918 influenza virus coding sequence, we used reverse genetics to generate an influenza virus bearing all eight gene segments of the pandemic virus to study the properties associated with its extraordinary virulence. In stark contrast to contemporary human influenza H1N1 viruses, the 1918 pandemic virus had the ability to replicate in the absence of trypsin, caused death in mice and embryonated chicken eggs, and displayed a high-growth phenotype in human bronchial epithelial cells. Moreover, the coordinated expression of the 1918 virus genes most certainly confers the unique high-virulence phenotype observed with this pandemic virus.
DOI: 10.1038/nature04230
2005
Cited 978 times
Characterization of the 1918 influenza virus polymerase genes
The influenza A viral heterotrimeric polymerase complex (PA, PB1, PB2) is known to be involved in many aspects of viral replication and to interact with host factors, thereby having a role in host specificity. The polymerase protein sequences from the 1918 human influenza virus differ from avian consensus sequences at only a small number of amino acids, consistent with the hypothesis that they were derived from an avian source shortly before the pandemic. However, when compared to avian sequences, the nucleotide sequences of the 1918 polymerase genes have more synonymous differences than expected, suggesting evolutionary distance from known avian strains. Here we present sequence and phylogenetic analyses of the complete genome of the 1918 influenza virus, and propose that the 1918 virus was not a reassortant virus (like those of the 1957 and 1968 pandemics), but more likely an entirely avian-like virus that adapted to humans. These data support prior phylogenetic studies suggesting that the 1918 virus was derived from an avian source. A total of ten amino acid changes in the polymerase proteins consistently differentiate the 1918 and subsequent human influenza virus sequences from avian virus sequences. Notably, a number of the same changes have been found in recently circulating, highly pathogenic H5N1 viruses that have caused illness and death in humans and are feared to be the precursors of a new influenza pandemic. The sequence changes identified here may be important in the adaptation of influenza viruses to humans.
DOI: 10.3201/eid1201.050979
2006
Cited 971 times
1918 Influenza: the Mother of All Pandemics
The "Spanish" influenza pandemic of 1918-1919, which caused approximately 50 million deaths worldwide, remains an ominous warning to public health. Many questions about its origins, its unusual epidemiologic features, and the basis of its pathogenicity remain unanswered. The public health implications of the pandemic therefore remain in doubt even as we now grapple with the feared emergence of a pandemic caused by H5N1 or other virus. However, new information about the 1918 virus is emerging, for example, sequencing of the entire genome from archival autopsy tissues. But, the viral genome alone is unlikely to provide answers to some critical questions. Understanding the 1918 pandemic and its implications for future pandemics requires careful experimentation and in-depth historical analysis.
DOI: 10.1146/annurev.pathmechdis.3.121806.154316
2008
Cited 969 times
The Pathology of Influenza Virus Infections
Influenza viruses are significant human respiratory pathogens that cause both seasonal, endemic infections and periodic, unpredictable pandemics. The worst pandemic on record, in 1918, killed approximately 50 million people worldwide. Human infections caused by H5N1 highly pathogenic avian influenza viruses have raised concern about the emergence of another pandemic. The histopathology of fatal influenza virus pneumonias as documented over the past 120 years is reviewed here. Strikingly, the spectrum of pathologic changes described in the 1918 influenza pandemic is not significantly different from the histopathology observed in other less lethal pandemics or even in deaths occurring during seasonal influenza outbreaks.
DOI: 10.1126/science.1124513
2006
Cited 884 times
Structure and Receptor Specificity of the Hemagglutinin from an H5N1 Influenza Virus
The hemagglutinin (HA) structure at 2.9 angstrom resolution, from a highly pathogenic Vietnamese H5N1 influenza virus, is more related to the 1918 and other human H1 HAs than to a 1997 duck H5 HA. Glycan microarray analysis of this Viet04 HA reveals an avian alpha2-3 sialic acid receptor binding preference. Introduction of mutations that can convert H1 serotype HAs to human alpha2-6 receptor specificity only enhanced or reduced affinity for avian-type receptors. However, mutations that can convert avian H2 and H3 HAs to human receptor specificity, when inserted onto the Viet04 H5 HA framework, permitted binding to a natural human alpha2-6 glycan, which suggests a path for this H5N1 virus to gain a foothold in the human population.
DOI: 10.1038/nature06945
2008
Cited 868 times
The genomic and epidemiological dynamics of human influenza A virus
The evolutionary interaction between influenza A virus and the human immune system, manifest as 'antigenic drift' of the viral haemagglutinin, is one of the best described patterns in molecular evolution. However, little is known about the genome-scale evolutionary dynamics of this pathogen. Similarly, how genomic processes relate to global influenza epidemiology, in which the A/H3N2 and A/H1N1 subtypes co-circulate, is poorly understood. Here through an analysis of 1,302 complete viral genomes sampled from temperate populations in both hemispheres, we show that the genomic evolution of influenza A virus is characterized by a complex interplay between frequent reassortment and periodic selective sweeps. The A/H3N2 and A/H1N1 subtypes exhibit different evolutionary dynamics, with diverse lineages circulating in A/H1N1, indicative of weaker antigenic drift. These results suggest a sink-source model of viral ecology in which new lineages are seeded from a persistent influenza reservoir, which we hypothesize to be located in the tropics, to sink populations in temperate regions.
DOI: 10.1016/j.chom.2010.05.009
2010
Cited 734 times
Influenza Virus Evolution, Host Adaptation, and Pandemic Formation
Newly emerging or "re-emerging" viral diseases continue to pose significant global public health threats. Prototypic are influenza viruses that are major causes of human respiratory infections and mortality. Influenza viruses can cause zoonotic infections and adapt to humans, leading to sustained transmission and emergence of novel viruses. Mechanisms by which viruses evolve in one host, cause zoonotic infection, and adapt to a new host species remain unelucidated. Here, we review the evolution of influenza A viruses in their reservoir hosts and discuss genetic changes associated with introduction of novel viruses into humans, leading to pandemics and the establishment of seasonal viruses.
DOI: 10.1126/science.275.5307.1793
1997
Cited 662 times
Initial Genetic Characterization of the 1918 “Spanish” Influenza Virus
The "Spanish" influenza pandemic killed at least 20 million people in 1918-1919, making it the worst infectious pandemic in history. Understanding the origins of the 1918 virus and the basis for its exceptional virulence may aid in the prediction of future influenza pandemics. RNA from a victim of the 1918 pandemic was isolated from a formalin-fixed, paraffin-embedded, lung tissue sample. Nine fragments of viral RNA were sequenced from the coding regions of hemagglutinin, neuraminidase, nucleoprotein, matrix protein 1, and matrix protein 2. The sequences are consistent with a novel H1N1 influenza A virus that belongs to the subgroup of strains that infect humans and swine, not the avian subgroup.
DOI: 10.1016/j.jmb.2005.11.002
2006
Cited 575 times
Glycan Microarray Analysis of the Hemagglutinins from Modern and Pandemic Influenza Viruses Reveals Different Receptor Specificities
Influenza A virus specificity for the host is mediated by the viral surface glycoprotein hemagglutinin (HA), which binds to receptors containing glycans with terminal sialic acids. Avian viruses preferentially bind to alpha2-3-linked sialic acids on receptors of intestinal epithelial cells, whereas human viruses are specific for the alpha2-6 linkage on epithelial cells of the lungs and upper respiratory tract. To define the receptor preferences of a number of human and avian H1 and H3 viruses, including the 1918 H1N1 pandemic strains, their hemagglutinins were analyzed using a recently described glycan array. The array, which contains 200 carbohydrates and glycoproteins, not only revealed clear differentiation of receptor preferences for alpha2-3 and/or alpha2-6 sialic acid linkage, but could also detect fine differences in HA specificity, such as preferences for fucosylation, sulfation and sialylation at positions 2 (Gal) and 3 (GlcNAc, GalNAc) of the terminal trisaccharide. For the two 1918 HA variants, the South Carolina (SC) HA (with Asp190, Asp225) bound exclusively alpha2-6 receptors, while the New York (NY) variant, which differed only by one residue (Gly225), had mixed alpha2-6/alpha2-3 specificity, especially for sulfated oligosaccharides. Only one mutation of the NY variant (Asp190Glu) was sufficient to revert the HA receptor preference to that of classical avian strains. Thus, the species barrier, as defined by the receptor specificity preferences of 1918 human viruses compared to likely avian virus progenitors, can be circumvented by changes at only two positions in the HA receptor binding site. The glycan array thus provides highly detailed profiles of influenza receptor specificity that can be used to map the evolution of new human pathogenic strains, such as the H5N1 avian influenza.
DOI: 10.1126/science.1222213
2012
Cited 569 times
An Overlapping Protein-Coding Region in Influenza A Virus Segment 3 Modulates the Host Response
Influenza A virus (IAV) infection leads to variable and imperfectly understood pathogenicity. We report that segment 3 of the virus contains a second open reading frame ("X-ORF"), accessed via ribosomal frameshifting. The frameshift product, termed PA-X, comprises the endonuclease domain of the viral PA protein with a C-terminal domain encoded by the X-ORF and functions to repress cellular gene expression. PA-X also modulates IAV virulence in a mouse infection model, acting to decrease pathogenicity. Loss of PA-X expression leads to changes in the kinetics of the global host response, which notably includes increases in inflammatory, apoptotic, and T lymphocyte-signaling pathways. Thus, we have identified a previously unknown IAV protein that modulates the host response to infection, a finding with important implications for understanding IAV pathogenesis.
DOI: 10.1073/pnas.96.4.1651
1999
Cited 566 times
Origin and evolution of the 1918 “Spanish” influenza virus hemagglutinin gene
The "Spanish" influenza pandemic killed over 20 million people in 1918 and 1919, making it the worst infectious pandemic in history. Here, we report the complete sequence of the hemagglutinin (HA) gene of the 1918 virus. Influenza RNA for the analysis was isolated from a formalin-fixed, paraffin-embedded lung tissue sample prepared during the autopsy of a victim of the influenza pandemic in 1918. Influenza RNA was also isolated from lung tissue samples from two additional victims of the lethal 1918 influenza: one formalin-fixed, paraffin-embedded sample and one frozen sample obtained by in situ biopsy of the lung of a victim buried in permafrost since 1918. The complete coding sequence of the A/South Carolina/1/18 HA gene was obtained. The HA1 domain sequence was confirmed by using the two additional isolates (A/New York/1/18 and A/Brevig Mission/1/18). The sequences show little variation. Phylogenetic analyses suggest that the 1918 virus HA gene, although more closely related to avian strains than any other mammalian sequence, is mammalian and may have been adapting in humans before 1918.
DOI: 10.1038/nature05181
2006
Cited 525 times
Genomic analysis of increased host immune and cell death responses induced by 1918 influenza virus
The influenza pandemic of 1918-19 was responsible for about 50 million deaths worldwide. Modern histopathological analysis of autopsy samples from human influenza cases from 1918 revealed significant damage to the lungs with acute, focal bronchitis and alveolitis associated with massive pulmonary oedema, haemorrhage and rapid destruction of the respiratory epithelium. The contribution of the host immune response leading to this severe pathology remains largely unknown. Here we show, in a comprehensive analysis of the global host response induced by the 1918 influenza virus, that mice infected with the reconstructed 1918 influenza virus displayed an increased and accelerated activation of host immune response genes associated with severe pulmonary pathology. We found that mice infected with a virus containing all eight genes from the pandemic virus showed marked activation of pro-inflammatory and cell-death pathways by 24 h after infection that remained unabated until death on day 5. This was in contrast with smaller host immune responses as measured at the genomic level, accompanied by less severe disease pathology and delays in death in mice infected with influenza viruses containing only subsets of 1918 genes. The results indicate a cooperative interaction between the 1918 influenza genes and show that study of the virulence of the 1918 influenza virus requires the use of the fully reconstructed virus. With recent concerns about the introduction of highly pathogenic avian influenza viruses into humans and their potential to cause a worldwide pandemic with disastrous health and economic consequences, a comprehensive understanding of the global host response to the 1918 virus is crucial. Moreover, understanding the contribution of host immune responses to virulent influenza virus infections is an important starting point for the identification of prognostic indicators and the development of novel antiviral therapies.
DOI: 10.1128/jvi.79.23.14933-14944.2005
2005
Cited 492 times
Pathogenicity of Influenza Viruses with Genes from the 1918 Pandemic Virus: Functional Roles of Alveolar Macrophages and Neutrophils in Limiting Virus Replication and Mortality in Mice
ABSTRACT The Spanish influenza pandemic of 1918 to 1919 swept the globe and resulted in the deaths of at least 20 million people. The basis of the pulmonary damage and high lethality caused by the 1918 H1N1 influenza virus remains largely unknown. Recombinant influenza viruses bearing the 1918 influenza virus hemagglutinin (HA) and neuraminidase (NA) glycoproteins were rescued in the genetic background of the human A/Texas/36/91 (H1N1) (1918 HA/NA:Tx/91) virus. Pathogenesis experiments revealed that the 1918 HA/NA:Tx/91 virus was lethal for BALB/c mice without the prior adaptation that is usually required for human influenza A H1N1 viruses. The increased mortality of 1918 HA/NA:Tx/91-infected mice was accompanied by (i) increased (>200-fold) viral replication, (ii) greater influx of neutrophils into the lung, (iii) increased numbers of alveolar macrophages (AMs), and (iv) increased protein expression of cytokines and chemokines in lung tissues compared with the levels seen for control Tx/91 virus-infected mice. Because pathological changes in AMs and neutrophil migration correlated with lung inflammation, we assessed the role of these cells in the pathogenesis associated with 1918 HA/NA:Tx/91 virus infection. Neutrophil and/or AM depletion initiated 3 or 5 days after infection did not have a significant effect on the disease outcome following a lethal 1918 HA/NA:Tx/91 virus infection. By contrast, depletion of these cells before a sublethal infection with 1918 HA/NA:Tx/91 virus resulted in uncontrolled virus growth and mortality in mice. In addition, neutrophil and/or AM depletion was associated with decreased expression of cytokines and chemokines. These results indicate that a human influenza H1N1 virus possessing the 1918 HA and NA glycoproteins can induce severe lung inflammation consisting of AMs and neutrophils, which play a role in controlling the replication and spread of 1918 HA/NA:Tx/91 virus after intranasal infection of mice.
DOI: 10.1126/science.1093373
2004
Cited 449 times
Structure of the Uncleaved Human H1 Hemagglutinin from the Extinct 1918 Influenza Virus
The 1918 "Spanish" influenza pandemic represents the largest recorded outbreak of any infectious disease. The crystal structure of the uncleaved precursor of the major surface antigen of the extinct 1918 virus was determined at 3.0 angstrom resolution after reassembly of the hemagglutinin gene from viral RNA fragments preserved in 1918 formalin-fixed lung tissues. A narrow avian-like receptor-binding site, two previously unobserved histidine patches, and a less exposed surface loop at the cleavage site that activates viral membrane fusion reveal structural features primarily found in avian viruses, which may have contributed to the extraordinarily high infectivity and mortality rates observed during 1918.
DOI: 10.1038/nature04239
2005
Cited 447 times
Large-scale sequencing of human influenza reveals the dynamic nature of viral genome evolution
Influenza viruses are remarkably adept at surviving in the human population over a long timescale. The human influenza A virus continues to thrive even among populations with widespread access to vaccines, and continues to be a major cause of morbidity and mortality. The virus mutates from year to year, making the existing vaccines ineffective on a regular basis, and requiring that new strains be chosen for a new vaccine. Less-frequent major changes, known as antigenic shift, create new strains against which the human population has little protective immunity, thereby causing worldwide pandemics. The most recent pandemics include the 1918 'Spanish' flu, one of the most deadly outbreaks in recorded history, which killed 30-50 million people worldwide, the 1957 'Asian' flu, and the 1968 'Hong Kong' flu. Motivated by the need for a better understanding of influenza evolution, we have developed flexible protocols that make it possible to apply large-scale sequencing techniques to the highly variable influenza genome. Here we report the results of sequencing 209 complete genomes of the human influenza A virus, encompassing a total of 2,821,103 nucleotides. In addition to increasing markedly the number of publicly available, complete influenza virus genomes, we have discovered several anomalies in these first 209 genomes that demonstrate the dynamic nature of influenza transmission and evolution. This new, large-scale sequencing effort promises to provide a more comprehensive picture of the evolution of influenza viruses and of their pattern of transmission through human and animal populations. All data from this project are being deposited, without delay, in public archives.
DOI: 10.5858/134.2.235
2010
Cited 377 times
Pulmonary Pathologic Findings of Fatal 2009 Pandemic Influenza A/H1N1 Viral Infections
In March 2009, a novel swine-origin influenza A/H1N1 virus was identified. After global spread, the World Health Organization in June declared the first influenza pandemic in 41 years.To describe the clinicopathologic characteristics of 34 people who died following confirmed A/H1N1 infection with emphasis on the pulmonary pathology findings.We reviewed medical records, autopsy reports, microbiologic studies, and microscopic slides of 34 people who died between May 15 and July 9, 2009, and were investigated either by the New York City Office of Chief Medical Examiner (32 deaths) or through the consultation service of a coauthor (2 deaths).Most of the 34 decedents (62%) were between 25 and 49 years old (median, 41.5 years). Tracheitis, bronchiolitis, and diffuse alveolar damage were noted in most cases. Influenza viral antigen was observed most commonly in the epithelium of the tracheobronchial tree but also in alveolar epithelial cells and macrophages. Most cases were reverse transcription-polymerase chain reaction positive for influenza. Histologic and microbiologic autopsy evidence of bacterial pneumonia was detected in 55% of cases. Underlying medical conditions including cardiorespiratory diseases and immunosuppression were present in 91% of cases. Obesity (body mass index, >30) was noted in 72% of adult and adolescent cases.The pulmonary pathologic findings in fatal disease caused by the novel pandemic influenza virus are similar to findings identified in past pandemics. Superimposed bacterial infections of the respiratory tract were common. Preexisting obesity, cardiorespiratory diseases, and other comorbidities also were prominent findings among the decedents.
