Abstract
Interferon-induced transmembrane proteins (IFITM1, 2 and 3) are important antiviral proteins that are active against many viruses, including influenza A virus (IAV), dengue virus (DENV), Ebola virus (EBOV), Zika virus (ZIKV) and severe acute respiratory syndrome coronavirus (SARS-CoV). IFITM proteins exhibit specificity in activity, but their distinct mechanisms of action and regulation are unclear. Since S-palmitoylation and cholesterol homeostasis are crucial for viral infections, we investigated IFITM interactions with cholesterol by photoaffinity crosslinking in mammalian cells along with molecular dynamic simulations and nuclear magnetic resonance analysis in vitro. These studies suggest that cholesterol can directly interact with S-palmitoylated IFITMs in cells and alter the conformation of IFITMs in membrane bilayers. Notably, we discovered that the S-palmitoylation levels regulate differential IFITM protein interactions with cholesterol in mammalian cells and specificity of antiviral activity towards IAV, SARS-CoV-2 and EBOV. Our studies suggest that modulation of IFITM S-palmitoylation levels and cholesterol interaction influence host susceptibility to different viruses.
Graphical Abstract
INTRODUCTION
Interferons (IFNs) provide an important host response to infection and induce the expression of many genes that protect against diverse pathogens1. Amongst the hundreds of IFN-stimulated genes, interferon-induced transmembrane proteins (IFITMs) have emerged as crucial IFN effectors that prevent viral infection of host cells2. In particular, IFITM1, 2 and 3 have been shown to restrict infection by many enveloped viruses including IAV, DENV, EBOV, ZIKV, SARS-CoV, hepatitis C virus (HCV), human immunodeficiency virus (HIV) and others in mammalian cell lines3–5 and mouse models6,7. Notably, single nucleotide polymorphisms (SNPs) in humans, such as rs12252-C, which encodes a truncated form of IFITM3, have been associated with severe H1N1 IAV infection7. Also, IFITM3 SNP rs34481144, which results in lower mRNA expression, has been correlated with reduced CD8+ T cells in patient airways and increased IAV infection severity8. Moreover, IFITM3 SNP rs12252-C has been identified as a risk factor for severity of SARS-CoV-2 infection amongst COVID-19 hospitalized patients in two independent studies9,10. IFITMs are also expressed in embryonic stem cells independent of IFN stimulation and are important for their resistance to viral infections11. IFITMs reduce susceptibility of placental trophoblasts to viral infections and inhibit trophoblast cell fusion, an essential process for fetal development mediated by syncytin, a retroviral envelope-like protein12,13. These significant studies in cellular and animal models as well as preliminary clinical observations highlight an important function for IFITM3 in host immunity against viral infections. Beyond antiviral immunity, IFITM3 has been shown to be critical in phosphoinositide 3-kinase (PI3K) signal amplification for rapid expansion of B cells with high affinity to antigen14. IFITM3 is also implicated in neuroinflammatory conditions as it regulates γ-secretase activity and amyloid-β production in patients with late-onset Alzheimer’s disease15. These studies highlight broader roles of IFITMs in host physiology and disease, which requires a more detailed understanding of their mechanisms of action and regulation.
IFITM1, 2 and 3 exhibit unique cellular properties and are highly regulated by posttranslational modifications2,16. All three IFITM proteins show antiviral activity, but IFITM3 is the most active against many viruses2,3,4. In mammalian cells, IFITM2 and IFITM3 are distributed through early and late endosomes and lysosomes whereas IFITM1, which has a truncated N terminus localizes to the plasma membrane2,4. The cellular localization of IFITMs is an important determinant of their antiviral mechanism specificity, since enveloped viruses enter cells at different sites (for example, plasma membrane versus endocytic pathway) to deliver their genetic contents for replication in host cells. IFITMs are small type IV single-pass transmembrane proteins with amphipathic helices that interact with the inner membrane leaflet17,18 and are post-translationally modified by S-palmitoylation, phosphorylation, and ubiquitination. IFITM3 is S-palmitoylated at membrane-juxtaposed Cys residues (Cys71, 72 and 105)19, but Cys72 is the major site of fatty acid-modification20,21 and the most important for antiviral activity20,22. Recent molecular dynamic simulation and NMR studies show that fatty-acylation at Cys72 stabilizes IFITM3 amphipathic helix-membrane interaction23. Notably, Cys72 is highly conserved across IFITM orthologs in mice, bats, and humans20,23,24. Live-cell imaging studies also show that Cys72 is essential for the trafficking of IFITM3-positive vesicles with incoming virus particles during infection22. In addition to fatty-acylation, Tyr20 phosphorylation regulates IFITM3 plasma membrane localization and endocytosis, whereas Lys ubiquitination at residues 24, 83, 88, and 104, and especially at Lys24, is important for IFITM3 trafficking and turnover in cells25.
The antiviral activity of IFITMs is also regulated by other protein and lipid interactions. For example, proteomic analysis of the S-palmitoylated IFITM3 interactome has identified many membrane-associated protein interaction partners26–28, including the p97/VCP ATPase that contributes to IFITM3 lysosomal turnover and antiviral activity26,29,30. Further, yeast two-hybrid screening revealed IFITM3 interaction with vesicle-membrane-protein-associated protein A (VAPA), which has been implicated in cholesterol accumulation in endolysosomal compartments and IFITM3 antiviral activity30,31. However, other studies indicate that high cholesterol accumulation in these compartments by U18666A pretreatment or down-regulation of Niemann-Pick C1 (NPC1) does not inhibit IAV hemagglutinin-mediated viral membrane fusion32. Nonetheless, membrane cholesterol composition plays a critical role in viral infection by altering membrane mechanical properties and modulating membrane curvature during hemifusion or lipid mixing33. Furthermore, specific interactions of viral fusion proteins with cholesterol and other lipids are crucial for successful viral entry into cells33,34. Recent live-cell imaging studies show that IFITM3 colocalize with virus particles and traffics to lysosomes but does not inhibit membrane lipid mixing or hemifusion22,35. Despite its lack of effect on viral membrane hemifusion with endosomal membranes22,32, IFITM3 could potentially block fusion pore formation and release of viral genetic material in the cytosol. It has been suggested that the IFITM3 amphiphatic helices could modulate membrane curvature and stiffness to block fusion pore formation, a process that can be promoted by the presence of cholesterol in model membranes17,36. However, whether cholesterol directly binds IFITMs and affects their antiviral activity is unclear.
To investigate IFITM-cholesterol interactions, we employed chemical biology tools together with in silico and structural approaches. To characterize cholesterol interaction with IFITMs in mammalian cells, we synthesized a cholesterol photoaffinity reporter (x-alk-chol) and performed proteomic as well as targeted protein photocrosslinking studies. These studies demonstrated that x-alk-chol can photocrosslink IFITM3 expressed in IFN-α-stimulated cells. Subsequent mutagenesis studies showed that S-palmitoylation and a cholesterol binding motif in the transmembrane domain of IFITM3 can influence x-alk-chol photoaffinity labeling and antiviral activity. Our molecular dynamics simulation suggests that cholesterol alters the conformation of IFITM3 in membrane bilayers, which was supported by NMR studies of truncated IFITM3 protein constructs reconstituted in membrane bicelles. Surprisingly, IFITM2 was photocrosslinked by x-alk-chol much less efficiently compared to IFITM3 even though both contain the identified cholesterol binding motif and are more than 80 percent identical at the protein level. Our subsequent analysis showed that the IFITM S-palmitoylation levels are consistent with their x-alk-chol photocrosslinking and specificity of antiviral activity towards IAV, SARS-CoV-2 and EBOV. Collectively, our results suggest that cholesterol can directly interact with IFITMs in mammalian cells, modulate S-palmitoylated IFITM conformations in membrane bilayers, and are consistent with specificity in antiviral activity.
