Innate immune control of influenza virus interspecies adaptation via ifitm3

Innate immune control of influenza virus interspecies adaptation via ifitm3

Play all audios:

Loading...

ABSTRACT Influenza virus pandemics are caused by viruses from animal reservoirs that adapt to efficiently infect and replicate in human hosts. Here, we investigate whether Interferon-Induced


Transmembrane Protein 3 (IFITM3), a host antiviral factor with known human deficiencies, plays a role in interspecies virus infection and adaptation. We find that IFITM3-deficient mice and


human cells can be infected with low doses of avian influenza viruses that fail to infect WT counterparts, identifying a new role for IFITM3 in controlling the minimum infectious virus dose


threshold. Remarkably, influenza viruses passaged through _Ifitm3_−/− mice exhibit enhanced host adaptation, a result that is distinct from viruses passaged in mice deficient for interferon


signaling, which exhibit attenuation. Our data demonstrate that IFITM3 deficiency uniquely facilitates potentially zoonotic influenza virus infections and subsequent adaptation, implicating


IFITM3 deficiencies in the human population as a vulnerability for emergence of new pandemic viruses. SIMILAR CONTENT BEING VIEWED BY OTHERS IKKΕ ISOFORM SWITCHING GOVERNS THE IMMUNE


RESPONSE AGAINST EV71 INFECTION Article Open access 02 June 2021 DEFINING THE BALANCE BETWEEN OPTIMAL IMMUNITY AND IMMUNOPATHOLOGY IN INFLUENZA VIRUS INFECTION Article 02 May 2024 IFITM3


RESTRICTS VIRUS-INDUCED INFLAMMATORY CYTOKINE PRODUCTION BY LIMITING NOGO-B MEDIATED TLR RESPONSES Article Open access 08 September 2022 INTRODUCTION Respiratory virus emergence from animal


reservoirs into the human population is a continuous worldwide health threat. The influenza pandemics of 1918, 1957, 1968, and 2009 arose from viruses of avian or swine origin that adapted


to replicate robustly in humans1,2. Likewise, infection of humans with coronaviruses, such as MERS-CoV, SARS-CoV, and SARS-CoV-2, likely resulted from animal-to-human transmission events


followed by virus adaptation to the human respiratory tract and other niches3. Despite these notable examples, major outbreaks of new viruses could be considered rare given the routine


contact between humans and wildlife throughout the world4. This discrepancy suggests that natural impediments to interspecies virus transmission or adaptation exist in human cells.


Conversely, genetic defects in such host defense mechanisms could provide opportunities for viruses to enter and more efficiently adapt to humans. Here, we tested the hypothesis that


deficiencies in the innate immunity protein, Interferon-Induced Transmembrane Protein 3 (IFITM3), facilitate interspecies infection and adaptation by influenza viruses. IFITM3 is expressed


at low levels in most cell types and is highly upregulated by interferons upon virus infection5,6. IFITM3 associates with cell membranes through a transmembrane domain and an


_S_-palmitoylated amphipathic helix domain6,7. The amphipathic helix alters membrane curvature, lipid composition, and fluidity in a manner that disfavors virus-to-cell membrane fusion,


endowing IFITM3 with the ability to block the release of virus genomes into the cytoplasm of cells7,8,9,10,11,12. As such, IFITM3 limits infection by a multitude of enveloped viruses in


vitro, including influenza virus, Zika virus, and SARS-CoV-25,13,14,15. In humans, there are two deleterious single nucleotide polymorphisms (SNPs) in the _IFITM3_ gene that negatively


impact its splicing or gene promoter efficacy16,17,18. These SNPs are unexpectedly common in the human population, with 20% of Chinese individuals being homozygous for the splicing SNP


(rs12252-C), and 4% of Europeans being homozygous for the promoter SNP (rs34481144-A)18. Human IFITM3 deficiencies are associated with heightened severity of influenza, and data are emerging


that IFITM3 deficiencies are a risk factor for severe COVID-1916,17,18,19,20. Indeed, we and other groups have observed that _Ifitm3_−/− mice experience more severe influenza virus and


SARS-CoV-2 infections, mimicking human IFITM3 defects15,16,21,22,23. Interestingly, IFITM3 deficiencies in humans may have the most profound effects in emergent virus infections, e.g.,


pandemic 2009 H1N1 influenza virus16,17,24, avian H7N9 influenza virus19, HIV25, and SARS-CoV-216,17,18,19,20. Some studies examining seasonal influenza have not observed major impacts of


the _IFITM3_ SNPs on infection severity26,27,28, suggesting that pre-existing adaptive immunity may compensate for diminished IFITM3 activity in some circumstances. We hypothesized that


IFITM3 deficiency removes a major block to initial zoonotic influenza virus infections and to subsequent viral replication, thus promoting virus evolution and the emergence of


species-specific adaptive mutations. Herein, we demonstrate that IFITM3 deficiency not only lowers the infectious dose threshold for influenza viruses in mice and human cells but also


promotes influenza virus adaptation to a new species. These functions of IFITM3 could not be inferred from its known roles, as virus passaging in the absence of interferon signaling, which


also enhances virus replication, resulted in virus attenuation. Our work indicates that human IFITM3 deficiencies are a unique “Achilles heel” for the emergence of new pandemic viruses and


should be considered in pandemic prevention efforts. RESULTS IFITM3 DEFICIENCY LOWERS THE MINIMUM INFECTIOUS DOSE OF AVIAN INFLUENZA VIRUSES IN MICE Intrinsic immunity is presumed to be


among the factors responsible for preventing infection when virus inoculums are below a minimum infectious dose threshold. However, this fundamental concept lacks concrete experimental


evidence. IFITM3 limits the severity of influenza virus infections in humans and mice16,17,21,22,23, but whether it influences the minimum viral dose required for productive infection in


vivo has not been tested. We infected WT and _Ifitm3__−/−_ mice with doses of 1, 10, or 50 TCID50 of H5N1 avian influenza virus and measured lung viral loads at day 3 post-infection.


