Next Article in Journal
Aging Activates the Immune System and Alters the Regenerative Capacity in the Zebrafish Heart
Next Article in Special Issue
10th Anniversary of Cells—Advances in Plant, Algae and Fungi Cell Biology
Previous Article in Journal
Environmental and Circadian Regulation Combine to Shape the Rhythmic Selenoproteome
Previous Article in Special Issue
Comparative Genomics and Physiological Investigation of a New Arthrospira/Limnospira Strain O9.13F Isolated from an Alkaline, Winter Freezing, Siberian Lake
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Legume Lectins with Different Specificities as Potential Glycan Probes for Pathogenic Enveloped Viruses

1
UMR 152 PharmaDev, Institut de Recherche et Développement, Faculté de Pharmacie, Université Paul Sabatier, 35 Chemin des Maraîchers, F-31062 Toulouse, France
2
Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium
3
UMR Virologie, INRA, ANSES, Ecole Nationale Vétérinaire d’Alfort, F-94700 Maisons-Alfort, France
*
Author to whom correspondence should be addressed.
Cells 2022, 11(3), 339; https://doi.org/10.3390/cells11030339
Submission received: 17 December 2021 / Revised: 13 January 2022 / Accepted: 18 January 2022 / Published: 20 January 2022

Abstract

:
Pathogenic enveloped viruses are covered with a glycan shield that provides a dual function: the glycan structures contribute to virus protection as well as host cell recognition. The three classical types of N-glycans, in particular complex glycans, high-mannose glycans, and hybrid glycans, together with some O-glycans, participate in the glycan shield of the Ebola virus, influenza virus, human cytomegalovirus, herpes virus, human immunodeficiency virus, Lassa virus, and MERS-CoV, SARS-CoV, and SARS-CoV-2, which are responsible for respiratory syndromes. The glycans are linked to glycoproteins that occur as metastable prefusion glycoproteins on the surface of infectious virions such as gp120 of HIV, hemagglutinin of influenza, or spike proteins of beta-coronaviruses. Plant lectins with different carbohydrate-binding specificities and, especially, mannose-specific lectins from the Vicieae tribe, such as pea lectin and lentil lectin, can be used as glycan probes for targeting the glycan shield because of their specific interaction with the α1,6-fucosylated core Man3GlcNAc2, which predominantly occurs in complex and hybrid glycans. Other plant lectins with Neu5Ac specificity or GalNAc/T/Tn specificity can also serve as potential glycan probes for the often sialylated complex glycans and truncated O-glycans, respectively, which are abundantly distributed in the glycan shield of enveloped viruses. The biomedical and therapeutical potential of plant lectins as antiviral drugs is discussed.

Graphical Abstract

1. Introduction

Many pathogenic viruses for humans are so-called enveloped viruses with a lipid bilayer that allows the infectious virions to fuse with the cell membrane, followed by the entry and replication of the viral genetic material into the host cells. Ebola virus (EBOV), influenza virus (IV), herpes simplex virus (HSV), human immunodeficiency virus (HIV), human cytomegalovirus (HCMV), Lassa virus (LASV), and the beta-coronaviruses responsible for the Middle East respiratory syndrome (MERS-CoV) and the severe acute respiratory syndrome (i.e., SARS-CoV and SARS-CoV-2) belong to this group of pathogenic enveloped viruses [1]. Among the surface glycoproteins that are embedded in the lipid bilayer of enveloped viruses, so-called fusion glycoproteins play a key role in mediating the recognition of infectious virions by the host cell membrane receptors and their subsequent anchorage to the host cells [2]. Energetically driven conformational changes occurring in the metastable fusion proteins, which usually occur in a prefusion state, are responsible for an enhanced exposure of the receptor-binding domain (RBD) of the fusion proteins that favors their recognition by the host cell receptors [1]. Fusion proteins of enveloped viruses usually consist of the non-covalent association of three monomers to build a homotrimeric structure exposed on the surface of the virions. However, depending on the enveloped viruses, the structure, shape, and size of the monomers building the homotrimers are highly variable from one virus to another (Table 1). Other surface glycoproteins, such as the so-called B glycoprotein from HSV [3], and E proteins from flaviviruses responsible for some severe diseases, including chikungunya virus (CHIV), dengue virus (DENV), and Zika virus (ZIV), also contribute to the glycan shield covering the infectious virions [4,5,6].
Table 1. Structural properties of fusion protein and E glycoprotein associations of enveloped viruses as parts of the glycan shield covering infectious virions.
Table 1. Structural properties of fusion protein and E glycoprotein associations of enveloped viruses as parts of the glycan shield covering infectious virions.
Enveloped VirusHomotrimerMonomerPDB Entry *Reference
Ebola virus (EBOV)Homotrimer 7JPH[7]
Influenza virus (IV)Hemagglutinin A
Homotrimer
hemagglutinin A6Y5G[8]
Human cytomegalovirus (HCMV)HomotrimerB glycoprotein5CXF[9]
Herpes simplex virus (HSV)HomotrimerB glycoprotein2GUM[3]
Human immunodeficiency virus (HIV)Homotrimergp1404TVP[10]
Lassa virus (LASV)HomotrimerGPC glycoprotein5VK2[11]
Middle east respiratory syndrome
coronavirus (MERS-CoV)
SpikeS protein5W9H[12]
Severe acute respiratory syndrome
coronavirus-1 (SAR-CoV)
SpikeS protein6ACD[13]
Severe acute respiratory syndrome
coronavirus-2 (SARS-CoV-2)
SpikeS protein6VXX[14]
Chikungunya virus (CHIV)HomodimerE protein3N40[4]
Dengue virus (DENV)HomodimerE protein1UZG[5]
Zika virus (ZIV)HomodimerE protein57BUB[6]
* A single PDB code is indicated but several PDB entries are available at the PDB.
Although N- and O-glycans decorate the fusion glycoproteins, the three classical types of N-glycans, including complex-type glycans, high-mannose-type glycans, and hybrid-type glycans, are predominantly distributed along the fusion proteins of pathogenic enveloped viruses. In addition, a high proportion of complex glycans are α1,6-fucosylated on the first GlcNAc linked to the Asn residue and often sialylated on their terminal Gal residues [15]. Moreover, the extreme diversity of complex glycans appears as a characteristic of enveloped viruses. In addition to the N-glycans, O-glycans have been identified on the envelope glycoproteins of infectious virions, especially in pathogenic coronaviruses such as SARS-CoV-2 [16]. In fact, most of the Ser and Thr residues of unoccupied NXT/S glycosylation sites of SARS-CoV-2 are O-glycosylated by short Gal/GalNAc/T/Tn-containing O-glycan chains [17]. However, the O-glycan content of SARS-CoV-2 is much lower than the level of N-glycans.
Lectins are known as a group of carbohydrate-binding proteins of non-immune origin that are widely distributed in plants. Many lectins have been studied for their role in the protection of plants against pathogens, aiming to resolve the function of the lectins inside different plant tissues. In addition, these carbohydrate-binding proteins have been proven to be important tools for glycobiology, allowing for the investigation of the importance of protein–carbohydrate interactions. Several plant lectins have been reported as potent molecules with anti-infectivity properties for RNA viruses including pathogenic enveloped viruses. Depending on their carbohydrate-binding specificity, lectins can recognize and bind particular types of glycan structures present in the glycan shield of viruses. Over the past decades, lectins from different legume species, referred to as legume lectins, have been studied in great detail. Despite the fact that legume lectins represent a large family of proteins with important similarity in their amino acid sequences, these lectins show remarkable variability in their carbohydrate-binding properties. Many legume lectins have been reported to recognize glycoconjugates on cells and viruses and can discriminate between diverse glycan structures, making them interesting research tools for glycomic research [18].
The primary purpose of this review was to give an overview of the types of glycans present in the glycan shield of different pathogenic enveloped viruses and how legume lectins with different specificities can act as carbohydrate-binding agents (CBAs) for these viruses. Finally, biomedical perspectives for plant lectins with antiviral properties are also discussed.

2. The Glycan Shield of Pathogenic Enveloped Viruses

Glycoproteins that are part of the glycan shield that covers the enveloped viruses are modified with three types of N-glycans including complex glycans, high-mannose glycans, and hybrid glycans (Figure 1):
  • Complex-type N-glycans are most abundant on all the envelope proteins except for the gp120 and hemagglutinin from HIV and IV, respectively, which predominantly contain high-mannose-type N-glycans. Complex glycans exhibit a high diversity in their glycan structure, including bi-, tri-, and tetra-antennary glycans which are often sialylated on their terminal Gal residues and fucosylated on sub-terminal GlcNAc residues. Most of these complex N-glycans possess an α1,6-fucosylated Man3GlcNAc2 core;
  • High-mannose N-glycans are less abundant and offer less diversity than complex N-glycans because they consist exclusively of Man residues. High-mannose N-glycans from enveloped viruses include oligomannosides containing 4–9 (Man4–9) Man residues, and all of them possess a non-fucosylated Man3GlcNAc2 core;
  • Hybrid N-glycans are least abundant on enveloped viruses.
A detailed study of the N-glycan structures occurring on the beta-coronaviruses, MERS-CoV, SARS-CoV, and SARS-CoV-2, confirmed the high heterogeneity of the complex glycans of the S glycoprotein forming the spikes and revealed important differences depending on the type of beta-coronavirus [15]. In addition, although some of the N-glycosylation sites, NXT/S, are often occupied with variable proportions of complex and high-mannose N-glycans, the complex N-glycans are largely predominant [16,27,28,29].
Furthermore, a few O-glycans also occur, especially on the S protein from beta-coronaviruses [16]. Interestingly, the Thr and Ser residues of N-glycosylation sites unoccupied by N-glycans are modified with short O-glycan chains [17]. Usually, these O-glycans are less exposed on the surface of S proteins, mainly due to the fact of their smaller size compared to the large and highly exposed N-glycans [27].
Both the homotrimeric organization of the fusion proteins and the homodimeric organization of E glycoproteins on the surface of pathogenic enveloped viruses favor the exposure of their glycan shield (Figure 2 and Figure 3). However, the distribution of N-glycans, especially at the top of the fusion protein homotrimer, provides areas devoid of glycans allowing for the recognition of pathogenic viruses by the corresponding DPP4 and ACE2 receptors located on the host cells. These glycan-free areas, which correspond to the so-called RBDs of S proteins from MERS-CoV, SARS-CoV, and SARS-CoV-2, contribute to the infectious potential developed by the pathogenic beta-coronaviruses [28,29,30].
In spite of the glycan-free character of the RBDs, it should be noted that these areas are surrounded by glycan chains that should be accessible to CBAs, such as lectins, which could hamper the proper recognition of RBDs by their corresponding host cell receptors (Figure 3) [14].

3. Plant Lectins with Different Specificities Are Potential CBAs for Pathogenic Enveloped Viruses

Lectins from higher plants offer extreme diversity in terms of structural organization and recognition of simple and complex glycans [31]. Owing to the high diversity that characterizes the glycan shield of pathogenic enveloped viruses, the heterogeneous group of Man-specific lectins and, especially, the group of two-chain legume lectins, emerges as a potential tool for specific targeting of the N-glycan shield of enveloped viruses. Two-chain legume lectins form a particular group of Man-specific lectins that display an enhanced affinity for complex N-glycans possessing an α1,6-fucosylated trimannoside core Man3GlcNAc2 [32,33]. Seed lectins from pea (Pisum sativum) (PsA), lentil (Lens culinaris) (LcA), Cyprus-vetch (Lathyrus ochrus) (LoL-I/II), and faba bean (Vicia faba) (VfA) belong to this group of two-chain lectins [34,35,36,37]. They are built from the non-covalent association of two identical monomers built up from a heavy (β-chain) and a light (α-chain) subunit and possess an overall jelly roll structure similar to that of Con A, the single-chain Man-binding lectin from Jackbean (Canavalia ensiformis) [38]. A detailed crystallographic study of the Lathyrus ochrus isolectin-II (LoL-II) in complex with an octasaccharide derived from the human lactotransferrin (PDB code 1LGC) [39] revealed that the enhanced affinity of Vicieae lectins towards the α1,6-fucosylated Man3GlcNAc2 core depends on the direct interaction of the α1,6-linked Fuc residue with some of the amino acid residues forming the carbohydrate-binding site (CBS) of the lectin via a few hydrogen bonds (Figure 4).
In addition, complexes of pea lectin and Lathyrus ochrus lectin with non-fucosylated trisaccharides, indicated that Vicieae lectins also interact with one of the terminal Man of the trimannosyl core from the non-fucosylated N-glycans [40]. In this respect, LoL-I from L. ochrus seeds interacted with the trisaccharide Manα1,3Manβ1,4GlcNAc in such a way that the terminal Man residue occupies the monosaccharide-binding site of the lectin (Figure 5).
This binding pattern allows Vicieae lectins to readily interact with the trimannosyl core from the three types of complex, high-mannose, and hybrid N-glycans, irrespective of the possible α1,6-fucosylation on the first GlcNAc residue of the N-glycan chain.
A survey of the glycan array analyses performed by the Consortium for Functional Glycomics (CFG) (http://www.functionalglycomics.org (accessed on 15 December 2021)) for PsA, LcA, and VfA, all members of the two-chain lectins from the Vicieae tribe, yielded the best results with glycans possessing the a1,6-fucosylated Man3GlcNAc2 core. As an example, most of the top five glycans displaying the best affinity for PsA, LcA, and VfA occur in the glycan shield covering the pathogenic enveloped viruses (Figure 6).
Other Man-specific lectins, such as the GNA-related lectins from different families of monocot plants, including Liliaceae, Amaryllidaceae, Polygonaceae, and Orchidaceae, preferentially interact with high-mannose glycans that contain a non-fucosylated Man3GlcNAc2 core. As an example, the top five glycans interacting with GNA in glycan array experiments mainly consist of high-mannose glycans (Figure 7). These high-mannose glycans were present in all the investigated enveloped viruses.
In addition, the N-glycans of pathogenic enveloped viruses are often sialylated on their terminal Gal antennae residues, which offers another potential recognition target for lectins that specifically recognize terminal sialylated Gal residues. The black elderberry (Sambucus nigra) bark lectin I (SNA-I) specifically interacts with these sialylated termini. In this respect, the top five glycans interacting with SNA-I in glycan array experiments contained sialylated Gal residues that occur in the glycan shield of enveloped viruses (Figure 8).
Finally, GalNAc/T/Tn lectins that recognize O-glycans, such as jacalin from Artocarpus integer, PNA from peanut (Arachis hypogaea), and Morniga-G from Morus nigra, should especially interact with the few O-glycans exposed at the surface of beta-coronaviruses [41]. The top five O-glycans interacting with Morniga-G in glycan array experiments are present in the glycan shield of the SARS-CoV-2 particles (Figure 9).