DOI: 10.1016/s0046-8177(03)00367-8
2003
Cited 375 times
Lung pathology of severe acute respiratory syndrome (SARS): a study of 8 autopsy cases from Singapore
Severe acute respiratory syndrome (SARS) is an infectious condition caused by the SARS-associated coronavirus (SARS-CoV), a new member in the family Coronaviridae. To evaluate the lung pathology in this life-threatening respiratory illness, we studied postmortem lung sections from 8 patients who died from SARS during the spring 2003 Singapore outbreak. The predominant pattern of lung injury in all 8 cases was diffuse alveolar damage. The histology varied according to the duration of illness. Cases of 10 or fewer days' duration demonstrated acute-phase diffuse alveolar damage (DAD), airspace edema, and bronchiolar fibrin. Cases of more than 10 days' duration exhibited organizing-phase DAD, type II pneumocyte hyperplasia, squamous metaplasia, multinucleated giant cells, and acute bronchopneumonia. In acute-phase DAD, pancytokeratin staining was positive in hyaline membranes along alveolar walls and highlighted the absence of pneumocytes. Multinucleated cells were shown to be both type II pneumocytes and macrophages by pancytokeratin, thyroid transcription factor-1, and CD68 staining. SARS-CoV RNA was identified by reverse transcriptase-polymerase chain reaction in 7 of 8 cases in fresh autopsy tissue and in 8 of 8 cases in formalin-fixed, paraffin-embedded lung tissue, including the 1 negative case in fresh tissue. Understanding the pathology of DAD in SARS patients may provide the basis for therapeutic strategies. Further studies of the pathogenesis of SARS may reveal new insight into the mechanisms of DAD.
DOI: 10.1002/ana.410340507
1993
Cited 366 times
Correlation between magnetic resonance imaging findings and lesion development in chronic, active multiple sclerosis
Abstract Magnetic resonance imaging is a highly sensitive method for the detection of the lesions of multiple sclerosis and renders possible the study and the evolution of early lesions. Previous reports on magnetic resonance imaging following gandolinium‐diethylenetriamine pentaacetic acid (Gd‐DTPA) injection demonstrated that new lesions can be recognized by contrast enhancement. The pathological basis of these observations is uncertain. We have had the opportunity to study at autopsy the brain of a patient with chronic progressive multiple sclerosis who suffered acute worsening leading to death. Magnetic resonance imaging performed 10 days and 4 weeks prior to death showed new Gd‐DTPA‐enhanced lesions in the posterior hemispheric white matter adjacent to the lateral ventricles. Light microscopic examination of these areas demonstrated them to be fresh lesions comprising intense inflammatory activity and dense perivascular cuffs within an edematous lesion center and a striking parenchymal mononuclear cell infiltration at the margins of the lesions. Lesions that were demonstrated by increased signal on T2‐weighted images, but were not enhanced following administration of Gd‐DTPA, were all of the chronic type, either inactive or active. None of these showed the intense inflammatory activity of the acute lesions and most displayed fibrous astrogliosis.
DOI: 10.1128/jvi.79.17.11533-11536.2005
2005
Cited 366 times
A Single Amino Acid Substitution in 1918 Influenza Virus Hemagglutinin Changes Receptor Binding Specificity
The receptor binding specificity of influenza viruses may be important for host restriction of human and avian viruses. Here, we show that the hemagglutinin (HA) of the virus that caused the 1918 influenza pandemic has strain-specific differences in its receptor binding specificity. The A/South Carolina/1/18 HA preferentially binds the alpha2,6 sialic acid (human) cellular receptor, whereas the A/New York/1/18 HA, which differs by only one amino acid, binds both the alpha2,6 and the alpha2,3 sialic acid (avian) cellular receptors. Compared to the conserved consensus sequence in the receptor binding site of avian HAs, only a single amino acid at position 190 was changed in the A/New York/1/18 HA. Mutation of this single amino acid back to the avian consensus resulted in a preference for the avian receptor.
DOI: 10.1371/journal.ppat.1000076
2008
Cited 359 times
The Evolutionary Genetics and Emergence of Avian Influenza Viruses in Wild Birds
We surveyed the genetic diversity among avian influenza virus (AIV) in wild birds, comprising 167 complete viral genomes from 14 bird species sampled in four locations across the United States. These isolates represented 29 type A influenza virus hemagglutinin (HA) and neuraminidase (NA) subtype combinations, with up to 26% of isolates showing evidence of mixed subtype infection. Through a phylogenetic analysis of the largest data set of AIV genomes compiled to date, we were able to document a remarkably high rate of genome reassortment, with no clear pattern of gene segment association and occasional inter-hemisphere gene segment migration and reassortment. From this, we propose that AIV in wild birds forms transient "genome constellations," continually reshuffled by reassortment, in contrast to the spread of a limited number of stable genome constellations that characterizes the evolution of mammalian-adapted influenza A viruses.
DOI: 10.1371/journal.pbio.0030300
2005
Cited 356 times
Whole-Genome Analysis of Human Influenza A Virus Reveals Multiple Persistent Lineages and Reassortment among Recent H3N2 Viruses
Understanding the evolution of influenza A viruses in humans is important for surveillance and vaccine strain selection. We performed a phylogenetic analysis of 156 complete genomes of human H3N2 influenza A viruses collected between 1999 and 2004 from New York State, United States, and observed multiple co-circulating clades with different population frequencies. Strikingly, phylogenies inferred for individual gene segments revealed that multiple reassortment events had occurred among these clades, such that one clade of H3N2 viruses present at least since 2000 had provided the hemagglutinin gene for all those H3N2 viruses sampled after the 2002-2003 influenza season. This reassortment event was the likely progenitor of the antigenically variant influenza strains that caused the A/Fujian/411/2002-like epidemic of the 2003-2004 influenza season. However, despite sharing the same hemagglutinin, these phylogenetically distinct lineages of viruses continue to co-circulate in the same population. These data, derived from the first large-scale analysis of H3N2 viruses, convincingly demonstrate that multiple lineages can co-circulate, persist, and reassort in epidemiologically significant ways, and underscore the importance of genomic analyses for future influenza surveillance.
DOI: 10.1073/pnas.112338099
2002
Cited 354 times
Cellular transcriptional profiling in influenza A virus-infected lung epithelial cells: The role of the nonstructural NS1 protein in the evasion of the host innate defense and its potential contribution to pandemic influenza
The NS1 protein of influenza A virus contributes to viral pathogenesis, primarily by enabling the virus to disarm the host cell type IFN defense system. We examined the downstream effects of NS1 protein expression during influenza A virus infection on global cellular mRNA levels by measuring expression of over 13,000 cellular genes in response to infection with wild-type and mutant viruses in human lung epithelial cells. Influenza A/PR/8/34 virus infection resulted in a significant induction of genes involved in the IFN pathway. Deletion of the viral NS1 gene increased the number and magnitude of expression of cellular genes implicated in the IFN, NF-kappaB, and other antiviral pathways. Interestingly, different IFN-induced genes showed different sensitivities to NS1-mediated inhibition of their expression. A recombinant virus with a C-terminal deletion in its NS1 gene induced an intermediate cellular mRNA expression pattern between wild-type and NS1 knockout viruses. Most significantly, a virus containing the 1918 pandemic NS1 gene was more efficient at blocking the expression of IFN-regulated genes than its parental influenza A/WSN/33 virus. Taken together, our results suggest that the cellular response to influenza A virus infection in human lung cells is significantly influenced by the sequence of the NS1 gene, demonstrating the importance of the NS1 protein in regulating the host cell response triggered by virus infection.
DOI: 10.1056/nejmp0904819
2009
Cited 350 times
The Persistent Legacy of the 1918 Influenza Virus
Descendants of the H1N1 influenza A virus that caused the catastrophic and historic pandemic of 1918–1919 continue to contribute their genes to new viruses, causing new pandemics, epidemics, and epizootics. Drs. David Morens, Jeffery Taubenberger, and Anthony Fauci discuss the influenza A events of the past 91 years.
DOI: 10.1016/j.vaccine.2008.07.025
2008
Cited 316 times
Pathology of human influenza revisited
The pathology of human influenza has been studied most intensively during the three pandemics of the last century, the last of which occurred in 1968. It is important to revisit this subject because of the recent emergence of avian H5N1 influenza in humans as well as the threat of a new pandemic. Uncomplicated human influenza virus infection causes transient tracheo-bronchitis, corresponding with predominant virus attachment to tracheal and bronchial epithelial cells. The main complication is extension of viral infection to the alveoli, often with secondary bacterial infection, resulting in severe pneumonia. Complications in extra-respiratory tissues such as encephalopathy, myocarditis, and myopathy occur occasionally. Sensitive molecular and immunological techniques allow us to investigate whether these complications are a direct result of virus infection or an indirect result of severe pneumonia. Human disease from avian influenza virus infections is most severe for subtype H5N1, but also has been reported for H7 and H9 subtypes. In contrast to human influenza viruses, avian H5N1 virus attaches predominantly to alveolar and bronchiolar epithelium, corresponding with diffuse alveolar damage as the primary lesion. Viremia and extra-respiratory complications appear to be more common for infections with avian H5N1 virus than with human influenza viruses. Further understanding and comparison of the pathology of human and avian influenza virus infections only can be achieved by directed and careful pathological analysis of additional influenza cases.
DOI: 10.1126/science.aai8128
2017
Cited 290 times
IgG antibodies to dengue enhanced for FcγRIIIA binding determine disease severity
Dengue virus (DENV) infection in the presence of reactive, non-neutralizing immunoglobulin G (IgG) (RNNIg) is the greatest risk factor for dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS). Progression to DHF/DSS is attributed to antibody-dependent enhancement (ADE); however, because only a fraction of infections occurring in the presence of RNNIg advance to DHF/DSS, the presence of RNNIg alone cannot account for disease severity. We discovered that DHF/DSS patients respond to infection by producing IgGs with enhanced affinity for the activating Fc receptor FcγRIIIA due to afucosylated Fc glycans and IgG1 subclass. RNNIg enriched for afucosylated IgG1 triggered platelet reduction in vivo and was a significant risk factor for thrombocytopenia. Thus, therapeutics and vaccines restricting production of afucosylated, IgG1 RNNIg during infection may prevent ADE of DENV disease.
DOI: 10.1128/mbio.00417-16
2016
Cited 287 times
Evaluation of Antihemagglutinin and Antineuraminidase Antibodies as Correlates of Protection in an Influenza A/H1N1 Virus Healthy Human Challenge Model
Despite long-term investment, influenza continues to be a significant worldwide problem. The cornerstone of protection remains vaccination, and approved vaccines seek to elicit a hemagglutination inhibition (HAI) titer of ≥1:40 as the primary correlate of protection. However, recent poor vaccine performance raises questions regarding the protection afforded and whether other correlates of protection should be targeted. A healthy volunteer challenge study was performed with a wild-type 2009 A(H1N1)pdm influenza A challenge virus at the NIH Clinical Center to evaluate two groups of participants with HAI titers of ≥1:40 and <1:40. The primary objective was to determine whether participants with HAI titers of ≥1:40 were less likely to develop mild to moderate influenza disease (MMID) after intranasal inoculation. HAI titers of ≥1:40 were protective against MMID but did not reduce the incidence of symptoms alone. Although the baseline HAI titer correlated with some reduction in disease severity measures, overall, the baseline NAI titer correlated more significantly with all disease severity metrics and had a stronger independent effect on outcome. This study demonstrates the importance of examining other immunological correlates of protection rather than solely HAI titers. This challenge study confirms the importance of NAI titer as a correlate and for the first time establishes that it can be an independent predictor of reduction of all aspects of influenza disease. This suggests that NAI titer may play a more significant role than previously thought and that neuraminidase immunity should be considered when studying susceptibility after vaccination and as a critical target in future influenza vaccine platforms.This study represents the first time the current gold standard for evaluating influenza vaccines as set by the U.S. Food and Drug Administration and the European Medicines Agency Committee for Medicinal Products for Human Use, a "protective" hemagglutination inhibition (HAI) titer of ≥1:40, has been evaluated in a well-controlled healthy volunteer challenge study since the cutoff was established. We used our established wild-type influenza A healthy volunteer human challenge model to evaluate how well this antibody titer predicts a reduction in influenza virus-induced disease. We demonstrate that although higher HAI titer is predictive of some protection, there is stronger evidence to suggest that neuraminidase inhibition (NAI) titer is more predictive of protection and reduced disease. This is the first time NAI titer has been clearly identified in a controlled trial of this type to be an independent predictor of a reduction in all aspects of influenza.
DOI: 10.1371/journal.ppat.1000012
2008
Cited 249 times
Multiple Reassortment Events in the Evolutionary History of H1N1 Influenza A Virus Since 1918
The H1N1 subtype of influenza A virus has caused substantial morbidity and mortality in humans, first documented in the global pandemic of 1918 and continuing to the present day. Despite this disease burden, the evolutionary history of the A/H1N1 virus is not well understood, particularly whether there is a virological basis for several notable epidemics of unusual severity in the 1940s and 1950s. Using a data set of 71 representative complete genome sequences sampled between 1918 and 2006, we show that segmental reassortment has played an important role in the genomic evolution of A/H1N1 since 1918. Specifically, we demonstrate that an A/H1N1 isolate from the 1947 epidemic acquired novel PB2 and HA genes through intra-subtype reassortment, which may explain the abrupt antigenic evolution of this virus. Similarly, the 1951 influenza epidemic may also have been associated with reassortant A/H1N1 viruses. Intra-subtype reassortment therefore appears to be a more important process in the evolution and epidemiology of H1N1 influenza A virus than previously realized.
DOI: 10.1038/s41590-020-00828-7
2020
Cited 242 times
Proinflammatory IgG Fc structures in patients with severe COVID-19
Severe acute respiratory syndrome coronavirus 2 infections can cause coronavirus disease 2019 (COVID-19), which manifests with a range of severities from mild illness to life-threatening pneumonia and multi-organ failure. Severe COVID-19 is characterized by an inflammatory signature, including high levels of inflammatory cytokines, alveolar inflammatory infiltrates and vascular microthrombi. Here we show that patients with severe COVID-19 produced a unique serologic signature, including an increased likelihood of IgG1 with afucosylated Fc glycans. This Fc modification on severe acute respiratory syndrome coronavirus 2 IgGs enhanced interactions with the activating Fcγ receptor FcγRIIIa; when incorporated into immune complexes, Fc afucosylation enhanced production of inflammatory cytokines by monocytes, including interleukin-6 and tumor necrosis factor. These results show that disease severity in COVID-19 correlates with the presence of proinflammatory IgG Fc structures, including afucosylated IgG1. COVID-19 is often characterized by a hyperinflammatory syndrome. Wang and colleagues show that low levels of IgG fucosylation enhance interactions with activating Fcγ receptors, boosting the inflammatory cytokines associated with severe COVID-19.
DOI: 10.1177/00333549101250s305
2010
Cited 218 times
Influenza: The Once and Future Pandemic
Influenza A viruses infect large numbers of warm-blooded animals, including wild birds, domestic birds, pigs, horses, and humans. Influenza viruses can switch hosts to form new lineages in novel hosts. The most significant of these events is the emergence of antigenically novel influenza A viruses in humans, leading to pandemics. Influenza pandemics have been reported for at least 500 years, with inter-pandemic intervals averaging approximately 40 years.
DOI: 10.1056/nejmp2002106
2020
Cited 217 times
Escaping Pandora’s Box — Another Novel Coronavirus
Escaping Pandora’s Box — Another Novel Coronavirus With luck, public health control measures may be able to put the Covid-19 demons back in the jar. If they do not, we face a daunting challenge equ...
DOI: 10.1093/cid/cit725
2013
Cited 202 times
The Natural History of Influenza Infection in the Severely Immunocompromised vs Nonimmunocompromised Hosts
Medical advances have led to an increase in the world's population of immunosuppressed individuals. The most severely immunocompromised patients are those who have been diagnosed with a hematologic malignancy, solid organ tumor, or who have other conditions that require immunosuppressive therapies and/or solid organ or stem cell transplants.Medically attended patients with a positive clinical diagnosis of influenza were recruited prospectively and clinically evaluated. Nasal washes and serum were collected. Evaluation of viral shedding, nasal and serum cytokines, clinical illness, and clinical outcomes were performed to compare severely immunocompromised individuals to nonimmunocompromised individuals with influenza infection.Immunocompromised patients with influenza had more severe disease/complications, longer viral shedding, and more antiviral resistance while demonstrating less clinical symptoms and signs on clinical assessment.Immunocompromised patients are at risk for more severe or complicated influenza induced disease, which may be difficult to prevent with existing vaccines and antiviral treatments. Specific issues to consider when managing a severely immunocompromised host include the development of asymptomatic shedding, multi-drug resistance during prolonged antiviral therapy, and the potential high risk of pulmonary involvement.ClinicalTrials.gov identifier NCT00533182.