Infection with 1 TCID50 of H5N1 influenza virus resulted in significant viral replication in the lungs of _Ifitm3__−/−_ mice, approaching titers of 104 TCID50/mL, while live virus was not


detected in the lungs of WT mice (Fig. 1a). Doses of 10 and 50 TCID50 caused detectable viral loads in both WT and KO mice, with KO mice showing significantly higher viral titers at both


doses (Fig. 1a). Examination of lung IL-6 and IFNβ as measures of inflammation induced by viral replication substantiated these titer results. Both lung IL-6 and IFNβ were nearly


undetectable in WT mice infected with 1 TCID50 but were significantly above baseline in all other samples, including _Ifitm3__−/−_ mice infected with only 1 TCID50 (Fig. 1b, c). We next


infected WT and KO mice with a second avian virus using doses of 1 and 10 TCID50. We again observed that 1 TCID50 of the H7N3 virus was sufficient to infect _Ifitm3__−/−_ mice as indicated


by robust replication and induction of IL-6 and IFNβ, whereas the same viral dose did not productively infect WT mice (Fig. 1d–f). A dosage of 10 TCID50 resulted in productive virus


replication in both WT and _Ifitm3__−/−_ mice, though viral titers and cytokine induction were each higher in the KO animals (Fig. 1d–f). IFITM3 DEFICIENCY LOWERS THE MINIMUM INFECTIOUS DOSE


OF AVIAN INFLUENZA VIRUSES IN HUMAN CELLS To extend our conclusions to a human system, we tested a broad panel of influenza viruses isolated from animals in a relevant human cell type, A549


lung epithelial cells. For our study, we compared infection rates of 11 avian, 3 swine, and 2 human influenza viruses representing distinct and diverse hemagglutinin protein (HA) subtypes


(strain details in Supplementary Table 1 and HA-based phylogenetic tree in Supplementary Fig. 1). Notably, 2 of the swine viruses are known to have infected humans making them confirmed


zoonotic viruses29,30. We first examined single-cycle infections of these viruses in the absence of trypsin in control A549 cells versus those in which IFITM3 was depleted by shRNA knockdown


(Fig. 2a). The gating strategy for determining infected A549 cells is included in Supplementary Fig. 2a. The IFITM3-knockdown cells were universally infected at a higher rate (Fig. 2b, c, e


and Supplementary Fig. 3), demonstrating that each of these diverse viruses was restricted by IFITM3 in human cells. Furthermore, IFITM3-knockdown cells maintained higher infection


susceptibility than control cells after treatment with type I interferon (Fig. 2b, c, e and Supplementary Fig. 3), demonstrating that IFITM3 plays a critical and non-redundant role in the


human antiviral interferon response against diverse influenza viruses. Since macrophages are also a relevant target of influenza virus31, we examined infection of differentiated human THP-1


cells. Once again, infection by the human-, avian-, and swine-origin viruses was significantly potentiated in the human macrophages lacking IFITM3 with or without interferon treatment (Fig. 


2b, d, e; Supplementary Fig. 2b and Supplementary Fig. 4). Increased infection was also observed for IFITM3 KO HAP1 fibroblasts (Supplementary Fig. 2c and Supplementary Fig. 5) and for


IFITM1/2/3 KO HeLa cells (Supplementary Fig. 2d, Supplementary Fig. 6). Of note, we determined sialic acid receptor binding preferences for viruses we tested and found that the human- and


swine-origin viruses showed a preference for a-2,6-linked sialic acid while many of the avian viruses showed a preference for a-2,3-linked sialic acid (Supplementary Table 1). Despite these


differences in receptor binding preference, all the viruses we examined were able to infect every cultured human cell type that we tested (Fig. 2b, e and Supplementary Figs. 2–6). Overall,


these data demonstrate that IFITM3 deficiency broadly increases infection of human cells with influenza viruses of animal origin. To further examine the impact of IFITM3 on minimum


infectious dosing, we infected A549 lung cells with or without IFITM3 knockdown using H5N1 or H7N3 avian influenza viruses at a range of doses from MOI 0.001 to 10. We observed that infected


cells could be detected at lower viral doses for both H5N1 (Fig. 3a) and H7N3 (Fig. 3b) when IFITM3 was deficient. To further examine the effects of IFITM3, we infected both control and


IFITM3-knockdown A549 cells with a selection of viruses, including 2 human-origin, 3 swine-origin (2 with known zoonotic transmission29,30), and 2 avian-origin viruses at an MOI of 1. The


cells were incubated with TPCK-trypsin to allow for multiple rounds of infection, and at 48 h post infection, we collected the supernatants to determine viral titers by TCID50 assay. For all


viruses, we measured significantly higher titers produced by IFITM3-knockdown versus control cells (Fig. 3c). Along with data from Fig. 1, these data establish that IFITM3 prevents


animal-origin influenza virus infection in vitro and in vivo when the virus dose is below a minimum threshold and that influenza virus replicates to higher titers in the absence of IFITM3.


These data overall demonstrate that IFITM3 increases the minimum infectious dose necessary to achieve a productive influenza virus infection. IFITM3 DEFICIENCY FACILITATES THE ADAPTATION OF


INFLUENZA VIRUSES TO A NEW HOST SPECIES IN VIVO Several factors have been proposed to favor inter-species adaptation of a virus to a new host, including levels of virus replication and


infectious doses. Given that IFITM3 deficiency impacts both of these features, we tested whether its absence allows influenza virus to adapt more readily in vivo. With biosafety in mind, we


did not attempt to adapt avian influenza virus to a mammalian host since such adaptations could increase virus replication across mammalian species, including humans, and could thus be


considered dual-use research of concern32. Rather, we sought to adapt viruses of human origin to mice, which has been performed safely for nearly a century in the influenza virus research


field32. Initially, we performed lung-to-lung passaging of influenza virus strain A/Victoria/361/2011 (H3N2) 10 times through WT or _Ifitm3_−/− mice using intranasal infection and allowing 3


days for the virus to replicate between successive passages (Fig. 4a). We chose this strain because preliminary experiments indicated that it infects mice, but replicates to lower levels


than commonly used mouse-adapted strains, thus providing a virus with significant potential for adaptation to mice. We propagated passages 1, 5, and 10 from both WT and _Ifitm3_−/− mice, as


well as the parental virus stock (passage 0), in MDCK cells. We then used these expanded stocks to challenge WT mice with equal virus doses. 1000 TCID50 of passages 1, 5, and 10 from WT and


KO mice, along with parental virus (passage 0), were used to infect WT mice for 7 days, at which point mice were sacrificed to measure viral titers and inflammatory cytokines (Fig. 4b). To


reiterate, passaged viruses, whether generated in WT or KO mice, were tested for adaptation in WT animals. We found that, as expected, the ability to replicate in mouse lungs was unchanged


for the earliest passage of virus from WT or KO mice as compared to the parental virus (Fig. 4c). However, passage 5 from KO mice exhibited an upward trend in viral titers and KO passage 10


showed a statistically significant, >1 log average increase in lung viral titers (Fig. 4c). In contrast, virus passages 5 and 10 from WT mice did not show significantly enhanced


replication capacity compared to the parental virus (Fig. 4c). As an independent metric for corroborating virus replication, we measured levels of the inflammatory cytokines IL-6 and IFNβ in


lungs during infection, which mirrored viral loads, with IFITM3 KO passage 10 inducing a statistically significant increase in both IL-6 (Fig. 4d) and IFNβ (Fig. 4e) compared to infection


with parent virus. Since virus adaptation is a stochastic process, we independently repeated our H3N2 virus passaging and testing a second time and obtained similar results indicating


enhanced adaptation occurring in IFITM3 KOs (Fig. 4f–h). Interestingly, the animals infected with both of the H3N2 virus passage series did not lose weight despite increased viral titers


seen for the IFITM3 KO-passaged virus stocks, suggesting only partial adaptation. We also tested the passaged virus stocks for replication fitness in A549 human lung cells versus LET1 mouse


lung cells. We found that the IFITM3 KO-passaged viruses showed increased replication in the murine LET1 cells compared to the parent virus, but replication in human A549 cells was not


changed (Supplementary Fig. 7). These results are in agreement with expectations that mouse adaptation would not provide a gain of function for infection and replication in human cells.