4. Man-Specific and Neu5Ac-Specific Lectins as Potential CBAs for Pathogenic Enveloped Viruses

Plant lectins with different carbohydrate-binding specificities have been identified as CBAs for pathogenic enveloped virus including HIV, HCMV, the hepatitis C virus HCV, HSV, IV, and the coronaviruses MERS-CoV, SARS-CoV, and SARS-CoV-2. Targets for these CBAs are the glycan structures present on the envelope proteins of pathogenic enveloped viruses (Table 2).
Table 2. List of plant lectins identified as carbohydrate-binding agents (CBAs) for envelope proteins from pathogenic enveloped viruses.
Table 2. List of plant lectins identified as carbohydrate-binding agents (CBAs) for envelope proteins from pathogenic enveloped viruses.
LectinPlant SpeciesCarbohydrate-
Binding
Specificity
Targeted Envelope
Protein
VirusRef.
APAAllium porumManS-proteinSARS-CoV[41]
AUAAllium ursinumManS-proteinSARS-CoV[41]
BanLecMusa acuminataMangp120HIV[42,43,44]
hemagglutininIV[45]
E-glycoproteinHCMV[46]
E-glycoproteinEBOV[46,47]
E-glycoproteinLASV[46]
Con ACanavalia ensiformisMangp120HIV[48]
S-proteinSARS-CoV-2[49]
Succinyl-Con ACanavalia ensiformisManS-proteinMers-CoV[50]
S-proteinSARS-CoV[50]
S-proteinSARS-CoV-2[50]
ConBrCanavalia brasiliensisManS-proteinSARS-CoV-2[51]
ConMCanavalia maritimaManS-proteinSARS-CoV-2[51]
CLACladastris luteaManS-proteinSARS-CoV[41]
CHACymbidium hybridManβ-glycoproteinHCMV[52]
E-glycoproteinHCV[52]
gp120HIV[52]
S-proteinSARS-CoV[53]
DSLDatura stramoniumNeu5Ac-Gal/GalNAcS-proteinMERS-CoV[50]
S-proteinSARS-CoV[50]
S-proteinSARS-CoV-2[50]
DLasLDioclea lasiocarpaManS-proteinSARS-CoV-2[51]
DSclerLDioclea sclerocarpaManS-proteinSARS-CoV-2[51]
EHAEpipactis helleborineManβ-glycoproteinHCMV[52]
gp120HIV[52]
hemagglutininIV[52]
GNAGalanthus nivalisManβ-glycoproteinHCMV[53]
E-glycoproteinHCV[54,55]
gp120HIV[53]
S-proteinSARS-CoV[41]
S-proteinSARS-CoV-2[56]
hemagglutininIV[57]
HHAHyppeastrum hybridManβ-glycoproteinHCMV[53]
E-glycoproteinHCV[52]
gp120HIV[52]
S-proteinSARS-CoV[41]
hemagglutininIV[57]
HorcolinHordeum vulgareMangp120HIV[58]
IRAIris hybridGalNAc/GalS-proteinSARS-CoV[41]
FRILLablab purpureusManS-proteinSARS-CoV[59]
hemagglutininIV[59]
LcALens culinarisManS-proteinMERS-CoV[50]
S-proteinSARS-CoV[50]
S-proteinSARS-CoV-2[50]
LOAListera ovataManβ-glycoproteinHCMV[53]
gp120HIV[53]
MALMaackia amurensisNeu5AcS-proteinSARS-CoV-2[60]
Morniga-GMorus nigraGalS-proteinSARS-CoV[41]
Morniga-MMorus nigraManS-proteinSARS-CoV[41]
NPANarcissus pseudonarcissusManβ-glycoproteinHCMV[53]
gp120HIV[53]
NictabaNicotiana tabacum(GlcNAc)nS-proteinSARS-CoV[41,61]
OrysataOryza sativaMangp120HIV[62]
S-proteinSARS-CoV[62]
PHAPhaseolus vulgarisComplex glycansS-proteinMERS-CoV[50]
S-proteinSARS-CoV[50]
S-proteinSARS-CoV-2[50]
PCAPolygonatum cyrtonemaMangp120HIV[63]
SSLSambucus sieboldianaNeu5Ac-Gal/GalNAcS-proteinSARS-CoV[50]
S-proteinSARS-CoV-2[50]
TLC IITulipa hybridManS-proteinSARS-CoV[41]
TDLTyphonium divaricatumManE-glycoproteinHSV[64]
UDAUrtica dioica(GlcNAc)nβ-glycoproteinHCMV[52]
E-glycoproteinHCV[51]
gp120HIV[52]
S-proteinSARS-CoV[41]
hemagglutininIV[57]
ML IIVicum albumGal/GalNAcS-proteinSARS-CoV[41]
ML IIIViscum albumGal/GalNAcS-proteinSARS-CoV[41]
WGATriticum aestivumGlcNAc/Neu5AcS-proteinMERS-CoV[50]
S-proteinSARS-CoV[41,50]
S-proteinSARS-CoV-2[50,65]
GNAmaizeZea maysManS-proteinSARS-CoV[66]
In addition to CBAs from higher plants, it should be noted that other lectins isolated from algae and Cyanobacteria (formerly classified as blue algae), which essentially recognize high-mannose glycans, have been identified as potential CBAs for pathogenic enveloped viruses (Table 3). In this respect, griffithsin, the Man-specific lectin purified from the red alga Griffithsia sp. [67], was investigated in detail as a relevant CBA for targeting the envelope protein from pathogenic enveloped viruses [68] because of its high affinity for oligomannosides [69] (Figure 10).
Table 3. List of algal and cyanobacterial lectins identified as carbohydrate-binding agents (CBAs) for envelope proteins from pathogenic enveloped viruses.
Table 3. List of algal and cyanobacterial lectins identified as carbohydrate-binding agents (CBAs) for envelope proteins from pathogenic enveloped viruses.
LectinAlgal SpeciesCarbohydrate-
Binding
Specificity
Targeted Envelope
Protein
VirusRef.
AMLAmantia multifidaFetuin, mannanhemagglutininIV[52]
E-glycoproteinHSV[52]
gp120HIV[52]
BSLBryothamnion seaforthiiFetuin, mucingp120HIV[52]
E-glycoproteinHSV[52]
hemagglutininIV[52]
ESA-2Eucheuma serraManhemagglutininIV[70]
GCLGrateloupia chiangiiManhemagglutininIV[71]
E-glycoproteinHSV[71]
GriffithsinGriffithsia sp. Mangp120HIV[72]
E-glycoproteinHCV[73]
S-proteinMERS-CoV[74]
S-proteinSARS-CoV[68]
S-proteinSARS-CoV-2[75]
HMLHypnea musciformisThyroglobulin, mucingp120HIV[52]
hemagglutininIV[52]
E-glycoproteinHSV[51]
HTL-40Halimeda renschiiManhemagglutininIV[76]
KAA-2Kappaphycus alvareziiManhemagglutininIV[77]
gp120HIV[78]
MELMeristiella echinocarpaMannanhemagglutininIV[52]
hemagglutininIV[52]
SfLSolieria filiformisMannanE-glycoproteinHSV[52]
gp120HIV[52]
hemagglutininIV[52]
BCABoodlea coactaManE-glycoproteinHSV[79]
hemagglutininIV[79]
LectinCyanobacterial SpeciesCarbohydrate-
Binding
Specificity
Targeted Envelope
Protein
VirusRef.
MVNMicrocystis aeruginosaMangp120HIV[80,81]
E-glycoproteinHSV[81]
MVLMicrocystis viridisMangp120HIV[82]
E-glycoproteinHCV[83]
Cyanovirin-NNostoc ellipsosporumMangp120HIV[49,84]
(CV-N) E-glycoprotein 1,2EBOV[85,86]
hemagglutininIV[86]
E-glycoproteinHCV[87]
E-glycoproteinHSV[88]
S-proteinSARS-CoV-2[89]
Oscillatoria agardhiiMangp120HIV[90]
OAAScytonema variumMangp120HIV[91]
SVNScytonema varium E-glycoproteinDENV[92]
E-glycoproteinEBOV[93]
Although cyanobacterial lectins exhibit similar antiviral activities against pathogenic enveloped viruses, compared to other lectins from higher plants, they readily differ by the different fold and the smaller size of their structural scaffolds [93,94]. Moreover, cyanobacteria contain other small metabolites that could be used as valuable tools for combating enveloped viruses and, especially SARS-CoV-2 responsible for the COVID-19 pandemic [95].

5. How Can the Infectivity of Pathogenic Enveloped Viruses Be Affected by Lectins?

The glycan-mediated interaction of lectins with pathogenic enveloped viruses has a direct effect on virus infectivity, essentially by interfering with the recognition of their corresponding host cell receptors via different mechanisms. However, a dichotomy must be introduced between experiments performed in vitro on cultured cells and experiments achieved in vivo on animals.
Experimental studies performed in vitro, especially on HIV-infected cultured cells, have focused on higher plant lectins and cyanobacterial lectins [96]. Mannose-specific lectins have been recognized as the most efficient inhibitors of the HIV entry into the target cells by interacting with the glycan shield of gp120 and gp41, preventing their recognition by the CD4 receptors present on CD4+ T cells. Mannose-specific lectins of monocot plant species, such as Cymbidium hybrid (CHA), Epipactis helleborine (EHA), Hippeastrum hybrid (HHA), Galanthus nivalis (GNA), Listera ovata (LOA), and Narcissus pseudonarcissus (NPA), have been widely investigated in this respect by Balzarini and co-workers [53,54]. Other lectins with similar Man-binding specificity like BanLec from Musa acuminata [43] and Con A from Canavalia ensiformis [97] or different carbohydrate-binding specificities, such as the (GlcNAc)n-specific lectins Nictaba from Nicotiana tabacum [62], UDA from Urtica dioica [53], and WGA from Triticum aestivum [98], were also identified as potential inhibitors of the HIV entry into target cells in vitro and the syncytium formation resulting from the fusion of HIV-infected and HIV-uninfected CD4+ T lymphocytes [99,100].
A rather different situation can occur under in vivo conditions due to the multiplicity of cells susceptible to interacting with the virus. In addition, to block the entry of HIV particles into the cells and syncytium formation, plant lectins interfere with other mechanisms of virus infection and transmission, for example, by preventing the recognition of high-mannose glycans of gp120 by the DC-SIGN receptor of dendritic cells [101,102,103] or by blocking the transmission of DC-SIGN-captured virions to the CD4+ T lymphocytes [102]. In addition, as reported in [96], interaction with lectins of different carbohydrate-binding specificities can result in cytotoxic side effects on host cells, e.g., caspase-dependent apoptotic responses, due to the activation of different signaling pathways leading to apoptotic and necrotic responses that result from the recognition of surface-exposed N- and O-glycans by lectins.
The effects of plant lectins on other pathogenic enveloped viruses have been more scarcely investigated. Plant lectins were identified as blocking agents for the entry of IV [45,52,57], HSV [57], HCMV [47,53,54], EBOV [47,48], LASV [47], and the coronaviruses MERS-CoV [51], SARS-CoV [42,51,54,62,63,104], and SARS-CoV-2 [50,51,52,58,60,61,66] in their corresponding host cells. However, depending on the viruses, the envelope glycoprotein(s) targeted by plant lectins are extremely diverse as mentioned in Table 2. In this respect, an engineered banana lectin, BanLec, which has lost its mitogenic potential but retained its mannose-binding property, interacted with the envelope E-glycoprotein and inhibited both the entry and replication of Ebola virus in cell cultures [47,48]. The cyanobacterial lectin, cyanovirin-N (CV-N), also displayed similar inhibition towards Ebola virus [85]. Similarly, lentil lectin, LcA, inhibited the early steps of the host cell infection by SARS-CoV-2 and variants B.1.1.7 (α variant), B.1.351 (β variant), and P1 (γ variant), by blocking the recognition of their spike S protein by the ACE2 receptor [51].
The effects resulting from the binding of plant lectins on the different enveloped viruses also depend on the mechanisms of infection and transmission of these viruses, which may differ from those developed by HIV. In addition to preventing the entry and replication of Ebola viruses into the host cells in cell cultures, the engineered BanLec lectin, pre-administered to virus-infected mice, were highly protective against a lethal EBOV infection in vivo (~80% of mice protected) [48]. The engineered BanLec lectin was similarly efficient for protecting influenza virus-infected mice, by inhibiting the virus-endosome fusion occurring after the exogenous lectin has been internalized in the late endosomal/lysosomal compartment of the host cells [46]. Both plant lectins and griffithsin, the Man-specific lectin from the red alga Griffithsia sp., also inhibited the entry of SARS-CoV and MERS-CoV, respectively, in virus-infected cultured cells [42,74]. The Neu5Ac-specific Maackia amurensis lectin, MAL, inhibited the interaction of the SARS-CoV-2 S protein with the ACE2 receptor in cell cultures and decreased the expression of inflammatory mediators associated with COVID-19 disease progression [61].
The effects of plant lectins on coronaviruses were also investigated in virus-infected mice models. The (GlcNAc)n-specific lectin from the stinging nettle (Urtica dioica) (UDA), was shown to prevent virus entry and replication in a dose-dependent manner and reduced the virus infectivity significantly in a lethal SARS-CoV BALB/c mouse model [105]. Under in vivo conditions, the lectin from the hyacinth bean (Lallab purpureus), FRIL, neutralized H1N1 influenza by aggregating and trapping virions in the late endosomes of the host cells, thus preventing their nuclear internalization [60]. The lectin similarly neutralized SARS-CoV-2 by preventing viral protein production and cytotoxic effects on the host cells.
Algal and cyanobacterial Man-specific lectins, such as griffithsin and cyanovirin-N, also inhibited the entry of HIV and other enveloped viruses in the host cells in vitro and exerted in vivo cytotoxic effects very similar to those of plant lectins [74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,105,106,107].

6. Biomedical Perspectives for Antiviral Lectins

Depending on their affinity towards surface-exposed glycans of enveloped viruses, plant lectins are considered as potential CBAs useful for combating viral infections, even though little evidence exists to date for their efficacy as relevant therapeutic tools [97,108,109,110,111,112]. Beyond their possible use as well-adapted tools for the diagnosis of viral infection, the therapeutic use of plant lectins as virus blockers faces practical and functional challenges which mainly concern (1) their large-scale production and (2) their unwanted immunomodulatory properties.
In most higher plants, lectins of different specificities that could be used as virus blockers occur as storage proteins in seeds and other vegetative organs such as tubers and rhizomes [113]. Man-specific two-chain (LcA, PsA, VfA, and LoL-I/II) and single-chain lectins (Con A, PHA, and SBA) from the Fabaceae are sequestered in the protein bodies of the cotyledonary cells in rather low amounts [114]. Accordingly, the extraction yield of legume seed lectins is rather low, in the range 50–80 mg/100 g (dry weight) seed [115]. However, the degree of purity of the extracted lectins is excellent since the introduction of affinity chromatography techniques using carbohydrate-immobilized columns. Different strategies have been developed recently to improve the extraction yield of griffithsin, the Man-specific lectin from the red alga Griffithsia sp., for the purpose of obtaining a large-scale production of the lectin able to supply the quantities of lectins needed for therapeutic applications [116,117,118,119,120,121]. These strategies are based on the continuous improvement of yields obtained from the high-level expression and extraction of griffithsin from transformed tobacco (Nicotiana benthamiana) leaves. In addition, griffithsin is easily purified and recovered from ensiled dried tobacco leaves, which allows for a low-cost production of lectin quickly adaptable to demand.
Most plant lectins consist of oligomeric structures built up from the non-covalent association of 15–20 kDa monomers in dimers and tetramers, more rarely in hexamers or octamers [31]. Depending on their structural organization, plant lectins usually exhibit a high degree of resistance to the degradation by trypsin-like proteases together with a pronounced capacity to trigger the synthesis of specific anti-lectin IgG. In this respect, IgG-binding epitopes have been identified on the molecular surface of Man-specific two-chain lectins from the Vicieae tribe [122,123], and monoclonal antibodies that specifically recognize lentil and Lathyrus ochrus lectins, were easily prepared [124,125]. Even Man-specific dietary lectins, such as BanLec from banana and ASA from garlic, have been reported to induce an immune response since specific anti-lectin IgG have been identified in the serum of banana and garlic consumers [126,127]. Through a specific interaction with the plant lectins associated to the enveloped viruses, these IgG could eventually neutralize the effects of lectins in the host cells. The Man-specific algal griffithsin and cyanobacterial lectins, such as cyanovirin-N and scytovirin, could overcome this challenge because of the small size of their composing monomers [128,129]. In addition to griffithsin, grifonin-1 (GRFN-1), an even smaller peptide of eighteen amino acids derived from griffithsin, has proven its efficacy as a blocking agent against HIV [130].
Initially recognized as potent mitogenic proteins [131,132,133], plant lectins have been known for a long time as non-specific immune-modulatory proteins that are susceptible to interaction with various cell surface glycoproteins/glycolipids and for interfering with various signaling pathways triggering cytotopathologic effects on the targeted cells. Although most plant lectins with antiviral activity activate different sets of T lymphocytes and, more scarcely, B lymphocytes, they also activate both the apoptotic and necrotic pathways in many other types of healthy and cancer cells [134,135,136,137,138,139,140,141,142,143]. The cellular activation mediated by plant lectins on healthy and transformed cells elicits the release of various chemokines and/or cytokines that are, in turn, susceptible to interfere with the cytokine stimulation associated with the viral infection, e.g., HIV infection [59]. However, plant lectins readily differ from each other by their capacity to elicit a cytokine production, some of them, such as PHA, Con A, and cyanovirin-N, being more active to induce the synthesis and release of activation markers [144,145], while Man-specific GNA-like lectins, such as GNA and HHA, were virtually incapable of triggering a relevant cytokine production [145,146]. Recently, a single-point mutation performed on an engineered banana lectin, BanLec, and an engineered Malaysian banana lectin, Malay BanLec, allowed to produce an active Man-binding lectin significantly devoid of mitogenic/cytotoxic activity [46,47]. If applicable for other Man-specific lectins, this point mutation approach would be an elegant way to attenuate or suppress the unwanted mitogenic/cytotoxic effects of lectins on target cells.
In spite of these limitations hampering the use of plant lectins as CBAs for combating pathogenic enveloped viruses, some ex vivo applications of plant lectins have been successfully developed. An important decrease in the plasma load with Ebola virus was achieved by extracorporeal affinity plasmapheresis of the contaminated blood through a GNA-immobilized matrix [147]. Recently, ex-vivo plasmapheresis on a Man-specific lectin-immobilized column of blood spoiled by the MERS-CoV and the Marburg viruses has proven its efficacity to purge the blood samples from virus particles [148]. Although essentially theoretical, the risk of a possible transfusion transmission of SARS-CoV-2 with spoiled blood samples should be avoided by a simple lectin plasmapheresis step of the suspected blood samples [149,150]. Another ex vivo application of plant lectins has been proposed on the web (Pittsburgh University, 2020) in the form of a nasal spray of griffithsin that could be used to prevent the infection by SARS-CoV-2 and other pathogenic enveloped viruses, e.g., in immune-compromised people. A lectin spray could also be used to detect the enveloped viruses on various domestic surfaces, such as doorknobs, handrails, computers, and cooking tools, under UV illumination after labeling with specific anti-lectin antibodies coupled to a fluorochrome.

7. Bioinformatics

Atomic coordinates of fusion proteins, B glycoproteins, and E glycoproteins were taken from the Protein Data Bank (PDB): 7JPH (EBOV) [5], 6Y5G (IV) [6], 5CXF (HCMV) [7], 2GUM (HSV) [8], 4TVP (HIV) [9], 5VK2 (LASV) [10], 5W9H (MERS-CoV) [11], 6ACD (SARS-CoV) [12], 6VXX (SARS-CoV-2) [13], 3N40 (CHIV) [14], 1UZG (DENV) [15], and 7BUB (ZIV) [16].
The molecular surface of the lectins and envelope glycoproteins from pathogenic enveloped viruses were calculated and displayed with Chimera [151] and Chimera-X [152]. Assuming that putative N-glycosylation sites, NXT/S, of envelope glycoproteins are actually glycosylated, a classic N-glycan chain corresponding to the trimannoside core Man3GlcNAc2, was modeled using the GlyProt server (http://www.glycosciences.de/modeling/glyprot/php/main.php) (accessed on 22 December 2021) [153] and represented in CPK on the molecular surface of the envelope glycoproteins.
The illustrations of the high-mannose N-glycans, complex N-glycans, hybrid N-glycans, and O-glycans were built and represented with the DrawGlycan SNFG package for Mac [154]. Colored symbols were used to represent Fuc (red triangle), Gal (yellow circle), Glc (blue circle), GalNAc (yellow square), GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond).

8. Discussion

The glycan shield covering pathogenic enveloped viruses plays a role not only in the protection of viruses but also in various important mechanisms insuring the entry and replication of viruses in the host cells [30]. Thus, the recognition of the glycan shield by plant lectins provides a way to fight viral infection and, especially, the SARS-CoV-2 infection, by competing with the spike-mediated attachment of viral particles to the host cell virus receptors. However, due to the extreme diversity of N-glycan types covering enveloped viruses, especially beta-coronaviruses [15], plant lectins with different carbohydrate-binding specificities should be tested for this purpose. From experiments performed under in vitro and in vivo conditions, it follows that plant, algal, and cyanobacterial lectins with different carbohydrate-binding specificities represent well-adapted CBAs for blocking the entry of pathogenic enveloped viruses into the host cells. In this respect, Man-specific lectins of the Vicieae tribe, which specifically recognize the α1,6-fucosylated Man3GlcNAc2 core of N-glycans of the complex- and hybrid-type, are particularly relevant as glycan probes for the beta-coronaviruses MERS-CoV, SARS-CoV, and SARS-CoV-2 [51,106]. However, Man-specific lectins are not considered as replication blockers for coronaviruses, since they do not interfere with the coronavirus replication within the cell.
Despite the accumulating evidences that plant lectins and, especially, Man-specific plant lectins, could be used as tools for preventing infection by pathogenic enveloped virus, in particular SARS-CoV-2 responsible for the COVID-19 pandemic, some unwanted characteristics of plant lectins make these molecules difficult to use for a therapeutic purpose. Due to the fact of their high molecular size, which favors the synthesis of anti-lectin antibodies, and their mitogenic/cytotoxic properties, which interfere with the cytokine response of infected individuals, their use is limited to external treatments. However, the promising results obtained with a single-mutated engineered banana lectin, BanLec, which retains its carbohydrate-binding ability but loses its mitogenic property [46,47], could pave the way for the forthcoming production of innocuous mutated plant lectins available for a therapeutic use.
In addition to lectins, other small molecules could be used as blockers for the SARS-CoV-2/ACE2 interaction. Recently, some small molecular drugs, including dyes, glycosides, tannins, and immunosuppressors, were characterized as either spike or ACE2 binders susceptible to blocking the attachment of the SARS-CoV-2 spikes to ACE2 by interfering with the ligand and/or the receptor surface [155]. Depending on the N-glycan types that are linked to the receptor DPP4 for MERS-CoV and SARS-CoV viruses [156] and ACE2 for SARS-CoV-2 virus [157], plant lectins with Man-binding activity could interfere with the capture of the beta-coronavirus spikes by their corresponding host cell receptors. This dual activity of plant lectins towards the glycans of spikes and their receptors is of paramount importance for reinforcing the antiviral properties of plant lectins against pathogenic beta-coronaviruses.

Author Contributions

Conceptualization, P.R.; software, P.R.; validation, E.J.M.V.D. and P.R.; resources, A.B., B.K., M.S., J.S. and H.B.; writing—original draft preparation, P.R.; writing—review and editing, E.J.M.V.D. and P.R.; supervision, E.J.M.V.D. and P.R.; project administration, A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rey, F.A.; Lok, S.-M. Common features of enveloped viruses and implications for immunogen design for next-generation vaccines. Cell 2018, 172, 1319–1334. [Google Scholar] [CrossRef] [PubMed]
  2. Thorley, J.A.; McKeating, J.A.; Rappoport, J.Z. Mechanisms of viral entry: Seaking in the front door. Protoplasma 2010, 244, 15–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Heldwein, E.E.; Lou, H.; Bender, F.C.; Cohen, G.H.; Eisenberg, R.J.; Harrison, S.C. Crystal structure of glycoprotein B from herpes simplex virus 1. Science 2006, 313, 217–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Voss, J.E.; Vaney, M.-C.; Duquerroy, S.; Vonrhein, C.; Girard-Blanc, C.; Crublet, E.; Thompson, A.; Bricogne, G.; Rey, F.A. Glycoprotein organization of Chikungunya virus particles revealed by X-ray crystallography. Nature 2010, 468, 707–712. [Google Scholar] [CrossRef] [PubMed]
  5. Modis, Y.; Ogata, S.; Clements, D.; Harrison, S.C. Variable surface epitopes in the crystal structure of dengue virus type 3 envelope glycoprotein. J. Virol. 2005, 79, 1223–1231. [Google Scholar] [CrossRef] [Green Version]
  6. Zhang, S.; Loy, T.; Ng, T.S.; Lim, X.-N.; Chew, S.V.; Tan, T.Y.; Xu, M.; Kostyuchenko, V.A.; Tukijan, F.; Shi, J.; et al. A human antibody neutralizes different flaviviruses by using different mechanisms. Cell Rep. 2020, 31, 107584. [Google Scholar] [CrossRef] [PubMed]
  7. He, L.; Chaudhary, A.; Lin, X.; Sou, C.; Alkutkar, T.; Kumar, S.; Ngo, T.; Kosviner, E.; Ozorowski, G.; Stanfield, R.L.; et al. Single-component multilayer self-assembling nanoparticles presenting rationally designed glycoprotein trimers as Ebola virus vaccines. Nat. Commun. 2021, 12, 2633. [Google Scholar] [CrossRef] [PubMed]
  8. Benton, D.J.; Gamblin, S.; Rosenthal, P.B.; Skehel, J. Structural transitions in influenza haemagglutinin at membrane fusion pH. Nature 2020, 583, 150–153. [Google Scholar] [CrossRef] [PubMed]
  9. Burke, H.G.; Heldwein, E.E. Crystal structure of the Human cytomegalovirus glycoprotein B. PLoS Pathog. 2015, 11, e1005227. [Google Scholar] [CrossRef] [Green Version]
  10. Pancera, M.; Zhou, T.; Druz, A.; Georgiev, I.S.; Soto, C.; Gorman, J.; Huang, J.; Acharya, P.; Chuang, G.-Y.; Ofek, G.; et al. Structure and immune recognition of trimeric prefusion HIV-1 Env. Nature 2014, 514, 455–461. [Google Scholar] [CrossRef] [Green Version]
  11. Hastie, K.; Zandonatti, M.A.; Kleinfelter, L.M.; Heinrich, M.L.; Rowland, M.M.; Chandran, K.; Branco, L.M.; Robinson, J.E.; Garry, R.F.; Saphire, E.O. Structural basis for antibody-mediated neutralization of Lassa virus. Science 2017, 356, 923–928. [Google Scholar] [CrossRef] [Green Version]
  12. Pallesen, J.; Wang, N.; Corbett, K.S.; Wrapp, D.; Kirchdoerfer, R.N.; Turner, H.L.; Cottrell, C.A.; Becker, M.M.; Wang, L.; Shi, W.; et al. Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen. Proc. Natl. Acad. Sci. USA 2017, 114, E7348–E7357. [Google Scholar] [CrossRef] [Green Version]
  13. Song, W.; Gui, M.; Wang, X.; Xiang, Y. Cryo-EM structure of the SARS coronavirus spike glycoprotein in complex with its host cell receptor ACE2. PLoS Pathog. 2018, 14, e1007236. [Google Scholar] [CrossRef]
  14. Walls, A.C.; Park, Y.-J.; Tortorici, M.A.; Wall, A.; McGuire, A.T.; Veesler, D. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell 2020, 181, 281–292. [Google Scholar] [CrossRef] [PubMed]
  15. Chon, B.G.; Gautam, S.; Peng, W.; Huang, Y.; Goli, M.; Mechref, Y. Direct comparison of N-glycans and their isomers derived from spike glycoprotein 1 of MERS-CoV, SARS-CoV, and SARS-CoV-2. J. Proteom. Res. 2021, 20, 4357–4365. [Google Scholar]
  16. Shajahan, A.; Supekar, N.T.; Gleinich, A.S.; Azadi, P. Deducing the N- and O-glycosylation profile of the spike protein of novel coronavirus SARS-CoV-2. Glycobiology 2020, 30, 981–988. [Google Scholar] [CrossRef] [PubMed]
  17. Bagdonaite, I.; Thompson, A.J.; Wang, X.; Søgaard, M.; Fougeroux, C.; Frank, M.; Diedrich, J.K.; Yates III, J.R.; Salanti, A.; Vakhrushev, S.Y.; et al. Site-specific O-glycosylation analysis of SARS-CoV-2 spike protein produced in insect and Human cells. Viruses 2021, 13, 551. [Google Scholar] [CrossRef]
  18. Van Damme, E.J.M. 35 years in plant lectin research: A journey from basic science to applications in agriculture and medicine. Glycoconj. J. 2021, in press. [Google Scholar] [CrossRef] [PubMed]
  19. Collar, A.L.; Clarke, E.C.; Anaya, E.; Merrill, D.; Yarborough, S.; Anthony, S.M.; Kuhn, J.H.; Merle, C.; Theisen, M.; Bradfute, S.B. Comparison of N- and O-linked glycosylation patterns of ebolavirus glycoproteins. Virology 2017, 502, 39–47. [Google Scholar] [CrossRef]
  20. Luo, S.; Hu, K.; He, S.; Wang, P.; Zhang, M.; Huang, X.; Du, T.; Zheng, C.; Liu, Y.; Hu, Q. Contribution of N-linked glycans on HSV-2 gG to cell-cell fusion and viral entry. Virology 2015, 483, 72–82. [Google Scholar] [CrossRef] [Green Version]
  21. Smargiasso, N.; Nader, J.; Rioux, S.; Mazzucchelli, G.; Boutry, M.; De Pauw, E.; Chaumont, F.; Navarre, C. Exploring the N-glycosylation profile of glycoprotein B from Human cytomegalovirus expressed in CHO and Nicotiana tabacum BY-2 cells. Int. J. Mol. Sci. 2019, 20, 3741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Behrens, A.-J.; Vasiljevic, S.; Pritchard, L.K.; Harvey, D.J.; Andev, R.S.; Krumm, S.A.; Struwe, W.B.; Cupo, A.; Kumar, A.; Zitzmann, N.; et al. Composition and antigenic effects of individual glycan sites of a trimeric HIV-1 envelope glycoprotein. Cell Resp. 2016, 14, 2695–2706. [Google Scholar] [CrossRef] [Green Version]
  23. Li, J.; Liu, S.; Gao, Y.; Tian, S.; Yang, Y.; Ma, N. Comparison of N-linked glycosylation on hemagglutinins derived from chicken embryos and MDCK cells: A case of the production of a trivalent seasonal influenza vaccine. Appl. Microbiol. Biotechnol. 2021, 105, 3559–3572. [Google Scholar] [CrossRef]
  24. Lancaster, C.; Pristatsky, P.; Van Hoang, M.; Casimiro, D.R.; Schwartz, R.M.; Rustandi, R.; Ha, S. Characterization of N-glycosylation profiles from mammalian and insect cell derived chikungunya VLP. J. Chromatogr. B Analyt. Technol. Life Sci. 2016, 1032, 218–223. [Google Scholar] [CrossRef] [PubMed]
  25. Watanabe, Y.; Raghwani, J.; Allen, J.D.; Seabright, G.E.; Li, S.; Moser, F.; Huiskonen, J.T.; Strecker, T.; Bowden, T.A.; Crispin, M. Structure of the Lassa virus glycan shield provides a model for immunological resistance. Proc. Natl. Acad. Sci. USA 2018, 115, 7320–7325. [Google Scholar] [CrossRef] [Green Version]
  26. Pralow, A.; Nikolay, A.; Leon, A.; Genzel, Y.; Rapp, E.; Reich, U. Site-specific N-glycosylation analysis of animal cell culture-derived Zika virus proteins. Sci. Rep. 2021, 11, 5147. [Google Scholar] [CrossRef] [PubMed]
  27. Watanabe, Y.; Allen, J.D.; Wrapp, D.; McLellan, J.S.; Crispin, M. Site-specific glycan analysis of the SARS-CoV-2 spike. Science 2020, 369, 330–333. [Google Scholar] [CrossRef] [PubMed]
  28. Hatmal, M.M.; Alshaer, W.; Al-Hatamleh, M.A.I.; Hatmal, M.; Smadi, O.; Taha, M.O.; Oweida, A.J.; Boer, J.C.; Mohamud, R.; Plebanski, M. Comprehensive structural and molecular comparison of spike proteins of SARS-CoV-2, SARS-CoV and MERS-CoV, and their interactions with ACE2. Cells 2020, 9, 2638. [Google Scholar] [CrossRef] [PubMed]
  29. Zhao, P.; Praissman, J.L.; Grant, O.C.; Xiao, T.; Rosenbalm, K.E.; Aoki, K.; Kellman, B.P.; Bridger, R.; Barouch, D.H.; Brindley, M.A.; et al. Virus-receptor interactions of glycosylated SARS-CoV-2 spike and Human ACE2 receptor. Cell Host Microbe 2020, 28, 586–601. [Google Scholar] [CrossRef] [PubMed]
  30. Li, Y.; Liu, D.; Wang, Y.; Su, W.; Liu, G.; Dong, W. The importance of glycans of viral and host proteins in enveloped virus infection. Front. Immunol. 2021, 12, 638573. [Google Scholar] [CrossRef]
  31. Peumans, W.J.; Van Damme, E.J.M.; Barre, A.; Rougé, P. Classification of plant lectins in families of structurally and evolu- tionary related proteins. Adv. Exp. Med. Biol. 2001, 491, 27–54. [Google Scholar] [PubMed]
  32. Debray, H.; Decout, D.; Strecker, G.; Spik, G.; Montreuil, J. Specificity of twelve lectins towards oligosaccharides and glyco peptides related to N-glycosylproteins. Eur. J. Biochem. 1981, 117, 41–55. [Google Scholar] [CrossRef]
  33. Debray, H.; Rougé, P. The fine sugar specificity of the Lathyrus ochrus seed lectin and isolectins. FEBS Lett. 1984, 176, 120–124. [Google Scholar] [CrossRef] [Green Version]
  34. Einspahr, H.; Pareks, E.H.; Suguna, K.; Subramanian, E.; Suddath, F.L. The crystal structure of pea lectin at 3.0-Å resolution. J. Biol. Chem. 1986, 261, 16518–16527. [Google Scholar] [CrossRef]
  35. Foriers, A.; Van Driessche, E.; De Neve, R.; Kanarek, L.; Strosberg, A.D. The subunit structure and N-terminal sequences of theα- and β-subunits of the lentil lectin (Lens culinaris). FEBS Lett. 1977, 75, 237–240. [Google Scholar] [CrossRef] [Green Version]
  36. Bourne, Y.; Abergel, C.; Cambillau, C.; Frey, M.; Rougé, P.; Fontecilla-Camps, J.C. X-ray crystal structure determination and refinement at 1.9 Å resolution of isolectin I from the seeds of Lathyrus ochrus. J. Mol. Biol. 1990, 214, 571–584. [Google Scholar] [CrossRef]
  37. Reeke, G.N., Jr.; Becker, J.W. Three-dimensional structure of favin: Saccharide binding-cyclic permutation in leguminous lectins. Science 1986, 234, 1108–1111. [Google Scholar] [CrossRef] [PubMed]
  38. Hardman, K.D.; Ainsworth, C.F. Structure of concanavalin A at 2.4-Å resolution. Biochemistry 1972, 11, 4910–4919. [Google Scholar] [CrossRef]
  39. Bourne, Y.; Mazurier, J.; Legrand, D.; Rougé, P.; Montreuil, J.; Spik, G.; Cambillau, C. Structures of a legume lectin complexed with the human lactotransferrin N2 fragment, and with an isolated biantennary glycopeptide: Role of the fucose moiety. Structure 1994, 15, 209–219. [Google Scholar] [CrossRef] [Green Version]
  40. Bourne, Y.; Rougé, P.; Cambillau, C. X-ray structure of (α-Man(1-3)β-Man(1-4)GlcNAc)-lectin complex at 2.1-Å resolution. The role of water in sugar-lectin interaction. J. Biol. Chem. 1990, 265, 18161–18165. [Google Scholar] [CrossRef]
  41. Rougé, P.; Peumans, W.J.; Van Damme, E.J.M.; Barre, A.; Singh, T.; Wu, J.H.; Wu, A.M. Glycotope structures and intramole-cular affinity factors of plant lectins for Tn/T antigens. Adv. Exp. Med. Biol. 2011, 705, 143–154. [Google Scholar] [PubMed] [Green Version]
  42. Keyaerts, E.; Vijgen, L.; Pannecouque, C.; Van Damme, E.; Peumans, W.; Egberink, H.; Balzarini, J.; Van Ranst, M. Plant lectins are potent inhibitors of coronaviruses by interfering with two targets in the viral replication cycle. Antivir. Res. 2007, 75, 179–187. [Google Scholar] [CrossRef]
  43. Swanson, M.D.; Winter, H.C.; Goldstein, I.J.; Markovitz, D.M. A lectin isolated from banana is a potent inhibitor of HIV replication. J. Biol. Chem. 2010, 285, 8646–8655. [Google Scholar] [CrossRef] [Green Version]
  44. Swanson, M.D.; Boudreaux, D.M.; Salmon, L.; Clugh, J.; Winter, H.C.; Meagher, J.L.; André, S.; Murphy, P.V.; Oscarson, S.; Roy, R.; et al. Engineering a therapeutic lectin by uncoupling mitogenicity from antiviral activity. Cell 2015, 163, 746–758. [Google Scholar] [CrossRef] [Green Version]
  45. Mazalovska, M.; Kouokam, J.C. Lectins as promising therapeutics for the prevention and treatment of HIV and other potential coinfections. BioMed. Res. Int. 2018, 8, 3750646. [Google Scholar] [CrossRef]
  46. Covés-Datson, E.M.; King, S.R.; Legendre, M.; Gupta, A.; Chan, S.M.; Gitlin, E.; Kulkarni, V.V.; Pantaleón García, J.; Smee, D.F.; Lipka, E.; et al. A molecularly engineered antiviral banana lectin inhibits fusion and is efficacious against influenza virus infection in vivo. Proc. Natl. Acad. Sci. USA 2020, 117, 2122–2132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Covés-Datson, E.M.; King, S.R.; Legendre, M.; Swanson, M.D.; Gupta, A.; Claes, S.; Meagher, J.L.; Boonen, A.; Zhang, L.; Kalveram, B.; et al. Targeted disruption of pi-pi stacking in Malaysian banana lectin reduces mitogenicity while preserving antiviral activity. Sci. Rep. 2021, 11, 656. [Google Scholar] [CrossRef]
  48. Covés-Datson, E.M.; Dyall, J.; DeWald, L.E.; King, S.R.; Dube, D.; Legendre, M.; Nelson, E.; Drews, K.C.; Gross, R.; Gerhardt, D.M.; et al. Inhibition of Ebola virus by a molecularly engineered banana lectin. PLoS Negl. Trop. Dis. 2019, 13, e0007595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Witvrouw, M.; Fikkert, V.; Hantson, A.; Pannecouque, C.; O’Keefe, B.R.; McMahon, J.; Stamatatos, L.; de Clercq, E.; Bolmstedt, A. Resistance to human immunodeficiency virus type 1 to the high-mannose binding agents cyanovirin N and concanavalin A. J. Virol. 2005, 79, 7777–7784. [Google Scholar] [CrossRef] [Green Version]
  50. Jang, H.; Lee, D.-H.; Kang, H.G.; Lee, S.J. Concanavalin A targeting N-linked glycans in spike proteins influence viral interactions. Dalton Trans. 2020, 49, 13538–13543. [Google Scholar] [CrossRef]
  51. Wang, W.; Li, Q.; Wu, J.; Hu, Y.; Wu, G.; Yu, C.; Xu, K.; Liu, X.; Wang, Q.; Huang, W.; et al. Lentil lectin from Lens culinaris exhibit broad antiviral activities against SARS-CoV-2 variants. Emerg. Microbes Infect. 2021, 10, 1519–1529. [Google Scholar] [CrossRef] [PubMed]
  52. Gondim, A.C.S.; da Silva, S.R.; Mathys, L.; Noppen, S.; Liekens, S.; Sampaio, A.H.; Nagano, C.S.; Costa Rocha, C.R.; Nasci Mento, K.S.; Cavada, B.S.; et al. Potent antiviral activity of carbohydrate-specific algal and leguminous lectins from the Brazilian biodiversity. Med. Chem. Commun. 2019, 10, 390–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Balzarini, J.; Neyts, J.; Schols, D.; Hosoya, M.; Van Damme, E.; Peumans, W.; De Clercq, E. The mannose-specific plant lectins from Cymbidium hybrid and Epipactis helleborine and the (N-acetylglucosamine)n-specific plant lectin from Urtica dioica are potent and selective inhibitors of human immunodeficiency virus and cytomegalovirus replication in vitro. Antivir. Res. 1992, 18, 191–207. [Google Scholar]
  54. Balzarini, J.; Schols, D.; Neyts, J.; Van Damme, E.; Peumans, W.; De Clercq, E. Alpha-(1-3)- and alpha-(1-6)-D-mannose-specific plant lectins are markedly inhibitory to human immunodeficiency virus and cytomegalovirus infections in vitro. Antimicrob. Agents Chemother. 1991, 35, 410–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Bertaux, C.; Daelemans, D.; Meertens, L.; Cormier, E.G.; Reinus, J.F.; Peumans, W.J.; Van Damme, E.J.M.; Igarashi, T.; Oki, T.; Schols, D.; et al. Entry of hepatitis C virus and human immunodeficiency virus is selectively inhibited by carbohydrate-binding agents but not by polyanions. Virology 2007, 366, 40–50. [Google Scholar] [CrossRef]
  56. Ashfaq, U.; Masoud, M.; Khaliq, S.; Nawaz, Z.; Riazuddin, S. Inhibition of hepatitis C virus 3a genotype entry through Galanthus nivalis agglutinin. Virol. J. 2011, 8, 248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Luo, Y.; Xu, X.; Liu, J.; Li, J.; Sun, Y.; Liu, Z.; Liu, J.; Van Damme, E.; Balzarini, J.; Bao, J. A novel mannose-binding tuber lectin from Typhonium divaricatum (L.) Decne (family Araceae) with antiviral activity against HSV-II and anti-proliferative effect on human cancer cell lines. J. Biochem. Mol. Biol. 2007, 40, 358–367. [Google Scholar] [CrossRef]
  58. Amundson, D.E.; Shah, U.; de Necochea-Campion, R.; Jacobs, M.; LaRosa, S.P.; Fisher, C.J., Jr. Removal of COVID-19 spike protein, whole virus, exosomes, and exosomal microRNAs by the Hemopurifier® lectin-affinity cartridge in critically III patients with COVID-19 infection. Front. Med. 2021, 8, 744141. [Google Scholar] [CrossRef] [PubMed]
  59. Jayaprakash, N.G.; Singh, A.; Vivek, R.; Yadav, S.; Pathak, S.; Trivedi, J.; Jayaraman, N.; Nandi, D.; Mitra, D.; Surolia, A. The barley lectin, horcolin, binds high-mannose glycans in a multivalent fashion, enabling high-affinity, specific inhibition of cellular HIV infection. J. Biol. Chem. 2020, 295, 12111–12129. [Google Scholar] [CrossRef]
  60. Liu, Y.-M.; Shahed-Al-Mahmud, M.D.; Chen, X.; Chen, T.-H.; Liao, K.-S.; Lo, J.M.; Wu, Y.-M.; Ho, M.-C.; Wu, C.-Y.; Wong, C.-H.; et al. A carbohydrate-binding protein from the edible lallab beans effectively blocks the infections of influenza viruses and SARS-CoV-2. Cell Rep. 2020, 32, 108016. [Google Scholar] [CrossRef]
  61. Sheehan, S.A.; Hamilton, K.L.; Retzbach, E.P.; Balachandran, P.; Krishnan, H.; Leone, P.; Lopez-Gonzalez, M.; Suryavanshi, S.; Kumar, P.; Russo, R.; et al. Evidence that Maackia amurensis seed lectin (MASL) exerts pleiotropic actions on oral squamous cells with potential to inhibit SARS-CoV-2 infection and COVID-19 disease progression. Exp. Cell Res. 2021, 403, 112594. [Google Scholar] [CrossRef] [PubMed]
  62. Gordts, S.C.; Renders, M.; Férir, G.; Huskens, D.; Van Damme, E.J.M.; Peumans, W.; Balzarini, J.; Schols, D. NICTABA and UDA, two GlcNAc-binding lectins with unique antiviral activity profiles. J. Antimicrob. Chemother. 2015, 70, 1674–1685. [Google Scholar] [CrossRef] [Green Version]
  63. Al Atalah, B.; Fouquaert, E.; Vanderschaeghe, D.; Proost, P.; Balzarini, J.; Smith, D.F.; Rougé, P.; Lasanajak, Y.; Callewaert, N.; Van Damme, E.J.M. Expression analysis of the nucleocytoplasmic lectin ‘Orysata’ from rice in Pichia pastoris. FEBS J. 2011, 278, 2064–2079. [Google Scholar] [CrossRef] [PubMed]
  64. An, J.; Liu, J.-Z.; Wu, C.-F.; Li, J.; Dai, L.; Van Damme, E.; Balzarini, J.; De Clercq, E.; Chen, F.; Bao, J.-K. Anti-HIV I/II activity and molecular cloning of a novel mannose/sialic acid-binding lectin from rhizome of Polygonatum cyrtonema Hua. Acta Biochim. Biophys. Sin. 2006, 38, 70–78. [Google Scholar] [CrossRef] [Green Version]
  65. Vanderlinden, E.; Van Winkel, N.; Naesens, L.; Van Damme, E.J.M.; Persoons, L.; Schols, D. In vitro characterization of the carbohydrate-binding agents HHA, GNA, and UDA as inhibitors of influenza A and B virus replication. Antimicrob. Agents Chemother. 2021, 65, e01732-20. [Google Scholar] [CrossRef] [PubMed]
  66. Auth, J.; Fröba, M.; Grobe, M.; Rauch, P.; Ruetalo, N.; Schindler, M.; Morokutti-Kurz, M.; Fraf, P.; Dolischka, A.; Prieschl- Grassauer, E.; et al. Lectin from Triticum vulgaris (WGA) inhibits infection with SARS-CoV-2 and its variants of concern alpha and beta. Int. J. Mol. Sci. 2021, 22, 10205. [Google Scholar] [CrossRef]
  67. Mori, T.; O’Keefe, B.R.; Sowder, R.C., II; Bringans, S.; Gardella, R.; Berg, S.; Cochran, P.; Turpin, J.A.; Buckheit, R.W., Jr.; McMahon, J.B.; et al. Isolation and characterization of griffithsin, a novel HIV-inactivating protein, from the red alga Griffithsia sp. J. Biol. Chem. 2005, 280, 9345–9353. [Google Scholar] [CrossRef] [Green Version]
  68. O’Keefe, B.R.; Giomarelli, B.; Barnard, D.L.; Shenoy, S.R.; Chan, P.K.S.; McMahon, J.B.; Palmer, K.E.; Barnett, B.W.; Mey Erholz, D.K.; Wohlford-Lenane, C.L.; et al. Broad-spectrum in vitro activity and in vivo efficacy of the antiviral protein griffithsin against emerging viruses of the family Coronaviridae. J. Virol. 2010, 84, 2511–2521. [Google Scholar] [CrossRef] [Green Version]
  69. Moulaei, T.; Shenoy, S.R.; Giomarelli, B.; Thomas, C.; McMahon, J.B.; Dauter, Z.; O’Keefe, B.R.; Wlodawer, A. Monomerizationof viral entry inhibitor griffithsin elucidates the relationship between multivalent binding to carbohydrates and anti-HIV activity. Structure 2010, 18, 1104–1115. [Google Scholar] [CrossRef] [Green Version]
  70. Sato, Y.; Morimoto, K.; Kubo, T.; Sakaguchi, T.; Nishizono, A.; Hirayama, M.; Hori, K. Entry inhibition of influenza viruses with high mannose binding lectin ESA-2 from the red alga Eucheuma serra through the recognition of viral hemagglutinin. Mar. Drugs 2015, 13, 3454–3465. [Google Scholar] [CrossRef] [Green Version]
  71. Hwang, H.-J.; Han, J.-W.; Jeon, H.; Cho, K.; Kim, J.-H.; Lee, D.-S.; Han, J.W. Characterization of a novel mannose binding lectin with antiviral activities from red alga, Grateloupia chiangii. Biomolecules 2020, 10, 333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Xue, J.; Hoorelbeke, B.; Kagiampakis, I.; Demeler, B.; Balzarini, J.; Liwang, P.J. The griffithsin dimer is required for high-potency inhibition of HIV-1: Evidence for manipulation of the structure of gp120 as part of the griffithsin dimer mechanism. Antimicrob. Agents Chemother. 2013, 57, 3976–3989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Meuleman, P.; Albecka, A.; Belouzard, S.; Vercauteren, K.; Verhoye, L.; Wychowski, C.; Leroux-Roels, G.; Palmer, K.E.; Dubuisson, J. Griffithsin has antiviral activity against hepatitis C virus. Antimicrob. Agents Chemother. 2011, 55, 5159–5167. [Google Scholar] [CrossRef] [Green Version]
  74. Millet, J.K.; Séron, K.; Labitt, R.N.; Danneels, A.; Palmer, K.E.; Whittaker, G.R.; Dubuisson, J.; Belouzard, S. Middle East respiratory syndrome coronavirus infection is inhibited by griffithsin. Antivir. Res. 2016, 133, 1–8. [Google Scholar] [CrossRef] [PubMed]
  75. Alsaidi, S.; Cornejal, N.; Mahoney, O.; Melo, C.; Verma, N.; Bonnaire, T.; Chang, T.; O’Keefe, B.R.; Sailer, J.; Zydowsky, T.M.; et al. Griffithsin and carrageenan combination results in antiviral synergy against SARS- CoV-1 and 2 in a pseudoviral model. Mar. Drugs 2021, 19, 418. [Google Scholar] [CrossRef]
  76. Mu, J.; Hirayama, M.; Sato, Y.; Morimoto, K.; Hori, K. A novel high-mannose specific lectin from the green alga Halimeda renschii exhibits a potent anti-influenza virus activity through high-affinity binding to the viral hemagglutinin. Mar. Drugs 2017, 15, 255. [Google Scholar] [CrossRef] [Green Version]
  77. Sato, Y.; Morimoto, K.; Hirayama, M.; Hori, K. High-mannose-specific lectin (KAA-2) from the red alga Kappaphycus alvarezii potently inhibits influenza virus infection in a strain-independent manner. Biochem. Bipophys. Res. Commun. 2011, 405, 291–296. [Google Scholar] [CrossRef]
  78. Hirayama, M.; Shibata, H.; Imamura, K.; Sakaguchi, T.; Hori, K. High-mannose specific lectin and its recombinants from a Carrageenophyta Kappaphycus alvarezii represent a potent anti-HIV activity through high-affinity binding to the viral envelope glycoprotein gp120. Mar Biotechnol. 2016, 18, 144–160. [Google Scholar] [CrossRef]
  79. Sato, Y.; Hirayama, M.; Morimoto, K.; Yamamoto, N.; Okuyama, S.; Hori, K. High mannose-binding lectin with preference for the cluster of α1-2-mannose from the green alga Boodlea coacta is a potent entry inhibitor of HIV-1 and influenza viruses. J. Biol. Chem. 2011, 286, 19446–19458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Huskens, D.; Férir, G.; Vermeire, K.; Kehr, J.-C.; Balzarini, J.; Dittmann, E.; Schols, D. Microvirin, a novel α(1,2)-mannose-specific lectin isolated from Microcystis aeruginosa, has anti-HIV-1 activity comparable with that of cyanovirin-N but a much higher safety profile. J. Biol. Chem. 2010, 285, 24845–24854. [Google Scholar] [CrossRef] [Green Version]
  81. Shahid, M.; Qadir, A.; Yang, J.; Ahmad, I.; Zahid, H.; Mirza, S.; Windisch, M.P.; Shahzad-Ul-Hussan, S. An engineered microvirin with identical structural domains potently inhibits human immunodeficiency virus and hepatitis C virus cellular entry. Viruses 2020, 12, 199. [Google Scholar] [CrossRef] [Green Version]
  82. Ziólkowska, N.E.; Wlodawer, A. Structural studies of algal lectins with anti-HIV activity. Acta Biochim. Pol. 2006, 53, 617–626. [Google Scholar] [CrossRef] [Green Version]
  83. Kachko, A.; Loesgen, S.; Shahzad-Ul-Hussan, S.; Tan, W.; Zubkova, I.; Takeda, K.; Wells, F.; Rubin, S.; Bewley, C.A.; Major, M.E. Inhibition of hepatitis C virus by the cyanobacterial protein Microcystis viridis lectin: Mechanistic differences between the high-mannose specific lectins MVL, CV-N, and GNA. Mol. Pharm. 2013, 10, 4590–4602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Shenoy, S.R.; O’Keefe, B.R.; Bolmstedt, A.J.; Cartner, L.K.; Boyd, M.R. Selective interactions of the human immunodeficiency virus-inactivating protein cyanovirin-N with high-mannose oligosaccharides on gp120 and other glycoproteins. J. Pharmacol. Exp. Ther. 2001, 297, 704–710. [Google Scholar] [PubMed]
  85. Barrientos, L.G.; O’Keefe, B.R.; Bray, M.; Sanchez, A.; Gronenborn, A.M.; Boyd, M.R. Cyanovirin-N binds to the viral surface glycoprotein, GP1,2 and inhibits infectivity of Ebola virus. Antivir. Res. 2003, 58, 47–56. [Google Scholar] [CrossRef]
  86. Maier, I.; Schiestl, R.H.; Kontaxis, G. Cyanovirin-N binds viral envelope proteins at the low-affinity carbohydrate binding site without direct virus neutralization ability. Molecules 2021, 26, 3621. [Google Scholar] [CrossRef]
  87. Helle, F.; Wychowski, C.; Vu-Dac, N.; Gustafson, K.R.; Voisset, C.; Dubuisson, J. Cyanovirin-N inhibits hepatitis C virus entry by binding to envelope protein glycans. J. Biol. Chem. 2006, 281, 15177–25183. [Google Scholar] [CrossRef] [Green Version]
  88. Tiwari, V.; Shukla, S.; Shukla, D. A sugar binding protein cyanovirin-N blocks herpes simplex virus type-1 entry and cell fusion. Antivir. Res. 2009, 84, 67–75. [Google Scholar] [CrossRef] [Green Version]
  89. Naidoo, D.; Kar, P.; Roy, A.; Mutanda, T.; Bwapwa, J.; Sen, A.; Anandraj, A. 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 2021, 26, 5114. [Google Scholar] [CrossRef]
  90. Sato, Y.; Okuyama, S.; Hori, K. Primary structure and carbohydrate binding specificity of a potent anti-HIV lectin isolated from the filamentous cyanobacterium Oscillatoria agardhii. J. Biol. Chem. 2007, 282, 11021–11029. [Google Scholar] [CrossRef] [Green Version]
  91. Bokesch, H.R.; O’Keefe, B.R.; McKee, T.C.; Pannell, L.K.; Patterson, G.M.L.; Gardella, R.S.; Sowder, R.C., II; Turpin, J.; Watson, K.; Buckheit, R.W., Jr.; et al. A potent novel anti-HIV protein from the cultured cyanobacterium Scytonema varium. Biochemistry 2003, 42, 2578–2584. [Google Scholar] [CrossRef]
  92. Siqueira, A.S.; Lima, A.R.J.; de Sousa, R.C.; Santos, A.S.; da Silva Gonçalves Vianez Júnior, J.L.; Costa Gonçalves, E. Anti-dengue virus activity of scytovirin and evaluation of point mutation effects by molecular dynamics and binding free energy calculations. Biochem. Biophys. Res. Commun. 2017, 490, 1033–1038. [Google Scholar] [CrossRef]
  93. Garrison, A.R.; Giomarelli, B.G.; Lear-Rooney, C.M.; Saucedo, C.J.; Yellayi, S.; Krumpe, L.R.H.; Rose, M.; Paragas, J.; Bray, M.; Olinger, G.G.; et al. The cyanobacterial lectin scytovirin displays potent in vitro and in vivo activity against Zaire Ebola virus. Antivir. Res. 2014, 112, 1–7. [Google Scholar] [CrossRef] [Green Version]
  94. Koharudin, L.M.; Furey, W.; Gronenborn, A.M. Novel fold and carbohydrate specificity of the potent anti-HIV cyanobacterial lectin from Oscillatoria agardhii. J. Biol. Chem. 2011, 286, 1588–1597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Mazur-Marzec, H.; Ceglowska, M.; Konkel, R.; Pyrc, K. Antiviral cyanometabolites—A review. Biomolecules 2021, 11, 474. [Google Scholar] [CrossRef]
  96. François, K.O.; Balzarini, J. Potential of carbohydrate-binding agents as therapeutics against enveloped viruses. Med. Res. Rev. 2012, 32, 349–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Matsui, T.; Kobayashi, S.; Yoshida, O.; Ishii, S.; Abe, Y.; Yamamoto, N. Effects of succinylated concanavalin A on infectivity and syncytial formation of human immunodeficiency virus. Med. Microbiol. Immunol. 1990, 179, 225–235. [Google Scholar] [CrossRef]
  98. Banks, W.A.; Ibrahimi, F.; Farr, S.A.; Flood, S.A.; Morley, J.E. Effects of wheat germ agglutinin and aging on the regional brain uptake of HIV-1 gp120. Life Sci. 1999, 65, 81–89. [Google Scholar] [CrossRef]
  99. Balzarini, J.; Van Laethem, K.; Hatse, S.; Vermeire, K.; De Clercq, E.; Peumans, W.; Van Damme, E.; Vandamme, A.M.; Bolmstedt, A.; Schols, D. Profile of resistance of human immunodeficiency virus to mannose-specific plant lectins. J. Virol. 2004, 78, 10617–10627. [Google Scholar] [CrossRef] [Green Version]
  100. Balzarini, J. Targeting the glycans of gp120: A novel approach aimed at the Achilles heel of HIV. Lancet Infect. Dis. 2005, 5, 727–731. [Google Scholar] [CrossRef]
  101. Turville, S.G.; Vermeire, K.; Balzarini, J.; Schols, D. Sugar-binding proteins potently inhibit dendritic cell human immuno- deficiency virus type 1 (HIV-1) infection and dendritic-cell-directed HIV-1 transfer. J. Virol. 2005, 79, 13519–13527. [Google Scholar] [CrossRef] [Green Version]
  102. Balzarini, J.; Van Herrewege, Y.; Vermeire, K.; Vanham, G.; Schols, D. Carbohydrate-binding agents efficiently prevent dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN)-directed HIV-1 transmission to T lymphocytes. Mol. Pharmacol. 2007, 71, 3–11. [Google Scholar] [CrossRef]
  103. Auwerx, J.; François, K.O.; Vanstreels, E.; Van Laethem, K.; Daelemans, D.; Schols, D.; Balzarini, J. Capture and transmission of HIV-1 by the C-type lectin L-SIGN (DC-SIGNR) is inhibited by carbohydrate-binding agents and polyanions. Antivir. Res. 2009, 83, 61–70. [Google Scholar] [CrossRef]
  104. Hoorelbeke, B.; Van Damme, E.J.M.; Rougé, P.; Schols, D.; Van Laethem, K.; Fouquaert, E.; Balzarini, J. Differences in the mannose oligomer specificities of the closely related lectins from Galanthus nivalis and Zea mays strongly determine their eventual anti-HIV activity. Retrovirology 2011, 8, 10. [Google Scholar] [CrossRef] [Green Version]
  105. Kumaki, Y.; Wandersee, M.K.; Smith, A.J.; Zhou, Y.; Simmons, G.; Nelson, N.M.; Bailey, K.W.; Vest, Z.G.; Li, J.K.-K.; Chan, P.K.-S.; et al. Inhibition of severe acute respiratory syndrome coronavirus replication in a lethal SARS-CoV BALB/c mouse model by stinging nettle lectin, Urtica dioica agglutinin. Antivir. Res. 2011, 90, 22–32. [Google Scholar] [CrossRef]
  106. Barre, A.; Van Damme, E.J.M.; Simplicien, M.; Le Poder, S.; Konjklowski, B.; Benoist, H.; Peyrade, D.; Rougé, P. Man-specific lectins from plants, fungi, algae and cyanobacteria, as potential blockers for SARS-CoV, MERS-CoV and SARS-CoV-2 (COVID-19) coronaviruses: Biomedical perspectives. Cells 2021, 10, 1619. [Google Scholar] [CrossRef]
  107. Ahmed, N.; Jahan, R.; Nissapatorn, V.; Wilairatana, P.; Rahmatullah, M. Plant lectins as prospective antiviral biomolecules in the search for COVID-19 eradication strategies. Miomed. Pharmacother. 2022, 146, 112507. [Google Scholar] [CrossRef]
  108. Singh, R.S.; Walia, A.K.; Khattar, J.S.; Singh, D.P.; Kennedy, J.F. Cyanobacterial lectins characteristics and their role as antiviral agents. Int. J. Biol. Macromol. 2017, 102, 475–496. [Google Scholar] [CrossRef]
  109. Singh, R.S.; Walia, A.K. Lectins from red algae and their biomedical potential. J. Appl. Phycol. 2018, 30, 1833–1858. [Google Scholar] [CrossRef] [Green Version]
  110. Breitenbach Barroso Coelho, L.C.; Marcelino dos Santos Silva, P.; Felix de Oliveira, W.; De Moura, M.C.; Viana Pontual, E.; Soares Gomes, F.; Guedes Paiva, P.M.; Napoleão, T.H.; dos Santos Correia, M.T. Lectins as antimicrobial agents. J. Appl. Microbiol. 2018, 125, 1238–1252. [Google Scholar] [CrossRef] [Green Version]
  111. Carneiro, D.C.; Fernandez, L.G.; Monteiro-Cunha, J.P.; Benevides, R.G.; Cunha Lima, S.T. A patent review of the antimicrobial applications of lectins: Perspectives on therapy of infectious diseases. J. Appl. Microbiol. 2021; online ahead of print. [Google Scholar] [CrossRef]
  112. Martinez, D.; Amaral, D.; Markovitz, D.; Pinto, L. The use of lectins as tools to combat SARS-CoV-2. Curr. Pharm. Des. 2021, 27, 4212–4222. [Google Scholar] [CrossRef]
  113. Peumans, W.J.; Van Damme, E.J.M. Lectins as plant defense proteins. Plant Physiol. 1995, 109, 347–352. [Google Scholar] [CrossRef]
  114. Boisseau, C.; Moisand, A.; Père, D.; Rougé, P. Immunocytochemical localization of the Lathyrus ochrus (L.) DC. Seed lectin in seeds and seedlings. Plant Sci. 1985, 42, 25–34. [Google Scholar] [CrossRef]
  115. Rougé, P.; Sousa-Cavada, B. Isolation and partial characterization of two isolectins from Lathyrus ochrus (L.) DC. Seeds. Plant Sci. Lett. 1984, 37, 21–27. [Google Scholar]
  116. Fuqua, J.L.; Wanga, V.; Palmer, K.E. Improving the large-scale purification of the HIV microbicide, griffithsin. BMC Biotechnol. 2015, 15, 12. [Google Scholar] [CrossRef] [Green Version]
  117. Fuqua, J.L.; Hamorsky, K.; Khalsa, G.; Matoba, N.; Palmer, K.E. Bulk production of the antiviral lectin griffithsin. Plant Biotechnol. 2015, 13, 1160–1168. [Google Scholar] [CrossRef]
  118. Alam, A.; Jiang, L.; Kittleson, G.A.; Steadman, K.D.; Nandl, S.; Fuqua, J.L.; Palmer, K.E.; Tusé, D.; McDonald, K.A. Technoeconomic modeling of plant-based griffithsin manufacturing. Front. Bieng. Biotechnol. 2018, 6, 102. [Google Scholar] [CrossRef] [Green Version]
  119. Hoelscher, M.; Tiller, N.; The, A.Y.-H.; Wu, G.-Z.; Ma, J.K.-C.; Bock, R. High-level expression of the HIV entry inhibitor griffithsin from the plastid genome and retention of biological activity in dried tobacco leaves. Plant Mol. Biol. 2018, 97, 357–370. [Google Scholar] [CrossRef] [Green Version]
  120. Eapen, P.; Cates, J.; Mundell, R.; Palmer, K.E.; Fuqua, J.L. In preparation for outdoor pharming: Griffithsin can be expressed in Nicotiana excelsiana and retains activity after storage and silage. Front. Bioeng. Biotechnol. 2020, 8, 199. [Google Scholar] [CrossRef]
  121. Decker, J.S.; Menacho-Melgar, R.; Lynch, M.D. Low-cost, large-scale production of the anti-viral lectin griffithsin. Front. Bioeng. Biotechnol. 2020, 8, 1020. [Google Scholar] [CrossRef]
  122. Lueken, K.; Mazarguil, H.; Rougé, P. The identification of two peptide sequences of light subunits of the Lathyrus ochrus isolectins containing a sequential epitope. Immunol. Lett. 1988, 19, 309–312. [Google Scholar] [CrossRef]
  123. Lueken, K.; Liboz, T.; Mazarguil, H.; Rougé, P. Localization of amino acid sequence stretches containing a continuous epitope on the surface of the two Lathyrus ochrus isolectins. Immunol. Lett. 1989, 23, 223–226. [Google Scholar] [CrossRef]
  124. Kolberg, J.; Ayouba, A.; Rougé, P. Production and characterization of a mouse monoclonal antibody specific for lentil lectin. Biol. Chem. Hoppe-Seyler 1991, 372, 57–61. [Google Scholar]
  125. Lueken, K.; Kolberg, J.; Cambillau, C.; Bourne, Y.; Rougé, P. Monoclonal antibody 117, C-11 recognizes three exposed regions on the surface of the Lathyrus ochrus isolectin I. Immunol. Lett. 1991, 30, 47–52. [Google Scholar] [CrossRef]
  126. Koshte, V.L.; Aalbers, M.; Calkhoven, P.G.; Aalberse, R.C. The potent IgG4-inducing antigen in banana is a mannose-binding lectin, BanLec-I. Int. Arch. Allergy Immunol. 1992, 97, 17–24. [Google Scholar] [CrossRef]
  127. Tchernychev, B.; Rabinkov, A.; Mirelman, D.; Wilchek, M. Natural antibodies to dietary proteins—The existence of natural antibodies to alliinase (alliin lyase) and mannose-specific lectin from garlic (Allium sativum) in human serum. Immunol. Lett. 1995, 47, 53–57. [Google Scholar] [CrossRef]
  128. Bewley, C.A.; Gustafson, K.R.; Boyd, M.R.; Covell, D.G.; Bax, A.; Clore, G.M.; Gronenborn, A.M. Solution structure of cyanovirin-N, a potent HIV-inactivating protein. Nat. Struct. Biol. 1998, 5, 571–578. [Google Scholar] [CrossRef]
  129. Moulaei, T.; Botos, I.; Ziólkowska, N.E.; Bokesch, H.; Krumpe, L.R.; McKee, T.C.; O’Keefe, B.R.; Dauter, Z.; Wlodawer, A. Atomic-resolution crystal structure of the antiviral lectin scytovirin. Protein Sci. 2007, 16, 2756–2760. [Google Scholar] [CrossRef] [Green Version]
  130. Micevicz, E.D.; Cole, A.L.; Jung, C.L.; Phillips, M.L.; Pratikhya, P.; Sharma, S.; Waring, A.J.; Cole, A.M.; Ruchala, P. Grifonin-I: A small HIV-1 entry inhibitor derived from the algal lectin, griffithsin. PLoS ONE 2010, 5, e14360. [Google Scholar] [CrossRef] [Green Version]
  131. Cooper, H.L.; Rubin, A.D. Synthesis of nonribosomal RNA by lymphocytes: A response to phytohemagglutinin treatment. Science 1966, 152, 516–518. [Google Scholar] [CrossRef]
  132. Phillips, B.; Weisrose, E. The mitogenic response of human B lymphocytes to phytohaemagglutinin. Clin. Exp. Immunol. 1974, 16, 383–392. [Google Scholar]
  133. Lis, H.; Sharon, N. Biological properties of lectins. In The Lectins, Properties, Functions, and Applications in Biology and Medicine; Liener, I.E., Sharon, N., Goldstein, I.J., Eds.; Academic Press Inc.: Orlando, FL, USA; San Diego, CA, USA; New York, NY, USA; Austin TX, USA, 1986; pp. 265–291. [Google Scholar]
  134. Liu, T.; Wu, L.; Wang, H.; Chen, J.; Yang, C.; Bao, J.; Wu, C. Role of reactive oxygen species-mediated MAPK and NF-κB activation on Polygonatum cyrtonema lectin-induced apoptosis and autophagy in human lung adenocarcinoma A549 cells. J. Biochem. 2016, 160, 315–324. [Google Scholar] [CrossRef] [Green Version]
  135. Chowdhury, S.R.; Ray, U.; Chatterjee, B.; Roy, S.S. Targeted apoptosis in ovarian cancer cells through mitochondrial dysfunction in response to Sambucus nigra agglutinin. Cell Death Dis. 2017, 8, e2762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Poiroux, G.; Barre, A.; Van Damme, E.J.M.; Benoist, H.; Rougé, P. Plant lectins targeting O-glycans at the cell surface as tools for cancer diagnosis, prognosis and therapy. Int. J. Mol. Sci. 2017, 18, 1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Naik, S.; Rawat, R.S.; Khandai, S.; Kumar, M.; Jena, S.S.; Vijayalakshmi, M.A.; Kumar, S. Biochemical characterization of lectin from Indian hyacinth plant bulbs with potential inhibitory action against human cancer cells. Int. J. Biol. Macromol. 2017, 105, 1349–1356. [Google Scholar] [CrossRef] [PubMed]
  138. Jang, S.; Yayeh, T.; Leem, Y.-H.; Park, E.-M.; Ito, Y.; Oh, S. Concanavalin A induces cortical neuron apoptosis by causing ROS accumulation and tyrosine kinase activation. Neurochem. Res. 2017, 42, 3504–3514. [Google Scholar] [CrossRef]
  139. Islam, F.; Gopalan, V.; Lam, A.K.-Y.; Rashel Kabir, S. Pea lectin inhibits cell growth by inducing apoptosis in SW480 and SW48 cell lines. Int. J. Biol. Macromol. 2018, 117, 1050–1057. [Google Scholar] [CrossRef] [Green Version]
  140. Poiroux, G.; Barre, A.; Simplicien, M.; Pelofy, S.; Segui, B.; Van Damme, E.J.M.; Rougé, P.; Benost, H. Morniga-G, a T/Tn-specific lectin, induces leukemic cell death via caspase and DR5 receptor-dependent pathways. Int. J. Mol. Sci. 2019, 20, 230. [Google Scholar] [CrossRef] [Green Version]
  141. Bhutia, S.; Panda, P.K.; Sinha, N.; Praharaj, P.P.; Bhol, C.S.; Panigrahi, D.P.; Mahapatra, K.K.; Saha, S.; Patra, S.; Mishra, S.R.; et al. Plant lectins in cancer therapeutics: Targeting apoptosis and autophagy-dependent cell dath. Pharmacol. Res. 2019, 144, 8–18. [Google Scholar] [CrossRef]
  142. Lalli, R.C.; Kaur, K.; Chakraborti, A.; Srinivasan, R.; Ghosh, S. Maackia amurensis agglutinin induces apoptosis in cultured resistant human non-small lung cancer cells. Glycoconj. J. 2019, 36, 473–485. [Google Scholar] [CrossRef]
  143. Rashidbaghan, A.; Mostafaie, A.; Yazdani, Y.; Mansouri, K. Urtica dioica agglutinin (a plant lectin) has a caspase-dependent apoptosis induction effect on the acute lymphoblastic leukemia cell line. Cell Mol. Biol. 2020, 66, 121–126. [Google Scholar] [CrossRef]
  144. Balzarini, J.; Van Laethem, K.; Peumans, W.J.; Van Damme, E.J.; Bolmstedt, A.; Cago, F.; Schols, D. Mutational pathways, resistance profile, and side effects of cyanovirin relative to human immunodeficiency virus type 1 strains with N-glycan deletions in their gp120 envelopes. J. Virol. 2006, 80, 8411–8421. [Google Scholar] [CrossRef] [Green Version]
  145. Huskens, D.; Vermeire, K.; Vandermeulebroucke, E.; Balzarini, J. Safety concerns for the potential use of cyanovirin-N as a microbicidal anti-HIV agent. Int. J. Biochem. Cell Biol. 2008, 40, 2802–2814. [Google Scholar] [CrossRef]
  146. Balzarini, J.; François, K.O.; Van Laethem, K.; Hoorelbeke, B.; Renders, M.; Auwers, J.; Liekens, S.; Oki, T.; Igarashi, Y.; Schold, D. Pradimicin S, a highly-soluble non-peptidic small-size carbohydrate-binding antibiotic, is an anti-HIV drug lead for both microbicidal and systemic use. Antimicrob. Agents Chemother. 2010, 54, 1425–1435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Büttner, S.; Koch, B.; Dolnik, O.; Eickmann, M.; Freiwald, T.; Rudolf, S.; Engel, J.; Becker, S.; Ronco, C.; Geiger, H. Extracorporeal virus elimination for the treatment of severe Ebola virus disease—First experience with lectin affinity plasmapheresis. Blood Purif. 2014, 38, 286–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Koch, B.; Schult-Dietrich, P.; Büttner, S.; Dilmaghani, B.; Lohmann, D.; Baer, P.C.; Dietrich, U.; Geiger, H. Lectin affinity plasmapheresis for middle east respiratory syndrome-coronavirus and Marburg virus glycoprotein elimination. Blood Purif. 2018, 46, 126–133. [Google Scholar] [CrossRef]
  149. Leblanc, J.-F.; Germain, M.; Delage, G.; O’Brien, S.; Drews, S.J.; Lewin, A. Risk of transmission of severe acute respiratory syndrome coronavirus 2 by transfusion: A literature review. Transfusion 2020, 60, 3046–3054. [Google Scholar] [CrossRef]
  150. Gaussen, A.; Hornby, L.; Rockl, G.; O’Brien, S.; Delage, G.; Sapir-Pichhadze, R.; Drews, S.J.; Weiss, M.J.; Lewin, A. Evidence of SARS-CoV-2 infection in cells, tissues and organs and the risk of transmission through transplantation. Transplantation 2021, 105, 1405–1422. [Google Scholar] [CrossRef] [PubMed]
  151. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimera—A visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605–1612. [Google Scholar] [CrossRef] [Green Version]
  152. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Meng, E.C.; Couch, G.S.; Croll, T.I.; Morris, J.H.; Ferrin, T.E. UCSF ChimeraX: Structure visualization for researchers, educators, and developers. Protein Sci. 2021, 30, 70–82. [Google Scholar] [CrossRef]
  153. Böhm, M.; Bohne-Lang, A.; Frank, M.; Loss, A.; Rojas-Macias, M.A.; Lütteke, T. Glycosciences.DB: An annotated data collec- tion linking glycomics and proteomics data (2018 update). Nucleic Acids Res. 2019, 47, D1195–D1201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Cheng, K.; Zhou, Y.; Neelamegham, S. DrawGlycan-SNFG: A robust tool to render glycans and glycopeptides with frag mentation information. Glycobiology 2017, 27, 200–205. [Google Scholar]
  155. Day, C.J.; Bailly, B.; Guillon, P.; Dirr, L.; Jen, F.E.-C.; Spillings, B.L.; Mak, J.; Itzstein, M.; Haselhorst, T.; Jennings, M.P. Mul- tidisciplinary approaches identify compounds that bind to human ACE2 or SARS-CoV-2 spike protein as candidates to block SARS-CoV-2-ACE2 receptor interactions. mBio 2021, 12, e03681-20. [Google Scholar] [CrossRef] [PubMed]
  156. Kawasaki, N.; Lin, C.-W.; Inoue, R.; Khoo, K.-H.; Kawasaki, N.; Ma, B.Y.; Oka, S.; Ishiguro, M.; Sawada, T.; Ishida, H.; et al. Highly fucosylated N-glycan ligand for mannan-binding protein expressed specifically on CD26(DPPIV) isolated from a human colorectal carcinoma cell line, SW1116. Glycobiology 2009, 19, 437–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Allen, J.D.; Watanabe, Y.; Chawla, H.; Newby, M.L.; Crispin, M. Subtle influence of ACE2 glycan processing on SARS-CoV-2 recognition. J. Mol. Biol. 2021, 433, 166762. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Diversity of the different types of N-glycans forming the glycan shield covering the pathogenic enveloped viruses: Ebola virus (EBOV) [19], herpes simplex virus (HSV) [20], human cytomegalovirus (HCMV) [21], human immunodeficiency virus (HIV) [22], influenza virus (IV) [23], chikungunya virus (CHIV) [24], Lassa virus (LASV) [25], MERS-CoV (MERS-CoV) [15], SARS-CoV (SARS-CoV) [15], SARS-CoV-2 (SARS-CoV-2) [15], and Zika virus (ZIV) [26]. Symbols representing the glycan structures are as follows: GlcNAc (blue square), Gal (yellow circle), Man (green circle), Fuc (red triangle), and Neu5Ac (purple diamond).
Figure 1. Diversity of the different types of N-glycans forming the glycan shield covering the pathogenic enveloped viruses: Ebola virus (EBOV) [19], herpes simplex virus (HSV) [20], human cytomegalovirus (HCMV) [21], human immunodeficiency virus (HIV) [22], influenza virus (IV) [23], chikungunya virus (CHIV) [24], Lassa virus (LASV) [25], MERS-CoV (MERS-CoV) [15], SARS-CoV (SARS-CoV) [15], SARS-CoV-2 (SARS-CoV-2) [15], and Zika virus (ZIV) [26]. Symbols representing the glycan structures are as follows: GlcNAc (blue square), Gal (yellow circle), Man (green circle), Fuc (red triangle), and Neu5Ac (purple diamond).
Cells 11 00339 g001
Figure 2. Figure illustrating the extent of the glycan shield covering the envelope glycoproteins of pathogenic enveloped viruses. The illustrations show the lateral face of the molecular surface of the homotrimeric organization of the envelope glycoprotein of Ebola virus (EBOV) (PDB code 7JPH), influenza virus (IV) (PDB code 6Y5G), human cytomegalovirus (HCMV) (PDB code 5CXF), herpes simplex virus (HSV) (PDB code 2GUM), human immunodeficiency virus HIV (PDB code 4TVP), Lassa virus (LASV) (PDB code 5VK2), Middle East respiratory syndrome virus (MERS-CoV) (PDB code 5W9H), severe acute respiratory syndrome (SARS-CoV) (PDB code 6ACD), and severe acute respiratory syndrome-2 (SARS-CoV-2) (PDB code 6VXX). Lateral face of the molecular surface of the dimeric organization of the E glycoprotein of chikungunya virus CHIV (PDB code 3N40), dengue virus DENV (PDB code 1UZG), and Zika virus ZIV (PDB code 7BUB). Monomers forming the homotrimeric and homodimeric associations of envelope glycoproteins are colored differently, and N-glycan chains forming the glycan shield are represented by cyan colored balls.
Figure 2. Figure illustrating the extent of the glycan shield covering the envelope glycoproteins of pathogenic enveloped viruses. The illustrations show the lateral face of the molecular surface of the homotrimeric organization of the envelope glycoprotein of Ebola virus (EBOV) (PDB code 7JPH), influenza virus (IV) (PDB code 6Y5G), human cytomegalovirus (HCMV) (PDB code 5CXF), herpes simplex virus (HSV) (PDB code 2GUM), human immunodeficiency virus HIV (PDB code 4TVP), Lassa virus (LASV) (PDB code 5VK2), Middle East respiratory syndrome virus (MERS-CoV) (PDB code 5W9H), severe acute respiratory syndrome (SARS-CoV) (PDB code 6ACD), and severe acute respiratory syndrome-2 (SARS-CoV-2) (PDB code 6VXX). Lateral face of the molecular surface of the dimeric organization of the E glycoprotein of chikungunya virus CHIV (PDB code 3N40), dengue virus DENV (PDB code 1UZG), and Zika virus ZIV (PDB code 7BUB). Monomers forming the homotrimeric and homodimeric associations of envelope glycoproteins are colored differently, and N-glycan chains forming the glycan shield are represented by cyan colored balls.
Cells 11 00339 g002
Figure 3. Figure illustrating the extent of the glycan shield covering the envelope glycoproteins of pathogenic enveloped viruses. Illustrations show the top face of the molecular surface of the homotrimeric organization of the envelope glycoprotein of Ebola virus (EBOV) (PDB code), influenza virus (IV) (PDB code), human cytomegalovirus (HCMV) (PDB code 5CXF), herpes simplex virus (HSV) (PDB code 2GUM), human immunodeficiency virus (HIV) (PDB code 4TVP), Lassa virus (LASV) (PDB code 5VK2), Middle East respiratory syndrome virus (MERS-CoV) (PDB code 5W9H), severe acute respiratory syndrome (SARS-CoV) (PDB code 6ACD), and severe acute respiratory syndrome-2 (SARS-CoV-2) (PDB code 6VXX). Monomers forming the homotrimeric associations of envelope glycoproteins are colored differently, and N-glycan chains forming the glycan shield are represented by cyan colored balls.
Figure 3. Figure illustrating the extent of the glycan shield covering the envelope glycoproteins of pathogenic enveloped viruses. Illustrations show the top face of the molecular surface of the homotrimeric organization of the envelope glycoprotein of Ebola virus (EBOV) (PDB code), influenza virus (IV) (PDB code), human cytomegalovirus (HCMV) (PDB code 5CXF), herpes simplex virus (HSV) (PDB code 2GUM), human immunodeficiency virus (HIV) (PDB code 4TVP), Lassa virus (LASV) (PDB code 5VK2), Middle East respiratory syndrome virus (MERS-CoV) (PDB code 5W9H), severe acute respiratory syndrome (SARS-CoV) (PDB code 6ACD), and severe acute respiratory syndrome-2 (SARS-CoV-2) (PDB code 6VXX). Monomers forming the homotrimeric associations of envelope glycoproteins are colored differently, and N-glycan chains forming the glycan shield are represented by cyan colored balls.
Cells 11 00339 g003
Figure 4. Figure illustrating the interaction of the Man-specific lectin LoL-II from Lathyrus ochrus with an oligosaccharide chain. (A) Network of hydrogen bonds (black lines) anchoring N2 oligosaccharide (colored purple) to LoL-II isolectin from Lathyrus ochrus (PDB code 1LGC). Hydrophilic residues R38, N78, D81, G99, and N125 of the α-chain and E31 of the β-chain, which participate in hydrogen bonds, are colored orange and green, respectively. Aromatic residues Y77, Y100, F123, Y124, and W128 of the α-chain and F32 of the β-chain, involved in stacking interactions with the pyranose rings of the oligosaccharide, are colored yellow. The α1,6-linked fucose (Fuc), which participates in the H-bond network, is colored cyan. (B) Depiction of the N2 oligosaccharide using the symbol nomenclature for glycans: Fuc (red triangle), Gal (yellow circle), GalNAc (yellow square), GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond). (C) Molecular surface of the N2 oligosaccharide–Lo-LII complex, showing how the isolectin accommodates the oligosaccharide via a network of hydrogen bonds and stacking interactions. The groove harboring the N2 oligosaccharide and the central monosaccharide-binding site of the lectin are delineated with yellow, dashed lines.
Figure 4. Figure illustrating the interaction of the Man-specific lectin LoL-II from Lathyrus ochrus with an oligosaccharide chain. (A) Network of hydrogen bonds (black lines) anchoring N2 oligosaccharide (colored purple) to LoL-II isolectin from Lathyrus ochrus (PDB code 1LGC). Hydrophilic residues R38, N78, D81, G99, and N125 of the α-chain and E31 of the β-chain, which participate in hydrogen bonds, are colored orange and green, respectively. Aromatic residues Y77, Y100, F123, Y124, and W128 of the α-chain and F32 of the β-chain, involved in stacking interactions with the pyranose rings of the oligosaccharide, are colored yellow. The α1,6-linked fucose (Fuc), which participates in the H-bond network, is colored cyan. (B) Depiction of the N2 oligosaccharide using the symbol nomenclature for glycans: Fuc (red triangle), Gal (yellow circle), GalNAc (yellow square), GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond). (C) Molecular surface of the N2 oligosaccharide–Lo-LII complex, showing how the isolectin accommodates the oligosaccharide via a network of hydrogen bonds and stacking interactions. The groove harboring the N2 oligosaccharide and the central monosaccharide-binding site of the lectin are delineated with yellow, dashed lines.
Cells 11 00339 g004
Figure 5. Figure illustrating the interaction of the Man-specific lectin LoL-I from Lathyrus ochrus with a trimannoside. (A) Network of hydrogen bonds (black lines) anchoring the trisaccharide Manα1,3Manβ1,4GlcNAc (colored purple) to LoL-I isolectin from Lathyrus ochrus (PDB code 1LOG). Hydrophilic residues D81, G99, and N125 of the α-chain and G29, A30, and E31 of the β-chain, which participate in hydrogen bonds, are colored orange and green, respectively. Aromatic residues Y100, Y124, and W128 of the α-chain, involved in stacking interactions with the pyranose rings of the oligosaccharide, are colored yellow. (B) Illustration of the Man3GlcNAc2 oligosaccharide using the symbol nomenclature for glycans: Fuc (red triangle), Gal (yellow circle), GalNAc (yellow square), GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond), showing the sugar units that participate in the trisaccharide–LoL-I complex.
Figure 5. Figure illustrating the interaction of the Man-specific lectin LoL-I from Lathyrus ochrus with a trimannoside. (A) Network of hydrogen bonds (black lines) anchoring the trisaccharide Manα1,3Manβ1,4GlcNAc (colored purple) to LoL-I isolectin from Lathyrus ochrus (PDB code 1LOG). Hydrophilic residues D81, G99, and N125 of the α-chain and G29, A30, and E31 of the β-chain, which participate in hydrogen bonds, are colored orange and green, respectively. Aromatic residues Y100, Y124, and W128 of the α-chain, involved in stacking interactions with the pyranose rings of the oligosaccharide, are colored yellow. (B) Illustration of the Man3GlcNAc2 oligosaccharide using the symbol nomenclature for glycans: Fuc (red triangle), Gal (yellow circle), GalNAc (yellow square), GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond), showing the sugar units that participate in the trisaccharide–LoL-I complex.
Cells 11 00339 g005
Figure 6. Structures of N-glycans recognized by PsA. Top 5 N-glycans are arranged in decreasing order of affinity for PsA. The α1,6-fucosylated Man3GlcNAc2 core is delineated with a red square. Glycans occurring in the glycan shield of pathogenic enveloped viruses are indicated by a red star. Fuc (red triangle), Gal (yellow circle), GalNAc (yellow circle), GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond).
Figure 6. Structures of N-glycans recognized by PsA. Top 5 N-glycans are arranged in decreasing order of affinity for PsA. The α1,6-fucosylated Man3GlcNAc2 core is delineated with a red square. Glycans occurring in the glycan shield of pathogenic enveloped viruses are indicated by a red star. Fuc (red triangle), Gal (yellow circle), GalNAc (yellow circle), GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond).
Cells 11 00339 g006
Figure 7. Structures of N-glycans recognized by GNA. The top 5 N-glycans are arranged in decreasing order of affinity for GNA. The Man3GlcNAc2 core is delineated by a blue square. High-mannose glycans occurring in the glycan shield of pathogenic enveloped viruses are indicated by a red star. Gal (yellow circle), GalNAc (yellow circle), GlcNAc (blue square), Man (green circle).
Figure 7. Structures of N-glycans recognized by GNA. The top 5 N-glycans are arranged in decreasing order of affinity for GNA. The Man3GlcNAc2 core is delineated by a blue square. High-mannose glycans occurring in the glycan shield of pathogenic enveloped viruses are indicated by a red star. Gal (yellow circle), GalNAc (yellow circle), GlcNAc (blue square), Man (green circle).
Cells 11 00339 g007
Figure 8. Structures of N-glycans recognized by SNA-I. The top five sialylated N-glycans are arranged in decreasing order of affinity for SNA-I. The Man3GlcNAc2 core is delineated by a blue square. The fucosylated Man3GlcNAc2 core is indicated with a red circled. Sialylated glycans occurring in the glycan shield of pathogenic enveloped viruses are indicated by a red star. Fuc (red triangle), Gal (yellow circle), GalNAc (yellow circle, GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond).
Figure 8. Structures of N-glycans recognized by SNA-I. The top five sialylated N-glycans are arranged in decreasing order of affinity for SNA-I. The Man3GlcNAc2 core is delineated by a blue square. The fucosylated Man3GlcNAc2 core is indicated with a red circled. Sialylated glycans occurring in the glycan shield of pathogenic enveloped viruses are indicated by a red star. Fuc (red triangle), Gal (yellow circle), GalNAc (yellow circle, GlcNAc (blue square), Man (green circle), and sialic acid/Neu5Ac (purple diamond).
Cells 11 00339 g008
Figure 9. Structures of O-glycans recognized by Morniga-G. The top 5 O-glycans are arranged in decreasing order of affinity for PNA. The O-glycans occurring in the glycan shield of SARS-CoV-2 spike protein are indicated by a red star. Gal (yellow circle), GalNAc (yellow square), GlcNAc (blue square), and sialic acid/Neu5Ac (purple diamond).
Figure 9. Structures of O-glycans recognized by Morniga-G. The top 5 O-glycans are arranged in decreasing order of affinity for PNA. The O-glycans occurring in the glycan shield of SARS-CoV-2 spike protein are indicated by a red star. Gal (yellow circle), GalNAc (yellow square), GlcNAc (blue square), and sialic acid/Neu5Ac (purple diamond).
Cells 11 00339 g009
Figure 10. Figure illustrating how the Man-specific lectin griffithsin interacts with an oligomannoside. Network of hydrogen bonds (black, dashed lines) connecting the griffithsin monomer (colored violet) to a linear Man8 chain (colored purple) (PDB code 3LL2). Amino acid D residues participating in hydrogen bonds are labeled (i.e., D67, D109, and D112). Aromatic Y residues involved in stacking interactions with the sugar rings are labeled and colored orange (i.e., Y28, Y68, and Y110).
Figure 10. Figure illustrating how the Man-specific lectin griffithsin interacts with an oligomannoside. Network of hydrogen bonds (black, dashed lines) connecting the griffithsin monomer (colored violet) to a linear Man8 chain (colored purple) (PDB code 3LL2). Amino acid D residues participating in hydrogen bonds are labeled (i.e., D67, D109, and D112). Aromatic Y residues involved in stacking interactions with the sugar rings are labeled and colored orange (i.e., Y28, Y68, and Y110).
Cells 11 00339 g010
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Barre, A.; Van Damme, E.J.M.; Klonjkowski, B.; Simplicien, M.; Sudor, J.; Benoist, H.; Rougé, P. Legume Lectins with Different Specificities as Potential Glycan Probes for Pathogenic Enveloped Viruses. Cells 2022, 11, 339. https://doi.org/10.3390/cells11030339

AMA Style

Barre A, Van Damme EJM, Klonjkowski B, Simplicien M, Sudor J, Benoist H, Rougé P. Legume Lectins with Different Specificities as Potential Glycan Probes for Pathogenic Enveloped Viruses. Cells. 2022; 11(3):339. https://doi.org/10.3390/cells11030339

Chicago/Turabian Style

Barre, Annick, Els J. M. Van Damme, Bernard Klonjkowski, Mathias Simplicien, Jan Sudor, Hervé Benoist, and Pierre Rougé. 2022. "Legume Lectins with Different Specificities as Potential Glycan Probes for Pathogenic Enveloped Viruses" Cells 11, no. 3: 339. https://doi.org/10.3390/cells11030339

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop