key: cord-356188-rwf78stz authors: Oshansky, Christine M.; Thomas, Paul G. title: The human side of influenza date: 2012-07-01 journal: Journal of Leukocyte Biology DOI: 10.1189/jlb.1011506 sha: doc_id: 356188 cord_uid: rwf78stz A clear understanding of immunity in individuals infected with influenza virus is critical for the design of effective vaccination and treatment strategies. Whereas myriad studies have teased apart innate and adaptive immune responses to influenza infection in murine models, much less is known about human immunity as a result of the ethical and technical constraints of human research. Still, these murine studies have provided important insights into the critical correlates of protection and pathogenicity in human infection and helped direct the human studies that have been conducted. Here, we examine and review the current literature on immunity in humans infected with influenza virus, noting evidence offered by select murine studies and suggesting directions in which future research is most warranted. Influenza infection in humans can cause a wide range of disease symptoms, from asymptomatic to serious illness, characterized by sudden onset of fever, myalgia, and respiratory indications, including nonproductive cough, sore throat, and rhinitis. In addition, children infected with influenza commonly present with otitis media, nausea, and vomiting and more frequently, complain of rhinorrhea [1, 2] . Historically, the pediatric and elderly populations are considered vulnerable during influenza epidemics [3] [4] [5] [6] , and although still debated, studies suggest that children have a unique role in the spread of influenza during epidemics [7] [8] [9] . Children under 2 years old are more susceptible to secondary complications [10] , and infants with severe respiratory tract viral infection are often at greater danger for pulmonary dysfunction, including wheezing, allergy, and asthma later in life [11] [12] [13] . In contrast, elderly individuals are at higher risk for developing hemorrhagic bronchitis, pneumonia, and death when infected with influenza [14] , and this population generally has a less-robust immune response following influenza immunization. Although RSV infections are the leading cause of hospitalizations of children in the United States, influenza accounts for the highest incidence of infection in children under 2 years of age [15] . Influenza epidemics occur annually and can cause significant morbidity and mortality worldwide. During 1976 -2007, the CDC estimated that in the United States alone, annually, 8.5% (6309) of all "deaths with underlying pneumonia and influenza causes" (74, 363) were influenza-associated, and 2.1% (23, 607) of "deaths with underlying respiratory and circulatory causes" (1,132,319) were influenza-associated [16] . Furthermore, individuals with underlying respiratory and cardiac disease or diabetes mellitis, infected with influenza, are often at higher risk for developing hemorrhagic bronchitis or pneumonia and associated comorbidities. In 2008, it was estimated that there were 28,000 -111,500 deaths attributable to influenza in children Ͻ5 years of age globally, with developing countries carrying the largest burden [17] . In the United States, annual influenza epidemics account for an estimated $10 billion (12% of the total economic burden) in direct medical costs [18] and an estimated $44 -$163 million in hospitalization costs for children [19] . Introduction of a swine-origin influenza into the human population in 2009 quickly disseminated globally, causing the first pandemic of the 21st century. This novel H1N1 virus, hereafter referred to as A(H1N1)pdm09 [20] , disproportionately affected children and young adults [21, 22] . Observed mortality increased between four and 10 times in the pediatric population during the initial wave of A(H1N1)pdm09 infection in 2009 compared with previous influenza seasons [23] , whereas those individuals born prior to 1950 had pre-existing, cross-reactive antibodies against A(H1N1)pdm09 [24] . As a result, within the population of those with laboratory-confirmed A(H1N1)pdm09, 2% were older individuals (Ͼ60 years), whereas 79% were Ͻ30 years old. Risk factors for severe A(H1N1)pdm09 infection are similar to those for seasonal influenza but also include obesity. The majority of the severe or fatal cases have accompanying chronic illnesses, such as cardiac disease, chronic respiratory diseases, and diabetes [25] . Several studies suggest that pregnant and postpartum women are at increased risk for complications from infections with A(H1N1)pdm09 [26 -28] . Furthermore, severity of A(H1N1)pdm09 infection was related to the length of time between symptom onset and the initiation of antiviral treatment with NA inhibitors [29] . Influenza infections are associated with secondary bacterial infections, or superinfections, most notably, Streptococcus pneumoniae and Staphylococcus aureus (reviewed in ref. [30] ). In children, the most common secondary bacterial infections lead to acute otitis media, which is found in 50% of patients with symptomatic influenza infection, with five to six cases observed annually for every 100 children Ͻ2 years of age [10] . Since the emergence of A(H1N1)pdm09, approximately one-third of fatal infections are associated with bacterial coinfection [31, 32] , but this number includes cases where respiratory support is needed, adding to the risk of secondary infections. In fatal cases of those infected by A(H1N1)pdm09, diffuse alveolar damage alone, diffuse alveolar damage associated with necrotizing bronchiolitis, and diffuse alveolar damage with hemorrhage were found, and in most of the cases where necrotizing bronchiolitis was found, the individuals had bacterial coinfections [33] . Vaccination remains an effective and primary preventive strategy to avert influenza infection. The World Health Organization and CDC recommend that children and adults over the age of 6 months receive an influenza vaccination annually [34] . Currently, there are three licensed seasonal vaccines administered in the United States: 1) a TIV, administered by i.m. injection; 2) a LAIV, delivered intranasally; and 3) an i.d.-administered TIV preparation [35] . Each includes three circulating strains of influenza grown in eggs, reflecting annual surveillance data that predict which strains (A/H1, A/H3, and B) are most likely to be circulating the following season in northern and southern hemispheres. The i.m. TIV is approved for use in children and adults over 6 months of age, whereas the newer i.d. TIV is approved for adults aged 18 -64 years. LAIV is approved for use in healthy children and adults between 2 and 49 years of age. The current influenza vaccines work to induce memory recall responses, primarily via humoral immune responses against the HA and NA surface glycoproteins, and HA inhibition antibody responses following immunization correlates with protection against infection [36, 37] . The main component standardized in influenza vaccines is the HA protein, and it is well-characterized in safety trials and challenge studies that the amount of HA within the vaccine correlates with level of protection elicited by immunization [38, 39] . Beginning during the northern hemisphere's 2010 -2011 influenza season, a vaccine containing 60 g HA/vaccine strain, as opposed to 15 g in other preparations, was approved as an alternative TIV for elderly individuals (over age 65 years) [35] . Following vaccination, ASCs rapidly proliferate upon antigen exposure, and cell numbers peak 1 week postimmunization in healthy adults and children [40, 41] . IgG and IgA ASCs decline to low levels by 4 -6 weeks postimmunization [42, 43] . This increase in the total numbers of ASCs corre-sponds to nAb levels postvaccination, peaking at 4 weeks in adults and children [43] . LAIV elicits a strong serum and mucosal influenza-specific antibody response [44] , and studies in young children (6 -59 months of age) showed that those receiving LAIV had significantly reduced