ABSTRACT
Influenza D virus (IDV) utilizes bovines as a primary reservoir with periodical spillover to other hosts. We have previously demonstrated that IDV binds both 9-O-acetylated N-acetylneuraminic acid (Neu5,9Ac2) and 9-O-acetylated N-glycolylneuraminic acid (Neu5Gc9Ac). Bovines produce both Neu5,9Ac2 and Neu5Gc9Ac, while humans are genetically unable to synthesize Neu5Gc9Ac. 9-O-Acetylation of sialic acids is catalyzed by CASD1 via a covalent acetyl-enzyme intermediate. To characterize the role of Neu5,9Ac2 and Neu5Gc9Ac in IDV infection and determine which form of 9-O-acetylated sialic acids drives IDV entry, we took advantage of a CASD1 knockout (KO) MDCK cell line and carried out feeding experiments using synthetic 9-O-acetyl sialic acids in combination with the single-round and multi-round IDV infection assays. The data from our studies show that (i) CASD1 KO cells are resistant to IDV infection and lack of IDV binding to the cell surface is responsible for the failure of IDV replication; (ii) feeding CASD1 KO cells with Neu5,9Ac2 or Neu5Gc9Ac resulted in a dose-dependent rescue of IDV infectivity; and (iii) diverse IDVs replicated robustly in CASD1 KO cells fed with either Neu5,9Ac2 or Neu5Gc9Ac at a level similar to that in wild-type cells with a functional CASD1. These data demonstrate that IDV can utilize Neu5,9Ac2- or non-human Neu5Gc9Ac-containing glycan receptor for infection. Our findings provide evidence that IDV has acquired the ability to infect and transmit among agricultural animals that are enriched in Neu5Gc9Ac, in addition to posing a zoonotic risk to humans expressing only Neu5,9Ac2.
IMPORTANCE
Influenza D virus (IDV) has emerged as a multiple-species-infecting pathogen with bovines as a primary reservoir. Little is known about the functional receptor that drives IDV entry and promotes its cross-species spillover potential among different hosts. Here, we demonstrated that IDV binds exclusively to 9-O-acetylated N-acetylneuraminic acid (Neu5,9Ac2) and non-human 9-O-acetylated N-glycolylneuraminic acid (Neu5Gc9Ac) and utilizes both for entry and infection. This ability in effective engagement of both 9-O-acetylated sialic acids as functional receptors for infection provides an evolutionary advantage to IDV for expanding its host range. This finding also indicates that IDV has the potential to emerge in humans because Neu5,9Ac2 is ubiquitously expressed in human tissues, including lung. Thus, results of our study highlight a need for continued surveillance of IDV in humans, as well as for further investigation of its biology and cross-species transmission mechanism.
KEYWORDS: influenza D; receptor; CASD1; Neu5,9Ac2; Neu5Gc9Ac
INTRODUCTION
The Orthomyxoviridae family consists of four influenza virus-related genera: alpha influenza (influenza A), beta influenza (influenza B), gamma influenza (influenza C), and delta influenza (influenza D) (1). Influenza A (IAV) and B (IBV) viruses contain an eight single-stranded, negative-sense RNA genome, while influenza C (ICV) and D (IDV) viruses have a seven-segmented RNA genome with negative polarity. Both IAV and IBV contain two major surface glycoproteins: the hemagglutinin (HA) and the neuraminidase (NA). In contrast, ICV and IDV have only one major surface glycoprotein, the hemagglutinin-esterase-fusion (HEF) protein, which communicates with the cell surface receptor and drives viral entry.
IDV as a new type of influenza virus is the latest addition to the Orthomyxoviridae family after its first identification from a diseased pig experiencing influenza-like symptoms. Subsequent studies identified the cattle as a primary reservoir of IDV (2, 3). IDV has a broad host range as antibodies against IDV have been found in sheep, goat, horses, and camels (4–8). More recently, serological studies in wildlife showed the IDV antibody prevalence in wild boar, deer, and hedgehogs (6, 9), indicating that the host range of IDV may be extremely broad. Although no human infection with IDV is reported clinically, antibodies against IDV were detected in people in Italy and U.S., and viral RNA were found in the nasal washes of cattle handlers (2, 10–12) and swine farmers (13). Furthermore, IDV genome was detected in the bio-aerosol sampling of respiratory viruses in the airport (14) and hospital emergency room setting (15). Experimentally challenged small animals like ferret, mice, and guinea pigs also demonstrated robust IDV replication, as well as its transmission to naïve animals (2, 16–18). All these laboratory animals have been used extensively as model systems to study human IAVs and their pathogenesis. Thus, the zoonotic spillover potential and transmission of IDV in humans cannot be excluded (19).
Influenza viruses are known to use cell surface sialic acid-containing glycoconjugates as entry receptors for infection (20). Sialic acids are 9-carbon α-keto aldonic acids that are usually present at the terminal positions of glycoproteins or glycolipids extending outward from the cell surface (21, 22). There is a great diversity in sialic acids due to different substitutions that can occur at carbon 4, 5, 7, 8, and 9 positions (23). N-acetyl neuraminic acid (Neu5Ac) is the predominant form of sialic acid species in humans (24). Agricultural animals, such as cattle and swine, have the ability to convert it to N-glycolylneuraminic acid (Neu5Gc) (25). Neu5Gc is synthesized via the addition of a hydroxyl group to the N-acetyl group of Neu5Ac in its sugar nucleotide form by cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) encoded by CMAH gene (26). Neu5Gc differs from Neu5Ac by an additional oxygen atom in the N-acyl moiety. Humans lack a functional CMAH enzyme due to a naturally occurred 92-bp deletion in CMAH gene and thus cannot synthesize Neu5Gc (27). Addition of “O-acetyl” group at the C-4/7/8/9 position of sialic acids is a common modification widely occurring in nature (28). This modification takes place inside the Golgi system, which is believed to be catalyzed by an enzyme termed CAS1 domain containing 1 (CASD1) encoded by CASD1 gene (29).
ICV, distantly related to IDV, is known to utilize 9-O-acetylated N-acetylneuraminic acid (Neu5,9Ac2) for entry (30). Interestingly, ICV has a narrow tropism, while IDV has a broad host range (31). Using the glycan microarray-based approach, we have previously shown that IDV bound to both Neu5,9Ac2- and Neu5Gc9Ac-terminated glycans (32). The binding specificity of IDV to the 9-O-acetylated sialic acid-containing glycans observed in the glycan array was further confirmed by the receptor analog-mediated inhibition in the traditional red blood cell (RBC) agglutination and cell-based replication assays (32). However, there is no absolute correlation between receptor binding and its functionality in driving viral entry. Binding assays with the HA proteins of influenza A viruses from avian, human, and equine showed that these influenza viruses were capable of binding both Neu5Ac and Neu5Gc; however, only equine H7N7 is known to exclusively utilize Neu5Gc as a receptor (33, 34). We have previously demonstrated that IDV has evolved with the ability to bind both Neu5,9Ac2 and Neu5Gc9Ac. However, whether binding can mediate functional entry of IDV and whether both forms of 9-O-acetylated sialic acids (Neu5,9Ac2 and Neu5Gc9Ac) serve as functional entry receptors to promote IDV infection have remained unknown, which is the primary focus of this work.
In this study, we took advantage of a CASD1 (sialate O-acetyltransferase gene) knockout (KO) MDCK cell line and carried out feeding experiments with exogenous synthetic 9-O-acetyl sialic acids, in combination with the single-round and multi-round IDV infection assays, to delineate the role of Neu5,9Ac2 and Neu5Gc9Ac in IDV entry and infection. The results of our experiments showed that IDV can utilize both Neu5,9Ac2- and Neu5Gc9Ac-containing glycan receptors for infection. Our findings provide evidence that IDV has acquired the ability to infect and transmit among agricultural animals, such as bovines that are relatively enriched in Neu5Gc9Ac, in addition to posing a zoonotic risk to humans expressing only Neu5,9Ac2.
MATERIALS AND METHODS
Cells, virus, and antibodies
HEK-293T (Human embryonic kidney) and MDCK-ATL cells were cultured in Dulbecco’s modified Eagle medium (DMEM; Gibco, Invitrogen, USA) supplemented with 10% (vol/vol) fetal bovine serum (FBS; Gibco, USA) and 100 U/mL penicillin-streptomycin (Life Technologies, Carlsbad, CA, USA). The CASD1-KO MDCK and its parental MDCK-NBL-II were cultured in DMEM supplemented with 5% (vol/vol) FBS and 100 U/mL penicillin-streptomycin. Cells were cultured at 37°C under 5% CO2. Influenza D/swine/Oklahoma/1314/2011 (D/OK), D/bovine/Oklahoma/660/2013 (D/660), D/bovine/California/0363/2019 (D/CA), and D/swine/Kentucky/17TOSU1262/2017 (D/swine/Kentucky) were propagated in MDCK-ATL cells at 33°C. Anti-D/OK, anti-D/CA, and anti-D/660 rabbit polyclonal sera were generated in house following the standard immunization procedure with the inactivated viral cultures. Influenza C/Victoria/2/2012 was propagated in MDCK-ATL cells at 33°C. The Six-His tag mouse monoclonal antibody was obtained from Invitrogen (MA1-21315). Alexa Fluor 488 goat anti-mouse (A-11001) and Alexa fluor 488 goat anti-rabbit (A-11008) antibodies were purchased, respectively, from Invitrogen.
Development of single-round IDV reporter assays
Two single-round reporter assays were developed to study the receptor biology of IDV. The first assay is the green fluorescent protein (GFP)-based reporter assay. The GFP coding sequence was inserted between the 5′- and 3′-end sequences of the D/OK HEF viral cDNA flanked with human polymerase I promoter and polymerase I terminator sequences, respectively. Terminal 240 nucleotide sequences from both directions of the HEF segment were retained in the reporter plasmid for the efficient package of this reporter segment into virus particles, while the native initiation codon (ATG) in the HEF was changed into CTG. To produce IDV GFP reporter virus, seven plasmids (1 µg each) encoding each of seven D/OK genomic segments together with HEF-based GFP reporter plasmid were co-transfected in the cell monolayer comprising co-culture of HEK-293T and MDCK-ATL cells. The supernatant was collected 4–5 days post-transfection for further analysis.
A seven-plasmid Nano Luciferase (Nluc)-based single-round assay (D/OK-NLuc) was also developed for this study. Seventh segment of IDV genome produces two proteins NS1 and NS2 (also called NEP, nuclear export protein). The unspliced mRNA is translated into NS1 protein, while a splicing event produces NS2 protein. To generate a D/OK luciferase reporter virus, a single non-overlapping transcript expressing D/OK NS1 and NEP was separated by a porcine teschovirus 1 (PTV-1) self-cleaving element 2A. NLuc was linked to the C-terminal end of NS1 followed by the PTV-2A element (Fig. 4B). The reporter plasmid (D/OK-NLuc) replaced wild-type D/OK NS segment during the transfection. To produce IDV Nano-Luc reporter virus, seven plasmids (1 µg each) of D/OK (NS NLuc plasmid with the other six wild-type plasmids) were co-transfected into the co-culture consisting of HEK-293T and MDCK-ATL cells. At 4–5 days post-transfection, viral supernatant was collected for further analysis.
Flow cytometry assay and quantification of D/OK-GFP reporter virus
D/OK-GFP reporter virus was quantified using a conventional flow cytometry assay, and its titer was expressed as fluorescence units per milliliter (FU/mL). Briefly, 106 MDCK cells were seeded in a six-well plate. Next day, 1 mL of 10-fold serial dilutions of D/OK-GFP reporter virus was used to inoculate cells. After 1 hour of incubation at 37°C, inoculum was discarded, and cells were washed three times with 1× PBS. Cells were then incubated for 24 hours at 37°C in 1 mL of DMEM media containing 1 µg/mL TPCK-treated trypsin (Thermo Fisher: 20233). After 24 hours post-infection (hpi), cells were washed three times with 1× PBS followed by the digestion with trypsin-EDTA (Thermo Fisher: 25200056). Cells were pelleted down, washed once in 1× PBS, and resuspended in 2% paraformaldehyde. The percentage of GFP-positive cells was calculated using BD FACSymphony A3 cell analyzer (BD Biosciences) and the associated software. For further analysis, 10,000 cells were gated. Reporter virus titer was calculated as: FU/mL = (initial cell count × % GFP positive)/dilution, where initial cell count is the number of cells seeded, dilution is the corresponding virus dilution used for flow cytometry analysis, and percentage of GFP-positive cells is obtained from flow cytometry. For all the assays involving the D/OK-GFP reporter virus, 106 FU of reporter virus was used.
9-O-Acetyl sialic acid feeding assay
Synthetic Neu5,9Ac2 and Neu5Gc9Ac (35, 36) were purchased from ChemBind (custom synthesis) and were shown to have more than 95% purity by mass spectrometry and nuclear magnetic resonance spectroscopy. Approximately 105 CASD1 KO MDCK cells were seeded in a 96-well plate in DMEM complete media (5% FBS, 1% Pen-strep). Similar numbers of WT MDCK cells were seeded in separate wells. After overnight incubation, cells were washed once with 1× PBS and fed with various concentrations of Neu5,9Ac2 or Neu5Gc9Ac, or a mixture containing both in equal concentrations in DMEM complete media (5% FBS, 1% Pen-strep). Cells were incubated for 3 days without changing media. On day 3, cells were washed three to four times with 1× PBS and infected with reporter or wild-type virus.
Cell-binding assay with the recombinant receptor-binding domain of D/OK HEF
D/OK HEF receptor-binding domain (RBD) with a histidine (His) tag at the C-terminus was expressed in E. coli BL21 using a T7 RNA polymerase-IPTG induction system. The expressed RBD protein was purified by a nickel column (Qiagen), and the protein concentration was measured by the Bio-Rad Protein Assay. To determine viral HEF RBD protein binding to the plasma membrane of various cells, CASD1-KO or WT MDCK cells were seeded in 96-well plates. CASD1-KO cells were exogenously supplemented with 200 µM of Neu5,9Ac2 and/or Neu5Gc9Ac. On the third day, cells were washed and sequentially incubated with the recombinant His-tagged RBD (20 µg/mL) and the His-tag-specific mouse antibody (5 µg/mL) at room temperature. After three washes with 1× PBS, cells were incubated with Alexa-Fluor 488-labeled anti-mouse antibody (1:300 dilution; Abcam) for 30 min at room temperature. The nuclei were stained with 4′,6-diamidino-2- phenylindole (DAPI) for 5 min. The samples were imaged on a Nikon Ti eclipse fluorescence microscope, and the images were merged using the Image J software.
Single-round reporter virus infection assay
After 3 days of exogenous receptor feeding, cells were washed three times with 1× PBS. D/OK-GFP reporter virus was inoculated at 106 FU. After 1 hour of incubation at 37°C, cells were washed three times with 1× PBS and maintained in DMEM containing 1 µg/mL TPCK-treated trypsin. After 24 hours of incubation at 37°C, D/OK-GFP-infected cells were processed for flow cytometry analysis, as described above. For D/OK-NLuc reporter infection assay, cells were infected at the multiplicity of infection (MOI) of 0.1. Receptor-fed CASD1-KO cells infected with D/OK-NLuc were assessed for the luciferase activity using the Nano-Glo Luciferase Assay Kit (Promega). After 24 hpi, virus-infected cells in white culture plates and assay reagents were equilibrated at room temperature prior to the assay. Freshly prepared assay reagents were mixed with equal volume of cell culture media. Nluc was measured in a Synergy HT BioTek plate reader. Each individual experiment assayed samples in triplicate. The D/OK-GFP reporter assay was repeated nine times (n = 9), while the D/OK-NLuc reporter assay was conducted six times (n = 6).
Multi-round replication assay
CASD1-KO cells fed with 200 µM Neu5,9Ac2 and/or Neu5Gc9Ac were incubated for 3 days. Treated cells were washed and infected with 1 MOI of D/OK, D/660, D/CA, and D/swine/Kentucky. The 50% tissue culture infection dose per milliliter (TCID50/mL) of the inoculum was 106.33, 106.5, 106, and 106.83, respectively, for D/OK, D/660, DD/CA, and D/swine/Kentucky. Final infection volume was adjusted with DMEM to make the equal amount of infectious dose across different virus strains (1.0 MOI). After 1 hour of incubation at 33°C, cells were washed and maintained in 100 µL of DMEM containing 1 µg/mL TPCK trypsin at 33°C. The 100-µL media were collected every day and replaced with fresh DMEM containing 1 µg/mL TPCK trypsin during the 5-day experiment. Three independent experiments were conducted with each sample analyzed in triplicate. The inoculum was titrated using the traditional 50% tissue culture infection dose (TCID50) assay. Briefly, 1.5 × 104 MDCK ATL cells were seeded in 96-well plates. Serial 10-fold dilutions of viral supernatants were prepared in DMEM containing 1 µg/mL TPCK trypsin, and 100 µL was transferred into pre-seeded cell plates. The plate was then incubated at 33°C for 5 days. After 5 days, 25 µL of supernatant was used for the measurement of viral hemagglutination using 1% turkey RBC. Virus titer was calculated using the classical Reed-Muench method (37).
Immunofluorescence assay
WT cells, CASD1 KO, and receptor-fed CASD1-KO cells were fixed at 72 hours post virus infection with 80% acetone. These acetone-fixed virus-infected cells were incubated with 1% BSA in 1× PBS to block the non-specific binding. Rabbit polyclonal antibody against IDV D/OK, D/CA, and D/660 was used at 1:200 dilution. For D/swine/Kentucky infection, anti-D/OK rabbit antibody was used. After 1 hour of incubation at 37°C, cells were washed three times with 1× PBS followed by additional incubation with goat-anti rabbit Alexa-Fluor 488 antibody at 1:400 dilution (Thermo Fisher: A11008) for 45 min at 37°C. After three washes with 1× PBS, cells were counterstained with DAPI. Images were taken by the visualization under Nikon Ti eclipse fluorescence microscope, and images were merged using ImageJ software.