DOI: 10.1371/journal.ppat.1002998
2012
Cited 199 times
Identification of a Novel Splice Variant Form of the Influenza A Virus M2 Ion Channel with an Antigenically Distinct Ectodomain
Segment 7 of influenza A virus produces up to four mRNAs. Unspliced transcripts encode M1, spliced mRNA2 encodes the M2 ion channel, while protein products from spliced mRNAs 3 and 4 have not previously been identified. The M2 protein plays important roles in virus entry and assembly, and is a target for antiviral drugs and vaccination. Surprisingly, M2 is not essential for virus replication in a laboratory setting, although its loss attenuates the virus. To better understand how IAV might replicate without M2, we studied the reversion mechanism of an M2-null virus. Serial passage of a virus lacking the mRNA2 splice donor site identified a single nucleotide pseudoreverting mutation, which restored growth in cell culture and virulence in mice by upregulating mRNA4 synthesis rather than by reinstating mRNA2 production. We show that mRNA4 encodes a novel M2-related protein (designated M42) with an antigenically distinct ectodomain that can functionally replace M2 despite showing clear differences in intracellular localisation, being largely retained in the Golgi compartment. We also show that the expression of two distinct ion channel proteins is not unique to laboratory-adapted viruses but, most notably, was also a feature of the 1983 North American outbreak of H5N2 highly pathogenic avian influenza virus. In identifying a 14th influenza A polypeptide, our data reinforce the unexpectedly high coding capacity of the viral genome and have implications for virus evolution, as well as for understanding the role of M2 in the virus life cycle.
DOI: 10.1126/scitranslmed.abj7790
2021
Cited 148 times
Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is characterized by respiratory distress, multiorgan dysfunction, and, in some cases, death. The pathological mechanisms underlying COVID-19 respiratory distress and the interplay with aggravating risk factors have not been fully defined. Lung autopsy samples from 18 patients with fatal COVID-19, with symptom onset-to-death times ranging from 3 to 47 days, and antemortem plasma samples from 6 of these cases were evaluated using deep sequencing of SARS-CoV-2 RNA, multiplex plasma protein measurements, and pulmonary gene expression and imaging analyses. Prominent histopathological features in this case series included progressive diffuse alveolar damage with excessive thrombosis and late-onset pulmonary tissue and vascular remodeling. Acute damage at the alveolar-capillary barrier was characterized by the loss of surfactant protein expression with injury to alveolar epithelial cells, endothelial cells, respiratory epithelial basal cells, and defective tissue repair processes. Other key findings included impaired clot fibrinolysis with increased concentrations of plasma and lung plasminogen activator inhibitor-1 and modulation of cellular senescence markers, including p21 and sirtuin-1, in both lung epithelial and endothelial cells. Together, these findings further define the molecular pathological features underlying the pulmonary response to SARS-CoV-2 infection and provide important insights into signaling pathways that may be amenable to therapeutic intervention.
DOI: 10.4269/ajtmh.20-0849
2020
Cited 143 times
The Origin of COVID-19 and Why It Matters
The COVID-19 pandemic is among the deadliest infectious diseases to have emerged in recent history. As with all past pandemics, the specific mechanism of its emergence in humans remains unknown. Nevertheless, a large body of virologic, epidemiologic, veterinary, and ecologic data establishes that the new virus, SARS-CoV-2, evolved directly or indirectly from a β-coronavirus in the sarbecovirus (SARS-like virus) group that naturally infect bats and pangolins in Asia and Southeast Asia. Scientists have warned for decades that such sarbecoviruses are poised to emerge again and again, identified risk factors, and argued for enhanced pandemic prevention and control efforts. Unfortunately, few such preventive actions were taken resulting in the latest coronavirus emergence detected in late 2019 which quickly spread pandemically. The risk of similar coronavirus outbreaks in the future remains high. In addition to controlling the COVID-19 pandemic, we must undertake vigorous scientific, public health, and societal actions, including significantly increased funding for basic and applied research addressing disease emergence, to prevent this tragic history from repeating itself.
DOI: 10.1126/scitranslmed.aau5485
2019
Cited 138 times
The 1918 influenza pandemic: 100 years of questions answered and unanswered
This Review summarizes key findings about the “Spanish” influenza pandemic and addresses implications for current pandemic response and control, including vaccination optimization.
DOI: 10.1128/mbio.00812-20
2020
Cited 133 times
Pandemic COVID-19 Joins History’s Pandemic Legion
With great apprehension, the world is now watching the birth of a novel pandemic already causing tremendous suffering, death, and disruption of normal life. Uncertainty and dread are exacerbated by the belief that what we are experiencing is new and mysterious. However, deadly pandemics and disease emergences are not new phenomena: they have been challenging human existence throughout recorded history. Some have killed sizeable percentages of humanity, but humans have always searched for, and often found, ways of mitigating their deadly effects.
DOI: 10.1056/nejmp2118468
2022
Cited 115 times
Universal Coronavirus Vaccines — An Urgent Need
DOI: 10.1016/j.chom.2022.11.016
2023
Cited 64 times
Rethinking next-generation vaccines for coronaviruses, influenzaviruses, and other respiratory viruses
Viruses that replicate in the human respiratory mucosa without infecting systemically, including influenza A, SARS-CoV-2, endemic coronaviruses, RSV, and many other "common cold" viruses, cause significant mortality and morbidity and are important public health concerns. Because these viruses generally do not elicit complete and durable protective immunity by themselves, they have not to date been effectively controlled by licensed or experimental vaccines. In this review, we examine challenges that have impeded development of effective mucosal respiratory vaccines, emphasizing that all of these viruses replicate extremely rapidly in the surface epithelium and are quickly transmitted to other hosts, within a narrow window of time before adaptive immune responses are fully marshaled. We discuss possible approaches to developing next-generation vaccines against these viruses, in consideration of several variables such as vaccine antigen configuration, dose and adjuventation, route and timing of vaccination, vaccine boosting, adjunctive therapies, and options for public health vaccination polices.
DOI: 10.1073/pnas.031575198
2001
Cited 263 times
Sequence of the 1918 pandemic influenza virus nonstructural gene (NS) segment and characterization of recombinant viruses bearing the 1918 NS genes
The influenza A virus pandemic of 1918-1919 resulted in an estimated 20-40 million deaths worldwide. The hemagglutinin and neuraminidase sequences of the 1918 virus were previously determined. We here report the sequence of the A/Brevig Mission/1/18 (H1N1) virus nonstructural (NS) segment encoding two proteins, NS1 and nuclear export protein. Phylogenetically, these genes appear to be close to the common ancestor of subsequent human and classical swine strain NS genes. Recently, the influenza A virus NS1 protein was shown to be a type I IFN antagonist that plays an important role in viral pathogenesis. By using the recently developed technique of generating influenza A viruses entirely from cloned cDNAs, the hypothesis that the 1918 virus NS1 gene played a role in virulence was tested in a mouse model. In a BSL3+ laboratory, viruses were generated that possessed either the 1918 NS1 gene alone or the entire 1918 NS segment in a background of influenza A/WSN/33 (H1N1), a mouse-adapted virus derived from a human influenza strain first isolated in 1933. These 1918 NS viruses replicated well in tissue culture but were attenuated in mice as compared with the isogenic control viruses. This attenuation in mice may be related to the human origin of the 1918 NS1 gene. These results suggest that interaction of the NS1 protein with host-cell factors plays a significant role in viral pathogenesis.
DOI: 10.1073/pnas.100140097
2000
Cited 222 times
Characterization of the 1918 “Spanish” influenza virus neuraminidase gene
The "Spanish" influenza pandemic of 1918 was characterized by exceptionally high mortality, especially among young adults. The surface proteins of influenza viruses, hemagglutinin and neuraminidase, play important roles in virulence, host specificity, and the human immune response. The complete coding sequence of hemagglutinin was reported last year. This laboratory has now determined the complete coding sequence of the neuraminidase gene of the 1918 virus. Influenza RNA fragments were isolated from lung tissue of three victims of the 1918 flu; complete sequence was generated from A/Brevig Mission/1/18, with confirmatory sequencing carried out on A/South Carolina/1/18 and A/New York/1/18. The 1918 neuraminidase gene sequence was compared with other N1 subtype neuraminidase genes, including 9 N1 strains newly sequenced for this study. The 1918 neuraminidase shares many sequence and structural characteristics with avian strains, including the conserved active site, wild-type stalk length, glycosylation sites, and antigenic sites. Phylogenetically, the 1918 neuraminidase gene appears to be intermediate between mammals and birds, suggesting that it was introduced into mammals just before the 1918 pandemic.
DOI: 10.1099/vir.0.19302-0
2003
Cited 207 times
The origin of the 1918 pandemic influenza virus: a continuing enigma
Influenza A virus is a major public health threat, killing more than 30,000 per year in the USA alone, sickening millions and inflicting substantial economic costs. Novel influenza virus strains emerge periodically to which humans have little immunity, resulting in devastating pandemics. The 1918 pandemic killed nearly 700,000 Americans and 40 million people worldwide. Pandemics in 1957 and 1968, while much less devastating than 1918, also caused tens of thousands of deaths in the USA. The influenza A virus is capable of enormous genetic variability, both by continuous, gradual mutation and by reassortment of gene segments between viruses. Both the 1957 and 1968 pandemic strains are thought to have originated as reassortants, in which one or both human-adapted viral surface proteins were replaced by proteins from avian influenza virus strains. Analyses of the surface proteins of the 1918 pandemic strain, however, suggest that this strain may have had a different origin. The haemagglutinin gene segment of the virus may have come directly from an avian source different from those currently circulating. Alternatively, the virus, or some of its gene segments, may have evolved in an intermediate host before emerging as a human pathogen. Determining whether pandemic influenza virus strains can emerge via different pathways will affect the scope and focus of surveillance and prevention efforts.
DOI: 10.1098/rstb.2001.1020
2001
Cited 192 times
Integrating historical, clinical and molecular genetic data in order to explain the origin and virulence of the 1918 Spanish influenza virus
The Spanish influenza pandemic of 1918-1919 caused acute illness in 25-30% of the world's population and resulted in the death of 40 million people. The complete genomic sequence of the 1918 influenza virus will be deduced using fixed and frozen tissues of 1918 influenza victims. Sequence and phylogenetic analyses of the complete 1918 haemagglutinin (HA) and neuraminidase (NA) genes show them to be the most avian-like of mammalian sequences and support the hypothesis that the pandemic virus contained surface protein-encoding genes derived from an avian influenza strain and that the 1918 virus is very similar to the common ancestor of human and classical swine H1N1 influenza strains. Neither the 1918 HA genes nor the NA genes possessed mutations that are known to increase tissue tropicity, which accounts for the virulence of other influenza strains such as A/WSN/33 or fowl plague viruses. The complete sequence of the nonstructural (NS) gene segment of the 1918 virus was deduced and tested for the hypothesis that the enhanced virulence in 1918 could have been due to type I interferon inhibition by the NS1 protein. The results from these experiments were inconclusive. Sequence analysis of the 1918 pandemic influenza virus is allowing us to test hypotheses as to the origin and virulence of this strain. This information should help to elucidate how pandemic influenza strains emerge and what genetic features contribute to their virulence.
DOI: 10.1038/nrmicro1027
2004
Cited 187 times
Evidence of an absence: the genetic origins of the 1918 pandemic influenza virus
Annual outbreaks of influenza A infection are an ongoing public health threat and novel influenza strains can periodically emerge to which humans have little immunity, resulting in devastating pandemics. The 1918 pandemic killed at least 40 million people worldwide and pandemics in 1957 and 1968 caused hundreds of thousands of deaths. The influenza A virus is capable of enormous genetic variation, both by continuous, gradual mutation and by reassortment of genome segments between viruses. Both the 1957 and 1968 pandemic strains are thought to have originated as reassortants in which one or both human-adapted viral surface proteins were replaced by proteins from avian influenza strains. Analyses of the genes of the 1918 pandemic virus, however, indicate that this strain might have had a different origin. The haemagglutinin and nucleoprotein genome segments in particular are unlikely to have come directly from an avian source that is similar to those that are currently being sequenced. Determining whether a pandemic influenza virus can emerge by different mechanisms will affect the scope and focus of surveillance and prevention efforts.
DOI: 10.1371/journal.ppat.0020125
2006
Cited 182 times
Stochastic Processes Are Key Determinants of Short-Term Evolution in Influenza A Virus
Understanding the evolutionary dynamics of influenza A virus is central to its surveillance and control. While immune-driven antigenic drift is a key determinant of viral evolution across epidemic seasons, the evolutionary processes shaping influenza virus diversity within seasons are less clear. Here we show with a phylogenetic analysis of 413 complete genomes of human H3N2 influenza A viruses collected between 1997 and 2005 from New York State, United States, that genetic diversity is both abundant and largely generated through the seasonal importation of multiple divergent clades of the same subtype. These clades cocirculated within New York State, allowing frequent reassortment and generating genome-wide diversity. However, relatively low levels of positive selection and genetic diversity were observed at amino acid sites considered important in antigenic drift. These results indicate that adaptive evolution occurs only sporadically in influenza A virus; rather, the stochastic processes of viral migration and clade reassortment play a vital role in shaping short-term evolutionary dynamics. Thus, predicting future patterns of influenza virus evolution for vaccine strain selection is inherently complex and requires intensive surveillance, whole-genome sequencing, and phenotypic analysis.
DOI: 10.20506/rst.28.1.1879
2009
Cited 173 times
Pandemic influenza - including a risk assessment of H5N1
Influenza pandemics and epidemics have apparently occurred since at least the Middle Ages. When pandemics appear, 50% or more of an affected population can be infected in a single year, and the number of deaths caused by influenza can dramatically exceed what is normally expected. Since 1500, there appear to have been 13 or more influenza pandemics. In the past 120 years there were undoubted pandemics in 1889, 1918, 1957, 1968, and 1977. Although most experts believe we will face another influenza pandemic, it is impossible to predict when it will appear, where it will originate, or how severe it will be. Nor is there agreement about the subtype of influenza virus most likely to cause the next pandemic. The continuing spread of H5N1 highly pathogenic avian influenza viruses has heightened interest in pandemic prediction. Despite uncertainties in the historical record of the pre-virology era, study of previous pandemics may help guide future pandemic planning and lead to a better understanding of the complex ecobiology underlying the formation of pandemic strains of influenza A viruses.
DOI: 10.1073/pnas.0308391100
2004
Cited 167 times
Pathogenicity and immunogenicity of influenza viruses with genes from the 1918 pandemic virus
The 1918 influenza A H1N1 virus caused the worst pandemic of influenza ever recorded. To better understand the pathogenesis and immunity to the 1918 pandemic virus, we generated recombinant influenza viruses possessing two to five genes of the 1918 influenza virus. Recombinant influenza viruses possessing the hemagglutinin (HA), neuraminidase (NA), matrix (M), nonstructural (NS), and nucleoprotein (NP) genes or any recombinant virus possessing both the HA and NA genes of the 1918 influenza virus were highly lethal for mice. Antigenic analysis by hemagglutination inhibition (HI) tests with ferret and chicken H1N1 antisera demonstrated that the 1918 recombinant viruses antigenically most resembled A/Swine/Iowa/30 (Sw/Iowa/30) virus but differed from H1N1 viruses isolated since 1930. HI and virus neutralizing (VN) antibodies to 1918 recombinant and Sw/Iowa/30 viruses in human sera were present among individuals born before or shortly after the 1918 pandemic. Mice that received an intramuscular immunization of the homologous or Sw/Iowa/30-inactivated vaccine developed HI and VN antibodies to the 1918 recombinant virus and were completely protected against lethal challenge. Mice that received A/PR/8/34, A/Texas/36/91, or A/New Caledonia/20/99 H1N1 vaccines displayed partial protection from lethal challenge. In contrast, control-vaccinated mice were not protected against lethal challenge and displayed high virus titers in respiratory tissues. Partial vaccine protection mediated by baculovirus-expressed recombinant HA vaccines suggest common cross-reactive epitopes on the H1 HA. These data suggest a strategy of vaccination that would be effective against a reemergent 1918 or 1918-like virus.
2006
Cited 156 times
The origin and virulence of the 1918 "Spanish" influenza virus.
The "Spanish" influenza pandemic of 1918-19 caused acute illness in 25-30 percent of the world's population and resulted in the death of up to an estimated 40 million people. Using fixed and frozen lung tissue of 1918 influenza victims, the complete genomic sequence of the 1918 influenza virus has been deduced. Sequence and phylogenetic analysis of the completed 1918 influenza virus genes shows them to be the most avian-like among the mammalian-adapted viruses. This finding supports the hypotheses that (1) the pandemic virus contains genes derived from avian-like influenza virus strains and that (2) the 1918 virus is the common ancestor of human and classical swine H1N1 influenza viruses. The relationship of the 1918 virus with avian influenza viruses is further supported by recent work in which the 1918 hemagglutinin (HA) protein crystal structure was resolved. Neither the 1918 hemagglutinin (HA) nor the neuraminidase (NA) genes possess mutations known to increase tissue tropicity that account for the virulence of other influenza virus strains like A/WSN/33 or the highly pathogenic avian influenza H5 or H7 viruses. Using reverse genetics approaches, influenza virus constructs containing the 1918 HA and NA on a modern human influenza virus background were lethal in mice. The complete 1918 virus was even more virulent in mice. The genotypic basis of this virulence has not yet been elucidated. The complete sequence of the non-structural (NS) gene segment of the 1918 virus was deduced and also tested for the hypothesis that enhanced virulence in 1918 could have been due to type I interferon inhibition by the NS1 protein. Results from these experiments suggest that in human cells the 1918 NS1 is a very effective interferon antagonist, but the 1918 NS1 gene does not have the amino acid change that correlates with virulence in the H5N1 virus strains identified in 1997 in Hong Kong. Sequence analysis of the 1918 pandemic influenza virus is allowing us to test hypotheses as to the origin and virulence of this strain. This information should help elucidate how pandemic influenza virus strains emerge and what genetic features contribute to virulence in humans.