RESULTS AND DISCUSSION
Photoaffinity labeling of proteins in HeLa cells with a sterol reporter.
To investigate cholesterol binding of IFITMs in mammalian cells, we synthesized a bifunctional cholesterol analog (x-alk-chol) that has a diazirine in position 6 for crosslinking to interacting proteins on UV light exposure (Supplementary Figure 1). This cholesterol photoaffinity probe (x-alk-chol) also has a terminal alkyne handle on the alkyl side chain for CuI-catalyzed azide-alkyne cycloaddition (CuAAC) to azide-fluorophore for fluorescence detection or azide-biotin for affinity enrichment of interacting proteins (Supplementary Figure 1). Such photoaffinity cholesterol probes have been used to study sterol-protein interactions in cells previously37,38. Maestro modeling of x-alk-chol showed similar molecular topology as cholesterol (Supplementary Figure 1). To probe cholesterol-binding proteins in cells, HeLa cells were treated with x-alk-chol and irradiated with UV light for x-alk-chol crosslinking with interacting proteins. The cell lysate was then subjected to CuAAC with azide-rhodamine for in-gel fluorescence profiling of x-alk-chol crosslinked proteins (Figure 1A). Many proteins in HeLa cells were crosslinked with x-alk-chol in a UV-dependent and dose-dependent manner (Figure 1B, C). Cells delivered excess cholesterol following x-alk-chol treatment showed lower labeling suggesting potential cholesterol competition with x-alk-chol (Figure 1D).
Figure 1. In-gel profiling of x-alk-chol labeled proteins in HeLa cells.
A) Experimental scheme for labeling of proteins with x-alk-chol in live cells (created with Biorender.com). B) In-gel fluorescence profiling of x-alk-chol crosslinked HeLa cell proteins shows UV dependent x-alk-chol labeling of many proteins in HeLa cell proteome. Cells were incubated with x-alk-chol for 30 min and irradiated with UV light (365 nm) for 5 min following which cell lysates were reacted with azide-rhodamine for fluorescence gel scanning. C) In-gel fluorescence profiling of x-alk-chol concentration dependent labeling of HeLa cell proteins. Cells were incubated with different concentrations of x-alk-chol for 30 min and irradiated with UV light (365 nm) for 5 min following which cell lysates were reacted with azide-rhodamine for fluorescence gel scanning. In-gel fluorescence profiling shows x-alk-chol concentration dependent labeling of proteins. α-Tubulin western blotting shows comparable protein loading. D) In-gel fluorescence profiling of cholesterol competition with x-alk-chol labeling of proteins. Cells were incubated with x-alk-chol for 30 min following which cells were treated with 10 μM cholesterol in mβCD and then irradiated with UV light (365 nm) for 5 min. Then cell lysates were reacted with azide-rhodamine for fluorescence gel scanning. In-gel fluorescence profiling shows cholesterol competition with x-alk-chol labeling of proteins. α-Actin western blotting shows comparable protein loading.
Photoaffinity labeling identifies IFITM3 as a cholesterol binding immune associated protein.
To enrich and identify x-alk-chol labeled proteins in HeLa cells, we subjected the cell lysate to CuAAC with azide-biotin, affinity purified with streptavidin beads and subjected to on-bead protease digestion for mass spectrometric analysis (Figure 2A). A summary of the proteomics data revealed enrichment of 330 proteins in UV light irradiated samples including known cholesterol binding proteins like caveolin-1 (CAV1) and NPC1, among others (Figure 2A)39,40. Bioinformatic analysis of the enriched proteins suggest x-alk-chol labeling of endoplasmic reticulum (ER) and plasma membrane resident membrane proteins primarily (Figure 2B). Many of the high confidence x-alk-chol labeled proteins were previously identified in a proteome-labeling profile of trans-sterol probe (Supplementary Figure 2)37. Gene ontology analysis of the enriched proteins enriched identified many membrane-associated proteins involved in lipid biosynthesis, and metabolism (Supplementary Figure 3), To characterize cholesterol binding to IFITMs and other immune-associated proteins, we profiled x-alk-chol crosslinked proteins in IFN-α stimulated HeLa cells (Figure 2C). In addition to the proteins identified in non-stimulated cells, we recovered IFITM3 and many other immune-associated proteins from the proteomic data (Figure 2C). Gene ontology analysis of the proteins enriched in IFN-α stimulated cells identified many other membrane-associated proteins involved in immunity (Figure 2D, Supplementary Figure 3), which may warrant further investigation as many of these hits are important host factors involved in virus infection including SARS-CoV-241,42. These results show that x-alk-chol can photocrosslink many known cholesterol-binding proteins as well as capture many immune-associated proteins including IFITM3.
Figure 2. Proteomic analysis of x-alk-chol labeled proteins in HeLa cells.
A) Volcano plot showing x-alk-chol binding proteins in HeLa cells. Cells were treated with x-alk-chol (10 μM) for 30 min and UV (365 nm) irradiated for 5 min. The cell lysates were further reacted with azide-biotin for enrichment of x-alk-chol labeled proteins with streptavidin beads and identification by mass spectrometry. Volcano plot shows enrichment of many reported cholesterol binding proteins (in blue) for UV treated sample. The x-axis is the difference of means between the UV treated and untreated samples and the y-axis is the log of the probability of that difference determined by the t-test. The minimum values for a valid protein are p<0.05 and a difference of means of 2 (FDR=0.0001, S0=1, n=3 replicates). B) Gene Ontology (GO) analysis shows subcellular localization of proteins enriched in UV treated samples. C) Volcano plot showing x-alk-chol crosslinked proteins in interferon (IFN-α) stimulated HeLa cells. Volcano plot shows enrichment of many reported cholesterol binding proteins (in blue) and new cholesterol binding immunity associated proteins (in red) for UV treated sample. The x-axis is the difference of means between the UV treated and untreated samples and the y-axis is the log of the probability of that difference determined by the t-test. The minimum values for a valid protein are p<0.05 and a difference of means of 2 (FDR=0.0001, S0=1, n=3 replicates). D) Gene Ontology (GO) analysis shows subcellular localization of proteins enriched in Interferon and UV treated samples.
S-Palmitoylated IFITM3 interacts with membrane cholesterol.