Statistical analysis
Statistical analyses were performed in GraphPad Prism version 9. To test the statistical significance, two-tailed t-test assuming equal variance was performed for the reporter virus infection assay. For multi-round replication assay, two-way analysis of variance followed by Tukey’s multiple comparison test was performed. Statistically significant data are indicated by asterisk (*, P < 0.05; **, P < 0.01). Data are represented as mean ± standard error of mean (SEM).
RESULTS
CASD1-KO MDCK cells are resistant to IDV infection
To investigate what form of 9-O-acetylated sialic acids is utilized by IDV for entry into cells, we first developed an IDV D/OK-reporter virus by taking advantage of the reverse genetics system (RGS) we have already developed for the virus (38). Briefly, the pUC57mini-D/OK-HEF-reporter plasmid was created by inserting the GFP-coding sequence between the 5′- and 3′-end sequences of the D/OK HEF segment flanked with human polymerase I promoter and polymerase I terminator sequences, respectively (Fig. 1A). The native HEF sequences of 240 nucleotides at both the 5′ and 3' ends were retained to ensure the efficient package of the reporter segment (Fig. 1A). All the ATG codons before GFP translation initiation codon were changed into CTG codons, so the downstream GFP reporter protein can be expressed properly. Seven WT RGS plasmids and the GFP reporter plasmid (1 µg each) were co-transfected into cell monolayers of HEK-293T and MDCK-ATL co-cultured cells. Supernatants were collected after 4–5 days (passage 0) followed by further passaging in fresh MDCK-ATL cells. Fluorescence microscopy showed successful rescue and propagation of D/OK-GFP reporter virus (Fig. 1B); however, further passaging showed a progressive loss of reporter gene expression (Fig. 1B), which is likely caused by the inability of seven-segmented IDV to stably package the eighth segmented genome. Nevertheless, these data demonstrate that the IDV D/OK-GFP reporter establishes a single round of infection in target cells, thus providing a valid reporter virus to detect IDV infection in host cells.
Fig 1.
Generation of D/OK-GFP reporter virus and infection of WT and CASD1-KO MDCK cells. (A) Schematic representation of D/OK-GFP plasmid. The GFP open reading frame is inserted between the 5'- and 3'-ends of the D/OK-HEF cDNA, which is flanked by the Pol I terminator and the human RNA Pol I promoter. All ATG codons before the native GFP translation initiation codon in the construct were mutated to CTG so that GFP gene can be expressed in native form. (B) Microscopy images showing a successful rescue and propagation of D/OK-GFP reporter virus in passages 1–4. Infection of WT (C) and CASD1-KO (D) MDCK cells with the D/OK-GFP reporter virus is shown with images representative of three independent experiments. Scale bar represents 50 µm.
Based on the above observation, we selected passage 1 (P1) IDV D/OK-GFP reporter virus for this study. The titer of the D/OK-GFP was calculated by flow cytometry, and 106 FU/mL of the reporter virus was used in our assays. Following infection, WT MDCK cells showed strong GFP expression (Fig. 1C) under a fluorescence microscope. However, infection of CASD1-KO cells under the identical conditions resulted in no detectable fluorescence signals, indicating that CASD1-KO cell is refractory to IDV infection (Fig. 1D).
IDV requires 9-O-acetylated sialic acid for cell attachment
To further determine whether the failure of IDV infection of CASD1-KO cells was caused by the absence of a functional IDV receptor in our assay system, we performed the cell-binding assay with the receptor-binding domain of IDV D/OK HEF protein, which was produced and purified from E. coli. As shown in Fig. 2, binding of IDV RBD to WT MDCK cells resulted in strong fluorescence signals (Fig. 2A), which was in marked contrast with RBD binding to CASD1-KO cells where only background signals were observed (Fig. 2B). These data indicate that CASD1-KO cells lose the ability to express IDV’s 9-O-acetylated sialic acid receptors as reported previously (39), which explains the lack of IDV RBD binding to the cell surface of CASD1 KO cells and no detectable IDV D/OK-GFP reporter virus replication in these cells (Fig. 1D).
Fig 2.
Cell binding of the D/OK-HEF RBD to MDCK cells. Images in the top panel represent immunostaining of WT MDCK cells (A), CASD1-KO MDCK cells (B) without feeding with exogenous O-acetylated sialic acids, Neu5,9Ac2-fed CASD1-KO MDCK cells (C), and Neu5Gc9Ac-fed CASD1-KO MDCK cells (D). Note that following the incubation with D/OK-HEF RBD containing His-tag (20 µg/mL), washed cells were further incubated with mouse anti-His antibody and Alexa-Fluor 488-labeled anti-mouse antibody. Nuclear staining of WT MDCK (E), CASD1-KO MDCK cells without external receptor feeding (F), Neu5,9Ac2-fed CASD1-KO MDCK cells (G), and Neu5Gc9Ac-fed CASD1-KO MDCK cells (H), is shown in the middle panel. The bottom panel shows merged DAPI and fluorescence for WT MDCK (I), CASD1-KO MDCK (J), Neu5,9Ac2-fed (K), and Neu5Gc9Ac-fed CASD1-KO cells (L). Images representative of three independent experiments. Scale bar represents 50 µm.
Next, we examined whether exogenous feeding of CASD1-KO cells with IDV’s 9-O-acetylated sialic acid receptors rescues the IDV RBD binding to these cells. For this experiment, cells were fed with exogenously added synthetic receptors followed by surface binding assay with the IDV D/OK RBD. CASD1-KO cells were seeded in 96-well plates and treated with 200 µM of synthetic Neu5,9Ac2 or Neu5Gc9Ac. After 3 days, cells were stained using His-tagged D/OK RBD. As shown in Fig. 2C and D, both exogenous receptor analogs restored the IDV RBD binding to CASD1-KO cells to levels similar to that of RBD binding to WT cells (Fig. 2A). These results indicate that CASD1-KO cells are capable of metabolic uptake of external 9-O-acetylated sialic acids and that these sialic acids are efficiently displayed on the cell surface, accessible to binding by the IDV RBD. This feeding assay with exogenous 9-O-acetylated sialic acids was further explored in experiments below to study IDV receptor biology. Our results also indicate that IDV D/OK specifically binds to 9-O-acetylated sialic acids, and this interaction is essential for viral binding/entry.
IDV utilizes both Neu5,9Ac2 and Neu5Gc9Ac receptors for entry into host cells
Feeding experiments demonstrated that D/OK-RBD binds specifically to CASD1-KO cells only when synthetic 9-O-acetylated sialic acids were added in the culture media (Fig. 2C and D). Next, we asked whether binding to these receptors can lead to productive IDV entry and replication. In this experiment, we used the single-round D/OK-GFP reporter virus in combination with receptor feeding, as described above. CASD1-KO cells were fed with Neu5,9Ac2 or Neu5Gc9Ac at four different concentrations (100, 200, 400, and 600 µM), followed by extensive washings and infection with 106 FU of D/OK-GFP virus. After 18–24 hpi, percentage of GFP-positive cells was quantified using flow cytometry. Flow cytometry analysis showed that D/OK-GFP reporter virus utilized both receptor analogs effectively for entry and replication. Mean percentage of GFP-positive cells for Neu5,9Ac2 was 4.5% (100 µM), 19.1% (200 µM), 38.7% (400 µM), and 46.5% (600 µM) (Fig. 3A). Similarly, in Neu5Gc9Ac feeding experiment, mean percentage of GFP-positive cells was 1.9% (100 µM), 8.3% (200 µM), 22.6% (400 µM), and 31.5% (600 µM) (Fig. 3A). The increasing concentration of fed receptor resulted in a proportionate increase in the percentage of GFP-positive cells for both Neu5,9Ac2 and Neu5Gc9Ac receptors. A dose-dependent increase in GFP-positive cells was also qualitatively captured under fluorescence microscopy. With increasing Neu5,9Ac2 concentration, we observed a proportional increase in GFP-expressing cells (Fig. 3C through F). Similarly, GFP-expressing cells increased proportionally as concentration of Neu5Gc9Ac increased (Fig. 3G through J). As expected, CASD1-KO cells remained uninfected (Fig. 3A and L). As a negative control for the 9-O-acetyl sialic acid feeding experiments, CASD1-KO cells fed with synthetic Neu5Ac without 9-O-acetyl modification resulted in no detectable infection (Fig. 3M). It should be noted that infectivity levels in CASD1-KO cells fed with 400 µM (Fig. 3A and E) or 600 µM (Fig. 3A and F) of Neu5,9Ac2 or 600 µM of Neu5Gc9Ac (Fig. 3A and J) were very comparable to that observed in infection of WT MDCK cells (Fig. 3A and K). We noticed that D/OK-GFP reporter virus appeared to replicate more efficiently in CASD1-KO cells fed with Neu5,9Ac2 than with Neu5Gc9Ac at all four different concentrations tested (Fig. 3A and B), which was supported by statistical analysis (P < 0.04, P < 0.02, P < 0.08, P < 0.02, respectively, for 100, 200, 400, and 600 µM groups). Taken together, these data suggest that IDV D/OK can utilize both Neu5,9Ac2 and Neu5Gc9Ac receptors for entry into host cells.
