Influenza viruses continue to be a major global health threat. Severity and clinical outcome of influenza disease is determined by both viral and host factors. Viral factors have long been the subject of intense research and many molecular determinants have been identified. However, research into the host factors that protect or predispose to severe and fatal influenza A virus infections is lagging. The goal of this review is to highlight the recent insights into host determinants of influenza pathogenesis.

Influenza viruses are segmented negative-sense RNA viruses that belong to the Orthomyxoviridae family. Four types (A, B, C, and D) of influenza virus are known, but only types A and B can cause annual epidemics in humans. Influenza A virus (IAV) is divided into different subtypes based on the surface expression of the two surface glycoproteins: hemagglutinin (HA) and neuraminidase (NA). Each year ∼3–5 million people suffer from severe influenza infection, and of those, ∼250,000–500,000 succumb to the disease. The pathogenesis following IAV infection occurs in two phases. The first phase lasts between 1 and 3 days and determines the peak virus titer as well as the amount of inflammation associated with that. Depending on these two parameters, the second phase may lead to control of the virus or result in severe disease associated with acute respiratory distress syndrome and death. The clinical course and outcome of influenza pathogenesis is determined by both viral and host factors. T and B cell immunity against IAV is a key factor in protection from infection and disease. However, other host factors, such as age, sex, microbiome, and genetic variation, also modulate the clinical course and outcome of the disease. In this review, we will summarize the current literature for each of these host factors and discuss future studies for the field.

Adaptive immune responses play a key role in protection against IAV infection and disease (previously reviewed in Ref. 16). The humoral immune response can be broadly divided into virus neutralizing and nonneutralizing Abs. Neutralizing Abs target the HA protein of IAV that is required for attachment to and entry into cells. The mechanism of neutralizing activity falls into four major categories: blocking binding to sialic acids, inhibiting viral fusion, preventing release of progeny virus, and blocking proteolytic cleavage of the HA protein (7). For reasons that are not entirely clear, the majority of HA-specific Abs target the head domain of HA and block binding to sialic acids (5). Unfortunately, this region is also the most variable part of the protein and amenable to extensive amino acid substitution. Changes in the antigenic sites of the HA protein allows the virus to escape pre-existing Abs and reinfect individuals previously exposed to IAV. This phenomenon of immune escape is called antigenic drift. The stalk region of the HA protein is significantly more conserved, and Abs targeting this region often cross-react with other HA proteins, providing potential protection against antigenically divergent strains of IAV. B cells also produce nonneutralizing Abs that target the NA or matrix 2 (M2) protein. These Abs promote uptake of virus particles or virus-infected cells by macrophages and neutrophils and induce Ab-dependent cellular toxicity. Intriguingly, the baseline NA inhibitory Ab titer correlated more significantly with all disease severity metrics and had a stronger independent effect on outcome compared with the HA inhibitory Ab titer (8), suggesting that future universal vaccines should target both HA and NA protein on the surface of the virion. In addition to B cells, T cells also protect against severe IAV disease. CD4+ T cells provide the necessary T helper activity to generate high-affinity Abs, whereas CD8+ T cells, or cytotoxic T cells, can kill IAV-infected cells. After IAV infection, a fraction of the T cells remains at the site of infection and become tissue resident memory T cells (TRM) (9, 10). Like most T cells, TRM confer protection against infection with a heterologous strain of IAV (11, 12), such as during an IAV pandemic. In mice, TRM also prevent dissemination of IAV from the upper respiratory tract to the lungs (13). Thus, future IAV vaccines, including the universal influenza vaccine, should stimulate cross-reactive Abs as well as CD4+ and CD8+ T cells to protect against current and emerging strains of IAV.

Influenza infections cause high morbidity and mortality rates in the elderly and very young populations. Individuals >65 y of age are the most vulnerable to severe illness from influenza infection and account for ∼90% of all influenza and influenza-related deaths (14). During influenza epidemics, individuals >65 y are 4.5 times more likely to suffer from severe disease and nearly three times more likely to die as a result of the infection (15). Despite this, age-related changes in immune responses to influenza infection have not been fully explored (16). Immunosenescence is the gradual deterioration of the immune system that is caused by aging and results in increased susceptibility to viral and bacterial infections as well as attenuated vaccination responses (17, 18). Immunosenescence affects both the innate and adaptive immune response to infection. Monocytes from older human donors show reduced RIG-I like receptor signaling and significantly impaired type I IFN responses to IAV infection despite preserved inflammatory cytokine production. This defect is associated with dampened upregulation of antiviral IFN-stimulated genes and a corresponding increase in IAV gene expression (19). In concordance, ex vivo studies of plasmacytoid dendritic cells (DC) and monocyte-derived DC from older human donors show impaired IFN-α production in response to IAV (2022). Besides alterations in antiviral immune signaling, immunosenescence in innate immune cells is associated with reduced superoxide generation and phagocytosis in neutrophils and macrophages, reduced TLR expression and function in DC and macrophages, and increased PGE2 production in macrophages (reviewed in Ref. 23). Intriguingly, PGE2 is reported to inhibit type I IFN induction as well as Ag presentation and apoptosis, suggesting a possible mechanistic link between aging, PGE2 production, and attenuated innate immune cell function (24). The effects of aging on Ag presentation by DC are unclear (25). Some studies indicated that T cell priming by DC is maintained in the aging population (2628), whereas others show that it is impaired (2932). Despite the general decrease in innate immune cell function, aging is associated with an increase in inflammatory cytokine production; a condition described as senescence-associated secretory phenotype (19, 33). Senescence-associated secretory phenotype in turn may contribute to inflammaging, which is chronic, sterile, low-grade inflammation that contributes to the pathogenesis of age-related diseases (34). The basal proinflammatory environment associated with aging, combined with delayed innate immune responses following IAV infection of elderly individuals, leads to excess immunopathology, tissue damage, more persistent symptoms, and a higher likelihood of progression to acute respiratory distress syndrome (23, 3538).

The waning of the adaptive immune response has been extensively studied in aging research. At puberty, thymic involution leads to a reduction in the output of naive T cells, which becomes more profound as an individual gets older. This results in decreased numbers of naive T cells in the periphery of elderly individuals, restricts the ability of the adaptive immune system to mount a defense against new pathogens, and importantly, decreases responses to vaccination (39, 40). Aging is also associated with a clonal expansion of T cells because of repeated antigenic stimulation by CMV (41). Clonal expansion further diminishes the response to novel Ags. Additionally, Th cell function, overall B cell numbers, and Ab efficacy against specific pathogens also decline with age (40, 42, 43).

Murine studies also support age-related increases in IAV susceptibility that have been associated with elevated virus titers, prolonged weight loss, and altered cytokine dynamics. A reduction in the numbers and activity of virus-specific CD8+ T cells in aged mice is one of the factors that contribute to a higher virus load and severe disease. The rationale for this age-dependent decrease in T cell immunity is not well understood. Aging is also associated with a decline in T cell repertoire diversity, leading to impaired immunity to IAV in mice (44). Combined, these factors lead to prolonged virus replication and disease in aged animals (38). Similar to humans, aged mice also have lower Ab responses to vaccination than young adult mice (45). Overall, these studies indicate that an aged mouse model is a suitable model to study the role of aging on IAV pathogenesis. Specifically, separating the effects of aging from other host factors, such as changes in sex hormone levels or microbiome (see below), is key to understanding mechanistically how aging and inflammaging increases susceptibility to IAV disease. These new insights may help to identify at-risk populations as well as develop future therapies aimed at reducing the impact of immunosenescence.

The impact of sex differences on morbidity and mortality after IAV infection is influenced by multiple behavioral, environmental, and social factors. Studies comparing the male and female susceptibilities and disease outcomes after IAV infection have reported widely varied results, with some suggesting that young males are more susceptible whereas others suggest that females are at an increased risk for severe and fatal disease. Outcomes of IAV infection are also complicated by age, immunological and vaccine responses, the specific strain of IAV, and seasonal versus pandemic influenza infection (reviewed in Ref. 4650).

Females of reproductive age displayed a higher incidence of severe disease following avian H5N1 and H7N9 IAV infection and pandemic IAV infections, yet young (prepubescent) and elderly males experience more severe disease following seasonal IAV infection. In addition, female mice consistently show greater reductions in body mass, temperature, and survival as compared with males when infected with IAV. The increase in IAV disease in female mice strongly correlates with increased cytokine and chemokine production (5154). These data underscore the intertwined environment that may be further exacerbated in females of reproductive age (premenopause) by increased sex hormones. In contrast, IAV disease in prepubescent and elderly males, who display poor vaccination and adaptive immune responses, may be exacerbated by decreased testosterone levels.

Disease outcomes in males and females are greatly influenced by the expression of respective sex hormones. Reducing testosterone levels in young mice by gonadectomy leads to increased morbidity, clinical illness, and pulmonary pathologic condition, despite having no effect on viral replication (55, 56). Treatment of aged males with testosterone improved survival following infection but, once again, did not affect viral replication, suggesting that increased levels of testosterone protect against detrimental outcomes following IAV infection. In humans, reduced endogenous testosterone levels correlated with lower responses to influenza vaccination (57). In female mice, exogenous estrogen protected against infection-induced morbidity and mortality (53, 58, 59). Protection was associated with dampening of the proinflammatory immune response and tissue damage. However, other studies have shown that the anti-inflammatory effects of elevated estrogen levels led to increased morbidity after IAV infection (60). These conflicting findings may be attributable to differences in viruses used or the amounts of exogenous estrogen used in treatment. It is known that low levels of estrogen stimulate inflammation, whereas high levels of estrogen are anti-inflammatory (61, 62). Like estrogen, the effects of progesterone on IAV disease outcome are also inconclusive. Progesterone treatment prior to infection reduced excessive pulmonary inflammation, improved lung function, and promoted lung epithelium repair and faster recovery in female mice following IAV infection (63), whereas in a second study, onset of and duration of morbidity was earlier and greater following progesterone treatment (64). Collectively, these data illustrate that sex hormones modulate the immune response and, therefore, the outcome after IAV infection. Future studies will determine if the disparity in susceptibility to IAV disease between male and females is caused by sex hormones or other sex-dependent host factors. Interestingly, sex hormones decline with age after puberty, suggesting that some of the age-related effects on IAV disease maybe be due to lower levels of estrogen and testosterone, resulting in more inflammation in aging individuals. The underlying mechanism of immune modulation by sex hormones and the role of specific host genes is not well understood. Future studies should identify specific transcriptional networks, regulators, and effectors that regulate these sex-dependent differences in immunity and influenza pathogenesis.

Pregnant women are recognized to be at higher risk for IAV disease, and this became more apparent during the 2009 H1N1 pandemic. Pregnant women were about seven times more likely to suffer from severe IAV disease and two times more likely to die of IAV than were nonpregnant women. In the United States, pregnant women account for 5% of all IAV-related deaths. However, underlying pathways that cause susceptibility of pregnant women and predispose them to severe disease remain unidentified. In infected pregnant mice, the production of antiviral molecules and inflammatory cytokines such as type I IFNs was reduced and accompanied by a lack of innate immune response activation following infection with pandemic H1N1 IAV (65). This corroborates studies wherein infection of pregnant BALB/c mice with pandemic H1N1 IAV resulted in higher mortality and more severe histological lesions associated with dampened tissue repair compared with nonpregnant mice (6668). Aside from developing better virus vaccines, more effort should go into understanding the underlying causes (i.e., increased virus replication, altered inflammatory response, or delayed virus control and tissue repair) that predispose pregnant women to more severe disease after IAV infection.