S-palmitoylated membrane proteins have been suggested to partition into cholesterol-rich liquid-ordered membrane microdomains43,44. IFITM3 is S-palmitoylated at Cys71, 72 and 105 (Figure 3A). Western blot analysis of IFN-α stimulated and x-alk-chol treated HeLa cells showed a mobility shift for IFITM3 in UV-irradiated samples (Figure 3B) resembling that seen with CAV1, a known cholesterol binding protein (Figure 3B). To confirm x-alk-chol photocrosslinking to IFITM3, the cell lysate was subjected to CuAAC with azide-biotin and affinity purified with streptavidin beads. Western blot analysis showed significant enrichment of IFITM3 and CAV1 in the UV-irradiated sample (Figure 3B). Further treatment of cells with x-alk-chol in the presence of cholesterol decreases x-alk-chol labeling of IFITM3, suggesting cholesterol competition with reporter (Supplementary Figure 4). To validate IFITM3-cholesterol interaction, we performed x-alk-chol crosslinking studies with overexpressed HA tagged IFITM3 WT in HEK293T cells. In-gel fluorescence detection of anti-HA immunoprecipitated sample shows UV-dependent x-alk-chol crosslinking of HA-IFITM3 (Figure 3C). To identify the role of S-palmitoylation in IFITM3 cholesterol binding, we tested x-alk-chol labeling of IFITM3 PalmΔ construct, a Cys71, 72 and 105 to Ala triple mutant (Figure 3C, D, Supplementary Figure 5A, B). IFITM3 PalmΔ shows significantly less x-alk-chol labeling. Bioinformatic analysis of IFITM3 orthologues from great apes and rodentia shows that IFITM3 has a putative cholesterol binding domain. IFITM3 in great apes have a cholesterol consensus domain CARC 104KCLNIWALIL113 (in pink) which lies N-terminus to the transmembrane domain (Supplementary Figure 6). Many membrane proteins have CARC motifs or its mirror code CRAC motif to mediate interactions with cholesterol45,46. To investigate role of this motif in IFITM3-cholesterol interaction, we made a CARCΔ construct by replacing Lys104 with Ala and Trp109 with Ile. We observe that x-alk-chol photocrosslinking efficiency of CARCΔ is significantly decreased (Figure 3C, D, Supplementary Figure 5A, B), suggesting role of the motif in IFITM3-cholesterol interaction. Mouse IFITM3, which has the three Cys residues but not the CARC motif conserved, shows significantly lower x-alk-chol labeling in HEK293T cells (Supplementary Figure 7A, B). IFITM3 Lys104 to Ala and Trp109 to Ile single mutants also show reduction in x-alk-chol photocrosslinking (Supplementary Figure 8A, B). IFITM3 single Cys71 and Cys72 to Ala mutants show similar x-alk-chol photocrosslinking as IFITM3 WT but Cys105 to Ala mutant shows some increase in x-alk-chol photocrosslinking. This may be due to less steric interactions at the CARC domain on mutation of Cys105 to Ala (Supplementary Figure 9A, B). S-palmitoylation levels of IFITM3 CARCΔ were analyzed by metabolic labeling with alk-16 labeling19. IFITM3 CARCΔ shows similar S-palmitoylation levels as IFITM3 WT (Supplementary Figure 10). We also analyzed the subcellular localization of IFITM3 CARCΔ by co-expressing myc-tagged IFITM3 WT with HA-tagged IFITM3 CARCΔ. IFITM3 CARCΔ shows similar endolysosomal localization as IFITM3 WT (Supplementary Figure 11A, B). These results suggest that CARC domain of IFITM3 is important for its interaction with cholesterol but does not impact S-palmitoylation or subcellular localization.
Figure 3. IFITM3 S-Palmitoylation and cholesterol binding regulates antiviral activity.
A) S-palmitoylated IFITM3 in a membrane bilayer (created with Biorender.com). Key domains and post translational modifications are highlighted. Each secondary structure is colored differently, amphipathic helix 1 (AH1 from residue 62 to 67) in purple, amphipathic helix 2 (AH2 from residue 76 to 85) in green and transmembrane domain (TM from residue 109 to 131) in orange. Cholesterol binding motif in pink. B) Western blot analysis of x-alk-chol binding proteins CAV1 and IFITM3. Interferon (IFN-α) treated HeLa cells were incubated with x-alk-chol (10 µM) for 30 min and UV-irradiated for 5 min and after cell lysis, proteins were separated by SDS-PAGE. Western blot analysis shows slower migration rate for CAV1 and IFITM3 for UV treated sample, suggesting x-alk-chol crosslinking. Western blot analysis of streptavidin-biotin pull down of x-alk-chol crosslinked proteins. Cell lysates from input was reacted with azide-biotin for streptavidin pull down of x-alk-chol crosslinked proteins. Further western blot analysis shows UV dependent pull down of endogenous CAV1 and IFITM3. C) x-alk-chol labeling of overexpressed HA-tagged IFITM3 wildtype and cholesterol binding mutants in HEK293T cells. In-gel fluorescence profiling shows UV-dependent fluorescence signal for IFITM3 WT. IFITM3 PalmΔ and CARCΔ shows significantly lower fluorescence signal, suggesting lower level of x-alk-chol labeling. Anti-HA blot shows IFITM3 expression for each construct. D) Quantitative analysis of x-alk-chol labeling of IFITM3 constructs in C. The fluorescence signal normalized to protein level was plotted for each construct. Data represents the mean and s.e.m. of three independent experiments. P values were determined by one-way anova with a Dunnett’s multiple comparisons test. **P<.01, ***P<.001. E) Influenza A virus (IAV) infection of A549 IFITM1/2/3 KO cells stably expressing IFITM3 constructs. Virus nucleoprotein (NP) levels were examined by flow cytometry for percentage of infection analysis. Data represents the mean and s.e.m. for triplicates. F) rVSV-SARS-CoV-2 S infection of A549 IFITM1/2/3 KO cells stably expressing IFITM3 constructs. Viral Infectivity was examined by counting the eGFP-positive cells using fluorescent microscopy. Data represents the mean and s.e.m. for five independent experiments.
To analyze the significance of IFITM3 interaction with cholesterol, we evaluated antiviral activity of IFITM3 cholesterol binding mutants against IAV and SARS-CoV-2. A549 IFITM1/2/3 KO – ACE2 cells stably expressing IFITM3 constructs were used for the infection studies (Supplementary Figure 12A, B). Cells expressing IFITM3 WT shows antiviral activity against IAV whereas loss of function construct IFITM3 PalmΔ has little or no activity19. Cells expressing IFITM3 CARCΔ construct shows significant loss of resistance to IAV infection (Figure 3E, Supplementary Figure 13). HEK293T cells expressing these IFITM3 constructs show similar trend in antiviral activity against IAV (Supplementary Figure 14A, B). Next, we tested antiviral activity of these cell lines against a recombinant vesicular stomatitis virus bearing SARS-CoV-2 spike (rVSV-SARS-CoV-2 S)47, because recent studies show that SARS-CoV-2 spike protein interaction with cholesterol is important for virus entry and pathological syncytia formation34. Though IFITM3 is shown to be protective against SARS-CoV-2 infections in mice48, early cell based studies show conflicting results for both inhibition of SARS-CoV-2 pseudotyped virus infection and SARS-CoV-2 spike protein mediated cell-cell fusion34,49,50,51. Furthermore, although overexpression of IFITMs restrict SARS-CoV-2 infection, endogenous levels of IFITMs was suggested to have a proviral effect52,53. Our A549 IFITM1/2/3 KO cells provides an excellent system to evaluate the activity of IFITMs in SARS-CoV-2 entry (Supplementary Figure 12A, B). Cells expressing IFITM3 WT shows antiviral activity against rVSV-SARS-CoV-2 S whereas IFITM3 PalmΔ or CARCΔ mutants have minimal or no antiviral activity (Figure 3F). Thus, IFITM3 cholesterol interaction might play an important role in blocking virus fusion and release of genetic material in host cytosol during IAV and SARS-CoV-2 infection entry.
Cholesterol alters IFITM3 conformations in membrane bilayers from molecular dynamics simulations.