Overall, our data from both passage series demonstrate that influenza virus gained enhanced replication capacity and induction of inflammation when passed through IFITM3 deficient versus WT


hosts. We next tested whether IFITM3 deficiency is unique in its ability to facilitate virus adaptation or whether passaging in another immune-compromised system that allows enhanced virus


replication would also promote interspecies adaptation. The interferon system is particularly important in this regard as interferons widely inhibit virus replication33. Further, it has


become increasingly apparent throughout the recent COVID-19 pandemic that genetic and antibody-mediated interferon deficiencies are significantly more common in the human population than


previously appreciated34,35. We thus repeated our passaging and challenge regimen with influenza virus A/Victoria/361/2011 (H3N2), comparing passaging through WT versus _Stat1__−/−_ mice,


which lack signaling downstream of all interferon receptors (Supplementary Fig. 8a, b). Consistent with our previous results, we did not observe significant adaptation of the virus passed


through WT mice (Supplementary Fig. 8c). In contrast, passage 5 from the STAT1 deficient mice exhibited a downward trend in viral replication and _Stat1__−/−_ passage 10 showed a


statistically significant reduction in the ability to replicate in WT mice (Supplementary Fig. 8c). The titer data were supported by measurement of the inflammatory cytokine IL-6 in that


STAT1 KO passage 10 showed a significant decrease in IL-6 levels compared to other groups (Supplementary Fig. 8d). Remarkably, IL-6 levels were uncoupled from IFNβ levels (Supplementary Fig.


 8e), which were highest for the STAT1 KO-passaged virus, suggesting that this stock induces increased IFNβ, which may account for decreased virus replication. Additionally, when testing the


passaged virus’s ability to replicate in A549 versus LET1 cells, we saw that STAT1 KO-passaged viruses showed reduced replication in LET1 cells compared to the parent virus, with two out of


six samples showing replication below detection limits, while replication in A549 cells was unchanged (Supplementary Fig. 8f, g) Our findings are consistent with the principle that, in the


absence of selective pressures from the interferon system, viral interferon antagonism mechanisms become less active, resulting in virus attenuation when infecting WT systems. Importantly,


these results underscore how IFITM3 may be a unique vulnerability in innate immune response pathways that, when impaired, can promote viral adaptation to new hosts rather than viral


attenuation. To further investigate the generality of our findings on IFITM3, we repeated the passaging regimen in WT and _Ifitm3_−/− mice using pandemic influenza virus strain


A/California/04/2009 (H1N1), followed by adaptation testing in WT mice. Both WT passage 5 and 10 virus stocks were modestly enhanced in replicative capacity (Fig. 5a). We saw that both WT


and KO passage 10 virus stocks induced increased weight loss in infected mice compared to the parent H1N1, with the IFITM3 KO passage 10 mice inducing the most dramatic loss of body weight


(Fig. 5b). WT passage 5 and 10 virus stocks exhibited no significant changes in their IL-6 and IFNβ induction capacity (Fig. 5c, d). Consistent with our previous results, virus passaged 10


times in _Ifitm3_−/− mice replicated to higher titers compared to the parental strain and to the WT mouse passaged viruses (Fig. 5a). In addition to the highest viral titer, KO passage 10


also induced robust levels of IL-6 and IFNβ in infected lungs (Fig. 5c, d). In a second series of passages with the H1N1 virus, the most robust adaptation in terms of viral replication,


weight loss, and inflammatory cytokine induction was again seen for KO passage 10 (Fig. 5e–h). Notably, this adapted viral stock became highly virulent, with all infected mice meeting humane


endpoint criteria of >30% weight loss by day 6 post infection (Fig. 5f). Similar to our results with the H3N2 passages, infecting human A549 versus murine LET1 lung cells with the H1N1


passages showed increased replication and IFNβ induction in LET1 cells for the IFITM3 KO-passaged virus whereas we did not observe increased replication in the human cells (Supplementary


Fig. 9), again confirming that mouse adaptation of human influenza viruses does not result in a gain of function in terms of increasing virus replication in human cells. To investigate the


specific changes within the passaged viruses that may contribute to the observed mouse adaptation, we extracted viral RNA and sequenced the genomes of passages 5 and 10 from all H1N1 and


H3N2 series with comparison to parental viruses. Despite enhanced replication of the H3N2 IFITM3 KO passage 10 viruses, we did not observe changes to the consensus sequence of those viruses


( > 50% prevalence). Instead, we observed the emergence of minor variants, particularly in polymerase segments, that may contribute to the observed phenotypes (Supplementary Data 1, 2).


Notably, the second H3N2 passage series showed a trend toward adaptation for the virus passaged 10 times through WT mice in terms of virus replication, though this was not to the same


magnitude as the IFITM3 KO-passaged virus and was not statistically significant. This may also be due to the contribution of minor variants present within the quaispecies of that passaged


virus (Supplementary Data 1 and 2). The H3N2 virus passaged through _Stat1__−/−_ mice also showed the emergence of minor variants in polymerase segments that are distinct from those present


in the IFITM3 KO-passaged viruses (Supplementary Data 1 and 3). These results are consistent with the H3N2 stocks being in the early stages of adaptation as supported by a lack of weight


loss when infecting with these viruses and titers that, while increased for IFITM3 KO-passaged viruses, did not reach the levels achieved by the adapted H1N1 stocks. In contrast to the H3N2


sequences, the H1N1 passages showed clear changes within the consensus sequences in addition to minor variants (Supplementary Data 4 and 5). The trend toward enhanced replication of WT


passages 5 and 10 in passage series 1 was associated with two amino acid changes in the viral NP (D101G, R102G) and a single change in PB2 (E158G) that is a known polymerase