The role of the human microbiome on influenza infection and associated disease is currently not known but is an up-and-coming area of investigation. Does IAV infection alter the microbiome of the respiratory tract or other organs, and is there an effect of this perturbation? Similarly, does an altered respiratory or gut microbiome predispose to virus infection and severe disease? For example, colonization with certain pathogenic bacteria such as Staphylococcus or Streptococcus may affect the risk of secondary bacterial infections following primary IAV disease (69).

Most studies on the microbiome have been done in mice. The earliest studies showed that antibiotic treatment predisposes to severe IAV disease, likely through changes in IFN and TLR signaling (7072). The gut microbiome also supported a robust vaccine response in mice (73). More recently, it was shown that the bacterial metabolite desaminotyrosine protects from severe IAV through production of type I IFN (74). Gut microbiota from wild mice transferred into laboratory mice also increased survival after IAV infection (75). The mice that received wild-derived microbiota were significantly more resistant to lethal IAV infection compared with control mice or mice that received microbiota from laboratory mice. Resistance to severe disease was associated with reduced early weight loss, lower viral titers, and less inflammation. In future studies, it will be important to identify the component of the wild mouse microbiome (i.e., bacteria, virus, or other organisms) that increases resistance to severe IAV infection and to determine whether similar factors that confer protection against severe IAV disease exist in the human microbiome. There are many important and unanswered questions in the field regarding the relationship between microbiome and IAV. How does the microbiome, and its perturbations, impact human IAV disease? How does the microbiome protect against IAV, and what is the impact of viral and other host factors on microbiome-mediated protection against severe IAV disease?

During the 2009 H1N1 pandemic, obesity was recognized as a risk factor (odd ratio between 2 and 4) for complications from IAV infection (7680). Since then, the impact of obesity on severe IAV disease is diminished (81, 82) or not present (83, 84). Perhaps the emergence of pre-existing immunity to the 2009 H1N1 IAV in obese and lean individuals masks the detrimental effects of obesity on IAV disease. Obesity is also an important risk factor for severe IAV disease after H7N9 IAV infection (15, 85, 86). There is growing evidence that obesity affects the effectiveness of the adaptive immune system following IAV infection or vaccination. T cells from influenza-vaccinated obese adults are less activated when stimulated with vaccine strains of influenza (87, 88). Ex vivo, CD4+, and CD8+ T cells from overweight and obese individuals expressed lower levels of CD69, CD28, and CD40L, as well as the effector molecules IFN-γ and granzyme B, suggesting deficiencies in activation following stimulation with IAV (87). Also, vaccinated obese adults were twice as likely to report influenza infection and influenza-like illness than their healthy weight counterparts, despite similarly robust serological responses (89). A higher body mass index also correlated with the prolonged shedding of infectious virus from IAV-infected individuals (90, 91).

In animal models, diet-induced obese mice infected with IAV have increased mortality, greater lung inflammation and damage, higher numbers of cytotoxic CD8+ T cells in the lungs, and fewer suppressive T regulatory cells when compared with lean mice (92, 93). In secondary challenge studies, obese mice had impaired adaptive responses as indicated by decreased memory CD8+ T cells and production of IFN-γ. Additionally, obese mice had higher mortality following vaccination and challenge despite increased production of neutralizing and nonneutralizing IAV-specific Abs, similar to observations in humans (94). These animal models capture the influence of diet-induced obesity on IAV morbidity, and models of genetic predisposition to obesity also show similar outcomes. Compared with lean mice, global leptin receptor-deficient mice (ob/ob) and hypothalamic leptin receptor-knockout (LepRH−/−) mice display increased mortality, higher lung inflammation, and decreased viral clearance when infected with pandemic H1N1 IAV (92, 95). In contrast, mice depleted of leptin receptor specifically in macrophages and lung epithelial cells had similar outcomes compared with wild-type mice following IAV infection. This may be due to a critical role of leptin in other cell types, such as T cell metabolic or glycolytic activity and adaptive immunity (95). Interestingly, obesity is associated with a change in the gut microbiome composition in both mice and humans (9698). Future studies should determine how each of these two host factors independently and in combination affect IAV disease.

Considerable progress has been made with the identification of human genetic polymorphisms associated with severe or fatal IAV disease. After the first human genetic association study, performed shortly after the 1957 (H2N2) IAV pandemic (99, 100), it took nearly 40 y before the impact of host genetic variation on IAV disease was considered again (101103). The identification of host genetic factors associated with clinical outcome of IAV infection in humans is often complicated by the presence of pre-existing immunity in the population. Therefore, many of the host genetic factors identified to date were discovered during the 2009 H1N1 IAV pandemic or in infants and young children, who are immunologically naive for IAV. Currently, there are approximately 25 different host genes, whose genetic variation has been associated with the outcome after IAV infection in humans (Table I) (104). Some of these genes have been independently validated, providing strong evidence that the severity of IAV disease is genetically predisposed. This important observation allows for the identification of at-risk individuals who should be prioritized to receive one or more IAV vaccines annually and supports the discovery of additional host factors that modulate IAV outcome.

Table I.
Host genes associated with severe influenza virus in humans
Host GeneReported FindingsAnimal Studies
IFITM3 rs12252 was associated with severe IAV-H1H1 and IAV-H7N9 disease in some, but not all, cohorts (135144). rs34481144 was associated with reduced gene expression in CD8+ T cells and severe IAV disease (110). 
   
IRF7 Compound heterozygous null mutations in a single child who suffered life-threatening IAV infection (112
   
TMPRSS2 rs2070788 was associated with severe IAV-H1N1 disease in 162 cases and 247 controls (145). rs2070788 and rs383510 were associated with susceptibility to IAV-H7N9 in 102 patients and 106 heavily exposed controls (145). 
   
LGALS1 Identified in a single cohort of 102 H7N9 IAV patients and 106 heavily exposed poultry workers (146
   
SFTPA2 rs1965708 and rs1059046 were associated with severe disease in 93 patients infected with IAV-H1N1 (147). rs1965708 and rs1059046 were not associated with severe IAV-H1N1 disease in 320 infected patients and 115 controls (148). N/A 
   
SFTPB rs1130866 was associated with severe IAV-H1N1 disease in 380 patients (149). rs1130866 was not associated with severe IAV-H1N1 disease in 320 patients and 115 controls (148). − 
   
CD55 rs2564978 was associated with severe IAV-H1N1 disease in 425 patients (150) as well as severe IAV-H7N9 and IAV-H1N1 disease in 275 patients (137− 
   
C1QBP rs3786054 was associated with severe IAV-H1N1 disease in 91 patients and 98 household contacts (151). − 
   
FCGR2A rs1801274 was associated with severe IAV-H1N1 disease in 91 patients and 98 household contacts (151). rs1801274 was not associated with severe disease in 436 patients (55). N/A 
   
CPT2 Compound heterozygotes mutations were associated with increased risk for IAV-associated encephalopathy (115117− 
   
TNF rs909253 was associated with severe IAV-H1N1 disease in a cohort of 145 patients and 360 healthy contacts. Infection with IAV-H1N1 was associated with rs361525 and rs1800750 (152). 
   
IL1A rs17561 was associated with IAV-H1N1 infection in 167 patients and 192 controls (153− 
   
IL1B rs1143627 was associated with IAV-H1N1 infection in 167 patients and 192 controls (153). rs16944 and rs3136558 were associated with IAV-H1N1 infection in 145 patients and 360 asymptomatic healthy contacts (154). rs16944 was associated with reduced risk of IAV-H3N2 infection (155). − 
   
TLR3 rs5743313 was associated with death after IAV infection among 275 adults (137) and increased risk of pneumonia in children infected with IAV-H1N1 (156
   
KIR Killer-cell Ig-like receptors are associated with severe disease after IAV-H1N1 infection (157, 158N/A 
   
CCR5 CCR5∆32 was associated with higher mortality after IAV-H1N1 in 171 cases (159). CCR5∆32 was not associated with the risk of IAV-H1N1 infection or with severe disease in a cohort of 29 patients (160) and 330 patients (161). 
   
RPAIN rs8070740 was associated with severe IAV-H1N1 disease in 91 patients and 98 household contacts (151− 
   
DBR1 DBR1 mutations in unrelated patients result in brainstem infection with IAV (113− 
   
IL10 rs1800872 was associated with risk of infection with IAV-H3N2 (155). rs1800896 was associated with risk for IAV-related pneumonia (152). 
   
IL28 rs8099917 was associated with risk of ILI symptom after IAV-H3N2 (155− 
   
IL6 rs1818879 was associated with severe IAV-H1N1 infection in 145 patients and 360 asymptomatic healthy contacts (154
   
ST3GAL1 rs113350588 and rs1048479 were associated with increased risk of severe IAV-H1N1 disease among 356 subjects (162− 
   
IL17 rs2275913 was associated with risk of IAV-H3N2 infection in case-control study (155
   
NOS3 rs2070744 was associated with risk for IAV-related pneumonia (152− 
   
GATA2 Haplo-insufficiency of GATA2 was associated with severe IAV-H1N1 disease (163− 
Host GeneReported FindingsAnimal Studies
IFITM3 rs12252 was associated with severe IAV-H1H1 and IAV-H7N9 disease in some, but not all, cohorts (135144). rs34481144 was associated with reduced gene expression in CD8+ T cells and severe IAV disease (110). 
   
IRF7 Compound heterozygous null mutations in a single child who suffered life-threatening IAV infection (112
   
TMPRSS2 rs2070788 was associated with severe IAV-H1N1 disease in 162 cases and 247 controls (145). rs2070788 and rs383510 were associated with susceptibility to IAV-H7N9 in 102 patients and 106 heavily exposed controls (145). 
   
LGALS1 Identified in a single cohort of 102 H7N9 IAV patients and 106 heavily exposed poultry workers (146
   
SFTPA2 rs1965708 and rs1059046 were associated with severe disease in 93 patients infected with IAV-H1N1 (147). rs1965708 and rs1059046 were not associated with severe IAV-H1N1 disease in 320 infected patients and 115 controls (148). N/A 
   
SFTPB rs1130866 was associated with severe IAV-H1N1 disease in 380 patients (149). rs1130866 was not associated with severe IAV-H1N1 disease in 320 patients and 115 controls (148). − 
   
CD55 rs2564978 was associated with severe IAV-H1N1 disease in 425 patients (150) as well as severe IAV-H7N9 and IAV-H1N1 disease in 275 patients (137− 
   
C1QBP rs3786054 was associated with severe IAV-H1N1 disease in 91 patients and 98 household contacts (151). − 
   
FCGR2A rs1801274 was associated with severe IAV-H1N1 disease in 91 patients and 98 household contacts (151). rs1801274 was not associated with severe disease in 436 patients (55). N/A 
   
CPT2 Compound heterozygotes mutations were associated with increased risk for IAV-associated encephalopathy (115117− 
   
TNF rs909253 was associated with severe IAV-H1N1 disease in a cohort of 145 patients and 360 healthy contacts. Infection with IAV-H1N1 was associated with rs361525 and rs1800750 (152). 
   
IL1A rs17561 was associated with IAV-H1N1 infection in 167 patients and 192 controls (153− 
   
IL1B rs1143627 was associated with IAV-H1N1 infection in 167 patients and 192 controls (153). rs16944 and rs3136558 were associated with IAV-H1N1 infection in 145 patients and 360 asymptomatic healthy contacts (154). rs16944 was associated with reduced risk of IAV-H3N2 infection (155). − 
   
TLR3 rs5743313 was associated with death after IAV infection among 275 adults (137) and increased risk of pneumonia in children infected with IAV-H1N1 (156
   
KIR Killer-cell Ig-like receptors are associated with severe disease after IAV-H1N1 infection (157, 158N/A 
   
CCR5 CCR5∆32 was associated with higher mortality after IAV-H1N1 in 171 cases (159). CCR5∆32 was not associated with the risk of IAV-H1N1 infection or with severe disease in a cohort of 29 patients (160) and 330 patients (161). 
   