To explore the effect of cholesterol on IFITMs in membrane bilayers, we performed molecular dynamics (MD) simulations of apo-IFITM3 and C72, C105 S-palmitoylated (palm) IFITM3 in a 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) membrane bilayer with or without 20% cholesterol (Figure 4A, Supplementary Figure 15A and Supplementary table 1)(Pogozheva et al. 2022; Garst et al. 2021). We measured tilt angles of each helical domain with respect to the membrane normal (i.e., Z-axis), Z positions of the center of mass of each helix with respect to the bilayer center (i.e., Z=0), as well as the interaction patterns of each residue to quantify behaviors of each helix namely, AH1 (residues 62–67), AH2 (residues 76–85) and TM (residues 109–131). Each helical domain shows different behaviors on addition of cholesterol in DMPC membrane systems. However, we observed similar change in trends for the helices in both apo-IFITM3 and S-palm IFITM3. C72, 105 S-palm IFITM3 shows more interactions of AH1 and Loop2 (residues 86–108) with DMPC membrane than apo-IFITM3 (Figure 4B, Supplementary Figure 15B, Supplementary table 1). This is consistent with our previous studies, which showed that S-palmitoylation at Cys72 increased IFITM3 AH1 interaction with DMPC membranes54. In the presence of cholesterol in the membrane, IFITM3 interacts with cholesterol via the residues around AH1, Loop1 and TM domain. MD simulation of IFITM3 CARCΔ shows subtle changes in cholesterol binding (Supplementary Figure 16). In the presence of cholesterol, both apo- and S-palm IFITM3 AH1 shows further increase in membrane interactions as its tilt angles are closer to 90°, a completely horizontal orientation. AH1 has a tilt angle of 86.85° for apo-IFITM3 and 77.7° for S-palm IFITM3 in cholesterol containing systems versus 101.53° for apo-IFITM3 and 108.4° for S-palm IFITM3 in DMPC-only systems (Supplementary table 2). Additionally, AH1 domain was more deeply embedded into the membrane in the presence of cholesterol, thus making more frequent interactions with lipid tails (Figure 4B, C, Supplementary Figure 15B). AH2, on the other hand, responses differently to the presence of cholesterol in comparison to AH1. AH2 tilt angle shows values larger than 90°, meaning that it becomes more vertical in a membrane system with cholesterol (Supplementary table 2). In addition, presence of cholesterol does not affect the position of AH2 along the membrane normal in contrast to AH1 (Figure 4C). For TM domain, the tilt angle tends to decrease in the cholesterol membrane system for both apo-IFITM3 and S-palm IFITM3 (Figure 4D). Such tilt angle changes are known to occur to maximize a hydrophobic match between the lipid bilayer and the transmembrane domain55. Cholesterol is known to increase the thickness and order of a lipid bilayer56, as we observed for all cholesterol containing membrane systems (Supplementary Figure 17). Interestingly, we observed significant changes in the interaction pattern for Loop2 in membrane with and without cholesterol for both apo-IFITM3 and S-palm IFITM3. Loop2 is a part of the highly conserved CD225 domain which contains a basic patch consisting of R85, R87, and K88, as well as a 91GxxxG95 motif implicated in IFITM3 oligomerization and antiviral activity24,57. We see increased hydrophobic interactions in Loop2 in the DMPC system, whereas the cholesterol containing membrane system has more hydrophilic (protein-water and protein-lipid headgroup) interactions (Supplementary Figure 18–21). Since DMPC is a fully saturated lipid and is well-mixed with rigid cholesterol, it leads to a more ordered membrane phase. This makes it difficult for Loop 2 to insert into the membrane hydrophobic core and have hydrophobic interactions. Thus, cholesterol-induced changes in IFITM3-membrane interactions might in turn influence IFITM3 interactions with other lipids and proteins as well as its activity.
Figure 4. Molecular dynamics simulation of S-palmitoylated IFITM3 in cholesterol-containing membrane bilayer.
A) Simulation snapshots of C72, 105 S-palm IFITM3 in DMPC (top) and DMPC + Chol membrane bilayers (bottom). B) Interaction frequency of each residue of C72, 105 S-palm IFITM3 in DMPC (top) and DMPC + Chol membrane bilayers (bottom) interacting with surrounding environment, DMPC lipid headgroups (red), DMPC lipid tails (gray), water (blue), cholesterol headgroup (magenta), and cholesterol tails (pink). Each graph shows the interaction frequency within 4 Å from each residue. C) The distance of the center of mass for each helical domain (AH1, AH2 and TM) to the membrane center (Z=0) for apo-IFITM3 and C72, 105 S-palm IFITM3 in DMPC or DMPC + Chol. Data represents the mean and SE for triplicates. D) The averaged tilt angle of each helical domain for apo-IFITM3 and C72, 105 S-palm IFITM3 in DMPC or DMPC + Chol. Averaged tilt angle was measured by calculating the angle between the principal axis of each helix and the membrane normal (Z-axis). Data represents the mean and SE for triplicates.
To structurally characterize IFITM3 in a cholesterol-containing bilayer, we employed a bicelle membrane system. However, in the presence of full length IFITM3, the short chain lipid, 1,2-dihexanoyl-sn-glycero-3-phosphatidylcholine (DHPC) could not solubilize DMPC liposomes to form membrane bicelles. We, therefore, designed several truncated constructs for bicelle reconstitution. IFITM389−133, which excluded the amphipathic helices, expressed well after overnight induction, and were purified using His-affinity purification (Supplementary Figure 22A, B). We characterized 15N-labeled IFITM389−133 in bicelles with a protein to lipid ratio of 1:50 and final q value between 0.5 and 0.6 using 2D 1H-15N TROSY spectra (Supplementary Figure 23). We observed subtle changes in some specific residues of the TM domain though full assignment of the TM residues could not be performed due to low signal.
IFITMs show different S-palmitoylation levels, x-alk-chol labeling and antiviral activity.
We next analyzed cholesterol binding of human IFITM1, IFITM2 and IFITM3. All three IFITM proteins have conserved Cys71, 72, 105 and cholesterol binding motif (Figure 5A). We used in-gel fluorescence profiling to evaluate photocrosslinking of IFITMs with x-alk-chol. In-gel fluorescence detection and quantification normalized to protein levels showed x-alk-chol photocrosslinking to IFITM1 and IFITM3 but not with IFITM2 (Figure 5B, C, Supplementary Figure 24A, B). Differential photocrosslinking of IFITM2 and IFITM3 with x-alk-chol is surprising since the protein sequence alignment shows 83% sequence identity. They also showed similar subcellular localization by immunofluorescence confocal microscopy (Supplementary Figure 25). However, the S-palmitoylation levels of IFITM2 and IFITM3 by alk-16 labeling correlated with x-alk-chol photocrosslinking (Figure 5D, E, Supplementary Figure 24C, D). These results suggest that IFITM2 and IFITM3 interactions with cholesterol are consistent with their S-palmitoylation levels.
Figure 5. Cholesterol binding, S-palmitoylation and antiviral activity of IFITM proteins.
A) Alignment, and topology of IFITM proteins. The distinct residues in IFITM2 and IFITM3 are in cyan. IFITM3 has an additional Phe residue in red. CARC Lys, Trp and Leu residues are highlighted in pink. B) x-alk-chol labeling of overexpressed HA-tagged IFITM proteins in HEK293T cells. In-gel fluorescence profiling shows UV-dependent fluorescence signal for IFITMs. Anti-HA blot shows IFITM expression for each construct. The fluorescence band marked with asterisk (*) is used for further quantification. C) Quantitative analysis of x-alk-chol labeling of IFITM constructs in b, respectively. The fluorescence signal normalized to protein level was plotted for each construct. Data represents the mean and s.e.m. of three independent experiments. P values were determined by one-way anova with a Dunnett’s multiple comparisons test. 0.5<ns, ****P<.0001. D) S-palmitoylation analysis of overexpressed HA-tagged IFITM constructs in HEK293T cells. In-gel fluorescence profiling shows UV-dependent fluorescence signal for IFITMs. Anti-HA blot shows IFITM3 expression for each construct. E) Quantitative analysis of alk-16 labeling of IFITM3 constructs in d. The fluorescence signal normalized to protein level was plotted for each construct. Data represents the mean and s.e.m. of three independent experiments. P values were determined by one-way anova with a Dunnett’s multiple comparisons test. 0.5<ns, **P<.01, ***P<.001. F) Influenza A virus (IAV) infection of HEK293T and A549 IFITM1/2/3 KO cells expressing IFITM constructs. Cells were infected with IAV (MOI 10) for 6 h. Virus nucleoprotein (NP) levels were examined by flow cytometry using anti-NP staining for percentage of infection analysis. Data represents the mean and s.e.m. for three independent experiments. P values were determined by one-way anova with a Tukey’s multiple comparisons test. ***P<.001. G) SARS-CoV-2 entry in A549 IFITM1/2/3 KO cells stably expressing IFITMs. Cells were infected with rVSV-eGFP SARS-CoV-2 for 7 h. Viral Infectivity was examined by counting the eGFP-positive cells using fluorescent microscopy. Data represents the mean and s.e.m. for five independent experiments. P values were determined by one-way anova with a Tukey’s multiple comparisons test. 0.5<ns, **P<.01, ****P<.0001. H) EBOV entry in A549 IFITM1/2/3 KO cells stably expressing IFITMs. Cells were infected with rVSV-eGFP EBOV GP for 7 h. Viral Infectivity was examined by counting the eGFP-positive cells using fluorescent microscopy. Data represents the mean and s.e.m. for five independent experiments. P values were determined by one-way anova with a Tukey’s multiple comparisons test. 0.5<ns, **P<.01, ****P<.0001.
We then evaluated the antiviral activity of IFITM proteins with IAV, SARS-CoV-2 and EBOV entry. IAV and SARS-CoV-2 use sialylated EGFR58 and ACE259 as entry factor respectively, whereas EBOV, another virus entering from late endosomes, has been shown to use NPC1 as an entry factor60–62. For these experiments, we stably expressed the IFITM proteins in A549 IFITM1/2/3 KO – ACE2 cells (Supplementary Figure 26A, B). All IFITM proteins show comparable expression levels in A549 IFITM1/2/3 KO – ACE2 cells (Supplementary Figure 26C). Consistent with previous reports63,64, expression of plasma membrane resident IFITM1 did not inhibit infection by these viruses that enter through low pH endosomal compartments (Figure 5F, G, H). In contrast, we found that IFITM3 exhibits greater antiviral activity against IAV and rVSV-SARS2 S, whereas IFITM2 is more active against an rVSV bearing the EBOV spike glycoprotein, GP (rVSV-EBOV GP)65 (Figure 5F, G, H). These results suggest that differential interactions with cholesterol and S-palmitoylation impact the antiviral activity of IFITM proteins towards different viruses.
Conclusion
IFITMs have emerged as important IFN-induced effectors in host immunity against diverse viruses but require further mechanistic insight. Here, we explored IFITM-cholesterol interactions using chemical biology approaches and in silico, structural methods. We employed photoaffinity crosslinking to investigate direct cholesterol-protein interactions in cells. Our proteomic analysis of cholesterol photoaffinity crosslinking revealed many candidate sterol-interacting proteins, including endogenously expressed IFITM3 in interferon (IFN-α)-stimulated cells. Further targeted crosslinking studies identified a cholesterol binding motif adjacent to the transmembrane domain of IFITM3. Infection studies with IFITM3 cholesterol binding mutants suggest that IFITM3-cholesterol interaction can play an important role in blocking virus entry into host cells mediated by the IAV and SARS-CoV-2 spike glycoproteins. This is in agreement with recent in vitro work, which shows that cone-shaped lipids like cholesterol facilitate IFITM3-induced membrane curvature to inhibit viral fusion with host membranes36. Moreover, we show that S-palmitoylation of IFITM3 is crucial for cholesterol photoaffinity crosslinking in cells. Molecular dynamics simulation revealed potential effects of cholesterol on IFITM conformations and membrane interactions in lipid bilayers, which was supported in part by our NMR studies of IFITM3 transmembrane domain reconstituted in membrane bicelles. Our in silico studies also suggested that cholesterol may impact IFITM3 AH1 and Loop2 interactions with the membrane. This is particularly interesting since IFITM3 AH1 is important in inhibiting viral infection in cells17,36 and IFITM3 Loop2 contains motifs implicated in IFITM3 oligomerization and antiviral activity24,57. Additional studies with different IFITM proteins led to the surprising discovery that IFITM2 exhibits significantly less x-alk-chol crosslinking than IFITM3, even though both contain the conserved Cys and cholesterol binding domain.
Further analysis by metabolic labeling revealed that S-palmitoylation level of IFITM2 is significantly less than IFITM3, which may explain the differential x-alk-chol crosslinking of the IFITMs. Interestingly, infection studies in A549 cell lines shows that IFITM3 has greater antiviral activity against IAV and SARS-CoV-2 infection, whereas IFITM2 is more active against EBOV, that enter host cells through interactions with NPC1 in late endosomes60–62,66. This is in agreement with previous observations that IFITM3 restricts viruses like IAV or flaviviruses more efficiently than cathepsin-dependent viruses like EBOV60–62,66. Interestingly, amphotericin B, which can bind cholesterol, has been reported to inhibit IFITM3 restriction of IAV but not EBOV63, suggesting that differential engagement of cholesterol-rich membranes or endosomes by IFITMs can impact selective inhibition of IAV versus EBOV infection. Our results now provide direct evidence for this possibility, as we show that higher levels of S-palmitoylation enhance IFITM3 interaction with cholesterol and inhibits viruses like IAV and SARS-CoV-2, whereas IFITM2 exhibits lower S-palmitoylation levels and cholesterol interactions but more effectively inhibits EBOV, which may engage cellular membranes with less cholesterol that contain NPC1 transporter. Therefore, while both IFITM2 and IFITM3 are present in the endolysosomal pathway, their differential S-palmitoylation levels and cholesterol interactions may provide specificity against viruses that enter through different endosomal trafficking routes and membrane compartments. Therefore, it will be interesting to investigate the cellular and biochemical mechanisms that regulate differential palmitoylation and depalmitoylation of IFITMs.67,68 Our study highlights how direct cholesterol photoaffinity labeling in cells can reveal unpredicted covalent and non-covalent lipid regulation of IFN effectors that maybe harnessed to modulate their activity and/or specificity towards different viruses in the future.
Methods
Labeling of HeLa cell proteome with x-alk-chol.
HeLa cells were incubated with different concentrations (5, 10, 15 and 20 μM) of x-alk-chol in mβCD for 30 min. For cholesterol competition, cells were first treated with 10 μM of x-alk-chol for 30 min and were further treated with 10 μM cholesterol for 30 min. Cells were washed with PBS and UV treated for 5 min. Cells were lysed in 1% Brij 97 in 50 mM TEA, 150 mM NaCl pH 7.4, 1X Roche protease inhibitor, and 1,500 units/mL benzonase (EMD). Protein concentrations were determined by the BCA assay.
45 μl of the samples (50 μg of protein) were added to 5 μl of CuAAC reactant solution (0.5 μl of 10 mM azido-rhodamine (final concentration 100 μM), 1 μl of 50 mM freshly prepared CuSO4·5H2O in H2O (final concentration 1 mM), 1 μl of 50 mM freshly prepared TCEP (final concentration 1 mM) and 2.5 μl of 10 mM Tris[(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]amine (TBTA) (final concentration 500 μM)). The samples were rocked at room temperature for 1 h and protein was precipitated with methanol-chloroform-water mixture (4:1.5:3). After air-drying, the protein pellets were dissolved in 1X Laemmli sample buffer (20 μl) and were heated for 10 min at 95 °C and separated by gel electrophoresis.
Proteomic analysis.
HeLa cells were grown to confluency in 10 cm dishes. Cells were stimulated with IFN-α (100 µg/mL) overnight. Next cells were treated with x-alk-chol (10 μM) for 30 min and UV treated for 5 min. Cells were lysed with 4% SDS in 50 mM HEPES buffer with 150 mM NaCl. 2 mg of cell lysate was reacted with azido-biotin for 4 h (1 mM CuSO4, 1 mM TCEP, 200 μM TBTA, 200 μM azido-biotin) at 1 mg/mL. Protein was precipitated using methanol (8 mL), chloroform (3 mL), and water (6 mL) overnight at −20 °C. Protein pellets were dried for 1 h and solubilized in 200 μL of 4% SDS buffer. The sample was diluted to 1 mg/mL with 50 mM HEPES buffer. 25 μL of streptavidin beads were added to the samples and were nutated for 1 h. The beads were washed with 1% SDS, 5 M Urea, and PBS. Samples were split and half the beads were boiled in 20 μL of 4% SDS loading buffer and separated by SDS-PAGE for western blot analysis. The other half was suspended in 200 μL of 25 mM ammonium bicarbonate, reduced with 1 mM DTT for 30 min, and then alkylated with 50 mM iodoacetamide in the dark for 30 min. Then, the beads were washed with 200 μL of 25 mM ammonium bicarbonate and suspended in 50 μL of 25 mM ammonium bicarbonate. 0.1 μg of trypsin was added, and the samples were digested at 37 °C overnight. The supernatant was collected and dried on a speedvac and solubilized in 5% acetonitrile/1% formic acid for LC-MS/MS analysis.
Extracted tryptic peptides were desalted on a trap column following separation on a 12 cm/75 μm reversed phase C18 column (Nikkyo Technos Co., Ltd. Japan). A 180-minute gradient increasing from 10% B to% 45% B in 133 minutes (A: 0.1% Formic Acid, B: Acetonitrile/0.1% Formic Acid) were delivered at 200 nL/min. The liquid chromatography setup (Dionex, Boston, MA, USA) was connected to an Orbitrap XL (Thermo, San Jose, CA, USA) operated in top-8-CID-mode with MS spectra measured at a resolution of 60,000@m/z 400. Acquired tandem MS spectra were extracted using Maxquant queried against the Uniprot complete human database and processed using Perseus. Absent values were imputed based on the normal distribution of values. To determine if a protein was a valid hit, the false discovery rate was lower than 0.01% and the mean difference of the control and labeled samples had be greater than 2 with p<0.05 determined by the t-test. Database for Annotation, Visualization and Integrated Discovery (DAVID) was used for Gene Ontology (GO) enrichment analysis.
Supplementary Material
Acknowledgements
T.D. and E.H.G., acknowledges support by Tri-Institutional Program in Chemical Biology. H.C.H., W.I., and K.C. acknowledge grant support from NIH-NIGMS R01GM087544. K.C. was also supported by NIH R01AI134824. We thank R. He for the generation of IFITM1, 2, 3 knockout mammalian cell lines. We also thank S. Bhattacharya for help with NMR data collection at New York Structural Biology Center. We also thank Z. Wu for help with polarimetry measurements at Scripps Research. We also thank M. Griffin and X. Zhao for helpful discussions.
Footnotes
Supporting Information
This material is available free of charge via the internet at http://pubs.acs.org.
Cell lines and reagents, experimental methods for labeling of overexpressed IFITMs with reporter, infection, imaging, molecular dynamic simulation, IFITM3 expression and reconstitution in bicelles, small molecule characterization and proteomics data.
K.C. is a member of the scientific advisory boards of Integrum Scientific, LLC and Biovaxys Technology Corp. K.C. is a named co-inventor on a patent application covering the rVSV-SARS2 surrogate virus assigned to Albert Einstein College of Medicine. The other authors declare no competing financial interests.
References
- 1.MacMicking JD (2012) Interferon-inducible effector mechanisms in cell-autonomous immunity. Nat. Rev. Immunol 12, 367–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bailey CC, Zhong G, Huang I-C, and Farzan M (2014) IFITM-Family Proteins: The Cell’s First Line of Antiviral Defense. Annu. Rev. Virol 1, 261–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Brass AL, Huang I-C, Benita Y, John SP, Krishnan MN, Feeley EM, Ryan BJ, Weyer JL, van der Weyden L, Fikrig E et al. (2009) The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell 139, 1243–1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Perreira JM, Chin CR, Feeley EM, and Brass AL (2013) IFITMs restrict the replication of multiple pathogenic viruses. J. Mol. Biol 425, 4937–4955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Savidis G, Perreira JM, Portmann JM, Meraner P, Guo Z, Green S, and Brass AL (2016) The ifitms inhibit zika virus replication. Cell Rep 15, 2323–2330. [DOI] [PubMed] [Google Scholar]
- 6.Bailey CC, Huang I-C, Kam C, and Farzan M (2012) Ifitm3 limits the severity of acute influenza in mice. PLoS Pathog 8, e1002909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Everitt AR, Clare S, Pertel T, John SP, Wash RS, Smith SE, Chin CR, Feeley EM, Sims JS, Adams DJ et al. (2012) IFITM3 restricts the morbidity and mortality associated with influenza. Nature 484, 519–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Allen EK, Randolph AG, Bhangale T, Dogra P, Ohlson M, Oshansky CM, Zamora AE, Shannon JP, Finkelstein D, Dressen A 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gómez J, Albaiceta GM, Cuesta-Llavona E, García-Clemente M, López-Larrea C, Amado-Rodríguez L, López-Alonso I, Melón S, Alvarez-Argüelles ME, Gil-Peña H et al. (2020) The Interferon-induced transmembrane protein 3 gene (IFITM3) rs12252 C variant is associated with COVID-19. Cytokine 137, 155354. [DOI] [PubMed] [Google Scholar]
- 10.Zhang Y, Qin L, Zhao Y, Zhang P, Xu B, Li K, Liang L, Zhang C, Dai Y, Feng Y et al. (2020) Interferon-Induced Transmembrane Protein 3 Genetic Variant rs12252-C Associated With Disease Severity in Coronavirus Disease 2019. J. Infect. Dis 222, 34–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wu X, Dao Thi VL, Huang Y, Billerbeck E, Saha D, Hoffmann H-H, Wang Y, Silva LAV, Sarbanes S, Sun T et al. (2018) Intrinsic immunity shapes viral resistance of stem cells. Cell 172, 423–438.e25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Buchrieser J, Degrelle SA, Couderc T, Nevers Q, Disson O, Manet C, Donahue DA, Porrot F, Hillion K-H, Perthame E et al. (2019) IFITM proteins inhibit placental syncytiotrophoblast formation and promote fetal demise. Science 365, 176–180. [DOI] [PubMed] [Google Scholar]