activity-increasing adaptive mutation in mice36,37. The H1N1 series 1 KO passages 5 and 10 possessed the same adaptive PB2 E158G mutation seen in WT passages 5 and 10. However, the


heightened replication and increased induction of inflammatory cytokines by KO passage 10 relative to KO passage 5 was associated with a single change in the PA polymerase subunit (N373H),


representing a novel mutation not previously reported in mouse adaptation studies (Supplementary Data 4). The high virulence of H1N1 series 2 KO passage 10 was linked to a single consensus


sequence change, specifically an E158A mutation in PB2 (Supplementary Data 5) that, similarly to the PB2 E158G mutation detected in our first series of H1N1 passages, is a known


mouse-adaptive mutation37. Overall, our examinations of replication levels and weight loss, along with sequencing data, suggest that we’ve captured the two viruses (H3N2 and H1N1) at


different stages of adaptation, though in each case, this process occurs more rapidly in IFITM3 KO mice. Together with data for the H3N2 virus, our H1N1 passaging experiments demonstrate


that the adaptation of influenza viruses to a new host species is accelerated in the absence of IFITM3, fueled by mutations that readily emerge in the compromised host. DISCUSSION Three


fundamentally important discoveries regarding IFITM3 emerged from our study. First, IFITM3 raises the minimum infectious dose of influenza virus required to achieve a detectable infection in


human cells, as well as a productive infection in vivo. Second, IFITM3 serves as a critical barrier to pandemic virus emergence by limiting the adaptation of influenza viruses to new host


species, whereas global defects in interferon responses lead to virus attenuation. Third, IFITM3, whether basally expressed or induced by interferon, broadly restricts infection of human


cells by potentially zoonotic influenza viruses, and this function was independent of viral receptor binding preferences. Each of these findings supports the premise that human IFITM3


deficiencies represent a critical vulnerability for the entry and adaptation of animal-origin influenza viruses in the population. While we did not perform virus transmission experiments,


our work indirectly addresses the transmission of influenza viruses. Mice are unable to transmit influenza virus to one another, even when in close contact32. We have also observed in


unpublished work that IFITM3 KO mice, perhaps the most susceptible mouse model of influenza, do not transmit mouse-adapted influenza virus or mouse-adapted SARS-CoV-2 to non-infected,


cohoused KO animals. Nonetheless, we found that the minimal in vivo infectious dose of avian influenza virus, a critical aspect of the transmission process, was lowered in the absence of


IFITM3. Specifically, _Ifitm3_−/− animals showed significant replication of the virus in their lungs after infection with only 1 TCID50 of H5N1 or H7N3 virus, while titers were undetectable


in WT lungs when infected at the same dose. These results provide experimental evidence for the long-presumed concept that minimum infectious doses of the virus are set by levels of


resistance from the innate immune system. Specifically, we show that IFITM3 underlies sterilizing immunity that prevents infection at low influenza virus doses that are below a minimum


threshold. We designed our virus passaging experiments to model transmission of a zoonotic virus through a family or community in which deleterious _IFITM3_ SNPs are prevalent. One caveat to


this approach is that, while our engineered mice are fully devoid of IFITM3 production, deleterious SNPs in human _IFITM3_ substantially reduce IFITM3 levels or activity, but do not ablate


the protein16,17. In this regard, lung cells partially deficient in IFITM3 (IFITM3-knockdown A549 cells in Fig. 2) were indeed significantly more susceptible to diverse influenza virus


strains. While infections in humans with IFITM3 deficiencies may represent more nuanced scenarios, experimentation with complete KO mice allowed us to unambiguously identify important and


relevant functionality of IFITM3 in limiting interspecies infection and adaptation of viruses to a new host species in vivo, key findings that are likely germane to preventing pandemics in


humans. It is also interesting to note that distinct _IFITM3_ SNPs were independently selected in both Chinese and European populations. While the high prevalence of _IFITM3_ SNPs in these


groups is puzzling, given their association with increased viral disease severity, a negative role for IFITM3 in placental development may explain their selection and maintenance in these


populations. Indeed, IFITM3 prevents the endogenous retrovirus fusogen-mediated cell-to-cell fusion of placental trophoblasts required for proper placental architecture and function38,39. In


this way, IFITM3 mediates a significant portion of the fetal toxicity of interferons produced during pregnancy-associated infections38,40. Increased fetal survival during certain infections


in the absence of IFITM3 thus represents the prevailing theory for how IFITM3 deficiencies may provide a selective benefit despite increasing susceptibility to emergent virus infections


later in life. RNA viruses generally adapt to new host species by producing mutations arising from error-prone polymerases41. Beneficial mutations that allow the virus to replicate more


efficiently are enriched by outcompeting the parental virus and other mutants. Indeed, influenza virus and several other viruses replicate to higher titers in _Ifitm3_−/− versus WT


mice15,16,21,22,23,42,43,44, which represents a plausible mechanism for enhanced adaptation in these animals41. Similarly, our results indicate that IFITM3 deficiency also relaxes the


infection bottleneck present at initial inoculation. In our current study, mutations in the 2009 pandemic H1N1 virus passages that drive adaptation in mice were primarily observed in the


viral polymerase subunits. Indeed, PB2 mutations at the E158 position are consistently reported in mouse-adapted versions of this virus36,37. Moreover, the relative impact of E158 mutations


on polymerase activity is in accord with viral replication and virulence levels observed in our in vivo experiments. We also identified a novel N373H mutation in PA from one H1N1 passage


series, which emerged subsequent to the E158G mutation. The dual mutations were associated with enhanced replication in vivo compared to E158G alone. The mechanistic impact of the PA N373H


mutation remains to be determined and may provide future insights into influenza virus replication and adaptation. Interestingly, we do not see an overall increase in variants in our IFITM3


KO-passaged virus stocks. Rather, in all four of our IFITM3 KO passaging experiments, the stock generated from the 10th KO passage contains the fewest number of minor variants as compared to


parental virus and WT passages. This suggests that IFITM3 deficiency may promote viral adaptation by allowing adaptive variants to more efficiently outcompete other viruses in the


quasispecies, perhaps by allowing infection of more cells per animal as our in vitro experiments would suggest. The prevalence of genetic and immunological mechanisms that impair interferon


induction or signaling in humans garnered increased attention during the COVID-19 pandemic since 20% of all severe COVID-19 cases could be attributed to interferon defects34,35. Given that


