Abstract

Sialidases, or neuraminidases, are enzymes that catalyze the hydrolysis of sialic acid (Sia)-containing molecules, mostly removal of the terminal Sia (desialylation). By desialylation, sialidase can modulate the functionality of the target compound and is thus often involved in biological pathways. Inhibition of sialidases with inhibitors is an important approach for understanding sialidase function and the underlying mechanisms and could serve as a therapeutic approach as well. Transition-state analogues, such as anti-influenza drugs oseltamivir and zanamivir, are major sialidase inhibitors. In addition, difluoro-sialic acids were developed as mechanism-based sialidase inhibitors. Further, fluorinated quinone methide-based suicide substrates were reported. Sialidase product analogue inhibitors were also explored. Finally, natural products have shown competitive inhibiton against viral, bacterial, and human sialidases. This Perspective describes sialidase inhibitors with different mechanisms and their activities and future potential, which include transition-state analogue inhibitors, mechanism-based inhibitors, suicide substrate inhibitors, product analogue inhibitors, and natural product inhibitors.
1. Introduction
1.1. Sialic Acids, Sialylation, and Desialylation
Sialic acids (Sias) are carboxylic acid-containing 9-carbon monosaccharides that often present at the terminus of glycan structures of glycoproteins and glycolipids.1 Sias are attached to either galactose (Gal), N-acetyl galactosamine (GalNAc) unit via α2,3- or α2,6-linkage, or Sias via α2,8(9)-linkage, which is known as sialoform.2N-acetylneuraminic acid (Neu5Ac) is the most abundant form of Sias found in mammalian cells, which also exhibits remarkable structural diversity, with more than 50 different derivatives identified in nature.2 These structural variations can occur as a variety of O-substitutions (acetylation, lactylation, sulfation, and methylation) at the C-4, C-7, C-8, and C-9 position, or as N-acetyl, N-glycolyl, or hydroxy at the C-5 position (Figure 1). As terminal carbohydrates with a negative charge, Sias could (i) exert physicochemical effects on the glycoconjugates to which they are attached, (ii) serve as recognition sites, or (iii) mask recognition sites of the glycoconjugates, and are therefore involved in various biological processes.1−4 The levels and linkages of Sias, known as sialylation status, is a delicate balance between sialylation (attachment of Sias) and desialylation (removal of Sias), which is maintained by sialyltransferases and sialidases, respectively.3,4
Figure 1.
Sialic acids (Sias) and their linkages, cell surface sialylation and desialylation by sialidases.
Desialylation by sialidase alters the functionality of Sia-containing glycoconjugates and is thus often involved in a variety of biological processes.5 First, desialylation reduces the charge of the glycoconjugate and the entire cell surface, as Sia is a highly electronegative carbohydrate. Second, desialylation unmasks the glycoconjugate’s binding site for its molecular ligand of the partner, thus affecting the glycoprotein’s function as an enzyme or receptor. Further, desialylation affects the target glycoprotein’s folding, thus regulating its interaction with other molecules accordingly. Several biological processes have been clarified through desialylation that controls target glycoproteins’ function on the cell surface and their downstream signaling.5−8 Overall, desialylation could alter the active and inactive state of a protein and attenuate or augment its function in either physiological or pathological processes.9
1.2. Sialidases (Neuraminidases) in General
Sialidases (neuraminidases) catalyze the hydrolysis of Sia-containing substrates in either an exo fashion, where the terminal Sia is cleaved, or an endo fashion, where cleavage occurs within oligo-/polysialic acids. Most sialidases are exosialidases (EC 3.2.1.18) that hydrolyze the sialyl substrates with terminal Sias, but fewer are endosialidases (EC 3.2.1.129). On the basis of the catalytic mechanisms, there are three kinds of exosialidases: (i) hydrolytic sialidases, (ii) trans-sialidases, and (iii) anhydrosialidases (intramolecular trans-sialidases, EC 4.2.2.15). Hydrolytic sialidases release free Sias from oligosaccharides, glycolipids, and glycoproteins.10 In the presence of asialo-substrates, trans-sialidases transfer Sias from sialoglycoconjugates to acceptor molecules.11 Anhydrosialidases release 2,7-anhydro-α-N-acetylneuraminic acid (2,7-anhydro-Neu5Ac) from sialoglycoconjugates.12,13 Hydrolytic sialidases are generally active against α2,3-, α2,6-, and α2,8-linked substrates but with different preferences. While anhydrosialidases are specific for α2,3-linked substrates.
On the basis of the primary sequence similarity (carbohydrate active enzyme (CAZy) database, available at http://www.cazy.org), sialidases are also classified into five glycoside hydrolase (GH) families: 33, 34, 58, 83, and 156 (Table 1). The family GH33 includes human sialidase and bacterial hydrolytic neuraminidases, trans-sialidases, and anhydrosialidases. The family GH34 includes exclusively viral sialidases from influenza A and B viruses. The family GH58 comprises bacteriophage endosialidases, which are viral sialidases and infect nonhuman hosts. The GH83 family contains viral sialidases of the Paramyxoviridae family, which exhibits both neuraminidase and hemagglutinin activities and infects humans.14,15 The family GH156 is an exo-sialidase, identified recently from a freshwater hot spring environment.16 This enzyme hydrolyzes a variety of Sia-containing glycosides, typically α2,3-, α2,6-linked.16
Table 1. Classification of Sialidases.
| Exosialidase |
||||||
|---|---|---|---|---|---|---|
| Hydrolytic sialidases | Trans-sialidases | Anhydro-sialidases | Endosialidase | CAZy families | Sia-linkage specificity | |
| Human sialidase | × | GH33 | α2,3-, α2,6-, α2,8-Sia | |||
| Bacterial sialidase | × | × | × | × | GH33, GH156 | α2,3-, α2,6-, α2,8-Sia |
| Viral sialidase | × | × | GH34, GH58, GH83 | α2,3-, α2,6-Sia | ||
| Protozoa sialidase | × | × | GH33 | α2,3-Sia | ||
There are three sources for the sialidases in the human body, which are (i) directly produced by the body, (ii) supplied by microbial cells as part of natural microflora of the human body, and (iii) through infections, in which the pathogens bring their own sialidases for infection and replication. Human sialidases play important roles in human health, but their overexpression, activation, and mutations cause disorders and diseases. For example, increased expression of Neu1 was confirmed in human pulmonary airway epithelial and microvascular endothelial cells and fibroblasts, which is relevant to the lung pathologies.17 Mutations of lysosomal sialidase Neu1 cause the lysosomal storage disorder, a fulminant disease called sialidosis, which often develops before birth.18,19 Sialidases produced from the human gut microbiome may have a beneficial effect on humans, but some bacterial sialidases play pathogenic roles. For example, sialidase of intestinal microbiota targets the intestinal mucin glycoconjugates and plays a regulatory function of physiological and pathological pathways.20Clostridium perfringens sialidases could cause numerous diseases in humans and animals.21 Therefore, better understanding of sialidases produced from different organisms and viruses will facilitate clarification of pathological mechanisms and development of effective treatments for certain diseases.
Mammalian Sialidases
Mammalian sialidases are exo-sialidases catalyzing the hydrolysis of sialyl substrates and belong to the GH33 family. No mammalian sialidases have been found with trans-sialidase or anhydrosialidase activities to date. On the basis of their subcellular and tissue localization, mammalian sialidases are further classified into Neu1 (localized predominantly in lysosomes), Neu2 (cytosol), Neu3 (plasma membranes), and Neu4 (lysosomes or mitochondria and endoplasmic reticulum) sialidases (Table 2).22−25 The expression levels of the four mammalian sialidases are different. Neu1 is more highly expressed than Neu3 and Neu4, while Neu2 is less expressed in human tissues.26 These four sialidases also differ in substrate specificity, enzymatic properties, and sensitivity and relocation in response to cellular stimuli, suggesting different physiological and pathological roles they play.6,27 Neu1 is typically located in the lysosome, where it associates with its chaperone/transport protein, protective protein/cathepsin A (PPCA) and β-galactosidase, and is involved in the metabolism of sialyglycans.28 In addition, Neu1 could relocate to the plasma membrane upon stimulation.28−52 Neu253−55 and Neu456,57 are also found on the cell surface. Cell surface sialidases act as structural and functional modulators of various extracellular soluble and membrane-bound molecules in a variety of cell types (Table 2).32,38,39 Therefore, cell surface sialidases play very important roles in receptor activation and signaling pathways and could serve as potential therapeutic targets as well.
Table 2. Mammalian Sialidases in Different Subcellular Locations and Their Target Proteins.
| Neu1 | Neu2 | Neu3 | Neu4 | |
|---|---|---|---|---|
| Subcellular localization | Lysosome | Cytosol | Cell surface | Lysosome mitochondria endoplasmic reticulum |
| Relocation | Cell surface28−52 | Cell surface53−55 | NDa | Cell surface56,57 |
| GenBank no. | AAB96774.1 | CAB41449.1 | AAE69072.1 | AAH95117.1 |
| Expression levelb | 1 | 0.01%–0.03% | 5%–10% | 5%–10% |
| Targetsc | ApoB10058 | ATG590 | ApoB10058 | NDa |
| CD529 | CD18 (ItGB2)59−62 | |||
| CD18 (ItGB2)59−62 | CD42b (GPlbα)64,65 | |||
| CD31 (PECAM1)60,63 | EGFR30,78,79 | |||
| CD3660 | TGF-β/LAP91 | |||
| CD42b (GPlbα)64,65 | ||||
| CD4466−68 | ||||
| CD54 (ICAM1)61 | ||||
| CD64 (FCγR)69 | ||||
| CD104 (ITGB4)70 | ||||
| CD107a/b (LAMP-1, LAMP-2)71,72 | ||||
| CD140 (PDGFR)73 | ||||
| CD220 (IR)74−77 | ||||
| CD221 (IGF-1R)73,74 | ||||
| EGFR30,78,79 | ||||
| HGFR/Met80 | ||||
| MMP930,32,33,47,60,78−81 | ||||
| MUC178,82−85 | ||||
| TLR234,60 | ||||
| TLR334 | ||||
| TLR432,34−36,47,86−88 | ||||
| TLR781 | ||||
| TLR981 | ||||
| TrKA33,89 |
ND: no data available.
Referred from ref (9).
Modified from ref (92). Abbreviations: ApoB100, Apolipoprotein B100; ATG5, Autophagy related 5; CD protein, cluster of differentiation protein; EGFR, epidermal growth factor receptor; HGF, hepatocyte growth factor; IR, insulin receptor; LAP: latency-associated peptide; MMP9, matrix metallopeptidase 9; MUC1, mucin 1; TGF-β, transforming growth factor beta; TLR, toll-like receptor; TrkA, Tropomyosin receptor kinase A.
Although mammalian sialidases have different substrate specificities and properties, they share a common genomic organization.93,94 However, the overall amino acid identity of Neu1 compared to the other mammalian sialidases is about 19–24%, whereas Neu2, Neu3, and Neu4 show 34–40% homology to each other.6 X-ray structures were reported for human Neu2 in free form and in a complex with 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (DANA) inhibitor.95 The 3D structure of Neu2 is often used for the homology model of Neu1, Neu3, and Neu4, as there is no 3D structure reported for any of them.96 The main reason for the poor characterization the mammalian sialidases could be due to their membrane-bound structure.96 More detailed information on the subcellular distribution, substrate specificity, catalytic properties, and amino acid homologies of the four mammalian sialidases can be found in recent review articles.5−7,92,97−99 Despite the accumulating data, molecular mechanisms underlying mammalian sialidases involvement in cellular phenomena have yet to be fully elucidated.
Microbial Sialidases in the Human Body
Bacterial sialidases are found in the human gastrointestinal (GI) tract, respiratory tract, oral cavity, and reproductive tract.100 Bacteria bind to the host cells through modification of cell surface glycans by their hydrolytic sialidases.101 For example, human pathogens Clostridium (C) perfringens(102) and Vibrio cholerae(103) use their hydrolytic sialidases for mucosal infections. In the human GI tract, several bacterial commensals and pathogens have been identified to have sialidase activity.104−109 Respiratory tract bacteria, specifically Streptococcus (S) pneumoniae,110S. intermedius,111 and Haemophilus influenzae also have their own sialidases.112 In the oral cavity, hydrolytic sialidase-producing bacteria was also confirmed, namely S. oralis,113S. sanguinis, S. intermedius, S. mitis,114Porphyromonas gingivalis,115Actinomyces oris,116Tannerella forsythia,117 and Treponema denticola.118
Influenza A and B viruses belong to the Orthomyxoviridae family (GH34) and produce their own neuraminidase with hydrolytic activity.119 To date, 11 types of neuraminidase (N1–N11) have been identified in influenza A virus and one neuraminidase in the influenza B virus.119 Only N1 and N2 types of neuraminidases are related to human infections. The N1 type contains avian, swine, and human lineages, while N2 contains avian and human lineages.119 Paramyxovirus produces hemagglutinin-neuraminidase (HN) proteins, which belong to the GH83 family of viral sialidases, and are responsible for viral attachment and interaction with the fusion protein in viral infection.14,15
Trypanosoma (T) cruzi, T. brucei, and T. rangeli protozoa species also produce their own sialidases. T. cruzi and T. brucei sialidases are characterized with trans-sialidase activity, while the T. rangeli sialidase is strictly a hydrolytic sialidase.120−122T. cruzi trans-sialidases are more efficient for transferring Sia from the α-sialylglycoside donor to the β-galactopyranosyl unit in the acceptor than catalyzing the sialoside hydrolytic reaction.123 Therefore, T. cruzi trans-sialidase has been used for the synthesis of sialylated oligosaccharides.119 In the absence of a proper acceptor, trans-sialidases catalyze sialoside hydrolysis with retention of the configuration.124
Overall, sialidases catalyze the cleavage of terminal Sias (desialylation), which modulate the functionality of the Sia-containing molecules, and thus are often involved in physiological and pathological processes. Imbalance in the sialidase activity could cause diseases such as cancer, diabetes, heart disease, or neurodegenerative disorders.7,92 Therefore, sialidases could serve as potential therapeutic targets. Suitable sialidase inhibitors could be used as effective drugs for these diseases depending on which sialidase needs to be inhibited.125
1.3. Catalytic Mechanism of Sialidases
As mentioned previously, exosialidases are responsible for the hydrolysis of sialyl linkages in oligosaccharides, glycoproteins, and glycolipids. The catalytic mechanism of sialidases has been an important research subject. Several mechanisms have been discovered related to a specific group of sialidases.126,127 Typically, enzymatic hydrolysis of the glycosidic bond proceeds with either net retention or inversion of the anomeric configuration. Retention of the anomeric configuration is completed in two inverting steps, a double displacement mechanism, in which the catalytic residues act as the acid/base and nucleophile, respectively. Inversion of the anomeric configuration is completed in a single-step mechanism, in which the substrate and a water molecule are bound simultaneously. The GH33, GH34, and GH83 families of sialidases are exo-sialidases, and all perform hydrolysis with net retention of the anomeric configuration, which is completed via the formation and subsequent breakdown of a covalent intermediate to a conserved tyrosine active center.126,127 The GH58 family sialidase is an endo-polysialidase that acts with the inversion of the anomeric configuration.128 Interestingly, the newly identified GH156 family is an exo-sialidase but acts with inversion of the anomeric configuration of the released free Sia from a variety of α2–3- and α2–6-linked sialosides.16,129
Sialidases act with either retention or inversion of the anomeric configuration, depending on their intramolecular rearrangement. A number of studies on family GH33 sialidases have demonstrated that these enzymes operate through a two-step, double-displacement mechanism similar to the majority of retaining glycosidases but, involving the participation of a tyrosine residue as the catalytic nucleophile to form a covalent aryl-glycoside intermediate.130−132 Generally, a nucleophile pair of Tyr/Glu, acid/base aspartate, and the arginine triad are essential residues involved in the mechanism of catalytic cleavage among all types of sialidases. Initially, the positively charged arginines in the catalytic pocket are involved in the coordination of substrates by surrounding the negatively charged carboxylate group of Sia (Figure 2A). Meanwhile, the Tyr residue acts as a nucleophile to attach the anomeric center (C-2), which is assisted by the base Glu to enhance its nucleophilicity, yielding a semiplanar oxocarbenium transition state with the adjacent carbohydrate attached (Figure 2B) and then leading to the formation of an intermediate that is covalently bound to the active site (Figure 2C). Next, the water molecule activated by the Asp residue attacks the anomeric C-2 center in a trans addition to form another semiplanar oxocarbenium transition state with a water molecule attached (Figure 2D). In the final step, free Sia, as an α-anomer, is released from the active site of the sialidase (Figure 2E), completing the hydrolysis with retention of the anomeric configuration. Kinetic isotope effect (KIE) measurements with isotopically labeled natural substrate analogues were used to characterize the transition states of sialidase-catalyzed hydrolysis reactions, which have a pyranosyl ring in the 4H5 half-chair conformation coupled with the adjacent carbohydrate.133,134
Figure 2.

Catalytic mechanism of the hydrolytic sialidase with net retention of the anomeric configuration. (A) First step, Tyr residue acts as a nucleophile to attach the anomeric center (C-2), (B) semiplanar oxocarbenium transition state formation with the adjacent carbohydrate attached, (C) covalent intermediate that is bound to the active site, (D) another semiplanar oxocarbenium transition state formation with a water molecule attached, and (E) finally, release of free Sia as an α-anomer.