DOI: 10.1128/jvi.78.17.9499-9511.2004
2004
Cited 156 times
Global Host Immune Response: Pathogenesis and Transcriptional Profiling of Type A Influenza Viruses Expressing the Hemagglutinin and Neuraminidase Genes from the 1918 Pandemic Virus
ABSTRACT To understand more fully the molecular events associated with highly virulent or attenuated influenza virus infections, we have studied the effects of expression of the 1918 hemagglutinin (HA) and neuraminidase (NA) genes during viral infection in mice under biosafety level 3 (agricultural) conditions. Using histopathology and cDNA microarrays, we examined the consequences of expression of the HA and NA genes of the 1918 pandemic virus in a recombinant influenza A/WSN/33 virus compared to parental A/WSN/33 virus and to an attenuated virus expressing the HA and NA genes from A/New Caledonia/20/99. The 1918 HA/NA:WSN and WSN recombinant viruses were highly lethal for mice and displayed severe lung pathology in comparison to the nonlethal New Caledonia HA/NA:WSN recombinant virus. Expression microarray analysis performed on lung tissues isolated from the infected animals showed activation of many genes involved in the inflammatory response, including cytokine, apoptosis, and lymphocyte genes that were common to all three infection groups. However, consistent with the histopathology studies, the WSN and 1918 HA/NA:WSN recombinant viruses showed increased up-regulation of genes associated with activated T cells and macrophages, as well as genes involved in apoptosis, tissue injury, and oxidative damage that were not observed in the New Caledonia HA/NA:WSN recombinant virus-infected mice. These studies document clear differences in gene expression profiles that were correlated with pulmonary disease pathology induced by virulent and attenuated influenza virus infections.
DOI: 10.4049/jimmunol.153.3.1225
1994
Cited 153 times
Loss of HLA haplotype and B locus down-regulation in melanoma cell lines.
The expression of HLA class I molecules on tumor cells is vital for CD8+T cell recognition of tumor Ags. Loss of HLA class I Ag expression as a result of defective beta 2-microglobulin genes has been described in melanoma cells. To further evaluate mechanisms of tumor escape, HLA class I Ag expression was compared in 24 metastatic melanoma cell lines and 20 melanocyte strains by FACS analysis with use of allele-specific mAbs. Total loss of HLA class I Ag expression was not noted; instead, two relatively common phenomena were identified: 1) A variable degree of expression of HLA-B Ags by melanoma cell lines and melanocytes; however, HLA-A Ags were consistently expressed in all cell types. Furthermore, HLA-B locus Ag expression was detected in vivo in only one of six frozen section specimens obtained from six patients having metastatic melanoma. 2) Loss of allelic expression was noted in two of 14 HLA-A2 (14%) and one of three HLA-A29 (33%) melanoma cell lines and included a full haplotype, which suggests loss of a genomic fragment. Allele-specific PCR amplification demonstrated deletion of genes in linkage disequilibrium within the MHC class II, III, and I regions. Aberrations of HLA class I expression in tumor lines should be considered when assessing MHC-restricted phenomena in in vitro models.
DOI: 10.1016/j.ajpath.2014.08.030
2015
Cited 147 times
The Role of Viral, Host, and Secondary Bacterial Factors in Influenza Pathogenesis
Influenza A virus infections in humans generally cause self-limited infections, but can result in severe disease, secondary bacterial pneumonias, and death. Influenza viruses can replicate in epithelial cells throughout the respiratory tree and can cause tracheitis, bronchitis, bronchiolitis, diffuse alveolar damage with pulmonary edema and hemorrhage, and interstitial and airspace inflammation. The mechanisms by which influenza infections result in enhanced disease, including development of pneumonia and acute respiratory distress, are multifactorial, involving host, viral, and bacterial factors. Host factors that enhance risk of severe influenza disease include underlying comorbidities, such as cardiac and respiratory disease, immunosuppression, and pregnancy. Viral parameters enhancing disease risk include polymerase mutations associated with host switch and adaptation, viral proteins that modulate immune and antiviral responses, and virulence factors that increase disease severity, which can be especially prominent in pandemic viruses and some zoonotic influenza viruses causing human infections. Influenza viral infections result in damage to the respiratory epithelium that facilitates secondary infection with common bacterial pneumopathogens and can lead to secondary bacterial pneumonias that greatly contribute to respiratory distress, enhanced morbidity, and death. Understanding the molecular mechanisms by which influenza and secondary bacterial infections, coupled with the role of host risk factors, contribute to enhanced morbidity and mortality is essential to develop better therapeutic strategies to treat severe influenza. Influenza A virus infections in humans generally cause self-limited infections, but can result in severe disease, secondary bacterial pneumonias, and death. Influenza viruses can replicate in epithelial cells throughout the respiratory tree and can cause tracheitis, bronchitis, bronchiolitis, diffuse alveolar damage with pulmonary edema and hemorrhage, and interstitial and airspace inflammation. The mechanisms by which influenza infections result in enhanced disease, including development of pneumonia and acute respiratory distress, are multifactorial, involving host, viral, and bacterial factors. Host factors that enhance risk of severe influenza disease include underlying comorbidities, such as cardiac and respiratory disease, immunosuppression, and pregnancy. Viral parameters enhancing disease risk include polymerase mutations associated with host switch and adaptation, viral proteins that modulate immune and antiviral responses, and virulence factors that increase disease severity, which can be especially prominent in pandemic viruses and some zoonotic influenza viruses causing human infections. Influenza viral infections result in damage to the respiratory epithelium that facilitates secondary infection with common bacterial pneumopathogens and can lead to secondary bacterial pneumonias that greatly contribute to respiratory distress, enhanced morbidity, and death. Understanding the molecular mechanisms by which influenza and secondary bacterial infections, coupled with the role of host risk factors, contribute to enhanced morbidity and mortality is essential to develop better therapeutic strategies to treat severe influenza. Influenza viruses are important pathogens from a public health perspective, because they are common causes of human respiratory illness. Significantly, influenza infections are associated with high morbidity and mortality, especially in elderly persons, infants, and those with chronic diseases.1Wright P.F. Neumann G. Kawaoka Y. Orthomyxoviruses.in: Knipe D.M. Howley P.M. Lippincott Williams & Wilkins, Philadelphia2007: 1691-1740Google Scholar Human influenza virus infections are associated with endemically circulating strains causing annual or seasonal epidemic outbreaks (usually in the winter months) and occasional and unpredictably emerging pandemic strains, as well as zoonotic infections from avian and mammalian animal hosts. Epidemic influenza infection results in up to 49,000 deaths and 200,000 hospitalizations each year in the United States alone.2Thompson M.G. Shay D.K. Zhou H. Bridges C.B. Cheng P.Y. Burns E. Bresee J.S. Cox N.J. Estimates of deaths associated with seasonal influenza: United States, 1976-2007.MMWR Morb Mortal Wkly Rep. 2010; 59: 1057-1062PubMed Google Scholar However, pandemic strains of influenza can cause even higher mortality. For example, the 1918 Spanish influenza pandemic resulted in approximately 675,000 deaths in the United States in a population approximately one-third the size of the current population and approximately 50 million deaths globally.3Johnson N.P. Mueller J. Updating the accounts: global mortality of the 1918-1920 “Spanish” influenza pandemic.Bull Hist Med. 2002; 76: 105-115Crossref PubMed Google Scholar, 4Taubenberger J.K. Morens D.M. 1918 Influenza: the mother of all pandemics.Emerg Infect Dis. 2006; 12: 15-22Crossref PubMed Scopus (23) Google Scholar Since 1997, there has been heightened concern that avian influenza viruses associated with zoonotic outbreaks, such as H5N1 and H7N9, could adapt to achieve efficient transmissibility in humans and cause a pandemic.5Peiris J.S.M. Avian influenza viruses in humans.Rev Sci Tech. 2009; 28: 161-173PubMed Google Scholar Influenza virus infections are generally acute, self-limited infections.6Taubenberger J.K. Morens D.M. The pathology of influenza virus infections.Annu Rev Pathol. 2008; 3: 499-522Crossref PubMed Scopus (759) Google Scholar Clinically, influenza manifests as an acute respiratory disease characterized by the sudden onset of high fever, coryza, cough, headache, prostration, malaise, and inflammation of the upper respiratory tree and trachea. Acute symptoms and fever often persist for 7 to 10 days, and in most cases, the infection is self-limited. Generally, pneumonic involvement is not clinically prominent, although weakness and fatigue may linger for weeks. People of all ages are afflicted, but the prevalence is greatest in school-aged children; disease severity is greatest in infants, elderly persons, and those with underlying illnesses. Influenza A viral replication peaks approximately 48 hours after infection of the nasopharynx and declines thereafter, with little virus shed after approximately 6 days. The virus replicates in both the upper and lower respiratory tract. The diagnosis of influenza can be established by viral culture, demonstration of viral antigens, demonstration of viral genetic material (in clinical specimens), or changes in specific antibody titers in serum or respiratory secretions.7Taubenberger J.K. Layne S.P. Diagnosis of influenza virus: coming to grips with the molecular era.Mol Diagn. 2001; 6: 291-305Crossref PubMed Google Scholar Influenza is the leading cause of respiratory viral disease in all hospitalized patients >16 years.8Gaunt E.R. Harvala H. McIntyre C. Templeton K.E. Simmonds P. Disease burden of the most commonly detected respiratory viruses in hospitalized patients calculated using the disability adjusted life year (DALY) model.J Clin Virol. 2011; 52: 215-221Abstract Full Text Full Text PDF PubMed Scopus (66) Google Scholar People with underlying comorbidities, including chronic pulmonary or cardiac disease, immunosuppression, or diabetes mellitus, are at high risk of developing severe complications from influenza A viruses (IAVs). These may include hemorrhagic bronchitis, laryngotracheitis in young children, pneumonia (primary viral or secondary bacterial), and death.6Taubenberger J.K. Morens D.M. The pathology of influenza virus infections.Annu Rev Pathol. 2008; 3: 499-522Crossref PubMed Scopus (759) Google Scholar, 9Kuiken T. Taubenberger J.K. The pathology of human influenza revisited.Vaccine. 2008; 26: D59-D66Crossref PubMed Scopus (263) Google Scholar Obesity has recently been identified as an independent risk factor, and pregnancy has long been associated with increased risk.10Karlsson E.A. Marcelin G. Webby R.J. Schultz-Cherry S. Review on the impact of pregnancy and obesity on influenza virus infection.Influenza Other Respir Viruses. 2012; 6: 449-460Crossref PubMed Scopus (46) Google Scholar, 11Memoli M.J. Harvey H. Morens D.M. Taubenberger J.K. Influenza in pregnancy.Influenza Other Respir Viruses. 2013; 7: 1033-1039Crossref PubMed Scopus (37) Google Scholar Complications of influenza, including hemorrhagic bronchitis, diffuse alveolar damage, and pneumonia, can develop within hours. Fulminant fatal influenza viral pneumonia occasionally occurs, but most pneumonias are caused by secondary bacterial infections.12Chertow D.S. Memoli M.J. Bacterial coinfection in influenza: a grand rounds review.JAMA. 2013; 309: 275-282Crossref PubMed Scopus (286) Google Scholar, 13Morens D.M. Taubenberger J.K. Fauci A.S. Predominant role of bacterial pneumonia as a cause of death in pandemic influenza: implications for pandemic influenza preparedness.J Infect Dis. 2008; 198: 962-970Crossref PubMed Scopus (1149) Google Scholar Development of pneumonia, dyspnea, cyanosis, hemoptysis, pulmonary edema leading to acute respiratory distress syndrome, and death may proceed in as little as 48 hours after the onset of symptoms. Progression to severe disease, including development of acute respiratory distress, pneumonia, and death, is likely a multifactorial process involving viral, host, and bacterial factors (Figure 1). In this review, we will examine each of these underlying factors to discuss their contribution to influenza pathogenesis. Influenza viruses (of the family Orthomyxoviridae) are enveloped, negative-sense, single-stranded RNA viruses with segmented genomes. There are five genera, including Influenzavirus A, Influenzavirus B, Influenzavirus C, Thogotovirus, and Isavirus (infectious salmon anemia virus).1Wright P.F. Neumann G. Kawaoka Y. Orthomyxoviruses.in: Knipe D.M. Howley P.M. Lippincott Williams & Wilkins, Philadelphia2007: 1691-1740Google Scholar, 14Palese P. Shaw M.L. Orthomyxoviridae: The Viruses and Their Replication.in: Knipe D.M. Howley P.M. Lippincott, Williams & Wilkins, Philadelphia2007: 1647-1690Google Scholar, 15Taubenberger J.K. Kash J.C. Influenza virus evolution, host adaptation, and pandemic formation.Cell Host Microbe. 2010; 7: 440-451Abstract Full Text Full Text PDF PubMed Scopus (551) Google Scholar The Influenzavirus genera differ in host range and pathogenicity and diverged evolutionarily at least several thousand years ago.16Suzuki Y. Nei M. Origin and evolution of influenza virus hemagglutinin genes.Mol Biol Evol. 2002; 19: 501-509Crossref PubMed Scopus (86) Google Scholar Influenza A and B viruses have a similar structure, whereas influenza C is more divergent. Influenza A and B type viruses contain eight discrete single-stranded RNA gene segments, each encoding at least one protein. Influenza B and C viruses are predominantly human-adapted viruses, whereas IAVs naturally infect hundreds of warm-blooded animal hosts, including both avian and mammalian species. Wild aquatic birds are the major reservoir of IAV, where it causes predominantly asymptomatic gastrointestinal tract infections.17Webster R.G. Bean W.J. Gorman O.T. Chambers T.M. Kawaoka Y. Evolution and ecology of influenza A viruses.Microbiol Rev. 1992; 56: 152-179Crossref PubMed Google Scholar Mixed IAV infections and gene segment reassortment are common in wild aquatic birds. These data suggest that IAVs in wild birds form transient genome constellations without the strong selective pressure to be maintained as linked genomes, leading to the continual emergence of novel genotypes.18Dugan V.G. Chen R. Spiro D.J. Sengamalay N. Zaborsky J. Ghedin E. Nolting J. Swayne D.E. Runstadler J.A. Happ G.M. Senne D.A. Wang R. Slemons R.D. Holmes E.C. Taubenberger J.K. The evolutionary genetics and emergence of avian influenza viruses in wild birds.PLoS Pathog. 2008; 4: e1000076Crossref PubMed Scopus (315) Google Scholar This genetic and antigenic diversity of IAVs thus poses a significant risk of zoonotic infection, host switch events, and the generation of pandemic IAV strains. IAVs encode at least 13 proteins via alternative open reading frames, splicing, or ribosomal frame shifting.19Wise H.M. Hutchinson E.C. Jagger B.W. Stuart A.D. Kang Z.H. Robb N. Schwartzman L.M. Kash J.C. Fodor E. Firth A.E. Gog J.R. Taubenberger J.K. Digard P. Identification of a novel splice variant form of the influenza A virus M2 ion channel with an antigenically distinct ectodomain.PLoS Pathog. 2012; 8: e1002998Crossref PubMed Scopus (164) Google Scholar IAVs express three surface proteins: hemagglutinin (HA), neuraminidase (NA), and matrix 2. IAVs are classified, or subtyped, by antigenic or genetic characterization of the HA and NA glycoproteins. Eighteen different HA and 11 different NA subtypes are known,18Dugan V.G. Chen R. Spiro D.J. Sengamalay N. Zaborsky J. Ghedin E. Nolting J. Swayne D.E. Runstadler J.A. Happ G.M. Senne D.A. Wang R. Slemons R.D. Holmes E.C. Taubenberger J.K. The evolutionary genetics and emergence of avian influenza viruses in wild birds.PLoS Pathog. 2008; 4: e1000076Crossref PubMed Scopus (315) Google Scholar, 20Wu Y. Tefsen B. Shi Y. Gao G.F. Bat-derived influenza-like viruses H17N10 and H18N11.Trends Microbiol. 2014; 22: 183-191Abstract Full Text Full Text PDF PubMed Scopus (241) Google Scholar with 16 of the HAs and 9 of the NAs consistently found in avian hosts in various combinations (eg, H1N1 or H3N2).18Dugan V.G. Chen R. Spiro D.J. Sengamalay N. Zaborsky J. Ghedin E. Nolting J. Swayne D.E. Runstadler J.A. Happ G.M. Senne D.A. Wang R. Slemons R.D. Holmes E.C. Taubenberger J.K. The evolutionary genetics and emergence of avian influenza viruses in wild birds.PLoS Pathog. 2008; 4: e1000076Crossref PubMed Scopus (315) Google Scholar, 20Wu Y. Tefsen B. Shi Y. Gao G.F. Bat-derived influenza-like viruses H17N10 and H18N11.Trends Microbiol. 