Skip to main content

PERSPECTIVE article

Front. Cell. Infect. Microbiol., 23 September 2022
Sec. Virus and Host
This article is part of the Research Topic Lectins from plants, algae, fungi, bacteria and animals as potential therapeutic tools for SARS-CoV-2 and other pathogenic enveloped viruses, in a “one-health” perspective View all 5 articles

Engineering recombinantly expressed lectin-based antiviral agents

  • Department of Environmental Health Sciences, University of California Los Angeles, CA, Los Angeles, United States

Cyanovirin-N (CV-N), a lectin from Nostoc ellipsosporum was found an infusion inhibitory protein for human immunodeficiency virus (HIV)-1. A tandem-repeat of the engineered domain-swapped dimer bound specific sites at hemagglutinin (HA), Ebola and HIV spike glycoproteins as well as dimannosylated HA peptide, N-acetyl-D-glucosamine and high-mannose containing oligosaccharides. Among these, CV-N bound the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike protein at a dissociation constant (KD) of 18.6 µM (and KD=260 µM to RBD), which was low-affinity carbohydrate-binding as compared with the recognition of the other viral spikes. Binding of dimannosylated peptide to homo-dimeric CVN2 and variants of CVN2 that were pairing Glu-Arg residues sterically located close to its high-affinity carbohydrate binding sites, was measured using surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC). Binding affinity increased with polar interactions, when the mutated residues were used to substitute a single, or two disulfide bonds, in CVN2. Site-specific N-linked glycans on spikes were mediating the infection with influenza virus by broadly neutralizing antibodies to HA and lectin binding to HA was further investigated via modes of saturation transfer difference (STD)-NMR. Our findings showed that stoichiometry and the lectin’s binding affinity were revealed by an interaction of CVN2 with dimannose units and either the high- or low-affinity binding site. To understand how these binding mechanisms add to viral membrane fusion we compare our tested HA-derived peptides in affinity with SARS-CoV-2 glycoprotein and review lectins and their mechanisms of binding to enveloped viruses for a potential use to simulate neutralization ability.

Introduction

Cyanobacterial lectin CV-N displayed antiviral activity, which is mediated by nanomolar binding to high-mannose oligosaccharide modifications on envelope spike proteins (Mazur-Marzec et al., 2021), against human immunodeficiency virus 1 (HIV-1) (Boyd, 1997; Bolmstedt et al., 2001), influenza (O’Keefe et al., 2003), herpes virus, hepatitis C, severe acute respiratory syndrome coronavirus (SARS-CoV)-2 (Naidoo et al., 2021; Li et al., 2022), among others, and binds Ebola virus (Jensen et al., 2014). Structural analyses and binding affinity assays indicated cross-linking of two high- or low-affinity carbohydrate binding sites in a domain-swapped CVN2 dimer (Boyd, 1997; Bewley et al., 2002; Barrientos et al., 2006) by bivalent binding in the micromolar range to enhance avidity to viral envelope glycoproteins (Schilling et al., 2020) and to inhibit viral entry (Boyd, 1997; O’Keefe et al., 2003; Barrientos et al., 2006). Selective binding of the terminal-accessible disaccharide Manα1-2Manα on oligomannose-8 D1D3 arms and oligomannose-9 was comprised by two binding sites of differing affinities located on opposite protein protomers, thereby reaching nanomolar binding affinities (Bewley, 2001; Bewley and Otero-Quintero, 2001). Thus, CVN2 is considered a pseudo-antibody concerning its application to bind epitopes on HIV gp120 similar to virus-neutralizing antibodies (Shenoy et al., 2002; Keeffe et al., 2011).

Lectins from natural sources function as viral entry inhibitors

A class of cyanometabolites that exhibit antiviral effects were focused on lectins and polysaccharides (Mazur-Marzec et al., 2021), and specifically Griffithsin (GRFT), a red-alga-derived lectin showed broad binding to enveloped viruses in combination with sulfated polysaccharides against SARS-CoV-1 and 2 (Alam et al., 2021; Alsaidi et al., 2021). The use of other carbohydrate-binding agents, such as lectins from plants, fungi, and prokaryotes (Gupta et al., 2020), or anti-HIV antibiotics Pradimicin A (Tanabe-Tochikura et al., 1990) was limited due to unfavorable responses like immunogenicity, mitogenicity, hemagglutination, inflammatory activity, and cellular toxicity. Most of them showed antiviral activities against coronavirus though (Gupta et al., 2020). Wheat germ agglutinin (WGA) and lentil lectin were evaluated for antiviral efficacy through direct binding to SARS-CoV-2 Spike (S) protein (Auth et al., 2021; Wang et al., 2021) and neutralization activity against SARS-CoV-2 and its major variants of concern (Alpha, Beta) (Auth et al., 2021), and lentil lectin blocked binding to angiotensin-converting enzyme 2 (ACE2) receptor (Mazur-Marzec et al., 2021; Wang et al., 2021). The Lens culinaris-derived lentil lectin showed broader activity, weak hemagglutination activity at 1 mg/mL and no cytotoxicity activity, and no weight loss was found in the single injection mouse experiment (Wang et al., 2021). To systematically identify lectins that bind to the trimeric S protein and receptor-binding domain (RBD) of SARS-CoV-2, researchers searched for all annotated carbohydrate recognition domains (CRDs) of mouse C-type lectins, galectins and sialic acid-binding immunoglobulin-type lectins (Siglecs). Of 168 annotated CRDs, 143 lectin-CRDs were expressed as IgG2a-Fc fusion proteins from human HEK293F cells and two lectins, Clec4g and CD209c, were then selected (Hoffmann et al., 2021) that, like DC/L-SIGN, were evaluated to bind S protein of SARS-CoV-2 at the RBD with ACE2 interfering interaction or sterically blocking the receptor binding (Lempp et al., 2021; Thépaut et al., 2021).