Mammalian, bacteria, viruses, and fungi sialidases have different primary sequences but share a common catalytic domain.135 They all form a covalent intermediate with the substrates initially, but the later steps of the catalytic mechanisms for the various sialidases are different. Detailed mechanisms and structural features of different types of sialidases can be found in comprehensive reviews and monographs.102,136−142
2. Discussions
Sialidases or neuraminidases cause the desialylation of Sia-containing oligosaccharides or glycoconjugates in both physiological and pathological pathways and thus play key roles in health and disease.5,9 Sialidase inhibitors have been developed and used for studying the sialidase function and related biological mechanisms and disease processes and could serve as drugs for sialidase-related diseases, such as viral infection.7 Inhibition of viral neuraminidase activity is a practical approach for the treatment of influenza infection.143 Tamiflu (oseltamivir) and Relenza (Zanamivir) are potent inhibitors of influenza virus neuraminidase and have been used for the treatment of influenza A and B for decades.143,144 Selective inhibitors against bacterial sialidases have been extensively explored for antibacterial action.145 Inhibitors of human sialidases are recognized as important tools for studying the biological functions of human sialidases and regulating the related biological processes.97,146 Several selective human sialidase inhibitors have been developed and have shown therapeutic potential for diseases such as inflammation, diabetes, atherosclerosis, fibrosis, neurodegenerative diseases, and cardiovascular diseases.147 Each type of sialidase inhibitor shows unique activity and potential for different applications. There has been a large number of sialidase inhibitors reported so far, and this Perspective will not describe them all in detail. Instead, it describes the sialidase inhibitors based on their inhibition mechanisms, including (i) transition-state analogue inhibitors, (ii) mechanism-based inhibitors, (iii) suicide substrate inhibitors, (iv) product analogue inhibitors, and (v) natural product inhibitors. More detailed information about specific sialidase inhibitors from the past decade can be found in comprehensive reviews.97,144−149
2.1. Transition-State Analogue Sialidase Inhibitors
Transition-state analogues have been widely used as potent enzyme inhibitors by blocking the active site of the enzyme.150 They are based on the theory that the enzyme binds the substrate at the transition state with extraordinary affinity. If an inhibitor mimics the transition state structure, it should have high affinity to the target enzyme and could serve as highly potent and specific drugs. In many cases, sialidase inhibitors are proposed to mimic the transition state formed during the sialoside hydrolysis. DANA (Neu5Ac2en) was the first transition-state analogue sialidase inhibitor (Figure 3), which mimics the oxocarbenium ion-like transition state and exhibits moderate inhibitory activity toward influenza viral neuraminidases with Ki values in the micromolar range.151 In addition, DANA is a product of sialidase-catalyzed hydrolysis reactions. Streptococcus pneumoniae sialidase SpNanC specifically hydrolyzes α2,3-linked sialosides and generates the transition-state analogue inhibitor DANA.152 Also, influenza B virus neuraminidase could catalyze the formation of DANA.153 Later, DANA was used in the structure-based drug design of the anti-influenza drug zanamivir (Relenza, GlaxoSmithKline) by the substitution of the 4-hydroxyl moiety with a guanidino group in the 1990s (Figure 3A).154 On the other hand, oseltamivir, which has a carbocyclic scaffold with a 3-pentyl ether side chain as a transition-state analogue, was developed as a potent anti-influenza drug (Figure 3B).155 The 3-pentyl ether is in lieu of the glycerol side chain in Sia, to render hydrophobic interactions with the Glu276, Ala246, Arg224, and Ile222 residues in the NA active site.156 Since then, DANA has been used as the model compound for developing selective sialidase inhibitors by modification with different functional groups at C-4, C-5, C-7, and C-9 positions (Figure 3C). In addition, substituted pyran-carboxylic acids, cyclohexene-carboxylic acids, benzoic acids, tetrahydrofuran acids, pyrrolidine acids, and bicyclo[3.1.0]hexane scaffolds were also developed as transition-state analogue sialidase inhibitors (Figure 3D). Natural products such as Siastatin B (Figure 3E) isolated from a Streptomyces strain, resemble the transition state and inhibit sialidases from various microorganisms, animal tissues, and viruses.157 This section describes the transition-state analogue inhibitors against viral, bacterial, and human sialidases, respectively.
Figure 3.

Transition-state analogue sialidase inhibitors in different scaffolds. IC50 values are for influenza viral NA inhibition. (A) Influenza neuraminidase inhibitors with pyran scaffold, (B) influenza neuraminidase inhibitors with carbocyclic scaffold, (C) selective sialidase inhibitors by modifying C-4, -5, -7, and -9 positions, and (D) sialidase inhibitors with other kinds of scaffolds.
Transition-state Analogue Influenza Virus Neuraminidase (NA) Inhibitors
Influenza virus neuraminidase (NA) becomes a primary drug target for the prophylaxis and treatment of influenza infections. Influenza virus NA inhibitors are the most successfully studied sialidase inhibitors.143 DANA was the first influenza virus NA inhibitor reported as a transition state analogue of the enzymatic hydrolysis of the flu receptor sialoside (Figure 3).151 While DANA shows moderate inhibitory activity, it has been used as a lead for the discovery of potent influenza virus NA inhibitors. Of these, zanamivir, oseltamivir, laninamivir, and peramivir have been developed for the treatment and prophylaxis of human influenza viral infection (Figure 3A).158−165Zanamivir (4-guanidino-Neu5Ac2en) is an analogue of DANA, in which a positively charged guanidino group was introduced to replace the hydroxyl group at C-4 position. This modification resulted in a significant increase in binding affinity to NA. Laninamivir is structurally similar to zanamivir, but has methylation of the C-7 hydroxyl group. Both laninamivir and zanamivir have the pyran scaffold. Oseltamivir carboxylate was designed with the aim of simplifying synthesis, while also improving bioavailability. Specifically, a carbocyclic scaffold is used instead of the pyran of zanamivir and DANA. Also, the 3-pentyl ether side chain replaces the hydrophilic glycerol side chain and the amino group replaces the guanidino group. Unlike zanamivir, oseltamivir relies on strong hydrophobic interactions rather than polar interactions (Figure 3B). In addition, carbocyclic compounds resemble the oxo-carbenium transition state intermediate more closely166 and bind the target NA more tightly than the pyran derivatives.155,167 As a result, the carbocyclic analogue of DANA has doubled the potency of DANA.168Peramivir is a cyclopentane derivative, making it structurally unique among the other approved NA inhibitors. Still, it has the functional groups, a guanidino moiety of zanamivir and a hydrophobic side chain of oseltamivir. Several other scaffolds have also been explored, such as benzoic acid, tetrahydrofuran acid, pyrrolidine acid, spiro compound, and bicyclo[3.1.0]hexane, but none of them produce the expected antiviral infection in vivo (Figure 3D).169 However, most of NA inhibitors developed so far are derivatives of commercial drugs. More detailed information about influenza virus NA inhibitors can be found in recent comprehensive reviews.143,170,171
Transition-State Analogue Bacterial Sialidase Inhibitors
Pathogenic bacterial species, such as Vibrio (V) cholerae (causes cholera), S. neumoniae (causes otitis media in children), and Gram-positive anaerobic bacteria Clostridium perfringens (causes gas gangrene disease), utilize their own sialidases for pathogenicity.172C. perfringens is pathogenic to humans and livestock and often causes gangrene, necrotizing enterocolitis, and food poisoning worldwide.173 DANA has been used as the template for developing C. perfringens and V. cholera sialidase inhibitors. Modification with the azido group at the C-9 or C-5 position of DANA increases its selectivity for bacterial sialidases over human sialidases. For example, Neu5Gc9N32en (Figure 4a) was identified as a selective inhibitor against V. cholerae sialidase.174 It was suggested that the hydrophobic group at the C-9 position of DANA would interact hydrophobically with the target loop moiety of the enzyme. 9-Triazole-linked and 5-N-trifluoroacetyl derivatives of DANA transition state analogue (Figure 4b) were reported as selective inhibitors against V. Cholerae sialidase.175 In addition, 9-triazole-linked peptide derivatives of DANA transition state analogues (Figure 4c) selectively inhibited V. cholerae and A. ureafaciens sialidases.176 The in vivo activities and therapeutic applications of these compounds deserve further investigation.
Figure 4.
Transition-state analogue bacterial sialidase inhibitors with modification at C-9 and C-5 position of DANA. (a) Neu5Gc9N32en, (b) 9-Triazole-linked and 5-N-trifluoroacetyl derivative of DANA, and (c) 9-triazole-linked peptide derivatives of DANA.
Transition-State Analogue Human Sialidase Inhibitors
Human sialidases catalyze the removal of Sia residues from glycoproteins and glycolipids. Four human sialidases (Neu1–4, belonging to the family GH33) have been identified and were found to be involved in atherosclerosis, cancer, diabetes, and neurodegenerative diseases.77,177,178 These four isoenzymes vary in their tissue expression, subcellular location, and substrate specificity (Table 2); however, their precise biochemical roles in different biological processes have not been fully investigated. Selective sialidase inhibitors are important tools for studying the biological functions of human sialidases and elucidating their roles in the regulation of glycoconjugates. They are also expected to serve as potent drugs for human sialidase-related diseases. In the past, there had been no commercially available human sialidase inhibitors. Bacterial or viral sialidase inhibitors were often used to study human sialidases, but typically show broad or weak activity.167,168 There remains a high demand for potent and selective human sialidase inhibitors for biological studies of the role of human sialidase isoenzymes.
C-9 Pentylamido derivative of DANA is the first selective inhibitor of human sialidases reported, which has a micromolar IC50 against Neu1 over the other isoenzymes.179 Pioneered by Cairo’s group, several selective inhibitors of human sialidases have been developed based on the DANA scaffold (Figure 4C).146 These selective inhibitors of human Neu1, Neu2, Neu3, and Neu4 isoenzymes were developed by modifying DANA at the C-4, C-5, C-7, and C-9 position and combining these modifications.174,179−185 Mostly, amide formation, oxime, or hydrazide formation and click chemistry were used for these modifications. The major DANA-based transition-state analogue human sialidase inhibitors are summarized in Table 3. Most recently, Bourguet et al. extensively described the structures and stereoselective inhibitors of human sialidases.97 Based on the already known inhibitors of human sialidases, a structure–activity relationship at C-4, C-5, C-7, and C-9 position is discussed in detail for the development of potent and selective inhibitors.97 More detailed information about these selective human sialidase inhibitors can be found in recent comprehensive reviews.143,170,171 These sialidase inhibitors represent useful tools for elucidating the roles of human sialidases in health and disease. Their in vivo activities and therapeutic applications deserve further investigation.
Table 3. Transition-State Analogue Human Sialidase Inhibitorsa.

| position | functional group | human sialidase selectivity | other sialidase selectivity | reference |
|---|---|---|---|---|
| (I)-C-4 (R1) | o | Neu2 > Neu3 | ND* | (184) |
| p | Neu3 > Neu2 | influenza virus | (181) | |
| (I)-C-4/5 (R1/R2) | p and d | Neu2 > Neu1 > Neu4 | ND | (182) |
| (I)-C-4/9 (R1/R3) | p and s | Neu3 | ND | (181) |
| p and t | Neu3 | ND | (181) | |
| (I)-C-5 (R2) | b | Neu1 > Neu3 | ND | (179,182) |
| c | Neu1 | ND | (182) | |
| e | Neu1 | ND | (182) | |
| h | Neu2 | ND | (182) | |
| i | Neu2 | ND | (182) | |
| (I)-C-5/9 (R2/R3) | b and b | Neu1 > Neu2 | ND | (182) |
| b and a | Neu1 > Neu2 | ND | (182) | |
| g and o | Neu2 | V. Cholerare | (174) | |
| f and o | Neu2 | V. Cholerare | (174) | |
| (I)-C-9 (R3) | b | Neu1 > Neu3 | ND | (182) |
| c | Neu1 > Neu3 | ND | (182) | |
| e | Neu1 | ND | (180) | |
| q | Neu3 | ND | (181) | |
| r | Neu3 | ND | (183) | |
| s | Neu3 | ND | (181) | |
| u | Neu4 | ND | (184) | |
| v | Neu4 | ND | (184) | |
| w | Neu3 | ND | (181) | |
| (II)-C-7 (R2) | k | Neu2 | ND | (180) |
| m | Neu2 > Neu3 | ND | (180) | |
| n | Neu2 = Neu3 | ND | (180) | |
| (II)-C-4/7 (R1/R2) | o and j | Neu3 | ND | (180,185) |
| o and l | Neu2 = Neu3 | ND | (180) | |
| o and n | Neu2 = Neu3 | ND | (180) |
ND: no data available.
2.2. Mechanism-Based Sialidase Inhibitors - 2,3-Difluoro-N-Acteylneuraminic Acid Derivatives
Carbohydrate fluorination, in which fluorine is used to replace a hydroxyl group, has been widely used for studying glycan-protein interactions186 and developing carbohydrate-based drugs.186,187 Earlier studies demonstrated that C-3-fluorinated N-acetylneuraminic acid worked as a competitive inhibitor for bacterial and viral sialidases.188 In addition, C-3-fluorinated sialosides were reported to inhibit C. perfringens bacterial sialidase189 and the activities of both hemagglutinins and neuraminidases of the influenza virus.189,190 A number of studies have demonstrated that family GH33 (CAZy) sialidases catalyze the hydrolysis in a two-step, double-displacement mechanism, in which a tyrosine residue serves as the catalytic nucleophile to form a unique covalent aryl-glycoside intermediate.191,192 2,3-Difluoro-N-acetylneuraminic acid (2,3-diF-Neu5Ac) was developed as a probe to confirm the covalent aryl-glycoside intermediate in T. cruzi trans-sialidase, where it attenuates glycosylation (k1) and deglycosylation (k2) rates in the catalytic cycle of the sialidases (Scheme 1).193,194 Specifically, the 3-F inductively destabilizes the formation of a positive charge during the transition states, thereby reducing the rates of glycosylation (k1) and deglycosylation (k2) (Scheme 1). However, the introduction of an anomeric fluorine, a good “leaving group”, could counteract the rate-retarding C3–F’s effect during glycosylation and increase k1 only. Another study obtained the cocrystal structure of a covalent intermediate complex at 1.2 Å resolution by cocrystallizing C. perfringens NanI sialidase with 2,3-diF-Neu5Ac.195 These demonstrate that 2,3-diF-Neu5Ac functions as a covalent inhibitor of sialidase, which is also called a mechanism-based inhibitor. 2α,3ax-diF-Neu5Ac was also demonstrated as a covalent influenza virus NA inhibitor, and the covalent adduct formed between the hydroxyl group of Tyr406 of NA and 2α,3ax-diF-Neu5Ac.196 Therefore, mechanism-based inhibitors are useful tools to trap and probe reaction intermediates in enzymatic reactions and also the active sites of the enzymes.
Scheme 1. Sialidase-Catalyzed Reaction with Mechanism-Based Inhibitor 2,3-diF-Neu5Ac in Glycosylation (k1) and Deglycosylation (k2) Steps.
2,3-diF-Sias have the potential to be developed into therapeutics as a novel class of sialidase inhibitors. It was reported that inactivation of T. cruzi trans-sialidase by 2,3-diF-Neu5Ac requires very high concentrations of inhibitor (5 mM), which was considered to be largely attributable to the rapid turnover of the covalent intermediate (high k2). The stability (half-life) of the covalent intermediate is the key to the inhibitory properties of this class of compound.191 Toward this aim, Hader et al. investigated the contribution by each hydroxyl group of Neu5Ac toward intermediate stabilization of sialidase-catalyzed hydrolysis.197 So far, several difluoro-Sias were investigated against some parasite trans-sialidases,131,192,198 bacterial sialidases,195,199 influenza A viral neuraminidases,196,200 and human cytosolic sialidase human Neu2 as well (Table 4).199 2(equatorial), 3(equatorial)-DiF-fluoro-Sia with a C4-guanidinium group showed superior in vitro anti-influenza A virus efficacy compared to its C4-ammonium or its 2(equatorial), 3(axial)-diF-Sia counterparts, which is comparable to zanamivir.200
Table 4. Mechanism-Based Sialidase Inhibitorsa.

| other
position |
||||||
|---|---|---|---|---|---|---|
| 2-position (R1) | 3-position (F) | 4-R2 | 5-R3 | 9-R4 | sialidase selectivity | ref |
| Fax | Feq | OH | Ac | OH | ND | (188) |
| -OPhNO2(eq) | Fax | OH | Ac | OH | influenza bacterial | (189) |
| -ODSPE(eq) | Fax | OH | Ac | OH | influenza | (189, 190) |
| Feq | Feq | OH | Ac | OH | influenza | (196, 200) |
| bacterial | (195, 199) | |||||
| protozoa | (192, 198) | |||||
| hNeu2 | (199) | |||||
| Feq | OH | Ac | N3 | hNeu2 | (199) | |
| Feq | OH | Gc | OH | hNeu2 | (199) | |
| Feq | Fax | OH | Ac | OH | bacterial | (199) |
| Fax | OH | Ac | N3 | bacterial | (199) | |
| Fax | guanidino | Ac | OH | influenza | (200) | |
ND: data not available; ax, axial; eq, equatorial.
Drug resistance has increased drastically to the current anti influenza therapy. Hence, it is urgent to develop potent broad-spectrum antiviral agents that can overcome viral resistance and treat a variety of viral infections. Mechanism-based covalent neuraminidase inhibitors, such as difluoro-Sia,200 have the potential to achieve both goals. However, possible nonspecific covalent bond formation with other biomolecules could cause side effects and toxicity. In particular, 3Fax-Neu5Ac could be converted to the corresponding CMP-Sias donor substrate in vivo, which could shut down the synthesis of sialylated glycan epitopes. Paulson and co-workers reported the peracetylated analog (P-3Fax-Neu5Ac) as a cell-permeable specific inhibitor of the sialyltransferases.201 This compound has well-known nephrotoxicity, which is a major barrier to its potential therapeutic use.202 Li et al. explored the selectivity of 2,3-difluoro-Sia by modifications at C-5 and/or C-9 position as well as varying C-3 fluorine stereochemistry (axial or equatorial).189 As a result, compounds with an axial fluorine at the C-3 position showed better inhibition (up to 100-fold) against bacterial sialidases compared to their C-3 equatorial counterparts. In addition, 9-azido-modified 2,3-diF-Neu5Ac showed increased inhibitory activity against bacterial sialidases; however, C-5-modification showed reduced inhibitory activity. Specifically, 9-azido-9-deoxy-2(equatorial),3(axial)-diF-Neu5Ac (2e3aDFNeu5Ac9N3) was developed as an effective inhibitor with a long effective duration selectively against C. perfringens (CpNanI) and V. cholerae sialidases (Table 4).199 Overall, increasing the specificity of binding to the target sites and reducing off-target toxicity are key factors for developing covalent sialidase inhibitors as potential drugs.
2.3. Suicide Substrate Inhibitors of Sialidase: Fluorinated Quinone Methide-Based Inhibitors
Suicide substrate inhibitors are a class of irreversible inhibitors that react with the enzyme residues through its reactive moieties generated during enzymatic reaction. 2-Difluoromethylphenyl glycosides were first reported as suicide substrate inhibitors of glycosidases in 1990 by Danzin et al., also called mechanism-based inhibitors.203 Specifically, the phenol aglycone is hydrolytically released by its target enzyme and subsequently transformed into fluorinated quinone methide. This aglycone is a highly reactive electrophilic species that could form a covalent bond with the nucleophilic amino acid residue of its target enzyme and irreversibly inhibits the activity of the enzyme (Figure 5a). Several suicide substrate inhibitors have been developed to glycosidases, including galactosidases204,205 and N-acetyl glucosaminidase.206 Later, 2-difluoromethyl-4-nitrophenyl glycoside of αNeu5Ac was reported as an irreversible inhibitor of trans-sialidase with an IC50 of 0.6 mM.207 This suicide substrate inhibitor prevents T. cruzi infection of mammalian cells and could serve as a lead compound for developing chemotherapeutics against Chagas disease. Kai et al. made a library of 2-difluoromethylphenyl-sialosides and identified a potent and selective inhibitor for V. cholerae and Neu2 sialidase (Figure 5a).208
Figure 5.

(a) Fluorinated quinone methide-based suicide substrate sialidase inhibitors and their covalent inhibition mechanism and (b) macrocycle-based suicide substrate sialidase inhibitor.
The activation efficiencies of suicide substrate inhibitors depend on the kcat of their target enzymes, and thus, they are called kcat inhibitors.209 Nevertheless, the inhibition efficiencies of this type of inhibitor depend on three factors: (i) the activations of trifluoromethylphenol groups to form respective reactive quinone methides, (ii) subsequent reaction with amino acid residues at the sialidase active site, and (iii) the diffusion of the difluoromethylphenol and reactive quinone group from the cavity of the active site of the sialidase.210,211 To overcome this diffusion problem, Kai et al. designed a macrocycle-based suicide substrate sialidase inhibitor by adding a covalent bond between the Sia and aglycone moiety (Figure 5b).212 By tethering with Sia, the difluoromethylphenol-type aglycone moiety of this inhibitor could stay within the active site of the sialidase after enzymatic cleavage of the sialoside bond and could form a covalent bond with a nucleophilic amino acid side chain of the sialidase. Inhibition assays for various sialidases showed that the irreversible inhibition of this macrocyclic compound depends on the kcat of the sialidase. Those sialidases with small kcat values (influenza viruses, Clostridium, Trypanosoma cruzi, and Neu2) were inhibited irreversibly, while those with high kcat values (S. typhimurium neuraminidase) were not affected by the inhibitor.208 Overall, suicide substrate sialidase inhibitors can be a versatile tool to elucidate the catalytic mechanism of a target enzyme and serve as drug candidates for certain sialidase targets and diseases.
2.4. Product Analogue Sialidase Inhibitors
Feedback inhibition is a normal biochemical process to control enzymatic reactions. In this process, the final product inhibits the enzyme and stops the reactions. Therefore, product analogues had been explored for developing novel enzyme inhibitors, which can be used to regulate enzyme activity and study enzyme function as well. Free Neu5Ac is a weak inhibitor of sialidases.213 Several Sia analogues that mimic the free Sia product structure and its enzyme binding features were developed as sialidase inhibitors and are discussed in this section.
Phosphonic Acid and Sulfo Acid Analogues of Sia
Sialidase active sites contain the triarginyl cluster, which are highly conserved across all known sialidases and could form strong electrostatic interactions with the anomeric carboxyl group of Sias and sialidase inhibitors.214 For example, in the case of influenza A NA, this triarginyl cluster consists of Arg118, Arg292, and Arg371.196,215 In the case of C. perfringens NanI sialidase, the arginine triad consists of Arg266, Arg555, and Arg615. Replacement of the carboxyl group with a phosphono group was proposed to improve the sialidase inhibitory activity, and therefore, phosphonic acid analogues of Sia were developed as sialidase inhibitors (Figure 6a).216−219 Obviously, the stronger electrostatic interactions of the anomeric phosphono with the conserved active site NA triarginyl cluster contribute to the increased activity. Interestingly, DANA phosphonates with a C-4-amino/guanidino group were also reported, which showed more potent activity against the NAs of avian and human influenza viruses, including the oseltamivir-resistant strains.220
Figure 6.

Phosphonic acid and sulfo acid analogues of Sia as product analogue sialidase inhibitors.
Neu5Ac-derived compounds bearing an anomeric sulfo functional group were also reported as a sialidase inhibitor (Figure 6b).221 For example, 2-decarboxy-2-deoxy-2-sulfo-N-acetylneuraminic acid was reported as a potent inhibitor of avian-origin H5N1 neuraminidase and drug-resistant His275Tyr NA as compared to the transition state analogue DANA. The sulfo-Neu5Ac analogue was demonstrated as a more potent inhibitor of influenza NA (up to 40-fold) and bacterial sialidase (up to 8.5-fold) relative to the corresponding anomeric phosphonic acids. The anomeric sulfo functional group could enhance electrostatic interactions with the triarginyl cluster. In addition, it serves as a strong electron withdrawing group that could destabilize oxocarbenium ion formation. These results confirm that anomeric sulfo modification offers another type of sialidase inhibitor, its sialidase selectivity deserves further investigation.
2,7-Anhydro-N-acetylneuraminic Acid (2,7-Anhydro-Neu5Ac) Derivatives
2,7-Anhydro-N-acetylneuraminic acid (2,7-anhydro-Neu5Ac, 1) (Figure 7) was initially found in rat urine222 and human wet cerumen223 as another kind of free Sia. It was found to be the product from the hydrolysis of sialosides catalyzed by intramolecular trans-sialidase (IT-sialidase L) from Macrobdella decora (leech),224,225 Gram-positive human pathogenic bacterium S. pneumoniae,226 and Gram-positive human gut commensal Ruminoccocus (R) gnavus.227 It was shown that 2,7-anhydro-Neu5Ac serves as a sole carbon source for the growth of R. gnavus in the Sia-rich host gut environment.228 Interestingly, a recent study indicates that 2,7-anhydro-Neu5Ac derivatives were selective sialidase inhibitors against S. pneumoniae sialidases SpNanB and SpNanC.229 On the basis of crystal structure analysis, several 2,7-anhydro-Neu5Ac derivatives were designed, synthesized, and tested for inhibitory activities against several GH33 family sialidases (Figure 7). 2,7-Anhydro-Neu5TFA (2) showed some inhibitory activity against SpNanA, SpNanB, AuSialidase, and VcSialidase. 2,7-Anhydro-Neu5Cyclohexyl (4) showed noticeable inhibitory activity against SpNanA, SpNanB, and SpNanC. This study demonstrated an effective product analogue strategy for exploring potential selective inhibitors of intramolecular trans-sialidases.
Figure 7.

Structures of 2,7-anhydro-Neu5Ac (1) and its derivatives 2–4.229.
2.5. Natural Products as Sialidase Inhibitors
The development of novel sialidase inhibitors has been largely based on synthetic compounds. Natural products provide diverse chemical scaffolds for drug discovery. A variety of natural compounds have been screened for antisialidase activity, specifically, diarylheptanoid katsumadain A, flavonoids artocarpin, apigenin, luteolin, gossypetin, oligostilbenes viniferin C and pedicularioside, and phenylpropanoid crenatoside. Among these natural compounds, artocarpin, kaempferol, and quercetin analogues were found to be the most potent sialidase inhibitors.148 This section summarizes major natural product sialidase inhibitors.
Natural Product Viral Sialidase Inhibitors
In the past, various natural compounds have been studied for influenza neuraminidase inhibition. A 2012 review illuminated the research efforts of the first decade of the 21st century (2000–2011), focusing on the structure and influenza neuraminidase inhibition activity of natural products.148 Approximately 150 natural product compounds were tested for their influenza neuraminidase-inhibiting potential during this period. Among those, flavonoids and (oligo)stilbenes were the most prominent scaffolds. A 2019 review updated recent discoveries of natural products as neuraminidase inhibitors by highlighting their structure, function, and inhibition mechanism.149 About 267 plant secondary metabolites were tested from 2011 to 2017 for their neuraminidase inhibition activity. More detailed information about various natural neuraminidase inhibitors and neuraminidase inhibition assays can be found in these two comprehensive reviews.148,149 Overall, natural compounds may serve as good lead structures for the discovery and development of potent viral neuraminidase inhibitors.
Natural Product Bacterial Sialidase Inhibitors
Bacterial sialidases play important roles in the pathogenesis of bacterial infection. Various natural compounds were tested for bacterial sialidase inhibition as well. Siastatin B (SB) was initially reported as an inhibitor of Streptomyces sialidase.157 SB has a 6-acetamido-3-piperidine carboxylate structure, which is similar with N-acetylneuraminic acid (Figure 8a). SB also shows inhibitory activity against C. perfringens sialidase activity.21,232,233 A recent study demonstrated that SB reduced the growth and survival rate of strain F4969 in the presence of Caco-2 cells.234 A curcumin derivative, 7-(3,4-dihydroxyphenyl)-5-hydroxy-1-(3-hydroxy-4-methoxyphenyl) hepta-1,4,6-trien-3-one (Figure 8b), was reported to inhibit S. pneumoniae NanA, V. cholerae, and C. perfringens sialidases.235 Another research indicated that the flavonoid diplacone showed inhibitory activity against C. perfringens sialidases (Cp-NanI).236 In addition, prenylated isoflavone237 and chromenone derivatives238 obtained from Flemingia philippinensis exhibit significant inhibition against bacterial sialidase. Park et al. reported phenolic metabolite Evernic acid (Figure 8c), isolated from the methanol extract of Usnea longissimi, displayed dose-dependent inhibition against bacterial sialidase.239 Interestingly, artocarpin and katsumadain A show inhibitory activity against both influenza and S. pneumoniae sialidases.240 Other curcumin and flavanoid derivatives were explored as bacterial sialidase inhibitors.235,236,241 Overall, natural products provide an alternative resource for the development of new bacterial sialidase inhibitors.
Figure 8.
Natural product bacterial sialidase inhibitors. (a) siastatin B, (b) 7-(3,4-dihydroxyphenyl)-5-hydroxy-1-(3-hydroxy-4-methoxyphenyl) hepta-1,4,6-trien-3-one, (c) evernic acid, (d) prenylated isoflavone, (e) chromenone, (f) artocarpin, (g) diplacone, and (h) katsumadain A.
Natural Product Protozoa Sialidase Inhibitors
The protozoan T. cruzi trans-sialidase (TcTS) is an attractive drug target for Chagas’ disease. Therefore, TcTS inhibitors could be used as therapeutics for the treatment of Chagas’ disease. DANA shows an IC50 value of several hundred micromolar against TcTS. Flavonoid and anthraquinone derivatives show strong inhibitory activity against TcTS.242 Specifically, 6-chloro-9,10-dihydro-4,5,7-trihydroxy-9,10-dioxo-2-anthracenecarboxylic acid (Figure 9a) was reported as a specific TcTS inhibitor with a IC50 value of 0.58 μM.242 The structure–activity relationship (SAR) analysis of the flavonoids revealed that apigenin has the minimal structure necessary for inhibition and may serve as a lead for drug discovery against Chagas’ disease.242
Figure 9.

Natural product protozoan sialidase inhibitor, (a) 6-chloro-9,10-dihydro-4,5,7-trihydroxy-9,10-dioxo-2-anthracenecarboxylic acid and human sialidase inhibitors, (b) Feddeiketone B, (c) 2,3-dihydroxy-1-(4-hydroxy-3,5-dimethoxyphenyl)-l-propanone, and (d) syringylglycerol.
Human Sialidase Natural Product Inhibitors
Natural compounds have been explored for human sialidase inhibition in recent studies. Albrecht et al. described the identification and evaluation of human Neu1 inhibitor extracted from Olyra latifolia L.243 Specifically, Feddeiketone B (Figure 9b), 2,3-dihydroxy-1-(4-hydroxy-3,5-dimethoxyphenyl)-l-propanone (Figure 9c), and syringylglycerol (Figure 9d) show inhibition effects on the cell membrane Neu1 sialidase activity. These compounds present structural similarities with DANA, and further investigation may be valuable for elucidating the biological functions of human sialidase and exploring potent human sialidase inhibitors.
3. Summary and Future Perspective
Sialidases (neuraminidases) catalyze the removal of Sia residue from sialoglycans and modulate their biological activity and thus are involved in numerous physiological and pathological processes. Expression of sialidases has been confirmed in a variety of organisms and viruses. In humans, four subtypes of sialidases (Neu1–4) have been identified; however, their functions have not been fully clarified. Sialidase inhibitors are highly needed for analysis of the precise function of sialidases and related physiological and pathological processes and development of novel drugs against specific sialidase. Over the past decades, sialidase inhibitors have received a great deal of interest, and various sialidase inhibitors have been developed and even used as drugs, such as anti-influenza drugs. Most sialidase inhibitors are developed by mimicking the transition state of sialidase-catalyzed hydrolysis reactions. In addition, mechanism-based inhibitors, suicide substrate inhibitors, and product analogue inhibitors have been developed. Various natural products have also been extensively isolated and tested for influenza virus, bacterial, and human sialidase inhibition. Nature provides an abundance of structurally diverse chemical scaffolds for lead structures in drug development. It is expected that new pharmacophore models from natural products will be identified, which will provide insights into the sialidase binding site, therefore helping develop selective and potent inhibitors of each sialidase isoenzyme.
Influenza virus neuraminidase inhibitors (oseltamivir, zanamivir, laninamivir, and peramivir) have been widely used in the treatment of influenza infection. However, the new strains of influenza virus are becoming resistant to current neuraminidase inhibitors, presenting serious threats to public health. Therefore, new neuraminidase inhibitors against drug-resistant influenza strains are in high demand. Bacterial pathogens produce sialidases for invasion, infection, and replication. Secondary pneumococcal infections cause severe complications in influenza patients. Therefore, development of inhibitors against both viral and bacterial sialidases could be of great interest. Dual inhibitors acting on both neuraminidases of S. pneumoniae and the influenza virus were demonstrated recently.240,244,245 In addition, it was observed that viral neuraminidase inhibitor oseltamivir has neuropsychiatric side effects.246 Therefore, for development of inhibitors of sialidases produced by pathogens, it is essential to test their activity on human sialidases and to know if they have side effects in vivo.
The surface of an influenza virus particle holds about 50 tetrameric neuraminidase spikes,247 each spike is a homotetrameric enzyme that could bind four sialosides.248 Therefore, multivalent influenza virus neuraminidase inhibitors have been proposed and tested.249 To date, dimeric, trimeric, tetrameric, and polymeric zanamivir derivatives linked through the C-7 hydroxyl group were prepared and showed outstanding antiviral potency. Previous multivalent influenza virus neuraminidase inhibitors were summarized in a 2007 review paper.249 Since then, several multivalent influenza virus neuraminidase inhibitors have been reported.250−252 Conjugation of the transition-state analogue DANA to polymeric scaffolds, on the other hand, produces highly potent inhibitors of bacterial sialidases.253 More than 4 orders of magnitude are added to the inhibitory potency of each clustered DANA for S. pneumoniae or B. thetaiotaomicron sialidases. This extends the multivalent concept to this important class of bacterial sialidases. This multivalent inhibition strategy provides interesting perspectives for other sialidase families, such as parasitic or human sialidases.
Human sialidases (Neu1–4) play important roles in many physiological processes but are also involved in numerous diseases and disorders.7 Therefore, human sialidases are promising pharmacological targets. Selective inhibitors of individual human sialidases are essential for a specific disease. Structure-based drug design is highly expected. Except for Neu2 sialidase, there are no 3D structure reported for Neu1, Neu3, and Neu4, which makes selective inhibitor design more difficult. Protein homology modeling based on the crystal structure of the Neu2 enzyme is used for human sialidase inhibitor development.95 It was found that the binding mode of the glycerol group of DANA is different between human, viral, and bacterial sialidases.242 This difference may play a role in substrate specificity and provides a new insight for designing selective sialidase inhibitors.
The overexpression and activation of Neu1 cause disorders and thus are of interest for regulation. On the other hand, cell surface relocation of Neu1 has been confirmed in different cell types including immune cells, where it could regulate the sialylation of several receptors and subsequent signaling pathways.28−52 The extensive review by Pshezhetsky et al. describes the key pathways in which desialylation of cell surface receptors by Neu1 modulates cellular signaling and molecular targeting.5 Therefore, regulation of Neu1 on the cell surface is highly desired. In other words, the cell surface Neu1 selective inhibitor is required to specifically regulate its activity on the cell surface and subsequent signaling pathways. Interestingly, natural product feddeiketone B, 2,3-dihydroxy-1-(4-hydroxy-3,5-dimethoxyphenyl)-l-propanone, and syringylglycerol (Figure 9) show inhibitory effects against Neu1-mediated sialidase activity at the plasma membrane.149 Further investigation of these compounds against Neu1 on the live cell surface is expected as they may be valuable for elucidating the biological functions of cell surface Neu1 and useful for regulating its subsequent signaling pathways. However, special attention should be paid when using natural compounds that contain catechol and quinone since they are known to contribute to false positives as pan-assay interference compounds (PAINs).254,255 They are known to interfere with bioassays via different mechanisms. For example, catechols can chelate metals and is reactive in the oxidized form to nucleophilic amino acids in proteins, such as cysteine and lysine.254
Overall, sialidases are involved in numerous physiological and pathological processes and thus are potential therapeutic targets. Development of sialidase inhibitors is important for studying the functions of sialidases and developing therapeutic drugs, but only a few selective inhibitors of sialidase have been developed. New selective sialidase inhibitors with novel scaffolds and new mechanisms of inhibition are highly expected. The important research areas to develop selective sialidase inhibitors are to (i) understand the mechanism of action, (ii) define cellular location of action, and (iii) identify the active site of each sialidase. In particular, determining the 3D structure of a sialidase is fundamental for understanding its function and properties and designing selective inhibitors. We expect that this Perspective gives an in-depth insight into several aspects of inhibitory mechanisms of sialidases and their inhibitors. Additionally, providing ample references about sialidase inhibitors with different mechanisms may be helpful for the ongoing study of sialidases and development of therapeutic agents.
Acknowledgments
The authors acknowledge the research support by research grant from the National Institutes of Health under award no. 1R15GM144881-01, Faculty Research Development Grant from Cleveland State University, and the Research Fund from the Center for Gene Regulation in Health and Disease (GRHD) at Cleveland State University.
Glossary
Abbreviations Used
- 2,7-anhydro-Neu5Ac
2,7-Anhydro-N-acetylneuraminic acid
- ApoB100
Apolipoprotein B100
- ATG5
Autophagy related 5
- ax
axial
- CAZy
carbohydrate active enzyme
- CD protein
cluster of differentiation protein
- 2,3-diF-Neu5Ac
2,3-difluoro-N-acetylneuraminic acid
- EGFR
epidermal growth factor receptor, eq: equatorial, Gal: galactose, GalNAc: N-acetyl galactosamine, GH, glycoside hydrolase
- GI
gastrointestinal
- HGF
hepatocyte growth factor
- HN
hemagglutinin-neuraminidase
- IR
insulin receptor
- LAP
latency-associated peptide
- MMP9
matrix metallopeptidase 9
- MUC1
mucin 1
- NA
neuraminidase
- Neu5Ac
N-acetylneuraminic acid
- Neu5Ac2en2
2-deoxy-2,3-didehydro-N-acetylneuraminic acid
- PPCA
protective protein/cathepsin A
- Sias
sialic acids
- SB
siastatin B
- TGF-b
transforming growth factor beta
- TLR
toll-like receptor
- TrkA
tropomyosin receptor kinase A.
Biographies
Joseph M. Keil earned a Bachelors in Pharmaceutical Science at Cleveland State University in 2018. He is currently a Clinical and Bioanalytical Chemistry Ph.D. graduate student in the department of Chemistry at Cleveland State University. His research interests include sialic acid derivative synthesis, sialidase inhibitor synthesis, recombinant thrombomodulin expression, and its glyco-engineering.
Garrett R. Rafn is a senior undergraduate student at Cleveland State University studying Health Science and Pre-Medicine. He is currently working as a research assistant in the Sun lab, Department of Chemistry and Center for Gene Regulation in Health and Disease (GRHD) at Cleveland State University.
Isaac M. Turan received his certification by the American Chemical Society in 2018 along with his Bachelor’s degree in Chemistry from Cleveland State University. He is currently engaged in graduate studies and is pursuing a Ph.D. in Clinical and Bioanalytical Chemistry with a focus on Cellular and Molecular Medicine. His interests and specialties include synthetic chemistry and NMR characterization and evaluation of sialidase inhibitors and substrates.
Majdi A. Aljohani is a third-year Clinical and Bioanalytical Chemistry Ph.D. student at Cleveland State University. He received a bachelor’s degree in Medical Laboratory Sciences from the University of Tabuk and a Master’s degree in Biomedical Sciences from Long Island University in New York, USA. He is interested in Clinical Chemistry, Immunology, and Immunohematology.