- 13.Zani A, Zhang L, McMichael TM, Kenney AD, Chemudupati M, Kwiek JJ, Liu S-L, and Yount JS (2019) Interferon-induced transmembrane proteins inhibit cell fusion mediated by trophoblast syncytins. J. Biol. Chem 294, 19844–19851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lee J, Robinson ME, Ma N, Artadji D, Ahmed MA, Xiao G, Sadras T, Deb G, Winchester J, Cosgun KN et al. (2020) IFITM3 functions as a PIP3 scaffold to amplify PI3K signalling in B cells. Nature 588, 491–497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hur J-Y, Frost GR, Wu X, Crump C, Pan SJ, Wong E, Barros M, Li T, Nie P, Zhai Y et al. (2020) The innate immunity protein IFITM3 modulates γ-secretase in Alzheimer’s disease. Nature 586, 735–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhao X, Li J, Winkler CA, An P, and Guo J-T (2018) IFITM genes, variants, and their roles in the control and pathogenesis of viral infections. Front. Microbiol 9, 3228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Chesarino NM, Compton AA, McMichael TM, Kenney AD, Zhang L, Soewarna V, Davis M, Schwartz O, and Yount JS (2017) IFITM3 requires an amphipathic helix for antiviral activity. EMBO Rep 18, 1740–1751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ling S, Zhang C, Wang W, Cai X, Yu L, Wu F, Zhang L, and Tian C (2016) Combined approaches of EPR and NMR illustrate only one transmembrane helix in the human IFITM3. Sci. Rep 6, 24029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yount JS, Moltedo B, Yang Y-Y, Charron G, Moran TM, López CB, and Hang HC (2010) Palmitoylome profiling reveals S-palmitoylation-dependent antiviral activity of IFITM3. Nat. Chem. Biol 6, 610–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Percher A, Ramakrishnan S, Thinon E, Yuan X, Yount JS, and Hang HC (2016) Mass-tag labeling reveals site-specific and endogenous levels of protein S-fatty acylation. Proc Natl Acad Sci USA 113, 4302–4307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Thinon E, Fernandez JP, Molina H, and Hang HC (2018) Selective Enrichment and Direct Analysis of Protein S-Palmitoylation Sites. J. Proteome Res 17, 1907–1922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Spence JS, He R, Hoffmann H-H, Das T, Thinon E, Rice CM, Peng T, Chandran K, and Hang HC (2019) IFITM3 directly engages and shuttles incoming virus particles to lysosomes. Nat. Chem. Biol 15, 259–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Benfield CT, MacKenzie F, Ritzefeld M, Mazzon M, Weston S, Tate EW, Teo BH, Smith SE, Kellam P, Holmes EC et al. (2020) Bat IFITM3 restriction depends on S-palmitoylation and a polymorphic site within the CD225 domain. Life Sci. Alliance 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.John SP, Chin CR, Perreira JM, Feeley EM, Aker AM, Savidis G, Smith SE, Elia AEH, Everitt AR, Vora M et al. (2013) The CD225 domain of IFITM3 is required for both IFITM protein association and inhibition of influenza A virus and dengue virus replication. J. Virol 87, 7837–7852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chesarino NM, McMichael TM, Hach JC, and Yount JS (2014) Phosphorylation of the antiviral protein interferon-inducible transmembrane protein 3 (IFITM3) dually regulates its endocytosis and ubiquitination. J. Biol. Chem 289, 11986–11992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wu X, Spence JS, Das T, Yuan X, Chen C, Zhang Y, Li Y, Sun Y, Chandran K, Hang HC et al. (2020) Site-Specific Photo-Crosslinking Proteomics Reveal Regulation of IFITM3 Trafficking and Turnover by VCP/p97 ATPase. Cell Chem. Biol 27, 571–585.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hubel P, Urban C, Bergant V, Schneider WM, Knauer B, Stukalov A, Scaturro P, Mann A, Brunotte L, Hoffmann HH, Schoggins JW et al. (2019) A protein-interaction network of interferon-stimulated genes extends the innate immune system landscape. Nat. Immunol 20, 493–502. [DOI] [PubMed] [Google Scholar]
- 28.Peng T, and Hang HC (2015) Bifunctional fatty acid chemical reporter for analyzing S-palmitoylated membrane protein-protein interactions in mammalian cells. J. Am. Chem. Soc 137, 556–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Fu B, Wang L, Li S, and Dorf ME (2017) ZMPSTE24 defends against influenza and other pathogenic viruses. J. Exp. Med 214, 919–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Amini-Bavil-Olyaee S, Choi YJ, Lee JH, Shi M, Huang I-C, Farzan M, and Jung JU (2013) The antiviral effector IFITM3 disrupts intracellular cholesterol homeostasis to block viral entry. Cell Host Microbe 13, 452–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kühnl A, Musiol A, Heitzig N, Johnson DE, Ehrhardt C, Grewal T, Gerke V, Ludwig S, and Rescher U (2018) Late Endosomal/Lysosomal Cholesterol Accumulation Is a Host Cell-Protective Mechanism Inhibiting Endosomal Escape of Influenza A Virus. MBio 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Desai TM, Marin M, Chin CR, Savidis G, Brass AL, and Melikyan GB (2014) IFITM3 restricts influenza A virus entry by blocking the formation of fusion pores following virus-endosome hemifusion. PLoS Pathog 10, e1004048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Teissier E, and Pécheur E-I (2007) Lipids as modulators of membrane fusion mediated by viral fusion proteins. Eur. Biophys. J 36, 887–899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sanders DW, Jumper CC, Ackerman PJ, Bracha D, Donlic A, Kim H, Kenney D, Castello-Serrano I, Suzuki S, Tamura T et al. (2020) SARS-CoV-2 Requires Cholesterol for Viral Entry and Pathological Syncytia Formation. BioRxiv [DOI] [PMC free article] [PubMed]
- 35.Suddala KC, Lee CC, Meraner P, Marin M, Markosyan RM, Desai TM, Cohen FS, Brass AL, and Melikyan GB (2019) Interferon-induced transmembrane protein 3 blocks fusion of sensitive but not resistant viruses by partitioning into virus-carrying endosomes. PLoS Pathog 15, e1007532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Guo X, Steinkühler J, Marin M, Li X, Lu W, Dimova R, and Melikyan GB (2021) Interferon-Induced Transmembrane Protein 3 Blocks Fusion of Diverse Enveloped Viruses by Altering Mechanical Properties of Cell Membranes. ACS Nano [DOI] [PMC free article] [PubMed]
- 37.Hulce JJ, Cognetta AB, Niphakis MJ, Tully SE, and Cravatt BF (2013) Proteome-wide mapping of cholesterol-interacting proteins in mammalian cells. Nat. Methods 10, 259–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Castellano BM, Thelen AM, Moldavski O, Feltes M, van der Welle REN, Mydock-McGrane L, Jiang X, van Eijkeren RJ, Davis OB, Louie SM et al. (2017) Lysosomal cholesterol activates mTORC1 via an SLC38A9-Niemann-Pick C1 signaling complex. Science 355, 1306–1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kwon HJ, Abi-Mosleh L, Wang ML, Deisenhofer J, Goldstein JL, Brown MS, and Infante RE (2009) Structure of N-terminal domain of NPC1 reveals distinct subdomains for binding and transfer of cholesterol. Cell 137, 1213–1224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hoop CL, Sivanandam VN, Kodali R, Srnec MN, and van der Wel PCA (2012) Structural characterization of the caveolin scaffolding domain in association with cholesterol-rich membranes. Biochemistry 51, 90–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Daniloski Z, Jordan TX, Wessels H-H, Hoagland DA, Kasela S, Legut M, Maniatis S, Mimitou EP, Lu L, Geller E et al. (2021) Identification of Required Host Factors for SARS-CoV-2 Infection in Human Cells. Cell 184, 92–105.e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Schneider WM, Luna JM, Hoffmann H-H, Sánchez-Rivera FJ, Leal AA, Ashbrook AW, Le Pen J, Michailidis E, Ricardo-Lax I, Peace A et al. (2020) Genome-scale identification of SARS-CoV-2 and pan-coronavirus host factor networks. BioRxiv [DOI] [PMC free article] [PubMed]
- 43.Levental I, Grzybek M, and Simons K (2010) Greasing their way: lipid modifications determine protein association with membrane rafts. Biochemistry 49, 6305–6316. [DOI] [PubMed] [Google Scholar]
- 44.Lorent JH, Diaz-Rohrer B, Lin X, Spring K, Gorfe AA, Levental KR, and Levental I (2017) Structural determinants and functional consequences of protein affinity for membrane rafts. Nat. Commun 8, 1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Fantini J, Di Scala C, Evans LS, Williamson PTF, and Barrantes FJ (2016) A mirror code for protein-cholesterol interactions in the two leaflets of biological membranes. Sci. Rep 6, 21907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fantini J, and Barrantes FJ (2013) How cholesterol interacts with membrane proteins: an exploration of cholesterol-binding sites including CRAC, CARC, and tilted domains. Front. Physiol 4, 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dieterle ME, Haslwanter D, Bortz RH, Wirchnianski AS, Lasso G, Vergnolle O, Abbasi SA, Fels JM, Laudermilch E, Florez C et al. (2020) A Replication-Competent Vesicular Stomatitis Virus for Studies of SARS-CoV-2 Spike-Mediated Cell Entry and Its Inhibition. Cell Host Microbe 28, 486–496.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zani A, Kenney AD, Kawahara J, Eddy AC, Wang X-L, Kc M, Lu M, Hemann EA, Li J, Peeples ME et al. (2021) Interferon-induced transmembrane protein 3 (IFITM3) limits lethality of SARS-CoV-2 in mice. BioRxiv [DOI] [PMC free article] [PubMed]
- 49.Zang R, Case JB, Yutuc E, Ma X, Shen S, Gomez Castro MF, Liu Z, Zeng Q, Zhao H, Son J et al. (2020) Cholesterol 25-hydroxylase suppresses SARS-CoV-2 replication by blocking membrane fusion. Proc Natl Acad Sci USA 117, 32105–32113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhao X, Zheng S, Chen D, Zheng M, Li X, Li G, Lin H, Chang J, Zeng H, and Guo J-T (2020) LY6E Restricts Entry of Human Coronaviruses, Including Currently Pandemic SARS-CoV-2. J. Virol 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Buchrieser J, Dufloo J, Hubert M, Monel B, Planas D, Rajah MM, Planchais C, Porrot F, Guivel-Benhassine F, Van der Werf S et al. (2020) Syncytia formation by SARS-CoV-2-infected cells. EMBO J 39, e106267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Shi G, Kenney AD, Kudryashova E, Zani A, Zhang L, Lai KK, Hall-Stoodley L, Robinson RT, Kudryashov DS, Compton AA et al. (2021) Opposing activities of IFITM proteins in SARS-CoV-2 infection. EMBO J 40, e106501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Prelli Bozzo C, Nchioua R, Volcic M, Koepke L, Krüger J, Schütz D, Heller S, Stürzel CM, Kmiec D, Conzelmann C et al. (2021) IFITM proteins promote SARS-CoV-2 infection and are targets for virus inhibition in vitro. Nat. Commun 12, 4584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Garst E, Lee H, Das T, Bhattacharya S, Percher A, Wiewiora R, Witte IP, Li Y, Goger M, Peng T et al. (2020) Site-specific lipidation enhances IFITM3 membrane interactions and antiviral activity. BioRxiv [DOI] [PMC free article] [PubMed]
- 55.Rao BD, Shrivastava S, and Chattopadhyay A (2017) Hydrophobic mismatch in membranes: when the tail matters, in Membrane organization and dynamics (Chattopadhyay A., Ed.), pp 375–387. Springer International Publishing, Cham. [Google Scholar]
- 56.de Meyer F, and Smit B (2009) Effect of cholesterol on the structure of a phospholipid bilayer. Proc Natl Acad Sci USA 106, 3654–3658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Rahman K, Coomer CA, Majdoul S, Ding SY, Padilla-Parra S, and Compton AA (2020) Homology-guided identification of a conserved motif linking the antiviral functions of IFITM3 to its oligomeric state. eLife 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Eierhoff T, Hrincius ER, Rescher U, Ludwig S, and Ehrhardt C (2010) The epidermal growth factor receptor (EGFR) promotes uptake of influenza A viruses (IAV) into host cells. PLoS Pathog 6, e1001099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, Zhang Q, Shi X, Wang Q, Zhang L et al. (2020) Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 581, 215–220. [DOI] [PubMed] [Google Scholar]
- 60.Carette JE, Raaben M, Wong AC, Herbert AS, Obernosterer G, Mulherkar N, Kuehne AI, Kranzusch PJ, Griffin AM, Ruthel G et al. (2011) Ebola virus entry requires the cholesterol transporter Niemann-Pick C1. Nature 477, 340–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Côté M, Misasi J, Ren T, Bruchez A, Lee K, Filone CM, Hensley L, Li Q, Ory D, Chandran K et al. (2011) Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection. Nature 477, 344–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Wang H, Shi Y, Song J, Qi J, Lu G, Yan J, and Gao GF (2016) Ebola Viral Glycoprotein Bound to Its Endosomal Receptor Niemann-Pick C1. Cell 164, 258–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wrensch F, Karsten CB, Gnirß K, Hoffmann M, Lu K, Takada A, Winkler M, Simmons G, and Pöhlmann S (2015) Interferon-Induced Transmembrane Protein-Mediated Inhibition of Host Cell Entry of Ebolaviruses. J. Infect. Dis 212 Suppl 2, S210–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Winstone H, Lista MJ, Reid AC, Bouton C, Pickering S, Galao RP, Kerridge C, Doores KJ, Swanson CM, and Neil SJD (2021) The Polybasic Cleavage Site in SARS-CoV-2 Spike Modulates Viral Sensitivity to Type I Interferon and IFITM2. J. Virol 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wong AC, Sandesara RG, Mulherkar N, Whelan SP, and Chandran K (2010) A forward genetic strategy reveals destabilizing mutations in the Ebolavirus glycoprotein that alter its protease dependence during cell entry. J. Virol 84, 163–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Huang I-C, Bailey CC, Weyer JL, Radoshitzky SR, Becker MM, Chiang JJ, Brass AL, Ahmed AA, Chi X, Dong L et al. (2011) Distinct patterns of IFITM-mediated restriction of filoviruses, SARS coronavirus, and influenza A virus. PLoS Pathog 7, e1001258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.McMichael TM, Zhang L, Chemudupati M, Hach JC, Kenney AD, Hang HC, and Yount JS (2017) The palmitoyltransferase ZDHHC20 enhances interferon-induced transmembrane protein 3 (IFITM3) palmitoylation and antiviral activity. J. Biol. Chem 292, 21517–21526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Linder ME, and Deschenes RJ (2007) Palmitoylation: policing protein stability and traffic. Nat. Rev. Mol. Cell Biol 8, 74–84. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.