2014; 22: 183-191Abstract Full Text Full Text PDF PubMed Scopus (241) Google Scholar HA is a glycoprotein that functions both as the viral receptor-binding protein and as the fusion protein. HA binds to sialic acid (N-acetyl neuraminic acid) bound to underlying sugars on the tips of host cell glycoproteins. IAVs have HAs with varying specificities for the disaccharide consisting of sialic acids (SAs) and the penultimate sugar (galactose or N-acetylgalactosamine) with different glycosidic bonds. IAVs adapted to birds typically have HA receptor binding specificity for α2,3 SA, whereas HAs from IAVs adapted to humans have higher specificity for α2,6 SA.1Wright P.F. Neumann G. Kawaoka Y. Orthomyxoviruses.in: Knipe D.M. Howley P.M. Lippincott Williams & Wilkins, Philadelphia2007: 1691-1740Google Scholar, 14Palese P. Shaw M.L. Orthomyxoviridae: The Viruses and Their Replication.in: Knipe D.M. Howley P.M. Lippincott, Williams & Wilkins, Philadelphia2007: 1647-1690Google Scholar After receptor binding, the virus is internalized. The endosomal compartment's acidic pH leads to an HA conformational change, facilitating fusion of the viral and endosomal membranes, release of viral ribonucleoproteins (RNPs) into the cytoplasm, and their subsequent transport to the nucleus. Viral HA is a homotrimer, and each monomer undergoes proteolytic cleavage to generate HA1 and HA2 polypeptide chains before activation. IAV does not encode a protease and requires exogenous serine proteases (trypsin-like enzymes) for activation that recognize a conserved Q/E-X-R motif found at the HA cleavage site.21Chen J. Lee K.H. Steinhauer D.A. Stevens D.J. Skehel J.J. Wiley D.C. Structure of the hemagglutinin precursor cleavage site, a determinant of influenza pathogenicity and the origin of the labile conformation.Cell. 1998; 95: 409-417Abstract Full Text Full Text PDF PubMed Scopus (401) Google Scholar In humans and other mammals, Clara tryptase, produced by cells of the bronchiolar epithelium, likely serves this role. IAVs of H5 and H7 subtypes can acquire insertional mutations at the HA cleavage site, which change their protease recognition site to a furin-like recognition sequence R-X-R/K-R. This polybasic cleavage site broadens protease specificity, allowing for intracellular cleavage activation, and systemic replication of such viruses in poultry, resulting in the emergence of highly pathogenic avian influenza. NA is a glycoprotein with neuraminidase (sialidase) enzymatic activity required for cleavage of these host cell SAs, allowing newly produced virions to be released and to cleave SAs from viral glycoproteins to prevent aggregation of nascent viral particles. The surface glycoproteins HA and NA are the major antigenic targets of the humoral immune response to IAV, and NA is the target of the antiviral drugs oseltamivir and zanamivir. The matrix 1 protein is the most abundant structural protein. Localized beneath the viral membrane, it interacts with the cytoplasmic domains of the surface glycoproteins HA and NA and also with the viral RNP complexes. The small protein matrix 2 is a proton channel necessary for viral replication and is the target of the adamantane class of antiviral drugs.22Hayden F.G. Antivirals for influenza: historical perspectives and lessons learned.Antiviral Res. 2006; 71: 372-378Crossref PubMed Scopus (83) Google Scholar Matrix 2 functions as a low pH gated ion channel that lowers the pH of the virion after internalization and leads to membrane fusion with the lysogenic vacuole. The viral RNPs are released into the cytoplasm and are then translocated to the nucleus to initiate viral RNA synthesis. IAVs are segmented, negative-stranded viruses, and viral RNPs consist of each RNA gene segment, encapsidated by the single-stranded RNA binding protein nucleoprotein (NP) and associated with three viral polymerase proteins that comprise the RNA-dependent RNA polymerase-polymerase basic protein 2 (PB2), polymerase basic protein 1 (PB1), and polymerase acidic protein (PA). The polymerase proteins form a heterotrimer bound to short hairpin structures formed by the complementary terminal 5′ and 3′ untranslated regions of each RNA segment. PB1 is the RNA-dependent RNA polymerase, and PB2 functions in mRNA synthesis by binding host mRNA caps. Although PA is necessary for a functional polymerase complex, including endonucleolytic cleavage of host pre-mRNAs, its biological roles remain less well understood. NP plays important roles in transcription, and in the trafficking of RNPs between the cytoplasm and nucleus. IAV is dependent on the RNA processing machinery of the host cell, and transcription and replication occur in the host nucleus. Several non-structural proteins play important roles in the viral replicative cycle. The non-structural protein 1 has a variety of functions, including double-stranded RNA binding and antagonism of host cell type I interferon responses.14Palese P. Shaw M.L. Orthomyxoviridae: The Viruses and Their Replication.in: Knipe D.M. Howley P.M. Lippincott, Williams & Wilkins, Philadelphia2007: 1647-1690Google Scholar The NS2 (or NEP) protein enables nuclear export of viral RNP complexes. PB1-F2 targets the mitochondrial inner membrane and may play a role in apoptosis during IAV infection.23Conenello G.M. Palese P. Influenza A virus PB1-F2: a small protein with a big punch.Cell Host Microbe. 2007; 2: 207-209Abstract Full Text Full Text PDF PubMed Scopus (55) Google Scholar PAX is a newly discovered protein that functions to repress cellular gene expression and modulates IAV pathogenicity by an unknown mechanism.24Jagger B.W. Wise H.M. Kash J.C. Walters K.A. Wills N.M. Xiao Y.L. Dunfee R.L. Schwartzman L.M. Ozinsky A. Bell G.L. Dalton R.M. Lo A. Efstathiou S. Atkins J.F. Firth A.E. Taubenberger J.K. Digard P. An overlapping protein-coding region in influenza A virus segment 3 modulates the host response.Science. 2012; 337: 199-204Crossref PubMed Scopus (455) Google Scholar The viral RNA-dependent RNA polymerase lacks proofreading, and IAVs are thus evolutionarily dynamic viruses with high mutation rates that range from approximately 1 × 10−3 to 8 × 10−3 substitutions per site per year.25Chen R. Holmes E.C. Avian influenza virus exhibits rapid evolutionary dynamics.Mol Biol Evol. 2006; 23: 2336-2341Crossref PubMed Scopus (176) Google Scholar Mutations that alter amino acids in the antigenic portions of the surface glycoproteins HA and NA may allow IAVs to evade preexisting immunity. These mutations are especially important in human seasonal IAV strains, which are subjected to strong population immunological pressures. Anti-HA antibodies can prevent receptor binding, can neutralize infection, and are effective at preventing reinfection with the same strain. This selective mutation in the antigenic domains of HA and NA has been termed antigenic drift26Murphy B.R. Clements M.L. The systemic and mucosal immune response of humans to influenza A virus.Curr Top Microbiol Immunol. 1989; 146: 107-116PubMed Google Scholar and is the basis for the need to yearly update the annual influenza vaccine formulation.1Wright P.F. Neumann G. Kawaoka Y. Orthomyxoviruses.in: Knipe D.M. Howley P.M. Lippincott Williams & Wilkins, Philadelphia2007: 1691-1740Google Scholar The high mutation rate can also result in the rapid establishment of antiviral drug-resistant populations, including resistance to NA inhibitors and adamantanes. Clinical studies have shown that NA-resistant viruses can rapidly acquire mutations in NA after initiation of treatment,27Memoli M.J. Hrabal R.J. Hassantoufighi A. Eichelberger M.C. Taubenberger J.K. Rapid selection of oseltamivir- and peramivir-resistant pandemic H1N1 virus during therapy in 2 immunocompromised hosts.Clin Infect Dis. 2010; 50: 1252-1255Crossref PubMed Scopus (139) Google Scholar, 28Memoli M.J. Hrabal R.J. Hassantoufighi A. Jagger B.W. Sheng Z.M. Eichelberger M.C. Taubenberger J.K. Rapid selection of a transmissible multidrug-resistant influenza A/H3N2 virus in an immunocompromised host.J Infect Dis. 2010; 201: 1397-1403Crossref PubMed Scopus (40) Google Scholar without apparent loss of fitness. Global circulation of such human IAVs bearing antiviral resistance mutations has been observed for different strains of influenza viruses in the past decade.29Samson M. Pizzorno A. Abed Y. Boivin G. Influenza virus resistance to neuraminidase inhibitors.Antiviral Res. 2013; 98: 174-185Crossref PubMed Scopus (270) Google Scholar Because the IAV genome consists of eight RNA segments, coinfection of one host cell with two different IAVs can result in progeny viruses containing gene segments derived from both parental viruses. When the process of genetic reassortment involves the gene segments encoding the HA and/or NA genes, it is termed antigenic shift. Reassortment is an important feature in IAV evolution18Dugan V.G. Chen R. Spiro D.J. Sengamalay N. Zaborsky J. Ghedin E. Nolting J. Swayne D.E. Runstadler J.A. Happ G.M. Senne D.A. Wang R. Slemons R.D. Holmes E.C. Taubenberger J.K. The evolutionary genetics and emergence of avian influenza viruses in wild birds.PLoS Pathog. 2008; 4: e1000076Crossref PubMed Scopus (315) Google Scholar, 30Rambaut A. Pybus O.G. Nelson M.I. Viboud C. Taubenberger J.K. Holmes E.C. The genomic and epidemiological dynamics of human influenza A virus.Nature. 2008; 453: 615-619Crossref PubMed Scopus (715) Google Scholar and host switch.15Taubenberger J.K. Kash J.C. Influenza virus evolution, host adaptation, and pandemic formation.Cell Host Microbe. 2010; 7: 440-451Abstract Full Text Full Text PDF PubMed Scopus (551) Google Scholar Considering viral factors specifically, the molecular basis of virulence is also usually polygenic. Because IAV strains can move between hosts to cause either self-limited spillover infections or stably adapt to new hosts, viral pathogenesis varies by host and must be considered when examining viral virulence factors. The processes by which IAV switch hosts, particularly those changes associated with adaptation of avian IAV to mammalian and ultimately human hosts, have been extensively studied, but are still only partially understood.15Taubenberger J.K. Kash J.C. Influenza virus evolution, host adaptation, and pandemic formation.Cell Host Microbe. 2010; 7: 440-451Abstract Full Text Full Text PDF PubMed Scopus (551) Google Scholar Ultimately, influenza viruses must be able to infect target cells, replicate, and be transmitted efficiently to be adapted to a particular host. However, influenza pathogenesis does not require transmissibility, and zoonotic infections of nonadapted, or partially host-adapted, viruses can cause severe disease, such as observed in human infections with avian H5N1 and H7N9 viruses.5Peiris J.S.M. Avian influenza viruses in humans.Rev Sci Tech. 2009; 28: 161-173PubMed Google Scholar, 31Morens D.M. Taubenberger J.K. Fauci A.S. H7N9 avian influenza A virus and the perpetual challenge of potential human pandemicity.MBio. 2013; 4 (e00445–13)Crossref Scopus (51) Google Scholar It is likely that viral adaptive mutations enhancing replication, pathogenicity, and transmissibility in a particular host are polygenic traits driven by mutational pressures that are independent and possibly even competing.15Taubenberger J.K. Kash J.C. Influenza virus evolution, host adaptation, and pandemic formation.Cell Host Microbe. 2010; 7: 440-451Abstract Full Text Full Text PDF PubMed Scopus (551) Google Scholar, 32Herfst S. Schrauwen E.J. Linster M. Chutinimitkul S. de Wit E. Munster V.J. Sorrell E.M. Bestebroer T.M. Burke D.F. Smith D.J. Rimmelzwaan G.F. Osterhaus A.D. Fouchier R.A. Airborne transmission of influenza A/H5N1 virus between ferrets.Science. 2012; 336: 1534-1541Crossref PubMed Scopus (1190) Google Scholar, 33Imai M. Watanabe T. Hatta M. Das S.C. Ozawa M. Shinya K. Zhong G. Hanson A. Katsura H. Watanabe S. Li C. Kawakami E. Yamada S. Kiso M. Suzuki Y. Maher E.A. Neumann G. Kawaoka Y. Experimental adaptation of an influenza H5 HA confers respiratory droplet transmission to a reassortant H5 HA/H1N1 virus in ferrets.Nature. 2012; 486: 420-428Crossref PubMed Scopus (1094) Google Scholar In seasonal influenza, rapid mutation of the antigenic regions of the HA results in antigenic drift and the necessity to continually reformulate the vaccine strains selected for inclusion. Emergence of antigenically drifted variants has been associated with more severe annual epidemics (eg, the 1947 H1N1 epidemic) and more recent H3N2 variants in 1997 and 2003 to 2004.30Rambaut A. Pybus O.G. Nelson M.I. Viboud C. Taubenberger J.K. Holmes E.C. The genomic and epidemiological dynamics of human influenza A virus.Nature. 2008; 453: 615-619Crossref PubMed Scopus (715) Google Scholar, 34Kilbourne E.D. Smith C. Brett I. Pokorny B.A. Johansson B. Cox N. The total influenza vaccine failure of 1947 revisited: major intrasubtypic antigenic change can explain failure of vaccine in a post-World War II epidemic.Proc Natl Acad Sci U S A. 2002; 99: 10748-10752Crossref PubMed Scopus (107) Google Scholar, 35Holmes E.C. Ghedin E. Miller N. Taylor J. Bao Y. St George K. Grenfell B.T. Salzberg S.L. Fraser C.M. Lipman D.J. Taubenberger J.K. Whole-genome analysis of human influenza A virus reveals multiple persistent lineages and reassortment among recent H3N2 viruses.PLoS Biol. 2005; 3: e300Crossref PubMed Scopus (305) Google Scholar Lack of protective immunity to pandemic IAV with antigenically novel HAs is also a likely factor contributing to enhanced morbidity and mortality in pandemics.36Morens D.M. Taubenberger J.K. Fauci A.S. The persistent legacy of the 1918 influenza virus.N Engl J Med. 2009; 361: 225-229Crossref PubMed Scopus (280) Google Scholar, 37Simonsen L. Clarke M.J. Schonberger L.B. Arden N.H. Cox N.J. Fukuda K. Pandemic versus epidemic influenza mortality: a pattern of changing age distribution.J Infect Dis. 1998; 178: 53-60Crossref PubMed Scopus (564) Google Scholar Certain H5 or H7 strains of IAV can develop an insertional polybasic mutation at the HA cleavage site in poultry to become highly pathogenic avian influenza. These viruses have enhanced pathogenicity because HA activation can use intracellular furin-like proteases, allowing virus to replicate systemically.1Wright P.F. Neumann G. Kawaoka Y. Orthomyxoviruses.in: Knipe D.M. Howley P.M. Lippincott Williams & Wilkins, Philadelphia2007: 1691-1740Google Scholar These viruses typically cause severe illness and high mortality in gallinaceous poultry, such as chickens and turkeys, but may be nonpathogenic in ducks, further supporting the species specificity of viral pathogenesis.38Sturm-Ramirez K.M. Hulse-Post D.J. Govorkova E.A. Humberd J. Seiler P. Puthavathana P. Buranathai C. Nguyen T.D. Chaisingh A. Long H.T. Naipospos T.S. Chen H. Ellis T.M. Guan Y. Peiris J.S. Webster R.G. Are ducks contributing to the endemicity of highly pathogenic H5N1 influenza virus in Asia?.J Virol. 2005; 79: 11269-11279Crossref PubMed Scopus (393) Google Scholar Human zoonotic infections with highly pathogenic avian influenza H5N1 viruses have been documented in 650 patients in 16 countries since 2003, with 386 deaths (59% case fatality rate; World Health Organization, http://www.who.int/influenza/human_animal_interface/EN_GIP_20140124CumulativeNumberH5N1cases.pdf?ua=1, last accessed August 13, 2014). The HA proteins encoded by the last four pandemic viruses (1918 H1N1, 1957 H2N2, 1968 H3N2, and 2009 H1N1) cause enhanced disease in mice compared to seasonal influenza H1N1 or H3N2 subtypes when expressed on a chimeric IAV in which the other seven genes were derived from a seasonal H1N1 human virus. These pandemic HA-expressing viruses had an expanded cellular tropism to infect alveolar epithelial cells and macrophages, and this was correlated with an inability of these zoonotically derived HAs to be neutralized by lung surfactant protein D in v
DOI: 10.1097/ccm.0b013e3181ceb25b
2010
Cited 145 times
The 1918 influenza pandemic: Lessons for 2009 and the future
The 1918 to 1919 H1N1 influenza pandemic is among the most deadly events in recorded human history, having killed an estimated 50 to 100 million persons. Recent H5N1 avian influenza epizootics associated with sporadic human fatalities have heightened concern that a new influenza pandemic, one at least as lethal as that of 1918, could be developing. In early 2009, a novel pandemic H1N1 influenza virus appeared, but it has not exhibited unusually high pathogenicity. Nevertheless, because this virus spreads globally, some scientists predict that mutations will increase its lethality. Therefore, to accurately predict, plan, and respond to current and future influenza pandemics, we must first better-understand the events and experiences of 1918. Although the entire genome of the 1918 influenza virus has been sequenced, many questions about the pandemic it caused remain unanswered. In this review, we discuss the origin of the 1918 pandemic influenza virus, the pandemic's unusual epidemiologic features and the causes and demographic patterns of fatality, and how this information should impact our response to the current 2009 H1N1 pandemic and future pandemics. After 92 yrs of research, fundamental questions about influenza pandemics remain unanswered. Thus, we must remain vigilant and use the knowledge we have gained from 1918 and other influenza pandemics to direct targeted research and pandemic influenza preparedness planning, emphasizing prevention, containment, and treatment.