Structural features for targeting glycoproteins

Surface-expressed membrane glycoproteins hemagglutinin (HA) and neuraminidase (NA) on influenza A virus are known for tetherin antagonism in a strain-specific manner (Gnirss et al., 2015), as they facilitate recognition of host receptor binding sites (Knossow and Skehel, 2006; Schmidt et al., 2015). HA binding is reported for broadly neutralizing antibodies, lectins, and also specific antibodies with highly variant antigenic sites (Knossow and Skehel, 2006; Schmidt et al., 2015; Wu and Wilson, 2017; Maier et al., 2021), but conserved epitopes on the homotrimeric membrane glycoprotein at the globular head domain (Bizebard et al., 1995; Fleury et al., 1998; Fleury et al., 1999; Krause et al., 2011; Benjamin et al., 2014; Schmidt et al., 2015; Nogales et al., 2018; Qiu et al., 2020) and stem region (Ekiert et al., 2009; Corti et al., 2011; Ekiert et al., 2011; Lee et al., 2012; Nachbagauer et al., 2014; Chen et al., 2021).

Current vaccines are effective but strain specific due to their focus on the immunodominant globular head domain of the HA (Krause et al., 2011; Schmidt et al., 2015; Raymond et al., 2018; Liao et al., 2020; Qiu et al., 2020). Contrarily, non-neutralizing antibodies destabilized the HA stem region, resulting in antibody-dependent enhancement of influenza disease, and enhanced virus fusion kinetics and manifestation of the respiratory disease in preclinical studies by treatment with two monoclonal antibodies (mAbs) following H3N2 viral challenge (Winarski et al., 2019). Structural features allowed mapping of single amino-acid mutations on HA (HA1 and HA2; where both are linked by disulfide bridges) that increase resistance to broad antibodies to H1 strains and show escape from antibody neutralization. Those antibodies targeting the H1 HA stalk [FI6v3 (Corti et al., 2011) and C179 (Dreyfus et al., 2013)] were broader, harder to escape, but less mutationally tolerating than other antibodies which targeted the head domain (Doud et al., 2018; Chen et al., 2021). Several studies showed evidence that it is possible to select antigenic mutants with broad antibodies, demonstrating that these epitopes are not entirely resistant to change (Doud et al., 2018; Wu et al., 2020a). More stem-specific antibodies were directed against a chimeric mono-glycosylated HA vaccine comprised of consensus sequences of avian H5 and H1 strains. The monosaccharide N-acetylglucosamine (GlcNAc) at the HA glycosite was attributed to better neutralization and cross-protection against H1, H3, H5, and H7 strains and subtypes, and overall vaccine efficacy was increased when the recombinant HA antigen-based vaccine was combined with a glycolipid adjuvant (Liao et al., 2020). Interestingly, N-linked glycans on the H5 antigen globular head domain and glycan-unmasking at the stem region elicit broad neutralizing antibodies to cross-protect against various H5N1 clades of virus infection (Chen et al., 2021); while the mechanism of introducing additional N-glycosylation sites was recently also applied to the modification of SARS-CoV-2 S glycoprotein (Galili, 2020; Lin et al., 2021). Glycans found on the SARS-CoV-2 S trimer (Watanabe et al., 2020) revealed few, but invariant interactions with human neutralizing antibodies to SARS-CoV-2 WT, the Alpha, Delta, Lambda, and Omicron variants (Wang et al., 2022), from which many were isolated, investigated during the current pandemics (Robbiani et al., 2020; Barnes et al., 2020a) and classified according to their binding capacities for epitopes on S trimer. The related “up” conformations of RBD (Lv et al., 2020; Pinto et al., 2020; Wu et al., 2020b), and down conformation of the RBD of the closed, prefusion S trimer (Zost et al., 2020a; Barnes et al., 2020b) were addressed. Around 2191 structures of SARS-CoV-2 S protein were published in the protein data bank until January 2022 and glycosylation varied according to the method of analyses used for structural classification of correlates, as well as immune pressure (Cao et al., 2020; Ju et al., 2020; Kreer et al., 2020; Shi et al., 2020; Walls et al., 2020a; Walls et al., 2020b; Wu et al., 2020c; Yu et al., 2020; Zost et al., 2020b). A comparison of CV-N binding to HA, HA top and RBD on human 2019-nCoV (Wuhan-Hu-1-2019 novel coronavirus) was examined (Maier, 2022). Using cryo-electron microscopy and binding assays, Pinto and coworkers described a mAb S309 and a class 3 mAb, that potentially recognized an epitope and glycan (N343 on SARS-CoV-2) that was found to be conserved within Sarbecovirus subgenus, without competing with receptor attachment (Pinto et al., 2020). The deletion of the glycosylation site at N165 on S protein by mutation N165A induced up-state to RBD. The solvent released N234-glycans also had direct contacts with the up-RBD in the WT and G614 S trimers, the state which is favored upon binding to ACE2 (Wang et al., 2022).

Mechanisms of cyanovirin-N binding to viral spikes

Influenza A antigen binding (H3N2) (Benjamin et al., 2014; Zost et al., 2017; Qiu et al., 2020) to one high-affinity binding site in CV-N, or two high-affinity binding sites in covalently linked dimeric CVN2 was determined to have equilibrium dissociation constants (KD)=5.7 nM and KD=2.7 nM, respectively. Selected immunoglobulin classes addressed specific and invariant structural patterns, however, which provided a substrate for affinity maturation in the membrane-anchored HA regions (Lingwood et al., 2012; Otterstrom et al., 2014). Our knowledge is incomplete on the variety and localization of epitopes on the stem of HA1 and HA2 that both involve epitope structures for glycan-targeting by highly neutralizing antibodies as compared with lectin binding (Ekiert et al., 2009).