RPAIN rs8070740 was associated with severe IAV-H1N1 disease in 91 patients and 98 household contacts (151− 
   
DBR1 DBR1 mutations in unrelated patients result in brainstem infection with IAV (113− 
   
IL10 rs1800872 was associated with risk of infection with IAV-H3N2 (155). rs1800896 was associated with risk for IAV-related pneumonia (152). 
   
IL28 rs8099917 was associated with risk of ILI symptom after IAV-H3N2 (155− 
   
IL6 rs1818879 was associated with severe IAV-H1N1 infection in 145 patients and 360 asymptomatic healthy contacts (154
   
ST3GAL1 rs113350588 and rs1048479 were associated with increased risk of severe IAV-H1N1 disease among 356 subjects (162− 
   
IL17 rs2275913 was associated with risk of IAV-H3N2 infection in case-control study (155
   
NOS3 rs2070744 was associated with risk for IAV-related pneumonia (152− 
   
GATA2 Haplo-insufficiency of GATA2 was associated with severe IAV-H1N1 disease (163− 

N/A, not applicable because of ortholog not present in mice. +, validated in mouse models; −, not validated in mouse models.

The effects of genetic variation in the IFN-induced transmembrane protein 3 (IFITM3) gene on IAV disease was first identified during the 2009 H1N1 pandemic. IFITM3 is a potent antiviral protein that blocks release of the viral genome into the cell’s cytoplasm by preventing viral fusion with the endosomal membrane (105107). Two groups reported on a polymorphism (rs12252_C) that predisposed to severe and fatal H1N1 IAV (101, 102). Similar findings were reported for Chinese patients infected with the emergent avian H7N9 IAV (108). The rs12252_C polymorphism was predicted to effect RNA splicing and truncate the IFITM3 protein. However, next-generation sequencing on cells derived from individuals with all three genotypes showed that full-length IFITM3 mRNA was present in all genotypes (109). Thus, the mechanism by which rs12252_C affects susceptibility to IAV disease remains unknown. Recently, a second polymorphism in the IFITM3 gene, rs34481144, was associated with severe IAV disease in predominantly non-Chinese populations (110). This polymorphism is located in the promotor region of the IFITM3 gene and affects IFITM3 expression level in CD8+ T cells, which is consistent with an earlier study in mice showing that Ifitm3 expression protect TRM from IAV infection (111).

The significance of innate immunity and antiviral host defense for protection against severe IAV disease is further highlighted by the discovery of a 2.5-y-old patient who suffered from severe IAV disease (112). Next-generation sequencing of her genome, as well as her parents, identified two missense mutations in the host gene IRF7. These two rare mutations impaired the function of IRF7 protein and diminished the amplification of type I and III IFN postinfection. Interestingly, the impaired IRF7 function did not lead to severe disease after RSV, VZV, and CMV infection, suggesting redundant pathways or mechanisms of protection against these pathogens. This same group also identified rare mutations in the gene debranching RNA lariats 1 (DBR1) that result in severe disease after IBV, HSV, and norovirus infection (113). DBR1 is an enzyme that is involved in the degradation of RNA lariats (leftover molecules from RNA splicing) (114). Abrogation of DBR1 activity leads to elevated levels of RNA lariats, attenuation of IFN signaling, and increased severity of disease. Besides host genes that are important for innate and antiviral immunity, genetic variation in genes associated with cellular homeostasis can also predispose to severe clinical outcomes after IAV infection. Carnitine palmitoyltransferase II (CPTII) is a mitochondrial membrane protein required for fatty acid oxidation. Missense mutations in CPTII produce a temperature sensitive form of the CPTII protein, which, upon high fever induced by severe IAV disease, becomes inactive. This results in a buildup of long-chain fatty acids, leading to influenza-associated encephalopathy (115117).

Animal models, such as mice, offer an alternative to identify and study host factors associated with severe and fatal IAV disease. The first antiviral host gene Mx1 was identified in 1962 after comparing IAV-resistant and susceptible mouse strains (118, 119). Our work and that of others have since identified many genetic loci associated with severe disease after pathogenic IAV infection in mice (120130) and chickens (131). In some cases, the host gene within the loci (Hc, Ifi35, Lst1, and Mx1) has been identified (120, 126, 132, 133). Ifi35 was recently shown to exacerbate weight loss and disease in mice after IAV infection (133). The expression of Ifi35 increases the production of the proinflammatory cytokine IL-12p80 (a homodimer of IL-12p40). Neutralization of IL-12p80 ameliorated weight loss postinfection. The significance of these host genes and associated mechanisms in human IAV needs to be investigated. Ongoing developments in next-generation sequencing and genome editing will accelerate the identification of host genes and associated polymorphisms that contribute to differences in clinical outcome after IAV infection.

A significant advantage of the mouse model is the ability to study gene function in the context of primary IAV infection and identify the mechanism of severe disease. Analysis of the literature identified several hundred citations containing a Kaplan–Meier survival analysis comparing IAV induced mortality between wild-type and gene-deficient mouse strains. One hundred seventy different host genes have been studied to date for susceptibility or resistance against different strains of IAV (Fig. 1). Many host genes in this analysis (Ifnar1, Ifnl1, Stat1, Ifitm3, Sfpta1, and Irf7) are involved in innate and antiviral immune pathways and are required for survival after IAV infection. These findings recapitulate what is observed in humans that are deficient in key components of the IFN and antiviral immune system. Deletion of host genes involved in the complement pathway, C3, C4, and C5, also increase the susceptibility to infection. A second set of host genes (Fig. 1, green box) exacerbates IAV disease, and deletion of these genes protect the mice from severe and fatal IAV infection. These genes are essential for IAV replication (Tmprss2, Ipo7) and are involved in inflammation (Par1, Tnfaip3, Nos2, Ptges2, and Ifi35) or tissue homeostasis (Epg5, Atg14, and Atg7). The negative impact of inflammatory gene expression on IAV disease is consistent with the idea of immunopathology contributing to the severity of IAV disease. The third group (Fig. 1, blue box) includes Ccr2, Tlr3, Tlr4, and Myd88, and their role during IAV pathogenesis is conflicting. Variation in virus strain, microbiome, or laboratory environment may contribute to the differences in disease outcome of these gene-deficient mice. The fourth and final group of host genes (Fig. 1, gray box) does not affect survival after IAV infection. Despite the challenges in reporting negative results, it is important for the field that these results are published or deposited in a publicly available database. This avoids duplication and allows for a systems-wide analysis to study and infer gene function and activity in the context of different microbial infections. Overall, this comprehensive analysis of the different host factors modulating the outcome after IAV infection reveals three things. First, genes involved in the innate and antiviral immune response are important for protection against IAV. However, genes involved in inflammation often exacerbate disease, although these effects may depend on other host and viral factors that contribute to the immune status of the animal. Secondly, future studies should include more genetically modified mouse strains to identify additional host genes that modulate IAV disease, but perhaps more importantly to identify the cell type (using conditional knockouts) and molecular mechanism that is driving this difference in disease outcome. Finally, it is important to evaluate gene activity in different contexts, including aging, obesity, and hosts (i.e., different animal models or mouse strains). An elegant example of the significance of host genetic variation on gene activity is a study of the antiviral host gene Mx1, which protects against IAV in a C57BL/6 mouse strain but not in a DBA/2J mouse strain (134). The analyses of gene activity and function in different contexts associated with severe IAV disease will further enhance our understanding of IAV disease and facilitate the transition of basic science in animal models to clinical practice and development of host-targeted therapies against IAV.

FIGURE 1.

The different host genes that have been tested in gene-knockout mice in IAV pathogenesis models. 2–6Number of publications reporting on the role of the host gene on survival after intranasal IAV challenge. *The significance of Ipo7 depends on the strain of IAV used. A table containing the names of the host genes, the associated IAV phenotypes, and a web-link to the paper in NCBI is available upon request.

FIGURE 1.

The different host genes that have been tested in gene-knockout mice in IAV pathogenesis models. 2–6Number of publications reporting on the role of the host gene on survival after intranasal IAV challenge. *The significance of Ipo7 depends on the strain of IAV used. A table containing the names of the host genes, the associated IAV phenotypes, and a web-link to the paper in NCBI is available upon request.

Close modal

Influenza pathogenesis occurs in two phases (Fig. 2). The first phase starts immediately postinfection and lasts for anywhere between 1 and 3 days. It is during this phase that much of the clinical course and outcome of the disease is determined. Reduced or no pre-existing immunity will result in high peak viral titer and thus more inflammation. This scenario occurs often during IAV pandemics or in the very young and elderly population. In contrast, pre-existing immunity or effective antiviral immunity will limit early virus replication and thus produce fewer clinical symptoms and disease. Depending on the amount of virus and inflammation after this phase, the second phase may lead to control of the virus or result in severe disease associated with acute respiratory stress syndrome and death. Control of IAV replication often occurs in healthy adults who are able to limit peak viral titer in phase 1 or mount a strong adaptive immune response in phase 2. Occasionally, the immune response becomes dysregulated, causing severe and sometimes fatal disease in young adults. In contrast, older individuals who are obese or have underlying comorbidities are less likely to control virus replication at early and late time points during infection, resulting in more inflammation and severe IAV disease. Excessive inflammation hampers tissue repair and has a negative effect on T and B cell immune responses. Virus and host factors including age, microbiome, obesity, and sex (sex hormones) can modulate each of these processes to produce the spectrum of IAV disease that we see to date. Development of effective and broadly protective IAV vaccines and antiviral therapies, including host-directed therapies, are key areas for future investigations to prevent severe IAV disease.

FIGURE 2.

Host factors that modulate influenza virus pathogenesis.

FIGURE 2.

Host factors that modulate influenza virus pathogenesis.

Close modal

We thank Drs. Shin and Shafiuddin for discussion and reviewing our manuscript.

This work was supported by National Institutes of Health (NIH) Grants R01- AI118938 and R21-AI137450 (to A.C.M.B.) and NIH/National Institute of Allergy and Infectious Diseases Training Grant T32-AI007172, which supported A.P.G.

Abbreviations used in this article:

CPTII

carnitine palmitoyltransferase II

DBR1

debranching RNA lariats 1

DC

dendritic cell

HA

hemagglutinin

IAV

influenza A virus

IFITM3

IFN-induced transmembrane protein 3

NA

neuraminidase

TRM

tissue resident memory T cell.