DOI: 10.1086/651605
2010
Cited 143 times
Rapid Selection of Oseltamivir‐ and Peramivir‐Resistant Pandemic H1N1 Virus during Therapy in 2 Immunocompromised Hosts
Pandemic 2009 H1N1 virus isolates containing the neuraminidase inhibitor resistance mutation H275Y have been reported. We describe rapid selection for the H275Y resistance mutation during therapy in 2 immunocompromised individuals at 9 and 14 days of therapy, as well as the first described case of clinically significant resistance to peramivir.
DOI: 10.1093/cid/ciu924
2014
Cited 141 times
Validation of the Wild-type Influenza A Human Challenge Model H1N1pdMIST: An A(H1N1)pdm09 Dose-Finding Investigational New Drug Study
Healthy volunteer wild-type influenza challenge models offer a unique opportunity to evaluate multiple aspects of this important virus. Such studies have not been performed in the United States in more than a decade, limiting our capability to investigate this virus and develop countermeasures. We have completed the first ever wild-type influenza A challenge study under an Investigational New Drug application (IND). This dose-finding study will lead to further development of this model both for A(H1N1)pdm09 and other strains of influenza.Volunteers were admitted to an isolation unit at the National Institutes of Health Clinical Center for a minimum of 9 days. A reverse genetics, cell-based, Good Manufacturing Practice (GMP)-produced, wild-type A(H1N1)pdm09 virus was administered intranasally. Escalating doses were given until a dose was reached that produced disease in a minimum of 60% of volunteers.An optimal dose of 10(7) tissue culture infectious dose 50 was reached that caused mild to moderate influenza disease in 69% of individuals with mean viral shedding for 4-5 days and significant rises in convalescent influenza antibody titers. Viral shedding preceded symptoms by 12-24 hours and terminated 2-3 days prior to symptom resolution, indicating that individuals may be infectious before symptom development. As expected, nasal congestion and rhinorrhea were most common, but interestingly, fever was observed in only 10% of individuals.This study represents the first healthy volunteer influenza challenge model using a GMP-produced wild-type virus under an IND. This unique clinical research program will facilitate future studies of influenza pathogenesis, animal model validation, and the rapid, efficient, and cost-effective evaluation of efficacy of novel vaccines and therapeutics. Clinical Trials Registration.NCT01646138.
DOI: 10.1038/nbt.1593
2009
Cited 135 times
Synthetic viruses: a new opportunity to understand and prevent viral disease
Rapid progress in DNA synthesis and sequencing is spearheading the deliberate, large-scale genetic alteration of organisms. These new advances in DNA manipulation have been extended to the level of whole-genome synthesis, as evident from the synthesis of poliovirus, from the resurrection of the extinct 1918 strain of influenza virus and of human endogenous retroviruses and from the restructuring of the phage T7 genome. The largest DNA synthesized so far is the 582,970 base pair genome of Mycoplasma genitalium, although, as yet, this synthetic DNA has not been 'booted' to life. As genome synthesis is independent of a natural template, it allows modification of the structure and function of a virus's genetic information to an extent that was hitherto impossible. The common goal of this new strategy is to further our understanding of an organism's properties, particularly its pathogenic armory if it causes disease in humans, and to make use of this new information to protect from, or treat, human viral disease. Although only a few applications of virus synthesis have been described as yet, key recent findings have been the resurrection of the 1918 influenza virus and the generation of codon- and codon pair-deoptimized polioviruses.
DOI: 10.1073/pnas.1111179108
2011
Cited 134 times
Autopsy series of 68 cases dying before and during the 1918 influenza pandemic peak
The 1918 to 1919 “Spanish” influenza pandemic virus killed up to 50 million people. We report here clinical, pathological, bacteriological, and virological findings in 68 fatal American influenza/pneumonia military patients dying between May and October of 1918, a period that includes ∼4 mo before the 1918 pandemic was recognized, and 2 mo (September–October 1918) during which it appeared and peaked. The lung tissues of 37 of these cases were positive for influenza viral antigens or viral RNA, including four from the prepandemic period (May–August). The prepandemic and pandemic peak cases were indistinguishable clinically and pathologically. All 68 cases had histological evidence of bacterial pneumonia, and 94% showed abundant bacteria on Gram stain. Sequence analysis of the viral hemagglutinin receptor-binding domain performed on RNA from 13 cases suggested a trend from a more “avian-like” viral receptor specificity with G222 in prepandemic cases to a more “human-like” specificity associated with D222 in pandemic peak cases. Viral antigen distribution in the respiratory tree, however, was not apparently different between prepandemic and pandemic peak cases, or between infections with viruses bearing different receptor-binding polymorphisms. The 1918 pandemic virus was circulating for at least 4 mo in the United States before it was recognized epidemiologically in September 1918. The causes of the unusually high mortality in the 1918 pandemic were not explained by the pathological and virological parameters examined. These findings have important implications for understanding the origins and evolution of pandemic influenza viruses.
DOI: 10.1128/jvi.01203-13
2013
Cited 133 times
Molecular Basis for Broad Neuraminidase Immunity: Conserved Epitopes in Seasonal and Pandemic H1N1 as Well as H5N1 Influenza Viruses
Influenza A viruses, including H1N1 and H5N1 subtypes, pose a serious threat to public health. Neuraminidase (NA)-related immunity contributes to protection against influenza virus infection. Antibodies to the N1 subtype provide protection against homologous and heterologous H1N1 as well as H5N1 virus challenge. Since neither the strain-specific nor conserved epitopes of N1 have been identified, we generated a panel of mouse monoclonal antibodies (MAbs) that exhibit different reactivity spectra with H1N1 and H5N1 viruses and used these MAbs to map N1 antigenic domains. We identified 12 amino acids essential for MAb binding to the NA of a recent seasonal H1N1 virus, A/Brisbane/59/2007. Of these, residues 248, 249, 250, 341, and 343 are recognized by strain-specific group A MAbs, while residues 273, 338, and 339 are within conserved epitope(s), which allows cross-reactive group B MAbs to bind the NAs of seasonal H1N1 and the 1918 and 2009 pandemic (09pdm) H1N1 as well as H5N1 viruses. A single dose of group B MAbs administered prophylactically fully protected mice against lethal challenge with seasonal and 09pdm H1N1 viruses and resulted in significant protection against the highly pathogenic wild-type H5N1 virus. Another three N1 residues (at positions 396, 397, and 456) are essential for binding of cross-reactive group E MAbs, which differ from group B MAbs in that they do not bind 09pdm H1N1 viruses. The identification of conserved N1 epitopes reveals the molecular basis for NA-mediated immunity between H1N1 and H5N1 viruses and demonstrates the potential for developing broadly protective NA-specific antibody treatments for influenza.
DOI: 10.1128/jvi.02565-08
2009
Cited 123 times
Different Evolutionary Trajectories of European Avian-Like and Classical Swine H1N1 Influenza A Viruses
In 1979, a lineage of avian-like H1N1 influenza A viruses emerged in European swine populations independently from the classical swine H1N1 virus lineage that had circulated in pigs since the Spanish influenza pandemic of 1918. To determine whether these two distinct lineages of swine-adapted A/H1N1 viruses evolved from avian-like A/H1N1 ancestors in similar ways, as might be expected given their common host species and origin, we compared patterns of nucleotide and amino acid change in whole genome sequences of both groups. An analysis of nucleotide compositional bias across all eight genomic segments for the two swine lineages showed a clear lineage-specific bias, although a segment-specific effect was also apparent. As such, there appears to be only a relatively weak host-specific selection pressure. Strikingly, despite each lineage evolving in the same species of host for decades, amino acid analysis revealed little evidence of either parallel or convergent changes. These findings suggest that although adaptation due to evolutionary lineages can be distinguished, there are functional and structural constraints on all gene segments and that the evolutionary trajectory of each lineage of swine A/H1N1 virus has a strong historical contingency. Thus, in the context of emergence of an influenza A virus strain via a host switch event, it is difficult to predict what specific polygenic changes are needed for mammalian adaptation.
DOI: 10.1128/jvi.06203-11
2012
Cited 115 times
Reassortment and Mutation of the Avian Influenza Virus Polymerase PA Subunit Overcome Species Barriers
The emergence of new pandemic influenza A viruses requires overcoming barriers to cross-species transmission as viruses move from animal reservoirs into humans. This complicated process is driven by both individual gene mutations and genome reassortments. The viral polymerase complex, composed of the proteins PB1, PB2, and PA, is a major factor controlling host adaptation, and reassortment events involving polymerase gene segments occurred with past pandemic viruses. Here we investigate the ability of polymerase reassortment to restore the activity of an avian influenza virus polymerase that is normally impaired in human cells. Our data show that the substitution of human-origin PA subunits into an avian influenza virus polymerase alleviates restriction in human cells and increases polymerase activity in vitro. Reassortants with 2009 pandemic H1N1 PA proteins were the most active. Mutational analyses demonstrated that the majority of the enhancing activity in human PA results from a threonine-to-serine change at residue 552. Reassortant viruses with avian polymerases and human PA subunits, or simply the T552S mutation, displayed faster replication kinetics in culture and increased pathogenicity in mice compared to those containing a wholly avian polymerase complex. Thus, the acquisition of a human PA subunit, or the signature T552S mutation, is a potential mechanism to overcome the species-specific restriction of avian polymerases and increase virus replication. Our data suggest that the human, avian, swine, and 2009 H1N1-like viruses that are currently cocirculating in pig populations set the stage for PA reassortments with the potential to generate novel viruses that could possess expanded tropism and enhanced pathogenicity.
DOI: 10.1128/jvi.01677-12
2012
Cited 109 times
Evolutionary Conservation of the PA-X Open Reading Frame in Segment 3 of Influenza A Virus
ABSTRACT PA-X is a fusion protein of influenza A virus encoded in part from a +1 frameshifted X open reading frame (X-ORF) in segment 3. We show that the X-ORFs of diverse influenza A viruses can be divided into two groups that differ in selection pressure and likely function, reflected in the presence of an internal stop codon and a change in synonymous diversity. Notably, truncated forms of PA-X evolved convergently in swine and dogs, suggesting a strong species-specific effect.
DOI: 10.1111/irv.12055
2012
Cited 100 times
Influenza in pregnancy
Please cite this paper as: Memoli et al. (2012) Influenza in pregnancy. Influenza and Other Respiratory Viruses 00(00), 000–000. The 2009 pandemic served as a strong reminder that influenza‐induced disease can have a great impact on certain at‐risk populations and that pregnant women are one such important population. The increased risk of fatal and severe disease in these women was appreciated more than 500 years ago, and during the last century, pregnant women and their newborns have continued to be greatly affected by both seasonal and pandemic influenza. In this review, we briefly discuss the data collected both before and after the 2009 pandemic as it relates to the impact of influenza on pregnant women and their fetuses/newborns, as well as risk variables, clinical features, clues to pathophysiologic mechanisms, and approaches to treatment and prevention.
DOI: 10.1369/0022155414531549
2014
Cited 98 times
Characterizing and Diminishing Autofluorescence in Formalin-fixed Paraffin-embedded Human Respiratory Tissue
Tissue autofluorescence frequently hampers visualization of immunofluorescent markers in formalin-fixed paraffin-embedded respiratory tissues. We assessed nine treatments reported to have efficacy in reducing autofluorescence in other tissue types. The three most efficacious were Eriochrome black T, Sudan black B and sodium borohydride, as measured using white light laser confocal Λ2 (multi-lambda) analysis. We also assessed the impact of steam antigen retrieval and serum application on human tracheal tissue autofluorescence. Functionally fitting this Λ2 data to 2-dimensional Gaussian surfaces revealed that steam antigen retrieval and serum application contribute minimally to autofluorescence and that the three treatments are disparately efficacious. Together, these studies provide a set of guidelines for diminishing autofluorescence in formalin-fixed paraffin-embedded human respiratory tissue. Additionally, these characterization techniques are transferable to similar questions in other tissue types, as demonstrated on frozen human liver tissue and paraffin-embedded mouse lung tissue fixed in different fixatives.
DOI: 10.1111/his.14201
2020
Cited 94 times
Insights into pathogenesis of fatal COVID‐19 pneumonia from histopathology with immunohistochemical and viral RNA studies
We describe post-mortem pulmonary histopathologic findings of COVID-19 pneumonia in patients with a spectrum of disease course, from rapid demise to prolonged hospitalisation.Histopathologic findings in post-mortem lung tissue from eight patients who died from COVID-19 pneumonia were reviewed. Immunohistochemistry (IHC) and next-generation sequencing (NGS) were performed to detect virus. Diffuse alveolar damage (DAD) was seen in all cases with a spectrum of acute phase and/or organising phase. IHC with monoclonal antibodies against SARS-CoV-2 viral nucleoprotein and spike protein detected virus in areas of acute but not organising DAD, with intracellular viral antigen and RNA expression seen predominantly in patients with duration of illness less than 10 days. Major vascular findings included thrombi in medium- and large-calibre vessels, platelet microthrombi detected by CD61 IHC and fibrin microthrombi.Presence of SARS-CoV-2 viral RNA by NGS early in the disease course and expression of viral antigen by IHC exclusively in the acute, but not in the organising phase of DAD, suggests that the virus may play a major role in initiating the acute lung injury of DAD, but when DAD progresses to the organising phase the virus may have been cleared from the lung by the patient's immune response. These findings suggest the possibility of a major change during the disease course of COVID-19 pneumonia that may have therapeutic implications. Frequent thrombi and microthrombi may also present potential targets for therapeutic intervention.
DOI: 10.1126/scitranslmed.abb1469
2020
Cited 84 times
New coronavirus outbreak: Framing questions for pandemic prevention
We need to understand and quantify the dominant variables that govern the SARS-CoV-2 outbreak, rather than relying exclusively on confirmed cases and their geospatial spread.
DOI: 10.1128/mbio.02284-17
2018
Cited 79 times
Evaluation of Preexisting Anti-Hemagglutinin Stalk Antibody as a Correlate of Protection in a Healthy Volunteer Challenge with Influenza A/H1N1pdm Virus
ABSTRACT Influenza virus hemagglutinin (HA) surface glycoprotein is currently the primary target of licensed influenza vaccines. Recently, broadly reactive antibodies that target the stalk region of the HA have become a major focus of current novel vaccine development. These antibodies have been observed in humans after natural infection with influenza A virus, but the data are limited. Using samples and data from the uniquely controlled setting of an influenza A/H1N1 virus human challenge study of healthy volunteers, we performed a secondary analysis that for the first time explores the role of anti-HA stalk antibody as a human correlate of protection. An anti-HA stalk antibody enzyme-linked immunosorbent assay (ELISA) was performed on samples from 65 participants challenged with a 2009 H1N1pdm virus. Pre- and postchallenge anti-HA stalk titers were then correlated with multiple outcome measures to evaluate anti-HA stalk antibody titer as a correlate of protection. Anti-HA stalk antibody titers were present before challenge and rose in response to challenge in 64% of individuals. Those individuals with higher titers at baseline were less likely to develop shedding, but not less likely to develop symptoms. Similar to the hemagglutination inhibition (HAI) titer, the baseline anti-HA stalk antibody titer did not independently predict a decrease in the severity of influenza disease, while the antineuraminidase (neuraminidase inhibition [NAI]) titer did. As a correlate of protection, the naturally occurring anti-HA stalk antibody titer is predictive of a reduction of certain aspects of disease similar to HAI titer, but the NAI titer is the only identified correlate that is an independent predictor of a reduction of all assessed influenza clinical outcome measures. IMPORTANCE This is the first study to evaluate preexisting anti-HA stalk antibodies as a predictor of protection. We use a healthy volunteer influenza challenge trial for an examination of the role such antibodies play in protection. This study demonstrates that anti-HA stalk antibodies are naturally generated in response to an infection, but there is significant variability in response. Similar to antibodies that target the HA head, baseline anti-HA stalk antibody titer is a correlate of protection in terms of reduced shedding, but it is not a predictor of reduced clinical disease or an independent predictor of disease severity. These results, in the context of the limited data available in humans, suggest that vaccines that induce anti-HA stalk antibodies could play a role in future vaccine strategies, but alone, this target may be insufficient to induce a fully protective vaccine and overcome some of the issues identified with current vaccines.