incidence of influenza infection [45] . Although limited data exist, influenza-specific ASCs and nAb titers are increased to a greater degree following TIV immunization compared with LAIV vaccination [43, 46, 47] , but the role of neutralization of LAIV occurring in antigen-experienced individuals is not well understood. Influenza immunization is effective, and studies in healthy children Ͻ15 years of age have shown TIV efficacies ranging from 31% to 90% (reviewed in ref. [48] ). Since 2010, universal influenza vaccination is recommended, i.e., for all individuals at least 6 months of age, but traditional efforts remain focused on those individuals at greatest risk of serious disease: young children, elderly, individuals with pulmonary or cardiovascular disorders, and those who are immunocompromised or pregnant [35] . Vaccination programs also elicit indirect benefits, including herd immunity, by which immunized individuals protect those who are immunocompromised or other nonvaccinated individuals [49, 50] . This effect was observed in a retrospective epidemiological study in Japan and the United States examining mortality and influenza vaccination rates between 1949 and 1998 [51] . As children have long been considered to have an important role in the spread of influenza during epidemics [7] [8] [9] , Japanese officials legislated compulsory vaccination of school-aged children in 1977, and as the policy changed to optional immunization in 1987, it became clear that mortality rates were inversely correlated with overall vaccination coverage. However, recent reviews of vaccine studies in various populations have concluded that overestimation of the benefits of vaccination is common, particularly within the elderly population [52, 53] , but studies that take into account selection biases do suggest that vaccination is associated with lower risk of hospitalization for pneumonia or influenza, as well as all-cause mortality [52, 54, 55] . Furthermore, in October 2009, the United States commenced a national influenza H1N1/2009 vaccination campaign, and it is estimated that ϳ41.2% and 43% of the U.S. population received the 2009 -2010 and 2010 -2011 influenza vaccines, respectively [56] . Following implementation of this campaign, the frequency of positive influenza cultures reported to the CDC declined quickly. Immunization and herd immunity resulting from prior exposure of A(H1N1)pdm09 likely resulted in decreased cases of influenza during the 2010 -2011 influenza season. Undoubtedly, influenza vaccines can be improved, particularly for the most vulnerable populations. Continuous epidemiological surveillance of circulating influenza strains is required for keeping pace with antigenic drift and shift (reviewed in ref. [57] ). Antigenic drift is an endless battle, whereby viral variants emerge as a result of point mutations within the viral genome, escaping antibody-mediated viral neutralization. Whereas many mutations occur throughout the viral genome, the point mutations within the HA or NA proteins can change antibody-binding domains, such that individuals no longer have adequate, protective B cell memory responses. In contrast, major qualitative changes in antigenicity as a result of antigenic shift occur as a result of genetic reassortment and replacement of one or more influenza gene segments. This, in turn, creates a new influenza virus subtype, to which little or no immunity may exist within the population. Antigenic shift remains a chief concern, in that highly pathogenic influenza viruses could potentially gain efficient human-to-human transmission. As influenza viruses frequently undergo antigenic drift and less frequently, shift, a desirable vaccine candidate would be able to induce highly specific, cross-reactive antibodies and T cell responses. One strategy used in vaccine improvement includes the use of adjuvant to increase antigen uptake by APCs and enhance cytokine production. Neither of the U.S.-licensed TIVs nor LAIV include adjuvant, but there are several licensed vaccines available outside of the United States that use oil and water emulsions for seasonal influenza as well as for A/H5N1 (reviewed in ref. [58] ). For instance, Novartis Vaccines and Diagnostics (Switzerland) manufactures Fluad using MF59 adjuvant, and GlaxoSmithKline Biologicals (Belgium) produces Pandemrix, which includes AS03. Fluad has been used in Europe since 1997 and was recently approved for use in Canada in adults 65 years and older. MF59 and AS03 are both squalene-based with surfactants included to stabilize the emulsions. Clinical trials of MF59-adjuvanted vaccines are well-tolerated and elicit higher antibody responses in adults [59 -61] and in children [62] [63] [64] compared with unadjuvanted vaccine preparations. The concept of a universal influenza vaccine, where one vaccine elicits protection against multiple influenza subtypes, is becoming a reality. The influenza M2 ion channel is expressed at high levels on the surface of influenza-infected cells and has a conserved ectodomain (M2e) that is 24 aa in length [65] . Animal studies have demonstrated improved viral clearance and cross-protection upon lethal challenge in mice following passive M2 mAb transfer or M2-immunized mice (reviewed in ref. [66] ). Phase I clinical safety trials evaluating VAX102, manufactured by VaxInnate, have reported increased M2e antibody responses [67] . Creation of a universal influenza vaccine that targets conserved regions of the HA stem region is particularly appealing, and several groups have demonstrated encouraging results using broadly neutralizing mAb to regions of HA that are cross-reactive between Group 1 and Group 2 influenza strains [68 -70] . Structural analyses indicate that these antibodies bind to conserved pockets of the A helix of the HA stalk, located below the trimeric head region [71] [72] [73] . It is therefore within the realm of possibility to focus immunization strategies to these conserved regions in the HA and M2 proteins to elicit effective antibody, and presumably T cell, responses in the case of emergent influenza viruses. Still, it remains a mystery as to why these conserved epitopes have not generated a high level of cross-protection in populations repeatedly immunized or infected with diverse viral strains. Factors that limit influenza vaccine efficacy in humans include age, immunocompetence, and pre-existing antibody levels from previous infections or immunizations. Whereas key lessons have been learned using animal models of influenza infection, studies done using human participants are invaluable for describing the unique and often complex characteristics of the human immune response to infection and immunization. Here, we describe central findings obtained from human influenza challenge studies, natural infections, and ex vivo characterization of human immune cells ( Table 1 ). Figure 1 indicates where questions remain for the human immune response to infection. Effective control and clearance of influenza infection can involve most components of the innate and adaptive immune responses. In mammals, the cellular innate immune system acts as a first-line defense consisting of multipotent hematopoietic stem cells that differentiate into NK cells, mast cells, eosinophils, and basophils, as well as macrophages, neutrophils, and DCs, collectively known as phagocytic cells. Phagocytes function as APCs, engulfing and digesting opsonized pathogens and apoptotic cells. These cells essentially act as scavengers to uptake toxic metabolic byproducts and produce myriad inflammatory mediators, which ideally, result in the killing of viruses, bacteria, and parasites. However, abundant inflammation often has a role in creating immunopathology and can contribute to diverse inflammatory diseases, including asthma [123] [124] [125] . In the absence of protective immune memory, the innate response limits initial viral replication, while the adaptive immune response develops. Influenza primarily infects epithelial cells lining the respiratory