1
Zens
,
K. D.
,
D. L.
Farber
.
2015
.
Memory CD4 T cells in influenza.
Curr. Top. Microbiol. Immunol.
386
:
399
421
.
2
Chen
,
X.
,
S.
Liu
,
M. U.
Goraya
,
M.
Maarouf
,
S.
Huang
,
J. L.
Chen
.
2018
.
Host immune response to influenza a virus infection.
Front Immunol.
9
:
320
.
3
Nussing
,
S.
,
S.
Sant
,
M.
Koutsakos
,
K.
Subbarao
,
T. H. O.
Nguyen
,
K.
Kedzierska
.
2018
.
Innate and adaptive T cells in influenza disease.
Front Med.
12
:
34
47
.
4
Chiu
,
C.
,
P. J.
Openshaw
.
2015
.
Antiviral B cell and T cell immunity in the lungs.
Nat. Immunol.
16
:
18
26
.
5
Angeletti
,
D.
,
J. W.
Yewdell
.
2018
.
Understanding and manipulating viral immunity: antibody immunodominance enters center stage.
Trends Immunol.
39
:
549
561
.
6
Takahashi
,
Y.
,
T.
Onodera
,
Y.
Adachi
,
M.
Ato
.
2017
.
Adaptive B cell responses to influenza virus infection in the lung.
Viral Immunol.
30
:
431
437
.
7
Corti
,
D.
,
E.
Cameroni
,
B.
Guarino
,
N. L.
Kallewaard
,
Q.
Zhu
,
A.
Lanzavecchia
.
2017
.
Tackling influenza with broadly neutralizing antibodies.
Curr Opin Virol.
24
:
60
69
.
8
Memoli
,
M. J.
,
P. A.
Shaw
,
A.
Han
,
L.
Czajkowski
,
S.
Reed
,
R.
Athota
,
T.
Bristol
,
S.
Fargis
,
K.
Risos
,
J. H.
Powers
, et al
.
2016
.
Evaluation of antihemagglutinin and antineuraminidase antibodies as correlates of protection in an influenza A/H1N1 virus healthy human challenge model.
MBio.
7
:
e00417–16
.
9
Pizzolla
,
A.
,
T. H.
Nguyen
,
S.
Sant
,
J.
Jaffar
,
T.
Loudovaris
,
S. I.
Mannering
,
P. G.
Thomas
,
G. P.
Westall
,
K.
Kedzierska
,
L. M.
Wakim
.
2018
.
Influenza-specific lung-resident memory T cells are proliferative and polyfunctional and maintain diverse TCR profiles.
J. Clin. Invest.
128
:
721
733
.
10
Wakim
,
L. M.
,
J.
Smith
,
I.
Caminschi
,
M. H.
Lahoud
,
J. A.
Villadangos
.
2015
.
Antibody-targeted vaccination to lung dendritic cells generates tissue-resident memory CD8 T cells that are highly protective against influenza virus infection.
Mucosal Immunol.
8
:
1060
1071
.
11
Wilkinson
,
T. M.
,
C. K.
Li
,
C. S.
Chui
,
A. K.
Huang
,
M.
Perkins
,
J. C.
Liebner
,
R.
Lambkin-Williams
,
A.
Gilbert
,
J.
Oxford
,
B.
Nicholas
, et al
.
2012
.
Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans.
Nat. Med.
18
:
274
280
.
12
Wu
,
T.
,
Y.
Hu
,
Y. T.
Lee
,
K. R.
Bouchard
,
A.
Benechet
,
K.
Khanna
,
L. S.
Cauley
.
2014
.
Lung-resident memory CD8 T cells (TRM) are indispensable for optimal cross-protection against pulmonary virus infection.
J. Leukoc. Biol.
95
:
215
224
.
13
Pizzolla
,
A.
,
T. H. O.
Nguyen
,
J. M.
Smith
,
A. G.
Brooks
,
K.
Kedzieska
,
W. R.
Heath
,
P. C.
Reading
,
L. M.
Wakim
.
2017
.
Resident memory CD8(+) T cells in the upper respiratory tract prevent pulmonary influenza virus infection.
Sci Immunol.
2
.
14
Thompson
,
W. W.
,
D. K.
Shay
,
E.
Weintraub
,
L.
Brammer
,
N.
Cox
,
L. J.
Anderson
,
K.
Fukuda
.
2003
.
Mortality associated with influenza and respiratory syncytial virus in the United States.
JAMA
289
:
179
186
.
15
Mertz
,
D.
,
T. H.
Kim
,
J.
Johnstone
,
P. P.
Lam
,
M.
Science
,
S. P.
Kuster
,
S. A.
Fadel
,
D.
Tran
,
E.
Fernandez
,
N.
Bhatnagar
,
M.
Loeb
.
2013
.
Populations at risk for severe or complicated influenza illness: systematic review and meta-analysis.
BMJ
347
:
f5061
.
16
Liu
,
W. M.
,
B. A.
van der Zeijst
,
C. J.
Boog
,
E. C.
Soethout
.
2011
.
Aging and impaired immunity to influenza viruses: implications for vaccine development.
Hum. Vaccin.
7
(
Suppl.
):
94
98
.
17
Carr
,
E. J.
,
J.
Dooley
,
J. E.
Garcia-Perez
,
V.
Lagou
,
J. C.
Lee
,
C.
Wouters
,
I.
Meyts
,
A.
Goris
,
G.
Boeckxstaens
,
M. A.
Linterman
,
A.
Liston
.
2016
.
The cellular composition of the human immune system is shaped by age and cohabitation.
Nat Immunol.
17
:
461
468
.
18
Shaw
,
A. C.
,
D. R.
Goldstein
,
R. R.
Montgomery
.
2013
.
Age-dependent dysregulation of innate immunity.
Nat. Rev. Immunol.
13
:
875
887
.
19
Pillai
,
P. S.
,
R. D.
Molony
,
K.
Martinod
,
H.
Dong
,
I. K.
Pang
,
M. C.
Tal
,
A. G.
Solis
,
P.
Bielecki
,
S.
Mohanty
,
M.
Trentalange
, et al
.
2016
.
Mx1 reveals innate pathways to antiviral resistance and lethal influenza disease.
Science
352
:
463
466
.
20
Prakash
,
S.
,
S.
Agrawal
,
J. N.
Cao
,
S.
Gupta
,
A.
Agrawal
.
2013
.
Impaired secretion of interferons by dendritic cells from aged subjects to influenza: role of histone modifications.
Age (Dordr).
35
:
1785
1797
.
21
Sridharan
,
A.
,
M.
Esposo
,
K.
Kaushal
,
J.
Tay
,
K.
Osann
,
S.
Agrawal
,
S.
Gupta
,
A.
Agrawal
.
2011
.
Age-associated impaired plasmacytoid dendritic cell functions lead to decreased CD4 and CD8 T cell immunity.
Age (Dordr).
33
:
363
376
.
22
Canaday
,
D. H.
,
N. A.
Amponsah
,
L.
Jones
,
D. J.
Tisch
,
T. R.
Hornick
,
L.
Ramachandra
.
2010
.
Influenza-induced production of interferon-alpha is defective in geriatric individuals.
J. Clin. Immunol.
30
:
373
383
.
23
Solana
,
R.
,
R.
Tarazona
,
I.
Gayoso
,
O.
Lesur
,
G.
Dupuis
,
T.
Fulop
.
2012
.
Innate immunosenescence: effect of aging on cells and receptors of the innate immune system in humans.
Semin Immunol.
24
:
331
341
.
24
Xu
,
X. J.
,
J. S.
Reichner
,
B.
Mastrofrancesco
,
W. L.
Henry
Jr.
,
J. E.
Albina
.
2008
.
Prostaglandin E2 suppresses lipopolysaccharide-stimulated IFN-beta production.
J. Immunol.
180
:
2125
2131
.
25
Wong
,
C.
,
D. R.
Goldstein
.
2013
.
Impact of aging on antigen presentation cell function of dendritic cells.
Curr. Opin. Immunol.
25
:
535
541
.
26
Komatsubara
,
S.
,
B.
Cinader
,
S.
Muramatsu
.
1986
.
Functional competence of dendritic cells of ageing C57BL/6 mice.
Scand. J. Immunol.
24
:
517
525
.
27
Tesar
,
B. M.
,
W. E.
Walker
,
J.
Unternaehrer
,
N. S.
Joshi
,
A.
Chandele
,
L.
Haynes
,
S.
Kaech
,
D. R.
Goldstein
.
2006
.
Murine [corrected] myeloid dendritic cell-dependent toll-like receptor immunity is preserved with aging. [Published erratum appears in 2007 Aging Cell. 6: 129.]
Aging Cell.
5
:
473
486
.
28
Wong
,
C. P.
,
K. R.
Magnusson
,
E.
Ho
.
2010
.
Aging is associated with altered dendritic cells subset distribution and impaired proinflammatory cytokine production.
Exp. Gerontol.
45
:
163
169
.
29
Pereira
,
L. F.
,
A. P.
de Souza
,
T. J.
Borges
,
C.
Bonorino
.
2011
.
Impaired in vivo CD4+ T cell expansion and differentiation in aged mice is not solely due to T cell defects: decreased stimulation by aged dendritic cells.
Mech Ageing Dev.
132
:
187
194
.
30
Li
,
G.
,
M. J.
Smithey
,
B. D.
Rudd
,
J.
Nikolich-Zugich
.
2012
.
Age-associated alterations in CD8alpha+ dendritic cells impair CD8 T-cell expansion in response to an intracellular bacterium.
Aging Cell.
11
:
968
977
.
31
Moretto
,
M. M.
,
E. M.
Lawlor
,
I. A.
Khan
.
2008
.
Aging mice exhibit a functional defect in mucosal dendritic cell response against an intracellular pathogen.
J. Immunol.
181
:
7977
7984
.
32
Grolleau-Julius
,
A.
,
E. K.
Harning
,
L. M.
Abernathy
,
R. L.
Yung
.
2008
.
Impaired dendritic cell function in aging leads to defective antitumor immunity.
Cancer Res.
68
:
6341
6349
.
33
McElhaney
,
J. E.
,
R. B.
Effros
.
2009
.
Immunosenescence: what does it mean to health outcomes in older adults?
Curr. Opin. Immunol.
21
:
418
424
.
34
Xia
,
S.
,
X.
Zhang
,
S.
Zheng
,
R.
Khanabdali
,
B.
Kalionis
,
J.
Wu
,
W.
Wan
,
X.
Tai
.
2016
.
An update on inflamm-aging: mechanisms, prevention, and treatment.
J. Immunol. Res.
2016
:
8426874
.
35
Le Saux
,
S.
,
C. M.
Weyand
,
J. J.
Goronzy
.
2012
.
Mechanisms of immunosenescence: lessons from models of accelerated immune aging.
Ann. N Y Acad. Sci.
1247
:
69
82
.
36
Cavanagh
,
M. M.
,
C. M.
Weyand
,
J. J.
Goronzy
.
2012
.
Chronic inflammation and aging: DNA damage tips the balance.
Curr Opin Immunol.
24
:
488
493
.
37
Ginaldi
,
L.
,
M. F.
Loreto
,
M. P.
Corsi
,
M.
Modesti
,
M.
De Martinis
.
2001
.
Immunosenescence and infectious diseases.
Microbes Infect.
3
:
851
857
.
38
Toapanta
,
F. R.
,
T. M.
Ross
.
2009
.
Impaired immune responses in the lungs of aged mice following influenza infection.
Respir. Res.
10
:
112
.
39
Franceschi