Fig 3.
Dose-dependent enhancement of D/OK-GFP infectivity in 9-O-acetyl sialic acid-fed CASD1-KO MDCK cells. (A) Percentages of GFP-positive cells infected with IDV D/OK GFP reporter virus at 24 hpi with exogenous Neu5,9Ac2 and Neu5Gc9Ac treatment. WT- and CASD1-KO (without exogenous receptor feeding) MDCK cells were used as internal controls. (B) Ratio of percentage of GFP-positive cells observed between Neu5,9Ac2-fed and Neu5Gc9Ac-fed CASD1-KO cells (Neu5,9Ac2/Neu5Gc9Ac). Fluorescence images of D/OK-GFP reporter virus-infected CASD1-KO cells in the presence of 100 (C), 200 (D), 400 (E), and 600 µM (F) of Neu5,9Ac2 or 100 (G), 200 (H), 400 (I), and 600 µM (J) of Neu5Gc9Ac. Fluorescence images of D/OK-GFP reporter virus-infected WT (K), CASD1-KO (L), and CASD1-KO fed with Neu5Ac (M) are also shown. Bar graphs denote the mean of nine independent experiments with error bar representing SEM. Statistical significance is indicated by * (P < 0.05) and ** (P < 0.001).
Combination of Neu5,9Ac2 and Neu5Gc9Ac receptors enhances IDV replication
GFP expression levels within cells are not uniform, and analysis is often confounded by the inability to distinguish low GFP expression levels and autofluorescence that may arise due to cellular components like flavin. Thus, we generated a more sensitive and specific Nluc-based reporter virus (D/OK-NLuc). The seventh segment of IDV genome produces two proteins, NS1 and NEP, through a canonical splicing event. The unspliced mRNA is translated as NS1, while the spliced transcript gives rise to NEP (Fig. 4A). To generate IDV D/OK-Nluc reporter virus, a single non-overlapping transcript expressing D/OK NS1 and NEP was separated by using the PTV-1 self-cleaving element 2A peptide. Nluc was linked to the C-terminal end of NS1 followed by PTV-2A element (Fig. 4B) and the entire open reading frame of NEP. D/OK-NLuc reporter plasmid in substitution of WT NS plasmid was used together with six other RGS plasmids (PB2, PB1, PA, HEF, NP, and M) for transfection of co-cultures of HEK-293T and WT MDCK cells to generate D/OK-NLuc reporter virus, which was subsequently used for infection of 9-O-acetyl sialic acid-fed CASD1 KO cells to determine how the presence of both Neu5,9Ac2- and Neu5Gc9Ac-containing receptors on cell surface influences IDV D/OK replication and infectivity. In this experiment, CASD1-KO cells were fed with a range of concentrations of individual Neu5,9Ac2 or Neu5Gc9Ac or an equimolar mixture of both (with an identical total concentration across conditions) for 3 days, followed by infection with D/OK-NLuc reporter virus (0.1 MOI) and the measurement of NanoLuc luciferase (RLU) as a surrogate of viral titer. For example, 200 µM single 9-O-acetylated sialic acid was used for cell feeding, while 200 µM mixed 9-O-acetylated sialic acids (combination) for cell feeding consisted of 100 µM of each of the two.
Fig 4.
D/OK-NLuc reporter virus infection in 9-O-acetyl sialic acid-fed CASD1-KO MDCK cells. Schematic representation of WT (A) and modified (B) IDV NS segment. (C) Levels of Nluc expression detected in cell culture supernatants of CASD1-KO MDCK cells in the absence or in the presence of different 9-O-acetyl sialic acids. WT MDCK cells infected with IDV D/OK-NLuc reporter virus were included as control. (D) Ratio of RLUs observed between Neu5,9Ac2-fed and Neu5Gc9Ac-fed CASD1-KO cells (Neu5,9Ac2/Neu5Gc9Ac) following infection with D/OK-NLuc reporter virus. Bar graphs denote the mean of six independent experiments with error bar representing standard error of mean. Statistical significance is indicated by * (P < 0.05) and ** (P < 0.001). NCR, non-coding region. 2A: porcine teschovirus 2A sequence.
D/OK-NLuc titers (RLU) increased in CASD1-KO cells in a dose-dependent fashion in response to exogenous 9-O-acetylated sialic acid feeding. Mean RLUs were 2,844, 16,340, 40,973, and 61,744 for cells fed with Neu5,9Ac2 at 100, 200, 400, and 600 µM concentrations, respectively (Fig. 4C). Similarly, in the Neu5Gc9Ac feeding experiment, the mean RLUs were 2,851, 11,145, 40,382, and 64,628 for cells fed with Neu5Gc9Ac at 100, 200, 400, and 600 µM concentrations, respectively (Fig. 4C). Notably, in the presence of 600 µM Neu5,9Ac2 or Neu5Gc9Ac, viral titers in CASD1-KO cells reached to a level similar to that observed in WT MDCK cells infected with D/OK-Nluc reporter virus. An interesting note is that the observed differences in viral titers between Neu5,9Ac2- and Neu5Gc9Ac-fed cells became insignificant (P < 0.99, P < 0.059, P < 0.93, and P < 0.69 for 100, 200, 400, and 600 µM, respectively) for the D/OK-Nluc reporter virus when compared to the D/OK-GFP reporter virus system, especially when higher concentrations of 9-O-acetyl sialic acids were used (400 and 600 µM) (Fig. 4C and D versus Fig. 3A). Nevertheless, D/OK-Nluc reporter virus appeared to replicate slightly better in cells fed with lower concentrations of Neu5,9Ac2 (100 and 200 µM) than cells fed with Neu5Gc9Ac (Fig. 4C and D), similar to the results observed using the D/OK-GFP reporter virus system (Fig. 3A). Finally, we observed that the combination of two 9-O-acetyl sialic acids in exogenous feeding significantly enhanced IDV D/OK-Nluc reporter virus replication in CASD1-KO cells when compared to cells individually treated with each of the compounds at all four different concentrations tested (100, 200, 400, and 600 µM) (Fig. 4C). The viral titer differences between the mixtures of the two and single compound (Neu5,9Ac2 and Neu5Gc9Ac) groups were statistically significant at all concentration groups (Fig. 4C), which indicate that the combination of two receptors can confer the replication advantage to IDV D/OK-Nluc reporter virus in CASD1-KO cells. Overall, these data recapitulated what has been observed previously using the IDV D/OK GFP reporter virus (Fig. 4), thereby emphasizing that IDV can utilize either receptor for productive entry.
IDVs of swine and bovine origin use both Neu5,9Ac2 and Neu5Gc9Ac equally well in a multi-round replication assay
To investigate if diverse IDV strains differ in their ability to utilize Neu5,9Ac2 or Neu5Gc9Ac for entry and infection, we used a traditional multi-round replication assay with four representative IDV strains isolated from swine or bovine. D/OK and D/swine/KY originated from swine, while D/660 and D/CA were isolated from cattle. CASD1-KO cells were fed with 200 µM of a single compound (Neu5,9Ac2 or Neu5Gc9Ac) and then infected (1.0 MOI) with different IDVs. D/OK virus replicated to higher titers in Neu5,9Ac2-fed cells than Neu5Gc9Ac-fed cells. Although mean viral titers were similar until day 2 for both Neu5,9Ac2- and Neu5Gc9Ac-treated cells, D/OK had nearly one-log higher titers from day 3 onward in Neu5,9Ac2-fed cells. Statistically significant differences were observed (P < 0.02) at day 4 for D/OK viral titer with Neu5,9Ac2 (mean titer of 105) than Neu5Gc9Ac (mean titer of 104.2) (Fig. 5A). D/OK in WT cells had the highest titer at day 5 (mean titer of 106) (Fig. 5A). Like swine-originated D/OK virus, D/swine/KY had slightly better replication kinetics in Neu5,9Ac2 than Neu5Gc9Ac-fed cells during the initial 3 days, although no statistical difference was observed. The highest viral titer was observed at day 3 with mean titer of 106.9 for Neu5,9Ac2 and 106.6 for Neu5Gc9Ac-fed cells (Fig. 5B). D/swine/KY propagated to high titers in WT MDCK cells, with peak of infection at day 2 (mean titer of 107.4) (Fig. 5B). D/660, at day 1, had slightly better replication in Neu5,9Ac2-fed cells (Fig. 5C). However, at days 2 and 3, viral titers were similar between Neu5,9Ac2- and Neu5Gc9Ac-fed cells. Interestingly, at days 4 and 5, Neu5Gc9Ac-fed cells showed higher replication of D/600 virus (Fig. 5C). D/660 propagated on WT MDCK with peak titer of 106 on day 2 post-infection (Fig. 5C). Despite notable differences in viral titers between Neu5,9Ac2- and Neu5Gc9Ac-fed cells, no statistically significant differences were revealed at all time points. D/CA showed higher replication in Neu5Gc9Ac compared to Neu5,9Ac2-fed cells after day 3 (Fig. 5D). Notably, at day 5, there was significant difference in replication of D/CA in Neu5Gc9Ac-treated cells than Neu5,9Ac2-fed cells (P < 0.00007) with mean viral titers of 105.5 and 105.1, respectively, for Neu5Gc9Ac- and Neu5,9Ac2-fed cells (Fig. 5D). The peak viral titer was at day 3 with mean titers of 105.7 and 105.6 for Neu5Gc9Ac- and Neu5,9Ac2-fed cells, respectively (Fig. 5D), while peak titers of D/CA in WT MDCK were observed at day 2 (mean viral titer of 105.5) (Fig. 5D). CASD1-KO cells without any receptor feeding were refractory to infections by all four IDV strains (Fig. 5A through D). Taken together, multi-round replication results confirmed the results of D/OK-based single-round replication assay that diverse IDVs utilize both receptors for productive entry and infection in host cells.