DOI: 10.1128/mbio.01044-15
2015
Cited 77 times
An Intranasal Virus-Like Particle Vaccine Broadly Protects Mice from Multiple Subtypes of Influenza A Virus
Influenza virus infections are a global public health problem, with a significant impact of morbidity and mortality from both annual epidemics and pandemics. The current strategy for preventing annual influenza is to develop a new vaccine each year against specific circulating virus strains. Because these vaccines are unlikely to protect against an antigenically divergent strain or a new pandemic virus with a novel hemagglutinin (HA) subtype, there is a critical need for vaccines that protect against all influenza A viruses, a so-called "universal" vaccine. Here we show that mice were broadly protected against challenge with a wide variety of lethal influenza A virus infections (94% aggregate survival following vaccination) with a virus-like particle (VLP) vaccine cocktail. The vaccine consisted of a mixture of VLPs individually displaying H1, H3, H5, or H7 HAs, and vaccinated mice showed significant protection following challenge with influenza viruses expressing 1918 H1, 1957 H2, and avian H5, H6, H7, H10, and H11 hemagglutinin subtypes. These experiments suggest a promising and practical strategy for developing a broadly protective "universal" influenza vaccine.The rapid and unpredictable nature of influenza A virus evolution requires new vaccines to be produced annually to match circulating strains. Human infections with influenza viruses derived from animals can cause outbreaks that may be associated with high mortality, and such strains may also adapt to humans to cause a future pandemic. Thus, there is a large public health need to create broadly protective, or "universal," influenza vaccines that could prevent disease from a wide variety of human and animal influenza A viruses. In this study, a noninfectious virus-like particle (VLP) vaccine was shown to offer significant protection against a variety of influenza A viruses in mice, suggesting a practical strategy to develop a universal influenza vaccine.
DOI: 10.1101/cshperspect.a038695
2019
Cited 74 times
The 1918 Influenza Pandemic and Its Legacy
Just over a century ago in 1918-1919, the "Spanish" influenza pandemic appeared nearly simultaneously around the world and caused extraordinary mortality-estimated at 50-100 million fatalities-associated with unexpected clinical and epidemiological features. The pandemic's sudden appearance and high fatality rate were unprecedented, and 100 years later still serve as a stark reminder of the continual threat influenza poses. Sequencing and reconstruction of the 1918 virus have allowed scientists to answer many questions about its origin and pathogenicity, although many questions remain. Several of the unusual features of the 1918-1919 pandemic, including age-specific mortality patterns and the high frequency of severe pneumonias, are still not fully understood. The 1918 pandemic virus initiated a pandemic era still ongoing. The descendants of the 1918 virus remain today as annually circulating and evolving influenza viruses causing significant mortality each year. This review summarizes key findings and unanswered questions about this deadliest of human events.
DOI: 10.1038/s41541-020-0174-9
2020
Cited 62 times
Efficacy of FLU-v, a broad-spectrum influenza vaccine, in a randomized phase IIb human influenza challenge study
Abstract FLU-v, developed by PepTcell (SEEK), is a peptide vaccine aiming to provide a broadly protective cellular immune response against influenza A and B. A randomized, double-blind, placebo-controlled, single-center, phase IIb efficacy and safety trial was conducted. One hundred and fifty-three healthy individuals 18–55 years of age were randomized to receive one or two doses of adjuvanted FLU-v or adjuvanted placebo subcutaneously on days −43 and −22, prior to intranasal challenge on day 0 with the A/California/04/2009/H1N1 human influenza A challenge virus. The primary objective of the study was to identify a reduction in mild to moderate influenza disease (MMID) defined as the presence of viral shedding and clinical influenza symptoms. Single-dose adjuvanted FLU-v recipients ( n = 40) were significantly less likely to develop MMID after challenge vs placebo ( n = 42) (32.5% vs 54.8% p = 0.035). FLU-v should continue to be evaluated and cellular immunity explored further as a possible important correlate of protection against influenza.
DOI: 10.2105/ajph.2021.306310
2021
Cited 43 times
A Centenary Tale of Two Pandemics: The 1918 Influenza Pandemic and COVID-19, Part I
Separated by a century, the influenza pandemic of 1918 and the COVID-19 pandemic of 2019–2021 are among the most disastrous infectious disease emergences of modern times. Although caused by unrelated viruses, the two pandemics are nevertheless similar in their clinical, pathological, and epidemiological features, and in the civic, public health, and medical responses to combat them. Comparing and contrasting the two pandemics, we consider what lessons we have learned over the span of a century and how we are applying those lessons to the challenges of COVID-19.
DOI: 10.1038/s41467-022-29614-9
2022
Cited 29 times
Archival influenza virus genomes from Europe reveal genomic variability during the 1918 pandemic
The 1918 influenza pandemic was the deadliest respiratory pandemic of the 20th century and determined the genomic make-up of subsequent human influenza A viruses (IAV). Here, we analyze both the first 1918 IAV genomes from Europe and the first from samples prior to the autumn peak. 1918 IAV genomic diversity is consistent with a combination of local transmission and long-distance dispersal events. Comparison of genomes before and during the pandemic peak shows variation at two sites in the nucleoprotein gene associated with resistance to host antiviral response, pointing at a possible adaptation of 1918 IAV to humans. Finally, local molecular clock modeling suggests a pure pandemic descent of seasonal H1N1 IAV as an alternative to the hypothesis of origination through an intrasubtype reassortment.
DOI: 10.1016/s1084-8592(97)80032-x
1997
Cited 156 times
Optimization of the isolation and amplification of RNA from formalin-fixed, paraffin-embedded tissue: The armed forces institute of pathology experience and literature review*, **
DOI: 10.1006/viro.2000.0495
2000
Cited 147 times
The 1918 Influenza Virus: A Killer Comes into View
In the fall and winter of 1918–1919, an influenza pandemic of unprecedented virulence swept the globe leaving 40 million or more dead in its wake. The virus responsible for this catastrophe was not isolated at the time, and it seemed that this very lethal infectious agent was lost for study. However, it has recently become possible to study the genetic features of the 1918 “Spanish” influenza virus using frozen and fixed archival autopsy tissue. Gene sequences of the 1918 virus can be used to frame hypotheses about the origin of the 1918 virus and to look for clues to its virulence. The study of the 1918 virus is not just one of historical curiosity. Since influenza viruses continually evolve by mechanisms of antigenic shift and drift, new influenza strains, as emerging pathogens, continue to threaten human populations. Pandemic influenza A viruses have emerged twice since 1918, in 1957 and 1968. The risk of future influenza pandemics is high. An understanding of the genetic makeup of the most virulent influenza strain in history may facilitate prediction and prevention of such future pandemics. Recent reviews dealing with more historical aspects of the 1918 pandemic are listed under Selected Reading.
DOI: 10.1016/s1286-4579(00)01351-4
2001
Cited 144 times
The 1918 Spanish influenza:integrating history and biology
In 1918 an influenza pandemic killed 40 million people. It is now possible to study the genetic features of the 1918 virus. Such analyses will try to answer questions about the origin and the unusual virulence of this pandemic virus.
DOI: 10.3201/eid0910.020789
2003
Cited 129 times
1918 Influenza Pandemic Caused by Highly Conserved Viruses with Two Receptor-Binding Variants
Abstract The “Spanish influenza pandemic swept the globe in the autumn and winter of 1918–19, and resulted in the deaths of approximately 40 million people. Clinically, epidemiologically, and pathologically, the disease was remarkably uniform, which suggests that similar viruses were causing disease around the world. To assess the homogeneity of the 1918 pandemic influenza virus, partial hemagglutinin gene sequences have been determined for five cases, including two newly identified samples from London, United Kingdom. The strains show 98.9% to 99.8% nucleotide sequence identity. One of the few differences between the strains maps to the receptor-binding site of hemagglutinin, suggesting that two receptor-binding configurations were co-circulating during the pandemic. The results suggest that in the early stages of an influenza A pandemic, mutations that occur during replication do not become fixed so that a uniform “consensus” strain circulates for some time.
DOI: 10.1073/pnas.212519699
2002
Cited 122 times
Existing antivirals are effective against influenza viruses with genes from the 1918 pandemic virus
The 1918 influenza pandemic caused more than 20 million deaths worldwide. Thus, the potential impact of a re-emergent 1918 or 1918-like influenza virus, whether through natural means or as a result of bioterrorism, is of significant concern. The genetic determinants of the virulence of the 1918 virus have not been defined yet, nor have specific clinical prophylaxis and/or treatment interventions that would be effective against a re-emergent 1918 or 1918-like virus been identified. Based on the reported nucleotide sequences, we have reconstructed the hemagglutinin (HA), neuraminidase (NA), and matrix (M) genes of the 1918 virus. Under biosafety level 3 (agricultural) conditions, we have generated recombinant influenza viruses bearing the 1918 HA, NA, or M segments. Strikingly, recombinant viruses possessing both the 1918 HA and 1918 NA were virulent in mice. In contrast, a control virus with the HA and NA from a more recent human isolate was unable to kill mice at any dose tested. The recombinant viruses were also tested for their sensitivity to U.S. Food and Drug Administration-approved antiinfluenza virus drugs in vitro and in vivo. Recombinant viruses possessing the 1918 NA or both the 1918 HA and 1918 NA were inhibited effectively in both tissue culture and mice by the NA inhibitors, zanamivir and oseltamivir. A recombinant virus possessing the 1918 M segment was inhibited effectively both in tissue culture and in vivo by the M2 ion-channel inhibitors amantadine and rimantadine. These data suggest that current antiviral strategies would be effective in curbing the dangers of a re-emergent 1918 or 1918-like virus.
DOI: 10.1128/jvi.78.22.12462-12470.2004
2004
Cited 114 times
Novel Origin of the 1918 Pandemic Influenza Virus Nucleoprotein Gene
ABSTRACT The nucleoprotein (NP) gene of the 1918 pandemic influenza A virus has been amplified and sequenced from archival material. The NP gene is known to be involved in many aspects of viral function and to interact with host proteins, thereby playing a role in host specificity. The 1918 NP amino acid sequence differs at only six amino acids from avian consensus sequences, consistent with reassortment from an avian source shortly before 1918. However, the nucleotide sequence of the 1918 NP gene has more than 170 differences from avian strain consensus sequences, suggesting substantial evolutionary distance from known avian strain sequences. Both the gene and protein sequences of the 1918 NP fall within the mammalian clade upon phylogenetic analysis. The evolutionary distance of the 1918 NP sequences from avian and mammalian strain sequences is examined, using several different parameters. The results suggest that the 1918 strain did not retain the previously circulating human NP. Nor is it likely to have obtained its NP by reassortment with an avian strain similar to those now characterized. The results are consistent with the existence of a currently unknown host for influenza, with an NP similar to current avian strain NPs at the amino acid level but with many synonymous nucleotide differences, suggesting evolutionary isolation from the currently characterized avian influenza virus gene pool.
DOI: 10.1177/135965350701200s02.1
2005
Cited 113 times
Discovery and Characterization of the 1918 Pandemic Influenza Virus in Historical Context
The 2005 completion of the entire genome sequence of the 1918 H1N1 pandemic influenza virus represents both a beginning and an end. Investigators have already begun to study the virus in vitro and in vivo to better understand its properties, pathogenicity, transmissibility and elicitation of host responses. Although this is an exciting new beginning, characterization of the 1918 virus also represents the culmination of over a century of scientific research aiming to understand the causes of pandemic influenza. In this brief review we attempt to place in historical context the identification and sequencing of the 1918 virus, including the alleged discovery of a bacterial cause of influenza during the 1889–1893 pandemic, the controversial detection of ‘filter-passing agents’ during the 1918–1919 pandemic, and subsequent breakthroughs in the 1930s that led to isolation of human and swine influenza viruses, greatly influencing the development of modern virology.
DOI: 10.1128/jvi.02683-07
2008
Cited 112 times
Homologous Recombination Is Very Rare or Absent in Human Influenza A Virus
ABSTRACT To determine the extent of homologous recombination in human influenza A virus, we assembled a data set of 13,852 sequences representing all eight segments and both major circulating subtypes, H3N2 and H1N1. Using an exhaustive search and a nonparametric test for mosaic structure, we identified 315 sequences (∼2%) in five different RNA segments that, after a multiple-comparison correction, had statistically significant mosaic signals compatible with homologous recombination. Of these, only two contained recombinant regions of sufficient length (&gt;100 nucleotides [nt]) that the occurrence of homologous recombination could be verified using phylogenetic methods, with the rest involving very short sequence regions (15 to 30 nt). Although this secondary analysis revealed patterns of phylogenetic incongruence compatible with the action of recombination, neither candidate recombinant was strongly supported. Given our inability to exclude the occurrence of mixed infection and template switching during amplification, laboratory artifacts provide an alternative and likely explanation for the occurrence of phylogenetic incongruence in these two cases. We therefore conclude that, if it occurs at all, homologous recombination plays only a very minor role in the evolution of human influenza A virus.
DOI: 10.1007/s00705-007-0994-1
2007
Cited 106 times
Using RRT-PCR analysis and virus isolation to determine the prevalence of avian influenza virus infections in ducks at Minto Flats State Game Refuge, Alaska, during August 2005
This study describes surveillance for avian influenza viruses (AIV) in the Minto Flats State Game Refuge, high-density waterfowl breeding grounds in Alaska. Five hundred paired cloacal samples from dabbling ducks (Northern Pintail, Mallard, Green Wing Teal, and Widgeon) were placed into ethanol and viral transport medium (VTM). Additional ethanol-preserved samples were taken. Of the ethanol-preserved samples, 25.6% were AIV RNA-positive by real-time RT-PCR. The hemagglutinin (HA) and neuraminidase (NA) subtypes were determined for 38 of the first-passage isolates, and four first-passage isolates could not be definitively subtyped. Five influenza A virus HA–NA combinations were identified: H3N6, H3N8, H4N6, H8N4, and H12N5. Differences in the prevalence of AIV infections by sex and by age classes of Northern Pintail and Mallard ducks were detected, but the significance of these differences is undefined. In the 500 paired samples, molecular screening detected positive birds at a higher rate than viral isolation (χ2 = 8.35, p = 0.0035, df = 1); however, 20 AIV isolates were recovered from PCR-negative ducks. Further research is warranted to compare the two screening protocols’ potential for estimating true prevalence in wild birds. Our success during 2005 indicates Minto Flats will be a valuable study site for a longitudinal research project designed to gain further insight into the natural history, evolution, and ecology of AIV in wild birds.
DOI: 10.1001/jama.297.18.2025
2007
Cited 106 times
The Next Influenza Pandemic
Our website uses cookies to enhance your experience. By continuing to use our site, or clicking "Continue," you are agreeing to our Cookie Policy | Continue JAMA HomeNew OnlineCurrent IssueFor Authors Podcasts Clinical Reviews Editors' Summary Medical News Author Interviews More Publications JAMA JAMA Network Open JAMA Cardiology JAMA Dermatology JAMA Health Forum JAMA Internal Medicine JAMA Neurology JAMA Oncology JAMA Ophthalmology JAMA Otolaryngology–Head & Neck Surgery JAMA Pediatrics JAMA Psychiatry JAMA Surgery Archives of Neurology & Psychiatry (1919-1959) JN Learning / CMESubscribeJobsInstitutions / LibrariansReprints & Permissions Terms of Use | Privacy Policy | Accessibility Statement 2023 American Medical Association. All Rights Reserved Search All JAMA JAMA Network Open JAMA Cardiology JAMA Dermatology JAMA Forum Archive JAMA Health Forum JAMA Internal Medicine JAMA Neurology JAMA Oncology JAMA Ophthalmology JAMA Otolaryngology–Head & Neck Surgery JAMA Pediatrics JAMA Psychiatry JAMA Surgery Archives of Neurology & Psychiatry Input Search Term Sign In Individual Sign In Sign inCreate an Account Access through your institution Sign In Purchase Options: Buy this article Rent this article Subscribe to the JAMA journal
DOI: 10.3201/eid1201.051442
2006
Cited 103 times
Influenza Revisited
DOI: 10.1016/j.virol.2012.06.003
2012
Cited 85 times
Protection against a lethal H5N1 influenza challenge by intranasal immunization with virus-like particles containing 2009 pandemic H1N1 neuraminidase in mice
Highly pathogenic H5N1 influenza shares the same neuraminidase (NA) subtype with the 2009 pandemic (H1N1pdm09), and cross-reactive NA immunity might protect against or mitigate lethal H5N1 infection. In this study, mice were either infected with a sublethal dose of H1N1pdm09 or were vaccinated and boosted with virus-like particles (VLP) consisting of the NA and matrix proteins, standardized by NA activity and administered intranasally, and were then challenged with a lethal dose of HPAI H5N1 virus. Mice previously infected with H1N1pdm09 survived H5N1 challenge with no detectable virus or respiratory tract pathology on day 4. Mice immunized with H5N1 or H1N1pdm09 NA VLPs were also fully protected from death, with a 100-fold and 10-fold reduction in infectious virus, respectively, and reduced pathology in the lungs. Human influenza vaccines that elicit not only HA, but also NA immunity may provide enhanced protection against the emergence of seasonal and pandemic viruses.
DOI: 10.1128/jvi.01848-10
2011
Cited 85 times
Pandemic Swine-Origin H1N1 Influenza A Virus Isolates Show Heterogeneous Virulence in Macaques
The first influenza pandemic of the new millennium was caused by a newly emerged swine-origin influenza virus (SOIV) (H1N1). This new virus is characterized by a previously unknown constellation of gene segments derived from North American and Eurasian swine lineages and the absence of common markers predictive of human adaptation. Overall, human infections appeared to be mild, but an alarming number of young individuals presented with symptoms atypical for seasonal influenza. The new SOIV also showed a sustained human-to-human transmissibility and higher reproduction ratio than common seasonal viruses, altogether indicating a higher pathogenic potential for this newly emerged virus. To study the virulence of the SOIV, we used a recently established cynomolgus macaque model and compared parameters of clinical disease, virology, host responses, and pathology/histopathology with a current seasonal H1N1 virus. We here show that infection of macaques with two genetically similar but clinically distinct SOIV isolates from the early stage of the pandemic (A/Mexico/4108/2009 and A/Mexico/InDRE4487/2009) resulted in upper and lower respiratory tract infections and clinical disease ranging from mild to severe pneumonia that was clearly advanced over the mild infection caused by A/Kawasaki/UTK-4/2009, a current seasonal strain. Unexpectedly, we observed heterogeneity among the two SOIV isolates in virus replication, host transcriptional and cytokine responses, and disease progression, demonstrating a higher pathogenic potential for A/Mexico/InDRE4487/2009. Differences in virulence may explain more severe disease, as was seen with certain individuals infected with the emerged pandemic influenza virus. Thus, the nonhuman primate model closely mimics influenza in humans.