Reza Sahebjam-Atabaki earned a Bachelors in Biology and Chemistry double major at Cleveland State University in 2013. He is a second-year graduate student at Cleveland State University now, pursuing a Ph.D. degree in Clinical and Bioanalytical Chemistry. His research interests include cell surface sialidase profiling and inhibition in epidermal cells.
Xue-Long Sun received a Ph.D. in Pharmaceutical Chemistry from Kitasato University School of Pharmacy (Japan) in 1997. Then, he had worked on synthesis of sialidase and sialyltransferase inhibitors at RIKEN Institute (Japan) as a basic science special research fellow (Drs. Chi-Huey Wong, Osamu Kanie, and Yukishige Ito) and antithrombotic biomaterials at Emory University School of Medicine as a NIH-funded research fellow (Dr. Elliot L. Chaikof). He started his academic career at Emory University School of Medicine (2002–2006) and has moved to Cleveland State University as a Full Professor of Medicinal Chemistry (primary) and Biomedical Engineering (secondary) now. His group is working on Sia-focused glycomics, chemical glycobiology and medicinal chemistry, glycopolymers, and glyco-liposomes-based biomaterials. Furthermore, he has been working recombinant thrombomodulin expression and glyco-engineering research.
The authors declare no competing financial interest.
References
- Ghosh S.Sialic Acid and Biology of Life: An Introduction. In Sialic Acids and Sialoglycoconjugates in the Biology of Life, Health and Disease; Elsevier, 2020; pp 1–61, 10.1016/b978-0-12-816126-5.00001-9. [DOI] [Google Scholar]
- Schauer R.; Kamerling J. P.. Exploration of the Sialic Acid World. In Advances in Carbohydrate Chemistry and Biochemistry; Academic Press Inc., 2018; Vol. 75, pp 1–213, 10.1016/bs.accb.2018.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao H; Chen X.. General Consideration on Sialic Acid Chemistry. In Carbohydrate Microarrays. Methods in Molecular Biology (Methods and Protocols); Humana Press, 2012; Vol. 808, pp 31–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohen M.; Varki A. The Sialome-Far More than the Sum of Its Parts. OMICS. 2010, 14 (4), 455–464. 10.1089/omi.2009.0148. [DOI] [PubMed] [Google Scholar]
- Pshezhetsky A. v.; Ashmarina L. I. Desialylation of Surface Receptors as a New Dimension in Cell Signaling. Biochemistry (Moscow) 2013, 78, 736–745. 10.1134/S0006297913070067. [DOI] [PubMed] [Google Scholar]
- Miyagi T.; Yamaguchi K. Mammalian Sialidases: Physiological and Pathological Roles in Cellular Functions. Glycobiology 2012, 22 (7), 880–896. 10.1093/glycob/cws057. [DOI] [PubMed] [Google Scholar]
- Glanz V. Yu.; Myasoedova V. A.; Grechko A. v.; Orekhov A. N. Sialidase Activity in Human Pathologies. Eur. J. Pharmacol. 2019, 842, 345–350. 10.1016/j.ejphar.2018.11.014. [DOI] [PubMed] [Google Scholar]
- Bennasroune A.; Romier-Crouzet B.; Blaise S.; Laffargue M.; Efremov R. G.; Martiny L.; Maurice P.; Duca L. Elastic Fibers and Elastin Receptor Complex: Neuraminidase-1 Takes the Center Stage. Matrix Biology 2019, 84, 57–67. 10.1016/j.matbio.2019.06.007. [DOI] [PubMed] [Google Scholar]
- Wei M.; Wang P. G. Desialylation in Physiological and Pathological Processes: New Target for Diagnostic and Therapeutic Development. Prog. Mol. Biol. Transl. Sci. 2019, 162, 25–57. 10.1016/bs.pmbts.2018.12.001. [DOI] [PubMed] [Google Scholar]
- Monti E.; Bonten E.; Bresciani R.; Venerando B.; Borsani G.; Schauer R.; Tettamanti G. Sialidases in Vertebrates. Advances in Carbohydrate Chemistry and Biochemistry 2010, 64, 403–479. 10.1016/S0065-2318(10)64007-3. [DOI] [PubMed] [Google Scholar]
- Schenkman S.; Eichinger D.; Pereira M. E. A.; Nussenzweig V. Structural and Functional Properties of Trypanosoma Trans -Sialidase. Annu. Rev. Microbiol. 1994, 48 (1), 499–523. 10.1146/annurev.mi.48.100194.002435. [DOI] [PubMed] [Google Scholar]
- Tailford L. E.; Owen C. D.; Walshaw J.; Crost E. H.; Hardy-Goddard J.; le Gall G.; de Vos W. M.; Taylor G. L.; Juge N. Discovery of Intramolecular Trans-Sialidases in Human Gut Microbiota Suggests Novel Mechanisms of Mucosal Adaptation. Nat. Commun. 2015, 6 (1), 7624. 10.1038/ncomms8624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crost E. H.; Tailford L. E.; Monestier M.; Swarbreck D.; Henrissat B.; Crossman L. C.; Juge N. The Mucin-Degradation Strategy of Ruminococcus Gnavus : The Importance of Intramolecular Trans -Sialidases. Gut Microbes. 2016, 7 (4), 302–312. 10.1080/19490976.2016.1186334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takimoto T.; Taylor G. L.; Connaris H. C.; Crennell S. J.; Portner A. Role of the Hemagglutinin-Neuraminidase Protein in the Mechanism of Paramyxovirus-Cell Membrane Fusion. J. Virol. 2002, 76 (24), 13028–13033. 10.1128/JVI.76.24.13028-13033.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lawrence M. C.; Borg N. A.; Streltsov V. A.; Pilling P. A.; Epa V. C.; Varghese J. N.; McKimm-Breschkin J. L.; Colman P. M. Structure of the Haemagglutinin-Neuraminidase from Human Parainfluenza Virus Type III. J. Mol. Biol. 2004, 335 (5), 1343–1357. 10.1016/j.jmb.2003.11.032. [DOI] [PubMed] [Google Scholar]
- Chuzel L.; Ganatra M. B.; Rapp E.; Henrissat B.; Taron C. H. Functional Metagenomics Identifies an Exosialidase with an Inverting Catalytic Mechanism That Defines a New Glycoside Hydrolase Family (GH156). J. Biol. Chem. 2018, 293 (47), 18138–18150. 10.1074/jbc.RA118.003302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luzina I. G.; Lockatell V.; Hyun S. W.; Kopach P.; Kang P. H.; Noor Z.; Liu A.; Lillehoj E. P.; Lee C.; Miranda-Ribera A.; Todd N. W.; Goldblum S. E.; Atamas S. P. Elevated Expression of NEU1 Sialidase in Idiopathic Pulmonary Fibrosis Provokes Pulmonary Collagen Deposition, Lymphocytosis, and Fibrosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2016, 310 (10), L940–L954. 10.1152/ajplung.00346.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonten E.; van der Spoel A.; Fornerod M.; Grosveld G.; D’Azzo A. Characterization of Human Lysosomal Neuraminidase Defines the Molecular Basis of the Metabolic Storage Disorder Sialidosis. Genes Dev. 1996, 10 (24), 3156–3169. 10.1101/gad.10.24.3156. [DOI] [PubMed] [Google Scholar]
- Pshezhetsky A. v.; Richard C.; Michaud L.; Igdoura S.; Wang S.; Elsliger M.-A.; Qu J.; Leclerc D.; Gravel R.; Dallaire L.; Potier M. Cloning, Expression and Chromosomal Mapping of Human Lysosomal Sialidase and Characterization of Mutations in Sialidosis. Nat. Genet. 1997, 15 (3), 316–320. 10.1038/ng0397-316. [DOI] [PubMed] [Google Scholar]
- Juge N.; Tailford L.; Owen C. D. Sialidases from Gut Bacteria: A Mini-Review. Biochem. Soc. Trans. 2016, 44 (1), 166–175. 10.1042/BST20150226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J.; McClane B. A. Contributions of NanI Sialidase to Caco-2 Cell Adherence by Clostridium Perfringens Type A and C Strains Causing Human Intestinal Disease. Infect. Immun. 2014, 82 (11), 4620–4630. 10.1128/IAI.02322-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Comelli E. M.; Amado M.; Lustig S. R.; Paulson J. C. Identification and Expression of Neu4, a Novel Murine Sialidase. Gene. 2003, 321, 155–161. 10.1016/j.gene.2003.08.005. [DOI] [PubMed] [Google Scholar]
- Tringali C.; Papini N.; Fusi P.; Croci G.; Borsani G.; Preti A.; Tortora P.; Tettamanti G.; Venerando B.; Monti E. Properties of Recombinant Human Cytosolic Sialidase HsNEU2. J. Biol. Chem. 2004, 279 (5), 3169–3179. 10.1074/jbc.M308381200. [DOI] [PubMed] [Google Scholar]
- Zanchetti G.; Colombi P.; Manzoni M.; Anastasia L.; Caimi L.; Borsani G.; Venerando B.; Tettamanti G.; Preti A.; Monti E.; Bresciani R. Sialidase NEU3 Is a Peripheral Membrane Protein Localized on the Cell Surface and in Endosomal Structures. Biochem. J. 2007, 408 (2), 211–219. 10.1042/BJ20070503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonten E. J.; Campos Y.; Zaitsev V.; Nourse A.; Waddell B.; Lewis W.; Taylor G.; d’Azzo A. Heterodimerization of the Sialidase NEU1 with the Chaperone Protective Protein/Cathepsin A Prevents Its Premature Oligomerization. J. Biol. Chem. 2009, 284 (41), 28430–28441. 10.1074/jbc.M109.031419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hata K.; Koseki K.; Yamaguchi K.; Moriya S.; Suzuki Y.; Yingsakmongkon S.; Hirai G.; Sodeoka M.; von Itzstein M.; Miyagi T. Limited Inhibitory Effects of Oseltamivir and Zanamivir on Human Sialidases. Antimicrob. Agents Chemother. 2008, 52 (10), 3484–3491. 10.1128/AAC.00344-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smutova V.; Albohy A.; Pan X.; Korchagina E.; Miyagi T.; Bovin N.; Cairo C. W.; Pshezhetsky A. v. Structural Basis for Substrate Specificity of Mammalian Neuraminidases. PLoS. One. 2014, 9 (9), e106320 10.1371/journal.pone.0106320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonten E. J.; Annunziata I.; D’Azzo A. Lysosomal Multienzyme Complex: Pros and Cons of Working Together. Cell. Mol. Life Sci. 2014, 71 (11), 2017–2032. 10.1007/s00018-013-1538-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kijimoto-Ochiai S.; Matsumoto-Mizuno T.; Kamimura D.; Murakami M.; Kobayashi M.; Matsuoka I.; Ochiai H.; Ishida H.; Kiso M.; Kamimura K.; Koda T. Existence of NEU1 Sialidase on Mouse Thymocytes Whose Natural Substrate Is CD5. Glycobiology. 2018, 28 (5), 306–317. 10.1093/glycob/cwy009. [DOI] [PubMed] [Google Scholar]
- Gilmour A. M.; Abdulkhalek S.; Cheng T. S. W.; Alghamdi F.; Jayanth P.; O’Shea L. K.; Geen O.; Arvizu L. A.; Szewczuk M. R. A Novel Epidermal Growth Factor Receptor-Signaling Platform and Its Targeted Translation in Pancreatic Cancer. Cell Signal. 2013, 25 (12), 2587–2603. 10.1016/j.cellsig.2013.08.008. [DOI] [PubMed] [Google Scholar]
- Abdulkhalek S.; Szewczuk M. R. Neu1 Sialidase and Matrix Metalloproteinase-9 Cross-Talk Regulates Nucleic Acid-Induced Endosomal TOLL-like Receptor-7 and −9 Activation, Cellular Signaling and pro-Inflammatory Responses. Cell Signal. 2013, 25 (11), 2093–2105. 10.1016/j.cellsig.2013.06.010. [DOI] [PubMed] [Google Scholar]
- Abdulkhalek S.; Amith S. R.; Franchuk S. L.; Jayanth P.; Guo M.; Finlay T.; Gilmour A.; Guzzo C.; Gee K.; Beyaert R.; Szewczuk M. R. Neu1 Sialidase and Matrix Metalloproteinase-9 Cross-Talk Is Essential for Toll-like Receptor Activation and Cellular Signaling. J. Biol. Chem. 2011, 286 (42), 36532–36549. 10.1074/jbc.M111.237578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jayanth P.; Amith S. R.; Gee K.; Szewczuk M. R. Neu1 Sialidase and Matrix Metalloproteinase-9 Cross-Talk Is Essential for Neurotrophin Activation of Trk Receptors and Cellular Signaling. Cell Signal. 2010, 22 (8), 1193–1205. 10.1016/j.cellsig.2010.03.011. [DOI] [PubMed] [Google Scholar]
- Amith S. R.; Jayanth P.; Franchuk S.; Siddiqui S.; Seyrantepe V.; Gee K.; Basta S.; Beyaert R.; Pshezhetsky A. v.; Szewczuk M. R. Dependence of Pathogen Molecule-Induced Toll-like Receptor Activation and Cell Function on Neu1 Sialidase. Glycoconj. J. 2009, 26 (9), 1197–1212. 10.1007/s10719-009-9239-8. [DOI] [PubMed] [Google Scholar]
- Karmakar J.; Roy S.; Mandal C. Modulation of TLR4 Sialylation Mediated by a Sialidase Neu1 and Impairment of Its Signaling in Leishmania Donovani Infected Macrophages. Front. Immunol. 2019, 10, 2360. 10.3389/fimmu.2019.02360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allendorf D. H.; Franssen E. H.; Brown G. C. Lipopolysaccharide Activates Microglia via Neuraminidase 1 Desialylation of Toll-like Receptor 4. J. Neurochem. 2020, 155 (4), 403–416. 10.1111/jnc.15024. [DOI] [PubMed] [Google Scholar]
- Pshezhetsky A. v.; Hinek A. Where Catabolism Meets Signaling: Neuraminidase 1 as a Modulator of Cell Receptors. Glycoconj. J. 2011, 28 (7), 441–452. 10.1007/s10719-011-9350-5. [DOI] [PubMed] [Google Scholar]
- Hinek A.; Pshezhetsky A. v.; von Itzstein M.; Starcher B. Lysosomal Sialidase (Neuraminidase-1) Is Targeted to the Cell Surface in a Multiprotein Complex That Facilitates Elastic Fiber Assembly. J. Biol. Chem. 2006, 281 (6), 3698–3710. 10.1074/jbc.M508736200. [DOI] [PubMed] [Google Scholar]
- Liang F.; Seyrantepe V.; Landry K.; Ahmad R.; Ahmad A.; Stamatos N. M.; Pshezhetsky A. v. Monocyte Differentiation Up-Regulates the Expression of the Lysosomal Sialidase, Neu1, and Triggers Its Targeting to the Plasma Membrane via Major Histocompatibility Complex Class II-Positive Compartments. J. Biol. Chem. 2006, 281 (37), 27526–27538. 10.1074/jbc.M605633200. [DOI] [PubMed] [Google Scholar]
- Nan X.; Carubelli I.; Stamatos N. M. Sialidase Expression in Activated Human T Lymphocytes Influences Production of IFN-γ. J. Leukoc. Biol. 2007, 81 (1), 284–296. 10.1189/jlb.1105692. [DOI] [PubMed] [Google Scholar]
- Stamatos N. M.; Carubelli I.; van de Vlekkert D.; Bonten E. J.; Papini N.; Feng C.; Venerando B.; DˈAzzo A.; Cross A. S.; Wang L.-X.; Gomatos P. J. LPS-Induced Cytokine Production in Human Dendritic Cells Is Regulated by Sialidase Activity. J. Leukoc. Biol. 2010, 88 (6), 1227–1239. 10.1189/jlb.1209776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Avila F.; Tringali C.; Papini N.; Anastasia L.; Croci G.; Massaccesi L.; Monti E.; Tettamanti G.; Venerando B. Identification of Lysosomal Sialidase NEU1 and Plasma Membrane Sialidase NEU3 in Human Erythrocytes. J. Cell. Biochem. 2013, 114 (1), 204–211. 10.1002/jcb.24355. [DOI] [PubMed] [Google Scholar]
- Alghamdi F.; Guo M.; Abdulkhalek S.; Crawford N.; Amith S. R.; Szewczuk M. R. A Novel Insulin Receptor-Signaling Platform and Its Link to Insulin Resistance and Type 2 Diabetes. Cell Signal. 2014, 26 (6), 1355–1368. 10.1016/j.cellsig.2014.02.015. [DOI] [PubMed] [Google Scholar]
- Chen G.-Y.; Brown N. K.; Wu W.; Khedri Z.; Yu H.; Chen X.; van de Vlekkert D.; D’Azzo A.; Zheng P.; Liu Y. Broad and Direct Interaction between TLR and Siglec Families of Pattern Recognition Receptors and Its Regulation by Neu1. Elife. 2014, 3, e04066 10.7554/eLife.04066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maurice P.; Baud S.; Bocharova O. v.; Bocharov E. v.; Kuznetsov A. S.; Kawecki C.; Bocquet O.; Romier B.; Gorisse L.; Ghirardi M.; Duca L.; Blaise S.; Martiny L.; Dauchez M.; Efremov R. G.; Debelle L. New Insights into Molecular Organization of Human Neuraminidase-1: Transmembrane Topology and Dimerization Ability. Sci. Rep. 2016, 6 (1), 38363. 10.1038/srep38363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scandolera A.; Odoul L.; Salesse S.; Guillot A.; Blaise S.; Kawecki C.; Maurice P.; el Btaouri H.; Romier-Crouzet B.; Martiny L.; Debelle L.; Duca L. The Elastin Receptor Complex: A Unique Matricellular Receptor with High Anti-Tumoral Potential. Front. Pharmacol. 2016, 7, 32. 10.3389/fphar.2016.00032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Abdulkhalek S.; Guo M.; Amith S. R.; Jayanth P.; Szewczuk M. R. G-Protein Coupled Receptor Agonists Mediate Neu1 Sialidase and Matrix Metalloproteinase-9 Cross-Talk to Induce Transactivation of TOLL-like Receptors and Cellular Signaling. Cell Signal. 2012, 24 (11), 2035–2042. 10.1016/j.cellsig.2012.06.016. [DOI] [PubMed] [Google Scholar]
- Lukong K. E.; Seyrantepe V.; Landry K.; Trudel S.; Ahmad A.; Gahl W. A.; Lefrancois S.; Morales C. R.; Pshezhetsky A. v. Intracellular Distribution of Lysosomal Sialidase Is Controlled by the Internalization Signal in Its Cytoplasmic Tail. J. Biol. Chem. 2001, 276 (49), 46172–46181. 10.1074/jbc.M104547200. [DOI] [PubMed] [Google Scholar]
- Shkandina T.; Herrmann M.; Bilyy R. Sweet Kiss of Dying Cell: Sialidase Activity on Apoptotic Cell Is Able to Act toward Its Neighbors. Autoimmunity 2012, 45 (8), 574–578. 10.3109/08916934.2012.719951. [DOI] [PubMed] [Google Scholar]
- Wu Y.; Lan C.; Ren D.; Chen G.-Y. Induction of Siglec-1 by Endotoxin Tolerance Suppresses the Innate Immune Response by Promoting TGF-Β1 Production. J. Biol. Chem. 2016, 291 (23), 12370–12382. 10.1074/jbc.M116.721258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sasaki N.; Itakura Y.; Toyoda M. Sialylation Regulates Myofibroblast Differentiation of Human Skin Fibroblasts. Stem Cell Res. Ther. 2017, 8 (1), 81. 10.1186/s13287-017-0534-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wahart A.; Hocine T.; Albrecht C.; Henry A.; Sarazin T.; Martiny L.; el Btaouri H.; Maurice P.; Bennasroune A.; Romier-Crouzet B.; Blaise S.; Duca L. Role of Elastin Peptides and Elastin Receptor Complex in Metabolic and Cardiovascular Diseases. FEBS. J. 2019, 286 (15), 2980–2993. 10.1111/febs.14836. [DOI] [PubMed] [Google Scholar]
- van der Wal D. E.; Davis A. M.; Mach M.; Marks D. C. The Role of Neuraminidase 1 and 2 in Glycoprotein Ibα-Mediated Integrin ΑIIbβ3 Activation. Haematologica. 2020, 105 (4), 1081–1094. 10.3324/haematol.2019.215830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nath S.; Mandal C.; Chatterjee U.; Mandal C. Association of Cytosolic Sialidase Neu2 with Plasma Membrane Enhances Fas-Mediated Apoptosis by Impairing PI3K-Akt/MTOR-Mediated Pathway in Pancreatic Cancer Cells. Cell Death Dis. 2018, 9 (2), 210. 10.1038/s41419-017-0191-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kijimoto-Ochiai S.; Doi N.; Fujii M.; Go S.; Kabayama K.; Moriya S.; Miyagi T.; Koda T. Possible Association of Neu2 with Plasma Membrane Fraction from Mouse Thymus Exhibited Sialidase Activity with Fetuin at PH 7.0 but Not at PH 4.5. Microbiol. Immunol. 2013, 57 (8), 569–582. 10.1111/1348-0421.12076. [DOI] [PubMed] [Google Scholar]
- Shiozaki K.; Yamaguchi K.; Takahashi K.; Moriya S.; Miyagi T. Regulation of Sialyl Lewis Antigen Expression in Colon Cancer Cells by Sialidase NEU4. J. Biol. Chem. 2011, 286 (24), 21052–21061. 10.1074/jbc.M111.231191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Finlay T. M.; Jayanth P.; Amith S. R.; Gilmour A.; Guzzo C.; Gee K.; Beyaert R.; Szewczuk M. R. Thymoquinone from Nutraceutical Black Cumin Oil Activates Neu4 Sialidase in Live Macrophage, Dendritic, and Normal and Type I Sialidosis Human Fibroblast Cells via GPCR Gαi Proteins and Matrix Metalloproteinase-9. Glycoconj. J. 2010, 27 (3), 329–348. 10.1007/s10719-010-9281-6. [DOI] [PubMed] [Google Scholar]
- Demina E. P.; Smutova V.; Pan X.; Fougerat A.; Guo T.; Zou C.; Chakraberty R.; Snarr B. D.; Shiao T. C.; Roy R.; Orekhov A. N.; Miyagi T.; Laffargue M.; Sheppard D. C.; Cairo C. W.; Pshezhetsky A. v. Neuraminidases 1 and 3 Trigger Atherosclerosis by Desialylating Low-Density Lipoproteins and Increasing Their Uptake by Macrophages. J. Am. Heart. Assoc. 2021, 10 (4), e018756 10.1161/JAHA.120.018756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng C.; Zhang L.; Almulki L.; Faez S.; Whitford M.; Hafezi-Moghadam A.; Cross A. S. Endogenous PMN Sialidase Activity Exposes Activation Epitope on CD11b/CD18 Which Enhances Its Binding Interaction with ICAM-1. J. Leukoc. Biol. 2011, 90 (2), 313–321. 10.1189/jlb.1210708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kawecki C.; Bocquet O.; Schmelzer C. E. H.; Heinz A.; Ihling C.; Wahart A.; Romier B.; Bennasroune A.; Blaise S.; Terryn C.; Linton K. J.; Martiny L.; Duca L.; Maurice P. Identification of CD36 as a New Interaction Partner of Membrane NEU1: Potential Implication in the pro-Atherogenic Effects of the Elastin Receptor Complex. Cell. Mol. Life Sci. 2019, 76 (4), 791–807. 10.1007/s00018-018-2978-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bocquet O.; Tembely D.; Rioult D.; Terryn C.; Romier B.; Bennasroune A.; Blaise S.; Sartelet H.; Martiny L.; Duca L.; Maurice P. Characterization of Novel Interactions with Membrane NEU1 Highlights New Regulatory Functions for the Elastin Receptor Complex in Monocyte Interaction with Endothelial Cells. Cell Biosci. 2021, 11 (1), 206. 10.1186/s13578-021-00718-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howlader Md. A.; Li C.; Zou C.; Chakraberty R.; Ebesoh N.; Cairo C. W. Neuraminidase-3 Is a Negative Regulator of LFA-1 Adhesion. Front. Chem. 2019, 7, 791. 10.3389/fchem.2019.00791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee C.; Liu A.; Miranda-Ribera A.; Hyun S. W.; Lillehoj E. P.; Cross A. S.; Passaniti A.; Grimm P. R.; Kim B.-Y.; Welling P. A.; Madri J. A.; DeLisser H. M.; Goldblum S. E. NEU1 Sialidase Regulates the Sialylation State of CD31 and Disrupts CD31-Driven Capillary-like Tube Formation in Human Lung Microvascular Endothelia. J. Biol. Chem. 2014, 289 (13), 9121–9135. 10.1074/jbc.M114.555888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J.; van der Wal D. E.; Zhu G.; Xu M.; Yougbare I.; Ma L.; Vadasz B.; Carrim N.; Grozovsky R.; Ruan M.; Zhu L.; Zeng Q.; Tao L.; Zhai Z.; Peng J.; Hou M.; Leytin V.; Freedman J.; Hoffmeister K. M.; Ni H. Desialylation Is a Mechanism of Fc-Independent Platelet Clearance and a Therapeutic Target in Immune Thrombocytopenia. Nat. Commun. 2015, 6 (1), 7737. 10.1038/ncomms8737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jansen A. J. G.; Josefsson E. C.; Rumjantseva V.; Liu Q. P.; Falet H.; Bergmeier W.; Cifuni S. M.; Sackstein R.; von Andrian U. H.; Wagner D. D.; Hartwig J. H.; Hoffmeister K. M. Desialylation Accelerates Platelet Clearance after Refrigeration and Initiates GPIbα Metalloproteinase-Mediated Cleavage in Mice. Blood. 2012, 119 (5), 1263–1273. 10.1182/blood-2011-05-355628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katoh S. Critical Involvement of CD44 in T Helper Type 2 Cell-Mediated Eosinophilic Airway Inflammation in a Mouse Model of Acute Asthma. Front. Immunol. 2022, 12, 811600. 10.3389/fimmu.2021.811600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faller C. E.; Guvench O. Terminal Sialic Acids on CD44 N-Glycans Can Block Hyaluronan Binding by Forming Competing Intramolecular Contacts with Arginine Sidechains. Proteins: Struct. Funct. Genet. 2014, 82 (11), 3079–3089. 10.1002/prot.24668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katoh S.; Maeda S.; Fukuoka H.; Wada T.; Moriya S.; Mori A.; Yamaguchi K.; Senda S.; Miyagi T. A Crucial Role of Sialidase Neu1 in Hyaluronan Receptor Function of CD44 in T Helper Type 2-Mediated Airway Inflammation of Murine Acute Asthmatic Model. Clin. Exp. Immunol. 2010, 161 (2), 233–241. 10.1111/j.1365-2249.2010.04165.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seyrantepe V.; Iannello A.; Liang F.; Kanshin E.; Jayanth P.; Samarani S.; Szewczuk M. R.; Ahmad A.; Pshezhetsky A. v. Regulation of Phagocytosis in Macrophages by Neuraminidase 1. J. Biol. Chem. 2010, 285 (1), 206–215. 10.1074/jbc.M109.055475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uemura T.; Shiozaki K.; Yamaguchi K.; Miyazaki S.; Satomi S.; Kato K.; Sakuraba H.; Miyagi T. Contribution of Sialidase NEU1 to Suppression of Metastasis of Human Colon Cancer Cells through Desialylation of Integrin Β4. Oncogene. 2009, 28 (9), 1218–1229. 10.1038/onc.2008.471. [DOI] [PubMed] [Google Scholar]
- Zanoteli E.; de Vlekkert D. v.; Bonten E. J.; Hu H.; Mann L.; Gomero E. M.; Harris A. J.; Ghersi G.; d'Azzo A. Muscle Degeneration in Neuraminidase 1-Deficient Mice Results from Infiltration of the Muscle Fibers by Expanded Connective Tissue. Biochim. Biophys. Acta. Mol. Basis Dis. 2010, 1802 (7–8), 659–672. 10.1016/j.bbadis.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yogalingam G.; Bonten E. J.; van de Vlekkert D.; Hu H.; Moshiach S.; Connell S. A.; D’Azzo A. Neuraminidase 1 Is a Negative Regulator of Lysosomal Exocytosis. Dev. Cell. 2008, 15 (1), 74–86. 10.1016/j.devcel.2008.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hinek A.; Bodnaruk T. D.; Bunda S.; Wang Y.; Liu K. Neuraminidase-1, a Subunit of the Cell Surface Elastin Receptor, Desialylates and Functionally Inactivates Adjacent Receptors Interacting with the Mitogenic Growth Factors PDGF-BB and IGF-2. Am. J. Pathol. 2008, 173 (4), 1042–1056. 10.2353/ajpath.2008.071081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arabkhari M.; Bunda S.; Wang Y.; Wang A.; Pshezhetsky A. v.; Hinek A. Desialylation of Insulin Receptors and IGF-1 Receptors by Neuraminidase-1 Controls the Net Proliferative Response of L6Myoblasts to Insulin. Glycobiology. 2010, 20 (5), 603–616. 10.1093/glycob/cwq010. [DOI] [PubMed] [Google Scholar]
- Blaise S.; Romier B.; Kawecki C.; Ghirardi M.; Rabenoelina F.; Baud S.; Duca L.; Maurice P.; Heinz A.; Schmelzer C. E. H.; Tarpin M.; Martiny L.; Garbar C.; Dauchez M.; Debelle L.; Durlach V. Elastin-Derived Peptides Are New Regulators of Insulin Resistance Development in Mice. Diabetes. 2013, 62 (11), 3807–3816. 10.2337/db13-0508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fougerat A.; Pan X.; Smutova V.; Heveker N.; Cairo C. W.; Issad T.; Larrivée B.; Medin J. A.; Pshezhetsky A. v. Neuraminidase 1 Activates Insulin Receptor and Reverses Insulin Resistance in Obese Mice. Mol. Metab. 2018, 12, 76–88. 10.1016/j.molmet.2018.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dridi L.; Seyrantepe V.; Fougerat A.; Pan X.; Bonneil É.; Thibault P.; Moreau A.; Mitchell G. A.; Heveker N.; Cairo C. W.; Issad T.; Hinek A.; Pshezhetsky A. v. Positive Regulation of Insulin Signaling by Neuraminidase 1. Diabetes. 2013, 62 (7), 2338–2346. 10.2337/db12-1825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lillehoj E. P.; Hyun S. W.; Feng C.; Zhang L.; Liu A.; Guang W.; Nguyen C.; Luzina I. G.; Atamas S. P.; Passaniti A.; Twaddell W. S.; Puché A. C.; Wang L.-X.; Cross A. S.; Goldblum S. E. NEU1 Sialidase Expressed in Human Airway Epithelia Regulates Epidermal Growth Factor Receptor (EGFR) and MUC1 Protein Signaling. J. Biol. Chem. 2012, 287 (11), 8214–8231. 10.1074/jbc.M111.292888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mozzi A.; Forcella M.; Riva A.; Difrancesco C.; Molinari F.; Martin V.; Papini N.; Bernasconi B.; Nonnis S.; Tedeschi G.; Mazzucchelli L.; Monti E.; Fusi P.; Frattini M. NEU3 Activity Enhances EGFR Activation without Affecting EGFR Expression and Acts on Its Sialylation Levels. Glycobiology. 2015, 25 (8), 855–868. 10.1093/glycob/cwv026. [DOI] [PubMed] [Google Scholar]
- Romier B.; Ivaldi C.; Sartelet H.; Heinz A.; Schmelzer C. E. H.; Garnotel R.; Guillot A.; Jonquet J.; Bertin E.; Guéant J.-L.; Alberto J.-M.; Bronowicki J.-P.; Amoyel J.; Hocine T.; Duca L.; Maurice P.; Bennasroune A.; Martiny L.; Debelle L.; Durlach V.; Blaise S. Production of Elastin-Derived Peptides Contributes to the Development of Nonalcoholic Steatohepatitis. Diabetes. 2018, 67 (8), 1604–1615. 10.2337/db17-0490. [DOI] [PubMed] [Google Scholar]
- Abdulkhalek S.; Szewczuk M. R. Neu1 Sialidase and Matrix Metalloproteinase-9 Cross-Talk Regulates Nucleic Acid-Induced Endosomal TOLL-like Receptor-7 and −9 Activation, Cellular Signaling and pro-Inflammatory Responses. Cell Signal. 2013, 25 (11), 2093–2105. 10.1016/j.cellsig.2013.06.010. [DOI] [PubMed] [Google Scholar]
- Luzina I. G.; Lillehoj E. P.; Lockatell V.; Hyun S. W.; Lugkey K. N.; Imamura A.; Ishida H.; Cairo C. W.; Atamas S. P.; Goldblum S. E. Therapeutic Effect of Neuraminidase-1-Selective Inhibition in Mouse Models of Bleomycin-Induced Pulmonary Inflammation and Fibrosis. J. Pharmacol. Exp. Ther. 2021, 376 (1), 136–146. 10.1124/jpet.120.000223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lillehoj E. P.; Guang W.; Hyun S. W.; Liu A.; Hegerle N.; Simon R.; Cross A. S.; Ishida H.; Luzina I. G.; Atamas S. P.; Goldblum S. E. Neuraminidase 1-Mediated Desialylation of the Mucin 1 Ectodomain Releases a Decoy Receptor That Protects against Pseudomonas Aeruginosa Lung Infection. J. Biol. Chem. 2019, 294 (2), 662–678. 10.1074/jbc.RA118.006022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hyun S. W.; Liu A.; Liu Z.; Cross A. S.; Verceles A. C.; Magesh S.; Kommagalla Y.; Kona C.; Ando H.; Luzina I. G.; Atamas S. P.; Piepenbrink K. H.; Sundberg E. J.; Guang W.; Ishida H.; Lillehoj E. P.; Goldblum S. E. The NEU1-Selective Sialidase Inhibitor, C9-Butyl-Amide-DANA, Blocks Sialidase Activity and NEU1-Mediated Bioactivities in Human Lung in Vitro and Murine Lung in Vivo. Glycobiology. 2016, 26 (8), 834–849. 10.1093/glycob/cww060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lillehoj E. P.; Hyun S. W.; Liu A.; Guang W.; Verceles A. C.; Luzina I. G.; Atamas S. P.; Kim K. C.; Goldblum S. E. NEU1 Sialidase Regulates Membrane-Tethered Mucin (MUC1) Ectodomain Adhesiveness for Pseudomonas Aeruginosa and Decoy Receptor Release. J. Biol. Chem. 2015, 290 (30), 18316–18331. 10.1074/jbc.M115.657114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Natori Y.; Nasui M.; Edo K.; Sato S.; Sakurai T.; Kizaki T.; Kihara-Negishi F. NEU1 Sialidase Controls Gene Expression and Secretion of IL-6 and MCP-1through NF- κ B Pathway in 3T3-L1 Adipocytes. J. Biochem. 2017, 162 (2), 137–143. 10.1093/jb/mvx006. [DOI] [PubMed] [Google Scholar]
- Amith S. R.; Jayanth P.; Franchuk S.; Finlay T.; Seyrantepe V.; Beyaert R.; Pshezhetsky A. v.; Szewczuk M. R. Neu1 Desialylation of Sialyl α-2,3-Linked β-Galactosyl Residues of TOLL-like Receptor 4 Is Essential for Receptor Activation and Cellular Signaling. Cell Signal. 2010, 22 (2), 314–324. 10.1016/j.cellsig.2009.09.038. [DOI] [PubMed] [Google Scholar]
- Feng C.; Stamatos N. M.; Dragan A. I.; Medvedev A.; Whitford M.; Zhang L.; Song C.; Rallabhandi P.; Cole L.; Nhu Q. M.; Vogel S. N.; Geddes C. D.; Cross A. S. Sialyl Residues Modulate LPS-Mediated Signaling through the Toll-Like Receptor 4 Complex. PLoS. One. 2012, 7 (4), e32359 10.1371/journal.pone.0032359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woronowicz A.; Amith S. R.; de Vusser K.; Laroy W.; Contreras R.; Basta S.; Szewczuk M. R. Dependence of Neurotrophic Factor Activation of Trk Tyrosine Kinase Receptors on Cellular Sialidase. Glycobiology. 2007, 17 (1), 10–24. 10.1093/glycob/cwl049. [DOI] [PubMed] [Google Scholar]
- Satyavarapu E. M.; Nath S.; Mandal C. Desialylation of Atg5 by Sialidase (Neu2) Enhances Autophagosome Formation to Induce Anchorage-Dependent Cell Death in Ovarian Cancer Cells. Cell Death Discovery 2021, 7 (1), 26. 10.1038/s41420-020-00391-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karhadkar T. R.; Meek T. D.; Gomer R. H. Inhibiting Sialidase-Induced TGF- β 1 Activation Attenuates Pulmonary Fibrosis in Mice. J. Pharmacol. Exp. Ther. 2021, 376 (1), 106–117. 10.1124/jpet.120.000258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lillehoj E. P.; Luzina I. G.; Atamas S. P. Mammalian Neuraminidases in Immune-Mediated Diseases: Mucins and Beyond. Front. Immunol. 2022, 13, 883079. 10.3389/fimmu.2022.883079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Achyuthan K. E.; Achyuthan A. M. Comparative Enzymology, Biochemistry and Pathophysiology of Human Exo-α-Sialidases (Neuraminidases). Comp. Biochem. Physiol. B. Biochem. Mol. Biol. 2001, 129 (1), 29–64. 10.1016/S1096-4959(01)00372-4. [DOI] [PubMed] [Google Scholar]
- Monti E.; Preti A.; Venerando B.; Borsani G. Recent Development in Mammalian Sialidase Molecular Biology. Neurochem. Res. 2002, 27, 649–663. 10.1023/A:1020276000901. [DOI] [PubMed] [Google Scholar]
- Chavas L. M. G.; Tringali C.; Fusi P.; Venerando B.; Tettamanti G.; Kato R.; Monti E.; Wakatsuki S. Crystal Structure of the Human Cytosolic Sialidase Neu2. J. Biol. Chem. 2005, 280 (1), 469–475. 10.1074/jbc.M411506200. [DOI] [PubMed] [Google Scholar]
- Magesh S.; Suzuki T.; Miyagi T.; Ishida H.; Kiso M. Homology Modeling of Human Sialidase Enzymes NEU1, NEU3 and NEU4 Based on the Crystal Structure of NEU2: Hints for the Design of Selective NEU3 Inhibitors. J. Mol. Graph Model. 2006, 25 (2), 196–207. 10.1016/j.jmgm.2005.12.006. [DOI] [PubMed] [Google Scholar]
- Bourguet E.; Figurska S.; Fra̧czek M. M. Human Neuraminidases: Structures and Stereoselective Inhibitors. J. Med. Chem. 2022, 65 (4), 3002–3025. 10.1021/acs.jmedchem.1c01612. [DOI] [PubMed] [Google Scholar]
- Monti E.; Miyagi T. Structure and Function of Mammalian Sialidases. Top. Curr. Chem. 2012, 366, 183–208. 10.1007/128_2012_328. [DOI] [PubMed] [Google Scholar]
- Miyagi T.; Takahashi K.; Yamamoto K.; Shiozaki K.; Yamaguchi K. Biological and Pathological Roles of Ganglioside Sialidases. Prog. Mol. Biol. Transl. Sci. 2018, 156, 121–150. 10.1016/bs.pmbts.2017.12.005. [DOI] [PubMed] [Google Scholar]
- Lewis A. L.; Lewis W. G. Host Sialoglycans and Bacterial Sialidases: A Mucosal Perspective. Cell Microbiol. 2012, 14 (8), 1174–1182. 10.1111/j.1462-5822.2012.01807.x. [DOI] [PubMed] [Google Scholar]
- Parker R. B.; Kohler J. J. Regulation of Intracellular Signaling by Extracellular Glycan Remodeling. ACS. Chem. Biol. 2010, 5 (1), 35–46. 10.1021/cb9002514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roggentin P.; Schauer R.; Hoyer L. L.; Vimr E. R. The Sialidase Superfamily and Its Spread by Horizontal Gene Transfer. Mol. Microbiol. 1993, 9 (5), 915–921. 10.1111/j.1365-2958.1993.tb01221.x. [DOI] [PubMed] [Google Scholar]
- Vimr E. R.; Lawrisuk L.; Galen J.; Kaper J. B. Cloning and Expression of the Vibrio Cholerae Neuraminidase Gene NanH in Escherichia Coli. J. Bacteriol. 1988, 170 (4), 1495–1504. 10.1128/jb.170.4.1495-1504.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park K.-H.; Kim M.-G.; Ahn H.-J.; Lee D.-H.; Kim J.-H.; Kim Y.-W.; Woo E.-J. Structural and Biochemical Characterization of the Broad Substrate Specificity of Bacteroides Thetaiotaomicron Commensal Sialidase. Biochim. Biophys. Acta. Proteins Proteom. 2013, 1834 (8), 1510–1519. 10.1016/j.bbapap.2013.04.028. [DOI] [PubMed] [Google Scholar]
- Huang Y.-L.; Chassard C.; Hausmann M.; von Itzstein M.; Hennet T. Sialic Acid Catabolism Drives Intestinal Inflammation and Microbial Dysbiosis in Mice. Nat. Commun. 2015, 6 (1), 8141. 10.1038/ncomms9141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamamoto T.; Ugai H.; Nakayama-Imaohji H.; Tada A.; Elahi M.; Houchi H.; Kuwahara T. Characterization of a Recombinant Bacteroides Fragilis Sialidase Expressed in Escherichia Coli. Anaerobe. 2018, 50, 69–75. 10.1016/j.anaerobe.2018.02.003. [DOI] [PubMed] [Google Scholar]
- Tanaka H.; Ito F.; Iwasaki T. Purification and Characterization of a Sialidase from Bacteroides Fragilis SBT3182. Biochem. Biophys. Res. Commun. 1992, 189 (1), 524–529. 10.1016/0006-291X(92)91589-I. [DOI] [PubMed] [Google Scholar]
- Derrien M.; Vaughan E. E.; Plugge C. M.; de Vos W. M. Akkermansia Muciniphila Gen. Nov., Sp. Nov., a Human Intestinal Mucin-Degrading Bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54 (5), 1469–1476. 10.1099/ijs.0.02873-0. [DOI] [PubMed] [Google Scholar]
- van Passel M. W. J.; Kant R.; Zoetendal E. G.; Plugge C. M.; Derrien M.; Malfatti S. A.; Chain P. S. G.; Woyke T.; Palva A.; de Vos W. M.; Smidt H. The Genome of Akkermansia Muciniphila, a Dedicated Intestinal Mucin Degrader, and Its Use in Exploring Intestinal Metagenomes. PLoS. One. 2011, 6 (3), e16876 10.1371/journal.pone.0016876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu G.; Kiefel M. J.; Wilson J. C.; Andrew P. W.; Oggioni M. R.; Taylor G. L. Three Streptococcus Pneumoniae Sialidases: Three Different Products. J. Am. Chem. Soc. 2011, 133 (6), 1718–1721. 10.1021/ja110733q. [DOI] [PubMed] [Google Scholar]
- Yamaguchi M.; Hirose Y.; Nakata M.; Uchiyama S.; Yamaguchi Y.; Goto K.; Sumitomo T.; Lewis A. L.; Kawabata S.; Nizet V. Evolutionary Inactivation of a Sialidase in Group B Streptococcus. Sci. Rep. 2016, 6 (1), 28852. 10.1038/srep28852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tuyau J. E.; Sims W. Neuraminidase Activity in Human Oral Strains of Haemophili. Arch. Oral Biol. 1974, 19 (9), 817–819. 10.1016/0003-9969(74)90171-X. [DOI] [PubMed] [Google Scholar]
- Singh A. K.; Woodiga S. A.; Grau M. A.; King S. J.. Streptococcus Oralis Neuraminidase Modulates Adherence to Multiple Carbohydrates on Platelets. Infect. Immun. 2017, 85 ( (3), ), 10.1128/IAI.00774-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beighton D.; Whiley R. A. Sialidase Activity of the “Streptococcus Milleri Group” and Other Viridans Group Streptococci. J. Clin. Microbiol. 1990, 28 (6), 1431–1433. 10.1128/jcm.28.6.1431-1433.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li C.; Kurniyati; Hu B.; Bian J.; Sun J.; Zhang W.; Liu J.; Pan Y.; Li C. Abrogation of Neuraminidase Reduces Biofilm Formation, Capsule Biosynthesis, and Virulence of Porphyromonas Gingivalis. Infect. Immun. 2012, 80 (1), 3–13. 10.1128/IAI.05773-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Do T.; Henssge U.; Gilbert S. C.; Clark D.; Beighton D. Evidence for Recombination between a Sialidase (NanH) of Actinomyces Naeslundii and Actinomyces Oris, Previously Named Actinomyces Naeslundii Genospecies 1 and 2. FEMS. Microbiol. Lett. 2008, 288 (2), 156–162. 10.1111/j.1574-6968.2008.01336.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roy S.; Honma K.; Douglas C. W. I.; Sharma A.; Stafford G. P. Role of Sialidase in Glycoprotein Utilization by Tannerella Forsythia. Microbiology (N.Y.) 2011, 157 (11), 3195–3202. 10.1099/mic.0.052498-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurniyati K.; Zhang W.; Zhang K.; Li C. A Surface-Exposed Neuraminidase Affects Complement Resistance and Virulence of the Oral Spirochaete Treponema Denticola. Mol. Microbiol. 2013, 89 (5), 842–856. 10.1111/mmi.12311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wohlbold T.; Krammer F. In the Shadow of Hemagglutinin: A Growing Interest in Influenza Viral Neuraminidase and Its Role as a Vaccine Antigen. Viruses. 2014, 6 (6), 2465–2494. 10.3390/v6062465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buschiazzo A.; Amaya M. F.; Cremona M. L.; Frasch A. C.; Alzari P. M. The Crystal Structure and Mode of Action of Trans-Sialidase, a Key Enzyme in Trypanosoma Cruzi Pathogenesis. Mol. Cell 2002, 10 (4), 757–768. 10.1016/S1097-2765(02)00680-9. [DOI] [PubMed] [Google Scholar]
- Amaya M. F.; Watts A. G.; Damager I.; Wehenkel A.; Nguyen T.; Buschiazzo A.; Paris G.; Frasch A. C.; Withers S. G.; Alzari P. M. Structural Insights into the Catalytic Mechanism of Trypanosoma Cruzi Trans-Sialidase. Structure. 2004, 12 (5), 775–784. 10.1016/j.str.2004.02.036. [DOI] [PubMed] [Google Scholar]
- Taylor G.; Crennell S.; Takimoto T.; Portner A. Crystal Structure of the Multifunctional Paramyxovirus Hemagglutinin-Neuraminidase. Nat. Struct. Biol. 2000, 7 (11), 1068–1074. 10.1038/81002. [DOI] [PubMed] [Google Scholar]
- Schenkman S.; Jiang M.-S.; Hart G. W.; Nussenzweig V. A Novel Cell Surface Trans-Sialidase of Trypanosoma Cruzi Generates a Stage-Specific Epitope Required for Invasion of Mammalian Cells. Cell. 1991, 65 (7), 1117–1125. 10.1016/0092-8674(91)90008-M. [DOI] [PubMed] [Google Scholar]
- Todeschini A. R.; Mendonça-Previato L.; Previato J. O.; Varki A.; van Halbeek H. Trans-Sialidase from Trypanosoma Cruzi Catalyzes Sialoside Hydrolysis With Retention of Configuration. Glycobiology. 2000, 10 (2), 213–221. 10.1093/glycob/10.2.213. [DOI] [PubMed] [Google Scholar]
- Lipničanová S.; Chmelová D.; Ondrejovič M.; Frecer V.; Miertuš S. Diversity of Sialidases Found in the Human Body - A Review. Int. J. Biol. Macromol. 2020, 148, 857–868. 10.1016/j.ijbiomac.2020.01.123. [DOI] [PubMed] [Google Scholar]
- Watts A. G.; Damager I.; Amaya M. L.; Buschiazzo A.; Alzari P.; Frasch A. C.; Withers S. G. Trypanosoma Cruzi Trans-Sialidase Operates through a Covalent Sialyl-Enzyme Intermediate: Tyrosine Is the Catalytic Nucleophile. J. Am. Chem. Soc. 2003, 125 (25), 7532–7533. 10.1021/ja0344967. [DOI] [PubMed] [Google Scholar]
- Damager I.; Buchini S.; Amaya M. F.; Buschiazzo A.; Alzari P.; Frasch A. C.; Watts A.; Withers S. G. Kinetic and Mechanistic Analysis of Trypanosoma Cruzi Trans-Sialidase Reveals a Classical Ping-Pong Mechanism with Acid/Base Catalysis. Biochemistry. 2008, 47 (11), 3507–3512. 10.1021/bi7024832. [DOI] [PubMed] [Google Scholar]
- Morley T. J.; Willis L. M.; Whitfield C.; Wakarchuk W. W.; Withers S. G. A New Sialidase Mechanism. J. Biol. Chem. 2009, 284 (26), 17404–17410. 10.1074/jbc.M109.003970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bule P.; Chuzel L.; Blagova E.; Wu L.; Gray M. A.; Henrissat B.; Rapp E.; Bertozzi C. R.; Taron C. H.; Davies G. J. Inverting Family GH156 Sialidases Define an Unusual Catalytic Motif for Glycosidase Action. Nat. Commun. 2019, 10 (1), 4816. 10.1038/s41467-019-12684-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zechel D. L.; Withers S. G. Glycosidase Mechanisms: Anatomy of a Finely Tuned Catalyst. Acc. Chem. Res. 2000, 33 (1), 11–18. 10.1021/ar970172+. [DOI] [PubMed] [Google Scholar]
- Watts A. G.; Withers S. G. The Synthesis of Some Mechanistic Probes for Sialic Acid Processing Enzymes and the Labeling of a Sialidase from Trypanosoma Rangeli. Can. J. Chem. 2004, 82 (11), 1581–1588. 10.1139/v04-125. [DOI] [Google Scholar]
- Watts A. G.; Oppezzo P.; Withers S. G.; Alzari P. M.; Buschiazzo A. Structural and Kinetic Analysis of Two Covalent Sialosyl-Enzyme Intermediates on Trypanosoma Rangeli Sialidase. J. Biol. Chem. 2006, 281 (7), 4149–4155. 10.1074/jbc.M510677200. [DOI] [PubMed] [Google Scholar]
- Chan J.; Lewis A. R.; Indurugalla D.; Schur M.; Wakarchuk W.; Bennet A. J. Transition State Analysis of Vibrio Cholerae Sialidase-Catalyzed Hydrolyses of Natural Substrate Analogues. J. Am. Chem. Soc. 2012, 134 (8), 3748–3757. 10.1021/ja208564y. [DOI] [PubMed] [Google Scholar]
- Vocadlo D. J.; Davies G. J. Mechanistic Insights into Glycosidase Chemistry. Curr. Opin. Chem. Biol. 2008, 12 (5), 539–555. 10.1016/j.cbpa.2008.05.010. [DOI] [PubMed] [Google Scholar]
- Taylor G. Sialidases: Structures, Biological Significance and Therapeutic Potential. Curr. Opin. Struct. Biol. 1996, 6 (6), 830–837. 10.1016/S0959-440X(96)80014-5. [DOI] [PubMed] [Google Scholar]
- Corfield T. Bacterial Sialidases-Roles in Pathogenicity and Nutrition. Glycobiology. 1992, 2 (6), 509–521. 10.1093/glycob/2.6.509. [DOI] [PubMed] [Google Scholar]
- Achyuthan K. E.; Achyuthan A. M. Comparative Enzymology, Biochemistry and Pathophysiology of Human Exo-α-Sialidases (Neuraminidases). Comp. Biochem. Physiol. B. Biochem. Mol. Biol. 2001, 129 (1), 29–64. 10.1016/S1096-4959(01)00372-4. [DOI] [PubMed] [Google Scholar]
- Juge N.; Tailford L.; Owen C. D. Sialidases from Gut Bacteria: A Mini-Review. Biochem. Soc. Trans. 2016, 44, 166–175. 10.1042/BST20150226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peltota V. T.; Mccullers J. A. Respiratory Viruses Predisposing to Bacterial Infections: Role of Neuraminidase. Pediatr. Infect. Dis. J. 2004, 23 (1), S87–S97. 10.1097/01.inf.0000108197.81270.35. [DOI] [PubMed] [Google Scholar]
- Giacopuzzi E.; Bresciani R.; Schauer R.; Monti E.; Borsani G. New Insights on the Sialidase Protein Family Revealed by a Phylogenetic Analysis in Metazoa. PLoS. One. 2012, 7 (8), e44193 10.1371/journal.pone.0044193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Monti E.; Bonten E.; D’Azzo A.; Bresciani R.; Venerando B.; Borsani G.; Schauer R.; Tettamanti G. Sialidases in Vertebrates. Advances in Carbohydrate Chemistry and Biochemistry 2010, 64, 403–479. 10.1016/S0065-2318(10)64007-3. [DOI] [PubMed] [Google Scholar]
- Lipničanová S.; Chmelová D.; Ondrejovič M.; Frecer V.; Miertuš S. Diversity of Sialidases Found in the Human Body - A Review. Int. J. Biol. Macromol. 2020, 148, 857–868. 10.1016/j.ijbiomac.2020.01.123. [DOI] [PubMed] [Google Scholar]
- Gubareva L.; Mohan T. Antivirals Targeting the Neuraminidase. Cold Spring Harb. Perspect. Med. 2022, 12 (1), a038455. 10.1101/cshperspect.a038455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Terrier O.; Slama-Schwok A. Anti-Influenza Drug Discovery and Development: Targeting the Virus and Its Host by All Possible Means. Adv. Exp. Med. Biol. 2021, 1322, 195–218. 10.1007/978-981-16-0267-2_8. [DOI] [PubMed] [Google Scholar]
- Wang Y. H. Sialidases From Clostridium Perfringens and Their Inhibitors. Front Cell Infect. Microbiol.. 2020, 9, 462. 10.3389/fcimb.2019.00462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cairo C. W. Inhibitors of the Human Neuraminidase Enzymes. MedChemComm. 2014, 5, 1067–1074. 10.1039/C4MD00089G. [DOI] [Google Scholar]
- Bowles W. H. D.; Gloster T. M. Sialidase and Sialyltransferase Inhibitors: Targeting Pathogenicity and Disease. Front. Mol. Biosci. 2021, 8, 705133. 10.3389/fmolb.2021.705133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grienke U.; Schmidtke M.; von Grafenstein S.; Kirchmair J.; Liedl K. R.; Rollinger J. M. Influenza Neuraminidase: A Druggable Target for Natural Products. Nat. Prod. Rep. 2012, 29, 11–36. 10.1039/C1NP00053E. [DOI] [PubMed] [Google Scholar]
- Singh N.; Anjum N.; Chandra R. Combating Influenza: Natural Products as Neuraminidase Inhibitors. Phytochem. Rev. 2019, 18 (1), 69–107. 10.1007/s11101-018-9581-1. [DOI] [Google Scholar]
- Silverman R. B.The Organic Chemistry of Drug Design and Drug Action, 2nd ed.; Academic Press: San Diego, CA, 2004. [Google Scholar]
- Meindl P.; Bodo G.; Palese P.; Schulman J.; Tuppy H. Inhibition of Neuraminidase Activity by Derivatives of 2-Deoxy-2,3-Dehydro-N-Acetylneuraminic Acid. Virology. 1974, 58 (2), 457–463. 10.1016/0042-6822(74)90080-4. [DOI] [PubMed] [Google Scholar]
- Shukla A. K.; Schröder C.; Nöhle U.; Schauer R. Natural Occurrence and Preparation of O-Acylated 2,3-Unsaturated Sialic Acids. Carbohydr. Res. 1987, 168 (2), 199–209. 10.1016/0008-6215(87)80026-5. [DOI] [PubMed] [Google Scholar]
- Burmeister W. P.; Henrissat B.; Bosso C.; Cusack S.; Ruigrok R. W. H. Influenza B Virus Neuraminidase Can Synthesize Its Own Inhibitor. Structure. 1993, 1 (1), 19–26. 10.1016/0969-2126(93)90005-2. [DOI] [PubMed] [Google Scholar]
- von Itzstein M.; Wu W.-Y.; Kok G. B.; Pegg M. S.; Dyason J. C.; Jin B.; van Phan T.; Smythe M. L.; White H. F.; Oliver S. W.; Colman P. M.; Varghese J. N.; Ryan D. M.; Woods J. M.; Bethell R. C.; Hotham V. J.; Cameron J. M.; Penn C. R. Rational Design of Potent Sialidase-Based Inhibitors of Influenza Virus Replication. Nature. 1993, 363 (6428), 418–423. 10.1038/363418a0. [DOI] [PubMed] [Google Scholar]
- Kim C. U.; Lew W.; Williams M. A.; Liu H.; Zhang L.; Swaminathan S.; Bischofberger N.; Chen M. S.; Mendel D. B.; Tai C. Y.; Laver W. G.; Stevens R. C. Influenza Neuraminidase Inhibitors Possessing a Novel Hydrophobic Interaction in the Enzyme Active Site: Design, Synthesis, and Structural Analysis of Carbocyclic Sialic Acid Analogues with Potent Anti-Influenza Activity. J. Am. Chem. Soc. 1997, 119 (4), 681–690. 10.1021/ja963036t. [DOI] [PubMed] [Google Scholar]
- Collins P. J.; Haire L. F.; Lin Y. P.; Liu J.; Russell R. J.; Walker P. A.; Skehel J. J.; Martin S. R.; Hay A. J.; Gamblin S. J. Crystal Structures of Oseltamivir-Resistant Influenza Virus Neuraminidase Mutants. Nature. 2008, 453 (7199), 1258–1261. 10.1038/nature06956. [DOI] [PubMed] [Google Scholar]
- Umezawa H.; Aoyagi T.; Komiyama T.; Morishima H.; Hamada M.; Takeuchi T. Purification and Characterization of a Sialidase Inhibitor, Siastatin, Produced by Streptomyces. J. Antibiot. (Tokyo). 1974, 27 (12), 963–969. 10.7164/antibiotics.27.963. [DOI] [PubMed] [Google Scholar]
- Eyer L.; Hruska K. Antiviral Agents Targeting the Influenza Virus: A Review and Publication Analysis. Vet. Med. (Praha). 2013, 58 (3), 113–185. 10.17221/6746-VETMED. [DOI] [Google Scholar]
- Burnham A. J.; Baranovich T.; Govorkova E. A. Neuraminidase Inhibitors for Influenza B Virus Infection: Efficacy and Resistance. Antiviral Res. 2013, 100 (2), 520–534. 10.1016/j.antiviral.2013.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chairat K.; Tarning J.; White N. J.; Lindegardh N. Pharmacokinetic Properties OfAnti-Influenza Neuraminidase Inhibitors. J. Clin. Pharmacol. 2013, 53 (2), 119–139. 10.1177/0091270012440280. [DOI] [PubMed] [Google Scholar]
- Cheng C.-K.; Tsai C.-H.; Shie J.-J.; Fang J.-M. From Neuraminidase Inhibitors to Conjugates: A Step towards Better Anti-Influenza Drugs?. Future Med. Chem. 2014, 6 (7), 757–774. 10.4155/fmc.14.30. [DOI] [PubMed] [Google Scholar]
- Feng E.; Ye D.; Li J.; Zhang D.; Wang J.; Zhao F.; Hilgenfeld R.; Zheng M.; Jiang H.; Liu H. Recent Advances in Neuraminidase Inhibitor Development as Anti-Influenza Drugs. ChemMedChem. 2012, 7 (9), 1527–1536. 10.1002/cmdc.201200155. [DOI] [PubMed] [Google Scholar]
- Holodniy M.; Kamali A. Influenza Treatment and Prophylaxis with Neuraminidase Inhibitors: A Review. Infect. Drug. Resist. 2013, 6, 187–193. 10.2147/IDR.S36601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muthuri S. G.; Venkatesan S.; Myles P. R.; Leonardi-Bee J.; al Khuwaitir T. S. A.; al Mamun A.; Anovadiya A. P.; Azziz-Baumgartner E.; Báez C.; Bassetti M.; Beovic B.; Bertisch B.; Bonmarin I.; Booy R.; Borja-Aburto V. H.; Burgmann H.; Cao B.; Carratala J.; Denholm J. T.; Dominguez S. R.; Duarte P. A. D.; Dubnov-Raz G.; Echavarria M.; Fanella S.; Gao Z.; Gérardin P.; Giannella M.; Gubbels S.; Herberg J.; Iglesias A. L. H.; Hoger P. H.; Hu X.; Islam Q. T.; Jiménez M. F.; Kandeel A.; Keijzers G.; Khalili H.; Knight M.; Kudo K.; Kusznierz G.; Kuzman I.; Kwan A. M. C.; Amine I. L.; Langenegger E.; Lankarani K. B.; Leo Y.-S.; Linko R.; Liu P.; Madanat F.; Mayo-Montero E.; McGeer A.; Memish Z.; Metan G.; Mickiene A.; Mikić D.; Mohn K. G. I.; Moradi A.; Nymadawa P.; Oliva M. E.; Ozkan M.; Parekh D.; Paul M.; Polack F. P.; Rath B. A.; Rodríguez A. H.; Sarrouf E. B.; Seale A. C.; Sertogullarindan B.; Siqueira M. M.; Skrȩt-Magierło J.; Stephan F.; Talarek E.; Tang J. W.; To K. K. W.; Torres A.; Törün S. H.; Tran D.; Uyeki T. M.; van Zwol A.; Vaudry W.; Vidmar T.; Yokota R. T. C.; Zarogoulidis P.; Nguyen-Van-Tam J. S. Effectiveness of Neuraminidase Inhibitors in Reducing Mortality in Patients Admitted to Hospital with Influenza A H1N1pdm09 Virus Infection: A Meta-Analysis of Individual Participant Data. Lancet. Respir. Med. 2014, 2 (5), 395–404. 10.1016/S2213-2600(14)70041-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moscona A. Neuraminidase Inhibitors for Influenza. N. Engl. J. Med. 2005, 353 (13), 1363–1373. 10.1056/NEJMra050740. [DOI] [PubMed] [Google Scholar]
- Smith B. J. A Conformational Study of 2-Oxanol: Insight into the Role of Ring Distortion on Enzyme-Catalyzed Glycosidic Bond Cleavage. J. Am. Chem. Soc. 1997, 119 (11), 2699–2706. 10.1021/ja9623020. [DOI] [Google Scholar]
- Kim C. U.; Lew W.; Williams M. A.; Wu H.; Zhang L.; Chen X.; Escarpe P. A.; Mendel D. B.; Laver W. G.; Stevens R. C. Structure-Activity Relationship Studies of Novel Carbocyclic Influenza Neuraminidase Inhibitors. J. Med. Chem. 1998, 41 (14), 2451–2460. 10.1021/jm980162u. [DOI] [PubMed] [Google Scholar]
- Vorwerk S.; Vasella A. Carbocyclic Analogues OfN-Acetyl-2,3-Didehydro-2-Deoxy-D-Neuraminic Acid (Neu5Ac2en, DANA): Synthesis and Inhibition of Viral and Bacterial Neuraminidases. Angew. Chem., Int. Ed. 1998, 37 (12), 1732–1734. . [DOI] [PubMed] [Google Scholar]
- Sudbeck E. A.; Jedrzejas M. J.; Singh S.; Brouillette W. J.; Air G. M.; Laver W. G.; Babu Y. S.; Bantia S.; Chand P.; Chu N.; Montgomery J. A.; Walsh D. A.; Luo M. Guanidinobenzoic Acid Inhibitors of Influenza Virus Neuraminidase 1 1Edited by R. Huber. J. Mol. Biol. 1997, 267 (3), 584–594. 10.1006/jmbi.1996.0885. [DOI] [PubMed] [Google Scholar]
- Laborda P.; Wang S.-Y.; Voglmeir J. Influenza Neuraminidase Inhibitors: Synthetic Approaches, Derivatives and Biological Activity. Molecules. 2016, 21 (11), 1513. 10.3390/molecules21111513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Świerczyńska M.; Mirowska-Guzel D. M.; Pindelska E. Antiviral Drugs in Influenza. Int. J. Environ. Res. Public Health. 2022, 19 (5), 3018. 10.3390/ijerph19053018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Severi E.; Hood D. W.; Thomas G. H. Sialic Acid Utilization by Bacterial Pathogens. Microbiology (N.Y.) 2007, 153 (9), 2817–2822. 10.1099/mic.0.2007/009480-0. [DOI] [PubMed] [Google Scholar]
- Freedman J.; Shrestha A.; McClane B. Clostridium Perfringens Enterotoxin: Action, Genetics, and Translational Applications. Toxins. (Basel). 2016, 8 (3), 73. 10.3390/toxins8030073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khedri Z.; Li Y.; Cao H.; Qu J.; Yu H.; Muthana M. M.; Chen X. Synthesis of Selective Inhibitors against V. Cholerae Sialidase and Human Cytosolic Sialidase NEU2. Org. Biomol. Chem. 2012, 10 (30), 6112–6120. 10.1039/c2ob25335f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hinou H.; Miyoshi R.; Takasu Y.; Kai H.; Kurogochi M.; Arioka S.; Gao X. D.; Miura N.; Fujitani N.; Omoto S.; Yoshinaga T.; Fujiwara T.; Noshi T.; Togame H.; Takemoto H.; Nishimura S. I. A Strategy for Neuraminidase Inhibitors Using Mechanism-Based Labeling Information. Chem. Asian. J. 2011, 6 (4), 1048–1056. 10.1002/asia.201000594. [DOI] [PubMed] [Google Scholar]
- Slack T. J.; Li W.; Shi D.; McArthur J. B.; Zhao G.; Li Y.; Xiao A.; Khedri Z.; Yu H.; Liu Y.; Chen X. Triazole-Linked Transition State Analogs as Selective Inhibitors against V. Cholerae Sialidase. Bioorg. Med. Chem. 2018, 26 (21), 5751–5757. 10.1016/j.bmc.2018.10.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shiga K.; Takahashi K.; Sato I.; Kato K.; Saijo S.; Moriya S.; Hosono M.; Miyagi T. Upregulation of Sialidase NEU3 in Head and Neck Squamous Cell Carcinoma Associated with Lymph Node Metastasis. Cancer. Sci. 2015, 106 (11), 1544–1553. 10.1111/cas.12810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C.; Chen J.; Liu Y.; Xu D. Sialic Acid Metabolism as a Potential Therapeutic Target of Atherosclerosis. Lipids Health Dis. 2019, 18 (1), 173. 10.1186/s12944-019-1113-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magesh S.; Moriya S.; Suzuki T.; Miyagi T.; Ishida H.; Kiso M. Design, Synthesis, and Biological Evaluation of Human Sialidase Inhibitors. Part 1: Selective Inhibitors of Lysosomal Sialidase (NEU1). Bioorg. Med. Chem. Lett. 2008, 18 (2), 532–537. 10.1016/j.bmcl.2007.11.084. [DOI] [PubMed] [Google Scholar]
- Zhang Y.; Albohy A.; Zou Y.; Smutova V.; Pshezhetsky A. v.; Cairo C. W. Identification of Selective Inhibitors for Human Neuraminidase Isoenzymes Using C4,C7-Modified 2-Deoxy-2,3-Didehydro-N-Acetylneuraminic Acid (DANA) Analogues. J. Med. Chem. 2013, 56 (7), 2948–2958. 10.1021/jm301892f. [DOI] [PubMed] [Google Scholar]
- Guo T.; Dätwyler P.; Demina E.; Richards M. R.; Ge P.; Zou C.; Zheng R.; Fougerat A.; Pshezhetsky A. v.; Ernst B.; Cairo C. W. Selective Inhibitors of Human Neuraminidase 3. J. Med. Chem. 2018, 61 (5), 1990–2008. 10.1021/acs.jmedchem.7b01574. [DOI] [PubMed] [Google Scholar]
- Guo T.; Héon-Roberts R.; Zou C.; Zheng R.; Pshezhetsky A. v.; Cairo C. W. Selective Inhibitors of Human Neuraminidase 1 (NEU1). J. Med. Chem. 2018, 61 (24), 11261–11279. 10.1021/acs.jmedchem.8b01411. [DOI] [PubMed] [Google Scholar]
- Zou Y.; Albohy A.; Sandbhor M.; Cairo C. W. Inhibition of Human Neuraminidase 3 (NEU3) by C9-Triazole Derivatives of 2,3-Didehydro-N-Acetyl-Neuraminic Acid. Bioorg. Med. Chem. Lett. 2010, 20 (24), 7529–7533. 10.1016/j.bmcl.2010.09.111. [DOI] [PubMed] [Google Scholar]
- Albohy A.; Zhang Y.; Smutova V.; Pshezhetsky A. v.; Cairo C. W. Identification of Selective Nanomolar Inhibitors of the Human Neuraminidase, NEU4. ACS. Med. Chem. Lett. 2013, 4 (6), 532–537. 10.1021/ml400080t. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howlader M. A.; Guo T.; Chakraberty R.; Cairo C. W. Isoenzyme-Selective Inhibitors of Human Neuraminidases Reveal Distinct Effects on Cell Migration. ACS. Chem. Biol. 2020, 15 (6), 1328–1339. 10.1021/acschembio.9b00975. [DOI] [PubMed] [Google Scholar]
- Linclau B.; Ardá A.; Reichardt N.-C.; Sollogoub M.; Unione L.; Vincent S. P.; Jiménez-Barbero J. Fluorinated Carbohydrates as Chemical Probes for Molecular Recognition Studies. Current Status and Perspectives. Chem. Soc. Rev. 2020, 49 (12), 3863–3888. 10.1039/C9CS00099B. [DOI] [PubMed] [Google Scholar]
- Delbrouck J. A.; Chêne L. P.; Vincent S. P.. Fluorosugars as Inhibitors of Bacterial Enzymes. In Fluorine in Life Sciences: Pharmaceuticals, Medicinal Diagnostics, and Agrochemicals; Elsevier, 2019; pp 241–279, 10.1016/B978-0-12-812733-9.00006-4. [DOI] [Google Scholar]
- Nakajima T.; Hori H.; Ohrui H.; Meguro H.; Ido T. Synthesis of N -Acetyl-3-Fluoro-Neuraminic Acids. Agric. Biol. Chem. 1988, 52 (5), 1209–1215. 10.1080/00021369.1988.10868800. [DOI] [Google Scholar]
- Sun X.-L.; Kanie Y.; Guo C.-T.; Kanie O.; Suzuki Y.; Wong C.-H. Syntheses of C-3-Modified Sialylglycosides as Selective Inhibitors of Influenza Hemagglutinin and Neuraminidase. Eur. J. Org. Chem. 2000, 2000 (14), 2643–2653. . [DOI] [Google Scholar]
- Guo C.-T.; Sun X.-L.; Kanie O.; Shortridge K. F.; Suzuki T.; Miyamoto D.; Hidari K. I.-P. J.; Wong C.-H.; Suzuki Y. An O-Glycoside of Sialic Acid Derivative That Inhibits Both Hemagglutinin and Sialidase Activities of Influenza Viruses. Glycobiology 2002, 12 (3), 183–190. 10.1093/glycob/12.3.183. [DOI] [PubMed] [Google Scholar]
- Zechel D. L.; Withers S. G. Glycosidase Mechanisms: Anatomy of a Finely Tuned Catalyst. Acc. Chem. Res. 2000, 33 (1), 11–18. 10.1021/ar970172+. [DOI] [PubMed] [Google Scholar]
- Watts A. G.; Oppezzo P.; Withers S. G.; Alzari P. M.; Buschiazzo A. Structural and Kinetic Analysis of Two Covalent Sialosyl-Enzyme Intermediates on Trypanosoma Rangeli Sialidase. J. Biol. Chem. 2006, 281 (7), 4149–4155. 10.1074/jbc.M510677200. [DOI] [PubMed] [Google Scholar]
- Watts A. G.; Damager I.; Amaya M. L.; Buschiazzo A.; Alzari P.; Frasch A. C.; Withers S. G. Trypanosoma Cruzi Trans-Sialidase Operates through a Covalent Sialyl-Enzyme Intermediate: Tyrosine Is the Catalytic Nucleophile. J. Am. Chem. Soc. 2003, 125 (25), 7532–7533. 10.1021/ja0344967. [DOI] [PubMed] [Google Scholar]
- Buchini S.; Buschiazzo A.; Withers S. G. A New Generation of SpecificTrypanosoma Cruzi Trans-Sialidase Inhibitors. Angew. Chem., Int. Ed. 2008, 47 (14), 2700–2703. 10.1002/anie.200705435. [DOI] [PubMed] [Google Scholar]
- Newstead S. L.; Potter J. A.; Wilson J. C.; Xu G.; Chien C.-H.; Watts A. G.; Withers S. G.; Taylor G. L. The Structure of Clostridium Perfringens NanI Sialidase and Its Catalytic Intermediates. J. Biol. Chem. 2008, 283 (14), 9080–9088. 10.1074/jbc.M710247200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vavricka C. J.; Liu Y.; Kiyota H.; Sriwilaijaroen N.; Qi J.; Tanaka K.; Wu Y.; Li Q.; Li Y.; Yan J.; Suzuki Y.; Gao G. F. Influenza Neuraminidase Operates via a Nucleophilic Mechanism and Can Be Targeted by Covalent Inhibitors. Nat. Commun. 2013, 4, 1491. 10.1038/ncomms2487. [DOI] [PubMed] [Google Scholar]
- Hader S.; Watts A. G. The Synthesis of a Series of Deoxygenated 2,3-Difluoro-N-Acteylneuraminic Acid Derivatives as Potential Sialidase Inhibitors. Carbohydr. Res. 2013, 374, 23–28. 10.1016/j.carres.2013.03.026. [DOI] [PubMed] [Google Scholar]
- Buchini S.; Gallat F. X.; Greig I. R.; Kim J. H.; Wakatsuki S.; Chavas L. M. G.; Withers S. G. Tuning Mechanism-Based Inactivators of Neuraminidases: Mechanistic and Structural Insights. Angew. Chem., Int. Ed. 2014, 53 (13), 3382–3386. 10.1002/anie.201309675. [DOI] [PubMed] [Google Scholar]
- Li W.; Santra A.; Yu H.; Slack T. J.; Muthana M. M.; Shi D.; Liu Y.; Chen X. 9-Azido-9-Deoxy-2,3-Difluorosialic Acid as a Subnanomolar Inhibitor against Bacterial Sialidases. J. Org. Chem. 2019, 84 (11), 6697–6708. 10.1021/acs.joc.9b00385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim J.-H.; Resende R.; Wennekes T.; Chen H.-M.; Bance N.; Buchini S.; Watts A. G.; Pilling P.; Streltsov V. A.; Petric M.; Liggins R.; Barrett S.; McKimm-Breschkin J. L.; Niikura M.; Withers S. G. Mechanism-Based Covalent Neuraminidase Inhibitors with Broad-Spectrum Influenza Antiviral Activity. Science. 2013, 340 (6128), 71–75. 10.1126/science.1232552. [DOI] [PubMed] [Google Scholar]
- Rillahan C. D.; Antonopoulos A.; Lefort C. T.; Sonon R.; Azadi P.; Ley K.; Dell A.; Haslam S. M.; Paulson J. C. Global Metabolic Inhibitors of Sialyl- and Fucosyltransferases Remodel the Glycome. Nat. Chem. Biol. 2012, 8 (7), 661–668. 10.1038/nchembio.999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Büll C.; Boltje T. J.; Balneger N.; Weischer S. M.; Wassink M.; van Gemst J. J.; Bloemendal V. R.; Boon L.; van der Vlag J.; Heise T.; den Brok M. H.; Adema G. J. Sialic Acid Blockade Suppresses Tumor Growth by Enhancing T-Cell-Mediated Tumor Immunity. Cancer. Res. 2018, 78 (13), 3574–3588. 10.1158/0008-5472.CAN-17-3376. [DOI] [PubMed] [Google Scholar]
- Halazy S.; Berges V.; Ehrhard A.; Danzin C. Ortho- and Para-(Difluoromethyl)Aryl-β-d-Glucosides: A New Class of Enzyme-Activated Irreversible Inhibitors of β-Glucosidases. Bioorg. Chem. 1990, 18 (3), 330–344. 10.1016/0045-2068(90)90007-R. [DOI] [Google Scholar]
- Janda K. D.; Lo L.-C.; Lo C.-H. L.; Sim M.-M.; Wang R.; Wong C.-H.; Lerner R. A. Chemical Selection for Catalysis in Combinatorial Antibody Libraries. Science. 1997, 275 (5302), 945–948. 10.1126/science.275.5302.945. [DOI] [PubMed] [Google Scholar]
- Kurogochi M.; Nishimura S.-I.; Lee Y. C. Mechanism-Based Fluorescent Labeling of β-Galactosidases. J. Biol. Chem. 2004, 279 (43), 44704–44712. 10.1074/jbc.M401718200. [DOI] [PubMed] [Google Scholar]
- Ichikawa M.; Ichikawa Y. A Mechanism-Based Affinity-Labeling Agent for Possible Use in Isolating N-Acetylglucosaminidase. Bioorg. Med. Chem. Lett. 2001, 11 (13), 1769–1773. 10.1016/S0960-894X(01)00300-6. [DOI] [PubMed] [Google Scholar]
- Carvalho S. T.; Sola-Penna M.; Oliveira I. A.; Pita S.; Gonçalves A. S.; Neves B. C.; Sousa F. R.; Freire-de-Lima L.; Kurogochi M.; Hinou H.; Nishimura S. I.; Mendonça-Previato L.; Previato J. O.; Todeschini A. R. A New Class of Mechanism-Based Inhibitors for Trypanosoma Cruzi Transsialidase and Their Influence on Parasite Virulence. Glycobiology. 2010, 20 (8), 1034–1045. 10.1093/glycob/cwq065. [DOI] [PubMed] [Google Scholar]
- Kai H.; Hinou H.; Nishimura S. I. Aglycone-Focused Randomization of 2-Difluoromethylphenyl-Type Sialoside Suicide Substrates for Neuraminidases. Bioorg. Med. Chem. 2012, 20 (8), 2739–2746. 10.1016/j.bmc.2012.02.001. [DOI] [PubMed] [Google Scholar]
- Rando R. R. Chemistry and Enzymology of kcat Inhibitors. Science. 1974, 185 (4148), 320–324. 10.1126/science.185.4148.320. [DOI] [PubMed] [Google Scholar]
- Ahmed V.; Liu Y.; Taylor S. D. Multiple Pathways for the Irreversible Inhibition of Steroid Sulfatase with Quinone Methide-Generating Suicide Inhibitors. ChemBioChem. 2009, 10 (9), 1457–1461. 10.1002/cbic.200900143. [DOI] [PubMed] [Google Scholar]
- Rempel B. P.; Withers S. G. Covalent Inhibitors of Glycosidases and Their Applications in Biochemistry and Biology. Glycobiology. 2008, 18 (8), 570–586. 10.1093/glycob/cwn041. [DOI] [PubMed] [Google Scholar]
- Kai H.; Hinou H.; Naruchi K.; Matsushita T.; Nishimura S. I. Macrocyclic Mechanism-Based Inhibitor for Neuraminidases. Chem.—Eur. J. 2013, 19 (4), 1364–1372. 10.1002/chem.201200859. [DOI] [PubMed] [Google Scholar]
- Schauer R.; Kamerling J. P.. Chemistry, Biochemistry and Biology of Sialic Acids. In New Comprehensive Biochemistry; Elsevier, 1997; Vol. 29b, pp 243–402, 10.1016/S0167-7306(08)60624-9. [DOI] [Google Scholar]
- von Itzstein M.Influenza Virus Sialidase - A Drug Discovery Target, 36th ed.; von Itzstein M., Ed.; Springer: Basel, 2012, 10.1007/978-3-7643-8927-7. [DOI] [Google Scholar]
- Kim C. U.; Lew W.; Williams M. A.; Liu H.; Zhang L.; Swaminathan S.; Bischofberger N.; Chen M. S.; Mendel D. B.; Tai C. Y.; Laver W. G.; Stevens R. C. Influenza Neuraminidase Inhibitors Possessing a Novel Hydrophobic Interaction in the Enzyme Active Site: Design, Synthesis, and Structural Analysis of Carbocyclic Sialic Acid Analogues with Potent Anti-Influenza Activity. J. Am. Chem. Soc. 1997, 119 (4), 681–690. 10.1021/ja963036t. [DOI] [PubMed] [Google Scholar]
- Wallimann K.; Vasella A. Phosphonic-Acid Analogues of the N-Acetyl-2-Deoxyneiiraniinic Acids: Synthesis and Inhibition of Vibrio Cholerae Sialidase. Helv. Chim. Acta 1990, 73 (5), 1359–1372. 10.1002/hlca.19900730523. [DOI] [Google Scholar]
- White C. L.; Janakiraman M. N.; Laver G. W.; Philippon C.; Vasella A.; Air G. M.; Luo M. A Sialic Acid-Derived Phosphonate Analog Inhibits Different Strains of Influenza Virus Neuraminidase with Different Efficiencies. J. Mol. Biol. 1995, 245 (5), 623–634. 10.1006/jmbi.1994.0051. [DOI] [PubMed] [Google Scholar]
- Gao J.; Martichonok V.; Whitesides G. M. Synthesis of a Phosphonate Analog of Sialic Acid (Neu5Ac) Using Indium-Mediated Allylation of Unprotected Carbohydrates in Aqueous Media. J. Org. Chem. 1996, 61 (26), 9538–9540. 10.1021/jo961290y. [DOI] [Google Scholar]
- Chan T.-H.; Xin Y.-C.; von Itzstein M. Synthesis of Phosphonic Acid Analogues of Sialic Acids (Neu5Ac and KDN) as Potential Sialidase Inhibitors. J. Org. Chem. 1997, 62 (11), 3500–3504. 10.1021/jo961891p. [DOI] [Google Scholar]
- Shie J.-J.; Fang J.-M.; Lai P.-T.; Wen W.-H.; Wang S.-Y.; Cheng Y.-S. E.; Tsai K.-C.; Yang A.-S.; Wong C.-H. A Practical Synthesis of Zanamivir Phosphonate Congeners with Potent Anti-Influenza Activity. J. Am. Chem. Soc. 2011, 133 (44), 17959–17965. 10.1021/ja207892q. [DOI] [PubMed] [Google Scholar]
- Vavricka C. J.; Muto C.; Hasunuma T.; Kimura Y.; Araki M.; Wu Y.; Gao G. F.; Ohrui H.; Izumi M.; Kiyota H. Synthesis of Sulfo-Sialic Acid Analogues: Potent Neuraminidase Inhibitors in Regards to Anomeric Functionality. Sci. Rep. 2017, 7 (1), 8239. 10.1038/s41598-017-07836-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schauer R.; Schröder C.; Shukla A. K.. New Techniques for the Investigation of Structure and Metabolism of Sialic Acids. In Advances in Experimental Medicine and Biology; 1984; Vol. 174, pp 75–86, 10.1007/978-1-4684-1200-0_7. [DOI] [PubMed] [Google Scholar]
- Suzuki M.; Suzuki A.; Yamakawa T.; Matsunaga E. Characterization of 2,7-Anhydro-N-Acetylneuraminic Acid in Human Wet Cerumen. J. Biochem. 1985, 97 (2), 509–515. 10.1093/oxfordjournals.jbchem.a135085. [DOI] [PubMed] [Google Scholar]
- Li Y. T.; Nakagawa H.; Ross S. A.; Hansson G. C.; Li S. C. A Novel Sialidase Which Releases 2,7-Anhydro-Alpha-N-Acetylneuraminic Acid from Sialoglycoconjugates. J. Biol. Chem. 1990, 265 (35), 21629–21633. 10.1016/S0021-9258(18)45787-0. [DOI] [PubMed] [Google Scholar]
- Chou M. Y.; Li S. C.; Kiso M.; Hasegawa A.; Li Y. T. Purification and Characterization of Sialidase L, a NeuAc Alpha 2->3Gal-Specific Sialidase. J. Biol. Chem. 1994, 269 (29), 18821–18826. 10.1016/S0021-9258(17)32240-8. [DOI] [PubMed] [Google Scholar]
- Xu G.; Potter J. A.; Russell R. J. M.; Oggioni M. R.; Andrew P. W.; Taylor G. L. Crystal Structure of the NanB Sialidase from Streptococcus Pneumoniae. J. Mol. Biol. 2008, 384 (2), 436–449. 10.1016/j.jmb.2008.09.032. [DOI] [PubMed] [Google Scholar]
- Tailford L. E.; Owen C. D.; Walshaw J.; Crost E. H.; Hardy-Goddard J.; le Gall G.; de Vos W. M.; Taylor G. L.; Juge N. Discovery of Intramolecular Trans-Sialidases in Human Gut Microbiota Suggests Novel Mechanisms of Mucosal Adaptation. Nat. Commun. 2015, 6 (1), 7624. 10.1038/ncomms8624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crost E. H.; Tailford L. E.; Monestier M.; Swarbreck D.; Henrissat B.; Crossman L. C.; Juge N. The Mucin-Degradation Strategy of Ruminococcus Gnavus : The Importance of Intramolecular Trans -Sialidases. Gut Microbes. 2016, 7 (4), 302–312. 10.1080/19490976.2016.1186334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao A.; Slack T. J.; Li Y.; Shi D.; Yu H.; Li W.; Liu Y.; Chen X. Streptococcus Pneumoniae Sialidase SpNanB-Catalyzed One-Pot Multienzyme (OPME) Synthesis of 2,7-Anhydro-Sialic Acids as Selective Sialidase Inhibitors. J. Org. Chem. 2018, 83 (18), 10798–10804. 10.1021/acs.joc.8b01519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J.; McClane B. A. The Sialidases of Clostridium Perfringens Type D Strain CN3718 Differ in Their Properties and Sensitivities to Inhibitors. Appl. Environ. Microbiol. 2014, 80 (5), 1701–1709. 10.1128/AEM.03440-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J.; Freedman J. C.; McClane B. A. NanI Sialidase, CcpA, and CodY Work Together To Regulate Epsilon Toxin Production by Clostridium Perfringens Type D Strain CN3718. J. Bacteriol. 2015, 197 (20), 3339–3353. 10.1128/JB.00349-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li J.; McClane B. A.. NanI Sialidase Can Support the Growth and Survival of Clostridium Perfringens Strain F4969 in the Presence of Sialyated Host Macromolecules (Mucin) or Caco-2 Cells. Infect. Immun. 2018, 86 ( (2), ), 10.1128/IAI.00547-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim B. R.; Park J.-Y.; Jeong H. J.; Kwon H.-J.; Park S.-J.; Lee I.-C.; Ryu Y. B.; Lee W. S. Design, Synthesis, and Evaluation of Curcumin Analogues as Potential Inhibitors of Bacterial Sialidase. J. Enzyme. Inhib. Med. Chem. 2018, 33 (1), 1256–1265. 10.1080/14756366.2018.1488695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee Y.; Ryu Y. B.; Youn H. S.; Cho J. K.; Kim Y. M.; Park J. Y.; Lee W. S.; Park K. H.; Eom S. H. Structural Basis of Sialidase in Complex with Geranylated Flavonoids as Potent Natural Inhibitors. Acta. Crystallogr. D. Biol. Crystallogr. 2014, 70 (5), 1357–1365. 10.1107/S1399004714002971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Y.; Curtis-Long M. J.; Yuk H. J.; Kim D. W.; Tan X. F.; Park K. H. Bacterial Neuraminidase Inhibitory Effects of Prenylated Isoflavones from Roots of Flemingia Philippinensis. Bioorg. Med. Chem. 2013, 21 (21), 6398–6404. 10.1016/j.bmc.2013.08.049. [DOI] [PubMed] [Google Scholar]
- Wang Y.; Kim J. Y.; Song Y. H.; Li Z. P.; Yoon S. H.; Uddin Z.; Ban Y. J.; Lee K. W.; Park K. H. Highly Potent Bacterial Neuraminidase Inhibitors, Chromenone Derivatives from Flemingia Philippinensis. Int. J. Biol. Macromol. 2019, 128, 149–157. 10.1016/j.ijbiomac.2019.01.105. [DOI] [PubMed] [Google Scholar]
- Ullah M.; Uddin Z.; Song Y. H.; Li Z. P.; Kim J. Y.; Ban Y. J.; Park K. H. Bacterial Neuraminidase Inhibition by Phenolic Compounds from Usnea Longissima. S. Afr. J. Bot. 2019, 120, 326–330. 10.1016/j.sajb.2018.10.020. [DOI] [Google Scholar]
- Walther E.; Xu Z.; Richter M.; Kirchmair J.; Grienke U.; Rollinger J. M.; Krumbholz A.; Saluz H. P.; Pfister W.; Sauerbrei A.; Schmidtke M. Dual Acting Neuraminidase Inhibitors Open New Opportunities to Disrupt the Lethal Synergism between Streptococcus Pneumoniae and Influenza Virus. Front. Microbiol. 2016, 7, 357. 10.3389/fmicb.2016.00357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee Y.; Youn H.-S.; Lee J.-G.; An J. Y.; Park K. R.; Kang J. Y.; Ryu Y. B.; Jin M. S.; Park K. H.; Eom S. H. Crystal Structure of the Catalytic Domain of Clostridium Perfringens Neuraminidase in Complex with a Non-Carbohydrate-Based Inhibitor, 2-(Cyclohexylamino)Ethanesulfonic Acid. Biochem. Biophys. Res. Commun. 2017, 486 (2), 470–475. 10.1016/j.bbrc.2017.03.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arioka S.; Sakagami M.; Uematsu R.; Yamaguchi H.; Togame H.; Takemoto H.; Hinou H.; Nishimura S.-I. Potent Inhibitor Scaffold against Trypanosoma Cruzi Trans-Sialidase. Bioorg. Med. Chem. 2010, 18 (4), 1633–1640. 10.1016/j.bmc.2009.12.062. [DOI] [PubMed] [Google Scholar]
- Albrecht C.; Akissi Z. L. E.; Yao-Kouassi P. A.; Alabdul Magid A.; Maurice P.; Duca L.; Voutquenne-Nazabadioko L.; Bennasroune A. Identification and Evaluation of New Potential Inhibitors of Human Neuraminidase 1 Extracted from Olyra Latifolia l.: A Preliminary Study. Biomedicines. 2021, 9 (4), 411. 10.3390/biomedicines9040411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffmann A.; Richter M.; von Grafenstein S.; Walther E.; Xu Z.; Schumann L.; Grienke U.; Mair C. E.; Kramer C.; Rollinger J. M.; Liedl K. R.; Schmidtke M.; Kirchmair J. Discovery and Characterization of Diazenylaryl Sulfonic Acids as Inhibitors of Viral and Bacterial Neuraminidases. Front. Microbiol. 2017, 8, 205. 10.3389/fmicb.2017.00205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grienke U.; Richter M.; Walther E.; Hoffmann A.; Kirchmair J.; Makarov V.; Nietzsche S.; Schmidtke M.; Rollinger J. M. Discovery of Prenylated Flavonoids with Dual Activity against Influenza Virus and Streptococcus Pneumoniae. Sci. Rep. 2016, 6, 27156. 10.1038/srep27156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen R.; Fang Z.; Huang Y. Neuropsychiatric Events in an Adult Patient with Influenza a (H3N2) Treated with Oseltamivir (Tamiflu): A Case Report. BMC. Infect. Dis. 2019, 19 (1), 224. 10.1186/s12879-019-3827-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murti K. G.; Webster R. G. Distribution of Hemagglutinin and Neuraminidase on Influenza Virions as Revealed by Immunoelectron Microscopy. Virology 1986, 149 (1), 36–43. 10.1016/0042-6822(86)90084-X. [DOI] [PubMed] [Google Scholar]
- Liu C.; Eichelberger M. C.; Compans R. W.; Air G. M. Influenza Type A Virus Neuraminidase Does Not Play a Role in Viral Entry, Replication, Assembly, or Budding. J. Virol. 1995, 69 (2), 1099–1106. 10.1128/jvi.69.2.1099-1106.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun X.-L. Recent Anti-Influenza Strategies in Multivalent Sialyloligosaccharides and Sialylmimetics Approaches. Curr. Med. Chem. 2007, 14 (21), 2304–2313. 10.2174/092986707781696582. [DOI] [PubMed] [Google Scholar]
- Fraser B. H.; Hamilton S.; Krause-Heuer A. M.; Wright P. J.; Greguric I.; Tucker S. P.; Draffan A. G.; Fokin V. v.; Sharpless K. B. Synthesis of 1,4-Triazole Linked Zanamivir Dimers as Highly Potent Inhibitors of Influenza A and B. Med. Chem. Commun. 2013, 4 (2), 383–386. 10.1039/C2MD20300F. [DOI] [Google Scholar]
- Fu L.; Bi Y.; Wu Y.; Zhang S.; Qi J.; Li Y.; Lu X.; Zhang Z.; Lv X.; Yan J.; Gao G. F.; Li X. Structure-Based Tetravalent Zanamivir with Potent Inhibitory Activity against Drug-Resistant Influenza Viruses. J. Med. Chem. 2016, 59 (13), 6303–6312. 10.1021/acs.jmedchem.6b00537. [DOI] [PubMed] [Google Scholar]
- Yan Z.-L.; Liu A.-Y.; Wei X.-X.; Zhang Z.; Qin L.; Yu Q.; Yu P.; Lu K.; Yang Y. Divalent Oseltamivir Analogues as Potent Influenza Neuraminidase Inhibitors. Carbohydr. Res. 2019, 477, 32–38. 10.1016/j.carres.2019.03.012. [DOI] [PubMed] [Google Scholar]
- Assailly C.; Bridot C.; Saumonneau A.; Lottin P.; Roubinet B.; Krammer E.; François F.; Vena F.; Landemarre L.; Alvarez Dorta D.; Deniaud D.; Grandjean C.; Tellier C.; Pascual S.; Montembault V.; Fontaine L.; Daligault F.; Bouckaert J.; Gouin S. G. Polyvalent Transition-State Analogues of Sialyl Substrates Strongly Inhibit Bacterial Sialidases. Chem.—Eur. J. 2021, 27 (9), 3142–3150. 10.1002/chem.202004672. [DOI] [PubMed] [Google Scholar]
- Baell J. B.; Holloway G. A. New Substructure Filters for Removal of Pan Assay Interference Compounds (PAINS) from Screening Libraries and for Their Exclusion in Bioassays. J. Med. Chem. 2010, 53 (7), 2719–2740. 10.1021/jm901137j. [DOI] [PubMed] [Google Scholar]
- Baell J. B. Feeling Nature’s PAINS: Natural Products, Natural Product Drugs, and Pan Assay Interference Compounds (PAINS). J. Nat. Prod. 2016, 79 (3), 616–628. 10.1021/acs.jnatprod.5b00947. [DOI] [PubMed] [Google Scholar]