Fig 5.
Replication kinetics of swine and bovine origin IDV strains in receptor-treated CASD1-KO MDCK cells. CASD1-KO MDCK cells were fed with synthetic Neu5,9Ac2 or Neu5Gc9Ac (200 µM) for 3 days. Treated cells were then infected (1 MOI) with D/OK (A), D/swine/KY (B), D/660 (C), and D/CA (D), respectively. After 1 hour of incubation at 33°C, cells were washed and maintained in 100 µL of DMEM containing 1 µg/mL of TPCK trypsin at 33°C. The 100-µL media were collected every day and replaced with fresh DMEM containing 1 µg/mL of TPCK trypsin during 5-day experiment. Supernatants were used to determine viral titers (TCID50/mL) using a traditional infectivity assay. Immunofluorescence images of WT MDCK cells infected (1 MOI) with D/660 (E), D/CA (F), D/OK (G), and D/swine/KY (H) are shown. Immunofluorescence images of Neu5,9Ac2-fed CASD1-KO MDCK cells infected (1 MOI) with D/660 (I), D/CA (J), D/OK (K), and D/swine/KY (L) are also provided. Similarly, immunofluorescence images of Neu5Gc9Ac-fed CASD1-KO MDCK cells infected (1 MOI) with D/660 (M), D/CA (N), D/OK (O), and D/swine/KY (P) are also shown. CASD1-KO MDCK cells without exogenous receptor feeding were refractory to infections by D/660 (Q), D/CA (R), D/OK (S), and D/swine/KY (T) natural isolates. Viral titers are represented as mean of six individual replicates, and error bar represents the standard error of mean. Statistical significance is indicated by * (P < 0.05). Merged (fluorescence and DAPI) immunofluorescence images are representative of six independent replicates. Scale bar represents 50 µm.
To validate the above finding through qualitatively demonstrating the presence of viral proteins in infected CASD1-KO cells, 9-O-acetylated sialic acid-fed cells infected with four representative strains of IDV were assessed by immunofluorescence at day 3 following infection using the whole virus-specific rabbit polyclonal sera. Consistent with measured viral titer data, WT MDCK cells infected with D/660 (Fig. 5E), D/CA (Fig. 5F), D/OK (Fig. 5G), and D/swine/KY (Fig. 5H) showed higher abundance of viral proteins. Both Neu5,9Ac2- (Fig. 5I through L, respectively, for D/660, D/CA, D/OK, and D/swine/KY) and Neu5Gc9Ac-fed cells (Fig. 5M through P, respectively, for D/660, D/CA, D/OK, and D/swine/KY) also showed the presence of viral proteins in infected receptor-treated CASD1-KO cells. CASD1-KO cells were refractory to D/660 (Fig. 5Q), D/CA (Fig. 5R), D/OK (Fig. 5S), and D/swine/KY (Fig. 5T) viruses.
Influenza C virus, like IDV, can utilize both Neu5,9Ac2 and Neu5Gc9Ac receptors for infection
Influenza C virus, like IDV, is a seven-segmented influenza virus with the negative polarity genome, and ICV has HEF that can bind to both Neu5Gc9Ac and Neu5,9Ac2 (32). Using influenza C/Victoria/2/2012 isolated from human (40), we performed the multi-round replication assay for ICV Victoria strain in CASD1-KO cells with and without feeding with 200 µM of Neu5,9Ac2 or Neu5Gc9Ac. We found that ICV, like IDV, utilized both receptors (Fig. 6), and there were no statistical differences in viral titers between Neu5,9Ac2- and Neu5Gc9Ac-fed cells. Viral titers of ICV in Neu5,9Ac2- or Neu5Gc9Ac-fed cells were approximately two logs less than those observed in WT MDCK cells with a native CASD1 (Fig. 6).
Fig 6.
Replication kinetics of human ICV C/Victoria/2/2012 strain in receptor-treated CASD1-KO MDCK cells. CASD1-KO MDCK cells were fed with synthetic Neu5,9Ac2 or Neu5Gc9Ac (200 µM) for 3 days. Treated cells were then infected with 1 MOI of C/Victoria/2/2012. After 1 hour of incubation at 33°C, cells were washed and maintained in 100 µL of DMEM containing 1 µg/mL of TPCK trypsin at 33°C. The 100-µL media were collected every day and replaced with fresh DMEM containing 1 µg/mL of TPCK trypsin during a 5-day experiment. Supernatants were used to determine viral titers (TCID50/mL) using a traditional infectivity assay. Viral titers are represented as mean of six individual replicates, and error bar represents the standard error of mean.
DISCUSSION
IDV was first isolated from a diseased pig with influenza-like symptoms in 2011 (2) and was officially designated as new subtype of influenza by the International Committee of Taxonomy of Viruses in 2016 (3, 41). Since its initial isolation, IDV has been shown to infect a variety of farm animals. Although direct human infection has not been established, antibodies against IDV have been reported in both swine and cattle handlers (2, 42, 43). Viral genome has also been detected in farmers who were in close contact to swine and bovine species (11, 13). As such, IDV has the potential to emerge into human population and cause influenza-like respiratory disease. To date, little is known regarding which form of naturally occurring 9-O-acetylated sialic acids (Neu5,9Ac2 and Neu5Gc9Ac) serves as a functional receptor for IDV entry into target cells.
Sialic acid O-acetylation occurs in the Golgi (29). Two classes of enzymes regulate O-acetylation: the sialic acid O-acetyltransferases (SOATs) and the sialic acid esterase (28). The only known mammalian SOAT is CASD1 (44). CASD1 enzyme encoded by CASD1 gene is known to catalyze the addition of O-acetyl group to sialic acids (29). O-Acetylation can occur at C-4, C7, C-8, and C-9, and reversible migration may occur under neutral-to-mild basic pH conditions (45, 46). MDCK cells are the most common cell line used for influenza research (47). The cell line with the CASD1 gene that was knocked out using CRISPR/Cas9 was validated for the lack of 9-O-acetyl sialic acid expression by using 9-O-acetyl sialic acid-specific probes in a previous study (39). This work also revealed that the CASD1-KO MDCK cells were resistant to IDV infection, while WT MDCK showed high infection levels. By leveraging this MDCK CASD1-KO cell lacking sialic acid 9-O-acetylation activity in combination with the exogenous 9-O-acetyl sialic acids feeding experiments involving single- and multiple-round viral replication assays, we demonstrate that IDV can utilize cell surface receptors containing Neu5,9Ac2 or Neu5Gc9Ac as a functional receptor for infection.
In this study, we demonstrated that IDV RBD binds to both Neu5,9Ac2 and Neu5Gc9Ac on cell surface, which correlates with IDV replication in CASD1-KO cells receiving exogenous 9-O-acetyl sialic acid feeding. Treatment of cells with exogenous sialic acid to restore sialic acid levels in hyposialylated cells has been employed in many published studies (48–52). Several studies have shown that humans, who do not have the ability to convert CMP-Neu5Ac to CMP-Neu5Gc, can incorporate diet-derived Neu5Gc into cells (25, 49, 50, 53–55). Thus, metabolic incorporation of sialic acid can occur both in vitro and in vivo. More recent studies (48, 56) showed that under hyposialylated conditions, cells can uptake exogenous Neu5,9Ac2 and display it in the plasma membrane, as confirmed by 9-O-acetylated sialic acid-binding probes. External sialic acids, such as Neu5,9Ac2, can reach the maximum concentration at the cell surface probably at day 3 post-feeding (56). By harnessing this system, we showed that receptor feeding of Neu5,9Ac2 and/or Neu5Gc9Ac can successfully restore infectivity of IDV in CASD1-KO cells. In addition to revealing the receptor usage by IDV, results of these experiments also validated the specificity of CASD1 inactivation by CRISPR-Cas9 since other enzymatic activities, such as the sialic acid conjugation to the glycans of glycoproteins and glycolipids by sialyltransferases, are apparently unaffected in CASD1-KO cells. In the single-round replication assays, we observed that IDV reporter viruses replicated slightly more efficiently in Neu5,9Ac2-fed cells than in Neu5Gc9Ac-fed cells. This could be due to the preference of IDV for Neu5,9Ac2 over Neu5Gc9Ac or the differential uptake and turnover rates between Neu5,9Ac2 and Neu5Gc9Ac, which warrants further investigation.