tract but can infect directly or be phagoctyosed by "professional" innate immune cells, DCs, and macrophages, which mature in response to viral stimuli or signals from other infected cells. After activation, mature DCs migrate to draining LNs, where they have a major role in antigen presentation to naïve T cells. Influenza infection of human monocyte-derived DCs can result in decreased DC maturation from NS1 suppression of type I IFN production [126] . Seasonal H1N1 viruses, including A(H1N1)pdm09, elicit weak, proinflammatory cytokine gene expression in human DCs [127] . We know from murine models that influenza contains ligands that activate several PRR signaling pathways, including TLR3 and retinoic acid-inducible gene I (RIG-I), which recognize dsRNA, and TLR7, which recognizes ssRNA [128 -130] . Type I (IFN-␣/␤) and type III (IFN-1-3) IFNs mediate expression of more than 300 ISGs via the JAK/STAT pathway and are important in the induction of antiviral responses against influenza viruses [131] . Stimulation of these pathways in DCs and macrophages leads to the induction of a potent antiviral response, including production of proinflammatory cytokines IL-6 and IFN-␣ within the respiratory tract by Day 2 following infection, paralleling the peak of viral replication, mucus production, and disease symptoms. TNF-␣ and IL-8 levels peak between Day 3 and 6 after infection as symptoms begin to subside [95] . Antimicrobial peptides, such as collectins, defensins, and cathelicidins, are important innate effectors in the human defense against influenza infection. Collectins, or collagenous lectins, are found primarily in mucosal secretions and include SP-A and SP-D; SP-D has been shown to inhibit HA activity by binding to N-linked, high-mannose oligosaccharides in a calcium-dependent manner, while enhancing activation of neutrophils in in vitro assays [132, 133] . In murine models, SP-A and SP-D have roles in promoting viral clearance from lungs and reducing inflammatory cytokines [134, 135] , but their role during human influenza infection is less well-defined. Influenza virus impairs calcium metabolism and subsequent calcium deactivation in neutrophils [136] , and interestingly, SP-D can protect neutrophils from calcium deactivation if preincubated with virus, presumably by inhibiting HA activity [132] . In humans, the only cathelicidin, human cathelicidin 18, the active form of which is LL-37, is found in diverse cell types, including neutrophils, NK cells, monocytes, B cells, ␥␦ T cells, mast cells, as well as epithelial cells lining the lungs [137, 138] . Like surfactants, LL-37 shows antiviral activity against influenza and can reduce viral titers in in vitro studies [139] . Defensins have a key role in host defense and act primarily to disrupt lipid membranes leading to lysis and subsequent death of bacteria, fungi, and enveloped viruses [140] . There are two classes of defensins, ␣-defensin and ␤-defensin, which were first described in models of host defense against bacterial pathogens. Both types are found in the respiratory and gastrointestinal tracts. The ␣-defensins in humans include HNP-1, -2, -3, and -4, all found in high concentrations within neutrophil granules and released within the lungs in response to proinflammatory signals during infection and inflammation [141] . In vitro, HNP-1 and HNP-2 act as chemokines for human monocytes [142] , and HNP-1, -2, and -3 can induce neutrophil internalization of influenza in vitro [143] . Moreover, the ␣-defensins may have a role in directly inactivating influenza virus as A/WSN preincubated with HNP-1 showed decreased infection in vitro in a dosedependent manner [144] . HNP-1 can also inhibit influenza virus replication via modulation of cellular pathways, including the PKC pathway [145] . Heterogeneous populations of monocytes comprise ϳ10% of blood leukocytes in humans (4% in the mouse), and their roles in controlling pathogens and inflammation can be characterized by functional cellular subsets. Human blood monocytes can be divided into three populations based primarily on cell surface expression of CD14 (LPS receptor) and CD16 (Fc␥RIII). The first subgroup consists of "classical monocytes", characterized by CD14 expression and lack of CD16 expression (CD14 ϩ CD16 Ϫ ). The CD14 ϩ CD16 Ϫ monocytes comprise 80 -90% of blood monocytes and resemble murine inflammatory Ly6C ϩ (Gr1 ϩ ) monocytes [146] . The second subgroup is defined by high expression of CD14 and CD16 (CD14 ϩ CD16 ϩ ) and release TNF following LPS stimulation [147] . The third monocytic subgroup is characterized by low expression of CD14 and high expression of CD16 (CD14 dim CD16 ϩ ), and at least one study has shown increased numbers in the blood of sepsis patients [148] . CD14 dim blood monocytes function as efficient producers of proinflammatory cytokines in response to viruses and nucleic acids [149, 150] . Few studies have examined the role of monocytes during influenza infection in humans, particularly regarding the specific subsets mentioned above, but comparison of IFN-␥ production from T cells cocultured with CD64 ϩ CD16 Ϫ and CD64 Ϫ CD16 ϩ monocytes [119, 120] Cellular immunity Class I HLA presents peptides from internal and external viral proteins. [121, 122] showed that CD64 Ϫ CD16 ϩ monocytes (equivalent to the CD14 ϩ CD16 ϩ monocytes) induced a greater response to influenza A antigen [151] . In vitro, human DCs and macrophages treated with type I or type III IFN prior to influenza infection result in potent antiviral activity and decreased viral replication [127] . Among DC populations, CD11c Ϫ CD14 Ϫ HLA-DR ϩ CD123 ϩ pDCs were found to produce more IFN-␣ in response to a high dose of A/PR8/8/34 (H1N1) [152] . Whereas increased numbers of total CD14 ϩ cells are observed in the blood of influenza-infected individuals, decreased blood myeloid DCs and pDCs were observed (defined in this study as HLA-DR ϩ CD14 Ϫ CD16 Ϫ CD11c ϩ and HLA-DR ϩ CD14 Ϫ CD16 Ϫ CD123 ϩ , respectively) [153] . However, increased numbers of pDCs were observed in nasal secretions compared with healthy controls within this cohort. The NS1 protein is a well-characterized antagonist of the host's antiviral response, acting primarily to circumvent IFN-␣/␤ responses (reviewed in ref. [154] ), and in vivo models show that its deletion results in virus attenuation and higher levels of IFN [155] . Type I IFN antagonism can result in inhibition of adaptive immunity by interfering with DC maturation and thus, the ability of DCs to activate T cells [126] . The NS1 protein from influenza B virus inhibits ISGylation, an important host antiviral effector mechanism, by interacting with the N-terminal domain of ISG15 [156] . Murine models have provided several clues regarding the actual role of monocytes during the innate immune response to influenza infection. In vivo, CCR2-dependent recruitment of inflammatory monocytes is critical for host defense and contributes to the innate immune response, as well as downstream adaptive responses [157] . During influenza infection, CCR2deficient mice have reduced monocyte and increased neutrophil recruitment, but, despite higher viral lung titers, in one model of infection these mice were protected from severe pneumonitis [158, 159] . In the lungs, CCR2-mediated recruit- ment of TNF and iNOS-producing DCs augments influenzaspecific T cell responses [160] . At least one murine study suggests that pregnant mice have increased infiltration of neutrophils (CD11b high Ly6G high MHCII Ϫ ) and macrophages (CD11b high Ly6G Ϫ MHCII high ) and elevated, proinflammatory cytokine levels, despite no change in viral titers in the lungs compared with nonpregnant animals [161] . In vitro, influenza infection of murine alveolar epithelial cells resulted in monocyte production of MCP-1 (CCL2, the ligand for CCR2) and RANTES, and monocyte migration was dependent on CCR2 expression [162] . Although some murine models suggest that neutrophils may have little role in mediating viral clearance [163] , type I interferon (IFN-␣/␤)-dependent generation of Ly6C hi monocytes following influenza infection may be necessary to limit excess neutrophil infiltration, preventing tissue damage caused by uncontrolled inflammation [164] . In addition, the newly described innate lymphoid cells have been implicated in having a role in airway integrity following influenza infection [165] . Thus, innate immune cells, particularly monocytes, have at least two roles during influenza infection: to boost the innate immune response by producing proinflammatory cytokines and to enhance influenza-specific T cell responses. However, the lack of clear correspondence between murine and human monocytic subsets has made the application of these data to the human situation difficult and highlights the need for specific studies focused on human innate immune responses. During RSV and influenza infection of infants, neutrophils are recruited to the upper and lower airways in high numbers [103, 104] , and fatal cases during the A(H1N1)pdm09 pandemic with necrotizing bronchiolitis had more neutrophils within the lung infiltrate [33] . The neutrophil chemoattractant IL-8, produced by bronchial epithelial cells, is elevated early during experimental human influenza infection [95] and is increased in the serum of A/H5N1-infected individuals [96] . Increased IL-8 levels in BALs of humans are associated with the infiltration of neutrophils and may have a role in neutrophil-mediated lung injury and development of acute respiratory distress syndrome [97] . Ex vivo influenza infection of PB-MCs has been shown to induce apoptosis in neutrophils [166] and peripheral blood monocytes [167] . Influenza infection of neutrophils resulted in increased hydrogen peroxide production, enhanced surface expression of Fas antigen and FasL, and secretion of FasL into the supernatant [166] . Interestingly, this study showed that influenza infection of neutrophils resulted in increased internalization of Escherichia coli, suggesting a role in controlling bacterial secondary infections in humans. Moreover, standard chemotaxis assays during experimental influenza infection [168 -170] or chemokine receptor expression during ex vivo influenza infection [105] demonstrated impaired chemotaxis of monocytes and granulocytes. Still, although all of these studies suggest that neutrophils can be regulated and recruited during human influenza infection, the specific protective and pathological roles that they play in vivo remain unclear. Nasopharygeal aspirates of infants infected with influenza have increased levels of proinflammatory cytokines such as IL-6, TNF-␣, IL-10, or IFN-␥ compared with RSV-or human metapneumovirus-infected infants [98 -100] , and MIP-1␣ levels are higher in infants with more severe influenza infections characterized by hypoxic bronchiolitis [101] . Variability between these studies is likely a result of seasonal differences observed year-to-year. Individuals infected with A/H5N1 have high viral loads detected within respiratory secretions and peripheral hypercytokinemia [96, 102] . Severe disease is associated with macrophage infiltration into the lungs, and chemoattractants of monocytes and macrophages, such as IFN-inducible protein 10 (IP-10), monokine induced by IFN-␥ (MIG), and MCP-1, were found to be elevated in the serum of influenza-infected individuals and were highest in those infected with A/H5N1 [96] . Therefore, peripheral (or local) hypercytokinemia likely contributes to enhanced immunopathogenesis during influenza disease. Although there is no difference in the duration of viral shedding in children hospitalized for A/H1N1 or A/H3N2 infections compared with those infected with influenza B virus, children with influenza A exhibited different serum cytokine profiles. Serum IFN-␥ levels were similar during acute-phase infection [171] , but levels were increased 3 weeks following influenza B infection compared with influenza A infection. Furthermore, serum IL-4 levels were significantly higher during the acute and recovery phases of influenza A infection. Influenza strain differences were also observed in umbilical cord blood lymphocytes treated with UV-inactivated viruses. UV-inactivated influenza B virus induced less IFN-␥, IL-4, or IL-10 relative to UV-inactivated A/H1N1, A/H3N2, and A/H2N2 [172] . Together, these data strongly suggest that innate cells and their associated responses have an important role in influenza disease manifestation, by providing protection and by contributing to pathological outcomes. The murine studies cited here clearly demonstrate that the local inflammatory environment can be dramatically different from the blood profile. The vast majority of experimental data in humans is restricted to blood leukocytes, with minimal emphasis placed on sites of infection. Further, we lack good, functional analysis of these cells in humans, and most studies provide correlational, cross-sectional observations. As a result, there is a significant gap in our knowledge of innate immune cells in affected tissues. The history of respiratory virus challenge studies dates back to the 1918 influenza pandemic [173] , and early studies in humans infected with influenza identified the principal correlate of protection against influenza infection as strain-specific, virus-neutralizing antibodies directed against the HA. These studies (reviewed in ref. [80] ) suggested that the presence of serum antibody is associated with protection, as illness is less frequently observed in those people with pre-existing nAb [74] . Subsequent studies showed that protection is also correlated with the development of serum and mucosal anti-NA or anti-HA IgG and IgA [37, 47, [75] [76] [77] [78] , but this protection can be incomplete, as individuals with detectable nAb can still be infected [79, 174] . Antibodies specific for the NA have been shown to reduce disease severity by restricting virus release from infected cells and enhancing viral clearance (reviewed in ref. [175] ). In addition to HA and NA, antibodies are produced to NP, matrix M2 protein, and PB1-F2 [176] , although the significance of these responses remains unclear. Of course, antigenic shift and antigenic drift can both thwart the efficacy of serological memory. The recent circulation of A(H1N1)pdm09 has provided a modern "experiment of nature", demonstrating the importance of pre-existing antibody from prior infection or vaccination [81] [82] [83] . Even prior to 2009, it was well-recognized that heterosubtypic immunity has a large role in protection against antigenic variants of influenza [84, 85] , and vaccine studies support the idea that cross-protective, adaptive immune responses occur [86] . Likewise, neutralizing polyclonal antibodies cross-reactive against H5 can also be produced in individuals vaccinated against seasonal influenza, as well as those naturally infected with A(H1N1)pdm09 [87, 88] . Compared with seasonal influenza infections prior to 2009, ϳ31% of B cell epitopes are conserved in A(H1N1)pdm09, with 17% of those conserved within the surface viral proteins [177] . The HA of A(H1N1)pdm09 contains sequences homologous with that of A/H1N1 (1918) demonstrated by the A/H1N1 (1918) mAb 2D1, which binds to the Sa antigenic site of the HA [89] . Furthermore, this particular antibody can neutralize HA activity in vitro, and mAb-treated mice show reduced viral titers when challenged with A(H1N1)pdm09 [90] . Most neutralizing antibodies bind to the exposed regions of the HA that surround the receptor-binding site and interfere with attachment to sialic acids on the cell surface. Therefore, generation of crossprotective neutralizing antibodies is elusive, as the external viral proteins are most exposed to immune pressure, but universal immunization against conserved HA stalk and M2e domains is promising [71] [72] [73] . Human PBMCs include at least three major subsets of DCs: CD141 ϩ mDCs, CD1c ϩ mDCs, and pDCs (reviewed in ref. [178] ). DCs process and present antigen to T cells and therefore have an important role in regulating the adaptive immune response. Depending on the local cytokine milieu, mDCs can differentiate into macrophages or tissue-specific DCs (i.e., epithelial Langerhans cells or interstitial DCs). As influenza primarily infects epithelial cells lining the respiratory tract, lung-resident DCs and macrophages are particularly important for efficient development of an adaptive immune response. Once activated, mDCs function as APCs to activate T cells; on the other hand, pDCs are major producers of type I IFN upon TLR activation. In response to influenza infection in vitro, human DCs and macrophages undergo maturation, secreting type I IFN and inducing T cell proliferation and IFN-␥ production [179 -181] . Cellular immune responses (CD8 ϩ CTLs and CD4 ϩ Th cells) are important for virus clearance in murine models of influenza infection, with CD4 ϩ and CD8 ϩ T cell-depleted mice displaying delayed viral clearance. The relevance of these observations remains somewhat controversial in humans. Regard-less, as in mice, the human lung contains resident respiratory virus-specific CD8 ϩ memory T cells [182] , which can effectively kill virus-infected cells. Human CD8 ϩ T cells recognize antigen in the context of surface HLA molecules and generally recognize a broad range of influenza epitopes, including those from structural and nonstructural influenza proteins [121] . Approximately 41% of CD4 ϩ and 69% of CD8 ϩ T cell epitopes are conserved in A(H1N1)pdm09 from the prior circulating seasonal H1N1 [177] . Furthermore, CD8 ϩ intraepithelial T cells can mount fast and efficient recall responses upon viral infection [183] , and immunization studies in humans have shown that T cell responses peak within 1 week after symptom onset [174] . In terms of pandemic viruses, individuals infected with A(H1N1)pdm09 were shown to have rapid, virus-specific CD8 ϩ T cell recall responses early during infection, but this CTL response wanes over time [184] . Likewise, vaccinated individuals, who were then naturally infected with A(H1N1)pdm09, had T cell responses increase by twofold early after infection [174] . Memory CD4 ϩ or CD8 ϩ T cells isolated from PBMCs of healthy individuals are also cross-reactive against proteins of heterologous viruses, including those from A(H1N1)pdm09 and A/H5N1 [91] [92] [93] [94] , but immunocompromised patients do not have lasting antibody or CD4 ϩ /CD8 ϩ memory responses following natural A(H1N1)pdm09 infection [185] . Furthermore, CTLs, isolated from A(H1N1)pdm09-or A/H5N1-inexperienced individuals with a memory CD45RA med CD62 ligand lo CCR7 Ϫ phenotype, will respond robustly to peptide pools and influenza-infected cells [34, 177, 186, 187] , suggesting that T cells may provide some level of protection against conserved epitopes [110] . Although nonconserved regions of the virus may effectively escape recognition by T cells [188] , the majority of T cell responses is elicited against conserved, internal viral proteins, generating much interest in vaccine or therapeutic strategies that involve T cell-mediated immunity. A frequently neglected population of lymphocytes, ␥␦ T cells comprise 1-5% of T cells in the blood of humans, the majority of which are V␥9V␦2 T cells. These ␥␦ T cells are considered to be a major innate-like T cell subset (reviewed in ref. [189] ), and in vitro studies suggest that activated human V␥9V␦2 T cells may have a role in the antiviral response by killing influenza-infected, monocyte-derived macrophages and producing high levels of IFN-␥ [190, 191] . In particular, neonates are known to have compromised cellular immune responses and represent an important target population for influenza immunization. Despite being such a critical target population, there are relatively few neonatal models of respiratory infections, and for ethical reasons, it is appropriately difficult to acquire data from human neonates. In the case of RSV infection, murine neonate models suggest that the age of primary viral infection can determine the immunopathology of a secondary viral infection [192] [193] [194] . During RSV infection, younger murine neonates (Յ7 days) will develop an immune response characterized by increased eosinophil and T cell recruitment to the lungs, enhanced airway hyper-responsiveness, and higher IL-13 levels upon reinfection 12 weeks later, whereas older mice (Ն4 weeks) develop an immune response characterized by decreased T cell and eosinophil recruitment to the lungs upon reinfection [192, 193] . Likewise, a neonatal mouse model of influenza A virus showed that early infection was associated with decreased CD8 ϩ T cell recruitment and function and enhanced airway inflammation [195] . These studies highlight an important concept, whereby infection too early results not in protective immunity but immunopathogenesis. Consistent with an age-dependent defect in adaptive immunity, during RSV and influenza infection of infants, few CD4 ϩ or CD8 ϩ T cells or CD56 ϩ NK cells are found in the upper and lower airways [100, 103, 104] , in contrast to the observed infiltration of lymphocytes in adult A/H5N1 influenza [106] and SARS CoV infections [196, 197] . Moreover, fatal cases of A(H1N1)pdm09 had increased numbers of CD8 ϩ T cells and granzyme B ϩ cells in their lungs [33] . This observation may represent a correlation with higher levels of viral antigen or may indicate a pathogenic role of exuberant CD8 ϩ T cell responses. On the other hand, the elderly have been shown to have reduced CTL activity compared with younger individuals, and this can have a negative impact on immunization efficacy [111] [112] [113] . However, prevaccination T cell responses do not correlate with postvaccination antibody titers [114] , and as the elderly also have decreased antibody responses following vaccination, T cell responses may provide a better correlate of vaccine efficacy in the elderly population [115] . Interestingly, unstimulated memory T cells from older individuals are in a higher state of differentiation than those of younger individuals, and it was shown that fewer senescent, influenza-specific T cells, characterized as killer cell lectin-like receptor subfamily G member 1 (KLRG1) hi CD57 hi , are associated with a more robust antibody response following TIV delivery [110] . The immunodominance hierarchy in HLA-A*02 individuals is often reported as directed primarily against the conserved influenza A M1 58 -66 (GILGFVFTL) [198, 199] . However, studies suggest that although this CD8 ϩ T cell response to M1 58 -66 is conserved in HLA-A*02 individuals, it may not be strictly immunodominant, as greater responses were observed against internal NP and polymerase PB1 subunit proteins in some individuals [122, 200] . In terms of HLA presentation of influenza peptides, the HLA-A*02:01-restricted M1 58 -66 peptide-specific CTL response is characterized by expression of V ␤ 17 TCR use [201] , and in HLA-A*02:01 individuals, V ␤ 17 ϩ CTLs were the dominant cells responding to in vitro influenza exposure. In contrast, cord blood from HLA-A*02:01 infants, thus having no prior influenza exposure, was characterized by a M1 58 -66 -specific CD8 ϩ T cell response that was less dependent on the presence of the CD8 ϩ V ␤ 17 ϩ T cell population [109] . Therefore, maturation of the CTL response upon subsequent influenza infections is likely related to effective clearance of viral infections and has important implications for therapeutic interventions. Although children beyond infancy who are considered high-risk for influenza infection, i.e., children with underlying diseases, such as asthma, sickle cell disease, and those receiving solid organ transplantation, have sig-nificant increases in serum nAb and T cell responses, children with systemic lupus erythematosus are unable to mount significant T cell responses following TIV immunization [108] . These findings suggest that the pathogenesis of respiratory viral infections in infants may be associated with the absence of immunological memory or a failure to develop an appropriate, well-regulated