IFITM3 expression is induced by interferon, such defects likely diminish its induction, as well as that of other antiviral proteins, which could allow increased replication of viruses and


enhanced adaptation to humans. However, passaging through interferon deficient systems often results in attenuated viruses that have lost interferon antagonism due to a lack of selective


pressure for their maintenance45,46. Indeed, our passaging of influenza virus in _Stat1_−/− animals resulted in virus attenuation, despite influenza virus replicating to high titers in


interferon deficient systems47. Additional studies on viral evolution in human populations and engineered animals will be required to address how broad interferon defects mechanistically


influence the adaptive evolution of influenza virus and other pathogens. While IFITM3 inhibited infection by all the influenza viruses in all cell types in our experiments, we observed that


the degree of inhibition varied to some extent depending on the individual viruses. Published work has suggested that differences of distinct influenza virus isolates in susceptibility to


IFITM3 may be due to differences in viral fusion pH optimums leading to fusion preferences in early or late endosomes where IFITM3 levels differ48. Importantly, influenza viruses capable of


fully evading IFITM3 restriction have not been identified to date. Likewise, unlike the well-characterized antagonism of interferon by the influenza virus NS1 protein47, influenza virus


antagonism of IFITM3 has not been reported. Thus, in contrast to interferon, IFITM3 is unlikely to contribute similar selective pressures on influenza virus, allowing the virus to


efficiently adapt in its absence. IFITM3 deficiency may, therefore, represent a particularly unique vulnerability for the entry and adaptation of viruses in a new host species. Broad testing


of IFITM3 status in the human population could bolster pandemic prevention efforts by allowing vulnerable individuals to receive greater vaccine coverage or exercise enhanced precautions


when encountering animal reservoirs of infection. METHODS Our research complies with all relevant ethical regulations and was approved by the Institutional Biosafety Committee (IBC) and


Institutional Animal Care and Use Committee (IACUC) of The Ohio State University under protocol number 2016A00000051-R2. CELL CULTURE Control and IFITM3 knockdown A549 cells were generated


by lentiviral shRNA-mediated targeting using previously described constructs and methods49. THP-1 IFITM3 knockout cells were generated via CRISPR-Cas9 targeting (provided by Dr. Anasuya


Sarkar of the Ohio State University). HeLa IFITM1/2/3 knockout and HAP1 IFITM3 knockout were purchased from ATCC (CRL-3452) and Horizon Discovery Biosciences (HZGHC004186c010), respectively.


MDCK cells were obtained from BEI Resources (NR-2628). LET1 cells were also obtained from BEI resources (NR-42941). Cells were maintained in DMEM medium (Fisher Scientific) supplemented


with 10% EquaFETAL bovine serum (Atlas Biologicals) except for the THP-1 cells, which were maintained in RPMI 1640 medium (Fisher Scientific) supplemented with 10% EquaFETAL bovine serum


(Atlas Biologicals). All cells were cultured in a humidified incubator at 37 °C with 5% CO2. VIRUS STOCKS AND IN VITRO INFECTION Influenza viruses A/Puerto Rico/8/34 (H1N1) (PR8, provided by


Dr. Thomas Moran of the Icahn School of Medicine at Mt. Sinai), A/Victoria/361/2011 (H3N2) (BEI Resources), and A/California/4/2009 (H1N1) (BEI Resources) were propagated in 10-day


embryonated chicken eggs (Charles River Laboratories) and titered on MDCK cells. The animal-origin viruses were collected by Andrew Bowman of the Ohio State University and were also


propagated in embryonated chicken eggs (Charles River Laboratories) and titered on MDCK cells. Infection of cell lines with the panel of animal-origin viruses (Fig. 2) was done at an MOI of


1 for 24 h in infection media without trypsin and represents a single-cycle infection. Cells that received Interferon-β pre-treatment were stimulated for 18 h with 100 units of recombinant


human Interferon-β protein (Millipore, IF014) prior to infection. All titration of virus stock was done using a TCID50 assay on MDCK cells, as previously described. Cells incubated with


TPCK-Trypsin (Worthington Biochemical Corporation, LS02123) were treated with 2 μg/mL for 48 h to allow for multicycle replication (Fig. 3 and Supplementary Figs 6-8). DETERMINATION OF VIRUS


RECEPTOR BINDING PREFERENCES 50 μL of horse (1%) or turkey (0.5%) RBCs (Innovative Research Inc, IHSRBC100P30M, and ITKRBC5P50ML, respectively) were added to 50 μL of 2-fold dilutions of


virus stocks in PBS and incubated for 1 h at room temperature to determine the hemagglutination titer for each virus. The turkey RBCs were either neuraminidase-treated to remove the α−2,3


sialyl linkages from glycoproteins and glycolipids or mock-treated. For neuraminidase treatment, 100 μL of a 5% Turkey RBC suspension prepared in PBS was treated with 15 units of α−2,3


neuraminidase S (New England Biolabs, P0743) for 1 h at 37 °C. The treated TRBCs were then diluted with PBS to a working concentration of 0.5% for the HA assay. MOUSE STUDIES All mice used


in this study were of the C57BL/6 J background. _Ifitm3__−/−_ mice with a 53 base pair deletion in exon 1 of the _Ifitm3_ gene were described previously23. WT mice for comparison to


_Ifitm3__−/−_ mice were obtained from Charles River Laboratories. _Stat1__−/−_ mice (Strain #: 012606) and complementary WT C57BL/6J mice (strain #007914) were obtained from The Jackson


Laboratory. Mice were housed in the Ohio State University’s Biomedical Research Tower vivarium, which is maintained at 68–76 degrees F, with a 12:12 light–dark cycle and humidity between


30–70%. Autoclaved individually ventilated cages (Allentown) were used for housing. Mice were fed irradiated natural ingredient chow ad libitum (Evnigo Teklad Diet 7912). Reverse osmosis


purified water was provided through an automated rack water system. Cages included 1⁄4 inch of corn cob bedding (Bed-o-Cobs, The Andersons) with cotton square nesting material. Male and


female mice between 6 and 10 weeks of age were used in our experiments. All mice in each individual experiment were age-matched. All mouse infections were performed intranasally under


anesthesia with isoflurane, delivering 25 μL of inoculum per nare. For virus passaging, mice were initially infected with doses of 1000 TCID50, and mouse organs were collected and


homogenized in 500 μL of sterile saline. Propagation and titering of passaged viruses, as well as parental control viruses, was done on MDCK cells. WT mice were infected with equal doses


(1000 TCID50) for testing of virus adaptation. Innoculums were prepared via dilution of viral stock in sterile PBS. For determining lung titers and cytokine levels in virus adaptation


studies, lungs were collected and homogenized in 1 mL of PBS, flash-frozen, and stored at −80 °C prior to titering on MDCK cells or analysis via ELISA. All procedures were approved by the