,
C.
,
M.
Bonafè
,
S.
Valensin
,
F.
Olivieri
,
M.
De Luca
,
E.
Ottaviani
,
G.
De Benedictis
.
2000
.
Inflamm-aging. An evolutionary perspective on immunosenescence.
Ann. N. Y. Acad. Sci.
908
:
244
254
.
40
Meyer
,
K. C.
2001
.
The role of immunity in susceptibility to respiratory infection in the aging lung.
Respir. Physiol.
128
:
23
31
.
41
Khan
,
N.
,
N.
Shariff
,
M.
Cobbold
,
R.
Bruton
,
J. A.
Ainsworth
,
A. J.
Sinclair
,
L.
Nayak
,
P. A.
Moss
.
2002
.
Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals.
J. Immunol.
169
:
1984
1992
.
42
Frasca
,
D.
,
R. L.
Riley
,
B. B.
Blomberg
.
2005
.
Humoral immune response and B-cell functions including immunoglobulin class switch are downregulated in aged mice and humans.
Semin. Immunol.
17
:
378
384
.
43
Ferguson
,
F. G.
,
A.
Wikby
,
P.
Maxson
,
J.
Olsson
,
B.
Johansson
.
1995
.
Immune parameters in a longitudinal study of a very old population of Swedish people: a comparison between survivors and nonsurvivors.
J. Gerontol. A Biol. Sci. Med. Sci.
50
:
B378
B382
.
44
Yager
,
E. J.
,
M.
Ahmed
,
K.
Lanzer
,
T. D.
Randall
,
D. L.
Woodland
,
M. A.
Blackman
.
2008
.
Age-associated decline in T cell repertoire diversity leads to holes in the repertoire and impaired immunity to influenza virus.
J Exp Med.
205
:
711
723
.
45
Asanuma
,
H.
,
K.
Hirokawa
,
M.
Uchiyama
,
Y.
Suzuki
,
C.
Aizawa
,
T.
Kurata
,
T.
Sata
,
S.
Tamura
.
2001
.
Immune responses and protection in different strains of aged mice immunized intranasally with an adjuvant-combined influenza vaccine.
Vaccine
19
:
3981
3989
.
46
Gubbels Bupp
,
M. R.
,
T.
Potluri
,
A. L.
Fink
,
S. L.
Klein
.
2018
.
The confluence of sex hormones and aging on immunity.
Front. Immunol.
9
:
1269
.
47
Gabriel
,
G.
,
P. C.
Arck
.
2014
.
Sex, immunity and influenza.
J. Infect. Dis.
209
(
Suppl. 3
):
S93
S99
.
48
Klein
,
S. L.
,
K. L.
Flanagan
.
2016
.
Sex differences in immune responses.
Nat. Rev. Immunol.
16
:
626
638
.
49
Klein
,
S. L.
,
A.
Jedlicka
,
A.
Pekosz
.
2010
.
The Xs and Y of immune responses to viral vaccines. [Published erratum appears in 2010 Lancet Infect. Dis. 10: 740.]
Lancet Infect. Dis.
10
:
338
349
.
50
Vom Steeg
,
L. G.
,
S. L.
Klein
.
2017
.
Sex steroids mediate bidirectional interactions between hosts and microbes.
Horm. Behav.
88
:
45
51
.
51
Larcombe
,
A. N.
,
R. E.
Foong
,
E. M.
Bozanich
,
L. J.
Berry
,
L. W.
Garratt
,
R. C.
Gualano
,
J. E.
Jones
,
L. F.
Dousha
,
G. R.
Zosky
,
P. D.
Sly
.
2011
.
Sexual dimorphism in lung function responses to acute influenza A infection.
Influenza Other Respir Viruses.
5
:
334
342
.
52
Hoffmann
,
J.
,
A.
Otte
,
S.
Thiele
,
H.
Lotter
,
Y.
Shu
,
G.
Gabriel
.
2015
.
Sex differences in H7N9 influenza A virus pathogenesis.
Vaccine.
33
:
6949
6954
.
53
Robinson
,
D. P.
,
M. E.
Lorenzo
,
W.
Jian
,
S. L.
Klein
.
2011
.
Elevated 17beta-estradiol protects females from influenza A virus pathogenesis by suppressing inflammatory responses.
PLoS Pathog.
7
:
e1002149
.
54
Lorenzo
,
M. E.
,
A.
Hodgson
,
D. P.
Robinson
,
J. B.
Kaplan
,
A.
Pekosz
,
S. L.
Klein
.
2011
.
Antibody responses and cross protection against lethal influenza A viruses differ between the sexes in C57BL/6 mice.
Vaccine.
29
:
9246
9255
.
55
Maestri
,
A.
,
V. A.
Sortica
,
D. L.
Ferreira
,
J.
de Almeida Ferreira
,
M. A.
Amador
,
W. A.
de Mello
,
S. E.
Santos
,
R. C.
Sousa
.
2016
.
The His131Arg substitution in the FCGR2A gene (rs1801274) is not associated with the severity of influenza A(H1N1)pdm09 infection.
BMC Res. Notes
9
:
296
.
56
Vom Steeg
,
L. G.
,
M. S.
Vermillion
,
O. J.
Hall
,
O.
Alam
,
R.
McFarland
,
H.
Chen
,
B.
Zirkin
,
S. L.
Klein
.
2016
.
Age and testosterone mediate influenza pathogenesis in male mice.
Am. J. Physiol. Lung Cell Mol. Physiol.
311
:
L1234
-
L1244
.
57
Furman
,
D.
,
B. P.
Hejblum
,
N.
Simon
,
V.
Jojic
,
C. L.
Dekker
,
R.
Thiébaut
,
R. J.
Tibshirani
,
M. M.
Davis
.
2014
.
Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination.
Proc. Natl. Acad. Sci. USA
111
:
869
874
.
58
Nguyen
,
D. C.
,
F.
Masseoud
,
X.
Lu
,
F.
Scinicariello
,
S.
Sambhara
,
R.
Attanasio
.
2011
.
17β-Estradiol restores antibody responses to an influenza vaccine in a postmenopausal mouse model.
Vaccine.
29
:
2515
2518
.
59
Robinson
,
D. P.
,
O. J.
Hall
,
T. L.
Nilles
,
J. H.
Bream
,
S. L.
Klein
.
2014
.
17β-estradiol protects females against influenza by recruiting neutrophils and increasing virus-specific CD8 T cell responses in the lungs.
J. Virol.
88
:
4711
4720
.
60
Pazos
,
M. A.
,
T. A.
Kraus
,
C.
Munoz-Fontela
,
T. M.
Moran
.
2012
.
Estrogen mediates innate and adaptive immune alterations to influenza infection in pregnant mice.
PLoS One
7
:
e40502
.
61
Biswas
,
D. K.
,
S.
Singh
,
Q.
Shi
,
A. B.
Pardee
,
J. D.
Iglehart
.
2005
.
Crossroads of estrogen receptor and NF-kappaB signaling.
Sci. STKE
2005
:
pe27
.
62
Au
,
A.
,
A.
Feher
,
L.
McPhee
,
A.
Jessa
,
S.
Oh
,
G.
Einstein
.
2016
.
Estrogens, inflammation and cognition.
Front Neuroendocrinol.
40
:
87
100
.
63
Hall
,
O. J.
,
N.
Limjunyawong
,
M. S.
Vermillion
,
D. P.
Robinson
,
N.
Wohlgemuth
,
A.
Pekosz
,
W.
Mitzner
,
S. L.
Klein
.
2016
.
Progesterone-based therapy protects against influenza by promoting lung repair and recovery in females.
PLoS Pathog.
12
:
e1005840
.
64
Davis
,
S. M.
,
L. M.
Sweet
,
K. H.
Oppenheimer
,
B. T.
Suratt
,
M.
Phillippe
.
2017
.
Estradiol and progesterone influence on influenza infection and immune response in a mouse model.
Am. J. Reprod. Immunol.
78
:
e12695
.
65
Engels
,
G.
,
A. M.
Hierweger
,
J.
Hoffmann
,
R.
Thieme
,
S.
Thiele
,
S.
Bertram
,
C.
Dreier
,
P.
Resa-Infante
,
H.
Jacobsen
,
K.
Thiele
, et al
.
2017
.
Pregnancy-related immune adaptation promotes the emergence of highly virulent H1N1 influenza virus strains in allogenically pregnant mice.
Cell Host Microbe.
21
:
321
333
.
66
Marcelin
,
G.
,
J. R.
Aldridge
,
S.
Duan
,
H. E.
Ghoneim
,
J.
Rehg
,
H.
Marjuki
,
A. C.
Boon
,
J. A.
McCullers
,
R. J.
Webby
.
2011
.
Fatal outcome of pandemic H1N1 2009 influenza virus infection is associated with immunopathology and impaired lung repair, not enhanced viral burden, in pregnant mice.
J. Virol.
85
:
11208
11219
.
67
Kim
,
H. M.
,
Y. M.
Kang
,
B. M.
Song
,
H. S.
Kim
,
S. H.
Seo
.
2012
.
The 2009 pandemic H1N1 influenza virus is more pathogenic in pregnant mice than seasonal H1N1 influenza virus.
Viral Immunol.
25
:
402
410
.
68
Chan
,
K. H.
,
A. J.
Zhang
,
K. K.
To
,
C. C.
Chan
,
V. K.
Poon
,
K.
Guo
,
F.
Ng
,
Q. W.
Zhang
,
V. H.
Leung
,
A. N.
Cheung
, et al
.
2010
.
Wild type and mutant 2009 pandemic influenza A (H1N1) viruses cause more severe disease and higher mortality in pregnant BALB/c mice.
PLoS One.
5
:
e13757
.
69
Mulcahy
,
M. E.
,
R. M.
McLoughlin
.
2016
.
Staphylococcus aureus and influenza A virus: partners in coinfection.
MBio.
7
:
e02068-16
.
70
Wu
,
S.
,
Z. Y.
Jiang
,
Y. F.
Sun
,
B.
Yu
,
J.
Chen
,
C. Q.
Dai
,
X. L.
Wu
,
X. L.
Tang
,
X. Y.
Chen
.
2013
.
Microbiota regulates the TLR7 signaling pathway against respiratory tract influenza A virus infection.
Curr Microbiol.
67
:
414
422
.
71
Ichinohe
,
T.
,
I. K.
Pang
,
Y.
Kumamoto
,
D. R.
Peaper
,
J. H.
Ho
,
T. S.
Murray
,
A.
Iwasaki
.
2011
.
Microbiota regulates immune defense against respiratory tract influenza A virus infection.
Proc. Natl. Acad. Sci. USA
108
:
5354
5359
.
72
Abt
,
M. C.
,
L. C.
Osborne
,
L. A.
Monticelli
,
T. A.
Doering
,
T.
Alenghat
,
G. F.
Sonnenberg
,
M. A.
Paley
,
M.
Antenus
,
K. L.
Williams
,
J.
Erikson
, et al
.
2012
.
Commensal bacteria calibrate the activation threshold of innate antiviral immunity.
Immunity.
37
:
158
170
.
73
Oh
,
J. Z.
,
R.
Ravindran
,
B.
Chassaing
,
F. A.
Carvalho
,
M. S.
Maddur
,
M.
Bower
,
P.
Hakimpour
,
K. P.
Gill
,
H. I.
Nakaya
,
F.
Yarovinsky
, et al
.
2014
.
TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination.
Immunity.
41
:
478
492
.
74
Steed
,
A. L.
,
G. P.
Christophi
,
G. E.
Kaiko
,
L.
Sun
,
V. M.
Goodwin
,
U.
Jain
,
E.
Esaulova
,
M. N.
Artyomov
,
D. J.
Morales
,