To investigate the binding of homogenously glycosylated targets on HA, we chemically synthesized di- and tri-mannose moieties via azido glycosylation (Salunke et al., 2011), therefore facilitating site-specific interactions with CV-N derivatives. Whereas the glycan shield on the membrane-distal HA top part induced high-affinity binding to CV-N, CVN2 binding to HA close to a stabilizing disulfide bridge of HA top [4 N-linked glycans at N54, N97, N181, N301 (Bizebard et al., 1995)] has further been observed at its low-affinity sites. HA binding to at least one high-affinity carbohydrate binding site (H) of CVN2 has been examined at KD of 275 nM (A/Wisconsin/67/05) (Schilling et al., 2020), and Ebola GP1,2 bound to 2H of CVN2 with affinities in the lower nanomolar range (KD=26 nM) as measured via surface plasmon resonance (SPR) (Maier et al., 2021). Higher density of glycans on HA protein achieved binding with polar Glu-Arg residues instead of cystines in CVN2L0, and an association between respective mannosylated peptides (KD=10 µM for dimannosylated HA peptide, DM) and CVN2L0 or mutated Glu-Arg pairing near the HA binding pockets (Schilling et al., 2020). Testing H on domains B, which was impacted by replacing a disulfide bond into ionic residues, and domain A forming the low-affinity carbohydrate binding site (L), the dimeric domain-swapped CVN2L0 molecule showed binding to HA top region and the whole molecule. Binding affinity was presented as SPR KD values: A variant with altered binding-affinity was expressed in E. coli, purified, and positively tested for binding to HA-protein (H3N2) and DM, and showed a conformational change upon binding HA with either H or L carbohydrate binding sites and KD1= 49 nM and KD2 = 8 µM (Schilling et al., 2020; Maier et al., 2021). Disulfide bond variants were created by substitution of Cys and insertion of polar residue pairs Glu – Arg with slightly decreasing thermal stability and helical structures in V4 and V5. As the number of disulfide bonds in vicinity to the glycan-targeting pocket in CVN2 decreased from 4 (CVN2L0) to 2 (variants V3-5), binding affinity to HA protein decreased (Schilling et al., 2020).

Next, we were also interested in investigating the potential binding of CV-N to SARS-CoV-2 spikes via its RBD and CVN2 to RBD, assuming specific targeting of possibly conserved carbohydrates on RBD S1 subunit (Pinto et al., 2020; Barnes et al., 2020a). SARS-CoV-2 glycosylation of S trimer was characterized by 14 N-linked complex, 6 N-linked hybrid-type, 2 high-mannose glycosylation sites, and O-linked serine-attached and O-linked threonine glycosylation sites (Pinto et al., 2020; Watanabe et al., 2020). The monomeric mutant CVN-E41A was suspected to destabilize pseudo-domain B or to interrupt connectivity with the second domain A (Maier, 2022), such as found in CV-N WT (Yang et al., 1999). CVN-E41A monomer revealed enhanced protein stability upon binding to S protein, although this mutation site abolished a contacting residue between H and L, and reduced strength of molecular binding to GlcNAc. Binding of CVN-E41A to SARS-CoV-2 S protein, bearing complex-type N-linked glycosylation and O-glycosylation, was achieved in the SPR at micromolar concentrations (Maier, 2022) through the rather small number of high-mannose containing oligosaccharides via SARS-CoV-2 RBD (Watanabe et al., 2020; Wang et al., 2022). CV-N WT binding to SARS-CoV-2 RBD was measured at weaker affinity (KD=260 µM) (Maier, 2022) as compared with binding of CV-N to HA (A/New York/55/04, KD=5.7 nM) (Maier et al., 2021) and binding to S protein [KD=18.6 µM, (Maier, 2022)]. In comparison, the biologically relevant human ACE2 interaction with the SARS-CoV-2 RBD was reported at KD=4.7 nM (Lan et al., 2020).

Discussion

Antiviral lectins showed neutralization ability alike broadly neutralizing antibodies to be restored by dimerization and to require two H, whereas a single binding site to HIV spike glycoproteins alone was not sufficient to reveal neutralization of enveloped viruses itself (Keeffe et al., 2011). These interactions were verified by generating knock-out variants in the binding site to correlate binding affinities to in silico predicted glycosylation (Maier et al., 2021). Thus, we aim to compare our previously tested chemically mannosylated HA peptides in binding affinity and specificity with short peptide sequences from SARS-related 2019 novel coronavirus (nCoV) spikes and SARS-CoV-2, which are naturally modified by different N-linked glycosylation sites (Kumar et al., 2020; Watanabe et al., 2020) and O-linked glycosylation (Watanabe et al., 2020, Table 1).

TABLE 1
www.frontiersin.org

Table 1 Literature on viral spike glycoproteins GP120, HA, and SARS-CoV (2) variants.

Binding to oligo-mannosides was usually measured by isothermal titration calorimetry (ITC) (Shenoy et al., 2002), and has been applied to CV-N, or mannose-dependent HIV-1 neutralizing 2G12, which bind overlapping epitopes on gp120 involving position 234 and the conserved N-linked GlcNAc at position N295 (Alexandre et al., 2010). More high-mannose glycans attached to N332 were recognized by 2G12 (Scanlan et al., 2002). Mannosylated peptides developed in this study were used as protein scaffolds for screening binding characteristics of antiviral agents by SPR and NMR. Saturation transfer difference (STD)-NMR spectroscopy allowed for characterization of carbohydrate binding by conformational selection (Angulo et al., 2010; Goldflam et al., 2012; Schilling et al., 2020). The STD effect was assigned to the alkyl side chain of T83 in the HA mono- or dimannosylated HA peptide (Schilling et al., 2020). The replacement of two disulfide bonds by exchange of cystines into Trp-Met pairs (forming CVN2L0-V3) may also be utilized to produce chemically induced dynamic nuclear polarization (CIDNP) signals after laser irradiation in the presence of a suitable radical pair-generating dye (Siebert et al., 1997; Schilling et al., 2020). Until today, this technique focused on a series of GlcNAc-binding plant lectins of increasing structural complexity (hevein, pseudohevein, Urtica dioica agglutinin and WGA), for which supporting structural NMR- or X-ray crystallography data was available (Siebert et al., 1997). Taken together, exposed N-linked glycans allowed interactions with, for example, C-type lectin receptor DC-SIGN through many viruses, such as SARS-CoV-2, Ebola, and HIV, and contributed to virus internalization and dissemination. In the context of the recent SARS-CoV-2 pandemic, involvement of DC-SIGN has been linked to severe cases of COVID-19 (Thépaut et al., 2021), possibly competing with neutralizing antibodies to the RBD-motif (Lempp et al., 2021). Binding of CVN-E41A to SARS-CoV-2 S protein, on the other hand, was without physiological relevance, but may trigger neutralization of coronavirus by antibodies targeting non-overlapping epitopes at the conserved site of RBD on S protein.