OSU IACUC and were performed in accordance with guidelines for the ethical use of animals. VIRUS SEQUENCING Viral RNA was extracted from MDCK-grown stocks using the RNeasy Mini Kit (Qiagen),


and cDNA was synthesized using the Superscript IV First-Strand Synthesis System (Invitrogen). The influenza A virus gene segments were amplified using modified universal primers in a


multi-segment PCR as previously described50. PCR products were purified using Agencourt AMPure XP beads according to the manufacturer’s protocol (BeckmanCoulter). Libraries were prepared


using the Nextera XT DNA Library Prep Kit (Illumina) according to the manufacturer’s protocol and sequenced using a MiSeq Reagent Kit v2 (300 cycles) on a MiSeq System (Illumina). Sequencing


reads were then quality trimmed and assembled using CLC Genomics Workbench (version 22.0.1). PHYLOGENETIC ANALYSIS The HA gene segments of the individual viruses used in this study were


downloaded from Genbank and used for analysis. Alignments and tree estimates for whole HA genomic segments were performed using Molecular Evolutionary Genetics Analysis (MEGA) software


v11.0.13. The sequences were aligned using the MUSCLE alignment. This was used to generate a neighbor-joining tree. WESTERN BLOTTING For detection of protein expression in cell lines,


samples were lysed in 1% SDS buffer (0.1 mM triethanolamine, 150 mM NaCl, 1% SDS (Sigma), pH 7.4) containing Roche Complete Protease Inhibitor Cocktail. The lysates were centrifuged at


16,000 × _g_ for 10 min, and soluble protein supernatants were used for Western blot analysis. Equal amounts of protein (30 μg) were separated by SDS-PAGE and transferred onto membranes.


Membranes were blocked with 10% non-fat milk in Phosphate-buffered saline with 0.1% Tween-20 (PBST) and probed with antibodies against IFITM1 (Cell Signaling Technology, #13126), IFITM2


(Cell Signaling Technology, #13530), IFITM3 (ProteinTech, 11714-1-AP), and GAPDH (Thermo Scientific, ZG003). Antibodies were used at dilutions of 1:1000 in PBST as determined by preliminary


experiments optimizing the protocol for western blotting. ELISAS ELISAs were performed on supernatant from mouse lung homogenates using the Mouse IL-6 Duoset ELISA kit from R&D Systems


(catalog # DY406) or the Mouse IFNβ Duoset ELISA kit from R&D Systems (catalog # DY8234-05) according to manufacturer’s instructions. FLOW CYTOMETRY Cells were fixed with


4%paraformaldehyde for 10 min at room temperature, permeabilized with 0.1% Triton X-100 in PBS, stained with an antibody against Influenza A Virus Nucleoprotein (Abcam, ab20343) conjugated


with Goat anti-Mouse IgG (H + L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor™ 647 (Thermo Scientific, Catalog # A-21236) resuspended in 2% FBS in PBS, and run on a BD FACS Canto II


flow cytometer. Antibodies were used at dilutions of 1:1000 in 0.1% Triton X-100 in PBS as determined by preliminary experiments optimizing the protocol for flow cytometry. Results were


analyzed using FlowJo Software (version 10.8.1). REPORTING SUMMARY Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article. DATA


AVAILABILITY All data are available in the main text or the supplementary materials. Source data are provided in this paper. The sequencing data generated in this study have been deposited


as the consensus sequence in the GenBank database under accession codes PQ384645—PQ384820.  Source data are provided in this paper. CHANGE HISTORY * _ 11 DECEMBER 2024 A Correction to this


paper has been published: https://doi.org/10.1038/s41467-024-54981-w _ REFERENCES * Bailey, E. S. et al. The continual threat of influenza virus infections at the human-animal interface:


What is new from a one health perspective? _Evol. Med. Public Health_ 2018, 192–198 (2018). Article  PubMed  PubMed Central  Google Scholar  * Taubenberger, J. K. & Kash, J. C. Influenza


virus evolution, host adaptation, and pandemic formation. _Cell Host Microbe_ 7, 440–451 (2010). Article  CAS  PubMed  PubMed Central  Google Scholar  * Enjuanes, L. et al. Nature of


viruses and pandemics: coronaviruses. _Curr. Res. Immunol._ 3, 151–158 (2022). Article  CAS  PubMed  PubMed Central  Google Scholar  * Dennehy, J. J. Evolutionary ecology of virus emergence.


_Ann. N. Y Acad. Sci._ 1389, 124–146 (2017). Article  ADS  PubMed  Google Scholar  * Brass, A. L. et al. The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile


virus, and dengue virus. _Cell_ 139, 1243–1254 (2009). Article  PubMed  PubMed Central  Google Scholar  * Yount, J. S. et al. Palmitoylome profiling reveals S-palmitoylation-dependent


antiviral activity of IFITM3. _Nat. Chem. Biol._ 6, 610–614 (2010). Article  CAS  PubMed  PubMed Central  Google Scholar  * Chesarino, N. M. et al. IFITM3 requires an amphipathic helix for


antiviral activity. _EMBO Rep._ 18, 1740–1751 (2017). Article  CAS  PubMed  PubMed Central  Google Scholar  * Guo, X. et al. Interferon-induced transmembrane protein 3 blocks fusion of


diverse enveloped viruses by altering mechanical properties of cell membranes. _ACS Nano_ 15, 8155–8170 (2021). Article  CAS  PubMed  PubMed Central  Google Scholar  * Li, K. et al. IFITM


proteins restrict viral membrane hemifusion. _PLoS Pathog._ 9, e1003124 (2013). Article  CAS  PubMed  PubMed Central  Google Scholar  * Desai, T. M. et al. IFITM3 restricts influenza A virus


entry by blocking the formation of fusion pores following virus-endosome hemifusion. _PLoS Pathog._ 10, e1004048 (2014). Article  PubMed  PubMed Central  Google Scholar  * Klein, S. et al.


IFITM3 blocks influenza virus entry by sorting lipids and stabilizing hemifusion. _Cell Host Microbe_ 31, 616–633.e620 (2023). Article  CAS  PubMed  Google Scholar  * Unali, G. et al.


Interferon-inducible phospholipids govern IFITM3-dependent endosomal antiviral immunity. _EMBO J._ 42, e112234 (2023). Article  CAS  PubMed  PubMed Central  Google Scholar  * Savidis, G. et


al. The IFITMS inhibit Zika virus replication. _Cell Rep._ 15, 2323–2330 (2016). Article  CAS  PubMed  Google Scholar  * Shi, G. et al. Opposing activities of IFITM proteins in SARS-CoV-2


infection. _EMBO J._ 40, e106501 (2021). Article  CAS  PubMed  Google Scholar  * Kenney, A. D. et al. Interferon-induced transmembrane protein 3 (IFITM3) limits lethality of SARS-CoV-2 in


mice. _EMBO Rep._ 24, e56660 (2023). Article  CAS  PubMed  PubMed Central  Google Scholar  * Everitt, A. R. et al. IFITM3 restricts the morbidity and mortality associated with influenza.


_Nature_ 484, 519–523 (2012). Article  CAS  PubMed  PubMed Central  Google Scholar  * Allen, E. K. et al. SNP-mediated disruption of CTCF binding at the IFITM3 promoter is associated with


risk of severe influenza in humans. _Nat. Med._ 23, 975–983 (2017). Article  CAS  PubMed  PubMed Central  Google Scholar  * Zani, A. & Yount, J. S. Antiviral protection by IFITM3 in


vivo. _Curr. Clin. Microbiol. Rep._ 5, 229–237 (2018). Article  PubMed  PubMed Central  Google Scholar  * Wang, Z. et al. Early hypercytokinemia is associated with interferon-induced


transmembrane protein-3 dysfunction and predictive of fatal H7N9 infection. _Proc. Natl Acad. Sci. USA_ 111, 769–774 (2014). Article  ADS  CAS  PubMed  Google Scholar  * Pecoraro, V.,


Cuccorese, M. & Trenti, T. Genetic polymorphisms of ACE1, ACE2, IFTM3, TMPRSS2 and TNFα genes associated with susceptibility and severity of SARS-CoV-2 infection: a systematic review and


meta-analysis. _Clin. Exp. Med_. 23, 1–14 (2023). * Bailey, C. C., Huang, I. C., Kam, C. & Farzan, M. Ifitm3 limits the severity of acute influenza in mice. _PLoS Pathog._ 8, e1002909


(2012). Article  CAS  PubMed  PubMed Central  Google Scholar  * Kenney, A. D. et al. Influenza virus replication in cardiomyocytes drives heart dysfunction and fibrosis. _Sci. Adv._ 8,


eabm5371 (2022). Article  CAS  PubMed  PubMed Central  Google Scholar  * Kenney, A. D. et al. IFITM3 protects the heart during influenza virus infection. _Proc. Natl Acad. Sci. USA_ 116,


18607–18612 (2019). Article  ADS  CAS  PubMed  PubMed Central  Google Scholar  * Zhang, Y. H. et al. Interferon-induced transmembrane protein-3 genetic variant rs12252-C is associated with


severe influenza in Chinese individuals. _Nat. Commun._ 4, 1418 (2013). Article  ADS  PubMed  Google Scholar  * Zhang, Y. H. et al. Interferon-induced transmembrane protein-3 rs12252-C is


associated with rapid progression of acute HIV-1 infection in Chinese MSM cohort. _Aids_ 29, 889–894 (2015). Article  CAS  PubMed  Google Scholar  * Randolph, A. G. et al. Evaluation of


IFITM3 rs12252 association with severe pediatric influenza infection. _J. Infect. Dis._ 216, 14–21 (2017). Article  CAS  PubMed  PubMed Central  Google Scholar  * Mills, T. C. et al. IFITM3


and susceptibility to respiratory viral infections in the community. _J. Infect. Dis._ 209, 1028–1031 (2014). Article  CAS  PubMed  Google Scholar  * Carter, T. C. et al. Pilot screening


study of targeted genetic polymorphisms for association with seasonal influenza hospital admission. _J. Med. Virol._ 90, 436–446 (2018). Article  CAS  PubMed  Google Scholar  * Sun, X. et


al. Pathogenesis and transmission of genetically diverse swine-origin H3N2 variant influenza A viruses from multiple lineages isolated in the United States, 2011-2016. _J. Virol._ 92,


e00665–18 (2018). Article  CAS  PubMed  PubMed Central  Google Scholar  * Lorbach, J. N. et al. Influenza vaccination of swine reduces public health risk at the swine-human interface.


_mSphere_ 6, e0117020 (2021). Article  PubMed  Google Scholar  * Meischel, T., Villalon-Letelier, F., Saunders, P. M., Reading, P. C. & Londrigan, S. L. Influenza A virus interactions


with macrophages: lessons from epithelial cells. _Cell Microbiol._ 22, e13170 (2020). Article  CAS  PubMed  Google Scholar  * Kamal, R. P., Katz, J. M. & York, I. A. Molecular


determinants of influenza virus pathogenesis in mice. _Curr. Top. Microbiol. Immunol._ 385, 243–274 (2014). PubMed  PubMed Central  Google Scholar  * Schoggins, J. W. Interferon-stimulated


genes: what do they all do? _Annu. Rev. Virol._ 6, 567–584 (2019). Article  CAS  PubMed  Google Scholar  * Zhang, Q. et al. Inborn errors of type I IFN immunity in patients with


life-threatening COVID-19. _Science_ 370, eabd4570 (2020). Article  CAS  PubMed  PubMed Central  Google Scholar  * Bastard, P. et al. Autoantibodies against type I IFNs in patients with


life-threatening COVID-19. _Science_ 370, eabd4585 (2020). Article  CAS  PubMed  PubMed Central  Google Scholar  * Zhou, B. et al. PB2 residue 158 is a pathogenic determinant of pandemic


H1N1 and H5 influenza a viruses in mice. _J. Virol._ 85, 357–365 (2011). Article  CAS  PubMed  Google Scholar  * Ilyushina, N. A. et al. Adaptation of pandemic H1N1 influenza viruses in


mice. _J. Virol._ 84, 8607–8616 (2010). Article  CAS  PubMed  PubMed Central  Google Scholar  * Buchrieser, J. et al. IFITM proteins inhibit placental syncytiotrophoblast formation and


promote fetal demise. _Science_ 365, 176–180 (2019). Article  ADS  CAS  PubMed  Google Scholar  * Zani, A. et al. Interferon-induced transmembrane proteins inhibit cell fusion mediated by


trophoblast syncytins. _J. Biol. Chem._ 294, 19844–19851 (2019). Article  CAS  PubMed  PubMed Central  Google Scholar  * Yockey, L. J. et al. Type I interferons instigate fetal demise after


Zika virus infection. _Sci. Immunol._ 3, eaao1680 (2018). Article  PubMed  PubMed Central  Google Scholar  * Smith, E. C. The not-so-infinite malleability of RNA viruses: viral and cellular


determinants of RNA virus mutation rates. _PLoS Pathog._ 13, e1006254 (2017). Article  PubMed  PubMed Central  Google Scholar  * Shi, G. et al. Rapalogs downmodulate intrinsic immunity and


promote cell entry of SARS-CoV-2. _J. Clin. Invest_. 132, 769–764 (2022). * Poddar, S., Hyde, J. L., Gorman, M. J., Farzan, M. & Diamond, M. S. The interferon-stimulated gene IFITM3


restricts infection and pathogenesis of arthritogenic and encephalitic alphaviruses. _J. Virol._ 90, 8780–8794 (2016). Article  CAS  PubMed  PubMed Central  Google Scholar  * Gorman, M. J.,