M. J.
Holtzman
, et al
.
2017
.
The microbial metabolite desaminotyrosine protects from influenza through type I interferon.
Science.
357
:
498
502
.
75
Rosshart
,
S. P.
,
B. G.
Vassallo
,
D.
Angeletti
,
D. S.
Hutchinson
,
A. P.
Morgan
,
K.
Takeda
,
H. D.
Hickman
,
J. A.
McCulloch
,
J. H.
Badger
,
N. J.
Ajami
, et al
.
2017
.
Wild mouse gut microbiota promotes host fitness and improves disease resistance.
Cell.
171
:
1015
1028.e13
.
76
Centers for Disease Control and Prevention (CDC)
.
2009
.
Hospitalized patients with novel influenza A (H1N1) virus infection - California, April-May, 2009.
MMWR Morb. Mortal. Wkly. Rep.
58
:
536
541
.
77
Louie
,
J. K.
,
M.
Acosta
,
M. C.
Samuel
,
R.
Schechter
,
D. J.
Vugia
,
K.
Harriman
,
B. T.
Matyas
.
2011
.
A novel risk factor for a novel virus: obesity and 2009 pandemic influenza A (H1N1).
Clin. Infect. Dis.
52
:
301
312
.
78
Hanslik
,
T.
,
P. Y.
Boelle
,
A.
Flahault
.
2010
.
Preliminary estimation of risk factors for admission to intensive care units and for death in patients infected with A(H1N1)2009 influenza virus, France, 2009-2010.
PLoS Curr.
2
:
RRN1150
.
79
Morgan
,
O. W.
,
A.
Bramley
,
A.
Fowlkes
,
D. S.
Freedman
,
T. H.
Taylor
,
P.
Gargiullo
,
B.
Belay
,
S.
Jain
,
C.
Cox
,
L.
Kamimoto
, et al
.
2010
.
Morbid obesity as a risk factor for hospitalization and death due to 2009 pandemic influenza A(H1N1) disease.
PLoS One.
5
:
e9694
.
80
Santa-Olalla Peralta
,
P.
,
M.
Cortes-García
,
M.
Vicente-Herrero
,
C.
Castrillo-Villamandos
,
P.
Arias-Bohigas
,
I.
Pachon-del Amo
,
M. J.
Sierra-Moros
;
Surveillance Group for New Influenza A(H1N1) Virus Investigation and Control Team in Spain
.
2010
.
Risk factors for disease severity among hospitalised patients with 2009 pandemic influenza A (H1N1) in Spain, April - December 2009.
Euro Surveill.
15
:
38
.
81
Cocoros
,
N. M.
,
T. L.
Lash
,
A.
DeMaria
Jr.
,
M.
Klompas
.
2014
.
Obesity as a risk factor for severe influenza-like illness.
Influenza Other Respir Viruses.
8
:
25
32
.
82
Karki
,
S.
,
D. J.
Muscatello
,
E.
Banks
,
C. R.
MacIntyre
,
P.
McIntyre
,
B.
Liu
.
2018
.
Association between body mass index and laboratory-confirmed influenza in middle aged and older adults: a prospective cohort study.
Int. J. Obes. (Lond.)
42
:
1480
1488
.
83
Yang
,
L.
,
K. P.
Chan
,
R. S.
Lee
,
W. M.
Chan
,
H. K.
Lai
,
T. Q.
Thach
,
K. H.
Chan
,
T. H.
Lam
,
J. S.
Peiris
,
C. M.
Wong
.
2013
.
Obesity and influenza associated mortality: evidence from an elderly cohort in Hong Kong.
Prev. Med.
56
:
118
123
.
84
Halvorson
,
E. E.
,
T. R.
Peters
,
J. A.
Skelton
,
C.
Suerken
,
B. M.
Snively
,
K. A.
Poehling
.
2018
.
Is weight associated with severity of acute respiratory illness?
Int J Obes
27
:
1582
1589
.
85
Louie
,
J. K.
,
M.
Acosta
,
K.
Winter
,
C.
Jean
,
S.
Gavali
,
R.
Schechter
,
D.
Vugia
,
K.
Harriman
,
B.
Matyas
,
C. A.
Glaser
, et al
California Pandemic (H1N1) Working Group
.
2009
.
Factors associated with death or hospitalization due to pandemic 2009 influenza A(H1N1) infection in California.
JAMA
302
:
1896
1902
.
86
Liu
,
B.
,
F.
Havers
,
E.
Chen
,
Z.
Yuan
,
H.
Yuan
,
J.
Ou
,
M.
Shang
,
K.
Kang
,
K.
Liao
,
F.
Liu
, et al
.
2014
.
Risk factors for influenza A(H7N9) disease–China, 2013.
Clin. Infect. Dis.
59
:
787
794
.
87
Paich
,
H. A.
,
P. A.
Sheridan
,
J.
Handy
,
E. A.
Karlsson
,
S.
Schultz-Cherry
,
M. G.
Hudgens
,
T. L.
Noah
,
S. S.
Weir
,
M. A.
Beck
.
2013
.
Overweight and obese adult humans have a defective cellular immune response to pandemic H1N1 influenza A virus.
Obesity (Silver Spring).
21
:
2377
2386
.
88
Painter
,
S. D.
,
I. G.
Ovsyannikova
,
G. A.
Poland
.
2015
.
The weight of obesity on the human immune response to vaccination.
Vaccine.
33
:
4422
4429
.
89
Neidich
,
S. D.
,
W. D.
Green
,
J.
Rebeles
,
E. A.
Karlsson
,
S.
Schultz-Cherry
,
T. L.
Noah
,
S.
Chakladar
,
M. G.
Hudgens
,
S. S.
Weir
,
M. A.
Beck
.
2017
.
Increased risk of influenza among vaccinated adults who are obese.
Int. J. Obes. (Lond.).
41
:
1324
1330
.
90
Yan
,
J.
,
M.
Grantham
,
J.
Pantelic
,
P. J.
Bueno de Mesquita
,
B.
Albert
,
F.
Liu
,
S.
Ehrman
,
D. K.
Milton
.
2018
.
Infectious virus in exhaled breath of symptomatic seasonal influenza cases from a college community.
Proc Natl Acad Sci U S A.
115
:
1081
1086
.
91
Maier
,
H. E.
,
R.
Lopez
,
N.
Sanchez
,
S.
Ng
,
L.
Gresh
,
S.
Ojeda
,
R.
Burger-Calderon
,
G.
Kuan
,
E.
Harris
,
A.
Balmaseda
,
A.
Gordon
.
2018
.
Obesity increases the duration of influenza a virus shedding in adults.
J. Infect. Dis.
218
:
1378
1382
.
92
Milner
,
J. J.
,
J.
Rebeles
,
S.
Dhungana
,
D. A.
Stewart
,
S. C.
Sumner
,
M. H.
Meyers
,
P.
Mancuso
,
M. A.
Beck
.
2015
.
Obesity increases mortality and modulates the lung metabolome during pandemic H1N1 influenza virus infection in mice.
J. Immunol.
194
:
4846
4859
.
93
Milner
,
J. J.
,
P. A.
Sheridan
,
E. A.
Karlsson
,
S.
Schultz-Cherry
,
Q.
Shi
,
M. A.
Beck
.
2013
.
Diet-induced obese mice exhibit altered heterologous immunity during a secondary 2009 pandemic H1N1 infection.
J. Immunol.
191
:
2474
2485
.
94
Sheridan
,
P. A.
,
H. A.
Paich
,
J.
Handy
,
E. A.
Karlsson
,
M. G.
Hudgens
,
A. B.
Sammon
,
L. A.
Holland
,
S.
Weir
,
T. L.
Noah
,
M. A.
Beck
.
2012
.
Obesity is associated with impaired immune response to influenza vaccination in humans.
Int. J. Obes. (Lond.).
36
:
1072
1077
.
95
Radigan
,
K. A.
,
L.
Morales-Nebreda
,
S.
Soberanes
,
T.
Nicholson
,
R.
Nigdelioglu
,
T.
Cho
,
M.
Chi
,
R. B.
Hamanaka
,
A. V.
Misharin
,
H.
Perlman
, et al
.
2014
.
Impaired clearance of influenza A virus in obese, leptin receptor deficient mice is independent of leptin signaling in the lung epithelium and macrophages.
PLoS One.
9
:
e108138
.
96
Turnbaugh
,
P. J.
,
F.
Backhed
,
L.
Fulton
,
J. I.
Gordon
.
2008
.
Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome.
Cell Host Microbe.
3
:
213
223
.
97
Ussar
,
S.
,
N. W.
Griffin
,
O.
Bezy
,
S.
Fujisaka
,
S.
Vienberg
,
S.
Softic
,
L.
Deng
,
L.
Bry
,
J. I.
Gordon
,
C. R.
Kahn
.
2015
.
Interactions between gut microbiota, host genetics and diet modulate the predisposition to obesity and metabolic syndrome. [Published erratum appears in 2016 Cell Metab. 23: 564–566.]
Cell Metab.
22
:
516
530
.
98
Turnbaugh
,
P. J.
,
R. E.
Ley
,
M. A.
Mahowald
,
V.
Magrini
,
E. R.
Mardis
,
J. I.
Gordon
.
2006
.
An obesity-associated gut microbiome with increased capacity for energy harvest.
Nature.
444
:
1027
1031
.
99
Watkin
,
I. J.
,
D.
Tills
,
R. B.
Heath
.
1975
.
Studies of the genetic susceptibility of individuals to infection with influenza viruses.
Humangenetik
30
:
75
79
.
100
Potter
,
C. W.
,
G. C.
Schild
.
1967
.
The incidence of HI antibody to Influenza virus A2/Singapore/ 1/57 in individuals of blood groups A and O.
J. Immunol.
98
:
1320
1325
.
101
Everitt
,
A. R.
,
S.
Clare
,
T.
Pertel
,
S. P.
John
,
R. S.
Wash
,
S. E.
Smith
,
C. R.
Chin
,
E. M.
Feeley
,
J. S.
Sims
,
D. J.
Adams
, et al
MOSAIC Investigators
.
2012
.
IFITM3 restricts the morbidity and mortality associated with influenza.
Nature
484
:
519
523
.
102
Zhang
,
Y. H.
,
Y.
Zhao
,
N.
Li
,
Y. C.
Peng
,
E.
Giannoulatou
,
R. H.
Jin
,
H. P.
Yan
,
H.
Wu
,
J. H.
Liu
,
N.
Liu
, et al
.
2013
.
Interferon-induced transmembrane protein-3 genetic variant rs12252-C is associated with severe influenza in Chinese individuals.
Nat. Commun.
4
:
1418
.
103
Horby
,
P.
,
H.
Sudoyo
,
V.
Viprakasit
,
A.
Fox
,
P. Q.
Thai
,
H.
Yu
,
S.
Davila
,
M.
Hibberd
,
S. J.
Dunstan
,
Y.
Monteerarat
, et al
.
2010
.
What is the evidence of a role for host genetics in susceptibility to influenza A/H5N1?
Epidemiol. Infect.
138
:
1550
1558
.
104
Kenney
,
A. D.
,
J. A.
Dowdle
,
L.
Bozzacco
,
T. M.
McMichael
,
C.
St Gelais
,
A. R.
Panfil
,
Y.
Sun
,
L. S.
Schlesinger
,
M. Z.
Anderson
,
P. L.
Green
, et al
.
2017
.
Human genetic determinants of viral diseases.
Annu. Rev. Genet.
51
:
241
263
.
105
Amini-Bavil-Olyaee
,
S.
,
Y. J.
Choi
,
J. H.
Lee
,
M.
Shi
,
I. C.
Huang
,
M.
Farzan
,
J. U.
Jung
.
2013
.