DOI: 10.1586/eri.10.24
2010
Cited 84 times
Methods for molecular surveillance of influenza
Molecular-based techniques for detecting influenza viruses have become an integral component of human and animal surveillance programs in the last two decades. The recent pandemic of the swine-origin influenza A virus (H1N1) and the continuing circulation of highly pathogenic avian influenza A virus (H5N1) further stress the need for rapid and accurate identification and subtyping of influenza viruses for surveillance, outbreak management, diagnosis and treatment. There has been remarkable progress on the detection and molecular characterization of influenza virus infections in clinical, mammalian, domestic poultry and wild bird samples in recent years. The application of these techniques, including reverse transcriptase-PCR, real-time PCR, microarrays and other nucleic acid sequencing-based amplifications, have greatly enhanced the capability for surveillance and characterization of influenza viruses.
DOI: 10.1126/science.323.5921.1560
2009
Cited 82 times
Pandemic Influenza: An Inconvenient Mutation
Seasonal influenza affects 10% of the population annually, killing up to one million persons worldwide. Pandemic viruses have even greater potential for mortality. We have several defenses, including personal and public health protective measures, vaccines immunologically matched to circulating strains, and two classes of antiviral drugs (neuraminidase inhibitors and adamantane ion-channel blockers). Our preventive options are limited by viral genetic diversity and a rapid viral mutation rate. Currently, two human influenza A subtypes (H1N1 and H3N2) and two influenza type B lineages cocirculate. About 425 million doses of trivalent influenza vaccine are produced annually, enough to protect less than 7% of the world's population. In the event of a pandemic, well-matched protective vaccines against a novel agent would not be available for at least several months, highlighting the importance of therapeutic options.
DOI: 10.2214/ajr.09.3365
2009
Cited 82 times
Imaging Findings in a Fatal Case of Pandemic Swine-Origin Influenza A (H1N1)
Imaging Findings in a Fatal Case of Pandemic Swine-Origin Influenza A (H1N1)Daniel J. Mollura1, Deborah S. Asnis2 3, Robert S. Crupi4, Rick Conetta5, David S. Feigin6, Mike Bray7, Jeffery K. Taubenberger8 and David A. Bluemke1 6Audio Available | Share
DOI: 10.1002/rmv.689
2011
Cited 78 times
Pandemic influenza: certain uncertainties
SUMMARY For at least five centuries, major epidemics and pandemics of influenza have occurred unexpectedly and at irregular intervals. Despite the modern notion that pandemic influenza is a distinct phenomenon obeying such constant (if incompletely understood) rules such as dramatic genetic change, cyclicity, “wave” patterning, virus replacement, and predictable epidemic behavior, much evidence suggests the opposite. Although there is much that we know about pandemic influenza, there appears to be much more that we do not know. Pandemics arise as a result of various genetic mechanisms, have no predictable patterns of mortality among different age groups, and vary greatly in how and when they arise and recur. Some are followed by new pandemics, whereas others fade gradually or abruptly into long‐term endemicity. Human influenza pandemics have been caused by viruses that evolved singly or in co‐circulation with other pandemic virus descendants and often have involved significant transmission between, or establishment of, viral reservoirs within other animal hosts. In recent decades, pandemic influenza has continued to produce numerous unanticipated events that expose fundamental gaps in scientific knowledge. Influenza pandemics appear to be not a single phenomenon but a heterogeneous collection of viral evolutionary events whose similarities are overshadowed by important differences, the determinants of which remain poorly understood. These uncertainties make it difficult to predict influenza pandemics and, therefore, to adequately plan to prevent them. Published 2011. This article is a US Government work and is in the public domain in the USA.
DOI: 10.1111/j.1750-2659.2010.00148.x
2010
Cited 76 times
Historical thoughts on influenza viral ecosystems, or behold a pale horse, dead dogs, failing fowl, and sick swine
To understand human influenza in a historical context of viral circulation in avian species, mammals, and in the environment.Historical review.Global events in a variety of circumstances over more than 3,000 years time.Comprehensive review of the historical literature including all major publications on pandemic and panzootic influenza.Influenza pandemics, panzootics, major epidemics and epizootics, and instances of interspecies transmission of influenza A.Extensive documentation of human and animal influenza over many centuries suggests that influenza A viruses have adapted to a variety of species and environmental milieu and are capable of switching between many different hosts under widely varying circumstances.The genetic elements of influenza A viruses circulate globally in an extensive ecosystem comprised of many avian and mammalian species and a spectrum of environments. Unstable gene constellations found in avian species become stable viruses only upon switching to secondary hosts, but may then adapt and circulate independently. It may be desirable to think of influenza A viruses as existing and evolving in a large ecosystem involving multiple hosts and environments. Implications for understanding human influenza are discussed.
DOI: 10.1098/rsob.120109
2012
Cited 75 times
Ribosomal frameshifting used in influenza A virus expression occurs within the sequence UCC_UUU_CGU and is in the +1 direction
Programmed ribosomal frameshifting is used in the expression of many virus genes and some cellular genes. In eukaryotic systems, the most well-characterized mechanism involves -1 tandem tRNA slippage on an X_XXY_YYZ motif. By contrast, the mechanisms involved in programmed +1 (or -2) slippage are more varied and often poorly characterized. Recently, a novel gene, PA-X, was discovered in influenza A virus and found to be expressed via a shift to the +1 reading frame. Here, we identify, by mass spectrometric analysis, both the site (UCC_UUU_CGU) and direction (+1) of the frameshifting that is involved in PA-X expression. Related sites are identified in other virus genes that have previously been proposed to be expressed via +1 frameshifting. As these viruses infect insects (chronic bee paralysis virus), plants (fijiviruses and amalgamaviruses) and vertebrates (influenza A virus), such motifs may form a new class of +1 frameshift-inducing sequences that are active in diverse eukaryotes.
DOI: 10.1093/infdis/jis745
2012
Cited 75 times
Age- and Sex-Specific Mortality Associated With the 1918–1919 Influenza Pandemic in Kentucky
Background. The reasons for the unusual age-specific mortality patterns of the 1918–1919 influenza pandemic remain unknown. Here we characterize pandemic-related mortality by single year of age in a unique statewide Kentucky data set and explore breakpoints in the age curves.
DOI: 10.1016/j.virol.2011.01.029
2011
Cited 70 times
The ability of pandemic influenza virus hemagglutinins to induce lower respiratory pathology is associated with decreased surfactant protein D binding
Pandemic influenza viral infections have been associated with viral pneumonia. Chimeric influenza viruses with the hemagglutinin segment of the 1918, 1957, 1968, or 2009 pandemic influenza viruses in the context of a seasonal H1N1 influenza genome were constructed to analyze the role of hemagglutinin (HA) in pathogenesis and cell tropism in a mouse model. We also explored whether there was an association between the ability of lung surfactant protein D (SP-D) to bind to the HA and the ability of the corresponding chimeric virus to infect bronchiolar and alveolar epithelial cells of the lower respiratory tract. Viruses expressing the hemagglutinin of pandemic viruses were associated with significant pathology in the lower respiratory tract, including acute inflammation, and showed low binding activity for SP-D. In contrast, the virus expressing the HA of a seasonal influenza strain induced only mild disease with little lung pathology in infected mice and exhibited strong in vitro binding to SP-D.
DOI: 10.1128/mbio.00201-12
2012
Cited 69 times
Reconstruction of the 1918 Influenza Virus: Unexpected Rewards from the Past
ABSTRACT The influenza pandemic of 1918–1919 killed approximately 50 million people. The unusually severe morbidity and mortality associated with the pandemic spurred physicians and scientists to isolate the etiologic agent, but the virus was not isolated in 1918. In 1996, it became possible to recover and sequence highly degraded fragments of influenza viral RNA retained in preserved tissues from several 1918 victims. These viral RNA sequences eventually permitted reconstruction of the complete 1918 virus, which has yielded, almost a century after the deaths of its victims, novel insights into influenza virus biology and pathogenesis and has provided important information about how to prevent and control future pandemics.
DOI: 10.1093/infdis/jiq067
2011
Cited 69 times
MultiDrug-Resistant 2009 Pandemic Influenza A(H1N1) Viruses Maintain Fitness and Transmissibility in Ferrets
The 2009 influenza A(H1N1) pandemic called attention to the limited influenza treatment options available, especially in individuals at high risk of severe disease. Neuraminidase inhibitor-resistant seasonal H1N1 viruses have demonstrated the ability to transmit well despite early data indicating that resistance reduces viral fitness. 2009 H1N1 pandemic viruses have sporadically appeared containing resistance to neuraminidase inhibitors and the adamantanes, but the ability of these viruses to replicate, transmit, and cause disease in mammalian hosts has not been fully characterized.Two pretreatment wild-type viruses and 2 posttreatment multidrug-resistant viruses containing the neuraminidase H275Y mutation collected from immunocompromised patients infected with pandemic influenza H1N1 were tested for viral fitness, pathogenicity, and transmissibility in ferrets.The pretreatment wild-type viruses and posttreatment resistant viruses containing the H275Y mutation all demonstrated significant pathogenicity and equivalent viral fitness and transmissibility.The admantane-resistant 2009 pandemic influenza A(H1N1) virus can develop the H275Y change in the neuraminidase gene conferring resistance to both oseltamivir and peramivir without any loss in fitness, transmissibility, or pathogenicity. This suggests that the dissemination of widespread multidrug resistance similar to neuraminidase inhibitor resistance in seasonal H1N1 is a significant threat.
DOI: 10.1371/journal.pone.0124706
2015
Cited 67 times
Antiviral Activity of the Human Cathelicidin, LL-37, and Derived Peptides on Seasonal and Pandemic Influenza A Viruses
Human LL-37, a cationic antimicrobial peptide, was recently shown to have antiviral activity against influenza A virus (IAV) strains in vitro and in vivo. In this study we compared the anti-influenza activity of LL-37 with that of several fragments derived from LL-37. We first tested the peptides against a seasonal H3N2 strain and the mouse adapted H1N1 strain, PR-8. The N-terminal fragment, LL-23, had slight neutralizing activity against these strains. In LL-23V9 serine 9 is substituted by valine creating a continuous hydrophobic surface. LL-23V9 has been shown to have increased anti-bacterial activity compared to LL-23 and we now show slightly increased antiviral activity compared to LL-23 as well. The short central fragments, FK-13 and KR-12, which have anti-bacterial activity did not inhibit IAV. In contrast, a longer 20 amino acid central fragment of LL-37 (GI-20) had neutralizing activity similar to LL-37. None of the peptides inhibited viral hemagglutination or neuraminidase activity. We next tested activity of the peptides against a strain of pandemic H1N1 of 2009 (A/California/04/09/H1N1 or "Cal09"). Unexpectedly, LL-37 had markedly reduced activity against Cal09 using several cell types and assays of antiviral activity. A mutant viral strain containing just the hemagglutinin (HA) of 2009 pandemic H1N1 was inhibited by LL-37, suggested that genes other than the HA are involved in the resistance of pH1N1. In contrast, GI-20 did inhibit Cal09. In conclusion, the central helix of LL-37 incorporated in GI-20 appears to be required for optimal antiviral activity. The finding that GI-20 inhibits Cal09 suggests that it may be possible to engineer derivatives of LL-37 with improved antiviral properties.
DOI: 10.1002/path.4145
2013
Cited 66 times
High‐throughput <scp>RNA</scp> sequencing of a formalin‐fixed, paraffin‐embedded autopsy lung tissue sample from the 1918 influenza pandemic
Abstract Most biopsy and autopsy tissues are formalin‐fixed and paraffin‐embedded ( FFPE ), but this process leads to RNA degradation that limits gene expression analysis. The RNA genome of the 1918 pandemic influenza virus was previously determined in a 9‐year effort by overlapping RT‐PCR from post‐mortem samples. Here, the full genome of the 1918 virus at 3000× coverage was determined in one high‐throughput sequencing run of a library derived from total RNA of a 1918 FFPE sample after duplex‐specific nuclease treatments. Bacterial sequences associated with secondary bacterial pneumonias were also detected. Host transcripts were well represented in the library. Compared to a 2009 pandemic influenza virus FFPE post‐mortem library, the 1918 sample showed significant enrichment for host defence and cell death response genes, concordant with prior animal studies. This methodological approach should assist in the analysis of FFPE tissue samples isolated over the past century from a variety of diseases.
DOI: 10.1056/nejmp1307009
2013
Cited 61 times
Pandemic Influenza Viruses — Hoping for the Road Not Taken
Since the avian H7N9 influenza A virus that has emerged in China has some types of mutations associated with circulation in humans, some experts predict that it will become pandemic. But firm evidence of such well-defined viral-evolutionary pathways is lacking.
DOI: 10.2105/ajph.2018.304631
2018
Cited 57 times
The Mother of All Pandemics Is 100 Years Old (and Going Strong)!
This year marks the 100th anniversary of the deadliest event in human history. In 1918-1919, pandemic influenza appeared nearly simultaneously around the globe and caused extraordinary mortality (an estimated 50-100 million deaths) associated with unexpected clinical and epidemiological features. The descendants of the 1918 virus remain today; as endemic influenza viruses, they cause significant mortality each year. Although the ability to predict influenza pandemics remains no better than it was a century ago, numerous scientific advances provide an important head start in limiting severe disease and death from both current and future influenza viruses: identification and substantial characterization of the natural history and pathogenesis of the 1918 causative virus itself, as well as hundreds of its viral descendants; development of moderately effective vaccines; improved diagnosis and treatment of influenza-associated pneumonia; and effective prevention and control measures. Remaining challenges include development of vaccines eliciting significantly broader protection (against antigenically different influenza viruses) that can prevent or significantly downregulate viral replication; more complete characterization of natural history and pathogenesis emphasizing the protective role of mucosal immunity; and biomarkers of impending influenza-associated pneumonia.
DOI: 10.1016/j.coi.2018.03.025
2018
Cited 55 times
Neuraminidase as an influenza vaccine antigen: a low hanging fruit, ready for picking to improve vaccine effectiveness
Neuraminidase (NA) plays an essential role in influenza virus replication, facilitating multicycle infection predominantly by releasing virions from infected cells. NA-inhibiting antibodies provide resistance to disease and NA-specific antibodies contribute to vaccine efficacy. The primary reason NA vaccine content and immunogenicity was not routinely measured in the past, was the lack of suitable assays to quantify NA and NA-specific antibodies. These are now available and with recent appreciation of its contribution to immunity, NA content of seasonal and pandemic vaccines is being considered. An added benefit of NA as a vaccine antigen is that many NA-specific antibodies bind to domains that are well conserved within a subtype, protecting against heterologous viruses. This suggests NA may be a good choice for inclusion in universal influenza vaccines.
DOI: 10.1093/cid/ciz141
2019
Cited 55 times
A Dose-finding Study of a Wild-type Influenza A(H3N2) Virus in a Healthy Volunteer Human Challenge Model
Abstract Background The development of vaccines and therapeutics has relied on healthy volunteer influenza challenge studies. A validated human infection model with wild-type A(H1N1)pdm09 was reported previously. Our objective was to characterize a wild-type influenza A/Bethesda/MM1/H3N2 challenge virus in healthy volunteers. Methods Participants received a single dose of a cell-based, reverse-genetics, Good Manufacturing Practices–produced wild-type influenza A(H3N2)2011 virus intranasally and were isolated at the National Institutes of Health Clinical Center for ≥9 days. Dose escalation was performed from 104 to 107 TCID50 (50% tissue culture infectious dose). Viral shedding and clinical disease were evaluated daily. Results Of 37 participants challenged, 16 (43%) had viral shedding and 27 (73%) developed symptoms, with 12 (32%) participants experiencing mild to moderate influenza disease (MMID), defined as shedding and symptoms. Only participants receiving 106 and 107 TCID50 experienced MMID at 44% and 40%, respectively. Symptom severity peaked on day 3, whereas most viral shedding occurred 1–2 days after challenge. Only 10 (29%) participants had a ≥4-fold rise in hemagglutination inhibition antibody titer after challenge. Conclusions The A/Bethesda/MM1/H3N2 challenge virus safely induced MMID in healthy volunteers, but caused less MMID than the A(H1N1)pdm09 challenge virus even at the highest dose. There was less detection of shedding though the incidence of symptoms was similar to A(H1N1)pdm09. Fewer serum anti-hemagglutinin (HA) antibody responses with less MMID indicate that preexisting immunity factors other than anti-HA antibody may limit shedding in healthy volunteers. This A/Bethesda/MM1/H3N2 challenge virus can be utilized in future studies to further explore pathogenesis and immunity and to evaluate vaccine candidates. Clinical Trials Registration NCT02594189