In the multi-round replication assays, using two representative IDV strains isolated from bovine and swine, we showed that both bovine- and swine-originated IDV can utilize Neu5,9Ac2 and Neu5Gc9Ac for entry and infection. IDV of swine origin showed higher replication titers in Neu5,9Ac2-fed cells, while bovine origin IDV strains showed higher viral titers in Neu5Gc9Ac-fed cells. Similarly, human ICV replicated well in both Neu5,9Ac2- and Neu5Gc9Ac-fed cells. Viral titers were slightly higher in Neu5,9Ac2-fed cells, which is congruent to our previously reported ICV glycan microarray data and ICV replication inhibition result involving the synthetic receptor analogs (32).
Both swine and bovine species have a functional CMAH enzyme, so they can convert Neu5Ac to Neu5Gc. Since this conversion is unidirectional (27), in swine and bovine, there is a higher expression of Neu5Gc than Neu5Ac. For IAV, the differential expression of sialic acids across various species plays a crucial role in determining the species specificity of influenza viruses and their tissue tropisms. Dogs, ferrets, seals, hedgehogs, certain species of new world monkey, and white-tailed deer have non-functional CMAH genes (57, 58) and thus cannot convert Neu5Ac to Neu5Gc. A study quantifying Neu5Gc expression in the lungs of cattle and swine showed that cattle possess higher levels of Neu5Gc expression (59). In wild pigs, there is a higher expression of Neu5Gc than Neu5Ac in the upper respiratory tract, transitioning to higher Neu5Ac and lower Neu5Gc in the lower respiratory tract. In contrast to wild pigs, adult domestic pig lung tissues express a higher amount of Neu5Gc throughout the respiratory tract, including high Neu5Gc in pig saliva (60–62). Elucidating the delicate interplay between Sia diversity (Neu5Ac and Neu5Gc) and selection for influenza viruses with high replication fitness and cross-species transmission efficiency in various animal species clearly warrants further work.
To date, limited studies are available on the quantification of O-acetylation expression in animal species. Mouse trachea and lung tissues were found to express high level of 9-O-acetylated Sias, while human submucosal glands and lung tissues contain significant amounts of 7,9- and 9-O-Ac Sias (63). Interestingly, mouse sera also have higher Neu5Gc, while rat sera contain both Neu5Ac and Neu5,9Ac2 in significant amounts (64). 7,9- and 9-O-Ac Sias are also expressed abundantly in the tracheal epithelium of pigs, horses, and dogs. Ducks were found to express 9-O-acetylated Sias in trachea and lung alveolar tissues as well as 7,9- and 9-O-Ac Sias in the intestine tissue (63). Despite progress, in-depth quantitative analysis of sialic acid species with various forms of O-acetylation in the respiratory tissues of humans and animals is needed in future study toward better understanding of the impact of modified Sias on evolution, replication fitness, and transmission effectiveness of influenza and other respiratory viruses utilizing this group of Sias as functional receptors for infection.
Results of our experiments reported in this study showed a general trend, in that two bovine IDVs replicated slightly more efficiently in Neu5Gc9Ac-treated cells, while an opposite trajectory was observed for the two swine IDVs. Whether the genetic polymorphisms in the HEF protein between two groups confer this phenotype (probably through receptor-binding preference or entry/fusion steps post-receptor binding) will be investigated in future study. Both IDV and ICV utilize Neu5Gc9Ac and Neu5,9Ac2 as functional receptors for their entry. The only notable difference between Neu5Gc9Ac and Neu5,9Ac2 is that the former molecule has an extra oxygen in the N-acyl group at the C-5 position. Our study indicates that this extra atom has no impact on the IDV binding to the 9-O-acetyl group in Neu5Gc9Ac. Thus, exploiting natural diversity of sialic acids with 9-O-acetylation may give the IDV or ICV evolutionary advantage to infect multiple host species, which has been already demonstrated in field study. 9-O-Acetylated Neu5Ac is ubiquitously expressed throughout the human body (28). In addition to the respiratory tract, including lung, Neu5,9Ac2 has been also found in brain, salivary gland, kidney, pancreas, circulation, and intestine human tissues (63). Furthermore, a growing number of studies have shown that Neu5,9Ac2 expression is significantly upregulated in patients with cancer, autoimmunity, and infection (65, 66). Based on the evidence presented in this study establishing that Neu5,9Ac2 serves as a functional receptor for IDV entry and infection, the potential emergence of IDV in humans, especially with underlying conditions, cannot be underestimated, which highlights a need for continued surveillance of IDV in farm animals and humans, as well as for further investigation of its biology and cross-species transmission mechanism.
ACKNOWLEDGMENTS
We acknowledge Dr. Ajit Varki (UCSD) and his group for valuable insights regarding the sialic acid feeding experiment. We also thank Dr. Brian Wasik (Cornell University) for his help in data interpretation concerning the CMAH functionality among different animal species.
This study was supported by NIH R01AI141889. The authors also thank the support provided by the William Robert Mills Chair Endowment Fund and Agricultural Experiment Station of the University of Kentucky.
The members of the Wang-Li lab are particularly appreciated for their great help in the proofreading of the manuscript and suggestions for experiments described in this study.
Contributor Information
Feng Li, Email: Feng.Li@uky.edu.
Martin Schwemmle, University Medical Center Freiburg, Freiburg, Germany.
REFERENCES
- 1. Krammer F, Smith GJD, Fouchier RAM, Peiris M, Kedzierska K, Doherty PC, Palese P, Shaw ML, Treanor J, Webster RG, García-Sastre A. 2018. Influenza. Nat Rev Dis Primers 4:3. doi: 10.1038/s41572-018-0002-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Hause BM, Ducatez M, Collin EA, Ran Z, Liu R, Sheng Z, Armien A, Kaplan B, Chakravarty S, Hoppe AD, Webby RJ, Simonson RR, Li F. 2013. Isolation of a novel swine influenza virus from Oklahoma in 2011 which is distantly related to human influenza C viruses. PLoS Pathog 9:e1003176. doi: 10.1371/journal.ppat.1003176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Hause BM, Collin EA, Liu R, Huang B, Sheng Z, Lu W, Wang D, Nelson EA, Li F. 2014. Characterization of a novel influenza virus in cattle and swine: proposal for a new genus in the Orthomyxoviridae family. mBio 5:e00031-14. doi: 10.1128/mBio.00031-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Murakami S, Odagiri T, Melaku SK, Bazartseren B, Ishida H, Takenaka-Uema A, Muraki Y, Sentsui H, Horimoto T. 2019. Influenza D virus infection in dromedary camels, Ethiopia. Emerg Infect Dis 25:1224–1226. doi: 10.3201/eid2506.181158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Salem E, Cook EAJ, Lbacha HA, Oliva J, Awoume F, Aplogan GL, Hymann EC, Muloi D, Deem SL, Alali S, Zouagui Z, Fèvre EM, Meyer G, Ducatez MF. 2017. Serologic evidence for influenza C and D virus among ruminants and camelids, Africa, 1991-2015. Emerg Infect Dis 23:1556–1559. doi: 10.3201/eid2309.170342 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Oliva J, Eichenbaum A, Belin J, Gaudino M, Guillotin J, Alzieu J-P, Nicollet P, Brugidou R, Gueneau E, Michel E, Meyer G, Ducatez MF. 2019. Serological evidence of influenza D virus circulation among cattle and small ruminants in France. Viruses 11:516. doi: 10.3390/v11060516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Quast M, Sreenivasan C, Sexton G, Nedland H, Singrey A, Fawcett L, Miller G, Lauer D, Voss S, Pollock S, Cunha CW, Christopher-Hennings J, Nelson E, Li F. 2015. Serological evidence for the presence of influenza D virus in small ruminants. Vet Microbiol 180:281–285. doi: 10.1016/j.vetmic.2015.09.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Nedland H, Wollman J, Sreenivasan C, Quast M, Singrey A, Fawcett L, Christopher-Hennings J, Nelson E, Kaushik RS, Wang D, Li F. 2018. Serological evidence for the co-circulation of two lineages of influenza D viruses in equine populations of the Midwest United States. Zoonoses Public Health 65:e148–e154. doi: 10.1111/zph.12423 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Gorin S, Fablet C, Quéguiner S, Barbier N, Paboeuf F, Hervé S, Rose N, Simon G. 2019. Assessment of influenza D virus in domestic pigs and wild boars in France: apparent limited spread within swine populations despite serological evidence of breeding sow exposure. Viruses 12:25. doi: 10.3390/v12010025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. White SK, Ma W, McDaniel CJ, Gray GC, Lednicky JA. 2016. Serologic evidence of exposure to influenza D virus among persons with occupational contact with cattle. J Clin Virol 81:31–33. doi: 10.1016/j.jcv.2016.05.017 [DOI] [PubMed] [Google Scholar]
- 11. Leibler JH, Abdelgadir A, Seidel J, White RF, Johnson WE, Reynolds SJ, Gray GC, Schaeffer JW. 2023. Influenza D virus exposure among US cattle workers: a call for surveillance. Zoonoses Public Health 70:166–170. doi: 10.1111/zph.13008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Trombetta CM. 2020. Influenza D virus: a potential threat for humans? Eur J Public Health 30:Supplement_5. doi: 10.1093/eurpub/ckaa165.1097 [DOI] [Google Scholar]
- 13. Borkenhagen LK, Mallinson KA, Tsao RW, Ha S-J, Lim W-H, Toh T-H, Anderson BD, Fieldhouse JK, Philo SE, Chong K-S, Lindsley WG, Ramirez A, Lowe JF, Coleman KK, Gray GC. 2018. Surveillance for respiratory and diarrheal pathogens at the human-pig interface in Sarawak, Malaysia. PLoS One 13:e0201295. doi: 10.1371/journal.pone.0201295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Bailey ES, Choi JY, Zemke J, Yondon M, Gray GC. 2018. Molecular surveillance of respiratory viruses with bioaerosol sampling in an airport. Trop Dis Travel Med Vaccines 4:11. doi: 10.1186/s40794-018-0071-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Choi JY, Zemke J, Philo SE, Bailey ES, Yondon M, Gray GC. 2018. Aerosol sampling in a hospital emergency room setting: a complementary surveillance method for the detection of respiratory viruses. Front Public Health 6:174. doi: 10.3389/fpubh.2018.00174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Ferguson L, Olivier AK, Genova S, Epperson WB, Smith DR, Schneider L, Barton K, McCuan K, Webby RJ, Wan X-F. 2016. Pathogenesis of influenza D virus in cattle. J Virol 90:5636–5642. doi: 10.1128/JVI.03122-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Sreenivasan C, Thomas M, Sheng Z, Hause BM, Collin EA, Knudsen DEB, Pillatzki A, Nelson E, Wang D, Kaushik RS, Li F. 2015. Replication and transmission of the novel bovine influenza D virus in a Guinea pig model. J Virol 89:11990–12001. doi: 10.1128/JVI.01630-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Oliva J, Mettier J, Sedano L, Delverdier M, Bourgès-Abella N, Hause B, Loupias J, Pardo I, Bleuart C, Bordignon PJ, Meunier E, Le Goffic R, Meyer G, Ducatez MF. 2020. Murine model for the study of influenza D virus. J Virol 94:e01662-19. doi: 10.1128/JVI.01662-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Vega-Rodriguez W, Ly H. 2023. Epidemiological, serological, and genetic evidence of influenza D virus infection in humans: is it a justifiable cause for concern? Virulence 14:2150443. doi: 10.1080/21505594.2022.2150443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Matrosovich M, Herrler G, Klenk HD. 2015. Sialic acid receptors of viruses. Top Curr Chem 367:1–28. doi: 10.1007/128_2013_466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Varki A. 2008. Sialic acids in human health and disease. Trends Mol Med 14:351–360. doi: 10.1016/j.molmed.2008.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Chen X, Varki A. 2010. Advances in the biology and chemistry of sialic acids. ACS Chem Biol 5:163–176. doi: 10.1021/cb900266r [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Varki NM, Varki A. 2007. Diversity in cell surface sialic acid presentations: implications for biology and disease. Lab Invest 87:851–857. doi: 10.1038/labinvest.3700656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Kooner AS, Yu H, Chen X. 2019. Synthesis of N-glycolylneuraminic acid (Neu5Gc) and its glycosides. Front Immunol 10:2004. doi: 10.3389/fimmu.2019.02004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Samraj AN, Pearce OMT, Läubli H, Crittenden AN, Bergfeld AK, Banda K, Gregg CJ, Bingman AE, Secrest P, Diaz SL, Varki NM, Varki A. 2015. A red meat-derived glycan promotes inflammation and cancer progression. Proc Natl Acad Sci U S A 112:542–547. doi: 10.1073/pnas.1417508112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Uno Y, Kawakami S, Ochiai K, Omi T. 2019. Molecular characterization of cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH) associated with the erythrocyte antigens in dogs. Canine Genet Epidemiol 6:9. doi: 10.1186/s40575-019-0076-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Kawanishi K, Dhar C, Do R, Varki N, Gordts P, Varki A. 2019. Human species-specific loss of CMP-N-acetylneuraminic acid hydroxylase enhances atherosclerosis via intrinsic and extrinsic mechanisms. Proc Natl Acad Sci U S A 116:16036–16045. doi: 10.1073/pnas.1902902116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Visser EA, Moons SJ, Timmermans S, de Jong H, Boltje TJ, Büll C. 2021. Sialic acid O-acetylation: from biosynthesis to roles in health and disease. J Biol Chem 297:100906. doi: 10.1016/j.jbc.2021.100906 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Baumann A-M, Bakkers MJG, Buettner FFR, Hartmann M, Grove M, Langereis MA, de Groot RJ, Mühlenhoff M. 2015. 9-O-acetylation of sialic acids is catalysed by CASD1 via a covalent acetyl-enzyme intermediate. Nat Commun 6:7673. doi: 10.1038/ncomms8673 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Rogers GN, Herrler G, Paulson JC, Klenk HD. 1986. Influenza C virus uses 9-O-acetyl-N-acetylneuraminic acid as a high affinity receptor determinant for attachment to cells. J Biol Chem 261:5947–5951. [PubMed] [Google Scholar]
- 31. Long JS, Mistry B, Haslam SM, Barclay WS. 2019. Host and viral determinants of influenza A virus species specificity. Nat Rev Microbiol 17:67–81. doi: 10.1038/s41579-018-0115-z [DOI] [PubMed] [Google Scholar]
- 32. Liu R, Sreenivasan C, Yu H, Sheng Z, Newkirk SJ, An W, Smith DF, Chen X, Wang D, Li F. 2020. Influenza D virus diverges from its related influenza C virus in the recognition of 9-O-acetylated N-acetyl- or N-glycolyl-neuraminic acid-containing glycan receptors. Virology 545:16–23. doi: 10.1016/j.virol.2020.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Chien Y-A, Alford BK, Wasik BR, Weichert WS, Parrish CR, Daniel S. 2023. Single particle analysis of H3N2 influenza entry differentiates the impact of the sialic acids (Neu5Ac and Neu5Gc) on virus binding and membrane fusion. J Virol 97:e0146322. doi: 10.1128/jvi.01463-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Broszeit F, Tzarum N, Zhu X, Nemanichvili N, Eggink D, Leenders T, Li Z, Liu L, Wolfert MA, Papanikolaou A, Martínez-Romero C, Gagarinov IA, Yu W, García-Sastre A, Wennekes T, Okamatsu M, Verheije MH, Wilson IA, Boons G-J, de Vries RP. 2019. N-glycolylneuraminic acid as a receptor for influenza A viruses. Cell Rep 27:3284–3294. doi: 10.1016/j.celrep.2019.05.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Ogura H, Furuhata K, Sato S, Anazawa K, Itoh M, Shitori Y. 1987. Synthesis of 9-O-acyl- and 4-O-acetyl-sialic acids. Carbohydr Res 167:77–86. doi: 10.1016/0008-6215(87)80269-0 [DOI] [PubMed] [Google Scholar]
- 36. Yu H, Cheng J, Ding L, Khedri Z, Chen Y, Chin S, Lau K, Tiwari VK, Chen X. 2009. Chemoenzymatic synthesis of GD3 oligosaccharides and other disialyl glycans containing natural and non-natural sialic acids. J Am Chem Soc 131:18467–18477. doi: 10.1021/ja907750r [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Reed LJ, Muench H. 1938. A simple method of estimating fifty per cent endpoints. Am J Epidemiol 27:493–497. doi: 10.1093/oxfordjournals.aje.a118408 [DOI] [Google Scholar]
- 38. Yu J, Liu R, Zhou B, Chou T-W, Ghedin E, Sheng Z, Gao R, Zhai S-L, Wang D, Li F. 2019. Development and characterization of a reverse-genetics system for influenza D virus. J Virol 93:e01186-19. doi: 10.1128/JVI.01186-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Barnard KN, Wasik BR, LaClair JR, Buchholz DW, Weichert WS, Alford-Lawrence BK, Aguilar HC, Parrish CR. 2019. Expression of 9-O- and 7,9-O-acetyl modified sialic acid in cells and their effects on influenza viruses. mBio 10:e02490-19. doi: 10.1128/mBio.02490-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Liu R, Sheng Z, Lin T, Sreenivasan C, Gao R, Thomas M, Druce J, Hause BM, Kaushik RS, Li F, Wang D. 2020. Genetic and antigenic characteristics of a human influenza C virus clinical isolate. J Med Virol 92:161–166. doi: 10.1002/jmv.25589 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Yu J, Li F, Wang D. 2021. The first decade of research advances in influenza D virus. J Gen Virol 102:jgv001529. doi: 10.1099/jgv.0.001529 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Trombetta CM, Marchi S, Manini I, Kistner O, Li F, Piu P, Manenti A, Biuso F, Sreenivasan C, Druce J, Montomoli E. 2020. Influenza D virus: serological evidence in the Italian population from 2005 to 2017. Viruses 12:30. doi: 10.3390/v12010030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Trombetta CM, Montomoli E, Di Bartolo I, Ostanello F, Chiapponi C, Marchi S. 2022. Detection of antibodies against influenza D virus in swine veterinarians in Italy in 2004. J Med Virol 94:2855–2859. doi: 10.1002/jmv.27466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Arming S, Wipfler D, Mayr J, Merling A, Vilas U, Schauer R, Schwartz-Albiez R, Vlasak R. 2011. The human Cas1 protein: a sialic acid-specific O-acetyltransferase? Glycobiology 21:553–564. doi: 10.1093/glycob/cwq153 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Kamerling JP, Schauer R, Shukla AK, Stoll S, Van Halbeek H, Vliegenthart JF. 1987. Migration of O-acetyl groups in N,O-acetylneuraminic acids. Eur J Biochem 162:601–607. doi: 10.1111/j.1432-1033.1987.tb10681.x [DOI] [PubMed] [Google Scholar]
- 46. Ji Y, Sasmal A, Li W, Oh L, Srivastava S, Hargett AA, Wasik BR, Yu H, Diaz S, Choudhury B, Parrish CR, Freedberg DI, Wang L-P, Varki A, Chen X. 2021. Reversible O-acetyl migration within the sialic acid side chain and its influence on protein recognition. ACS Chem Biol 16:1951–1960. doi: 10.1021/acschembio.0c00998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Tsai H-C, Lehman CW, Lin C-C, Tsai S-W, Chen C-M. 2019. Functional evaluation for adequacy of MDCK-lineage cells in influenza research. BMC Res Notes 12:101. doi: 10.1186/s13104-019-4134-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Oetke C, Brossmer R, Mantey LR, Hinderlich S, Isecke R, Reutter W, Keppler OT, Pawlita M. 2002. Versatile biosynthetic engineering of sialic acid in living cells using synthetic sialic acid analogues. J Biol Chem 277:6688–6695. doi: 10.1074/jbc.M109973200 [DOI] [PubMed] [Google Scholar]
- 49. Bardor M, Nguyen DH, Diaz S, Varki A. 2005. Mechanism of uptake and incorporation of the non-human sialic acid N-glycolylneuraminic acid into human cells. J Biol Chem 280:4228–4237. doi: 10.1074/jbc.M412040200 [DOI] [PubMed] [Google Scholar]
- 50. Tangvoranuntakul P, Gagneux P, Diaz S, Bardor M, Varki N, Varki A, Muchmore E. 2003. Human uptake and incorporation of an immunogenic nonhuman dietary sialic acid. Proc Natl Acad Sci U S A 100:12045–12050. doi: 10.1073/pnas.2131556100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Luchansky SJ, Bertozzi CR. 2004. Azido sialic acids can modulate cell-surface interactions. Chembiochem 5:1706–1709. doi: 10.1002/cbic.200400148 [DOI] [PubMed] [Google Scholar]
- 52. Cheng B, Dong L, Zhu Y, Huang R, Sun Y, You Q, Song Q, Paton JC, Paton AW, Chen X. 2019. 9-azido analogues of three sialic acid forms for metabolic remodeling of cell-surface sialoglycans. ACS Chem Biol 14:2141–2147. doi: 10.1021/acschembio.9b00556 [DOI] [PubMed] [Google Scholar]
- 53. Soulillou JP, Cozzi E, Bach JM. 2020. Challenging the role of diet-induced anti-Neu5Gc antibodies in human pathologies. Front Immunol 11:834. doi: 10.3389/fimmu.2020.00834 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Kawanishi K, Coker JK, Grunddal KV, Dhar C, Hsiao J, Zengler K, Varki N, Varki A, Gordts P. 2021. Dietary Neu5Ac intervention protects against atherosclerosis associated with human-like Neu5Gc loss-brief report. Arterioscler Thromb Vasc Biol 41:2730–2739. doi: 10.1161/ATVBAHA.120.315280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Bashir S, Fezeu LK, Leviatan Ben-Arye S, Yehuda S, Reuven EM, Szabo de Edelenyi F, Fellah-Hebia I, Le Tourneau T, Imbert-Marcille BM, Drouet EB, Touvier M, Roussel J-C, Yu H, Chen X, Hercberg S, Cozzi E, Soulillou J-P, Galan P, Padler-Karavani V. 2020. Association between Neu5Gc carbohydrate and serum antibodies against it provides the molecular link to cancer: French NutriNet-sante study. BMC Med 18:262. doi: 10.1186/s12916-020-01721-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Khedri Z, Xiao A, Yu H, Landig CS, Li W, Diaz S, Wasik BR, Parrish CR, Wang L-P, Varki A, Chen X. 2017. A chemical biology solution to problems with studying biologically important but unstable 9-O-acetyl sialic acids. ACS Chem Biol 12:214–224. doi: 10.1021/acschembio.6b00928 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Nemanichvili N, Spruit CM, Berends AJ, Gröne A, Rijks JM, Verheije MH, de Vries RP. 2022. Wild and domestic animals variably display Neu5Ac and Neu5Gc sialic acids. Glycobiology 32:791–802. doi: 10.1093/glycob/cwac033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Springer SA, Diaz SL, Gagneux P. 2014. Parallel evolution of a self-signal: humans and new world monkeys independently lost the cell surface sugar Neu5Gc. Immunogenetics 66:671–674. doi: 10.1007/s00251-014-0795-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Jahan M, Thomson PC, Wynn PC, Wang B. 2021. The non-human glycan, N-glycolylneuraminic acid (Neu5Gc), is not expressed in all organs and skeletal muscles of nine animal species. Food Chem 343:128439. doi: 10.1016/j.foodchem.2020.128439 [DOI] [PubMed] [Google Scholar]
- 60. Ji S, Wang F, Chen Y, Yang C, Zhang P, Zhang X, Troy FA, Wang B. 2017. Developmental changes in the level of free and conjugated sialic acids, Neu5Ac, Neu5Gc and KDN in different organs of pig: a LC-MS/MS quantitative analyses. Glycoconj J 34:21–30. doi: 10.1007/s10719-016-9724-9 [DOI] [PubMed] [Google Scholar]
- 61. Byrd-Leotis L, Liu R, Bradley KC, Lasanajak Y, Cummings SF, Song X, Heimburg-Molinaro J, Galloway SE, Culhane MR, Smith DF, Steinhauer DA, Cummings RD. 2014. Shotgun glycomics of pig lung identifies natural endogenous receptors for influenza viruses. Proc Natl Acad Sci U S A 111:E2241–E2250. doi: 10.1073/pnas.1323162111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Barnard KN, Alford-Lawrence BK, Buchholz DW, Wasik BR, LaClair JR, Yu H, Honce R, Ruhl S, Pajic P, Daugherity EK, Chen X, Schultz-Cherry SL, Aguilar HC, Varki A, Parrish CR. 2020. Modified sialic acids on mucus and erythrocytes inhibit influenza A virus hemagglutinin and neuraminidase functions. J Virol 94:e01567-19. doi: 10.1128/JVI.01567-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Wasik BR, Barnard KN, Ossiboff RJ, Khedri Z, Feng KH, Yu H, Chen X, Perez DR, Varki A, Parrish CR. 2017. Distribution of O-acetylated sialic acids among target host tissues for influenza virus. mSphere 2:e00379-16. doi: 10.1128/mSphere.00379-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Morimoto N, Nakano M, Kinoshita M, Kawabata A, Morita M, Oda Y, Kuroda R, Kakehi K. 2001. Specific distribution of sialic acids in animal tissues as examined by LC-ESI-MS after derivatization with 1,2-diamino-4,5-methylenedioxybenzene. Anal Chem 73:5422–5428. doi: 10.1021/ac0104328 [DOI] [PubMed] [Google Scholar]
- 65. Mandal C, Srinivasan GV, Chowdhury S, Chandra S, Mandal C, Schauer R, Mandal C. 2009. High level of sialate-O-acetyltransferase activity in lymphoblasts of childhood acute lymphoblastic leukaemia (ALL): enzyme characterization and correlation with disease status. Glycoconj J 26:57–73. doi: 10.1007/s10719-008-9163-3 [DOI] [PubMed] [Google Scholar]
- 66. Mandal C, Mandal C, Chandra S, Schauer R, Mandal C. 2012. Regulation of O-acetylation of sialic acids by sialate-O-acetyltransferase and sialate-O-acetylesterase activities in childhood acute lymphoblastic leukemia. Glycobiology 22:70–83. doi: 10.1093/glycob/cwr106 [DOI] [PubMed] [Google Scholar]