The antiviral effector IFITM3 disrupts intracellular cholesterol homeostasis to block viral entry. [Published erratum appears in 2013 Cell Host Microbe 14: 600–601.]
Cell Host Microbe
13
:
452
464
.
106
Desai
,
T. M.
,
M.
Marin
,
C. R.
Chin
,
G.
Savidis
,
A. L.
Brass
,
G. B.
Melikyan
.
2014
.
IFITM3 restricts influenza A virus entry by blocking the formation of fusion pores following virus-endosome hemifusion.
PLoS Pathog.
10
:
e1004048
.
107
Feeley
,
E. M.
,
J. S.
Sims
,
S. P.
John
,
C. R.
Chin
,
T.
Pertel
,
L. M.
Chen
,
G. D.
Gaiha
,
B. J.
Ryan
,
R. O.
Donis
,
S. J.
Elledge
,
A. L.
Brass
.
2011
.
IFITM3 inhibits influenza A virus infection by preventing cytosolic entry.
PLoS Pathog.
7
:
e1002337
.
108
Wang
,
Z.
,
A.
Zhang
,
Y.
Wan
,
X.
Liu
,
C.
Qiu
,
X.
Xi
,
Y.
Ren
,
J.
Wang
,
Y.
Dong
,
M.
Bao
, et al
.
2014
.
Early hypercytokinemia is associated with interferon-induced transmembrane protein-3 dysfunction and predictive of fatal H7N9 infection.
Proc Natl Acad Sci U S A.
111
:
769
774
.
109
Makvandi-Nejad
,
S.
,
H.
Laurenson-Schafer
,
L.
Wang
,
D.
Wellington
,
Y.
Zhao
,
B.
Jin
,
L.
Qin
,
K.
Kite
,
H. K.
Moghadam
,
C.
Song
, et al
.
2018
.
Lack of truncated IFITM3 transcripts in cells homozygous for the rs12252-C variant that is associated with severe influenza infection.
J. Infect. Dis.
217
:
257
262
.
110
Allen
,
E. K.
,
A. G.
Randolph
,
T.
Bhangale
,
P.
Dogra
,
M.
Ohlson
,
C. M.
Oshansky
,
A. E.
Zamora
,
J. P.
Shannon
,
D.
Finkelstein
,
A.
Dressen
, et al
.
2017
.
SNP-mediated disruption of CTCF binding at the IFITM3 promoter is associated with risk of severe influenza in humans.
Nat. Med.
23
:
975
983
.
111
Wakim
,
L. M.
,
N.
Gupta
,
J. D.
Mintern
,
J. A.
Villadangos
.
2013
.
Enhanced survival of lung tissue-resident memory CD8(+) T cells during infection with influenza virus due to selective expression of IFITM3.
Nat. Immunol.
14
:
238
245
.
112
Ciancanelli
,
M. J.
,
S. X.
Huang
,
P.
Luthra
,
H.
Garner
,
Y.
Itan
,
S.
Volpi
,
F. G.
Lafaille
,
C.
Trouillet
,
M.
Schmolke
,
R. A.
Albrecht
, et al
.
2015
.
Infectious disease. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency.
Science
348
:
448
453
.
113
Zhang
,
S. Y.
,
N. E.
Clark
,
C. A.
Freije
,
E.
Pauwels
,
A. J.
Taggart
,
S.
Okada
,
H.
Mandel
,
P.
Garcia
,
M. J.
Ciancanelli
,
A.
Biran
, et al
.
2018
.
Inborn errors of RNA lariat metabolism in humans with brainstem viral infection.
Cell.
172
:
952
965.e18
.
114
Montemayor
,
E. J.
,
A.
Katolik
,
N. E.
Clark
,
A. B.
Taylor
,
J. P.
Schuermann
,
D. J.
Combs
,
R.
Johnsson
,
S. P.
Holloway
,
S. W.
Stevens
,
M. J.
Damha
,
P. J.
Hart
.
2014
.
Structural basis of lariat RNA recognition by the intron debranching enzyme Dbr1.
Nucleic Acids Res.
42
:
10845
10855
.
115
Yao
,
D.
,
H.
Mizuguchi
,
M.
Yamaguchi
,
H.
Yamada
,
J.
Chida
,
K.
Shikata
,
H.
Kido
.
2008
.
Thermal instability of compound variants of carnitine palmitoyltransferase II and impaired mitochondrial fuel utilization in influenza-associated encephalopathy.
Hum. Mutat.
29
:
718
727
.
116
Mak
,
C. M.
,
C. W.
Lam
,
N. C.
Fong
,
W. K.
Siu
,
H. C.
Lee
,
T. S.
Siu
,
C. K.
Lai
,
C. Y.
Law
,
S. F.
Tong
,
W. T.
Poon
, et al
.
2011
.
Fatal viral infection-associated encephalopathy in two Chinese boys: a genetically determined risk factor of thermolabile carnitine palmitoyltransferase II variants.
J. Hum. Genet.
56
:
617
621
.
117
Chen
,
Y.
,
H.
Mizuguchi
,
D.
Yao
,
M.
Ide
,
Y.
Kuroda
,
Y.
Shigematsu
,
S.
Yamaguchi
,
M.
Yamaguchi
,
M.
Kinoshita
,
H.
Kido
.
2005
.
Thermolabile phenotype of carnitine palmitoyltransferase II variations as a predisposing factor for influenza-associated encephalopathy.
FEBS Lett.
579
:
2040
2044
.
118
Lindenmann
,
J.
1962
.
Resistance of mice to mouse-adapted influenza A virus.
Virology
16
:
203
204
.
119
Staeheli
,
P.
,
R.
Grob
,
E.
Meier
,
J. G.
Sutcliffe
,
O.
Haller
.
1988
.
Influenza virus-susceptible mice carry Mx genes with a large deletion or a nonsense mutation.
Mol. Cell. Biol.
8
:
4518
4523
.
120
Boon
,
A. C.
,
J.
deBeauchamp
,
A.
Hollmann
,
J.
Luke
,
M.
Kotb
,
S.
Rowe
,
D.
Finkelstein
,
G.
Neale
,
L.
Lu
,
R. W.
Williams
,
R. J.
Webby
.
2009
.
Host genetic variation affects resistance to infection with a highly pathogenic H5N1 influenza A virus in mice.
J. Virol.
83
:
10417
10426
.
121
Boon
,
A. C.
,
D.
Finkelstein
,
M.
Zheng
,
G.
Liao
,
J.
Allard
,
K.
Klumpp
,
R.
Webster
,
G.
Peltz
,
R. J.
Webby
.
2011
.
H5N1 influenza virus pathogenesis in genetically diverse mice is mediated at the level of viral load.
MBio
2
.
e00171-11
.
122
Boon
,
A. C.
,
R. W.
Williams
,
D. S.
Sinasac
,
R. J.
Webby
.
2014
.
A novel genetic locus linked to pro-inflammatory cytokines after virulent H5N1 virus infection in mice.
BMC Genomics
15
:
1017
.
123
Nedelko
,
T.
,
H.
Kollmus
,
F.
Klawonn
,
S.
Spijker
,
L.
Lu
,
M.
Heßman
,
R.
Alberts
,
R. W.
Williams
,
K.
Schughart
.
2012
.
Distinct gene loci control the host response to influenza H1N1 virus infection in a time-dependent manner.
BMC Genomics
13
:
411
.
124
Srivastava
,
B.
,
P.
Błazejewska
,
M.
Hessmann
,
D.
Bruder
,
R.
Geffers
,
S.
Mauel
,
A. D.
Gruber
,
K.
Schughart
.
2009
.
Host genetic background strongly influences the response to influenza a virus infections.
PLoS One
4
:
e4857
.
125
Bottomly
,
D.
,
M. T.
Ferris
,
L. D.
Aicher
,
E.
Rosenzweig
,
A.
Whitmore
,
D. L.
Aylor
,
B. L.
Haagmans
,
L. E.
Gralinski
,
B. G.
Bradel-Tretheway
,
J. T.
Bryan
, et al
.
2012
.
Expression quantitative trait Loci for extreme host response to influenza a in pre-collaborative cross mice.
G3 (Bethesda)
2
:
213
221
.
126
Ferris
,
M. T.
,
D. L.
Aylor
,
D.
Bottomly
,
A. C.
Whitmore
,
L. D.
Aicher
,
T. A.
Bell
,
B.
Bradel-Tretheway
,
J. T.
Bryan
,
R. J.
Buus
,
L. E.
Gralinski
, et al
.
2013
.
Modeling host genetic regulation of influenza pathogenesis in the collaborative cross.
PLoS Pathog.
9
:
e1003196
.
127
Trammell
,
R. A.
,
T. A.
Liberati
,
L. A.
Toth
.
2012
.
Host genetic background and the innate inflammatory response of lung to influenza virus.
Microbes Infect.
14
:
50
58
.
128
Blazejewska
,
P.
,
L.
Koscinski
,
N.
Viegas
,
D.
Anhlan
,
S.
Ludwig
,
K.
Schughart
.
2011
.
Pathogenicity of different PR8 influenza A virus variants in mice is determined by both viral and host factors. [Published erratum appears in 2014 Virology. 450–451: 369–370.]
Virology
412
:
36
45
.
129
Boivin
,
G. A.
,
J.
Pothlichet
,
E.
Skamene
,
E. G.
Brown
,
J. C.
Loredo-Osti
,
R.
Sladek
,
S. M.
Vidal
.
2012
.
Mapping of clinical and expression quantitative trait loci in a sex-dependent effect of host susceptibility to mouse-adapted influenza H3N2/HK/1/68.
J. Immunol.
188
:
3949
3960
.
130
Krementsov
,
D. N.
,
L. K.
Case
,
O.
Dienz
,
A.
Raza
,
Q.
Fang
,
J. L.
Ather
,
M. E.
Poynter
,
J. E.
Boyson
,
J. Y.
Bunn
,
C.
Teuscher
.
2017
.
Genetic variation in chromosome Y regulates susceptibility to influenza A virus infection.
Proc. Natl. Acad. Sci. U S A.
114
:
3491
3496
.
131
Ruiz-Hernandez
,
R.
,
W.
Mwangi
,
M.
Peroval
,
J. R.
Sadeyen
,
S.
Ascough
,
D.
Balkissoon
,
K.
Staines
,
A.
Boyd
,
J.
McCauley
,
A.
Smith
,
C.
Butter
.
2016
.
Host genetics determine susceptibility to avian influenza infection and transmission dynamics.
Sci. Rep.
6
:
26787
.
132
Leist
,
S. R.
,
H.
Kollmus
,
B.
Hatesuer
,
R. L.
Lambertz
,
K.
Schughart
.
2016
.
Lst1 deficiency has a minor impact on course and outcome of the host response to influenza A H1N1 infections in mice.
Virol. J.
13
:
17
.
133
Gounder
,
A. P.
,
C. C.
Yokoyama
,
N. N.
Jarjour
,
T. L.
Bricker
,
B. T.
Edelson
,
A. C. M.
Boon
.
2018
.
Interferon induced protein 35 exacerbates H5N1 influenza disease through the expression of IL-12p40 homodimer.
PLoS Pathog.
14
:
e1007001
.
134
Shin
,
D. L.
,
B.
Hatesuer
,
S.
Bergmann
,
T.
Nedelko
,
K.
Schughart
.
2015
.
Protection from severe influenza virus infections in mice carrying the Mx1 influenza virus resistance gene strongly depends on genetic background.
J Virol.
89
:
9998
10009
.
135
Gaio
,
V.
,
B.
Nunes
,
P.
Pechirra
,
P.
Conde
,
R.