T cell response. Undoubtedly, host genetics have a role in the outcome of many infectious diseases, including influenza. Mouse models have provided some clues regarding the role of the host genetic component on the inflammatory response in the respiratory tract. Briefly, these studies found that influenza-induced pathology is altered depending on the inbred mouse strain used for infection. The genetic background of the host also controls cumulative and maximal viral titer, underscoring the importance of limiting viral replication early during influenza infection [111, 202] . In humans, certain populations worldwide have been found to be more prone to severe disease during influenza infection. For instance, Aboriginal peoples have more severe illness and increased mortality during the A/H1N1 pandemics of 1918 and 2009 compared with those of non-Aboriginal descent [29, 116 -118] . It is important to note, however, that poor socioeconomic conditions and metabolic diseases, such as diabetes, are more common in indigenous populations [203] , and this may contribute to influenza-related disease. Moreover, HLA alleles are proposed to have some role during influenza infection, as differential CTL responses exist in individuals with varying HLA alleles [204] . Gene polymorphisms located in human chromosomes 1 and 17 were also determined to be associated with severe pneumonia during A(H1N1)pdm09 infection [120] . Interestingly, two of these SNPs are mapped to genes FCGR2A and C1QBP. The protein encoded by FCGR2A is involved in phagocytosis and clearance of immune complexes and expressed on the surface of phagocytic cells. The protein encoded by C1QBP is a critical factor involved in complement activation. Notably, presence of low-avidity antibodies and subsequent formation of immune complex-mediated complement activation in the lungs of patients with severe influenza infection are associated with severe disease following influenza infection [107] . Recently, "systems biology" strategies have emerged to improve vaccine efficacy, measuring expression patterns to expose protective molecular signatures following immunization or natural infection [46, 205] . Confirming in vitro mouse studies, which suggest that MBL can neutralize influenza [206, 207] , human polymorphisms identified within MBL2 are associated with poor antibody responses to influenza vaccination [119] . Although MBL deficiency has been observed to influence the outcome in several respiratory diseases, including pneumococcus infection, tuberculosis, and SARS CoV (reviewed in ref. [208] ), it did not have an obvious role in A(H1N1)pdm09 infection of naïve individuals [209] . These studies highlight the involvement of host ge-netics in development of enhanced disease, although they likely represent a small fraction of the potential regulatory host genetic elements. Studies of innate and adaptive immune responses to influenza virus infection have been restricted primarily to animal models of disease and inflammation. As our immunological tools become more sophisticated, more emphasis should be placed on human immunology as opposed to relying solely on animal infection models. Moreover, the vast majority of experimental data in humans is restricted to blood leukocytes, with minimal emphasis placed on the innate immune response, particularly at sites of infection. As a result, there is limited scientific knowledge regarding the specific roles of immune cells in affected tissues. Understanding the innate response in particular is critical to appreciate early determinants of pathogenicity and disease outcome during influenza infection. Influenza Prevention and Control Recommendations: Recommendations of the Advisory Committee on Immunization Practices (ACIP). CDC Influenza A and B virus infections in children Burden of influenza in children in the community The underrecognized burden of influenza in young children The effect of influenza on hospitalizations, outpatient visits, and courses of antibiotics in children Children hospitalized with 2009 novel influenza A(H1N1) in California Age-specific differences in influenza A epidemic curves: do children drive the spread of influenza epidemics? Estimating household and community transmission parameters for influenza Risk factors of influenza transmission in households Burden of interpandemic influenza in children younger than 5 years: a 25-year prospective study Wheezing, asthma, and pulmonary dysfunction 10 years after infection with respiratory syncytial virus in infancy Respiratory syncytial virus bronchiolitis in infancy is an important risk factor for asthma and allergy at age 7 Severe respiratory syncytial virus bronchiolitis in infancy and asthma and allergy at age 13 Influenza-associated hospitalizations in the United States Epidemiology of viral respiratory infections Estimates of deaths associated with seasonal influenza-United States Global burden of respiratory infections due to seasonal influenza in young children: a systematic review and metaanalysis The annual impact of seasonal influenza in the US: measuring disease burden and costs High costs of influenza: direct medical costs of influenza disease in young children Standardization of terminology of the pandemic A(H1N1) Severe respiratory disease concurrent with the circulation of H1N1 influenza Pneumonia and respiratory failure from swine-origin influenza A (H1N1) in Mexico Cross-reactive antibody responses to the 2009 pandemic H1N1 influenza virus Risk factors for severe outcomes follow Novel influenza A (H1N1) virus infections in three pregnant women-United States Severe 2009 H1N1 influenza in pregnant and postpartum women in California Correlates of severe disease in patients with 2009 pandemic influenza (H1N1) virus infection Insights into the interaction between influenza virus and pneumococcus Community-acquired pneumonia due to pandemic A(H1N1)2009 influenzavirus and methicillin resistant Staphylococcus aureus co-infection Bacterial coinfections in lung tissue specimens from fatal cases of 2009 pandemic influenza A (H1N1)-United States Lung pathology in fatal novel human influenza A (H1N1) infection Cross-recognition of avian H5N1 influenza virus by human cytotoxic T-lymphocyte populations directed to human influenza A virus Prevention and control of influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices (ACIP) Assessment of immunity to influenza using artifical challenge of normal volunteers with influenza virus Serum and nasal wash antibodies associated with resistance to experimental challenge with influenza A wild-type virus Safety of high doses of influenza vaccine and effect on antibody responses in elderly persons Safety and immunogenicity of a high dosage trivalent influenza vaccine among elderly subjects Parenteral vaccination against influenza does not induce a local antigen-specific immune response in the nasal mucosa An early humoral immune response in peripheral blood following parenteral inactivated influenza vaccination Systemic and mucosal immune responses in young children and adults after parenteral influenza vaccination Comparison of the influenza virus-specific effector and memory B-cell responses to immunization of children and adults with live attenuated or inactivated influenza virus vaccines Correlates of immune protection induced by live, attenuated, cold-adapted, trivalent, intranasal influenza virus vaccine Live attenuated versus inactivated influenza vaccine in infants and young children Systems biology of vaccination for seasonal influenza in humans Development and persistence of local and systemic antibody responses in adults given live attenuated or inactivated influenza A virus vaccine Inactivated influenza virus vaccines in children Herd immunity in adults against influenza-related illnesses with use of the trivalent-live attenuated influenza vaccine (CAIV-T) in children Effectiveness of influenza vaccination of children with recurrent respiratory tract infections in reducing respiratory-related morbidity within the households