Author contributions

The author designed and pursued the study, and described the work for the present manuscript.

Conflict of interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Alam, M. A., Parra-Saldivar, R., Bilal, M., Afroze, C. A., Ahmed, M. N, Iqbal, H. M. N., et al. (2021). Algae-derived bioactive molecules for the potential treatment of SARS-CoV-2. Molecules 26, 2134. doi: 10.3390/molecules26082134

PubMed Abstract | CrossRef Full Text | Google Scholar

Alexandre, K. B., Gray, E. S., Lambson, B. E., Moore, P. L., Choge, I. A., Mlisan, K., et al. (2010). Mannose-rich glycosylation patterns on HIV-1 subtype c gp120 and sensitivity to the lectins, griffithsin, cyanovirin-n and scytovirin. J. Virol. 402 (1), 187–96. doi: 10.1016/j.virol.2010.03.021

CrossRef Full Text | Google Scholar

Alsaidi, S., Cornejal, N., Mahoney, O., Melo, C., Verma, N., Bonnaire, T., et al. (2021). Griffithsin and carrageenan combination results in antiviral synergy against SARS-CoV-1 and 2 in a pseudoviral model. Mar. Drugs 19 (8), 418. doi: 10.3390/md19080418

PubMed Abstract | CrossRef Full Text | Google Scholar

Angulo, J., Enríquez-Navas, P. M., Nieto, P. M. (2010). Ligand-receptor binding affinities from saturation transfer difference (STD)-NMR spectroscopy: the binding isotherm of STD initial growth rates. Chemistry 16 (26), 7803–7812. doi: 10.1002/chem.200903528

PubMed Abstract | CrossRef Full Text | Google Scholar

Auth, J., Fröba, M., Große, M., Rauch, P., Ruetalo, N., Schindler, M., et al. (2021). Lectin from triticum vulgaris (WGA) inhibits infection with SARS-CoV-2 and its variants of concern alpha and beta. Int. J. Mol. Sci. 22, 10205. doi: 10.3390/ijms221910205

PubMed Abstract | CrossRef Full Text | Google Scholar

Barnes, C. O., Jette, C.A., Abernathy, M. E., Dam, K.-M. A., Esswein, S. R., Gristick, H. B, et al. (2020a). SARS-CoV-2 neutralizing antibody structures inform therapeutic strategies. Nature 588 (7839), 682–687. doi: 10.1038/s41586-020-2852-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Barnes, C. O., West, A. P., Jr, Huey-Tubman, K. E., Hoffmann, M. A. G., Sharaf, N. G., Hoffman, P. R, et al. (2020b). Structures of human antibodies bound to SARS-CoV-2 spike reveal common epitopes and recurrent features of antibodies. Cell 182 (4), 828–842. doi: 10.1016/j.cell.2020.06.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Barrientos, L. G., Matei, E., Lasala, F., Delgado, R., Gronenborn, A. (2006). Dissecting carbohydrate-Cyanovirin-N binding by structure-guided mutagenesis: functional implications for viral entry inhibition. Protein Eng. Des. Sel 19 (12), 525–535. doi: 10.1093/protein/gzl040

PubMed Abstract | CrossRef Full Text | Google Scholar

Benjamin, E., Wang, W., McAuliffe, J., Palmer-Hill, F., Kallewaard, N., Chen, Z., et al. (2014). A broadly neutralizing human monoclonal antibody directed against a novel conserved epitope on the influenza virus H3 hemagglutinin globular head. J. Virol. 88 (12), 6743–6750. doi: 10.1128/JVI.03562-13

PubMed Abstract | CrossRef Full Text | Google Scholar

Bewley, C. A. (2001). Solution structure of a cyanovirin-N:Man alpha 1-2Man alpha complex: structural basis for high-affinity carbohydrate-mediated binding to gp120. Structure 9 (10), 931–940. doi: 10.1016/S0969-2126(01)00653-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Bewley, C. A., Kiyonaka, S., Hamachi, I. (2002). Site-specific discrimination by cyanovirin-n for alpha-linked trisaccharides comprising the three arms of Man(8) and Man(9). J. Mol. Biol. 322 (4), 881–889. doi: 10.1016/S0022-2836(02)00842-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Bewley, C. A., Otero-Quintero, S. (2001). The potent anti-HIV protein cyanovirin-n contains two novel carbohydrate binding sites that selectively bind to Man(8) D1D3 and Man(9) with nanomolar affinity: implications for binding to the HIV envelope protein gp120. J. Amer Chem. Soc. 123 (17), 3892–3902. doi: 10.1021/ja004040e

CrossRef Full Text | Google Scholar

Bizebard, T., Gigant, B., Rigolet, P., Rasmussen, B., Diat, O., BÃseckei, P., et al. (1995). Structure of influenza virus haemagglutinin complexed with a neutralizing antibody. Nature 376 (6535), 92–94. doi: 10.1038/376092a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Bolmstedt, A. J., O’keefe, B. R., Shenoy, S. R., Mcmahon, J. B., Boyd, M. R, et al. (2001). Cyanovirin-n defines a new class of antiviral agent targeting n-linked, high-mannose glycans in an oligosaccharide-specific manner. Mol. Pharmacol. 59 (5), 949–954. doi: 10.1124/mol.59.5.949

PubMed Abstract | CrossRef Full Text | Google Scholar

Boyd, R. (1997). Discovery of cyanovirin-n, a novel human immunodeficiency virus-inactivating protein that binds viral surface envelope glycoprotein gp120: potential applications to microbicide development. Antimicrob. Agents Chemother. 41 (7), 1521–1530. doi: 10.1128/AAC.41.7.1521

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, Y., Su, B., Guo, X., Sun, W., Deng, Y., Bao, L., et al. (2020). Potent neutralizing antibodies against SARS-CoV-2 identified by high-throughput single-cell sequencing of convalescent patients’ b cells. Cell 182 (1), 73–84. doi: 10.1016/j.cell.2020.05.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, T. H., Yang, Y.-L., Jan, J.-T., Chen, C.-C., Wu, S.-C, et al. (2021). Site-specific glycan-Masking/Unmasking hemagglutinin antigen design to elicit broadly neutralizing and stem-binding antibodies against highly pathogenic avian influenza H5N1 virus infections. Front. Immunol. 12, 692700. doi: 10.3389/fimmu.2021.692700

PubMed Abstract | CrossRef Full Text | Google Scholar

Corti, D., Voss, J., Gamblin, S.J., Codoni, G., Macagno, A., Jarrossay, D., et al. (2011). A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza a hemagglutinins. Science 333 (6044), 850–856. doi: 10.1126/science.1205669

PubMed Abstract | CrossRef Full Text | Google Scholar

Doud, M. B., Lee, J. M., Bloom, J. D. (2018). How single mutations affect viral escape from broad and narrow antibodies to H1 influenza hemagglutinin. Nat. Commun. 9 (1), 1386. doi: 10.1038/s41467-018-03665-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Dreyfus, C., Ekiert, D. C., Wilson, I. A. (2013). Structure of a classical broadly neutralizing stem antibody in complex with a pandemic H2 influenza virus hemagglutinin. J. Virol. 87 (12), 7149–7154. doi: 10.1128/JVI.02975-12

PubMed Abstract | CrossRef Full Text | Google Scholar

Ekiert, D. C., Bhabha, G., Elsliger, M.-A., Friesen, R. H. E., Jongeneelen, M., Throsby, M., et al. (2009). Antibody recognition of a highly conserved influenza virus epitope. Science 324 (5924), 246–251. doi: 10.1126/science.1171491

PubMed Abstract | CrossRef Full Text | Google Scholar

Ekiert, D. C., Friesen, R. H. E., Bhabha, G., Kwaks, T., Jongeneelen, M., Yu, W., et al. (2011). A highly conserved neutralizing epitope on group 2 influenza a viruses. Science 333 (6044), 843–850. doi: 10.1126/science.1204839

PubMed Abstract | CrossRef Full Text | Google Scholar

Fleury, D., Wharton, S. A., Skehel, J. J., Knossow, M., Bizebard, T. (1998). Antigen distortion allows influenza virus to escape neutralization. Nat. Struct. Biol. 5 (2), 119–123. doi: 10.1038/nsb0298-119

PubMed Abstract | CrossRef Full Text | Google Scholar

Fleury, D., Barrère, B., Bizebard, T., Daniels, R. S., Skehel, J. J., Knossow, M., et al. (1999). A complex of influenza hemagglutinin with a neutralizing antibody that binds outside the virus receptor binding site. Nat. Struct. Biol. 6 (6), 530–534. doi: 10.1038/9299

PubMed Abstract | CrossRef Full Text | Google Scholar

Galili, U. (2020). Amplifying immunogenicity of prospective covid-19 vaccines by glycoengineering the coronavirus glycan-shield to present α-gal epitopes. Vaccine 38 (42), 6487–6499. doi: 10.1016/j.vaccine.2020.08.032

PubMed Abstract | CrossRef Full Text | Google Scholar

Gnirß, K., Zmora, P, Blazejewska, P, Winkler, M, Lins, A, Nehlmeier, I., et al. (2015). Tetherin sensitivity of influenza a viruses is strain specific: Role of hemagglutinin and neuraminidase. J. Virol. 89 (18), 9178–9188. doi: 10.1128/JVI.00615-15

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldflam, M., Tarragó, T., Gairí, M., Giralt, E. (2012). NMR studies of protein-ligand interactions. Meth Mol. Biol. 831, 233–259. doi: 10.1007/978-1-61779-480-3_14

CrossRef Full Text | Google Scholar

Gupta, R. K., Apte, G. R., Lokhande, K. B., Mishra, S., Pal, J. K. (2020). Carbohydrate-binding agents: Potential of repurposing for COVID-19 therapy. Curr. Protein Pept. Sci. 21 (11), 1085–1096. doi: 10.2174/1389203721666200918153717

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoffmann, D., Mereiter, S., Oh, Y. J., Monteil, V., Elder, E., Zhu, R., et al. (2021). Identification of lectin receptors for conserved SARS-CoV-2 glycosylation sites. EMBO J. 40 (19), e108375. doi: 10.15252/embj.2021108375

PubMed Abstract | CrossRef Full Text | Google Scholar

Jensen, S. M. R., Ruscetti, F. W., Rein, A., Bertolette, D. C., Saucedo, C. J., O'Keefe, B. R., et al. (2014). Differential inhibitory effects of cyanovirin-n, griffithsin, and scytovirin on entry mediated by envelopes of gammaretroviruses and deltaretroviruses. J. Virol. 88 (4), 2327–2332. doi: 10.1128/JVI.02553-13

PubMed Abstract | CrossRef Full Text | Google Scholar

Ju, B., Zhang, Q., Ge, J., Wang, R., Sun, J., Ge, X., et al. (2020). Human neutralizing antibodies elicited by SARS-CoV-2 infection. Nature 584 (7819), 115–119. doi: 10.1038/s41586-020-2380-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Keeffe, J. R., Gnanapragasam, P. N. P., Gillespie, S. K., Yong, J., Bjorkman, P. J., Mayo, S. L., et al. (2011). Designed oligomers of cyanovirin-n show enhanced HIV neutralization. Proc. Natl. Acad. Sci. U.S.A. 108 (34), 14079–14084. doi: 10.1073/pnas.1108777108

PubMed Abstract | CrossRef Full Text | Google Scholar

Knossow, M., Skehel, J. J. (2006). Variation and infectivity neutralization in influenza. Immunol 119 (1), 1–7. doi: 10.1111/j.1365-2567.2006.02421.x

CrossRef Full Text | Google Scholar

Krause, J. C., Tsibane, T., Tumpey, T. M., Huffman, C. J., Basler, C. F., Crowe Jr, J. E., et al. (2011). A broadly neutralizing human monoclonal antibody that recognizes a conserved, novel epitope on the globular head of the influenza H1N1 virus hemagglutinin. J. Virol. 85 (20), 10905–10908. doi: 10.1128/JVI.00700-11

PubMed Abstract | CrossRef Full Text | Google Scholar

Kreer, C., Zehner, M., Weber, T., Ercanoglu, M. S., Gieselmann, L., Rohde, C., et al. (2020). Longitudinal isolation of potent near-germline SARS-CoV-2-Neutralizing antibodies from COVID-19 patients. Cell 182 (4), 843–845. doi: 10.1016/j.cell.2020.06.044.

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, S., Maurya, V. K., Prasad, A. K., Bhatt, M. L. B., Saxena, S. K. (2020). Structural, glycosylation and antigenic variation between 2019 novel coronavirus (2019-nCoV) and SARS coronavirus (SARS-CoV). Virusdisease 31 (1), 13–21. doi: 10.1007/s13337-020-00571-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Lan, J., Ge, J., Yu, J., Shan, S., Zhou, H., Fan, S., et al. (2020). Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature 581 (7807), 215–220. doi: 10.1038/s41586-020-2180-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, P. S., Yoshida, R., Ekiert, D. C., Sakai, N., Suzuki, Y., Takada, A., et al. (2012). Heterosubtypic antibody recognition of the influenza virus hemagglutinin receptor binding site enhanced by avidity. Proc. Natl. Acad. Sci. U.S.A. 109 (42), 17040–17 45. doi: 10.1073/pnas.1212371109

PubMed Abstract | CrossRef Full Text | Google Scholar

Lempp, F. A., Soriaga, L. B., Montiel-Ruiz, M., Benigni, F., Noack, J., Park, Y.-J, et al. (2021). Lectins enhance SARS-CoV-2 infection and influence neutralizing antibodies. Nature 598, 342–347. doi: 10.1038/s41586-021-03925-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Liao, H. Y., Wang, S. -C., Ko, Y. -A., Lin, K. -I., Ma, C., Cheng, T. -J. R, et al. (2020). Chimeric hemagglutinin vaccine elicits broadly protective CD4 and CD8 T cell responses against multiple influenza strains and subtypes. Proc. Natl. Acad. Sci. U.S.A. 117 (30), 17757–17763. doi: 10.1073/pnas.2004783117

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, D., Wang, C., Wang, S., Mehmood, A., Gu, J., Cheng, X., et al. (2022). Discovery of a natural product with potent efficacy against SARS-CoV-2 by drug screening. Interdiscip. Sciences: Comput. Life Sci. 14, 55–63. doi: 10.1007/s12539-021-00477-w

CrossRef Full Text | Google Scholar

Lin, W. S., Chen, I.-C., Chen, H.-C., Lee, Y.-C., Wu, S.-C. (2021). Glycan masking of epitopes in the NTD and RBD of the spike protein elicits broadly neutralizing antibodies against SARS-CoV-2 variants. Front. Immunol. 12, 795741. doi: 10.3389/fimmu.2021.795741

PubMed Abstract | CrossRef Full Text | Google Scholar

Lingwood, D., McTamney, P. M., Yassine, H. M., Whittle, J. R. R., Guo, X., Boyington, J. C, et al. (2012). Structural and genetic basis for development of broadly neutralizing influenza antibodies. Nature 489 (7417), 566–570. doi: 10.1038/nature11371

PubMed Abstract | CrossRef Full Text | Google Scholar

Lv, Z., Deng, Y.-Q., Ye, Q., Cao, L., Sun, C.-Y., Fan, C., et al. (2020). Structural basis for neutralization of SARS-CoV-2 and SARS-CoV by a potent therapeutic antibody. Science 369 (6510), 1505–1509. doi: 10.1126/science.abc5881

PubMed Abstract | CrossRef Full Text | Google Scholar

Maier, I. (2022). Engineering antiviral agents via surface plasmon resonance. J. Vis. Exp. 184. doi: 10.3791/63541

CrossRef Full Text | Google Scholar

Maier, I., Schiestl, R. H., Kontaxis, G. (2021). Cyanovirin-n binds viral envelope proteins at the low-affinity carbohydrate binding site without direct virus neutralization ability. Molecules 26 (12), 3621. doi: 10.3390/molecules26123621

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazur-Marzec, H., Cegłowska, M., Konkel, R., Pyrc, K. (2021). Antiviral cyanometabolites–a review. Biomolecules 11 (3), 474. doi: 10.3390/biom11030474

PubMed Abstract | CrossRef Full Text | Google Scholar

Nachbagauer, R., Wohlbold, T. J., Hirsh, A., Hai, R., Sjursen, H., Palese, P., et al. (2014). Induction of broadly reactive anti-hemagglutinin stalk antibodies by an H5N1 vaccine in humans. J. Virol. 88 (22), 13260–13268. doi: 10.1128/JVI.02133-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Naidoo, D., Kar, P., Roy, A., Mutanda, T., Bwapwa, J., Sen, A., et al. (2021). Structural insight into the binding of cyanovirin-n with the spike glycoprotein, mpro and PLpro of SARS-CoV-2: Protein–protein interactions, dynamics simulations and free energy calculations. Molecules 26 (17), 5114. doi: 10.3390/molecules26175114

PubMed Abstract | CrossRef Full Text | Google Scholar

Nogales, A., Piepenbrink, M. S., Wang, J., Ortega, S., Basu, M., Fucile, C. F., et al. (2018). A highly potent and broadly neutralizing H1 influenza-specific human monoclonal antibody. Sci. Rep. 8 (1), 4374. doi: 10.1038/s41598-018-22307-8

PubMed Abstract | CrossRef Full Text | Google Scholar

O’Keefe, B. R., Smee, D. F., Turpin, J. A., Saucedo, C. J., Gustafson, K. R., Mori, T., et al. (2003). Potent anti-influenza activity of cyanovirin-n and interactions with viral hemagglutinin. Antimicrob. Agents Chemother. 47 (8), 2518–2525. doi: 10.1128/AAC.47.8.2518-2525.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

Otterstrom, J. J., Brandenburg, B., Koldijk, M. H., Juraszek, J., Tang, C., Mashaghi, S., et al. (2014). Relating influenza virus membrane fusion kinetics to stoichiometry of neutralizing antibodies at the single-particle level. Proc. Natl. Acad. Sci. U.S.A. 111 (48), E5143–E5148. doi: 10.1073/pnas.1411755111

PubMed Abstract | CrossRef Full Text | Google Scholar

Pinto, D., Park, Y.-J., Beltramello, M., Walls, A. C., Tortorici, M. A., Bianchi, S., et al. (2020). Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Nature 583 (7815), 290–295. doi: 10.1038/s41586-020-2349-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Qiu, Y., Stegalkina, S., Zhang, J., Boudanova, E., Park, A., Zhou, Y., et al. (2020). Mapping of a novel H3-specific broadly neutralizing monoclonal antibody targeting the hemagglutinin globular head isolated from an elite influenza virus-immunized donor exhibiting serological breadth. J. Virol. 94 (6), e01035–e01019. doi: 10.1128/JVI.01035-19

PubMed Abstract | CrossRef Full Text | Google Scholar

Raymond, D. D., Bajica, G., Ferdman, J., Suphaphiphat, P., Ethan, C., Settembre, M., et al. (2018). Conserved epitope on influenza-virus hemagglutinin head defined by a vaccine-induced antibody. Proc. Natl. Acad. Sci. U.S.A. 115 (1), 168–173. doi: 10.1073/pnas.1715471115

PubMed Abstract | CrossRef Full Text | Google Scholar

Robbiani, D. F., Gaebler, C., Muecksch, F., Lorenzi, J. C. C., Wang, Z., Cho, A., et al. (2020). Convergent antibody responses to SARS-CoV-2 in convalescent individuals. Nature 584 (7821), 437–442. doi: 10.1038/s41586-020-2456-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Salunke, S. B., Babu, N. S., Chen, C.-T. (2011). Iron(III) chloride as an efficient catalyst for stereoselective synthesis of glycosyl azides and a cocatalyst with Cu(0) for the subsequent click chemistry. Chem. Commun. (Camb) 47 (37), 10440–10442. doi: 10.1039/c1cc13370e

PubMed Abstract | CrossRef Full Text | Google Scholar

Scanlan, C. N., Pantophlet, R., Wormald, M. R, Saphire, E. O., Stanfield, R., Wilson, I. A., et al. (2002). The broadly neutralizing anti-human immunodeficiency virus type 1 antibody 2G12 recognizes a cluster of alpha1–>2 mannose residues on the outer face of gp120. J. Virol. 76 (14), 7306–7321. doi: 10.1128/JVI.76.14.7306-7321.2002

PubMed Abstract | CrossRef Full Text | Google Scholar

Schilling, P. E., Kontaxis, G., Dragosits, M., Schiestl, R. H., Becker, C. F.W., Maier, I., et al. (2020). Mannosylated hemagglutinin peptides bind cyanovirin-n independent of disulfide-bonds in complementary binding sites. RSC Adv. 10, 11079–11087. doi: 10.1039/D0RA01128B

PubMed Abstract | CrossRef Full Text | Google Scholar

Schmidt, A. G., Therkelsen, M. D., Stewart, S., Kepler, T. B, et al. (2015). Viral receptor-binding site antibodies with diverse germline origins. Cell 161 (5), 1026–1034. doi: 10.1016/j.cell.2015.04.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Shenoy, S. R., Barrientos, L. G., Ratner, D. M., O'Keefe, B. R., Seeberger, P. H., Gronenborn, A. M., et al. (2002). Multisite and multivalent binding between cyanovirin-n and branched oligomannosides: calorimetric and NMR characterization. Chem. Biol. 9 (10), 1109–1118. doi: 10.1016/S1074-5521(02)00237-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, R., Shan, C., Duan, X., Chen, Z., Liu, P., Song, J., et al. (2020). A human neutralizing antibody targets the receptor-binding site of SARS-CoV-2. Nature 584 (7819), 120–124. doi: 10.1038/s41586-020-2381-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Siebert, H. C., von der Lieth, C.-W., Kaptein, R., Beintema, J. J., Dijkstra, K., van Nuland, N., et al. (1997). Role of aromatic amino acids in carbohydrate binding of plant lectins: laser photo chemically induced dynamic nuclear polarization study of hevein domain- containing lectins. Proteins 28 (2), 268–284. doi: 10.1002/(SICI)1097-0134(199706)28:2<268::AID-PROT14>3.0.CO;2-G

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanabe-Tochikura, A., Tochikura, T. S., Yoshida, O., Oki, T., Yamamoto, N. (1990). Pradimicin a inhibition of human immunodeficiency virus: attenuation by mannan. Virology 176 (2), 467–473. doi: 10.1016/0042-6822(90)90016-K

PubMed Abstract | CrossRef Full Text | Google Scholar

Thépaut, M., Luczkowiak, J., Vivès, C., Labiod, N., Bally, I., Lasala, F., et al. (2021). DC/L-SIGN recognition of spike glycoprotein promotes SARS-CoV-2 trans-infection and can be inhibited by a glycomimetic antagonist. PloS Pathog. 17 (5), e1009576. doi: 10.1371/journal.ppat.1009576

PubMed Abstract | CrossRef Full Text | Google Scholar

Walls, A. C., Park, Y.-J., Tortorici, M. A., Wall, A., McGuire, A. T., Veesler, D., et al. (2020a). Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell 181 (2), 281–92.e6. doi: 10.1016/j.cell.2020.02.058

PubMed Abstract | CrossRef Full Text | Google Scholar

Walls, A. C., Fiala, B., Schäfer, A., Wrenn, S., Pham, M. N., Murphy, M., et al. (2020b). Elicitation of potent neutralizing antibody responses by designed protein nanoparticle vaccines for SARS-CoV-2. Cell 183 (5), 1367–1382. doi: 10.1016/j.cell.2020.10.043.

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, W., Li, Q., Wu, J., Hue, Y., Wua, G., Yu, C., et al. (2021). Lentil lectin derived from lens culinaris exhibit broad antiviral activities against SARS-CoV-2 variants. Emerging Microbes Infect 10, 1519–1529. doi: 10.1080/22221751.2021.1957720

CrossRef Full Text | Google Scholar

Wang, X., Chen, X., Tan, J., Yue, S., Zhou, R., Xu, Y., et al. (2022). 35B5 antibody potently neutralizes SARS-CoV-2 omicron by disrupting the n- glycan switch via a conserved spike epitope. Cell Host Microbe 30, 1–9. doi: 10.1016/j.chom.2022.03.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Watanabe, Y., Allen, J. D., Wrapp, D., McLellan, J. S., Crispin, M., et al. (2020). Site-specific glycan analysis of the SARS-CoV-2 spike. Science 369 (6501), 330–333. doi: 10.1126/science.abb9983

PubMed Abstract | CrossRef Full Text | Google Scholar

Winarski, K. L., Tang, J., Klenow, L., Lee, J., Coyle, E. M., Manischewitz, J., et al. (2019). Antibody-dependent enhancement of influenza disease promoted by increase in hemagglutinin stem flexibility and virus fusion kinetics. Proc. Natl. Acad. Sci. U.S.A. 116 (30), 15194–15199. doi: 10.1073/pnas.1821317116

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, N. C., Thompson, A.J., Lee, J. M., Su, W., Arlian, B. M., Xie, J, et al. (2020a). Different genetic barriers for resistance to HA stem antibodies in influenza H3 and H1 viruses. Science 368 (6497), 1335–1340. doi: 10.1126/science.aaz5143

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, N. C., Yuan, M., Liu, H., Lee, C.-C. D., Zhu, X., Bangaru, S., et al. (2020b). An alternative binding mode of IGHV3-53 antibodies to the SARS-CoV- 2 receptor binding domain. Cell Rep. 33 (3), 108274. doi: 10.1016/j.celrep.2020.108274

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, N. C., Yuan, M., Bangaru, S., Huang, D., Zhu, X., Lee, C.-C. D, et al. (2020c). A natural mutation between SARS-CoV-2 and SARS-CoV determines neutralization by a cross-reactive antibody. PloS Pathog. 16 (12), e1009089. doi: 10.1371/journal.ppat.1009089

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, N. C., Wilson, I. A. (2017). A perspective on the structural and functional constraints for immune evasion: Insights from influenza virus. J. Mol. Biol. 429 (17), 2694–2709. doi: 10.1016/j.jmb.2017.06.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, F., Bewley, C., Louis, J., Gustafson, K., Boyd, M., Gronenborn, A., et al. (1999). Crystal structure of cyanovirin-n, a potent HIV-inactivating protein, shows unexpected domain swapping. J. Mol. Biol. 288 (3), 403–412. doi: 10.1006/jmbi.1999.2693

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, F., Xiang, R., Deng, X., Wang, L., Yu, Z., Tian, S., et al. (2020). Receptor-binding domain-specific human neutralizing monoclonal antibodies against SARS-CoV and SARS-CoV-2. Signal Transduct Target Ther. 5 (1), 212. doi: 10.1038/s41392-020-00318-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Zost, S. J., Parkhouse, K., Gumina, M. E., Kangchon, K., Diaz Perez, S., Wilson, P. C, et al. (2017). Contemporary H3N2 influenza viruses have a glycosylation site that alters binding of antibodies elicited by egg-adapted vaccine strains. Proc. Natl. Acad. Sci. U.S.A. 114 (47), 12578–12583. doi: 10.1073/pnas.1712377114

PubMed Abstract | CrossRef Full Text | Google Scholar

Zost, S. J., Gilchuk, P., Case, J. B., Binshtein, E., Chen, R. E., Nkolola, J. P, et al. (2020a). Potently neutralizing and protective human antibodies against SARS-CoV-2. Nature 584 (7821), 443–449. doi: 10.1038/s41586-020-2548-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Zost, S. J., Gilchuk, P., Chen, R. E., Case, J., Reidy, J. X., Trivette, A, et al. (2020b). Rapid isolation and profiling of a diverse panel of human monoclonal 559 antibodies targeting the SARS-CoV-2 spike protein. Nat. Med. 26 (9), 1422–1427. doi: 10.1038/s41591-020-0998-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: lectin, cyanovirin-N, carbohydrate-binding agent, virus, SARS-CoV-2

Citation: Maier I (2022) Engineering recombinantly expressed lectin-based antiviral agents. Front. Cell. Infect. Microbiol. 12:990875. doi: 10.3389/fcimb.2022.990875

Received: 10 July 2022; Accepted: 25 August 2022;
Published: 23 September 2022.

Edited by:

Els Jm Van Damme, Ghent University, Belgium

Reviewed by:

Jay Trivedi, Rhode Island Hospital, United States

Copyright © 2022 Maier. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Irene Maier, irene.maier@meduniwien.ac.at

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.