Guiomar
,
C. M.
Dias
,
M.
Barreto
.
2016
.
Hospitalization risk due to respiratory illness associated with genetic variation at IFITM3 in patients with influenza A(H1N1)pdm09 infection: a case-control study.
PLoS One.
11
:
e0158181
.
136
Kim
,
Y. C.
,
B. H.
Jeong
.
2017
.
No correlation of the disease severity of influenza A virus infection with the rs12252 polymorphism of the interferon-induced transmembrane protein 3 gene.
Intervirology
60
:
69
74
.
137
Lee
,
N.
,
B.
Cao
,
C.
Ke
,
H.
Lu
,
Y.
Hu
,
C. H. T.
Tam
,
R. C. W.
Ma
,
D.
Guan
,
Z.
Zhu
,
H.
Li
, et al
.
2017
.
IFITM3, TLR3, and CD55 gene SNPs and cumulative genetic risks for severe outcomes in Chinese patients with H7N9/H1N1pdm09 influenza.
J. Infect. Dis.
216
:
97
104
.
138
Lopez-Rodriguez
,
M.
,
E.
Herrera-Ramos
,
J.
Sole-Violan
,
J. J.
Ruiz-Hernandez
,
L.
Borderias
,
J. P.
Horcajada
,
E.
Lerma-Chippirraz
,
O.
Rajas
,
M.
Briones
,
M. C.
Perez-Gonzalez
, et al
.
2016
.
IFITM3 and severe influenza virus infection. No evidence of genetic association.
Eur. J. Clin. Microbiol. Infect. Dis.
35
:
1811
1817
.
139
Pan
,
Y.
,
P.
Yang
,
T.
Dong
,
Y.
Zhang
,
W.
Shi
,
X.
Peng
,
S.
Cui
,
D.
Zhang
,
G.
Lu
,
Y.
Liu
, et al
.
2017
.
IFITM3 Rs12252-C variant increases potential risk for severe influenza virus infection in Chinese population.
Front Cell Infect Microbiol.
7
:
294
.
140
Prabhu
,
S. S.
,
T. T.
Chakraborty
,
N.
Kumar
,
I.
Banerjee
.
2018
.
Association between IFITM3 rs12252 polymorphism and influenza susceptibility and severity: a meta-analysis.
Gene
674
:
70
79
.
141
Randolph
,
A. G.
,
W. K.
Yip
,
E. K.
Allen
,
C. M.
Rosenberger
,
A. A.
Agan
,
S. A.
Ash
,
Y.
Zhang
,
T. R.
Bhangale
,
D.
Finkelstein
,
N. Z.
Cvijanovich
, et al
Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network Pediatric Influenza (PICFLU) Investigators
.
2017
.
Evaluation of IFITM3 rs12252 association with severe pediatric influenza infection.
J. Infect. Dis.
216
:
14
21
.
142
Xuan
,
Y.
,
L. N.
Wang
,
W.
Li
,
H. R.
Zi
,
Y.
Guo
,
W. J.
Yan
,
X. B.
Chen
,
P. M.
Wei
.
2015
.
IFITM3 rs12252 T>C polymorphism is associated with the risk of severe influenza: a meta-analysis.
Epidemiol Infect.
143
:
2975
2984
.
143
Yang
,
X.
,
B.
Tan
,
X.
Zhou
,
J.
Xue
,
X.
Zhang
,
P.
Wang
,
C.
Shao
,
Y.
Li
,
C.
Li
,
H.
Xia
,
J.
Qiu
.
2015
.
Interferon-inducible transmembrane protein 3 genetic variant rs12252 and influenza susceptibility and severity: a meta-analysis.
PLoS One.
10
:
e0124985
.
144
Carter
,
T. C.
,
S. J.
Hebbring
,
J.
Liu
,
J. D.
Mosley
,
C. M.
Shaffer
,
L. C.
Ivacic
,
S.
Kopitzke
,
E. L.
Stefanski
,
R.
Strenn
,
M. E.
Sundaram
, et al
.
2018
.
Pilot screening study of targeted genetic polymorphisms for association with seasonal influenza hospital admission.
J. Med. Virol.
90
:
436
446
.
145
Cheng
,
Z.
,
J.
Zhou
,
K. K.
To
,
H.
Chu
,
C.
Li
,
D.
Wang
,
D.
Yang
,
S.
Zheng
,
K.
Hao
,
Y.
Bossé
, et al
.
2015
.
Identification of TMPRSS2 as a susceptibility gene for severe 2009 pandemic A(H1N1) influenza and A(H7N9) influenza.
J. Infect. Dis.
212
:
1214
1221
.
146
Chen
,
Y.
,
J.
Zhou
,
Z.
Cheng
,
S.
Yang
,
H.
Chu
,
Y.
Fan
,
C.
Li
,
B. H.
Wong
,
S.
Zheng
,
Y.
Zhu
, et al
.
2015
.
Functional variants regulating LGALS1 (Galectin 1) expression affect human susceptibility to influenza A(H7N9).
Sci. Rep.
5
:
8517
.
147
Herrera-Ramos
,
E.
,
M.
López-Rodríguez
,
J. J.
Ruíz-Hernández
,
J. P.
Horcajada
,
L.
Borderías
,
E.
Lerma
,
J.
Blanquer
,
M. C.
Pérez-González
,
M. I.
García-Laorden
,
Y.
Florido
, et al
.
2014
.
Surfactant protein A genetic variants associate with severe respiratory insufficiency in pandemic influenza A virus infection.
Crit. Care
18
:
R127
.
148
Dudina
,
K. R.
,
M. M.
Kutateladze
,
N. O.
Bokova
,
O. O.
Znoiko
,
D. D.
Abramov
,
E. I.
Kelly
,
N. D.
Yuschuk
.
2015
.
Association of polymorphism genes of surfactant proteins in patients with influenza
.
Zh. Mikrobiol. Epidemiol. Immunobiol.
71
77
.
149
To
,
K. K. W.
,
J.
Zhou
,
Y. Q.
Song
,
I. F. N.
Hung
,
W. C. T.
Ip
,
Z. S.
Cheng
,
A. S. F.
Chan
,
R. Y. T.
Kao
,
A. K. L.
Wu
,
S.
Chau
, et al
.
2014
.
Surfactant protein B gene polymorphism is associated with severe influenza.
Chest.
145
:
1237
1243
.
150
Zhou
,
J.
,
K. K.
To
,
H.
Dong
,
Z. S.
Cheng
,
C. C.
Lau
,
V. K.
Poon
,
Y. H.
Fan
,
Y. Q.
Song
,
H.
Tse
,
K. H.
Chan
, et al
.
2012
.
A functional variation in CD55 increases the severity of 2009 pandemic H1N1 influenza A virus infection.
J. Infect. Dis.
206
:
495
503
.
151
Zúñiga
,
J.
,
I.
Buendía-Roldán
,
Y.
Zhao
,
L.
Jiménez
,
D.
Torres
,
J.
Romo
,
G.
Ramírez
,
A.
Cruz
,
G.
Vargas-Alarcon
,
C. C.
Sheu
, et al
.
2012
.
Genetic variants associated with severe pneumonia in A/H1N1 influenza infection.
Eur. Respir. J.
39
:
604
610
.
152
Romanova
,
E. N.
,
A. V.
Govorin
.
2013
.
TNF-α, IL-10, and eNOS gene polymorphisms in patients with influenza A/H1N1 complicated by pneumonia
.
Ter. Arkh.
85
:
58
62
.
153
Liu
,
Y.
,
S.
Li
,
G.
Zhang
,
G.
Nie
,
Z.
Meng
,
D.
Mao
,
C.
Chen
,
X.
Chen
,
B.
Zhou
,
G.
Zeng
.
2013
.
Genetic variants in IL1A and IL1B contribute to the susceptibility to 2009 pandemic H1N1 influenza A virus.
BMC Immunol.
14
:
37
.
154
Garcia-Ramirez
,
R. A.
,
A.
Ramirez-Venegas
,
R.
Quintana-Carrillo
,
A. E.
Camarena
,
R.
Falfan-Valencia
,
J. M.
Mejia-Arangure
.
2015
.
TNF, IL6, and IL1B polymorphisms are associated with severe influenza A (H1N1) virus infection in the Mexican population.
PLoS One.
10
:
e0144832
.
155
Rogo
,
L. D.
,
F.
Rezaei
,
S. M.
Marashi
,
M. S.
Yekaninejad
,
M.
Naseri
,
N.
Ghavami
,
T.
Mokhtari-Azad
.
2016
.
Seasonal influenza A/H3N2 virus infection and IL-1Beta, IL-10, IL-17, and IL-28 polymorphisms in Iranian population.
J. Med. Virol.
88
:
2078
2084
.
156
Esposito
,
S.
,
C. G.
Molteni
,
S.
Giliani
,
C.
Mazza
,
A.
Scala
,
L.
Tagliaferri
,
C.
Pelucchi
,
E.
Fossali
,
A.
Plebani
,
N.
Principi
.
2012
.
Toll-like receptor 3 gene polymorphisms and severity of pandemic A/H1N1/2009 influenza in otherwise healthy children.
Virol. J.
9
:
270
.
157
Aranda-Romo
,
S.
,
C. A.
Garcia-Sepulveda
,
A.
Comas-Garcia
,
F.
Lovato-Salas
,
M.
Salgado-Bustamante
,
A.
Gomez-Gomez
,
D. E.
Noyola
.
2012
.
Killer-cell immunoglobulin-like receptors (KIR) in severe A (H1N1) 2009 influenza infections.
Immunogenetics.
64
:
653
662
.
158
La
,
D.
,
C.
Czarnecki
,
H.
El-Gabalawy
,
A.
Kumar
,
A. F.
Meyers
,
N.
Bastien
,
J. N.
Simonsen
,
F. A.
Plummer
,
M.
Luo
.
2011
.
Enrichment of variations in KIR3DL1/S1 and KIR2DL2/L3 among H1N1/09 ICU patients: an exploratory study.
PLoS One
6
:
e29200
.
159
Falcon
,
A.
,
M. T.
Cuevas
,
A.
Rodriguez-Frandsen
,
N.
Reyes
,
F.
Pozo
,
S.
Moreno
,
J.
Ledesma
,
J.
Martinez-Alarcon
,
A.
Nieto
,
I.
Casas
.
2015
.
CCR5 deficiency predisposes to fatal outcome in influenza virus infection.
J. Gen. Virol.
96
:
2074
2078
.
160
Sironi
,
M.
,
R.
Cagliani
,
C.
Pontremoli
,
M.
Rossi
,
G.
Migliorino
,
M.
Clerici
,
A.
Gori
.
2014
.
The CCR5Δ32 allele is not a major predisposing factor for severe H1N1pdm09 infection.
BMC Res. Notes
7
:
504
.
161
Maestri
,
A.
,
M. C.
dos Santos
,
E. M.
Ribeiro-Rodrigues
,
W. A.
de Mello
,
R. C.
Sousa
,
S. E.
dos Santos
,
V. A.
Sortica
.
2015
.
The CCR5Δ32 (rs333) polymorphism is not a predisposing factor for severe pandemic influenza in the Brazilian admixed population.
BMC Res. Notes
8
:
326
.
162
Maestri
,
A.
,
V. A.
Sortica
,
L.
Tovo-Rodrigues
,
M. C.
Santos
,
L.
Barbagelata
,
M. R.
Moraes
,
W.
Alencar de Mello
,
L.
Gusmao
,
R. C.
Sousa
,
S.
Emanuel Batista Dos Santos
.
2015
.
Siaalpha2-3Galbeta1- receptor genetic variants are associated with influenza A(H1N1)pdm09 Severity.
PLoS One.
10
:
e0139681
.
163
Sologuren
,
I.
,
M. T.
Martinez-Saavedra
,
J.
Sole-Violan
,
E.
de Borges de Oliveira
Jr.
,
E.
Betancor
,
I.
Casas
,
C.
Oleaga-Quintas
,
M.
Martinez-Gallo
,
S. Y.
Zhang
,
J.
Pestano
, et al
.
2018
.
Lethal influenza in two related adults with inherited GATA2 deficiency.
J. Clin. Immunol.
38
:
513
526
.

The authors have no financial conflicts of interest.