The Japanese experience with vaccinating schoolchildren against influenza Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis Mortality benefits of influenza vaccination in elderly people: an ongoing controversy Influenza vaccination and mortality: differentiating vaccine effects from bias Effect of influenza vaccination on hospitalizations in persons aged 50 years and older FluVaxView: Influenza Vaccination Coverage Influenza vaccine: the challenge of antigenic drift Emulsion-based adjuvants for improved influenza vaccines The adjuvanted influenza vaccines with novel adjuvants: experience with the MF59-adjuvanted vaccine Trial of 2009 influenza A (H1N1) monovalent MF59-adjuvanted vaccine MF59 is a safe and potent vaccine adjuvant that enhances protection against influenza virus infection MF59-adjuvanted influenza vaccine (FLUAD) in children: safety and immunogenicity following a second year seasonal vaccination Enhanced immunogenicity of seasonal influenza vaccines in young children using MF59 adjuvant. Pediatr Oil-in-water emulsion adjuvant with influenza vaccine in young children Potent immunogenicity and efficacy of a universal influenza vaccine candidate comprising a recombinant fusion protein linking influenza M2e to the TLR5 ligand flagellin Universal M2 ectodomain-based influenza A vaccines: preclinical and clinical developments Safety and immunogenicity of a recombinant M2e-flagellin influenza vaccine (STF2.4xM2e) in healthy adults A highly conserved neutralizing epitope on group 2 influenza A viruses A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins Broadly neutralizing human antibody that recognizes the receptor-binding pocket of influenza virus hemagglutinin Antibody recognition of a highly conserved influenza virus epitope Structural and functional bases for broadspectrum neutralization of avian and human influenza A viruses A broadly neutralizing human monoclonal antibody that recognizes a conserved, novel epitope on the globular head of the influenza H1N1 virus hemagglutinin Investigation of volunteers infected with the influenza virus Comparison of long-term systemic and secretory antibody responses in children given live, attenuated, or inactivated influenza A vaccine Immunity to influenza A virus infection in young children: a comparison of natural infection, live cold-adapted vaccine, and inactivated vaccine Use of live cold-adapted influenza A H1N1 and H3N2 virus vaccines in seropositive adults New research on influenza: studies with normal volunteers. Combined Clinical Staff Conference at the National Institutes of Health Immune correlates of protection against influenza in the human challenge model Potential role of human challenge studies for investigation of influenza transmission Partial protection of seasonal trivalent inactivated vaccine against novel pandemic influenza A/H1N1 2009: case-control study in Mexico City Seasonal influenza vaccine and protection against pandemic (H1N1) 2009-associated illness among US military personnel Protective efficacy of seasonal influenza vaccination against seasonal and pandemic influenza virus infection during 2009 in Hong Kong Prior H1N1 influenza infection and susceptibility of Cleveland Family Study participants during the H2N2 pandemic of 1957: an experiment of nature Cross-subtype protection in humans during sequential, overlapping, and/or concurrent epidemics caused by H3N2 and H1N1 influenza viruses Kinetics of viral shedding and immune responses in adults following administration of cold-adapted influenza vaccine Heterosubtypic neutralizing antibodies are produced by individuals immunized with a seasonal influenza vaccine Broadly cross-reactive antibodies dominate the human B cell response against 2009 pandemic H1N1 influenza virus infection Neutralizing antibodies derived from the B cells of 1918 influenza pandemic survivors Naturally occurring human monoclonal antibodies neutralize both 1918 and 2009 pandemic influenza A (H1N1) viruses Memory T cells established by seasonal human influenza A infection cross-react with avian influenza A (H5N1) in healthy individuals Cutting edge: CD4 T cells generated from encounter with seasonal influenza viruses and vaccines have broad protein specificity and can directly recognize naturally generated epitopes derived from the live pandemic H1N1 virus In vitro responses to avian influenza H5 by human CD4 T cells Assessment of seasonal influenza A virus-specific CD4 T-cell responses to 2009 pandemic H1N1 swine-origin influenza A virus Local and systemic cytokine responses during experimental human influenza A virus infection. Relation to symptom formation and host defense Re-emergence of fatal human influenza A subtype H5N1 disease Interleukin 8 (IL-8) in the bronchoalveolar lavage fluid from patients with the adult respiratory distress syndrome (ARDS) and patients at risk for ARDS A comparison of cytokine responses in respiratory syncytial virus and influenza A infections in infants Differential production of inflammatory cytokines in primary infection with human metapneumovirus and with other common respiratory viruses of infancy Severe human lower respiratory tract illness caused by respiratory syncytial virus and influenza virus is characterized by the absence of pulmonary cytotoxic lymphocyte responses A comparison of epidemiologic and immunologic features of bronchiolitis caused by influenza virus and respiratory syncytial virus Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia Analysis of cells obtained by bronchial lavage of infants with respiratory syncytial virus infection Leucocyte populations in respiratory syncytial virus-induced bronchiolitis Chemokine receptor expression and chemotactic responsiveness of human monocytes after influenza A virus infection Avian influenza A (H5N1) infection in humans Humoral and cell-mediated immune responses to monovalent 2009 influenza A/H1N1 and seasonal trivalent influenza vaccines in high-risk children Influenza A antigen exposure selects dominant V␤17ϩ TCR in human CD8ϩ cytotoxic T cell responses Influenza-specific T cells from older people are enriched in the late effector subset and their presence inversely correlates with vaccine response Influenza A virus specific T cell immunity in humans during aging Influenza vaccine responses in older adults Role of humoral and cell-mediated immunity in protection from influenza disease after immunization of healthy elderly Discordance between antibody and T cell responses in recipients of trivalent inactivated influenza vaccine T cell responses are better correlates of vaccine protection in the elderly The 2009 pandemic H1N1 influenza and indigenous populations of the Americas and the Pacific Critical care services and 2009 H1N1 influenza in Australia and New Zealand Public Health Weekly Reports for Host single-nucleotide polymorphisms and altered responses to inactivated influenza vaccine Genetic variants associated with severe pneumonia in A/H1N1 influenza infection Human memory CTL response specific for influenza A virus is broad and multispecific HLA class I molecules consistently present internal influenza epitopes Immunity and immunopathology to viruses: what decides the outcome? Role of innate immune cells and their products in lung immunopathology Interactions between innate antiviral and atopic immunoinflammatory pathways precipitate and sustain asthma exacerbations in children Influenza virus evades innate and adaptive immunity via the NS1 protein Pandemic H1N1 2009 influenza A virus induces weak cytokine responses in human macrophages and dendritic cells and is highly sensitive to the antiviral actions of interferons Involvement of Toll-like receptor 3 in the immune response of lung epithelial cells to double-stranded RNA and influenza A virus Dendritic cells respond to influenza virus through TLR7-and PKR-independent pathways Recognition of single-stranded RNA viruses by Toll-like receptor 7 Interferon renders epithelial cells of the respiratory and gastrointestinal tracts resistant to viral infections Evidence for a protective role of pulmonary surfactant protein D (SP-D) against influenza A viruses Mechanism of binding of surfactant protein D to influenza A viruses: importance of binding to haemagglutinin to antiviral activity Surfactant protein D enhances clearance of influenza A virus from the lung in vivo Absence of SP-A modulates innate and adaptive defense responses to pulmonary influenza infection Effects of influenza A virus on human neutrophil calcium metabolism The peptide antibiotic LL-37/hCAP-18 is expressed in epithelia of the human lung where it has broad antimicrobial activity at the airway surface The human antimicrobial and chemotactic peptides LL-37 and ␣-defensins are expressed by specific lymphocyte and monocyte populations Antiviral activity and increased host defense against influenza infection elicited by the human cathelicidin LL-37 Interaction of human defensins with Escherichia coli. Mechanism of bactericidal activity Elevated levels of ␣-defensins in plasma and BAL fluid of patients with active pulmonary tuberculosis Monocytechemotactic activity of defensins from human neutrophils Mechanisms of anti-influenza activity of surfactant proteins A and D: comparison with serum collectins Direct inactivation of viruses by human granulocyte defensins ␣-Defensin inhibits influenza virus replication by cell-mediated mechanism(s) Blood monocytes consist of two principal subsets with distinct migratory properties The proinflammatory CD14ϩCD16ϩDRϩϩ monocytes are a major source of TNF The novel subset of CD14ϩ/CD16ϩ blood monocytes is expanded in sepsis patients Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors Differential regulation of Toll-like receptor-2, Toll-like receptor-4, CD16 and human leucocyte antigen-DR on peripheral blood monocytes during mild and severe dengue fever Identification of a novel dendritic cell-like subset of CD64(ϩ)/CD16(ϩ) blood monocytes Characterization of myeloid and plasmacytoid dendritic cells in human lung Differential recruitment of dendritic cells and monocytes to respiratory mucosal sites in children with influenza virus or respiratory syncytial virus infection The multifunctional NS1 protein of influenza A viruses Influenza A virus lacking the NS1 gene replicates in interferon-deficient systems Species-specific antagonism of host ISGylation by the influenza B virus NS1 protein Monocyte-mediated defense against microbial pathogens Contrasting effects of CCR5 and CCR2 deficiency in the pulmonary inflammatory response to influenza A virus Chemokine regulation of the inflammatory response to a low-dose influenza infection in CCR2Ϫ/Ϫ mice TNF/iNOS-producing dendritic cells are the necessary evil of lethal influenza virus infection Fatal outcome of pandemic H1N1 2009 influenza virus infection is associated with immunopathology and impaired lung repair, not enhanced viral burden in pregnant mice Alveolar epithelial cells direct monocyte transepithelial migration upon influenza virus infection: impact of chemokines and adhesion molecules CXCR2 is required for neutrophil recruitment to the lung during influenza virus infection, but is not essential for viral clearance Type I interferon signaling regulates Ly6C monocytes and neutrophils during acute viral pneumonia in mice nate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus Influenza A virus accelerates neutrophil apoptosis and markedly potentiates apoptotic effects of bacteria Programmed cell death (apoptosis) in human monocytes infected by influenza A virus Effect of virus infections on polymorph function in children Depressed monocyte chemotaxis during acute influenza infection Effects of infection with influenza virus on the function of polymorphonuclear leukocytes Differences in serum cytokine levels between influenza virus A and B infections in children Cytokine induction in human cord blood lymphocytes after pulsing with UV-inactivated influenza viruses Experiments to determine mode of spread of influenza Clinical and immunological characteristics of patients with 2009 pandemic influenza A (H1N1) virus infection after vaccination The role of the antibody response in influenza virus infection Antigenic fingerprinting of H5N1 avian influenza using convalescent sera and monoclonal antibodies reveals potential vaccine and diagnostic targets Pre-existing immunity against swine-origin H1N1 influenza viruses in the general human population Human dendritic cell deficiency: the missing ID? Influenza virus-infected dendritic cells stimulate strong proliferative and cytolytic responses from human CD8ϩ T cells Plasmacytoid dendritic cells activated by influenza virus and CD40L drive a potent TH1 polarization Influenza A virus-induced IFN-␣/␤ and IL-18 synergistically enhance IFN-␥ gene expression in human T cells Selective accumulation of differentiated CD8ϩ T cells specific for respiratory viruses in the human lung CD8 T cells with an intraepithelial phenotype upregulate cytotoxic function upon influenza infection in human lung Characterization of the human CD8ϩ T cell response following infection with 2009 pandemic influenza H1N1 virus Long term immune responses to pandemic influenza A/H1N1 infection in solid organ transplant recipients CD8ϩ T cell immunity to 2009 pandemic and seasonal H1N1 influenza viruses Cytotoxic T lymphocytes established by seasonal human influenza cross-react against 2009 pandemic H1N1 influenza virus Crossreactive CD8ϩ T-cell immunity between the pandemic H1N1-2009 and H1N1-1918 influenza A viruses ␥␦ T cell effector functions: a blend of innate programming and acquired plasticity Phosphoantigen-expanded human ␥␦ T cells display potent cytotoxicity against monocyte-derived macrophages infected with human and avian influenza viruses Type 1 responses of human V␥9V␦2 T cells to influenza A viruses Age at first viral infection determines the pattern of T cell-mediated disease during reinfection in adulthood The enhancement or prevention of airway hyperresponsiveness during reinfection with respiratory syncytial virus is critically dependent on the age at first infection and IL-13 production Exposure of neonates to respiratory syncytial virus is critical in determining subsequent airway response in adults Inchoate CD8ϩ T cell responses in neonatal mice permit influenza-induced persistent pulmonary dysfunction Severe acute respiratory syndrome complicated by spontaneous pneumothorax A major outbreak of severe acute respiratory syndrome in Hong Kong The magnitude and specificity of influenza A virus-specific cytotoxic T-lymphocyte responses in humans is related to HLA-A and -B phenotype The minimum peptide epitope from the influenza virus matrix protein. Extra and intracellular loading of HLA-A2 Systematic identification of immunodominant CD8ϩ T-cell responses to influenza A virus in HLA-A2 individuals Human HLA-A0201-restricted cytotoxic T lymphocyte recognition of influenza A is dominated by T cells bearing the V beta 17 gene segment Host genetic variation affects resistance to infection with a highly pathogenic H5N1 influenza A virus in mice Diabetes prevalence among American Indians and Alaska Natives and the overall population-United States Preferential HLA usage in the influenza virus-specific CTL response Systems vaccinology: learning to compute the behavior of vaccine induced immunity Complement-dependent neutralization of influenza virus by a serum mannose-binding lectin Human mannan-binding lectin inhibits the infection of influenza A virus without complement Mannose-binding lectin deficiency and respiratory tract infection No association between mannose-binding lectin deficiency and H1N1 2009 infection observed during the first season of this novel pandemic influenza virus The authors are funded in part by U.S. National Institutes of Health/National Institute of Allergy and Infectious Diseases contracts HHSN266200700005C (St. Jude Center of Excellence for Influenza Research and Surveillance) and HHSN272200800058C (Systems Influenza) and the American Lebanese Syrian Associated Charities (ALSAC).