Poddar, S., Farzan, M. & Diamond, M. S. The interferon-stimulated gene IFITM3 restricts west Nile virus infection and pathogenesis. _J. Virol._ 90, 8212–8225 (2016). Article  CAS  PubMed


  PubMed Central  Google Scholar  * Jabre, R., Sandekian, V. & Lemay, G. Amino acid substitutions in σ1 and μ1 outer capsid proteins are selected during mammalian reovirus adaptation to


Vero cells. _Virus Res._ 176, 188–198 (2013). Article  CAS  PubMed  Google Scholar  * Pérez-Cidoncha, M. et al. An unbiased genetic screen reveals the polygenic nature of the influenza virus


anti-interferon response. _J. Virol._ 88, 4632–4646 (2014). Article  PubMed  PubMed Central  Google Scholar  * García-Sastre, A. et al. Influenza A virus lacking the NS1 gene replicates in


interferon-deficient systems. _Virology_ 252, 324–330 (1998). Article  PubMed  Google Scholar  * Sun, X. et al. Constitutively expressed IFITM3 protein in human endothelial cells poses an


early infection block to human influenza viruses. _J. Virol._ 90, 11157–11167 (2016). Article  CAS  PubMed  PubMed Central  Google Scholar  * Lu, J. et al. The IFITM proteins inhibit HIV-1


infection. _J. Virol._ 85, 2126–2137 (2011). Article  CAS  PubMed  Google Scholar  * Zhou, B. et al. Single-reaction genomic amplification accelerates sequencing and vaccine production for


classical and Swine origin human influenza a viruses. _J. Virol._ 83, 10309–10313 (2009). Article  CAS  PubMed  PubMed Central  Google Scholar  Download references ACKNOWLEDGEMENTS The


authors thank Dr. Eugene Oltz (Ohio State University) for critical reading and editing of the manuscript. We also thank Dr. Ana Sarkar and Paul Consiglio (Ohio State University) for


providing IFITM3 KO THP-1 cells. Research in the Yount Laboratory is funded by NIH grants AI130110, HL168501, HL157215, HL154001, and AI151230. Research in the Hemann laboratory is funded by


NIH grant AI146141. A.D.K. and P.J.D. were funded by NIH training grant AI112542. AUTHOR INFORMATION AUTHORS AND AFFILIATIONS * Department of Microbial Infection and Immunity, The Ohio


State University College of Medicine, Columbus, OH, USA Parker J. Denz, Samuel Speaks, Adam D. Kenney, Adrian C. Eddy, Jonathan L. Papa, Jack Roettger, Sydney C. Scace, Emily A. Hemann, 


Adriana Forero & Jacob S. Yount * Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA Parker J. Denz, Samuel Speaks, Adam


D. Kenney, Adrian C. Eddy, Jonathan L. Papa, Jack Roettger, Sydney C. Scace, Emily A. Hemann, Adriana Forero, Andrew S. Bowman & Jacob S. Yount * Department of Infectious Diseases, St.


Jude Children’s Research Hospital, Memphis, TN, USA Adam Rubrum & Richard J. Webby * Department of Veterinary Preventive Medicine, The Ohio State University, Columbus, OH, USA Andrew S.


Bowman Authors * Parker J. Denz View author publications You can also search for this author inPubMed Google Scholar * Samuel Speaks View author publications You can also search for this


author inPubMed Google Scholar * Adam D. Kenney View author publications You can also search for this author inPubMed Google Scholar * Adrian C. Eddy View author publications You can also


search for this author inPubMed Google Scholar * Jonathan L. Papa View author publications You can also search for this author inPubMed Google Scholar * Jack Roettger View author


publications You can also search for this author inPubMed Google Scholar * Sydney C. Scace View author publications You can also search for this author inPubMed Google Scholar * Adam Rubrum


View author publications You can also search for this author inPubMed Google Scholar * Emily A. Hemann View author publications You can also search for this author inPubMed Google Scholar *


Adriana Forero View author publications You can also search for this author inPubMed Google Scholar * Richard J. Webby View author publications You can also search for this author inPubMed 


Google Scholar * Andrew S. Bowman View author publications You can also search for this author inPubMed Google Scholar * Jacob S. Yount View author publications You can also search for this


author inPubMed Google Scholar CONTRIBUTIONS P.J.D. organized and performed all experiments in the manuscript, analyzed data, and contributed to manuscript writing and editing of the


manuscript. S.S., A.D.K., A.C.E., J.L.P., J.R., and S.S. assisted with mouse colony genotyping/maintenance, virus titering, and mouse experiments. E.A.H. and A.F. provided conceptual input


and experimental design and edited the manuscript. R.J.W. and A.R. performed 2009 H1N1 and 2011 H3N2 virus sequencing and analysis. A.S.B. provided animal-origin influenza virus strains and


conceptual input. J.S.Y. conceived the study, supervised the experimental design and performance of experiments, analyzed and interpreted data, and wrote the manuscript. CORRESPONDING AUTHOR


Correspondence to Jacob S. Yount. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing interests. PEER REVIEW PEER REVIEW INFORMATION _Nature Communications_ thanks Tao


Dong, Xiangjie Sun and the other anonymous reviewer(s) for their contribution to the peer review of this work. A peer review file is available. ADDITIONAL INFORMATION PUBLISHER’S NOTE


Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. SUPPLEMENTARY INFORMATION SUPPLEMENTARY INFORMATION DESCRIPTION OF


ADDITIONAL SUPPLEMENTARY FILES SUPPLEMENTARY DATA 1 SUPPLEMENTARY DATA 2 SUPPLEMENTARY DATA 3 SUPPLEMENTARY DATA 4 SUPPLEMENTARY DATA 5 REPORTING SUMMARY TRANSPARENT PEER REVIEW FILE SOURCE


DATA SOURCE DATA RIGHTS AND PERMISSIONS OPEN ACCESS This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation,


distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and


indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit


line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use,


you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. Reprints and permissions ABOUT THIS


ARTICLE CITE THIS ARTICLE Denz, P.J., Speaks, S., Kenney, A.D. _et al._ Innate immune control of influenza virus interspecies adaptation via IFITM3. _Nat Commun_ 15, 9375 (2024).


https://doi.org/10.1038/s41467-024-53792-3 Download citation * Received: 22 August 2023 * Accepted: 20 October 2024 * Published: 30 October 2024 * DOI:


https://doi.org/10.1038/s41467-024-53792-3 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link Sorry, a shareable link is not


currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative