Next Article in Journal
Cytokine-Targeting Biologic Therapies for Alopecia Areata: A Comprehensive Review of Mechanism of Action, Clinical Efficacy, and Adverse Events
Previous Article in Journal
Unleashing the Power of Biologics: Exploring the Governance and Regulation of Membrane-Based Virus Purification (MVP) Technologies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sialic Acids in Health and Disease

by
Gerardo N. Guerrero-Flores
1,2,
Fayth M. Butler
3,4,
Veronica L. Martinez Marignac
5,
Guangyu Zhang
3,
Fabio J. Pacheco
1 and
Danilo S. Boskovic
3,6,*
1
Interdisciplinary Center for Research in Health and Behavioral Sciences, School of Medicine, Universidad Adventista del Plata, Libertador San Martín 3103, Argentina
2
Faculty of Medical Sciences, Universidad Nacional de Rosario (UNR), Rosario 3100, Argentina
3
Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
4
Center for Nutrition, Healthy Lifestyles, and Disease Prevention, School of Public Health, Loma Linda University, Loma Linda, CA 92350, USA
5
Interdisciplinary Laboratory of Biology and Molecular Genetics (IBIOGEM-CONICET), Diamante 3105, Argentina
6
Department of Earth and Biological Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
*
Author to whom correspondence should be addressed.
Biologics 2025, 5(2), 10; https://doi.org/10.3390/biologics5020010
Submission received: 19 December 2024 / Revised: 4 February 2025 / Accepted: 7 March 2025 / Published: 26 March 2025

Abstract

:
Vertebrate cell surfaces exhibit intricate arrangements of glycosaminoglycan polymers, which are primarily linked to lipids and proteins. Numerous soluble secreted proteins are also decorated with either individual sugar molecules or their polymers. The carbohydrate polymers commonly possess terminal nine-carbon sugars, known as sialic acids. Due to their widespread distribution and strategic positioning, sialic acids play a crucial role in mediating and regulating a wide range of physiologic processes and pathologic conditions. Human- or animal-based investigations predominantly concentrate on the effects of sialic acids during infections, inflammations, vascular disorders, or cancers. Further investigations encompass a variety of applications, including cell–cell interactions, signaling, host–pathogen interactions, and other biological functions associated with nutrition, metabolism, or genetic disorders. Nevertheless, future mechanistic investigations are needed to clarify the specific roles of sialic acids in these varied contexts, so that more effective interventions may be developed.

Graphical Abstract

1. Introduction

Cell surfaces are covered by a glycocalyx [1] composed of complex carbohydrate polymers (also known as glycans) anchored to cell membranes via glycoprotein or glycolipid linkages [2,3,4,5]. Biosynthetically derived from neuraminic acid [6,7], sialic acids represent a family of nine-carbon keto-aldonic acids (nonulosonic acids (NulOs)) [8]. These normally occupy the terminal positions of cell surface glycosaminoglycan moieties [9,10]. They are, however, also present freely in various biofluids or as modifiers of soluble secreted proteins [11,12,13]. Sialic acids are found in a wide range of animal phyla, including Platyhelminthes [14], Echinodermata [15], Cephalopoda [16], Crustacea [17], Cephalochordata [18], and Vertebrata [19]. In addition, sialic acids are also known to be associated with viruses, bacteria, protozoa, and fungi [20,21,22]. Consequently, it is not surprising that sialic acids are involved in a large variety of physiologic functions, such as scavenging of reactive oxygen species (ROS) [23], cell–cell interactions, receptor-binding, and cell signaling [9,13,21]. Further, sialic acids are also recognized to play a central role in some pathologic processes, like cardiovascular disorders and cancer [24,25,26].

1.1. Analysis of Sialic Acids

Analytical approaches to characterizing sialic acids include separation techniques, such as chromatography [27] and capillary electrophoresis [28], and detection methods such as colorimetry [29] and spectrophotometry [30]. Colorimetry and capillary electrophoresis are relatively straightforward to operate, but tend to have lower sensitivity and specificity compared to other methods [31]. High-performance liquid chromatography (HPLC) is commonly used for the separation of sialic acids [31,32], and is generally combined with ultraviolet [33] or fluorescence detectors [27,34]. Currently, the study of sialic acids is greatly facilitated by the application of mass spectrometric techniques for specific detection and quantification [35,36,37,38,39].
Mass spectrometry can provide qualitative (structural) and quantitative (molecular mass or concentration) information about specific molecules of interest [40]. Mass spectrometers comprise four main components [41,42]: (1) an ion source, enabling ionization of sample molecules; (2) a mass analyzer, which separates ions according to their mass-to-charge ratio (m/z); (3) a detector, which measures the separated ion abundances in the form of electrical signals, and (4) a recording device, which transforms the detector signal into a format suitable for further analysis and processing. The process itself unfolds in its respective stages. (a) The vaporization and ionization of the sample [43,44]. Various approaches may be used for ionization in the gas phase, including electron ionization, chemical ionization, electrospray ionization (ESI), photoionization, inductively coupled plasma, and matrix-assisted laser desorption ionization [45]. (b) Acceleration of ions through an electric field to reach the same kinetic energy [46]. (c) Separation and sorting of ions according to their specific m/z ratios [40,47]. (d) Fragmentation of ions to generate fragments, the distribution of which is characteristic for each molecule. This is typically accomplished in tandem mass spectrometry. (e) Detection and recording of ions, based on the ion abundance and relative intensity [43,45].
Technical advantages of mass spectrometry include high sensitivity, specific selectivity, and small sample requirements [35,48]. Consequently, mass spectrometry has become the preferred approach for sialic acid research [49]. As a result of continuing advancements, specific sialic acids are now being studied in a variety of organisms and human tissues. Moreover, their potential roles in human health and disease are also being investigated.

1.2. Structures of Sialic Acids

Sialic acids constitute a family of more than 50 distinct molecules, 15 of which have been identified in human tissues [9,24]. The differences between sialic acids are based on particular N-linked or O-linked modifying groups, such as N-acetyl-, N-glycolyl, and O-acetyl, or, less frequently, the O-lactyl, O-methyl, and O-sulfate groups (Figure 1) [10,25,50]. The modifications confer distinct properties to the nine-carbon sugars, which support their specific physiologic functions [12]. Two major structural features include an often-modified amino group at position 5 and a carboxyl group at position 1. This carboxyl group, with its characteristic pKa of about 2.2, confers a negative charge on the sialic acid within a wide physiologic pH range [10,13]. Esterification of the hydroxyl groups at positions 7, 8, or 9 with acetic acid, in the form of O-acetylation, occurs less frequently [10]. Other known modifications include sulfation or methylation at position 8, or lactylation at position 9. Substitutions may occur at multiple sites, as illustrated by 7,8,9-tri-O-acetyl-N-acetyl- and N-glycolylneuraminic acid [8,9,51].
Widely distributed in a variety of human tissues and fluids [52], sialic acids exhibit particular functional roles associated with their specific structural features [10]. The most common member of this family is N-acetylneuraminic acid (Neu5Ac), with an acetylated amino group at position 5 (Figure 2) [7,50]. Neu5Ac can be converted to a less common N-glycolylneuraminic acid (Neu5Gc) by cytidine monophosphate-N-acetylneuraminic acid hydroxylase (Cmah), which hydroxylates the acetyl group [9,50,53]. Another sialic acid member, mainly found in its free form [10,54,55], and generally expressed at lower levels than Neu5Ac or Neu5Gc in mammalian tissues, is 2-keto-3-deoxy-D-glycero-D-galacto-nononic acid (KDN) [8,36,55,56]. More in-depth structural and stereochemical sialic acid analyses are presented elsewhere [6,10,25,57,58,59,60].

1.3. Synthesis

In mammals, the synthesis of sialic acids involves more than 30 genes, encoding enzymes, and transporters allocated in various cellular compartments [61]. The synthesis of Neu5Ac begins in the cytosol with glucose, which enters the hexosamine biosynthetic pathway (HBP). This pathway phosphorylates glucose using ATP to produce the key metabolite uridine-5′-di-phospho-N-acetylglucosamine (UDP-GlcNAc) [60,62]. Then, the epimerase domain of UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) mediates the formation of the uncharged monosaccharide N-acetylmannosamine (ManNAc) [63], with simultaneous cleavage of UDP [64,65,66]. ManNAc is phosphorylated by the kinase domain of GNE, leading to ManNAc-6P [67]. Sialic acid synthase (NANS) converts ManNAc-6P to Neu5Ac-9P in a condensation reaction with phosphoenol-pyruvate (PEP) [51], and subsequently, N-acetylneuraminic-acid-phosphatase (NANP) mediates the dephosphorylation to produce free Neu5Ac [68]. The free monosaccharide is activated in the nucleus by coupling with cytidine monophosphate (CMP), via the action of CMP-N-acetylneuraminate synthetase (CMAS), producing CMP-Neu5Ac [10,69]. Then, the CMP-Neu5Ac is added to lipid or protein to form glycans in the Golgi apparatus [70,71], or transferred to carbohydrate chains of nascent glycoconjugates by sialyltransferases (Figure 3) [10,13,24,72]. At this point, the bound Neu5Ac may be further altered via O-acetylation or O-methylation [73], before the glycoconjugate is transferred to the cell surface [72].
The main precursor for KDN biosynthesis is mannose (Man). The synthetic pathway comprises a series of reactions, similar to those for the biosynthesis of Neu5Ac (Figure 4) [60,74,75,76]. (1) Man + ATP → Man-6-P + ADP; (2) Man-6-P + PEP → KDN-9-P + Pi; (3) KDN-9-P → KDN + Pi. Reaction 1, catalyzed by a hexokinase, represents the 6-O-phosphorylation of mannose to form D-mannose-6-phosphate (Man-6-P). Reaction 2, catalyzed by KDN-9-phosphate (KDN-9-P) synthetase, condenses Man-6-P and PEP to form KDN-9-P. Reaction 3, catalyzed by a phosphatase, involves dephosphorylation of KDN-9-P to yield free KDN [12,55,74].

1.4. Functions

Sialic acids are involved in a variety of physiologically important functions [24,77], such as (a) antioxidant protection [78], (b) cell surface interactions [1], (c) infant bone development and skeletal growth [79], (d) immune recognition [80], (e) brain development [79,81], and (f) learning and memory [82].
In GNE myopathy patients and in a GNE mutant mouse model, muscles were hyposialylated and proteins were highly modified via S-nitrosylation, while oxidative stress-responsive genes were significantly upregulated [83]. In both cases, reactive oxygen species (ROS) production was elevated with cellular hyposialylation [83]. However, increasing overall sialylation through the intake of extrinsic sialic acid reduced ROS and protein S-nitrosylation. Additionally, the oral antioxidant N-acetylcysteine ameliorated muscle atrophy and weakness in GNE-mutant mice [83,84]. Together, this line of evidence demonstrates that overall sialylation is associated with increased antioxidant protection and improved reserve capacity to handle incidental ROS.
A biallelic mutation in NANS is associated with infantile-onset severe developmental delay and skeletal dysplasia [79,85]. The NANS gene is responsible for encoding the synthase for Neu5Ac [79,86]. Consequently, the immediate precursor substrate builds up, so that body fluids from affected individuals will have elevated levels of N-acetyl-D-mannosamine [79]. Knockdown of nansa, a zebrafish ortholog for the human NANS gene, results in abnormal skeletal development, while exogenously added sialic acid partially rescues this skeletal phenotype. Thus, NANS-mediated synthesis of sialic acid is required for early skeletal growth [79].
The glycocalyx is central to immune recognition and checkpoint inhibition, which enables pathogen detection and activation of immune defenses [80,87]. This carbohydrate coat serves as a cellular barrier that can either facilitate or obstruct immune cell interactions. Depending on the cell type and the composition of the glycosaminoglycan layer, the immune system may identify it as representing “self”. Alternatively, the sialic acid-rich layer may also potentially shield pathogens or cancerous cells from immune detection by masking their antigen-decorated surfaces with a carbohydrate-rich layer [88]. Ordinarily, for cancer cells to establish malignant lesions, they first need to evade recognition by immune cells [89,90]. They achieve this by tailoring their glycocalyx, which covers their neoantigens and other ligands, protecting them from acquired immune system cells. This alters the phagocytic efforts of myeloid cells and changes the normal immune responses [90]. This explains the upregulation of transmembrane mucins and other highly sialylated glycoproteins in cancer cells, as a way of establishing a protective glycocalyx [80,91,92]. Accordingly, in mouse models, tumor cells with the most efficient surface glycocalyx shielding have been shown to have the highest metastatic potential [93].
The human brain grows rapidly in early life and requires adequate nutrition [94,95,96]. Breastfeeding significantly improves the cognitive development of infants [94,97,98]. These benefits have been attributed to human milk oligosaccharides (HMOSs), some of which are linked with sialic acids [94,97,99]. Sialic acids are recognized as essential molecules for the proper development of gangliosides, and therefore, as critical for brain development and function [95]. Dietary supplementation with sialic acid or sialylated oligosaccharides is known to enhance intelligence and cognitive performance in early and later life [95,96,100]. Similarly, exogenous supplementation of sialic acid in animal models increases its concentration in the frontal cortex and improves learning performance [101]. As a building block for neurons, dietary 3′-sialyllactose is associated with improved language development in infants [94]. Conversely, some intellectual developmental disorders are associated with genetic defects affecting the synthesis of endogenous Neu5Ac [79].

1.5. Genetic Disorders

1.5.1. UDP-GlcNAc 2-Epimerase/ManNAc Kinase (GNE) Myopathy

GNE myopathy is a hereditary myogenic disorder that results in muscle weakness due to a defect in the muscle itself. The GNE gene provides instructions for expression of the bifunctional GNE enzyme, which plays a key role in the synthesis of Neu5Ac [83,84,102]. The N-terminal epimerase domain of GNE uses UDP-GlcNAc as a substrate and epimerizes it into ManNAc by the epimerase domain, which is then subsequently phosphorylated by the kinase domain of the GNE enzyme [63,83].
Usually, GNE myopathy patients present with muscle symptoms between the ages of 15 and 35 [103]. The pathophysiology of the disease is not entirely understood, but hyposialylation of muscle glycans is thought to play an important role [84,102]. The typical presentation of bilateral foot drop in early adulthood is a consequence of weakness in the anterior tibialis muscles [102]. Over the subsequent decades, the disease gradually progresses, gradually affecting skeletal muscles throughout the body [102]. The quadriceps muscles tend to be relatively spared until the advanced stages of the disease [84,102,103].
Currently, a phase 2 clinical trial with ManNAc supplementation as a therapeutic intervention is ongoing for patients diagnosed with GNE myopathy [63,104]. Preliminary evidence of its long-term safety and biochemical efficacy, consistent with the intended mechanism of action, is reported in this cohort of patients [63]. Additionally, ManNAc supplementation restores the intracellular biosynthesis of Neu5Ac in affected subjects. Surprisingly, such an improvement is observed even in patients with homozygous GNE mutations in the kinase domain [104].

1.5.2. Free Sialic Acid Storage Disorder (FSASD)

Lysosomes are cytoplasmic organelles that contain a variety of hydrolases [105]. A genetic deficiency in one of these hydrolases can result in the accumulation of material to be degraded by the lysosome [106]. Sometimes, such lysosomal storage can be caused by the deficiency of an activator protein [107,108]. Alternatively, accumulation of lysosomal material can also occur due to a transporter deficiency, as seen in the Salla disease [105,109].
Damage to the transport mechanism of free Neu5Ac through the lysosomal membrane leads to lysosomal accumulation of Neu5Ac [110,111]. Lysosomal free sialic acid storage disorders constitute a spectrum of uncommon, autosomal recessive, neurodegenerative, and multisystemic disorders caused by defective sialic acid lysosomal membrane exporter Sialin (Figure 3) [112,113,114].
The biallelic pathogenic gene variants of SLC17A5, which encode Sialin, produce three forms of free sialic acid storage disorders: (a) Salla disease, (b) intermediate severe Salla disease, and (c) infantile free sialic acid storage disease [109,114]. Salla disease and infantile free sialic acid storage disease exemplify the mildest and the most severe manifestations of the FSASD phenotypic spectrum, respectively [109]. At six months of age, infants with Salla disease typically manifest hypotonia, ataxia, developmental delay, and, in most cases, hypomyelination [105,115]. This is followed by a gradual neurological decline in an otherwise near-normal lifespan [115]. In contrast, infantile free sialic acid storage disease is characterized by nonimmune hydrops fetalis (24%), hepatosplenomegaly, failure to thrive, severe developmental delay, cardiomegaly, club feet, increasingly coarse facial features, neurological deterioration, and premature death [112]. The recently described intermediate severe Salla disease represents individuals with an intermediate phenotype [115]. Patients usually present within the initial six months of life with severe hypotonia and developmental delay, gradually progressing to ataxia, spasticity, and epilepsy [109].

2. Diet

In addition to their endogenous production, sialic acids can also be obtained from dietary sources, absorbed by the intestine, and transported via circulation to tissues [104,116,117]. Most of the common sialic acids are obtained through dietary sources [118], either from red meat, in the form of lamb, pork, or beef, or from a variety of milk products [119,120]. Exogenous Neu5Ac can be picked up by cells via fluid pinocytosis and a lysosomal transporter [121]. In contrast to Neu5Ac, Neu5Gc cannot be synthesized by human tissues from endogenous precursors [122]. Instead, it is metabolically incorporated in the tissues of subjects who consume red meat [123,124,125]. Consequently, low levels of surface expression of diet-derived Neu5Gc occur on endothelial and some epithelial cells [53,126]. Once sialic acids are internalized by cells into the cytosol [116], they are integrated with endogenous glycoconjugates, of which a relatively small fraction is retained in the body, while the rest is metabolized and excreted in urine [126,127] or feces [104].
Unlike animals, plants are known to lack sialic acids [110,128,129]. However, they are rich sources of precursors for the synthesis of sialic acids [7,60,75,129]. In fish, O-acetylated Neu5Ac occurs frequently on N-glycans, while a smaller number of Neu5Acα2-8Neu5Ac structures are also observed [130]. Neu5Gc is seen very rarely in certain marine sources (fish eggs or echinoderms) [116,131,132] and is apparently not synthesized by birds or reptiles [133,134].
The precise metabolic fates and the relative contributions of sialic acids from endogenous and dietary sources in humans remain unknown, making the inferences from current models difficult [126]. Implications of dietary uptake and the utilization of Neu5Ac and Neu5Gc deserve further investigation.

Microbiome and Sialic Acids

Plant polysaccharides include long-chain glycans, as components of plant-based fiber, which animals generally cannot digest [135]. Fiber, however, is an essential source of carbon and energy that supports the maintenance and functioning of gut microbiota [136]. These gut bacteria, which are able to utilize various types of plant glycans [136], are categorized into seven predominant divisions: Actinobacteria, Bacteroidetes, Cyanobacteria, Firmicutes, Fusobacteria, Proteobacteria, and Verrucomicrobia. Of these, Bacteroidetes and Firmicutes represent more than 90% of the microbiome [137]. Gut bacteria are believed to be essential to human health by (a) conferring additional energy-harvesting capacity, (b) providing niche exclusion of pathogens, (c) producing vitamins and glycans, and (d) carrying out molecular communication activities, all of which form an integral part of human physiologic processes [136,138,139]. Thus, the quantity and quality of plant glycans consumed daily by the human host can have an impact on health [136]. Both pre-clinical and clinical studies indicate that different types of dietary plant glycans and their metabolic products contribute beneficial metabolic effects to the host [140]. For example, the fermentation of cellulose, hemicellulose, resistant starch, pectin, lignin, or various oligosaccharides can lead to the production of short-chain fatty acids (SCFAs) like acetic, propionic, or butyric acids, which, in turn, help to regulate glucose and lipid metabolism [140,141]. The release of these fatty acids can occur during digestion in the upper gastrointestinal tract but may also continue in the colon [142].
Studies of the microbiome composition in adults are mainly focused on the resulting health impacts, while the effects of dietary sialic acids in this context are largely overlooked [143]. Given the considerable dietary variation between different people, the effects of dietary sialic acids on the adult gut microbiome are often difficult to tease apart. Consequently, cohort studies focusing on particular dietary patterns, like the Adventist Health Study-2 [144] or the EPIC-OXFORD study [145], are potentially helpful in examining the impact of dietary sialic acids, like Neu5Ac or Neu5Gc, on the gut microbiome. Such information about the relationship between dietary sialic acids and the microbiome may reveal additional insights into several non-communicable diseases, like type 2 diabetes [146,147], cardiovascular disorders [148,149], or cancer [12,150,151,152].
Bacteria can release free sialic acids or use them as nutrient sources [153,154]. Gut microbiome symbionts, such as Ruminococcus gnavus, can scavenge Neu5Ac from mucins and convert it to 2,7-anhydro-Neu5Ac [1]. However, catabolism of 2,7-anhydro-Neu5Ac is not restricted to R. gnavus. Escherichia coli can also transport and catabolize sialic acids such as Neu5Ac, Neu5Gc, or KDN to satisfy its carbon and nitrogen needs. These sialic acids are transported via the transporter NanT and catabolized using the sialic acid aldolase NanA [155]. Glycosylhydrolases cleave the glycoketosidic linkages of sialic acid O-acceptor substrates through an exohydrolytic reaction [156]. In addition, bacterial sialidases can release Neu5Gc from red meat [153,157], helping to reduce the inflammatory effects of red meat consumption [143].

3. Interactions with Pathogens

Sialic acids act as receptors for various pathogens that produce infectious diseases [158]. Some pathogens can synthesize or appropriate sialic acids from their hosts and incorporate them into their own glycoconjugates and derivatives [159]. For example, bacteria produce adhesins or toxins that interact with sialic acids on host cells [157], viruses can use sialic acids to attach to and enter cells [160], and protozoa can use sialic acids to elude host immunity [161,162,163].

3.1. Sialic Acids and Bacteria

Some bacterial species can produce Neu5Ac and corresponding glycoconjugates. Others, in contrast, are merely able to use sialic acids as carbon sources. This capability to use sialic acids appears to be correlated with bacterial virulence in a range of infections, particularly in the profusely sialylated environment of the gut [143]. Bacteria obtain sialic acids in two ways: either via de novo synthesis, or from scavenging pathways using specific sialidases to cleave sialic acids from glycoconjugates [155,164]. As in eukaryotes, the de novo synthesis pathway in bacteria begins with the phosphorylation of glucose by a hexokinase [165].
In E. coli, in the NeuC pathway, glucose undergoes conversion into glucose-6-phosphate through the action of a hexokinase [166,167,168,169,170]. This is followed by the conversion of glucose-6-phosphate to fructose-6-phosphate by glucose-6-phosphate isomerase (PGI). Four reactions take place sequentially, facilitated by three enzymes: glucosamine-6-phosphate synthase (glmS), phosphoglucosamine mutase (glmM), and the bi-functional enzyme glucosamine-1-phosphate acetyltransferase/N-acetylglucosamine-1-phosphate uridyltransferase (glmU). The result of these reaction steps is the formation of UDP-GlcNAc [166,171]. The conversion of UDP-GlcNAc to ManNAc is catalyzed by UDP-GlcNAc 2-epimerase [172] and NeuC, which is responsible for the biosynthesis of the precursor of N-acetyl-D-mannosamine, followed by NeuB-catalyzed conversion of N-acetyl-D-mannosamine with phosphoenolpyruvate to the product Neu5Ac (Figure 5) [168,171,172,173,174,175,176]. Alternatively, a shortened synthetic pathway can utilize sialyl precursors scavenged from animal hosts to produce Neu5Ac [177], particularly during cell wall synthesis [165].
Some bacteria may be able to synthesize additional structurally related NulOs. Such bacterial NulOs include legionaminic acid [157], pseudaminic acid [178,179], fusaminic acid [180], and acinetaminic acid [181]. In this context, the naming convention typically follows a name derivative of the microbial species first reported as its producer [178]. For example, legionaminic acid was first observed in Legionella pneumophila, known for the respiratory infection Legionnaires disease [182]. Similarly, acinetaminic acid was first identified in the multi-drug-resistant pathogen Acinetobacter baumannii [154,183].

3.1.1. Porphyromonas gingivalis

Periodontitis is one of the most prevalent oral diseases [184], characterized by gradual damage of the periodontal ligament and surrounding tissues, which can potentially include alveolar bone, with ultimate tooth loss [185]. These conditions are characteristically associated with poor brushing and flossing habits, allowing polymicrobial colonization on the surfaces of teeth, in the form of a sticky biofilm [186]. Porphyromonas gingivalis serves as a keystone pathogen in the microbial community, responsible for the occurrence and progression of periodontitis [187,188,189].
The survival of P. gingivalis next to the oral mucosa depends on its dynamic interface and its cell membrane [190], which facilitates the release of wastes and the acquisition of nutrients. Once attached to the oral cavity, P. gingivalis utilizes the host’s amino acids and other carbon-containing molecules as energy sources and as building materials for its functions and growth [191,192]. Interestingly, this Gram-negative pathogen, although long described as asaccharolytic, can utilize sugars such as glucose for the biosynthesis of other intracellular macromolecules [193]. The cell membrane of P. gingivalis also contributes toward effective tissue colonization [190], by releasing vesicles with an arsenal of virulence factors, including lipopolysaccharides (LPS), fimbriae, gingipains, and others [194,195]. The host’s immune system responds to these by ramping up the production and release of inflammatory cytokines such as tumor necrosis factor (TNF)-α and interleukin (IL)-1β [196]. The interplay between the destructive microorganisms and the pro-inflammatory immune responses causes periodontal tissue degradation [190,197].
In addition, P. gingivalis can produce and release sialidases, which can also serve as virulence factors [156]. The infection may be sustained by the action of sialidases and sialopeptidases (the O-sialoglycoprotease hydrolyzes the sialic acid O-acceptor substrate through an endohydrolytic reaction [156]), cleaving the sialic acids from the host glycoproteins [156] and contributing to disease progression [189]. In a similar manner, P. gingivalis virulence may be further enhanced if the sialidases also regulate the activity of gingipains (cysteine proteases) [156,198]. Conversely, the inhibition of P. gingivalis sialidases may be strategically helpful in treating periodontitis [189].

3.1.2. Streptococcus pneumoniae

Streptococcus pneumoniae, commonly known as pneumococcus, is a Gram-positive extracellular opportunistic pathogen adapted to the human upper respiratory tract [199]. This bacterium is especially predominant in pre-school-age children and those attending daycare centers [199,200]. S. pneumoniae is responsible for a spectrum of diseases, ranging from mild respiratory tract infections, such as sinusitis or otitis media, to severe conditions, like pneumonia, septicemia, or meningitis [181,182,184]. Pneumococcal infections pose a significant threat to global health. Globally, deaths attributed to pneumococcal disease among children under the age of five have been estimated to reach around 1.8 to 2.0 million, causing about 11% of all deaths in this age group [200,201,202,203].
The primary source of S. pneumoniae spread is mediated by transmission from the host nasopharyngeal airway [203]. Upon entering the human nasopharynx, S. pneumoniae encounters mucus, which is rich in glycoconjugates with terminal sialic acid residues [202]. The respiratory tract is coated with mucin glycoproteins, which are rich in GlcNAc, N-acetylgalactosamine (GalNAc), galactose, and Neu5Ac [202,204]. These sugars are degraded by the glycan-specific metabolic machinery of S. pneumoniae [204], a process that includes the removal of terminal sialic acids, which also serve as carbon sources for the bacteria [204,205].
S. pneumoniae expresses up to three sialidases, including SpNanA, SpNanB, and/or SpNanC [201]. SpNanA, along with SpNanB, provides a source of sialic acids to the pathogen, and helps in host infection, nutrition, colonization, and biofilm formation [201,206,207]. SpNanC serves as a regulator of hydrolytic sialidases, such as SpNanA, catalyzing the production of the sialidase inhibitor Neu5Ac2en [201,208]. SpNanC is thought of as a potential marker of pneumococcal haemolytic uraemic syndrome in children [201], since its activity is linked to the development of this severe complication [201,209,210].
Continuing studies of these sialidases and their structure–function relationships have the potential to yield novel approaches to bacterial infections [188,192,193,194,195]. The development of selective inhibitors against sialidases opens up new perspectives for the treatment of S. pneumoniae [201].

3.1.3. Haemophilus influenzae

Haemophilus influenzae is a human pathogen that is adapted to its host [211,212]. It is classified into typeable strains, which produce a polysaccharide capsule (serotypes a to f), and nontypeable, noncapsulated strains of H. influenzae (NTHi) [211,213]. Healthy humans may asymptomatically carry H. influenzae in the upper respiratory tract [214]. Typable H. influenzae strains are associated with invasive forms of infection, such as sepsis or meningitis [215]. In contrast, NTHi strains are more often responsible for airway-associated inflammation, such as sinusitis, conjunctivitis, pharyngitis, otitis media, and pneumonia, in addition to possible bacteremia and meningitis [216]. Invasive NTHi, representing 43–91% of invasive isolates, is increasing in a number of countries, including Canada, the Netherlands, Portugal, Slovenia, and Sweden [217]. NTHi tends to cause severe invasive disease in neonates and in children, with significant comorbidities [216,218,219]. Approximately 10% of children from 2 to 4 years of age [220], and 17% of children in their first year, succumb to fatal NTHi infections [219,220,221]. The increased incidence of invasive NTHi is due to several factors, including higher vulnerability of some patients [217,220].
H. influenzae relies on its capacity to resist host defenses, including the complement system [212,222]. While evading both innate and adaptive immunity, H. influenzae causes recurrent and persistent infections [222]. Several factors contribute to the survival of NTHi [211,220], including the sialylation of the released lipooligosaccharide from outer membrane fragments during bacterial growth [215,220,223]. This modification protects specific epitopes of the lipooligosaccharide from bactericidal IgM in circulation [222,223]. Sialic acids, incorporated as the terminal residues on lipooligosaccharides, enable NTHi to evade antibacterial defenses, to persist within mucosal tissues, and to form a biofilm. Neither the underlying mechanisms involved in these processes, nor the role of sialic acid scavenged by NTHi during infection, are well understood yet [222,223]. However, since NTHi strains defective in sialic acid utilization are no longer virulent in animal models for otitis media, the sialic acid utilization pathway is a promising therapeutic target [223,224].

3.1.4. Pseudomonas aeruginosa

Pseudomonas aeruginosa is a free-living, motile, and aerobic Gram-negative bacterium [225,226]. It is an opportunistic infectious pathogen, generally associated with nosocomial infections, severe burns, chronic cystic fibrosis, cancer, AIDS, transplantation, and other immunosuppressed conditions [226].
P. aeruginosa lacks the de novo sialic acid synthetic pathway. However, it can take up monosaccharides from exogenous sources, including the glycolyl sialic acid derivative, Neu5Gc [227,228]. When sialic acids are incorporated by P. aeruginosa, the deposition of the complement C3b on the surface of the bacterial cell is reduced [228]. Furthermore, sialylated P. aeruginosa has an affinity for human CD33 receptors, including the immune cell surface receptors Siglecs 7 and 9, as a characteristic shared with other sialylated bacteria [227,229,230,231]. The sialylated P. aeruginosa also suppresses macrophage antimicrobial responses and inhibits phagosome maturation, thereby enabling its persistent viability and intracellular replication within macrophages [227]. In conclusion, acquisition of sialic acids by P. aeruginosa within the host contributes toward bacterial pathogenicity and further host infection by reducing complement deposition and by mimicking Siglec-dependent recognition.

3.1.5. Clostridium perfringens

Clostridium perfringens is the primary pathogen in intestinal and histotoxic infections, producing conditions such as gas gangrene and clostridial myonecrosis [232,233]. C. perfringens is also frequently associated with cases of food poisoning [234]. C. perfringens is known to be able to utilize sialic acids as carbon sources [232,235]. In addition to a variety of toxins [233], this Gram-positive anaerobic bacterium also produces a number of sialidases. Of these, NanH has a cytoplasmic location during the culture log-growth phase. In contrast, NanJ and NanI are secreted exosialidases [232,233,236]. Most C. perfringens strains produce all three sialidases. However, some C. perfringens strains produce only one or two of them. For strains that produce all three, NanI is typically responsible for 70% of total exosialidase activity [236]. When present, NanI enhances the colonization of C. perfringens in the intestinal tract [237] and increases the cytotoxic activity and interaction of several toxins with host cells [232].
The role of sialidases in histotoxic infections remains unclear [233]. Sialidases could enhance C. perfringens adherence to host cells through both nonspecific and specific mechanisms [233]. The nonspecific mechanisms include disruptions of physiologic host cell interactions. Terminal sialic acids promote endothelial barrier integrity and cellular interactions in epithelial monolayers [238]. Sialidases mediate the removal of terminal sialic groups, leading to barrier disruption and increasing access for C. perfringens [238,239]. Therefore, nonspecific effects of secreted NanI on both surface charges and epithelial barrier integrity could help to increase toxin binding and C. perfringens colonization [238].
The specific mechanisms of sialidase-mediated enhancement of C. perfringens infectivity include modifications to host cell surface receptors. Some strains, such as C. perfringens CN3718, display significantly enhanced adhesion to specific mammalian cells, including Caco-2 and HT-29 intestinal cell lines [240]. Similarly, toxins involved in C. perfringens intestinal infections bind to and affect only certain cells. These observations suggest that NanI sialidase modifies host cell surface adhesins and toxin receptors and trims adjacent molecules to enhance the accessibility of the adhesin or toxin receptors [233].
C. perfringens infections pose significant challenges, in spite of antibiotic treatments, due to the persistence of previously synthesized toxins [233,241]. Similarly, altered surface antigens in certain C. perfringens strains make it difficult to prepare comprehensively effective vaccines or neutralizing antibodies [234]. As a result, development of effective sialidase inhibitors represents novel treatment options for C. perfringens infections.

3.1.6. Neisseria gonorrhoeae

Gonorrhea poses a significant global health challenge, due to escalating infection rates, the emergence of antibiotic resistance, and evasion of defensive immune responses [242,243,244]. The exact mechanisms of N. gonorrhoeae-mediated disruptions of host immune responses are not well understood. It is known, however, that N. gonorrhoeae sialyltransferase attaches sialic acids, scavenged from the host, to its own abundant surface lipooligosaccharide [243]. The length and composition of these glycolipids are regulated by genes encoding the lipooligosaccharide glycosyltransferases [245,246]. The expression of these genes constitutes a primary mechanism of bacterial adaptation to challenging environments by altering their surface oligosaccharide composition [233,235]. N. gonorrhoeae, isolated from urethral infection, predominantly produces lipooligosaccharides, which can be sialylated by sialyltransferases for optimal genital tract infection [247,248]. In this context, CMP-sialic acid from the host can be used by N. gonorrhoeae sialyltransferases to attach sialic acid to its own acceptor hydroxyl groups [243]. Many aspects of the N. gonorrhoeae virulence mechanism, and its association with sialic acids, remain to be worked out.

3.2. Sialic Acids and Viral Interactions

The initial step in the viral life cycle is the attachment of virus particles to the cell surface [249]. This adherence is facilitated by the interaction of the virus with a specific receptor [250]. Receptor molecules are integral components of the cell membrane. The receptor determinant, which serves as the binding site for the virus, may consist of either a protein epitope or the carbohydrate molecule of a glycoprotein or glycolipid [160]. Soluble proteins in body fluids and mucus on respiratory and enteric epithelia may also contain carbohydrates and interfere with virus binding to the cell surface [251]. Attachment is attained by the virus binding to a cell surface receptor, usually via electrostatic interactions. Sometimes, co-receptors are also involved, which might promote post-attachment events in the entry process. Early contact between the viral attachment protein and a receptor is weak and reversible. However, as multiple viral attachment proteins interact with multiple receptor molecules, the binding becomes stronger and irreversible [252]. Thus, cells with a higher density or a higher number of receptors are more readily infected [253].

3.2.1. Influenza Virus

As members of Orthomyxoviridae, influenza viruses contain segmented, single-stranded, negative-sense RNAs packaged as ribonucleoproteins in enveloped virions [254,255]. Based on the antigenic diversity of the virion proteins, such as the matrix protein and the nucleocapsid protein, influenza viruses are further classified into three genera: A, B, and C [256,257,258]. Additional classification is based on the haemagglutinin and sialidase glycoproteins expressed on the viral envelope of influenza A and B viruses [258]. Avian influenza A viruses exhibit a strong affinity for α2,3-linked sialic acid as a receptor [258]. As a result, avian viruses tend to bind to Neu5Ac when linked to galactose by an α2,3 bond, while human viruses bind largely to Neu5Ac, linked to galactose via an α2,6 bond [259,260,261].
Usually characterized by annual seasonal epidemics, sporadic pandemic outbreaks involve influenza A virus strains of zoonotic origin [262]. The World Health Organization estimates that annual epidemics of influenza result in approximately 1 billion infections, 3–5 million cases of severe illness, and 300,000–500,000 fatalities [263]. Approximately 10% of the global population is affected by influenza annually, resulting in approximately half a million fatalities annually [264].
Influenza viruses enter cells via endocytosis, by both clathrin-independent and clathrin-dependent pathways [265]. The viral phospholipid bilayer, originating from the host membrane, includes hemagglutinin, which is expressed in the form of spikes on the virion surface [266,267]. Through the clathrin-dependent endocytosis pathway, hemagglutinin binds to sialic acids of host cell surface glycoproteins or glycolipids and mediates virus–host membrane fusion [266,268,269,270]. However, alternative endocytic pathways, such as macropinocytosis, may also be employed [265,271,272].
Furthermore, sialidases are also inserted into the viral phospholipid bilayer as spikes [266,267]. These sialidases are of crucial importance during viral invasion of the ciliated epithelium in human airways [273], specifically, in the removal of decoy receptors on mucins, cilia, and cellular glycocalyx [266,274]. Moreover, sialidase contributes to the pathogenicity of influenza through several additional mechanisms, all which stem from its ability to cleave the α-ketosidic linkage between the terminal sialic acid and the adjacent sugar residue [275]. Notably, influenza sialidase facilitates the release of sialic acid, which can serve as a carbon source for bacteria [273], explaining the frequent superinfection by bacteria following influenza viral infection [276,277].
It is anticipated that influenza vaccines will be introduced in the near future that simultaneously target both the viral hemagglutinin and the viral sialidase [278].

3.2.2. SARS-CoV-2

In response to the rapid global spread of Coronavirus disease 2019 (COVID-19), the World Health Organization (WHO) declared a public health emergency of international interest on 30 January 2020 [279]. This infectious disease is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has resulted in many deaths worldwide. By October 31, 2020, over 45 million confirmed COVID-19 cases and 1.2 million deaths had been reported globally [280]. While all coronaviruses infect the respiratory tract, SARS-CoV-2 also damages other organs, like the heart, gastrointestinal tract, liver, kidneys, and central nervous system. This can potentially lead to multi-organ failure [281,282].
The SARS-CoV-2 virus particle is a spherical, enveloped RNA virus containing a single-stranded genome. The viral envelope has an average diameter between 65 and 97 nanometers [281]. Like other coronaviruses, SARS-CoV-2 contains spike S protein trimers protruding from its envelope. The S protein is made up of 1273 amino acids comprising two subunits, S1 and S2. Binding of the S1 subunit to host cell receptors mediates viral entry and infection. The S1 subunit contains two distinct domains: an N-terminal domain and a C-terminal domain [283,284]. The glycan-binding domain located in the N-terminal domain interacts with glycolipids and glycoproteins on the host cell surface [285]. The C-terminal domain contains the receptor-binding domain that attaches to the ACE2 receptor, the primary entry point for SARS-CoV-2 and other coronaviruses [286].
Some coronaviruses are known to bind sialic acids, and SARS-CoV infection might also involve sialic acid interactions [287,288]. Computational predictions, modeling studies, and microscopic analyses indicate that SARS-CoV-2 may bind to sialic acids [289,290]. This binding capacity may explain the extremely high transmissibility of SARS-CoV-2, compared with other human coronaviruses [287,291]. Although gold nanoparticles conjugated with sialic acids have high avidity for the SARS-CoV-2 S1 protein (containing both the N-terminal domain and receptor-binding domain), their binding to model sialylated molecules appears to be relatively weak [292,293]. While numerous reports and reviews [294,295] discuss the potential role of sialic acids as co-receptors for SARS-CoV-2, the specific sialyl structure that is targeted by the spike protein remains unclear [296]. The receptor-binding domain of the SARS-CoV-2 spike protein is known to bind oligosaccharides containing sialic acids. Nonetheless, the preferential interactions seem to be with lipids that enable the virus to enter host cells [293]. Additionally, direct binding between sialic acid-containing oligosaccharides (α2,3 and α2,6 sialyl N-acetyllactosamine) and the N-terminal domain of the SARS-CoV-2 spike glycoprotein has been verified through saturation transfer difference nuclear magnetic resonance spectroscopy [297]. However, the relevance of this interaction for cellular entry still needs to be confirmed. More work is needed for an improved understanding of the infection mechanism and spread of SARS-CoV-2.

3.3. Sialic Acids and Fungi

Fungi are effective degraders of dead organic matter [298] and may impact the human ecosystem [20,299]. Occasionally, fungi may become pathogenic, leading to the development of disease, exploiting the host living tissue for survival [300]. Before that can happen, however, fungal cells must somehow withstand the human immune system defenses. Hence, fungal infections tend to occur in immunocompromised individuals. Despite this, fungi with polysaccharide capsules tend to be more infectious.

3.3.1. Cryptococcus neoformans

Cryptococcus neoformans is used as a model organism illustrating how sialic acids might contribute to the evasion of the human immune response [20,301]. This pathogenic fungus has a true polysaccharide capsule on the outside of its cell wall [302], serving as its major virulence factor [301,303]. Protection from the immune system is aided by sialic acids, and their 9-O-acetylated derivatives, which are a part of the fungal capsular polysaccharides [20,304]. These sialic acids appear to reduce the likelihood of phagocytosis by alveolar macrophages during the initial stages of infection, as the basidiospores are inhaled and gain entry to alveolar spaces [302,305].

3.3.2. Histoplasma capsulatum

Infection with Histoplasma capsulatum results in substantial global morbidity and mortality [306]. As a dimorphic ascomycete, this fungus grows in its hyphal form in the soil and bird and bat droppings [307]. Upon entry into the host, in the form of microconidia or hyphal fragments, H. capsulatum travels through the respiratory system inside phagocytes until it reaches the alveoli [308]. There, the yeast, inside or outside of host phagocytes, is transformed into the pathogenic phase, producing histoplasmosis [296,297]. While invading the host respiratory system, the yeast must evade immune-mediated and intracellular defenses [306]. Meanwhile, these yeast cells need to survive or evade the hostile anti-microbial environment within phagocytes [309,310], including potential exposure to reactive oxygen, such as hydrogen peroxide, and reactive nitrogen species [309,311]. Yeast cells actively inhibit phago-lysosomal fusion, thereby preventing exposure to the acidic hydrolytic enzymes of the lysosomes [308,312]. Moreover, H. capsulatum impedes the accumulation of vacuolar ATPase, which is ordinarily needed for proton accumulation in phagosomes, allowing the fungus to alkalinize the phagosomal pH to 6.5 [309,311,312]. If a suitable environment for growth and reproduction is located, the yeast may also disseminate and develop granulomas [313]. Subsequently, the pathogen is disseminated to the lymph nodes, and then to various organs, by phagocytes as its carriers [306,308,314].
Interactions between carbohydrates and lectins are considered to be pivotal to the recognition of target particles by phagocytes [311,315]. The expression of lectins by pathogenic microorganisms has been correlated to the organisms’ attachment and invasion of host tissues [311,316,317]. H. capsulatum expresses a 50 kDa lectin that recognizes sialic acid residues on laminin, a key protein of the basement membrane, potentially mediating surface interactions [318,319,320].
The identification and isolation of sialic acid-specific lectins from pathogenic fungi, particularly airborne species that cause severe infections, is highly desirable. It represents an opportunity to intervene at the initial stages of infection, as the microconidia interact with lung epithelial cells. If this initial stage is dependent on specific lectin structures, then several options for diagnosis and therapy could become available [320].

3.3.3. Candida albicans

Candida albicans commonly colonizes the skin, the oropharyngeal cavity, the gastrointestinal system, and the vaginal tract [321]. Infections by Candida species are widespread and are increasing in frequency [321,322,323], particularly during infancy or in old age [324,325]. A diverse array of conditions, encompassing environmental, local, and systemic factors, as well as hereditary predispositions, may alter Candida’s normally dormant state [321,325,326]. The signs and symptoms may correspond with a range of morbidities, from localized superficial mucocutaneous disorders to invasive diseases involving multiple organ systems [309,315]. The transition in pathophysiology from the onset through the progression of infection is also influenced by the particular virulence traits that ultimately lead to the development of candidiasis [321,327].
Three types of monosaccharides form carbohydrate chains, or glycans, within the Candida cell wall: d-glucose (Glc), N-acetyl-d-glucosamine (GlcNAc), and d-mannose (Man) [328]. Several reports document sialic acids as constituents of the Candida cell wall [20,304,329]. Sialic acids contribute to the negative charge of fungal cells and have a role in their specific interaction with the host tissue, including a potential role in pathogenicity [329]. Although several microorganisms infecting animals or humans produce sialidases for adhesion or invasion, C. albicans does not, implying that fungal sialidase is likely of no relevance in cutaneous or mucosal candidiasis [330]. Consequently, this implies that sialic acids are likely acquired by C. albicans from the host environment via sialyltransferases. Further investigations are needed into the role of surface sialylation of C. albicans, particularly as it relates to the infectivity of this organism.

3.4. Sialic Acids and Parasites

Parasites are significant contributors to human disease worldwide, particularly in less-developed nations. The clinical severity and outcomes of parasitic diseases are often strongly affected by the host’s immune status [331]. For example, helminths tend to elicit a protective helper T-lymphocyte type 2 host response, with production of IgE, eosinophilia, and mastocytosis [332], whereas protozoa induce a humoral and/or cellular immune response that is not usually associated with eosinophilia [333]. To overcome the immune defenses, parasites employ a variety of mechanisms. These may include (a) becoming “invisible” to the immune system by invading immune-privileged organs or cells, such as the central nervous system, eyes, or blood cells, or (b) decorating surface glycoconjugates with sialic acids, which may be directly synthesized or appropriated from the host [334]. Consequently, the T-cells and antibodies of the vertebrate’s immune system either cannot access the parasite, or do not easily recognize the parasite’s specific antigenic surfaces [335] for effective detection, attack, and subsequent elimination. Such evasive mechanisms are crucial for the parasite’s survival inside the host [159].

3.4.1. Trypanosoma cruzi

Trypanosoma cruzi is a protozoan parasite that causes Chagas disease [336]. In Latin America, nearly 30% of chronically infected people with Chagas disease manifest an intense systemic response that is frequently associated with damage to essential organs, such as the heart or the gastrointestinal tract. This suggests that a wide range of host–parasite interactions affect the course of the infection [163,337].
The protozoan parasite is carried by a triatomine insect [338]. After the infected insect has a blood meal, T. cruzi metacyclic trypomastigotes are discharged via the insect’s feces or urine. Then, the trypomastigotes can infect the mammalian host by penetrating the mucosa, or via discontinuous regions in the epithelium. Once inside the host, the protozoan parasite rapidly infects a variety of nucleated mammalian cells. The attachment to and invasion of host cells, and the formation of the parasitophorous vacuoles with T. cruzi, involves a collection of polymorphic glycosylphosphatidylinositol-anchored surface proteins, including mucins and trans-sialidases [339]. Using its arsenal, the parasite destabilizes both the host’s innate and adaptive immunity [340,341]. Though unable to synthesize sialic acids de novo, T. cruzi uses a unique trans-sialidase enzyme that cleaves terminal sialic acid residues from host cell surface glycoconjugates and transfers them onto parasite surface mucins, generating a protective/adhesive covering [342]. The sialylation of parasite-derived mucins has inhibitory effects on CD4 T cells [341], inducing suppression of their responses. Additionally, T. cruzi sheds trans-sialidase into circulation, as a mechanism of modifying the host’s surface sialic acid signature, exploiting the signaling and functional properties of mammalian host target cells [22]. After lysosomes merge with parasitophorous vacuoles, a complex network of antioxidant enzymes, including superoxide dismutase, shield the microorganism from reactive oxygen and nitrogen species [343]. The acidification associated with the lysosomal contents activates certain critical steps that allow the parasite to escape from the phagosome to the cytoplasm, where it differentiates into the replicative amastigote form. Following numerous duplications and rupture of the host cell membrane, the amastigotes differentiate into infective trypomastigotes that are capable of infecting adjacent cells and being transported by them to distant areas via circulation [343,344]. Based on screening studies, ciprofloxacin binds trans-sialidases, which are essential for T. cruzi survival [345]. This suggests that the antiparasitic effects of quinolones are, at least in part, caused by trans-sialidase inhibition [346].

3.4.2. Plasmodium spp.

Malaria can be caused by several Plasmodium species [347]. According to the most recent World Malaria Report, in 2023, there were 263 million cases, a slight increase from 252 million cases the year before. In 2023, malaria-related deaths were estimated to reach 597,000 [348]. The escalation in malaria cases is primarily attributed to (a) the developing resistance of the parasite to antimalarial medications, and (b) the rapid population growth in the most severely affected regions [347,349,350].
Infection is initiated during a blood meal by an infected Anopheles mosquito, when, along with anticoagulant saliva, 10–100 sporozoites are injected into the host circulation [351,352]. The parasites invade hepatocytes in the liver [353], where they undergo their first replication cycle, producing thousands of merozoites. These merozoites are released into the bloodstream [354], where they infect erythrocytes. The parasite encases itself within a parasitophorous vacuolar membrane and commences remodeling the host cell [355]. These processes entail both internal and external alterations that facilitate the parasite’s survival and proliferation within the red blood cell [356]. During the blood stage, the parasites replicate asexually, maturing into schizonts that undergo multiple rounds of nuclear division to produce merozoites [354,357]. Since red blood cells are not targeted by cytotoxic cells, the parasite is protected from the immune system [354,358]. While digesting host hemoglobin and replicating within the red blood cell, the parasite undergoes expansion and modifies the red blood cell membrane [162,354,358]. Red blood cells infected by P. falciparum become more rigid, whereas those infected by P. vivax become more flexible [352]. At this stage, a fraction of parasites differentiate into sexually competent gametocytes, which can then be transmitted via the mosquito vector [359].
The malaria pathogen has a relatively high selectivity for its respective host [360]. Of the five Plasmodium species that infect humans, P. falciparum and P. vivax are by far the most common and most lethal [361]. Both rely on sialic acids for the establishment of infection, the invasion of red blood cells, and the evasion of the immune system [354,357,362,363]. Reticulocyte invasion by P. vivax, however, is primarily dependent on the reticulocyte Duffy antigen/receptor for chemokines (DARC) [364]. P. falciparum, on the other hand, employs a more direct and critical dependence on erythrocyte sialic acid residues, mediated in part by two distinct sialic acid-binding protein ligands, expressed on the parasite surface and used during red blood cell invasion. These are (a) cysteine-rich protective antigen (PfCyRPA) [365], and (b) antigen 175 (EBA-175) [366,367]. The merozoite surface receptor EBA175 binds to the sialic acid residues of erythrocyte glycophorin A [162,362]. The binding activity of EBA175 is primarily restricted to Region II of the protein. Consequently, this domain is thought to present a potential target for the development of a malaria vaccine [368]. The expression of different forms of EBA and Rh proteins in field parasite isolates, including EBA175, EBA140, EBA181, Rh1, and Rh2, makes such efforts more challenging. Therefore, targeting EBA175 alone by a vaccine against P. falciparum is likely to be insufficient. Developing a multivalent vaccine, incorporating several key blood-stage proteins, should be more promising [368].

4. Inflammatory Processes

Several pathological conditions are characteristically associated with structural and functional changes in sialic acid expression [77]. Such alterations in sialylated glycans can readily be detected in histological sections using plant lectins or antiglycan antibodies [369]. Increased levels of both total and free sialic acid forms in almost any severe condition or inflammatory process are consistent with their potential role as markers of inflammatory disorders [49,56,370,371]. While the rationale supporting the prognostic value of sialic acids in pathological conditions is unclear, some potential explanations have been proposed. These include (a) activation of the hepatocyte-based synthesis and secretion of acute-phase proteins, including several sialoglycoproteins, (b) impaired membrane integrity due to cell damage, and (c) upregulated activities of sialidases and sialyltransferases [371]. These circumstances could lead to measurable changes in sialic acid levels or sialylation patterns, correlating with disease progression.

4.1. Cardiovascular Disease

Cardiovascular disease (CVD), with its behavioral, environmental, cardiometabolic, and social risk factors, represents the leading cause of global morbidity and premature mortality in Western developed countries [372]. Some of the risk factors that could be modified to help prevent or reduce CVD include a lack of adequate physical activity, a sedentary lifestyle, hyperlipidemia, obesity, hypertension, diabetes, and tobacco smoking [373].
Arterial walls comprise several cell types and a specialized extracellular matrix, all of which provide adjustable vascular elasticity and strength [374]. Vascular luminal surfaces are lined with a layer of endothelial cells, which themselves are covered with a dense, heavily sialylated glycocalyx [375]. These sialylated glycoconjugates participate in the regulation of cell–cell interactions for (a) cellular junctions between endothelial cells, and (b) contact between endothelial and circulating blood cells. They also play a role as vascular receptors that modulate the binding of soluble ligands and their signaling [376]. Additionally, sialic acids provide net negative surface charges to vascular endothelial cells and low-density lipoprotein (LDL) receptors. Thus, sialylation, or the covalent addition of sialic acids to the termini of glycoconjugates, and desialylation, or the removal of sialic acid from glycoconjugates, represent a pivotal part of sialic acid metabolism in a variety of physiological and pathological processes [89,377].
Since sialic acids coat all cell surfaces, many circulatory molecules and most molecular complexes, their alteration could have varied effects, potentially including a role in the pathogenesis of CVD [26,148,378]. Desialylation, for example, is recognized as one of the most important steps in the pro-atherogenic pathological cascade [379]. The deposition of cholesterol in cultured human aortic intimal cells is significantly increased when LDLs are treated with sialidase before addition to the culture [379]. The diminished sialic acid content of LDLs leads to increased formation of aggregates, which are readily taken up by smooth muscle cells. Moreover, some free sialic acids, such as the nonhuman Neu5Gc, could also be potential risk factors for CVD [380], or serve as biomarkers for it, as does Neu5Ac [381,382,383]. Neu5Gc accumulates on endothelial surfaces and in atherosclerotic plaques, where anti-Neu5Gc antibodies can trigger endothelial activation and induction of vascular inflammation [384]. Despite such in vitro studies, however, the clinical correlation between serum sialic acids, anti-Neu5Gc antibodies, and their relationship to CVD remains unclear (Table 1).
Comparing the content of sialic acids in the adipose tissue among individuals following habitually vegetarian or non-vegetarian dietary patterns, significantly higher levels of Neu5Ac were found in vegans and lacto-ovo-vegetarians relative to non-vegetarians [39]. These findings could be comparable with other studies reporting intrinsically protective cardiometabolic effects of Neu5Ac or ManNAc supplementation [380,385,386]. Despite this, the beneficial effects of dietary Neu5Ac in vegetarian subjects have not yet been established. However, the positive effects are frequently inferred, given the low risk of cardiovascular disease mortality among these individuals [387].
An excessive accumulation of abdominal adipose tissue is strongly associated with a prognosis of atherosclerosis [388]. Interestingly, it seems that diet could also indirectly affect sialic acids through alterations in BMI or adiposity. For example, KDN levels are lower with increasing BMI, independent of diet, even when comparing individuals following habitually vegetarian or non-vegetarian dietary patterns [39]. These findings are consistent with previous reports of lower sialic acids in obese individuals [386], suggesting that obesity is associated with a higher inflammatory tone. Moreover, in contrast to non-vegetarians, vegetarians have been reported to have a lower BMI and smaller waist circumference [389], which are risk factors for metabolic syndrome, diabetes [390], and cardiovascular disease [387,391]. Therefore, the effects of diet on sialic acids could be related, at least in part, to adiposity. Either way, a plant-based diet promotes a lower BMI and an anti-inflammatory state, both of which appear to be associated with higher sialic acids in adipose tissue. The sialic acids, in turn, could help to regulate or prevent the development of cardiovascular and other chronic diseases. Nevertheless, more detailed epidemiological and clinical studies are necessary to better elucidate the relationships between diet, BMI, and sialic acid levels.

4.2. Cancer

The immune system can be directed in various ways to identify and attack tumors [392]. Sialic acids shield host surfaces by resisting inappropriate immune reactions against self-antigens. This effectively suppresses the immune response. In this context, glycans, including terminal sialic acids, represent self-associated molecular patterns (SAMPs), which are recognized by self-pattern recognition receptors [393]. As a result, sialic acids play a pivotal role in modulating immune reactivity by serving as a self-recognition signal in several ways, including (a) via complement interactions, (b) via alteration of antibody-mediated clearance of pathogens, or (c) via sialic acid-binding immunoglobulin-like lectins (Siglecs) [394]. Consequently, strategies that disrupt sialic acid-mediated immune evasion, such as targeting Siglec interactions or modulating sialoglycan expression, hold promise for enhancing immune recognition and improving cancer therapies.
Table 1. Sialic acids and cardiometabolic diseases.
Table 1. Sialic acids and cardiometabolic diseases.
TitleApproachYearFindingsReference
AnimalHuman
Relation of serum sialic acid to blood coagulation activity in type 2 diabetes x2002Serum sialic acid level is positive correlated with (a) blood coagulation activity and (b) circulatory fibrinogen levels.[147]
High fat diet-induced inflammation and oxidative stress are attenuated by N-acetylneuraminic acid in ratsx 2005Neu5Ac could be useful for preventing inflammation and oxidative stress associated with hyperlipidemia.[395]
Relationship between Sialic acid and metabolic variables in Indian type 2 diabetic patients x2005Among Indian patients with type 2 diabetes, elevated levels of serum and urinary sialic acid, as well as microalbumin, are strongly associated with microvascular complications.[396]
Percentage of body fat and plasma glucose predict plasma sialic acid concentration in type 2 diabetes mellitus x2006Percentage of body fat correlates with plasma sialic acid levels and contributes to elevated sialic acid concentrations in patients with type 2 diabetes mellitus.[146]
Sialic acid and oxidizability of lipid and proteins and antioxidant status in patients with coronary artery disease x2007Patients with CAD show significant increases in total sialic acid levels and markers of oxidative stress. Furthermore, higher TSA levels correlate with greater CAD severity.[370]
N-Acetylneuraminic Acid Supplementation Prevents High Fat Diet-Induced Insulin Resistance in Rats through Transcriptional and Nontranscriptional Mechanismsx 2015Administering a low dose of sialic acids prevents insulin resistance in rats fed a high-fat diet.[397]
N-Acetylneuraminic acid attenuates hypercoagulation on high fat diet-induced hyperlipidemic ratsx 2015Data show that Neu5Ac prevents high-fat diet-induced high blood lipid levels and associated increased blood clotting in rats.[385]
Sialidase downregulation reduces non-HDL cholesterol, inhibits leukocyte transmigration, and attenuates atherosclerosis in ApoE knockout micex 2018Decreasing Neu1 expression or function reduces atherosclerosis in mice by substantially impacting lipid metabolism and inflammation.[398]
Supplementation with the Sialic Acid Precursor N-Acetyl-D-Mannosamine Breaks the Link between Obesity and Hypertensionxx2019Interventions targeting hyposialylated IgG and FcγRIIB, such as ManNAc supplementation, could potentially break link between obesity and hypertension and provide novel therapeutic approaches.[386]
Neuraminidases 1 and 3 trigger atherosclerosis by desialylating low-density lipoproteins and increasing their uptake by macrophagesxx2021Neuraminidases 1 and 3 initiate atherosclerosis and formation of aortic fatty streaks.[149]
Sialic acid metabolism as a potential therapeutic target of atherosclerosis Sialic acids Neu5Ac and KDN in adipose tissue samples from individuals following habitual vegetarian or non-vegetarian dietary patterns x2023Concentrations of Neu5Ac are significantly higher in vegans and lacto-ovo-vegetarians compared to non-vegetarians. Significant inverse association observed between KDN levels and body mass index.[39]
CAD, coronary artery disease; FcγRIIB, Fc gamma receptor IIB; KDN, 2-keto-3-deoxy-D-glycero-D-galacto-nononic acid; ManNAc, N-acetylmannosamine; Neu1, neuraminidase 1; Neu5Ac, N-acetylneuraminic acid; TSA, total sialic acids; x, report is based on animal study, human study or both, as indicated.
Lectins represent a family of immunomodulatory receptors located on the surfaces of granulocytes, monocytes, antigen-presenting cells, and natural killer cells. Commonly, Siglecs contain an immunoreceptor tyrosine-based inhibitory motif (ITIM) within the cytoplasmic domain [152,399,400]. A subset of these, Siglecs 14–16, do not possess an ITIM, but instead contain within their transmembrane region a lysine residue, which facilitates interactions with immunoreceptor tyrosine-based activatory motif (ITAM)-containing adaptor proteins [401].
The role of the immune system is unclear during tumor formation and progression, or in the development of resistance to and eradication of emerging neoplasias, late-stage tumors, and micrometastases [402]. The theory of immune surveillance holds that cells and tissues are constantly scanned by the immune system, and that such ever-alert immune surveillance is responsible for recognizing and eliminating most early cancer cells and nascent tumors [403]. The detection of abnormal cells may occur when aberrant surface expression of the sialylated glycans SLeA, SLeX, STn, or GM2 takes place (Figure 6) [91,399,404,405,406]. This can be a result of upregulated activity of sialyltransferases, leading to various surface sialic acids (Table 2) [92,407]. However, once Siglecs are bound to sialylated glycans on cancer cells, immunosuppressive signaling may also be promoted [408,409,410]. For instance, natural killer (NK) cell-mediated tumor cell death is inhibited by interactions between NK-expressed Siglec-7 or Siglec-9, and sialylated glycans, which serve as Siglec ligands on tumor cells [268].
Through a process of immunoediting [403], tumor cells are selected that mimic the glycosylation patterns of healthy cells using “self”-surface-sialylated glycan signals, as part of evasive strategies [416,417]. This mechanism might represent a potential immune checkpoint for cancer, since immune cells make aberrant sialoglycans prime ligands for Siglecs [418,419]. Thus, it is feasible to target the sialoglycan–Siglec glyco-immune checkpoint by employing Siglec-blocking antibodies. An alternative approach could be to reduce the ligand density by targeting sialoglycans [420]. It has been demonstrated, for example, that when the glyco-profile of a cancer cell is altered by hypersialylation (an increment in sialic acid residues of up to 40–60%) [416], occurring either by the upregulation of sialyltransferases, the downregulation of neuraminidases, or a combination of both, the tumor becomes more aggressive, leading to metastasis and chemoresistance [6,416]. This suggests that downregulation of the surface sialylation of cancer cells may render them less aggressive and more accessible to the immune system.

Colorectal Cancer

Colorectal cancer (CRC) is among the most common cancers and is associated with one of the highest worldwide mortality rates [421]. It is the third most commonly diagnosed cancer among men, and the second most common cancer among women [422]. Numerous studies report associations between dietary factors and total cancer risk [387,423,424,425,426,427]. Diet and nutrition are estimated to explain as much as 30–50% of the worldwide incidence of CRC [428]. Dietary approaches to cancer prevention are of high interest [387], because of their impact as risk modifiers within the multistep process of colorectal tumorigenesis [428]. In this context, vegetarian diets are associated with an overall lower incidence of colorectal cancer [387]. Conversely, consumption of red and processed meats is associated with an increased risk of colon and rectal cancers [429,430,431,432].
Multiple mechanisms explain the potential cancer-promoting effects associated with the consumption of red or processed meats. These include (a) DNA damage due to toxic and mutagenic effects of N-nitroso compounds produced during high-temperature grilling [433], (b) high dietary intake of salt and saturated fats [434], (c) pro-oxidant effects of heme and iron [435], (d) production of trimethylamine (TMA) by the gut microbiome, which is subsequently oxidized by hepatic enzymes to form trimethylamine N-oxide (TMAO) [150,436], (e) altered human gut microbiome composition [153], and (f) increased levels of contaminating cancer-causing viruses [437].
The hypothesis of xenosialitis as an aspect of chronic inflammation is being increasingly discussed [50]. The human body synthetizes its own sialic acids, in addition to exogenously obtained dietary sugars [55,72]. Such dietary sources can include Neu5Gc, especially from foods like red meat, milk, or dairy products [120,438,439]. It has been suggested that Neu5Gc could be a potential aggravating risk factor for carcinoma [436]. The proposed likely mechanism is based on mouse model studies [122,131,432]. The two principal contributors are (a) the fact that humans have a mutation in the gene CMAH, encoding cytidine monophosphate-N-acetylneuraminic acid hydroxylase (Cmah), which converts Neu5Ac to Neu5Gc [122,125,131,440], and (b) the fact that humans can metabolize dietary Neu5Gc and incorporate it during the formation of native human glycans in the place of the usually employed Neu5Ac [131,441]. Thus, multiple instances of potentially immunogenic Neu5Gc-containing glycan neoantigens can be expressed on the colon’s epithelial surfaces [131]. As a result, two different mechanisms may be activated: (a) the canonical NF-κB signaling pathway, which regulates the transcription of inflammatory factors, and (b) the in situ formation of antigen–antibody immunocomplexes [122]. Both processes promote systemic inflammation and subsequent tissue damage [442]. Consequently, each of these could explain the link between a red meat-rich diet and colon carcinoma.
This xenosialitis hypothesis was tested, with some mixed results. Since polyclonal rabbit anti-T cell IgG (ATG) is known to elicit an immune response in humans toward Neu5Gc [443], allograft recipients treated with animal-derived IgG were tested. Yet, in one study, no evidence was found that exposure to higher anti-Neu5Gc levels is associated with a higher incidence of colon carcinoma [53], possibly due to low cohort numbers. In contrast, in another study of 71 subjects, a positive association between total anti-Neu5Gc and CRC risk was reported [150]. However, a significant association between anti-Neu5Gc IgG and dietary red meat intake has not yet been demonstrated [150]. Interestingly, a different study reported a clear link between the levels and repertoire of serum anti-Neu5Gc IgG and the Neu5Gc from dietary red meat and dairy consumption [131]. Further, adequately powered investigations are needed to elucidate the links between diet, Neu5Gc, anti-Neu5Gc, and the risks of chronic diseases.

5. Conclusions

Sialic acids are versatile nine-carbon carbohydrates that play essential roles in human health and disease. Given their abundance on cell surfaces and secreted proteins, they mediate critical processes, such as facilitating cell–cell interactions, signaling mechanisms, immune responses, and developmental processes. Pathogens frequently exploit sialic acids to mimic host surfaces and evade immune responses. Conversely, changes in host sialylation patterns are known to be associated with inflammation, cardiovascular diseases, and cancers. Advances in analytical techniques, particularly mass spectrometry, have expanded understanding of sialic acid biology. Further investigations are needed to elucidate the diverse roles of sialic acids in physiologic and pathologic processes, in order to enable targeted therapeutic interventions.

Author Contributions

Conceptualization, D.S.B., G.N.G.-F. and F.J.P.; methodology, D.S.B. and G.N.G.-F.; writing original draft and preparation, D.S.B., G.N.G.-F. and F.J.P.; writing review and editing, D.S.B., G.N.G.-F., F.J.P., F.M.B., G.Z. and V.L.M.M.; visualization, G.N.G.-F. and D.S.B.; supervision, D.S.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by: (a) Grant for Research and School Partnerships (GRASP), Loma Linda University (#2170258), California, USA; (b) Doctoral scholarship to GNGF, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ministry of Science and Technology, Argentina (RESOL-2018-2704-APN-DIR#CONICET); (c) Research grant, School of Medicine, Adventist University of River Plate, Entre Rios, Argentina.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADPadenosine diphosphate
ATPadenosine triphosphate
CMASCMP-N-acetylneuraminate synthetase
Cmahcytidine monophosphate-N-acetylneuraminic acid hydroxylase; CMP
CMPcytidine monophosphate
CTPcytidine triphosphate
GalNAcN-acetylgalactosamine
GlcNAcN-acetylglucosamine
GNEUDP-GlcNAc 2-epimerase/ManNAc kinase
GD2disialoganglioside
GM2ganglioside-mono sialic acid 2
GlmMphosphoglucosamine mutase
GlmSglucosamine-6-phosphate synthase
GlmUglucosamine-1-phosphate acetyltransferase/N-acetylglucosamine-1-phosphate uridyltransferase
HBP hexosamine biosynthesis pathway
ITAMimmunoreceptor tyrosine-based activatory motif
ITIMimmunoreceptor tyrosine-based inhibitory motif
KDN2-keto-3-deoxy-D-glycero-D-galacto-nononic acid
LC-MS/MSliquid chromatography with tandem mass spectrometry
Manmannose
ManNAcN-acetylmannosamine
MSmass spectrometry
NANPN-acetylneuraminic acid phosphatase
NANSN-acetylneuraminic acid 9-phosphate synthase
Neuneuraminic acid
Neu5AcN-acetylneuraminic acid
Neu5GcN-glycolylneuraminic acid
NeuAsialic acid synthase A
NeuBsialic acid synthase B
NeuCsialic acid synthase C
NulOnonulosonic acid
PEPphosphoenolpyruvate
PGIglucose-6-phosphate isomerase
ROSreactive oxygen species
SLeAsialyl-Lewis A
SLeXsialyl-Lewis X
STnsialyl-Tn
SCFAsshort-chain fatty acids
UDPuridine diphosphate
SARS-CoV-2severe-acute-respiratory-syndrome-related coronavirus 2.

References

  1. Tarbell, J.M.; Cancel, L.M. The glycocalyx and its significance in human medicine. J. Intern. Med. 2016, 280, 97–113. [Google Scholar] [CrossRef]
  2. Lucas, T.M.; Gupta, C.; Altman, M.O.; Sanchez, E.; Naticchia, M.R.; Gagneux, P.; Singharoy, A.; Godula, K. Mucin-mimetic glycan arrays integrating machine learning for analyzing receptor pattern recognition by influenza A viruses. Chem 2021, 7, 3393–3411. [Google Scholar] [CrossRef]
  3. Ebong, E.E.; Macaluso, F.P.; Spray, D.C.; Tarbell, J.M. Imaging the Endothelial Glycocalyx In Vitro by Rapid Freezing/Freeze Substitution Transmission Electron Microscopy. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 1908–1915. [Google Scholar] [CrossRef] [PubMed]
  4. Du, J.; Zhang, Q.; Li, J.; Zheng, Q. LC-MS in combination with DMBA derivatization for sialic acid speciation and distribution analysis in fish tissues. Anal. Methods 2020, 12, 2221–2227. [Google Scholar] [CrossRef]
  5. Möckl, L.; Pedram, K.; Roy, A.R.; Krishnan, V.; Gustavsson, A.-K.; Dorigo, O.; Bertozzi, C.R.; Moerner, W.E. Quantitative Super-Resolution Microscopy of the Mammalian Glycocalyx. Dev. Cell 2019, 50, 57–72.e6. [Google Scholar] [CrossRef] [PubMed]
  6. Lewis, A.L.; Toukach, P.; Bolton, E.; Chen, X.; Frank, M.; Lütteke, T.; Knirel, Y.; Schoenhofen, I.; Varki, A.; Vinogradov, E.; et al. The SNFG Discussion Group. Cataloging natural sialic acids and other nonulosonic acids (NulOs), and their representation using the Symbol Nomenclature for Glycans. Glycobiology 2023, 33, 99–103. [Google Scholar] [CrossRef]
  7. Kooner, A.S.; Yu, H.; Chen, X. Synthesis of N-glycolylneuraminic acid (Neu5Gc) and its glycosides. Front. Immunol. 2019, 10, 2004. [Google Scholar] [CrossRef]
  8. Lewis, A.L.; Chen, X.; Schnaar, R.L.; Varki, A. Sialic acids and other Nonulosonic Acids. In Essential of Glycobiology, 4th ed.; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, NY, USA, 2022; pp. 316–344. [Google Scholar]
  9. Chen, X.; Varki, A. Advances in the biology and chemistry of sialic acids. ACS Chem. Biol. 2010, 5, 163–176. [Google Scholar] [CrossRef]
  10. Traving, C.; Schauer, R. Structure, function and metabolism of sialic acids. Cell. Mol. Life Sci. 1998, 54, 1330–1349. [Google Scholar] [CrossRef]
  11. Inoue, S.; Lin, S.L.; Chang, T.; Wu, S.H.; Yao, C.W.; Chu, T.Y.; Troy, F.A.; Inoue, Y. Identification of free deaminated sialic acid (2-keto-3-deoxy-D-glycero- D-galacto-nononic acid) in human red blood cells and its elevated expression in fetal cord red blood cells and ovarian cancer cells. J. Biol. Chem. 1998, 273, 27199–27204. [Google Scholar] [CrossRef]
  12. Inoue, S.; Kitajima, K.; Sato, C.; Go, S. Human KDN (Deaminated Neuraminic Acid) and Its Elevated Expression in Cancer Cells: Mechanism and Significance. In The Molecular Immunology of Complex Carbohydrates-3; Wu, A.M., Ed.; Springer: Boston, MA, USA, 2011; Volume 705, pp. 669–678. [Google Scholar]
  13. Schauer, R. Sialic acids: Fascinating sugars in higher animals and man. Zoology 2004, 107, 49–64. [Google Scholar] [CrossRef] [PubMed]
  14. Persat, F.; Bouhours, J.-F.; Petavy, A.-F.; Mojon, M. Gangliosides of Echinococcus multilocularis metacestodes. Biochim. Biophys. Acta BBA—Mol. Basis Dis. 1994, 1225, 297–303. [Google Scholar] [CrossRef]
  15. Yeşilyurt, B.; Şahar, U.; Deveci, R. Determination of the type and quantity of sialic acid in the egg jelly coat of the sea urchin Paracentrotus lividus using capillary LC-ESI-MS/MS. Mol. Reprod. Dev. 2015, 82, 115–122. [Google Scholar] [CrossRef] [PubMed]
  16. Saito, M.; Kitamura, H.; Sugiyama, K. Occurrence of gangliosides in the common squid and pacific octopus among protostomia. Biochim. Biophys. Acta BBA—Biomembr. 2001, 1511, 271–280. [Google Scholar] [CrossRef]
  17. Elayabharathi, T.; Vinoliya Josephine Mary, J.; Mary Mettilda Bai, S. Characterization of a novel O-acetyl sialic acid specific lectin from the hemolymph of the marine crab, Atergatis integerrimus (Lamarck, 1818). Fish Shellfish Immunol. 2020, 106, 1131–1138. [Google Scholar] [CrossRef] [PubMed]
  18. Guerardel, Y.; Chang, L.-Y.; Fujita, A.; Coddeville, B.; Maes, E.; Sato, C.; Harduin-Lepers, A.; Kubokawa, K.; Kitajima, K. Sialome analysis of the cephalochordate Branchiostoma belcheri, a key organism for vertebrate evolution. Glycobiology 2012, 22, 479–491. [Google Scholar] [CrossRef]
  19. Varki, A. Glycan-based interactions involving vertebrate sialic-acid-recognizing proteins. Nature 2007, 446, 1023–1029. [Google Scholar] [CrossRef]
  20. Eneva, R.; Engibarov, S.; Abrashev, R.; Krumova, E.; Angelova, M. Sialic acids, sialoconjugates and enzymes of their metabolism in fungi. Biotechnol. Biotechnol. Equip. 2021, 35, 346–357. [Google Scholar] [CrossRef]
  21. Ghosh, S. Sialic acid and biology of life: An introduction. In Sialic Acids and Sialoglycoconjugates in the Biology of Life, Health and Disease; Elsevier: Amsterdam, The Netherlands, 2020; pp. 1–61. [Google Scholar]
  22. Campetella, O.; Buscaglia, C.A.; Mucci, J.; Leguizamón, M.S. Parasite-host glycan interactions during Trypanosoma cruzi infection: Trans-Sialidase rides the show. Biochim. Biophys. Acta BBA—Mol. Basis Dis. 2020, 1866, 165692. [Google Scholar] [CrossRef]
  23. Iijima, R.; Takahashi, H.; Namme, R.; Ikegami, S.; Yamazaki, M. Novel biological function of sialic acid (N-acetylneuraminic acid) as a hydrogen peroxide scavenger. FEBS Lett. 2004, 561, 163–166. [Google Scholar] [CrossRef]
  24. Yang, H.; Lu, L.; Chen, X. An overview and future prospects of sialic acids. Biotechnol. Adv. 2021, 46, 107678. [Google Scholar] [CrossRef]
  25. Schauer, R. Achievements and challenges of sialic acid research. Glycoconj. J. 2000, 17, 485–499. [Google Scholar] [CrossRef] [PubMed]
  26. Gopaul, K.P.; Crook, M.A. Sialic acid: A novel marker of cardiovascular disease? Clin. Biochem. 2006, 39, 667–681. [Google Scholar] [CrossRef] [PubMed]
  27. Kim, H.J.; Schweiker, S.; Powell, K.; Levonis, S. An efficient and robust HPLC method to determine the sialylation levels of human epithelial cells. PLoS ONE 2022, 17, e0257178. [Google Scholar] [CrossRef]
  28. Szabo, Z.; Bones, J.; Guttman, A.; Glick, J.; Karger, B.L. Sialic acid speciation using capillary electrophoresis: Optimization of analyte derivatization and separation. Anal. Chem. 2012, 84, 7638–7642. [Google Scholar] [CrossRef]
  29. Tang, S.; Li, L.; Wang, R.; Regmi, S.; Zhang, X.; Yang, G.; Ju, J. A Schematic Colorimetric Assay for Sialic Acid Assay Based on PEG-Mediated Interparticle Crosslinking Aggregation of Gold Nanoparticles. Biosensors 2023, 13, 164. [Google Scholar] [CrossRef] [PubMed]
  30. Costa, J.B.S.; de Paula, N.T.G.; da Silva, P.A.B.; de Souza, G.C.S.; Paim, A.P.S.; Lavorante, A.F. A spectrophotometric procedure for sialic acid determination in milk employing a flow-batch analysis system with direct heating. Microchem. J. 2019, 147, 782–788. [Google Scholar] [CrossRef]
  31. Wang, M.-H.; Wang, Z.-F.; Yuan, M.; Yang, C.-G.; Wang, D.-L.; Wang, S.-Q. Changes in the Serum and Tissue Levels of Free and Conjugated Sialic Acids, Neu5Ac, Neu5Gc, and KDN in Mice after the Oral Administration of Edible Bird’s Nests: An LC–MS/MS Quantitative Analysis. Separations 2024, 11, 107. [Google Scholar] [CrossRef]
  32. Ye, L.; Mu, L.; Li, G.; Bao, Y. Assessing sialic acid content in food by hydrophilic chromatography-high performance liquid chromatography. J. Food Compos. Anal. 2020, 87, 103393. [Google Scholar] [CrossRef]
  33. Wang, L.; Wang, D.; Zhou, X.; Wu, L.; Sun, X.-L. Systematic investigation of quinoxaline derivatization of sialic acids and their quantitation applicability using high performance liquid chromatography. RSC Adv. 2014, 4, 45797–45803. [Google Scholar] [CrossRef]
  34. Reversed-Phase High-Performance Liquid Chromatography: A Robust Method for Quantitation of Fluorescently Labeled and Derivatized Sialic Acids Isolated from Mouse Liver. Available online: https://app.jove.com/v/21130/reversed-phase-high-performance-liquid-chromatography-robust-method?playlist=2655796&isFrc=true (accessed on 27 January 2025).
  35. Palmisano, G.; Larsen, M.R.; Packer, N.H.; Thaysen-Andersen, M. Structural analysis of glycoprotein sialylation-part II: LC-MS based detection. RSC Adv. 2013, 3, 22706–22726. [Google Scholar] [CrossRef]
  36. Wang, F.; Xie, B.; Wang, B.; Troy, F.A. LC-MS/MS glycomic analyses of free and conjugated forms of the sialic acids, Neu5Ac, Neu5Gc and KDN in human throat cancers. Glycobiology 2015, 25, 1362–1374. [Google Scholar] [CrossRef] [PubMed]
  37. van der Ham, M.; Prinsen, B.H.C.M.T.; Huijmans, J.G.M.; Abeling, N.G.G.M.; Dorland, B.; Berger, R.; de Koning, T.J.; de Sain-van der Velden, M.G.M. Quantification of free and total sialic acid excretion by LC-MS/MS. J. Chromatogr. B Analyt. Technol. Biomed. Life. Sci. 2007, 848, 251–257. [Google Scholar] [CrossRef]
  38. Zhou, S.; Dong, X.; Veillon, L.; Huang, Y.; Mechref, Y. LC-MS/MS analysis of permethylated N-glycans facilitating isomeric characterization. Anal. Bioanal. Chem. 2017, 409, 453–466. [Google Scholar] [CrossRef]
  39. Guerrero-Flores, G.N.; Pacheco, F.J.; Boskovic, D.S.; Pacheco, S.O.S.; Zhang, G.; Fraser, G.E.; Miles, F.L. Sialic acids Neu5Ac and KDN in adipose tissue samples from individuals following habitual vegetarian or non-vegetarian dietary patterns. Sci. Rep. 2023, 13, 12593. [Google Scholar] [CrossRef]
  40. Ho, C.; Lam, C.; Chan, M.; Cheung, R.; Law, L.; Lit, L.; Ng, K.; Suen, M.; Tai, H. Electrospray Ionisation Mass Spectrometry: Principles and Clinical Applications. Clin. Biochem. Rev. 2003, 24, 3–12. [Google Scholar]
  41. Mellon, F.A. MASS SPECTROMETRY|Principles and Instrumentation. In Encyclopedia of Food Sciences and Nutrition, 2nd ed.; Caballero, B., Ed.; Academic Press: Oxford, UK, 2003; pp. 3739–3749. ISBN 978-0-12-227055-0. [Google Scholar]
  42. Kaklamanos, G.; Aprea, E.; Theodoridis, G. 11—Mass spectrometry: Principles and instrumentation. In Chemical Analysis of Food, 2nd ed.; Pico, Y., Ed.; Academic Press: Cambridge, MA, USA, 2020; pp. 525–552. [Google Scholar]
  43. Banerjee, S.; Mazumdar, S. Electrospray Ionization Mass Spectrometry: A Technique to Access the Information beyond the Molecular Weight of the Analyte. Int. J. Anal. Chem. 2012, 2012, 282574. [Google Scholar] [CrossRef]
  44. Thomas, S.N.; French, D.; Jannetto, P.J.; Rappold, B.A.; Clarke, W.A. Liquid chromatography–tandem mass spectrometry for clinical diagnostics. Nat. Rev. Methods Primer 2022, 2, 96. [Google Scholar] [CrossRef]
  45. Bhardwaj, C.; Hanley, L. Ion sources for mass spectrometric identification and imaging of molecular species. Nat. Prod. Rep. 2014, 31, 756–767. [Google Scholar] [CrossRef]
  46. Malik, A.K.; Kumar, R. Heena Spectroscopy: Types. In Encyclopedia of Food and Health; Caballero, B., Finglas, P.M., Toldrá, F., Eds.; Academic Press: Oxford, UK, 2016; pp. 64–72. ISBN 978-0-12-384953-3. [Google Scholar]
  47. Ravelli, M.N.; Shriver, T.C.; Schoeller, D.A. Doubly labeled water. In Encyclopedia of Human Nutrition, 4th ed.; Caballero, B., Ed.; Academic Press: Oxford, UK, 2023; pp. 52–61. ISBN 978-0-323-90816-0. [Google Scholar]
  48. Nishikaze, T. Sialic acid derivatization for glycan analysis by mass spectrometry. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 2019, 95, 523–537. [Google Scholar] [CrossRef]
  49. Priego-Capote, F.; Orozco-Solano, M.I.; Calderón-Santiago, M.; Luque de Castro, M.D. Quantitative determination and confirmatory analysis of N-acetylneuraminic and N-glycolylneuraminic acids in serum and urine by solid-phase extraction on-line coupled to liquid chromatography-tandem mass spectrometry. J. Chromatogr. A 2014, 1346, 88–96. [Google Scholar] [CrossRef] [PubMed]
  50. Paul, A.; Padler-Karavani, V. Evolution of sialic acids: Implications in xenotransplant biology. Xenotransplantation 2018, 25, e12424. [Google Scholar] [CrossRef]
  51. Angata, T.; Varki, A. Chemical diversity in the sialic acids and related α-keto acids: An evolutionary perspective. Chem. Rev. 2002, 102, 439–469. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, C.; Chen, J.; Liu, Y.; Xu, D. Sialic acid metabolism as a potential therapeutic target of atherosclerosis. Lipids Health Dis. 2019, 18, 173. [Google Scholar] [CrossRef] [PubMed]
  53. Soulillou, J.P.; Süsal, C.; Döhler, B.; Opelz, G. No increase in colon cancer risk following induction with neu5Gc-bearing rabbit anti-T cell igg (ATG) in recipients of kidney transplants. Cancers 2018, 10, 324. [Google Scholar] [CrossRef]
  54. Inoue, S.; Kitajima, K.; Inoue, Y. Identification of 2-keto-3-deoxy-D-glycero-D-galactonononic acid (KDN, deaminoneuraminic acid) residues in mammalian tissues and human lung carcinoma cells. Chemical evidence of the occurrence of KDN glycoconjugates in mammals. J. Biol. Chem. 1996, 271, 24341–24344. [Google Scholar] [CrossRef]
  55. Inoue, S.; Kitajima, K. KDN (Deaminated neuraminic acid): Dreamful past and exciting future of the newest member of the sialic acid family. Glycoconj. J. 2006, 23, 277–290. [Google Scholar] [CrossRef]
  56. Chen, Y.; Pan, L.; Liu, N.; Troy, F.A.; Wang, B. LC-MS/MS quantification of N-acetylneuraminic acid, N-glycolylneuraminic acid and ketodeoxynonulosonic acid levels in the urine and potential relationship with dietary sialic acid intake and disease in 3-to 5-year-old children. Br. J. Nutr. 2014, 111, 332–341. [Google Scholar] [CrossRef]
  57. Li, D.; Lin, Q.; Luo, F.; Wang, H. Insights into the Structure, Metabolism, Biological Functions and Molecular Mechanisms of Sialic Acid: A Review. Foods 2024, 13, 145. [Google Scholar] [CrossRef]
  58. Guin, S.K.; Velasco-Torrijos, T.; Dempsey, E. Explorations in a galaxy of sialic acids: A review of sensing horizons, motivated by emerging biomedical and nutritional relevance. Sens. Diagn. 2022, 1, 10–70. [Google Scholar] [CrossRef]
  59. Vos, G.M.; Hooijschuur, K.C.; Li, Z.; Fjeldsted, J.; Klein, C.; de Vries, R.P.; Toraño, J.S.; Boons, G.-J. Sialic acid O-acetylation patterns and glycosidic linkage type determination by ion mobility-mass spectrometry. Nat. Commun. 2023, 14, 6795. [Google Scholar] [CrossRef] [PubMed]
  60. Chou, W.K.; Hinderlich, S.; Reutter, W.; Tanner, M.E. Sialic Acid Biosynthesis:  Stereochemistry and Mechanism of the Reaction Catalyzed by the Mammalian UDP-N-Acetylglucosamine 2-Epimerase. J. Am. Chem. Soc. 2003, 125, 2455–2461. [Google Scholar] [CrossRef] [PubMed]
  61. Wickramasinghe, S.; Medrano, J.F. Primer on genes encoding enzymes in sialic acid metabolism in mammals. Biochimie 2011, 93, 1641–1646. [Google Scholar] [CrossRef]
  62. Akella, N.M.; Ciraku, L.; Reginato, M.J. Fueling the fire: Emerging role of the hexosamine biosynthetic pathway in cancer. BMC Biol. 2019, 17, 52. [Google Scholar] [CrossRef]
  63. Carrillo, N.; Malicdan, M.C.; Leoyklang, P.; Shrader, J.A.; Joe, G.; Slota, C.; Perreault, J.; Heiss, J.D.; Class, B.; Liu, C.-Y.; et al. Safety and efficacy of N-acetylmannosamine (ManNAc) in patients with GNE myopathy: An open-label phase 2 study. Genet. Med. 2021, 23, 2067–2075. [Google Scholar] [CrossRef]
  64. Hagenhaus, V.; Gorenflos López, J.L.; Rosenstengel, R.; Neu, C.; Hackenberger, C.P.R.; Celik, A.; Weinert, K.; Nguyen, M.-B.; Bork, K.; Horstkorte, R.; et al. Glycation Interferes with the Activity of the Bi-Functional UDP-N-Acetylglucosamine 2-Epimerase/N-Acetyl-mannosamine Kinase (GNE). Biomolecules 2023, 13, 422. [Google Scholar] [CrossRef]
  65. Glaser, L. On the mechanism of N-acetylmannosamine formation. Biochim. Biophys. Acta 1960, 41, 534–536. [Google Scholar] [CrossRef]
  66. Comb, D.G.; Roseman, S. Enzymic synthesis of N-acetyl-D-mannosamine. Biochim. Biophys. Acta 1958, 29, 653–654. [Google Scholar] [CrossRef]
  67. Hinderlich, S.; Weidemann, W.; Yardeni, T.; Horstkorte, R.; Huizing, M. UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE), a master regulator of sialic acid synthesis. Top. Curr. Chem. 2015, 366, 97–137. [Google Scholar] [CrossRef]
  68. Willems, A.P.; Sun, L.; Schulz, M.A.; Tian, W.; Ashikov, A.; van Scherpenzeel, M.; Hermans, E.; Clausen, H.; Yang, Z.; Lefeber, D.J. Activity of N-acylneuraminate-9-phosphatase (NANP) is not essential for de novo sialic acid biosynthesis. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 1471–1479. [Google Scholar] [CrossRef]
  69. Teoh, S.T.; Ogrodzinski, M.P.; Ross, C.; Hunter, K.W.; Lunt, S.Y. Sialic acid metabolism: A key player in breast cancer metastasis revealed by metabolomics. Front. Oncol. 2018, 8, 174. [Google Scholar] [CrossRef]
  70. Kozutsumi, Y.; Kawano, T.; Yamakawa, T.; Suzuki, A. Participation of Cytochrome b5 in CMP-N- Acetylneuraminic Acid Hydroxylation in Mouse Liver Cytosol1. J. Biochem. 1990, 108, 704–706. [Google Scholar] [CrossRef] [PubMed]
  71. Kawano, T.; Kozutsumi, Y.; Takematsu, H.; Kawasaki, T.; Suzuki, A. Regulation of biosynthesis ofN-glycolylneuraminic acid-containing glycoconjugates: Characterization of factors required for NADH-dependent cytidine 5′monophosphate-N-acetylneuraminic acid hydroxylation. Glycoconj. J. 1993, 10, 109–115. [Google Scholar] [CrossRef] [PubMed]
  72. Li, Y.; Chen, X. Sialic acid metabolism and sialyltransferases: Natural functions and applications. Appl. Microbiol. Biotechnol. 2012, 94, 887–905. [Google Scholar] [CrossRef] [PubMed]
  73. Schauer, R.; Kelm, S.; Reuter, G.; Roggentin, P.; Shaw, L. Biochemistry and Role of Sialic Acids. In Biology of the Sialic Acids; Rosenberg, A., Ed.; Springer: Boston, MA, USA, 1995; pp. 7–67. [Google Scholar]
  74. Angata, T.; Nakata, D.; Matsuda, T.; Kitajima, K.; Troy, F.A. Biosynthesis of KDN (2-Keto-3-deoxy-d-glycero -d-galacto -nononic acid). J. Biol. Chem. 1999, 274, 22949–22956. [Google Scholar] [CrossRef]
  75. Crich, D.; Navuluri, C. Practical Synthesis of 2-Keto-3-deoxy-D- glycero-D-galactononulosonic Acid (KDN). Org. Lett. 2011, 13, 6288–6291. [Google Scholar] [CrossRef]
  76. Kawanishi, K.; Saha, S.; Diaz, S.; Vaill, M.; Sasmal, A.; Siddiqui, S.S.; Choudhury, B.; Sharma, K.; Chen, X.; Schoenhofen, I.C.; et al. Evolutionary conservation of human ketodeoxynonulosonic acid production is independent of sialoglycan biosynthesis. J. Clin. Investig. 2021, 131, e137681. [Google Scholar] [CrossRef]
  77. Varki, A. Sialic acids in human health and disease. Trends Mol. Med. 2008, 14, 351–360. [Google Scholar] [CrossRef]
  78. Pawluczyk, I.Z.A.; Najafabadi, M.G.; Brown, J.R.; Bevington, A.; Topham, P.S. Sialic acid supplementation ameliorates puromycin aminonucleoside nephrosis in rats. Lab. Investig. J. Tech. Methods Pathol. 2015, 95, 1019–1028. [Google Scholar] [CrossRef]
  79. van Karnebeek, C.D.M.; Bonafé, L.; Wen, X.-Y.; Tarailo-Graovac, M.; Balzano, S.; Royer-Bertrand, B.; Ashikov, A.; Garavelli, L.; Mammi, I.; Turolla, L.; et al. NANS-mediated synthesis of sialic acid is required for brain and skeletal development. Nat. Genet. 2016, 48, 777–784. [Google Scholar] [CrossRef]
  80. Möckl, L. The Emerging Role of the Mammalian Glycocalyx in Functional Membrane Organization and Immune System Regulation. Front. Cell Dev. Biol. 2020, 8, 253. [Google Scholar] [CrossRef]
  81. Wang, B. Molecular mechanism underlying sialic acid as an essential nutrient for brain development and cognition. Adv. Nutr. 2012, 3, 465–472. [Google Scholar] [CrossRef]
  82. Bian, D.; Wang, X.; Huang, J.; Chen, X.; Li, H. Maternal Neu5Ac Supplementation During Pregnancy Improves Offspring Learning and Memory Ability in Rats. Front. Nutr. 2021, 8, 641027. [Google Scholar] [CrossRef]
  83. Cho, A.; Christine, M.; Malicdan, V.; Miyakawa, M.; Nonaka, I.; Nishino, I.; Noguchi, S. Sialic acid deficiency is associated with oxidative stress leading to muscle atrophy and weakness in GNE myopathy. Hum. Mol. Genet. 2017, 26, 3081–3093. [Google Scholar] [CrossRef]
  84. Pogoryelova, O.; González Coraspe, J.A.; Nikolenko, N.; Lochmüller, H.; Roos, A. GNE myopathy: From clinics and genetics to pathology and research strategies. Orphanet J. Rare Dis. 2018, 13, 70. [Google Scholar] [CrossRef]
  85. den Hollander, B.; Rasing, A.; Post, M.A.; Klein, W.M.; Oud, M.M.; Brands, M.M.; de Boer, L.; Engelke, U.F.H.; van Essen, P.; Fuchs, S.A.; et al. NANS-CDG: Delineation of the Genetic, Biochemical, and Clinical Spectrum. Front. Neurol. 2021, 12, 668640. [Google Scholar] [CrossRef]
  86. Betenbaugh, M.J.; Yin, B.; Blake, E.; Kristoffersen, L.; Narang, S.; Viswanathan, K. N-Acetylneuraminic Acid Synthase (NANS). In Handbook of Glycosyltransferases and Related Genes; Taniguchi, N., Honke, K., Fukuda, M., Narimatsu, H., Yamaguchi, Y., Angata, T., Eds.; Springer: Tokyo, Japan, 2014; pp. 1523–1536. [Google Scholar]
  87. Jin, J.; Fang, F.; Gao, W.; Chen, H.; Wen, J.; Wen, X.; Chen, J. The Structure and Function of the Glycocalyx and Its Connection with Blood-Brain Barrier. Front. Cell. Neurosci. 2021, 15, 739699. [Google Scholar] [CrossRef]
  88. Houghton, A.N.; Guevara-Patiño, J.A. Immune recognition of self in immunity against cancer. J. Clin. Investig. 2004, 114, 468–471. [Google Scholar] [CrossRef]
  89. Li, F.; Ding, J. Sialylation is involved in cell fate decision during development, reprogramming and cancer progression. Protein Cell 2019, 10, 550–565. [Google Scholar] [CrossRef] [PubMed]
  90. Ghasempour, S.; Freeman, S.A. The glycocalyx and immune evasion in cancer. FEBS J. 2023, 290, 55–65. [Google Scholar] [CrossRef]
  91. Wang, M.; Zhu, J.; Lubman, D.M.; Gao, C. Aberrant glycosylation and cancer biomarker discovery: A promising and thorny journey. Clin. Chem. Lab. Med. CCLM 2019, 57, 407–416. [Google Scholar] [CrossRef] [PubMed]
  92. Hugonnet, M.; Singh, P.; Haas, Q.; von Gunten, S. The Distinct Roles of Sialyltransferases in Cancer Biology and Onco-Immunology. Front. Immunol. 2021, 12, 799861. [Google Scholar] [CrossRef]
  93. Le, T.; Ferling, I.; Qiu, L.; Nabaile, C.; Assunção, L.; Roskelley, C.D.; Grinstein, S.; Freeman, S.A. Redistribution of the glycocalyx exposes phagocytic determinants on apoptotic cells. Dev. Cell 2024, 59, 853–868.e7. [Google Scholar] [CrossRef]
  94. Cho, S.; Zhu, Z.; Li, T.; Baluyot, K.; Howell, B.R.; Hazlett, H.C.; Elison, J.T.; Hauser, J.; Sprenger, N.; Wu, D.; et al. Human milk 3′-Sialyllactose is positively associated with language development during infancy. Am. J. Clin. Nutr. 2021, 114, 588–597. [Google Scholar] [CrossRef]
  95. Schnaar, R.L.; Gerardy-Schahn, R.; Hildebrandt, H. Sialic Acids in the Brain: Gangliosides and Polysialic Acid in Nervous System Development, Stability, Disease, and Regeneration. Physiol. Rev. 2014, 94, 461–518. [Google Scholar] [CrossRef]
  96. Liu, F.; Simpson, A.B.; D’Costa, E.; Bunn, F.S.; van Leeuwen, S.S. Sialic acid, the secret gift for the brain. Crit. Rev. Food Sci. Nutr. 2023, 63, 9875–9894. [Google Scholar] [CrossRef]
  97. Röhrig, C.H.; Choi, S.S.H.; Baldwin, N. The nutritional role of free sialic acid, a human milk monosaccharide, and its application as a functional food ingredient. Crit. Rev. Food Sci. Nutr. 2017, 57, 1017–1038. [Google Scholar] [CrossRef] [PubMed]
  98. Pang, W.W.; Tan, P.T.; Cai, S.; Fok, D.; Chua, M.C.; Lim, S.B.; Shek, L.P.; Chan, S.-Y.; Tan, K.H.; Yap, F.; et al. Nutrients or nursing? Understanding how breast milk feeding affects child cognition. Eur. J. Nutr. 2020, 59, 609–619. [Google Scholar] [CrossRef]
  99. Wang, B.; Brand-Miller, J.; McVeagh, P.; Petocz, P. Concentration and distribution of sialic acid in human milk and infant formulas. Am. J. Clin. Nutr. 2001, 74, 510–515. [Google Scholar] [CrossRef]
  100. Rawal, P.; Zhao, L. Sialometabolism in Brain Health and Alzheimer’s Disease. Front. Neurosci. 2021, 15, 648617. [Google Scholar] [CrossRef]
  101. Wang, B.; Yu, B.; Karim, M.; Hu, H.; Sun, Y.; McGreevy, P.; Petocz, P.; Held, S.; Brand-Miller, J. Dietary sialic acid supplementation improves learning and memory in piglets2. Matern. Nutr. Optim. Infant Feed. Pract. 2007, 85, 561–569. [Google Scholar] [CrossRef]
  102. Carrillo, N.; Malicdan, M.C.; Huizing, M. GNE Myopathy: Etiology, Diagnosis, and Therapeutic Challenges. Neurotherapeutics 2018, 15, 900–914. [Google Scholar] [CrossRef]
  103. Mori-Yoshimura, M.; Oya, Y.; Yajima, H.; Yonemoto, N.; Kobayashi, Y.; Hayashi, Y.K.; Noguchi, S.; Nishino, I.; Murata, M. GNE myopathy: A prospective natural history study of disease progression. Neuromuscul. Disord. 2014, 24, 380–386. [Google Scholar] [CrossRef]
  104. Xu, X.; Wang, A.Q.; Latham, L.L.; Celeste, F.; Ciccone, C.; Malicdan, M.C.; Goldspiel, B.; Terse, P.; Cradock, J.; Yang, N.; et al. Safety, pharmacokinetics and sialic acid production after oral administration of N -acetylmannosamine (ManNAc) to subjects with GNE myopathy. Mol. Genet. Metab. 2017, 122, 126–134. [Google Scholar] [CrossRef] [PubMed]
  105. Ferreira, C.R.; Gahl, W.A. Lysosomal storage diseases. Transl. Sci. Rare Dis. 2017, 2, 1–71. [Google Scholar] [CrossRef]
  106. Liu, L.; Lee, W.-S.; Doray, B.; Kornfeld, S. Engineering of GlcNAc-1-Phosphotransferase for Production of Highly Phosphorylated Lysosomal Enzymes for Enzyme Replacement Therapy. Mol. Ther.—Methods Clin. Dev. 2017, 5, 59–65. [Google Scholar] [CrossRef] [PubMed]
  107. Ito, M. 3.11—Degradation of Glycolipids. In Comprehensive Glycoscience; Kamerling, H., Ed.; Elsevier: Oxford, UK, 2007; pp. 193–208. ISBN 978-0-444-51967-2. [Google Scholar]
  108. Xiao, C.; Toro, C.; Tifft, C. GM2 Activator Deficiency. In GeneReviews®; Adam, M.P., Feldman, J., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  109. Chapleau, A.; Mirchi, A.; Tran, L.T.; Poulin, C.; Bernard, G. Longitudinal Characterization of the Clinical Course of Intermediate-Severe Salla Disease. Pediatr. Neurol. 2023, 148, 133–137. [Google Scholar] [CrossRef] [PubMed]
  110. Schauer, R.; Kamerling, J.P. Exploration of the Sialic Acid World. In Advances in Carbohydrate Chemistry and Biochemistry; Elsevier: Amsterdam, The Netherlands, 2018; pp. 1–213. [Google Scholar]
  111. Tietze, F.; Seppala, R.; Renlund, M.; Hopwood, J.J.; Harper, G.S.; Thomas, G.H.; Gahl, W.A. Defective lysosomal egress of free sialic acid (N-acetylneuraminic acid) in fibroblasts of patients with infantile free sialic acid storage disease. J. Biol. Chem. 1989, 264, 15316–15322. [Google Scholar] [CrossRef]
  112. Adams, D.; Wasserstein, M. Free Sialic Acid Storage Disorders. In GeneReviews®; Adam, M.P., Feldman, J., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  113. Huizing, M.; Hackbarth, M.E.; Adams, D.R.; Wasserstein, M.; Patterson, M.C.; Walkley, S.U.; Gahl, W.A.; Adams, D.R.; Dobrenis, K.; Foglio, J.; et al. Free sialic acid storage disorder: Progress and promise. Neurosci. Lett. 2021, 755, 135896. [Google Scholar] [CrossRef]
  114. Sabir, M.S.; Leoyklang, P.; Hackbarth, M.E.; Pak, E.; Dutra, A.; Tait, R.; Pollard, L.; Adams, D.R.; Gahl, W.A.; Huizing, M.; et al. Generation and characterization of two iPSC lines derived from subjects with Free Sialic Acid Storage Disorder (FSASD). Stem Cell Res. 2024, 81, 103600. [Google Scholar] [CrossRef]
  115. Barmherzig, R.; Bullivant, G.; Cordeiro, D.; Sinasac, D.S.; Blaser, S.; Mercimek-Mahmutoglu, S. A New Patient with Intermediate Severe Salla Disease with Hypomyelination: A Literature Review for Salla Disease. Pediatr. Neurol. 2017, 74, 87–91.e2. [Google Scholar] [CrossRef]
  116. Tangvoranuntakul, P.; Gagneux, P.; Diaz, S.; Bardor, M.; Varki, N.; Varki, A.; Muchmore, E. Human uptake and incorporation of an immunogenic nonhuman dietary sialic acid. Proc. Natl. Acad. Sci. USA 2003, 100, 12045–12050. [Google Scholar] [CrossRef]
  117. Nöhle, U.; Schauer, R. Metabolism of Sialic Acids from Exogeneously Administered Sialyllactose and Mucin in Mouse and Rat. Biol. Chem. 1984, 365, 1457–1468. [Google Scholar] [CrossRef]
  118. Jahan, M.; Thomson, P.C.; Wynn, P.C.; Wang, B. Red Meat Derived Glycan, N-acetylneuraminic Acid (Neu5Ac) Is a Major Sialic Acid in Different Skeletal Muscles and Organs of Nine Animal Species—A Guideline for Human Consumers. Foods 2023, 12, 337. [Google Scholar] [CrossRef] [PubMed]
  119. Samraj, A.N.; Läubli, H.; Varki, N.; Varki, A. Involvement of a Non-Human Sialic Acid in Human Cancer. Front. Oncol. 2014, 4, 33. [Google Scholar] [CrossRef]
  120. Yao, H.L.; Conway, L.P.; Wang, M.M.; Huang, K.; Liu, L.; Voglmeir, J. Quantification of sialic acids in red meat by UPLC-FLD using indoxylsialosides as internal standards. Glycoconj. J. 2016, 33, 219–226. [Google Scholar] [CrossRef]
  121. Bardor, M.; Nguyen, D.H.; Diaz, S.; Varki, A. Mechanism of Uptake and Incorporation of the Non-human Sialic Acid N-Glycolylneuraminic Acid into Human Cells. J. Biol. Chem. 2005, 280, 4228–4237. [Google Scholar] [CrossRef]
  122. Dhar, C.; Sasmal, A.; Varki, A. From “Serum Sickness” to “Xenosialitis”: Past, Present, and Future Significance of the Non-human Sialic Acid Neu5Gc. Front. Immunol. 2019, 10, 807. [Google Scholar] [CrossRef]
  123. Sroga, J.M.; Wu, D.H.; Ma, F.; Tecle, E.; Ressler, I.B.; Maxwell, R.; Ferrari, R.; Whigham, L.; Gagneux, P.; Lindheim, S.R. Detection of the dietary xenoglycan N-glycolylneuraminic acid (Neu5Gc) and anti-Neu5Gc antibodies within reproductive tracts of male and female infertility subjects. Clin. Obstet. Gynecol. Reprod. Med. 2015, 1, 72–78. [Google Scholar] [CrossRef]
  124. Leviatan Ben-Arye, S.; Yu, H.; Chen, X.; Padler-Karavani, V. Profiling Anti-Neu5Gc IgG in Human Sera with a Sialoglycan Microarray Assay. J. Vis. Exp. 2017, 125, e56094. [Google Scholar] [CrossRef]
  125. Padler-karavani, V.; Yu, H.; Cao, H.; Karp, F.; Varki, N.; Chen, X. Diversity in specificity, abundance, and composition of anti-Neu5Gc antibodies in normal humans: Potential implications for disease. Glycobiology 2008, 18, 818–830. [Google Scholar] [CrossRef]
  126. Tran, C.; Turolla, L.; Ballhausen, D.; Buros, S.C.; Teav, T.; Gallart-Ayala, H.; Ivanisevic, J.; Faouzi, M.; Lefeber, D.J.; Ivanovski, I.; et al. The fate of orally administered sialic acid: First insights from patients with N-acetylneuraminic acid synthase deficiency and control subjects. Mol. Genet. Metab. Rep. 2021, 28, 100777. [Google Scholar] [CrossRef] [PubMed]
  127. Soares, C.O.; Grosso, A.S.; Ereño-Orbea, J.; Coelho, H.; Marcelo, F. Molecular Recognition Insights of Sialic Acid Glycans by Distinct Receptors Unveiled by NMR and Molecular Modeling. Front. Mol. Biosci. 2021, 8, 727847. [Google Scholar]
  128. Wang, B.; Brand-Miller, J. The role and potential of sialic acid in human nutrition. Eur. J. Clin. Nutr. 2003, 57, 1351–1369. [Google Scholar] [CrossRef]
  129. Zeleny, R.; Kolarich, D.; Strasser, R.; Altmann, F. Sialic acid concentrations in plants are in the range of inadvertent contamination. Planta 2006, 224, 222–227. [Google Scholar] [CrossRef]
  130. Ylönen, A.; Kalkkinen, N.; Saarinen, J.; Bøgwald, J.; Helin, J. Glycosylation analysis of two cysteine proteinase inhibitors from Atlantic salmon skin: Di-O-acetylated sialic acids are the major sialic acid species on N-glycans. Glycobiology 2001, 11, 523–531. [Google Scholar] [CrossRef] [PubMed]
  131. Bashir, S.; Fezeu, L.K.; Leviatan Ben-Arye, S.; Yehuda, S.; Reuven, E.M.; Szabo De Edelenyi, F.; Fellah-Hebia, I.; Le Tourneau, T.; Imbert-Marcille, B.M.; Drouet, E.B.; et al. Association between Neu5Gc carbohydrate and serum antibodies against it provides the molecular link to cancer: French NutriNet-Santé study. BMC Med. 2020, 18, 262. [Google Scholar] [CrossRef]
  132. Altman, M.O.; Gagneux, P. Absence of Neu5Gc and Presence of Anti-Neu5Gc Antibodies in Humans-An Evolutionary Perspective. Front. Immunol. 2019, 10, 789. [Google Scholar] [CrossRef]
  133. Schauer, R.; Srinivasan, G.V.; Coddeville, B.; Zanetta, J.-P.; Guérardel, Y. Low incidence of N-glycolylneuraminic acid in birds and reptiles and its absence in the platypus. Carbohydr. Res. 2009, 344, 1494–1500. [Google Scholar] [CrossRef]
  134. Peri, S.; Kulkarni, A.; Feyertag, F.; Berninsone, P.M.; Alvarez-Ponce, D. Phylogenetic Distribution of CMP-Neu5Ac Hydroxylase (CMAH), the Enzyme Synthetizing the Proinflammatory Human Xenoantigen Neu5Gc. Genome Biol. Evol. 2018, 10, 207–219. [Google Scholar] [CrossRef]
  135. Koropatkin, N.M.; Cameron, E.A.; Martens, E.C. How glycan metabolism shapes the human gut microbiota. Nat. Rev. Microbiol. 2012, 10, 323–335. [Google Scholar] [CrossRef]
  136. Tannock Gerald, W. Modulating the Gut Microbiota of Humans by Dietary Intervention with Plant Glycans. Appl. Environ. Microbiol. 2021, 87, e02757-20. [Google Scholar] [CrossRef] [PubMed]
  137. Adak, A.; Khan, M.R. An insight into gut microbiota and its functionalities. Cell. Mol. Life Sci. 2019, 76, 473–493. [Google Scholar] [CrossRef]
  138. El-Sayed, A.; Aleya, L.; Kamel, M. Microbiota’s role in health and diseases. Environ. Sci. Pollut. Res. 2021, 28, 36967–36983. [Google Scholar] [CrossRef]
  139. Ottman, N.; Smidt, H.; de Vos, W.M.; Belzer, C. The function of our microbiota: Who is out there and what do they do? Front. Cell. Infect. Microbiol. 2012, 2, 104. [Google Scholar] [CrossRef]
  140. Cronin, P.; Joyce, S.A.; O’Toole, P.W.; O’Connor, E.M. Dietary Fibre Modulates the Gut Microbiota. Nutrients 2021, 13, 1655. [Google Scholar] [CrossRef] [PubMed]
  141. Heimann, E.; Nyman, M.; Degerman, E. Propionic acid and butyric acid inhibit lipolysis and de novo lipogenesis and increase insulin-stimulated glucose uptake in primary rat adipocytes. Adipocyte 2015, 4, 81–88. [Google Scholar] [CrossRef]
  142. Lin, L.; Zhang, J. Role of intestinal microbiota and metabolites on gut homeostasis and human diseases. BMC Immunol. 2017, 18, 2. [Google Scholar] [CrossRef]
  143. Coker, J.K.; Moyne, O.; Rodionov, D.A.; Zengler, K. Carbohydrates great and small, from dietary fiber to sialic acids: How glycans influence the gut microbiome and affect human health. Gut Microbes 2021, 13, 1869502. [Google Scholar] [CrossRef]
  144. Butler, T.L.; Fraser, G.E.; Beeson, W.L.; Knutsen, S.F.; Herring, R.P.; Chan, J.; Sabaté, J.; Montgomery, S.; Haddad, E.; Preston-Martin, S.; et al. Cohort Profile: The Adventist Health Study-2 (AHS-2). Int. J. Epidemiol. 2008, 37, 260–265. [Google Scholar] [CrossRef]
  145. Davey, G.K.; Spencer, E.A.; Appleby, P.N.; Allen, N.E.; Knox, K.H.; Key, T.J. EPIC–Oxford:lifestyle characteristics and nutrient intakes in a cohort of 33 883 meat-eaters and 31 546 non meat-eaters in the UK. Public Health Nutr. 2003, 6, 259–268. [Google Scholar] [CrossRef]
  146. Englyst, N.A.; Crook, M.A.; Lumb, P.; Stears, A.J.; Masding, M.G.; Wootton, S.A.; Sandeman, D.D.; Byrne, C.D. Percentage of body fat and plasma glucose predict plasma sialic acid concentration in type 2 diabetes mellitus. Metabolism 2006, 55, 1165–1170. [Google Scholar] [CrossRef] [PubMed]
  147. Wakabayashi, I.; Masuda, H. Relation of serum sialic acid to blood coagulation activity in type 2 diabetes. Blood Coagul. Fibrinolysis 2002, 13, 691–696. [Google Scholar] [CrossRef]
  148. Cheeseman, J.; Badia, C.; Elgood-Hunt, G.; Gardner, R.A.; Trinh, D.N.; Monopoli, M.P.; Kuhnle, G.; Spencer, D.I.R.; Osborn, H.M.I. Elevated Concentrations of Neu5Ac and Neu5,9Ac2 in Human Plasma: Potential Biomarkers of Cardiovascular Disease. Glycoconj. J. 2023, 40, 645–654. [Google Scholar] [CrossRef]
  149. Demina, E.P.; Smutova, V.; Pan, X.; Fougerat, A.; Guo, T.; Zou, C.; Chakraberty, R.; Snarr, B.D.; Shiao, T.C.; Roy, R.; et al. Neuraminidases 1 and 3 Trigger Atherosclerosis by Desialylating Low-Density Lipoproteins and Increasing Their Uptake by Macrophages. J. Am. Heart Assoc. 2021, 10, e018756. [Google Scholar] [CrossRef]
  150. Samraj, A.N.; Bertrand, K.A.; Luben, R.; Khedri, Z.; Yu, H.; Nguyen, D.; Gregg, C.J.; Diaz, S.L.; Sawyer, S.; Chen, X.; et al. Polyclonal human antibodies against glycans bearing red meat-derived non-human sialic acid N-glycolylneuraminic acid are stable, reproducible, complex and vary between individuals: Total antibody levels are associated with colorectal cancer risk. PLoS ONE 2018, 13, e0197464. [Google Scholar] [CrossRef] [PubMed]
  151. Yabu, M.; Korekane, H.; Takahashi, H.; Ohigashi, H.; Ishikawa, O.; Miyamoto, Y. Accumulation of free Neu5Ac-containing complex-type N-glycans in human pancreatic cancers. Glycoconj. J. 2013, 30, 247–256. [Google Scholar] [CrossRef]
  152. Cornelissen, L.A.M.; Blanas, A.; van der Horst, J.C.; Kruijssen, L.; Zaal, A.; O’Toole, T.; Wiercx, L.; van Kooyk, Y.; van Vliet, S.J. Disruption of sialic acid metabolism drives tumor growth by augmenting CD8 + T cell apoptosis. Int. J. Cancer 2019, 144, 2290–2302. [Google Scholar] [CrossRef] [PubMed]
  153. Zaramela, L.S.; Martino, C.; Alisson-Silva, F.; Rees, S.D.; Diaz, S.L.; Chuzel, L.; Ganatra, M.B.; Taron, C.H.; Secrest, P.; Zuñiga, C.; et al. Gut bacteria responding to dietary change encode sialidases that exhibit preference for red meat-associated carbohydrates. Nat. Microbiol. 2019, 4, 2082–2089. [Google Scholar] [CrossRef]
  154. McDonald, N.D.; Boyd, E.F. Structural and Biosynthetic Diversity of Nonulosonic Acids (NulOs) That Decorate Surface Structures in Bacteria. Trends Microbiol. 2021, 29, 142–157. [Google Scholar] [CrossRef]
  155. Bell, A.; Severi, E.; Lee, M.; Monaco, S.; Latousakis, D.; Angulo, J.; Thomas, G.H.; Naismith, J.H.; Juge, N. Uncovering a novel molecular mechanism for scavenging sialic acids in bacteria. J. Biol. Chem. 2020, 295, 13724–13736. [Google Scholar] [CrossRef]
  156. Aruni, W.; Vanterpool, E.; Osbourne, D.; Roy, F.; Muthiah, A.; Dou, Y.; Fletcher, H.M. Sialidase and Sialoglycoproteases Can Modulate Virulence in Porphyromonas gingivalis. Infect. Immun. 2011, 79, 2779–2791. [Google Scholar] [CrossRef]
  157. Jennings, M.P.; Day, C.J.; Atack, J.M. How bacteria utilize sialic acid during interactions with the host: Snip, snatch, dispatch, match and attach: This article is part of the Bacterial Cell Envelopes collection. Microbiology 2022, 168, 001157. [Google Scholar] [CrossRef]
  158. Dedola, S.; Ahmadipour, S.; de Andrade, P.; Baker, A.N.; Boshra, A.N.; Chessa, S.; Gibson, M.I.; Hernando, P.J.; Ivanova, I.M.; Lloyd, J.E.; et al. Sialic acids in infection and their potential use in detection and protection against pathogens. RSC Chem. Biol. 2023, 5, 167. [Google Scholar] [CrossRef]
  159. Cavalcante, T.; Medeiros, M.M.; Mule, S.N.; Palmisano, G.; Stolf, B.S. The Role of Sialic Acids in the Establishment of Infections by Pathogens, with Special Focus on Leishmania. Front. Cell. Infect. Microbiol. 2021, 11, 671913. [Google Scholar] [CrossRef]
  160. Matrosovich, M.; Herrler, G.; Klenk, H.D. Sialic Acid Receptors of Viruses. In SialoGlyco Chemistry and Biology II; Springer: Cham, Switzerland, 2013; Volume 367, pp. 1–28. [Google Scholar] [CrossRef]
  161. Burzyńska, P.; Sobala, Ł.; Mikołajczyk, K.; Jodłowska, M.; Jaśkiewicz, E. Sialic Acids as Receptors for Pathogens. Biomolecules 2021, 11, 831. [Google Scholar] [CrossRef] [PubMed]
  162. Tolia, N.H.; Enemark, E.J.; Sim, B.K.L.; Joshua-Tor, L. Structural Basis for the EBA-175 Erythrocyte Invasion Pathway of the Malaria Parasite Plasmodium falciparum. Cell 2005, 122, 183–193. [Google Scholar] [CrossRef] [PubMed]
  163. Morrot, A.; Villar, S.R.; González, F.B.; Pérez, A.R. Evasion and Immuno-Endocrine Regulation in Parasite Infection: Two Sides of the Same Coin in Chagas Disease? Front. Microbiol. 2016, 7, 704. [Google Scholar] [CrossRef]
  164. Both, P.; Riese, M.; Gray, C.J.; Huang, K.; Pallister, E.G.; Kosov, I.; Conway, L.P.; Voglmeir, J.; Flitsch, S.L. Applications of a highly α2,6-selective pseudosialidase. Glycobiology 2018, 28, 261–268. [Google Scholar] [CrossRef]
  165. Haines-Menges, B.L.; Whitaker, W.B.; Lubin, J.B.; Boyd, E. Fidelma Host Sialic Acids: A Delicacy for the Pathogen with Discerning Taste. Microbiol. Spectr. 2015, 3, MBP-0005-2014. [Google Scholar] [CrossRef]
  166. Liu, F.; Dong, Q.; Myers, A.M.; Fromm, H.J. Expression of human brain hexokinase in Escherichiacoli: Purification and characterization of the expressed enzyme. Biochem. Biophys. Res. Commun. 1991, 177, 305–311. [Google Scholar] [CrossRef]
  167. Bell, A.; Severi, E.; Owen, C.D.; Latousakis, D.; Juge, N. Biochemical and structural basis of sialic acid utilization by gut microbes. J. Biol. Chem. 2023, 299, 102989. [Google Scholar] [CrossRef]
  168. Zhao, M.; Zhu, Y.; Wang, H.; Zhang, W.; Mu, W. Recent advances on N-acetylneuraminic acid: Physiological roles, applications, and biosynthesis. Synth. Syst. Biotechnol. 2023, 8, 509–519. [Google Scholar] [CrossRef] [PubMed]
  169. Lin, B.-X.; Qiao, Y.; Shi, B.; Tao, Y. Polysialic acid biosynthesis and production in Escherichia coli: Current state and perspectives. Appl. Microbiol. Biotechnol. 2016, 100, 1–8. [Google Scholar] [CrossRef]
  170. Feng, Y.; Cao, M.; Shi, J.; Zhang, H.; Hu, D.; Zhu, J.; Zhang, X.; Geng, M.; Zheng, F.; Pan, X.; et al. Attenuation of Streptococcus suis virulence by the alteration of bacterial surface architecture. Sci. Rep. 2012, 2, 710. [Google Scholar] [CrossRef] [PubMed]
  171. Rodríguez-Díaz, J.; Rubio-del-Campo, A.; Yebra, M.J. Regulatory insights into the production of UDP-N-acetylglucosamine by Lactobacillus casei. Bioengineered 2012, 3, 339–342. [Google Scholar] [CrossRef]
  172. Ko, T.-P.; Lai, S.-J.; Hsieh, T.-J.; Yang, C.-S.; Chen, Y. The tetrameric structure of sialic acid–synthesizing UDP-GlcNAc 2-epimerase from Acinetobacter baumannii: A comparative study with human GNE. J. Biol. Chem. 2018, 293, 10119–10127. [Google Scholar] [CrossRef]
  173. Zapata, G.; Crowley, J.M.; Vann, W.F. Sequence and expression of the Escherichia coli K1 neuC gene product. J. Bacteriol. 1992, 174, 315–319. [Google Scholar] [CrossRef]
  174. Vimr, E.R.; Aaronson, W.; Silver, R.P. Genetic analysis of chromosomal mutations in the polysialic acid gene cluster of Escherichia coli K1. J. Bacteriol. 1989, 171, 1106–1117. [Google Scholar] [CrossRef]
  175. Silver, R.P.; Vann, W.F.; Aaronson, W. Genetic and Molecular Analyses of Escherichia coli KI Antigen Genes. J. Bacteriol. 1984, 157, 568–575. [Google Scholar] [CrossRef]
  176. Vimr, E.R.; Kalivoda, K.A.; Deszo, E.L.; Steenbergen, S.M. Diversity of Microbial Sialic Acid Metabolism. Microbiol. Mol. Biol. Rev. 2004, 68, 132–153. [Google Scholar] [CrossRef]
  177. Vimr, E.; Lichtensteiger, C.; Steenbergen, S. Sialic acid metabolism’s dual function in Haemophilus influenzae. Mol. Microbiol. 2000, 36, 1113–1123. [Google Scholar] [CrossRef] [PubMed]
  178. Flack, E.K.; Chidwick, H.S.; Best, M.; Thomas, G.H.; Fascione, M.A. Synthetic Approaches for Accessing Pseudaminic Acid (Pse) Bacterial Glycans. ChemBioChem 2020, 21, 1397–1407. [Google Scholar] [CrossRef] [PubMed]
  179. Chidwick, H.S.; Flack, E.K.P.; Keenan, T.; Walton, J.; Thomas, G.H.; Fascione, M.A. Reconstitution and optimisation of the biosynthesis of bacterial sugar pseudaminic acid (Pse5Ac7Ac) enables preparative enzymatic synthesis of CMP-Pse5Ac7Ac. Sci. Rep. 2021, 11, 4756. [Google Scholar] [CrossRef]
  180. Wei, R.; Liu, H.; Li, X. De novo synthesis of novel bacterial monosaccharide fusaminic acid. J. Antibiot. 2019, 72, 420–431. [Google Scholar] [CrossRef]
  181. Kenyon, J.J.; Marzaioli, A.M.; De Castro, C.; Hall, R.M. 5,7-di-N-acetyl-acinetaminic acid: A novel non-2-ulosonic acid found in the capsule of an Acinetobacter baumannii isolate. Glycobiology 2015, 25, 644–654. [Google Scholar] [CrossRef]
  182. Hassan, M.I.; Lundgren, B.R.; Chaumun, M.; Whitfield, D.M.; Clark, B.; Schoenhofen, I.C.; Boddy, C.N. Total Biosynthesis of Legionaminic Acid, a Bacterial Sialic Acid Analogue. Angew. Chem. Int. Ed. Engl. 2016, 55, 12018–12021. [Google Scholar] [CrossRef]
  183. Tomás-Martínez, S.; Kleikamp, H.B.C.; Neu, T.R.; Pabst, M.; Weissbrodt, D.G.; van Loosdrecht, M.C.M.; Lin, Y. Production of nonulosonic acids in the extracellular polymeric substances of “Candidatus Accumulibacter phosphatis”. Appl. Microbiol. Biotechnol. 2021, 105, 3327–3338. [Google Scholar] [CrossRef]
  184. Eke, P.I.; Borgnakke, W.S.; Genco, R.J. Recent epidemiologic trends in periodontitis in the USA. Periodontol. 2000 2020, 82, 257–267. [Google Scholar] [CrossRef]
  185. Yu, S.; Fan, X.; Zheng, S.; Lin, L.; Liu, J.; Pan, Y.; Li, C. The sialidase inhibitor, DANA, reduces Porphyromonas gingivalis pathogenicity and exerts anti-inflammatory effects: An in vitro and in vivo experiment. J. Periodontol. 2021, 92, 286–297. [Google Scholar] [CrossRef]
  186. Bostanci, N.; Bao, K.; Greenwood, D.; Silbereisen, A.; Belibasakis, G.N. Chapter Six—Periodontal disease: From the lenses of light microscopy to the specs of proteomics and next-generation sequencing. In Advances in Clinical Chemistry; Makowski, G.S., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; Volume 93, pp. 263–290. [Google Scholar] [CrossRef]
  187. Chen, W.A.; Fletcher, H.M.; Gheorghe, J.D.; Oyoyo, U.; Boskovic, D.S. Platelet plug formation in whole blood is enhanced in the presence of Porphyromonas gingivalis. Mol. Oral Microbiol. 2020, 35, 251–259. [Google Scholar] [CrossRef]
  188. Chen, W.A.; Fletcher, H.M.; Payne, K.J.; Aka, S.; Thornburg, M.B.; Gheorghe, J.D.; Safi, S.B.; Shavlik, D.; Oyoyo, U.; Boskovic, D.S. Platelet and neutrophil responses to Porphyromonas gingivalis in human whole blood. Mol. Oral Microbiol. 2021, 36, 202–213. [Google Scholar] [CrossRef]
  189. Frey, A.M.; Satur, M.J.; Phansopa, C.; Honma, K.; Urbanowicz, P.A.; Spencer, D.I.R.; Pratten, J.; Bradshaw, D.; Sharma, A.; Stafford, G. Characterization of Porphyromonas gingivalis sialidase and disruption of its role in host–pathogen interactions. Microbiology 2019, 165, 1181–1197. [Google Scholar] [CrossRef] [PubMed]
  190. Chen, W.A.; Dou, Y.; Fletcher, H.M.; Boskovic, D.S. Local and Systemic Effects of Porphyromonas gingivalis Infection. Microorganisms 2023, 11, 470. [Google Scholar] [CrossRef] [PubMed]
  191. Lamont, R.J.; Jenkinson, H.F. Life Below the Gum Line: Pathogenic Mechanisms of Porphyromonas gingivalis. Microbiol. Mol. Biol. Rev. 1998, 62, 1244–1263. [Google Scholar] [CrossRef] [PubMed]
  192. Nemoto, T.K.; Ohara-Nemoto, Y. Exopeptidases and gingipains in Porphyromonas gingivalis as prerequisites for its amino acid metabolism. Jpn. Dent. Sci. Rev. 2016, 52, 22–29. [Google Scholar] [CrossRef] [PubMed]
  193. Moradali, M.F.; Davey, M.E. Metabolic plasticity enables lifestyle transitions of Porphyromonas gingivalis. Npj Biofilms Microbiomes 2021, 7, 46. [Google Scholar] [CrossRef]
  194. Nakayama, M.; Ohara, N. Molecular mechanisms of Porphyromonas gingivalis-host cell interaction on periodontal diseases. Jpn. Dent. Sci. Rev. 2017, 53, 134–140. [Google Scholar] [CrossRef]
  195. Mysak, J.; Podzimek, S.; Sommerova, P.; Lyuya-Mi, Y.; Bartova, J.; Janatova, T.; Prochazkova, J.; Duskova, J. Porphyromonas gingivalis: Major Periodontopathic Pathogen Overview. J. Immunol. Res. 2014, 2014, 476068. [Google Scholar] [CrossRef]
  196. Cekici, A.; Kantarci, A.; Hasturk, H.; Van Dyke, T.E. Inflammatory and immune pathways in the pathogenesis of periodontal disease. Periodontol. 2000 2014, 64, 57–80. [Google Scholar] [CrossRef]
  197. Werheim, E.R.; Senior, K.G.; Shaffer, C.A.; Cuadra, G.A. Oral Pathogen Porphyromonas gingivalis Can Escape Phagocytosis of Mammalian Macrophages. Microorganisms 2020, 8, 1432. [Google Scholar] [CrossRef]
  198. Cueno, M.E.; Kamio, N.; Imai, K.; Ohya, M.; Tamura, M.; Ochiai, K. Structural Significance of the β1K396 Residue Found in the Porphyromonas gingivalis Sialidase β-Propeller Domain: A Computational Study with Implications for Novel Therapeutics Against Periodontal Disease. OMICS J. Integr. Biol. 2014, 18, 591–599. [Google Scholar] [CrossRef] [PubMed]
  199. Weiser, J.N.; Ferreira, D.M.; Paton, J.C. Streptococcus pneumoniae: Transmission, colonization and invasion. Nat. Rev. Microbiol. 2018, 16, 355–367. [Google Scholar] [CrossRef] [PubMed]
  200. Loughran, A.J.; Orihuela, C.J.; Tuomanen, E.I. Streptococcus pneumoniae: Invasion and Inflammation. Microbiol. Spectr. 2019, 7, GPP3-0004-2018. [Google Scholar] [CrossRef]
  201. 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, 10798–10804. [Google Scholar] [CrossRef]
  202. Hentrich, K.; Löfling, J.; Pathak, A.; Nizet, V.; Varki, A.; Henriques-Normark, B. Streptococcus pneumoniae Senses a Human-like Sialic Acid Profile via the Response Regulator CiaR. Cell Host Microbe 2016, 20, 307–317. [Google Scholar] [CrossRef] [PubMed]
  203. Versluys, K.A.; Eurich, D.T.; Marrie, T.J.; Forgie, S.; Tyrrell, G.J. Invasive pneumococcal disease and long-term outcomes in children: A 20-year population cohort study. Lancet Reg. Health—Am. 2022, 14, 100341. [Google Scholar] [CrossRef] [PubMed]
  204. Werren, J.P.; Troxler, L.J.; Oyewole, O.R.-A.; Ramette, A.; Brugger, S.D.; Bruggmann, R.; van der Linden, M.; Nahm, M.H.; Gjuroski, I.; Casanova, C.; et al. Carbon Source-Dependent Changes of the Structure of Streptococcus pneumoniae Capsular Polysaccharide with Serotype 6F. Int. J. Mol. Sci. 2021, 22, 4580. [Google Scholar] [CrossRef]
  205. Mathew, B.J.; Gupta, P.; Naaz, T.; Rai, R.; Gupta, S.; Gupta, S.; Chaurasiya, S.K.; Purwar, S.; Biswas, D.; Vyas, A.K.; et al. Role of Streptococcus pneumoniae extracellular glycosidases in immune evasion. Front. Cell. Infect. Microbiol. 2023, 13, 1109449. [Google Scholar] [CrossRef]
  206. Trappetti, C.; Kadioglu, A.; Carter, M.; Hayre, J.; Iannelli, F.; Pozzi, G.; Andrew, P.W.; Oggioni, M.R. Sialic acid: A preventable signal for pneumococcal biofilm formation, colonization, and invasion of the host. J. Infect. Dis. 2009, 199, 1497–1505. [Google Scholar] [CrossRef]
  207. Manco, S.; Hernon, F.; Yesilkaya, H.; Paton, J.C.; Andrew, P.W.; Kadioglu, A. Pneumococcal neuraminidases A and B both have essential roles during infection of the respiratory tract and sepsis. Infect. Immun. 2006, 74, 4014–4020. [Google Scholar] [CrossRef]
  208. 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, 1718–1721. [Google Scholar] [CrossRef]
  209. Agarwal, H.S.; Latifi, S.Q. Streptococcus Pneumoniae-Associated Hemolytic Uremic Syndrome in the Era of Pneumococcal Vaccine. Pathogens 2021, 10, 727. [Google Scholar] [CrossRef] [PubMed]
  210. Parker, D.; Soong, G.; Planet, P.; Brower, J.; Ratner, A.J.; Prince, A. The NanA neuraminidase of Streptococcus pneumoniae is involved in biofilm formation. Infect. Immun. 2009, 77, 3722–3730. [Google Scholar] [CrossRef] [PubMed]
  211. Ng, P.S.K.; Day, C.J.; Atack, J.M.; Hartley-Tassell, L.E.; Winter, L.E.; Marshanski, T.; Padler-Karavani, V.; Varki, A.; Barenkamp, S.J.; Apicella, M.A.; et al. Nontypeable Haemophilus influenzae Has Evolved Preferential Use of N-Acetylneuraminic Acid as a Host Adaptation. mBio 2019, 10, e00422-19. [Google Scholar] [CrossRef] [PubMed]
  212. Riesbeck, K. Complement evasion by the human respiratory tract pathogens Haemophilus influenzae and Moraxella catarrhalis. FEBS Lett. 2020, 594, 2586–2597. [Google Scholar] [CrossRef]
  213. Pittman, M. Variation and Type Specificity in the Bacterial Species Hemophilus influenzae. J. Exp. Med. 1931, 53, 471–492. [Google Scholar] [CrossRef]
  214. Paulo, A.C.; Lança, J.; Almeida, S.T.; Hilty, M.; Sá-Leão, R. The upper respiratory tract microbiota of healthy adults is affected by Streptococcus pneumoniae carriage, smoking habits, and contact with children. Microbiome 2023, 11, 199. [Google Scholar] [CrossRef]
  215. Langereis, J.D.; de Jonge, M.I. Invasive Disease Caused by Nontypeable Haemophilus influenzae. Emerg. Infect. Dis. 2015, 21, 1711–1718. [Google Scholar] [CrossRef]
  216. Landwehr, K.R.; Granland, C.M.; Martinovich, K.M.; Scott, N.M.; Seppanen, E.J.; Berry, L.; Strickland, D.; Fulurija, A.; Richmond, P.C.; Kirkham, L.-A.S. An infant mouse model of influenza-driven nontypeable Haemophilus influenzae colonization and acute otitis media suitable for preclinical testing of novel therapies. Infect. Immun. 2024, 92, e00453-23. [Google Scholar] [CrossRef]
  217. Oliver, S.E.; Rubis, A.B.; Soeters, H.M.; Reingold, A.; Barnes, M.; Petit, S.; Farley, M.M.; Harrison, L.H.; Como-Sabetti, K.; Khanlian, S.A.; et al. Epidemiology of Invasive Nontypeable Haemophilus influenzae Disease—United States, 2008–2019. Clin. Infect. Dis. 2023, 76, 1889–1895. [Google Scholar] [CrossRef]
  218. Van Eldere, J.; Slack, M.P.E.; Ladhani, S.; Cripps, A.W. Non-typeable Haemophilus influenzae, an under-recognised pathogen. Lancet Infect. Dis. 2014, 14, 1281–1292. [Google Scholar] [CrossRef] [PubMed]
  219. Collins, S.; Litt, D.J.; Flynn, S.; Ramsay, M.E.; Slack, M.P.E.; Ladhani, S.N. Neonatal Invasive Haemophilus influenzae Disease in England and Wales: Epidemiology, Clinical Characteristics, and Outcome. Clin. Infect. Dis. 2015, 60, 1786–1792. [Google Scholar] [CrossRef]
  220. Phillips, Z.N.; Brizuela, C.; Jennison, A.V.; Staples, M.; Grimwood, K.; Seib, K.L.; Jennings, M.P.; Atack, J.M. Analysis of Invasive Nontypeable Haemophilus influenzae Isolates Reveals Selection for the Expression State of Particular Phase-Variable Lipooligosaccharide Biosynthetic Genes. Infect. Immun. 2019, 87, e00093-19. [Google Scholar] [CrossRef]
  221. Ladhani, S.; Slack, M.P.E.; Heath, P.T.; von Gottberg, A.; Chandra, M.; Ramsay, M.E.; European Union Invasive Bacterial Infection Surveillance Participants. Invasive Haemophilus influenzae Disease, Europe, 1996–2006. Emerg. Infect. Dis. 2010, 16, 455–463. [Google Scholar] [CrossRef] [PubMed]
  222. Jackson, M.D.; Wong, S.M.; Akerley, B.J. Sialic Acid Protects Nontypeable Haemophilus influenzae from Natural IgM and Promotes Survival in Murine Respiratory Tract. Infect. Immun. 2021, 89, e00676-20. [Google Scholar] [CrossRef] [PubMed]
  223. Heise, T.; Langereis, J.D.; Rossing, E.; de Jonge, M.I.; Adema, G.J.; Büll, C.; Boltje, T.J. Selective Inhibition of Sialic Acid-Based Molecular Mimicry in Haemophilus influenzae Abrogates Serum Resistance. Cell Chem. Biol. 2018, 25, 1279–1285.e8. [Google Scholar] [CrossRef]
  224. Jenkins, G.A.; Figueira, M.; Kumar, G.A.; Sweetman, W.A.; Makepeace, K.; Pelton, S.I.; Moxon, R.; Hood, D.W. Sialic acid mediated transcriptional modulation of a highly conserved sialometabolism gene cluster in Haemophilus influenzae and its effect on virulence. BMC Microbiol. 2010, 10, 48. [Google Scholar] [CrossRef]
  225. Kariminik, A.; Baseri-Salehi, M.; Kheirkhah, B. Pseudomonas aeruginosa quorum sensing modulates immune responses: An updated review article. Immunol. Lett. 2017, 190, 1–6. [Google Scholar] [CrossRef]
  226. Estahbanati, H.K.; Kashani, P.P.; Ghanaatpisheh, F. Frequency of Pseudomonas aeruginosa serotypes in burn wound infections and their resistance to antibiotics. Burns J. Int. Soc. Burn Inj. 2002, 28, 340–348. [Google Scholar] [CrossRef]
  227. Mukherjee, K.; Khatua, B.; Mandal, C. Sialic Acid-Siglec-E Interactions During Pseudomonas aeruginosa Infection of Macrophages Interferes with Phagosome Maturation by Altering Intracellular Calcium Concentrations. Front. Immunol. 2020, 11, 332. [Google Scholar] [CrossRef]
  228. Khatua, B.; Ghoshal, A.; Bhattacharya, K.; Mandal, C.; Saha, B.; Crocker, P.R.; Mandal, C. Sialic acids acquired by Pseudomonas aeruginosa are involved in reduced complement deposition and siglec mediated host-cell recognition. Febs Lett. 2010, 584, 555–561. [Google Scholar] [CrossRef] [PubMed]
  229. Avril, T.; Wagner, E.R.; Willison, H.J.; Crocker, P.R. Sialic acid-binding immunoglobulin-like lectin 7 mediates selective recognition of sialylated glycans expressed on Campylobacter jejuni lipooligosaccharides. Infect. Immun. 2006, 74, 4133–4141. [Google Scholar] [CrossRef]
  230. Carlin, A.F.; Lewis, A.L.; Varki, A.; Nizet, V. Group B streptococcal capsular sialic acids interact with siglecs (immunoglobulin-like lectins) on human leukocytes. J. Bacteriol. 2007, 189, 1231–1237. [Google Scholar] [CrossRef]
  231. Crocker, P.R.; Paulson, J.C.; Varki, A. Siglecs and their roles in the immune system. Nat. Rev. Immunol. 2007, 7, 255–266. [Google Scholar] [CrossRef] [PubMed]
  232. Wang, Y. Sialidases from Clostridium perfringens and Their Inhibitors. Front. Cell. Infect. Microbiol. 2020, 9, 462. [Google Scholar] [CrossRef]
  233. Li, J.; Uzal, F.A.; McClane, B.A. Clostridium perfringens Sialidases: Potential Contributors to Intestinal Pathogenesis and Therapeutic Targets. Toxins 2016, 8, 341. [Google Scholar] [CrossRef]
  234. Yao, P.Y.; Annamaraju, P. Clostridium perfringens Infection. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  235. Schauer, R.; Sommer, U.; Krüger, D.; van Unen, H.; Traving, C. The Terminal Enzymes of Sialic Acid Metabolism: Acylneuraminate Pyruvate-Lyases. Biosci. Rep. 1999, 19, 373–383. [Google Scholar] [CrossRef]
  236. Navarro, M.A.; Li, J.; McClane, B.A.; Morrell, E.; Beingesser, J.; Uzal, F.A. NanI Sialidase Is an Important Contributor to Clostridium perfringens Type F Strain F4969 Intestinal Colonization in Mice. Infect. Immun. 2018, 86. [Google Scholar] [CrossRef]
  237. Camargo, A.; Ramírez, J.D.; Kiu, R.; Hall, L.J.; Muñoz, M. Unveiling the pathogenic mechanisms of Clostridium perfringens toxins and virulence factors. Emerg. Microbes Infect. 2024, 13, 2341968. [Google Scholar] [CrossRef]
  238. Cioffi, D.L.; Pandey, S.; Alvarez, D.F.; Cioffi, E.A. Terminal sialic acids are an important determinant of pulmonary endothelial barrier integrity. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2012, 302, L1067–L1077. [Google Scholar] [CrossRef]
  239. Li, X.; Xiong, Y.; Qing, G.; Jiang, G.; Li, X.; Sun, T.; Liang, X. Bioinspired Saccharide-Saccharide Interaction and Smart Polymer for Specific Enrichment of Sialylated Glycopeptides. ACS Appl. Mater. Interfaces 2016, 8, 13294–13302. [Google Scholar] [CrossRef]
  240. Li, J.; Sayeed, S.; Robertson, S.; Chen, J.; McClane, B.A. Sialidases affect the host cell adherence and epsilon toxin-induced cytotoxicity of Clostridium perfringens type D strain CN3718. PLoS Pathog. 2011, 7, e1002429. [Google Scholar] [CrossRef]
  241. 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, e00547-17. [Google Scholar] [CrossRef] [PubMed]
  242. Belcher, T.; Rollier, C.S.; Dold, C.; Ross, J.D.C.; MacLennan, C.A. Immune responses to Neisseria gonorrhoeae and implications for vaccine development. Front. Immunol. 2023, 14, 1248613. [Google Scholar] [CrossRef]
  243. Cardenas, A.J.; Thomas, K.S.; Broden, M.W.; Ferraro, N.J.; Pires, M.M.; John, C.M.; Jarvis, G.A.; Criss, A.K. Neisseria gonorrhoeae scavenges host sialic acid for Siglec-mediated, complement-independent suppression of neutrophil activation. mBio 2024, 15, e00119-24. [Google Scholar] [CrossRef]
  244. Kirkcaldy, R.D.; Weston, E.; Segurado, A.C.; Hughes, G. Epidemiology of gonorrhoea: A global perspective. Sex. Health 2019, 16, 401–411. [Google Scholar] [CrossRef] [PubMed]
  245. Handing, J.W.; Criss, A.K. The lipooligosaccharide-modifying enzyme LptA enhances gonococcal defence against human neutrophils. Cell. Microbiol. 2015, 17, 910–921. [Google Scholar] [CrossRef]
  246. Cartee, J.C.; Joseph, S.J.; Weston, E.; Pham, C.D.; Thomas, J.C., IV; Schlanger, K.; St Cyr, S.B.; Farley, M.M.; Moore, A.E.; Tunali, A.K.; et al. Phylogenomic Comparison of Neisseria gonorrhoeae Causing Disseminated Gonococcal Infections and Uncomplicated Gonorrhea in Georgia, United States. Open Forum Infect. Dis. 2022, 9, ofac247. [Google Scholar] [CrossRef]
  247. Gulati, S.; Cox, A.; Lewis, L.A.; St. Michael, F.; Li, J.; Boden, R.; Ram, S.; Rice, P.A. Enhanced Factor H Binding to Sialylated Gonococci Is Restricted to the Sialylated Lacto-N-Neotetraose Lipooligosaccharide Species: Implications for Serum Resistance and Evidence for a Bifunctional Lipooligosaccharide Sialyltransferase in Gonococci. Infect. Immun. 2005, 73, 7390–7397. [Google Scholar] [CrossRef]
  248. Jen, F.E.-C.; Ketterer, M.R.; Semchenko, E.A.; Day, C.J.; Seib, K.L.; Apicella, M.A.; Jennings, M.P. The Lst Sialyltransferase of Neisseria gonorrhoeae Can Transfer Keto-Deoxyoctanoate as the Terminal Sugar of Lipooligosaccharide: A Glyco-Achilles Heel That Provides a New Strategy for Vaccines to Prevent Gonorrhea. mBio 2021, 12, e03666-20. [Google Scholar] [CrossRef]
  249. Gil-Farina, I.; Schmidt, M. Interaction of vectors and parental viruses with the host genome. Curr. Opin. Virol. 2016, 21, 35–40. [Google Scholar] [CrossRef] [PubMed]
  250. Maginnis, M.S. Virus–Receptor Interactions: The Key to Cellular Invasion. J. Mol. Biol. 2018, 430, 2590–2611. [Google Scholar] [CrossRef] [PubMed]
  251. Wallace, L.E.; Liu, M.; van Kuppeveld, F.J.M.; de Vries, E.; de Haan, C.A.M. Respiratory mucus as a virus-host range determinant. Trends Microbiol. 2021, 29, 983–992. [Google Scholar] [CrossRef] [PubMed]
  252. Payne, S. 3—Virus interactions with the cell. In Viruses, 2nd ed.; Payne, S., Ed.; Academic Press: Cambridge, MA, USA, 2023; pp. 25–37. [Google Scholar]
  253. Reeves, J.D.; Doms, R.W. CHAPTER 33—The Role of Chemokine Receptors in HIV Infection of Host Cells. In Handbook of Cell Signaling; Bradshaw, R.A., Dennis, E.A., Eds.; Academic Press: Burlington, VT, USA, 2003; pp. 191–196. [Google Scholar]
  254. Nicholls, J.M.; Moss, R.B.; Haslam, S.M. The use of sialidase therapy for respiratory viral infections. Antiviral Res. 2013, 98, 401–409. [Google Scholar] [CrossRef]
  255. Modrow, S.; Falke, D.; Truyen, U.; Schätzl, H. Viruses with Single-Stranded, Segmented, Negative-Sense RNA Genomes. In Molecular Virology; Modrow, S., Falke, D., Truyen, U., Schätzl, H., Eds.; Springer: Berlin/Heidelberg, Germany, 2013; pp. 437–520. [Google Scholar]
  256. Li, X.; Gu, M.; Zheng, Q.; Gao, R.; Liu, X. Packaging signal of influenza A virus. Virol. J. 2021, 18, 36. [Google Scholar] [CrossRef]
  257. Hutchinson, E.C. Influenza Virus. Trends Microbiol. 2018, 26, 809–810. [Google Scholar] [CrossRef]
  258. Kumlin, U.; Olofsson, S.; Dimock, K.; Arnberg, N. Sialic acid tissue distribution and influenza virus tropism. Influenza Other Respir. Viruses 2008, 2, 147–154. [Google Scholar] [CrossRef]
  259. Chen, Z.; Chen, W.; Wang, Q.; Qin, Y.; Wang, X.; Ma, T.; Zhang, P.; Li, X.; Wang, X.; Ding, L.; et al. Comparative analysis of sialic acid α2–3/6 galactose glycan-binding proteins in human saliva and serum. J. Mol. Struct. 2021, 1230, 129859. [Google Scholar] [CrossRef]
  260. Liu, M.; Huang, L.Z.X.; Smits, A.A.; Büll, C.; Narimatsu, Y.; van Kuppeveld, F.J.M.; Clausen, H.; de Haan, C.A.M.; de Vries, E. Human-type sialic acid receptors contribute to avian influenza A virus binding and entry by hetero-multivalent interactions. Nat. Commun. 2022, 13, 4054. [Google Scholar] [CrossRef]
  261. Hatakeyama, S.; Sakai-Tagawa, Y.; Kiso, M.; Goto, H.; Kawakami, C.; Mitamura, K.; Sugaya, N.; Suzuki, Y.; Kawaoka, Y. Enhanced Expression of an α2,6-Linked Sialic Acid on MDCK Cells Improves Isolation of Human Influenza Viruses and Evaluation of Their Sensitivity to a Neuraminidase Inhibitor. J. Clin. Microbiol. 2005, 43, 4139–4146. [Google Scholar] [CrossRef]
  262. Dawre, S.; Maru, S. Human respiratory viral infections: Current status and future prospects of nanotechnology-based approaches for prophylaxis and treatment. Life Sci. 2021, 278, 119561. [Google Scholar] [CrossRef] [PubMed]
  263. Krammer, F.; Smith, G.J.D.; Fouchier, R.A.M.; Peiris, M.; Kedzierska, K.; Doherty, P.C.; Palese, P.; Shaw, M.L.; Treanor, J.; Webster, R.G.; et al. Influenza. Nat. Rev. Dis. Primer 2018, 4, 3. [Google Scholar] [CrossRef]
  264. Javanian, M.; Barary, M.; Ghebrehewet, S.; Koppolu, V.; Vasigala, V.; Ebrahimpour, S. A brief review of influenza virus infection. J. Med. Virol. 2021, 93, 4638–4646. [Google Scholar] [CrossRef] [PubMed]
  265. Sulpiana; Amalia, R.; Atik, N. The Roles of Endocytosis and Autophagy at the Cellular Level During Influenza Virus Infection: A Mini-Review. Infect. Drug Resist. 2024, 17, 3199–3208. [Google Scholar] [CrossRef]
  266. McAuley, J.L.; Gilbertson, B.P.; Trifkovic, S.; Brown, L.E.; McKimm-Breschkin, J.L. Influenza Virus Neuraminidase Structure and Functions. Front. Microbiol. 2019, 10, 39. [Google Scholar] [CrossRef]
  267. Gallagher, J.R.; McCraw, D.M.; Torian, U.; Gulati, N.M.; Myers, M.L.; Conlon, M.T.; Harris, A.K. Characterization of Hemagglutinin Antigens on Influenza Virus and within Vaccines Using Electron Microscopy. Vaccines 2018, 6, 31. [Google Scholar] [CrossRef] [PubMed]
  268. Gottschalk, A. The Influenza Virus Neuraminidase. Nature 1958, 181, 377–378. [Google Scholar] [CrossRef]
  269. Wu, N.C.; Wilson, I.A. Influenza Hemagglutinin Structures and Antibody Recognition. Cold Spring Harb. Perspect. Med. 2020, 10, a038778. [Google Scholar] [CrossRef]
  270. Fitz, W.; Rosenthal, P.B.; Wong, C.H. Synthesis and inhibitory properties of a thiomethylmercuric sialic acid with application to the X-ray structure determination of 9-O-acetylsialic acid esterase from influenza C virus. Bioorg. Med. Chem. 1996, 4, 1349–1353. [Google Scholar] [CrossRef]
  271. Rossman, J.S.; Leser, G.P.; Lamb, R.A. Filamentous Influenza Virus Enters Cells via Macropinocytosis. J. Virol. 2012, 86, 10950–10960. [Google Scholar] [CrossRef]
  272. de Vries, E.; Tscherne, D.M.; Wienholts, M.J.; Cobos-Jiménez, V.; Scholte, F.; García-Sastre, A.; Rottier, P.J.M.; de Haan, C.A.M. Dissection of the Influenza A Virus Endocytic Routes Reveals Macropinocytosis as an Alternative Entry Pathway. PLOS Pathog. 2011, 7, e1001329. [Google Scholar] [CrossRef]
  273. Matrosovich, M.N.; Matrosovich, T.Y.; Gray, T.; Roberts, N.A.; Klenk, H.-D. Neuraminidase Is Important for the Initiation of Influenza Virus Infection in Human Airway Epithelium. J. Virol. 2004, 78, 12665–12667. [Google Scholar] [CrossRef]
  274. Ohuchi, M.; Asaoka, N.; Sakai, T.; Ohuchi, R. Roles of neuraminidase in the initial stage of influenza virus infection. Microbes Infect. 2006, 8, 1287–1293. [Google Scholar] [CrossRef] [PubMed]
  275. Giurgea, L.T.; Morens, D.M.; Taubenberger, J.K.; Memoli, M.J. Influenza Neuraminidase: A Neglected Protein and Its Potential for a Better Influenza Vaccine. Vaccines 2020, 8, 409. [Google Scholar] [CrossRef]
  276. Escuret, V.; Terrier, O. Co-infection of the respiratory epithelium, scene of complex functional interactions between viral, bacterial, and human neuraminidases. Front. Microbiol. 2023, 14, 1137336. [Google Scholar] [CrossRef]
  277. Peltola, V.T.; Murti, K.G.; McCullers, J.A. Influenza Virus Neuraminidase Contributes to Secondary Bacterial Pneumonia. J. Infect. Dis. 2005, 192, 249–257. [Google Scholar] [CrossRef]
  278. Creytens, S.; Pascha, M.N.; Ballegeer, M.; Saelens, X.; de Haan, C.A.M. Influenza Neuraminidase Characteristics and Potential as a Vaccine Target. Front. Immunol. 2021, 12, 786617. [Google Scholar] [CrossRef]
  279. Eurosurveillance Editorial Team. Note from the editors: World Health Organization declares novel coronavirus (2019-nCoV) sixth public health emergency of international concern. Eurosurveillance 2020, 25, 200131e. [Google Scholar] [CrossRef]
  280. Majumder, J.; Minko, T. Recent Developments on Therapeutic and Diagnostic Approaches for COVID-19. AAPS J. 2021, 23, 14. [Google Scholar] [CrossRef]
  281. Su, S.; Wong, G.; Shi, W.; Liu, J.; Lai, A.C.K.; Zhou, J.; Liu, W.; Bi, Y.; Gao, G.F. Epidemiology, Genetic Recombination, and Pathogenesis of Coronaviruses. Trends Microbiol. 2016, 24, 490–502. [Google Scholar] [CrossRef]
  282. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef]
  283. Wrapp, D.; Wang, N.; Corbett, K.S.; Goldsmith, J.A.; Hsieh, C.-L.; Abiona, O.; Graham, B.S.; McLellan, J.S. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020, 367, 1260–1263. [Google Scholar] [CrossRef]
  284. Casalino, L.; Gaieb, Z.; Goldsmith, J.A.; Hjorth, C.K.; Dommer, A.C.; Harbison, A.M.; Fogarty, C.A.; Barros, E.P.; Taylor, B.C.; McLellan, J.S.; et al. Beyond Shielding: The Roles of Glycans in the SARS-CoV-2 Spike Protein. ACS Cent. Sci. 2020, 6, 1722–1734. [Google Scholar] [CrossRef]
  285. Tortorici, M.A.; Walls, A.C.; Lang, Y.; Wang, C.; Li, Z.; Koerhuis, D.; Boons, G.-J.; Bosch, B.-J.; Rey, F.A.; de Groot, R.J.; et al. Structural basis for human coronavirus attachment to sialic acid receptors. Nat. Struct. Mol. Biol. 2019, 26, 481–489. [Google Scholar] [CrossRef]
  286. Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef]
  287. Milanetti, E.; Miotto, M.; Di Rienzo, L.; Nagaraj, M.; Monti, M.; Golbek, T.W.; Gosti, G.; Roeters, S.J.; Weidner, T.; Otzen, D.E.; et al. In-Silico Evidence for a Two Receptor Based Strategy of SARS-CoV-2. Front. Mol. Biosci. 2021, 8, 690655. [Google Scholar] [CrossRef]
  288. Vandelli, A.; Monti, M.; Milanetti, E.; Armaos, A.; Rupert, J.; Zacco, E.; Bechara, E.; Delli Ponti, R.; Tartaglia, G.G. Structural analysis of SARS-CoV-2 genome and predictions of the human interactome. Nucleic Acids Res. 2020, 48, 11270–11283. [Google Scholar] [CrossRef]
  289. Pronker, M.F.; Creutznacher, R.; Drulyte, I.; Hulswit, R.J.G.; Li, Z.; van Kuppeveld, F.J.M.; Snijder, J.; Lang, Y.; Bosch, B.-J.; Boons, G.-J.; et al. Sialoglycan binding triggers spike opening in a human coronavirus. Nature 2023, 624, 201–206. [Google Scholar] [CrossRef]
  290. Maity, S.; Acharya, A. Many Roles of Carbohydrates: A Computational Spotlight on the Coronavirus S Protein Binding. ACS Appl. Bio Mater. 2024, 7, 646–656. [Google Scholar] [CrossRef]
  291. Awasthi, M.; Gulati, S.; Sarkar, D.P.; Tiwari, S.; Kateriya, S.; Ranjan, P.; Verma, S.K. The Sialoside-Binding Pocket of SARS-CoV-2 Spike Glycoprotein Structurally Resembles MERS-CoV. Viruses 2020, 12, 909. [Google Scholar] [CrossRef]
  292. Baker, A.N.; Richards, S.-J.; Guy, C.S.; Congdon, T.R.; Hasan, M.; Zwetsloot, A.J.; Gallo, A.; Lewandowski, J.R.; Stansfeld, P.J.; Straube, A.; et al. The SARS-COV-2 Spike Protein Binds Sialic Acids and Enables Rapid Detection in a Lateral Flow Point of Care Diagnostic Device. ACS Cent. Sci. 2020, 6, 2046–2052. [Google Scholar] [CrossRef]
  293. Nguyen, L.; McCord, K.A.; Bui, D.T.; Bouwman, K.M.; Kitova, E.N.; Elaish, M.; Kumawat, D.; Daskhan, G.C.; Tomris, I.; Han, L.; et al. Sialic acid-containing glycolipids mediate binding and viral entry of SARS-CoV-2. Nat. Chem. Biol. 2022, 18, 81–90. [Google Scholar] [CrossRef]
  294. Cavezzi, A.; Menicagli, R.; Troiani, E.; Corrao, S. COVID-19, Cation Dysmetabolism, Sialic Acid, CD147, ACE2, Viroporins, Hepcidin and Ferroptosis: A Possible Unifying Hypothesis. F1000Research 2022, 11, 102. [Google Scholar] [CrossRef]
  295. Jiang, X.; Tan, M.; Xia, M.; Huang, P.; Kennedy, M.A. Intra-species sialic acid polymorphism in humans: A common niche for influenza and coronavirus pandemics? Emerg. Microbes Infect. 2021, 10, 1191–1199. [Google Scholar] [CrossRef]
  296. Dhar, C.; Sasmal, A.; Diaz, S.; Verhagen, A.; Yu, H.; Li, W.; Chen, X.; Varki, A. Are sialic acids involved in COVID-19 pathogenesis? Glycobiology 2021, 31, 1068–1071. [Google Scholar] [CrossRef]
  297. Unione, L.; Moure, M.J.; Lenza, M.P.; Oyenarte, I.; Ereño-Orbea, J.; Ardá, A.; Jiménez-Barbero, J. The SARS-CoV-2 Spike Glycoprotein Directly Binds Exogeneous Sialic Acids: A NMR View. Angew. Chem. 2022, 134, e202201432. [Google Scholar] [CrossRef]
  298. Frąc, M.; Hannula, S.E.; Bełka, M.; Jędryczka, M. Fungal Biodiversity and Their Role in Soil Health. Front. Microbiol. 2018, 9, 707. [Google Scholar] [CrossRef]
  299. Belvoncikova, P.; Splichalova, P.; Videnska, P.; Gardlik, R. The Human Mycobiome: Colonization, Composition and the Role in Health and Disease. J. Fungi 2022, 8, 1046. [Google Scholar] [CrossRef]
  300. Köhler, J.R.; Casadevall, A.; Perfect, J. The Spectrum of Fungi That Infects Humans. Cold Spring Harb. Perspect. Med. 2015, 5, a019273. [Google Scholar] [CrossRef]
  301. Rodrigues, M.L.; Dobroff, A.S.; Couceiro, J.N.D.S.; Alviano, C.S.; Schauer, R.; Travassos, L.R. Sialylglycoconjugates and sialyltransferase activity in the fungus Cryptococcus neoformans. Glycoconj. J. 2002, 19, 165–173. [Google Scholar] [CrossRef]
  302. Bose, I.; Reese, A.J.; Ory, J.J.; Janbon, G.; Doering, T.L. A Yeast under Cover: The Capsule of Cryptococcus neoformans. Eukaryot. Cell 2003, 2, 655–663. [Google Scholar] [CrossRef] [PubMed]
  303. Warwas, M.L.; Watson, J.N.; Bennet, A.J.; Moore, M.M. Structure and role of sialic acids on the surface of Aspergillus fumigatus conidiospores. Glycobiology 2007, 17, 401–410. [Google Scholar] [CrossRef]
  304. Alviano, C.S.; Travassos, L.R.; Schauer, R. Sialic acids in fungi. Glycoconj. J. 1999, 16, 545–554. [Google Scholar] [CrossRef]
  305. Maziarz, E.K.; Perfect, J.R. Cryptococcosis. Infect. Dis. Clin. North Am. 2016, 30, 179–206. [Google Scholar] [CrossRef]
  306. Horwath, M.C.; Fecher, R.A.; Deepe, G.S. Histoplasma capsulatum, lung infection and immunity. Future Microbiol. 2015, 10, 967–975. [Google Scholar] [CrossRef]
  307. Sil, A.; Andrianopoulos, A. Thermally Dimorphic Human Fungal Pathogens—Polyphyletic Pathogens with a Convergent Pathogenicity Trait. Cold Spring Harb. Perspect. Med. 2015, 5, a019794. [Google Scholar] [CrossRef] [PubMed]
  308. Valdez, A.F.; Miranda, D.Z.; Guimarães, A.J.; Nimrichter, L.; Nosanchuk, J.D. Pathogenicity & virulence of Histoplasma capsulatum—A multifaceted organism adapted to intracellular environments. Virulence 2022, 13, 1900. [Google Scholar] [CrossRef]
  309. Burgess, T.B.; Condliffe, A.M.; Elks, P.M. A Fun-Guide to Innate Immune Responses to Fungal Infections. J. Fungi 2022, 8, 805. [Google Scholar] [CrossRef]
  310. Erwig, L.P.; Gow, N.A.R. Interactions of fungal pathogens with phagocytes. Nat. Rev. Microbiol. 2016, 14, 163–176. [Google Scholar] [CrossRef]
  311. Guimaraes, A.J.; de Cerqueira, M.D.; Nosanchuk, J.D. Surface Architecture of Histoplasma Capsulatum. Front. Microbiol. 2011, 2, 225. [Google Scholar] [CrossRef]
  312. Uribe-Querol, E.; Rosales, C. Control of Phagocytosis by Microbial Pathogens. Front. Immunol. 2017, 8, 1368. [Google Scholar] [CrossRef] [PubMed]
  313. Mittal, J.; Ponce, M.G.; Gendlina, I.; Nosanchuk, J.D. Histoplasma Capsulatum: Mechanisms for Pathogenesis. Curr. Top. Microbiol. Immunol. 2019, 422, 157–191. [Google Scholar] [CrossRef]
  314. Huang, S.; Deepe, G.S., Jr. Notch regulates Histoplasma capsulatum clearance in mouse lungs during innate and adaptive immune response phases in primary infection. J. Leukoc. Biol. 2022, 112, 1137–1154. [Google Scholar] [CrossRef] [PubMed]
  315. Sharon, N. Carbohydrates as recognition determinants in phagocytosis and in lectin-mediated killing of target cells. Biol. Cell 1984, 51, 239–245. [Google Scholar] [CrossRef] [PubMed]
  316. Ielasi, F.S.; Alioscha-Perez, M.; Donohue, D.; Claes, S.; Sahli, H.; Schols, D.; Willaert, R.G. Lectin-Glycan Interaction Network-Based Identification of Host Receptors of Microbial Pathogenic Adhesins. mBio 2016, 7, e00584-16. [Google Scholar] [CrossRef]
  317. Singh, R.S.; Bhari, R.; Rana, V.; Tiwary, A.K. Immunomodulatory and therapeutic potential of a mycelial lectin from Aspergillus nidulans. Appl. Biochem. Biotechnol. 2011, 165, 624–638. [Google Scholar] [CrossRef]
  318. Duarte-Escalante, E.; Zenteno, E.; Taylor, M.L. Interaction of Histoplasma capsulatum Yeasts with Galactosylated Surface Molecules of Murine Macrophages. Arch. Med. Res. 2003, 34, 176–183. [Google Scholar] [CrossRef] [PubMed]
  319. Taylor, M.L.; Duarte-Escalante, E.; Pérez, A.; Zenteno, E.; Toriello, C. Histoplasma capsulatum yeast cells attach and agglutinate human erythrocytes. Med. Mycol. 2004, 42, 287–292. [Google Scholar] [CrossRef]
  320. Lehmann, F.; Tiralongo, E.; Tiralongo, J. Sialic acid-specific lectins: Occurrence, specificity and function. Cell. Mol. Life Sci. CMLS 2006, 63, 1331–1354. [Google Scholar] [CrossRef]
  321. Talapko, J.; Juzbašić, M.; Matijević, T.; Pustijanac, E.; Bekić, S.; Kotris, I.; Škrlec, I. Candida albicans—The Virulence Factors and Clinical Manifestations of Infection. J. Fungi 2021, 7, 79. [Google Scholar] [CrossRef]
  322. Sonesson, A.; Bartosik, J.; Christiansen, J.; Roscher, I.; Nilsson, F.; Schmidtchen, A.; Bäck, O. Sensitization to skin-associated microorganisms in adult patients with atopic dermatitis is of importance for disease severity. Acta Derm. Venereol. 2013, 93, 340–345. [Google Scholar] [CrossRef]
  323. Kobiela, A.; Frackowiak, J.E.; Biernacka, A.; Hovhannisyan, L.; Bogucka, A.E.; Panek, K.; Paul, A.A.; Lukomska, J.; Wang, X.; Giannoulatou, E.; et al. Exposure of Keratinocytes to Candida Albicans in the Context of Atopic Milieu Induces Changes in the Surface Glycosylation Pattern of Small Extracellular Vesicles to Enhance Their Propensity to Interact with Inhibitory Siglec Receptors. Front. Immunol. 2022, 13, 884530. [Google Scholar] [CrossRef]
  324. Flevari, A.; Theodorakopoulou, M.; Velegraki, A.; Armaganidis, A.; Dimopoulos, G. Treatment of invasive candidiasis in the elderly: A review. Clin. Interv. Aging 2013, 8, 1199–1208. [Google Scholar] [CrossRef]
  325. R, A.N.; Rafiq, N.B. Candidiasis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  326. Valentijn-Benz, M.; Nazmi, K.; Brand, H.S.; van’t Hof, W.; Veerman, E.C.I. Growth of Candida albicans in human saliva is supported by low-molecular-mass compounds. FEMS Yeast Res. 2015, 15, fov088. [Google Scholar] [CrossRef]
  327. Lopes, J.P.; Lionakis, M.S. Pathogenesis and virulence of Candida albicans. Virulence 2022, 13, 89–121. [Google Scholar] [CrossRef]
  328. Masuoka, J. Surface Glycans of Candida albicans and Other Pathogenic Fungi: Physiological Roles, Clinical Uses, and Experimental Challenges. Clin. Microbiol. Rev. 2004, 17, 281–310. [Google Scholar] [CrossRef]
  329. Soares, R.M.A.; de A. Soares, R.M.; Alviano, D.S.; Angluster, J.; Alviano, C.S.; Travassos, L.R. Identification of sialic acids on the cell surface of Candida albicans. Biochim. Biophys. Acta BBA—Gen. Subj. 2000, 1474, 262–268. [Google Scholar] [CrossRef]
  330. Roggentin, P.; Krug, G.; Schauer, R.; Brasch, J. Lack of sialidase activity in Candida albicans and Candida glabrata. Mycoses 1999, 42, 33–36. [Google Scholar] [CrossRef]
  331. Theel, E.S.; Pritt, B.S. Parasites. Microbiol. Spectr. 2016, 4, DMIH2-0013-2015. [Google Scholar] [CrossRef]
  332. Iwasaki, A.; Medzhitov, R. Control of adaptive immunity by the innate immune system. Nat. Immunol. 2015, 16, 343–353. [Google Scholar] [CrossRef]
  333. Coakley, G.; Buck, A.H.; Maizels, R.M. Host parasite communications—Messages from helminths for the immune system: Parasite communication and cell-cell interactions. Sens. Signal. Parasit. 2016, 208, 33–40. [Google Scholar] [CrossRef]
  334. Agusti, R.; Gallo-Rodriguez, C.; de Lederkremer, R.M. Trypanosoma cruzi trans-sialidase. A tool for the synthesis of sialylated oligosaccharides. Carbohydr. Res. 2019, 479, 48–58. [Google Scholar] [CrossRef]
  335. Schmid-Hempel, P. Immune defence, parasite evasion strategies and their relevance for ‘macroscopic phenomena’ such as virulence. Philos. Trans. R. Soc. B Biol. Sci. 2008, 364, 85–98. [Google Scholar] [CrossRef]
  336. Useche, Y.; Pérez, A.R.; de Meis, J.; Bonomo, A.; Savino, W. Central nervous system commitment in Chagas disease. Front. Immunol. 2022, 13, 975106. [Google Scholar] [CrossRef]
  337. da Silva Oliveira Barbosa, E.; Roggero, E.A.; González, F.B.; Fernández, R.D.V.; Carvalho, V.F.; Bottasso, O.A.; Pérez, A.R.; Villar, S.R. Evidence in Favor of an Alternative Glucocorticoid Synthesis Pathway During Acute Experimental Chagas Disease. Front. Endocrinol. 2020, 10, 866. [Google Scholar] [CrossRef]
  338. González, F.B.; Savino, W.; Pérez, A.R. Brain-Thymus Connections in Chagas Disease. Neuroimmunomodulation 2024, 31, 78–88. [Google Scholar] [CrossRef]
  339. Cardoso, M.S.; Reis-Cunha, J.L.; Bartholomeu, D.C. Evasion of the Immune Response by Trypanosoma cruzi during Acute Infection. Front. Immunol. 2016, 6, 659. [Google Scholar] [CrossRef]
  340. Savino, W.; Villa-Verde, D.M.; Mendes-da-Cruz, D.A.; Silva-Monteiro, E.; Perez, A.R.; Aoki Mdel, P.; Bottasso, O.; Guiñazú, N.; Silva-Barbosa, S.D.; Gea, S. Cytokines and cell adhesion receptors in the regulation of immunity to Trypanosoma cruzi. Cytokines Coming Age S. Am. 2007, 18, 107–124. [Google Scholar] [CrossRef]
  341. Flávia Nardy, A.; Freire-de-Lima, C.G.; Morrot, A. Immune Evasion Strategies of Trypanosoma cruzi. J. Immunol. Res. 2015, 2015, 178947. [Google Scholar] [CrossRef]
  342. Muiá, R.; Yu, H.; Prescher, J.; Hellman, U.; Bertozzi, C.; Campetella, O. Identification of glycoproteins targeted by Trypanosoma cruzi trans-sialidase, a virulence factor that disturbs lymphocyte glycosylation. Glycobiology 2010, 20, 833–842. [Google Scholar] [CrossRef]
  343. Piacenza, L.; Peluffo, G.; Alvarez, M.N.; Kelly, J.M.; Wilkinson, S.R.; Radi, R. Peroxiredoxins play a major role in protecting Trypanosoma cruzi against macrophage- and endogenously-derived peroxynitrite. Biochem. J. 2008, 410, 359–368. [Google Scholar] [CrossRef] [PubMed]
  344. Martín-Escolano, J.; Marín, C.; Rosales, M.J.; Tsaousis, A.D.; Medina-Carmona, E.; Martín-Escolano, R. An Updated View of the Trypanosoma cruzi Life Cycle: Intervention Points for an Effective Treatment. ACS Infect. Dis. 2022, 8, 1107–1115. [Google Scholar] [CrossRef]
  345. Schacht, P.; Arcieri, G.; Hullmann, R. Safety of oral ciprofloxacin: An update based on clinical trial results. Am. J. Med. 1989, 87, S98–S102. [Google Scholar] [CrossRef]
  346. Adasme, M.F.; Bolz, S.N.; Adelmann, L.; Salentin, S.; Haupt, V.J.; Moreno-Rodríguez, A.; Nogueda-Torres, B.; Castillo-Campos, V.; Yepez-Mulia, L.; De Fuentes-Vicente, J.A.; et al. Repositioned Drugs for Chagas Disease Unveiled via Structure-Based Drug Repositioning. Int. J. Mol. Sci. 2020, 21, 8809. [Google Scholar] [CrossRef]
  347. Goerdeler, F.; Seeberger, P.H.; Moscovitz, O. Unveiling the Sugary Secrets of Plasmodium Parasites. Front. Microbiol. 2021, 12, 712538. [Google Scholar] [CrossRef]
  348. Fact Sheet About Malaria. Available online: https://www.who.int/news-room/fact-sheets/detail/malaria (accessed on 28 January 2025).
  349. Hanboonkunupakarn, B.; White, N.J. The threat of antimalarial drug resistance. Trop. Dis. Travel Med. Vaccines 2016, 2, 1–5. [Google Scholar] [CrossRef]
  350. Rasmussen, C.; Alonso, P.; Ringwald, P. Current and emerging strategies to combat antimalarial resistance. Expert Rev. Anti Infect. Ther. 2022, 20, 353–372. [Google Scholar] [CrossRef]
  351. Chuang, Y.-M.; Agunbiade, T.A.; Tang, X.-D.; Freudzon, M.; Almeras, L.; Fikrig, E. The Effects of A Mosquito Salivary Protein on Sporozoite Traversal of Host Cells. J. Infect. Dis. 2020, 224, 544–553. [Google Scholar] [CrossRef]
  352. Suwanarusk, R.; Cooke, B.M.; Dondorp, A.M.; Silamut, K.; Sattabongkot, J.; White, N.J.; Udomsangpetch, R. The deformability of red blood cells parasitized by Plasmodium falciparum and P. vivax. J. Infect. Dis. 2004, 189, 190–194. [Google Scholar] [CrossRef]
  353. Valenciano, A.L.; Gomez-Lorenzo, M.G.; Vega-Rodríguez, J.; Adams, J.H.; Roth, A. In vitro models for human malaria: Targeting the liver stage. Trends Parasitol. 2022, 38, 758–774. [Google Scholar] [CrossRef]
  354. Deroost, K.; Pham, T.-T.; Opdenakker, G.; Van den Steen, P.E. The immunological balance between host and parasite in malaria. FEMS Microbiol. Rev. 2016, 40, 208–257. [Google Scholar] [CrossRef] [PubMed]
  355. Goldberg, D.E.; Zimmerberg, J. Hardly Vacuous: The Parasitophorous Vacuolar Membrane of Malaria Parasites. Trends Parasitol. 2020, 36, 138–146. [Google Scholar] [CrossRef] [PubMed]
  356. Molina-Franky, J.; Patarroyo, M.E.; Kalkum, M.; Patarroyo, M.A. The Cellular and Molecular Interaction Between Erythrocytes and Plasmodium falciparum Merozoites. Front. Cell. Infect. Microbiol. 2022, 12, 816574. [Google Scholar] [CrossRef]
  357. Cowman, A.F.; Crabb, B.S. Invasion of Red Blood Cells by Malaria Parasites. Cell 2006, 124, 755–766. [Google Scholar] [CrossRef]
  358. Ezema, C.A.; Okagu, I.U.; Ezeorba, T.P.C. Escaping the enemy’s bullets: An update on how malaria parasites evade host immune response. Parasitol. Res. 2023, 122, 1715–1731. [Google Scholar] [CrossRef]
  359. Venugopal, K.; Hentzschel, F.; Valkiūnas, G.; Marti, M. Plasmodium asexual growth and sexual development in the haematopoietic niche of the host. Nat. Rev. Microbiol. 2020, 18, 177–189. [Google Scholar] [CrossRef] [PubMed]
  360. Butcher, G.A.; Mitchell, G.H.; Cohen, S. Mechanism of Host Specificity in Malarial Infection. Nature 1973, 244, 40–42. [Google Scholar] [CrossRef]
  361. Habtamu, K.; Petros, B.; Yan, G. Plasmodium vivax: The potential obstacles it presents to malaria elimination and eradication. Trop. Dis. Travel Med. Vaccines 2022, 8, 27. [Google Scholar] [CrossRef]
  362. Jaskiewicz, E.; Jodłowska, M.; Kaczmarek, R.; Zerka, A. Erythrocyte glycophorins as receptors for Plasmodium merozoites. Parasit. Vectors 2019, 12, 317. [Google Scholar] [CrossRef]
  363. Alves-Rosa, M.F.; Tayler, N.M.; Dorta, D.; Coronado, L.M.; Spadafora, C.P. falciparum Invasion and Erythrocyte Aging. Cells 2024, 13, 334. [Google Scholar] [CrossRef]
  364. Moskovitz, R.; Pholcharee, T.; DonVito, S.M.; Guloglu, B.; Lowe, E.; Mohring, F.; Moon, R.W.; Higgins, M.K. Structural basis for DARC binding in reticulocyte invasion by Plasmodium vivax. Nat. Commun. 2023, 14, 3637. [Google Scholar] [CrossRef] [PubMed]
  365. Day, C.J.; Favuzza, P.; Bielfeld, S.; Haselhorst, T.; Seefeldt, L.; Hauser, J.; Shewell, L.K.; Flueck, C.; Poole, J.; Jen, F.E.-C.; et al. The essential malaria protein PfCyRPA targets glycans to invade erythrocytes. Cell Rep. 2024, 43, 114012. [Google Scholar] [CrossRef]
  366. Yang, P.-K.; Liang, X.-Y.; Lin, M.; Chen, J.-T.; Huang, H.-Y.; Lin, L.-Y.; Ehapo, C.S.; Eyi, U.M.; Zheng, Y.-Z.; Xie, D.-D.; et al. Population genetic analysis of the Plasmodium falciparum erythrocyte binding antigen-175 (EBA-175) gene in Equatorial Guinea. Malar. J. 2021, 20, 374. [Google Scholar] [CrossRef]
  367. Jiang, L.; Gaur, D.; Mu, J.; Zhou, H.; Long, C.A.; Miller, L.H. Evidence for erythrocyte-binding antigen 175 as a component of a ligand-blocking blood-stage malaria vaccine. Proc. Natl. Acad. Sci. USA 2011, 108, 7553–7558. [Google Scholar] [CrossRef] [PubMed]
  368. Takashima, E.; Otsuki, H.; Morita, M.; Ito, D.; Nagaoka, H.; Yuguchi, T.; Hassan, I.; Tsuboi, T. The Need for Novel Asexual Blood-Stage Malaria Vaccine Candidates for Plasmodium falciparum. Biomolecules 2024, 14, 100. [Google Scholar] [CrossRef]
  369. Estrada-Martínez, L.E.; Moreno-Celis, U.; Cervantes-Jiménez, R.; Ferriz-Martínez, R.A.; Blanco-Labra, A.; García-Gasca, T. Plant lectins as medical tools against digestive system cancers. Int. J. Mol. Sci. 2017, 18, 1403. [Google Scholar] [CrossRef] [PubMed]
  370. Serdar, Z.; Yeşilbursa, D.; Dirican, M.; Sarandöl, E.; Serdar, A. Sialic acid and oxidizability of lipid and proteins and antioxidant status in patients with coronary artery disease. Cell Biochem. Funct. 2007, 25, 655–664. [Google Scholar] [CrossRef]
  371. Volkhina, I.V.; Butolin, E.G. Clinical and Diagnostic Significance of Sialic Acids Determination in Biological Material. Biochem. Mosc. Suppl. Ser. B Biomed. Chem. 2022, 16, 165–174. [Google Scholar] [CrossRef]
  372. Roth, G.A.; Mensah, G.A.; Fuster, V. The Global Burden of Cardiovascular Diseases and Risks: A Compass for Global Action. J. Am. Coll. Cardiol. 2020, 76, 2980–2981. [Google Scholar] [CrossRef]
  373. Teo, K.K.; Rafiq, T. Cardiovascular Risk Factors and Prevention: A Perspective From Developing Countries. Can. J. Cardiol. 2021, 37, 733–743. [Google Scholar] [CrossRef]
  374. Cocciolone, A.J.; Hawes, J.Z.; Staiculescu, M.C.; Johnson, E.O.; Murshed, M.; Wagenseil, J.E. Elastin, arterial mechanics, and cardiovascular disease. Am. J. Physiol.-Heart Circ. Physiol. 2018, 315, H189–H205. [Google Scholar] [CrossRef]
  375. Milusev, A.; Rieben, R.; Sorvillo, N. The Endothelial Glycocalyx: A Possible Therapeutic Target in Cardiovascular Disorders. Front. Cardiovasc. Med. 2022, 9, 897087. [Google Scholar] [CrossRef]
  376. D’Addio, M.; Frey, J.; Otto, V.I. The manifold roles of sialic acid for the biological functions of endothelial glycoproteins. Glycobiology 2020, 30, 490–499. [Google Scholar] [CrossRef]
  377. Wei, M.; Wang, P.G. Desialylation in physiological and pathological processes: New target for diagnostic and therapeutic development. In Progress in Molecular Biology and Translational Science; Elsevier: Amsterdam, The Netherlands, 2019; Volume 162, pp. 25–57. [Google Scholar] [CrossRef]
  378. Gokmen, S.S.; Kilicli, G.; Ozcelik, F.; Ture, M.; Gulen, S. Association between serum total and lipid-bound sialic acid concentration and the severity of coronary atherosclerosis. J. Lab. Clin. Med. 2002, 140, 110–118. [Google Scholar] [CrossRef] [PubMed]
  379. Mezentsev, A.; Bezsonov, E.; Kashirskikh, D.; Baig, M.S.; Eid, A.H.; Orekhov, A. Proatherogenic Sialidases and Desialylated Lipoproteins: 35 Years of Research and Current State from Bench to Bedside. Biomedicines 2021, 9, 600. [Google Scholar] [CrossRef] [PubMed]
  380. Kawanishi, K.; Coker, J.K.; Grunddal, K.V.; Dhar, C.; Hsiao, J.; Zengler, K.; Varki, N.; Varki, A.; Gordts, P.L.S.M. Dietary Neu5Ac Intervention Protects Against Atherosclerosis Associated with Human-Like Neu5Gc Loss—Brief Report. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 2730–2739. [Google Scholar] [CrossRef] [PubMed]
  381. Li, M.-N.; Qian, S.-H.; Yao, Z.-Y.; Ming, S.-P.; Shi, X.-J.; Kang, P.-F.; Zhang, N.-R.; Wang, X.-J.; Gao, D.-S.; Gao, Q.; et al. Correlation of serum N-Acetylneuraminic acid with the risk and prognosis of acute coronary syndrome: A prospective cohort study. BMC Cardiovasc. Disord. 2020, 20, 404. [Google Scholar] [CrossRef]
  382. Zhang, L.; Wei, T.-T.; Li, Y.; Li, J.; Fan, Y.; Huang, F.-Q.; Cai, Y.-Y.; Ma, G.; Liu, J.-F.; Chen, Q.-Q.; et al. Functional Metabolomics Characterizes a Key Role for N-Acetylneuraminic Acid in Coronary Artery Diseases. Circulation 2018, 137, 1374–1390. [Google Scholar] [CrossRef]
  383. Li, C.; Zhao, M.; Xiao, L.; Wei, H.; Wen, Z.; Hu, D.; Yu, B.; Sun, Y.; Gao, J.; Shen, X.; et al. Prognostic Value of Elevated Levels of Plasma N-Acetylneuraminic Acid in Patients with Heart Failure. Circ. Heart Fail. 2021, 14, e008459. [Google Scholar] [CrossRef]
  384. Pham, T.; Gregg, C.J.; Karp, F.; Chow, R.; Padler-Karavani, V.; Cao, H.; Chen, X.; Witztum, J.L.; Varki, N.M.; Varki, A. Evidence for a novel human-specific xeno-auto-antibody response against vascular endothelium. Blood 2009, 114, 5225–5235. [Google Scholar] [CrossRef]
  385. Yida, Z.; Imam, M.U.; Ismail, M.; Wong, W.; Abdullah, M.A.; Ideris, A.; Ismail, N. N-Acetylneuraminic acid attenuates hypercoagulation on high fat diet-induced hyperlipidemic rats. Food Nutr. Res. 2015, 59, 29046. [Google Scholar] [CrossRef] [PubMed]
  386. Peng, J.; Vongpatanasin, W.; Sacharidou, A.; Kifer, D.; Yuhanna, I.S.; Banerjee, S.; Tanigaki, K.; Polasek, O.; Chu, H.; Sundgren, N.C.; et al. Supplementation with the Sialic Acid Precursor N-Acetyl-D-Mannosamine Breaks the Link Between Obesity and Hypertension. Circulation 2019, 140, 2005–2018. [Google Scholar] [CrossRef]
  387. Orlich, M.J.; Singh, P.N.; Sabaté, J.; Fan, J.; Sveen, L.; Bennett, H.; Knutsen, S.F.; Beeson, W.L.; Jaceldo-Siegl, K.; Butler, T.L.; et al. Vegetarian Dietary Patterns and the Risk of Colorectal Cancers. JAMA Intern. Med. 2015, 175, 767. [Google Scholar] [CrossRef]
  388. Liu, L.; Shi, Z.; Ji, X.; Zhang, W.; Luan, J.; Zahr, T.; Qiang, L. Adipokines, adiposity, and atherosclerosis. Cell. Mol. Life Sci. 2022, 79, 272. [Google Scholar] [CrossRef] [PubMed]
  389. Rizzo, N.S.; Sabaté, J.; Jaceldo-Siegl, K.; Fraser, G.E. Vegetarian Dietary Patterns Are Associated with a Lower Risk of Metabolic Syndrome: The Adventist Health Study 2. Diabetes Care 2011, 34, 1225–1227. [Google Scholar] [CrossRef]
  390. Tonstad, S.; Stewart, K.; Oda, K.; Batech, M.; Herring, R.P.; Fraser, G.E. Vegetarian diets and incidence of diabetes in the Adventist Health Study-2. Nutr. Metab. Cardiovasc. Dis. 2013, 23, 292–299. [Google Scholar] [CrossRef] [PubMed]
  391. Fraser, G.; Katuli, S.; Anousheh, R.; Knutsen, S.; Herring, P.; Fan, J. Vegetarian diets and cardiovascular risk factors in black members of the Adventist Health Study-2. Public Health Nutr. 2015, 18, 537–545. [Google Scholar] [CrossRef]
  392. Matar, P.; Alaniz, L.; Rozados, V.; Aquino, J.B.; Malvicini, M.; Atorrasagasti, C.; Gidekel, M.; Silva, M.; Scharovsky, O.G.; Mazzolini, G. Immunotherapy for liver tumors: Present status and future prospects. J. Biomed. Sci. 2009, 16, 30. [Google Scholar] [CrossRef]
  393. Varki, A. Letter to the Glyco-Forum: Since there are PAMPs and DAMPs, there must be SAMPs? Glycan “self-associated molecular patterns” dampen innate immunity, but pathogens can mimic them. Glycobiology 2011, 21, 1121–1124. [Google Scholar] [CrossRef]
  394. Lübbers, J.; Rodríguez, E.; van Kooyk, Y. Modulation of Immune Tolerance via Siglec-Sialic Acid Interactions. Front. Immunol. 2018, 9, 2807. [Google Scholar] [CrossRef]
  395. Pilling, D.; Karhadkar, T.R.; Gomer, R.H. High-Fat Diet–Induced Adipose Tissue and Liver Inflammation and Steatosis in Mice Are Reduced by Inhibiting Sialidases. Am. J. Pathol. 2021, 191, 131–143. [Google Scholar] [CrossRef] [PubMed]
  396. Nayak, B.S.; Bhaktha, G. Relationship between Sialic acid and metabolic variables in Indian type 2 diabetic patients. Lipids Health Dis. 2005, 4, 15. [Google Scholar] [CrossRef]
  397. Yida, Z.; Imam, M.U.; Ismail, M.; Ismail, N.; Azmi, N.H.; Wong, W.; Altine Adamu, H.; Md Zamri, N.D.; Ideris, A.; Abdullah, M.A. N-Acetylneuraminic Acid Supplementation Prevents High Fat Diet-Induced Insulin Resistance in Rats through Transcriptional and Nontranscriptional Mechanisms. BioMed Res. Int. 2015, 2015, 602313. [Google Scholar] [CrossRef]
  398. White, E.J.; Gyulay, G.; Lhoták, Š.; Szewczyk, M.M.; Chong, T.; Fuller, M.T.; Dadoo, O.; Fox-Robichaud, A.E.; Austin, R.C.; Trigatti, B.L.; et al. Sialidase down-regulation reduces non-HDL cholesterol, inhibits leukocyte transmigration, and attenuates atherosclerosis in ApoE knockout mice. J. Biol. Chem. 2018, 293, 14689–14706. [Google Scholar] [CrossRef] [PubMed]
  399. Munkley, J.; Scott, E. Targeting Aberrant Sialylation to Treat Cancer. Medicines 2019, 6, 102. [Google Scholar] [CrossRef]
  400. Rabinovich, G.A.; Croci, D.O. Regulatory Circuits Mediated by Lectin-Glycan Interactions in Autoimmunity and Cancer. Immunity 2012, 36, 322–335. [Google Scholar] [CrossRef]
  401. Rodrigues, E.; Macauley, M.S. Hypersialylation in cancer: Modulation of inflammation and therapeutic opportunities. Cancers 2018, 10, 207. [Google Scholar] [CrossRef] [PubMed]
  402. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  403. Croci, D.O.; Zacarías Fluck, M.F.; Rico, M.J.; Matar, P.; Rabinovich, G.A.; Scharovsky, O.G. Dynamic cross-talk between tumor and immune cells in orchestrating the immunosuppressive network at the tumor microenvironment. Cancer Immunol. Immunother. 2007, 56, 1687–1700. [Google Scholar] [CrossRef]
  404. Cagnoni, A.J.; Pérez Sáez, J.M.; Rabinovich, G.A.; Mariño, K.V. Turning-Off Signaling by Siglecs, Selectins, and Galectins: Chemical Inhibition of Glycan-Dependent Interactions in Cancer. Front. Oncol. 2016, 6, 109. [Google Scholar] [CrossRef]
  405. van de Wall, S.; Santegoets, K.C.M.; van Houtum, E.J.H.; Büll, C.; Adema, G.J. Sialoglycans and Siglecs Can Shape the Tumor Immune Microenvironment. Trends Immunol. 2020, 41, 274–285. [Google Scholar] [CrossRef]
  406. Büll, C.; den Brok, M.H.; Adema, G.J. Sweet escape: Sialic acids in tumor immune evasion. Biochim. Biophys. Acta BBA—Rev. Cancer 2014, 1846, 238–246. [Google Scholar] [CrossRef]
  407. Dall’Olio, F.; Malagolini, N.; Trinchera, M.; Chiricolo, M. Sialosignaling: Sialyltransferases as engines of self-fueling loops in cancer progression. Biochim. Biophys. Acta BBA—Gen. Subj. 2014, 1840, 2752–2764. [Google Scholar] [CrossRef]
  408. Sun, J.; Lu, Q.; Sanmamed, M.F.; Wang, J. Siglec-15 as an Emerging Target for Next-generation Cancer Immunotherapy. Clin. Cancer Res. 2021, 27, 680–688. [Google Scholar] [CrossRef] [PubMed]
  409. Altevogt, P.; Sammar, M.; Hüser, L.; Kristiansen, G. Novel insights into the function of CD24: A driving force in cancer. Int. J. Cancer 2021, 148, 546–559. [Google Scholar] [CrossRef] [PubMed]
  410. Stanczak, M.A.; Rodrigues Mantuano, N.; Kirchhammer, N.; Sanin, D.E.; Jacob, F.; Coelho, R.; Everest-Dass, A.V.; Wang, J.; Trefny, M.P.; Monaco, G.; et al. Targeting cancer glycosylation repolarizes tumor-associated macrophages allowing effective immune checkpoint blockade. Sci. Transl. Med. 2022, 14, eabj1270. [Google Scholar] [CrossRef]
  411. Diamantopoulou, S.; Stagiannis, K.D.; Vasilopoulos, K.; Barlas, P.; Tsegenidis, T.; Karamanos, N.K. Importance of high-performance liquid chromatographic analysis of serum N-acylneuraminic acids in evaluating surgical treatment in patients with early endometrial cancer. J. Chromatogr. B. Biomed. Sci. App. 1999, 732, 375–381. [Google Scholar] [CrossRef]
  412. Phitak, T.; Klangjorhor, J.; Pothacharoen, P.; Pruksakorn, D.; Kongtawelert, P. Level and distribution of secreted and cell-associated N-acetyl, N-glycolylneuraminic, and deaminoneuraminic acids in osteosarcoma cells isolated from patients. ScienceAsia 2017, 43, 15–25. [Google Scholar] [CrossRef]
  413. Yabu, M.; Korekane, H.; Hatano, K.; Kaneda, Y.; Nonomura, N.; Sato, C.; Kitajima, K.; Miyamoto, Y. Occurrence of free deaminoneuraminic acid (KDN)-containing complex-type N-glycans in human prostate cancers. Glycobiology 2013, 23, 634–642. [Google Scholar] [CrossRef]
  414. Tzanakakis, G.N.; Syrokou, A.; Kanakis, I.; Karamanos, N.K. Determination and distribution of N-acetyl- and N-glycolylneuraminic acids in culture media and cell associated glycoconjugates from human malignant mesothelioma and adenocarcinoma cells. Biomed. Chromatogr. 2006, 20, 434–439. [Google Scholar] [CrossRef]
  415. Tzanakakis, G.N.; Nikitovic, D.; Katonis, P.; Kanakis, I.; Karamanos, N.K. Expression and distribution of N-acetyl and N-glycolylneuraminic acids in secreted and cell-associated glycoconjugates by two human osteosarcoma cell lines. Biomed. Chromatogr. 2007, 21, 406–409. [Google Scholar] [CrossRef] [PubMed]
  416. Dobie, C.; Skropeta, D. Insights into the role of sialylation in cancer progression and metastasis. Br. J. Cancer 2021, 124, 76–90. [Google Scholar] [CrossRef] [PubMed]
  417. Ostrand-Rosenberg, S. Immune surveillance: A balance between protumor and antitumor immunity. Curr. Opin. Genet. Dev. 2008, 18, 11–18. [Google Scholar] [CrossRef]
  418. Smith, B.A.H.; Bertozzi, C.R. The clinical impact of glycobiology: Targeting selectins, Siglecs and mammalian glycans. Nat. Rev. Drug Discov. 2021, 20, 217–243. [Google Scholar] [CrossRef] [PubMed]
  419. Duan, S.; Paulson, J.C. Siglecs as Immune Cell Checkpoints in Disease. Annu. Rev. Immunol. 2020, 38, 365–395. [Google Scholar] [CrossRef]
  420. Läubli, H.; Borsig, L. Altered Cell Adhesion and Glycosylation Promote Cancer Immune Suppression and Metastasis. Front. Immunol. 2019, 10, 2120. [Google Scholar] [CrossRef]
  421. Anselmino, L.E.; Baglioni, M.V.; Malizia, F.; Laluce, N.C.; Etichetti, C.B.; Marignac, V.L.M.; Rozados, V.; Scharovsky, O.G.; Girardini, J.; Rico, M.J.; et al. Repositioning metformin and propranolol for colorectal and triple negative breast cancers treatment. Sci. Rep. 2021, 11, 8091. [Google Scholar] [CrossRef]
  422. Veettil, S.K.; Wong, T.Y.; Loo, Y.S.; Playdon, M.C.; Lai, N.M.; Giovannucci, E.L.; Chaiyakunapruk, N. Role of Diet in Colorectal Cancer Incidence: Umbrella Review of Meta-analyses of Prospective Observational Studies. JAMA Netw. Open 2021, 4, e2037341. [Google Scholar] [CrossRef]
  423. Buja, A.; Pierbon, M.; Lago, L.; Grotto, G.; Baldo, V. Breast Cancer Primary Prevention and Diet: An Umbrella Review. Int. J. Environ. Res. Public. Health 2020, 17, 4731. [Google Scholar] [CrossRef]
  424. Debras, C.; Chazelas, E.; Srour, B.; Kesse-Guyot, E.; Julia, C.; Zelek, L.; Agaësse, C.; Druesne-Pecollo, N.; Galan, P.; Hercberg, S.; et al. Total and added sugar intakes, sugar types, and cancer risk: Results from the prospective NutriNet-Santé cohort. Am. J. Clin. Nutr. 2020, 112, 1267–1279. [Google Scholar] [CrossRef]
  425. Gaesser, G.A. Whole Grains, Refined Grains, and Cancer Risk: A Systematic Review of Meta-Analyses of Observational Studies. Nutrients 2020, 12, 3756. [Google Scholar] [CrossRef] [PubMed]
  426. Orlich, M.J.; Mashchak, A.D.; Jaceldo-Siegl, K.; Utt, J.T.; Knutsen, S.F.; Sveen, L.E.; Fraser, G.E. Dairy foods, calcium intakes, and risk of incident prostate cancer in Adventist Health Study–2. Am. J. Clin. Nutr. 2022, 116, 314–324. [Google Scholar] [CrossRef] [PubMed]
  427. Penniecook-Sawyers, J.A.; Jaceldo-Siegl, K.; Fan, J.; Beeson, L.; Knutsen, S.; Herring, P.; Fraser, G.E. Vegetarian dietary patterns and the risk of breast cancer in a low-risk population. Br. J. Nutr. 2016, 115, 1790–1797. [Google Scholar] [CrossRef]
  428. Vargas, A.J.; Thompson, P.A. Diet and Nutrient Factors in Colorectal Cancer Risk. Nutr. Clin. Pract. 2012, 27, 613–623. [Google Scholar] [CrossRef] [PubMed]
  429. Aykan, N.F. Red meat and colorectal cancer. Oncol. Rev. 2015, 9, 38–44. [Google Scholar] [CrossRef]
  430. Oostindjer, M.; Alexander, J.; Amdam, G.V.; Andersen, G.; Bryan, N.S.; Chen, D.; Corpet, D.E.; De Smet, S.; Dragsted, L.O.; Haug, A.; et al. The role of red and processed meat in colorectal cancer development: A perspective. Meat Sci. 2014, 97, 583–596. [Google Scholar] [CrossRef]
  431. Sesink, A.L.A.; Termont, D.S.M.L.; Kleibeuker, J.H.; Meer, R.V.D. Red meat and colon cancer: Dietary haem, but not fat, has cytotoxic and hyperproliferative effects on rat colonic epithelium Aloys. Carcinogenesis 2000, 21, 1909–1915. [Google Scholar]
  432. Turner, N.D.; Lloyd, S.K. Association between red meat consumption and colon cancer: A systematic review of experimental results. Exp. Biol. Med. 2017, 242, 813–839. [Google Scholar] [CrossRef]
  433. EFSA Panel on Contaminants in the Food Chain (EFSA CONTAM Panel); Schrenk, D.; Bignami, M.; Bodin, L.; Chipman, J.K.; del Mazo, J.; Hogstrand, C.; Hoogenboom, L.; Leblanc, J.-C.; Nebbia, C.S.; et al. Risk assessment of N-nitrosamines in food. EFSA J. 2023, 21, e07884. [Google Scholar] [CrossRef]
  434. Wu, B.; Yang, D.; Yang, S.; Zhang, G. Dietary Salt Intake and Gastric Cancer Risk: A Systematic Review and Meta-Analysis. Front. Nutr. 2021, 8, 801228. [Google Scholar] [CrossRef]
  435. Fiorito, V.; Chiabrando, D.; Petrillo, S.; Bertino, F.; Tolosano, E. The Multifaceted Role of Heme in Cancer. Front. Oncol. 2020, 9, 1540. [Google Scholar] [CrossRef]
  436. Alisson-Silva, F.; Kawanishi, K.; Varki, A. Human risk of diseases associated with red meat intake: Analysis of current theories and proposed role for metabolic incorporation of a non-human sialic acid. Mol. Asp. Med. 2016, 51, 16–30. [Google Scholar] [CrossRef]
  437. zur Hausen, H. Red meat consumption and cancer: Reasons to suspect involvement of bovine infectious factors in colorectal cancer. Int. J. Cancer 2012, 130, 2475–2483. [Google Scholar] [CrossRef]
  438. Crisà, A.; Marchitelli, C.; Failla, S.; Contò, M. Determination of N-acetylneuraminic and N-glycolylneuraminic acids in unprocessed milk of four cattle breeds. J. Dairy Res. 2022, 89, 299–301. [Google Scholar] [CrossRef]
  439. Hinterseher, J.; Günther, J.; Zlatina, K.; Isernhagen, L.; Viergutz, T.; Wirthgen, E.; Hoeflich, A.; Vernunft, A.; Galuska, S.P. Milk Polysialic Acid Levels Rapidly Decrease in Line with the N-Acetylneuraminic Acid Concentrations during Early Lactation in Dairy Cows. Biology 2022, 12, 5. [Google Scholar] [CrossRef]
  440. Frei, R.; Roduit, C.; Ferstl, R.; Mahony, L.O.; Lauener, R.P. Exposure of children to rural lifestyle factors associated with protection against allergies induces an Anti-Neu5Gc antibody response. Front. Immunol. 2019, 10, 1628. [Google Scholar] [CrossRef]
  441. Leviatan Ben-Arye, S.; Schneider, C.; Yu, H.; Bashir, S.; Chen, X.; Von Gunten, S.; Padler-Karavani, V. Differential recognition of diet-derived Neu5Gc-Neoantigens on glycan microarrays by carbohydrate-specific pooled human IgG and IgA antibodies. Bioconjug. Chem. 2019, 30, 1565–1574. [Google Scholar] [CrossRef]
  442. He, E.; Quan, W.; Luo, J.; Liu, C.; Zheng, W.; Shen, Q. Absorption and Transport Mechanism of Red Meat-Derived N-glycolylneuraminic Acid and Its Damage to Intestinal Barrier Function through the NF-κB Signaling Pathway. Toxins 2023, 15, 132. [Google Scholar] [CrossRef]
  443. Salama, A.; Evanno, G.; Lim, N.; Rousse, J.; Le Berre, L.; Nicot, A.; Brouard, S.; Harris, K.M.M.; Ehlers, M.R.R.; Gitelman, S.E.E.; et al. Anti-Gal and anti-Neu5Gc responses in nonimmunosuppressed patients following treatment with rabbit anti-thymocyte polyclonal IgGs. Transplantation 2017, 101, 2501–2507. [Google Scholar] [CrossRef]
Figure 1. The molecular structure of sialic acid and its variants under various physiologic conditions. Sialic acids are characteristically carboxylate at group C1, attached to the C2 anomeric carbon. The wavy lines in C1 signify that the specific α or β anomeric configuration is not specified. The most common natural substituents (positions indicated in brackets) are a glycerol-like side chain (C7, 8, and 9) projecting out of the ring, and an acylated amino group at C5. Different groups with methyl, lactyl, acetyl, glycolyl, or sulfate can additionally diversify sialic acids. Prepared with ChemDraw, ver.: 23.1.2.7.
Figure 1. The molecular structure of sialic acid and its variants under various physiologic conditions. Sialic acids are characteristically carboxylate at group C1, attached to the C2 anomeric carbon. The wavy lines in C1 signify that the specific α or β anomeric configuration is not specified. The most common natural substituents (positions indicated in brackets) are a glycerol-like side chain (C7, 8, and 9) projecting out of the ring, and an acylated amino group at C5. Different groups with methyl, lactyl, acetyl, glycolyl, or sulfate can additionally diversify sialic acids. Prepared with ChemDraw, ver.: 23.1.2.7.
Biologics 05 00010 g001
Figure 2. The structures of the most common sialic acids. The graphical representation of these glycans was developed according to the Symbol Nomenclature for Glycans (SNFG) guidelines. Prepared with ChemDraw, ver.: 23.1.2.7. Neu, neuraminic acid; Neu5Ac, N-acetylneuraminic acid; Neu5Gc, N-glycolylneuraminic acid (Neu5Gc); KDN, 2-keto-3-deoxy-D-glycero-D-galacto-nononic acid.
Figure 2. The structures of the most common sialic acids. The graphical representation of these glycans was developed according to the Symbol Nomenclature for Glycans (SNFG) guidelines. Prepared with ChemDraw, ver.: 23.1.2.7. Neu, neuraminic acid; Neu5Ac, N-acetylneuraminic acid; Neu5Gc, N-glycolylneuraminic acid (Neu5Gc); KDN, 2-keto-3-deoxy-D-glycero-D-galacto-nononic acid.
Biologics 05 00010 g002
Figure 3. Sialic acid metabolism in eukaryote cells. (1) Sialic acid molecules found in the cytosol of eukaryotic cells are obtained from exogenous sources, such as glycolipids or glycoproteins. (2) Sialic acids can be produced from cytosolic UDP-GlcNAc molecules, biosynthesized from glucose in the hexosamine pathway. Finally, in the lysosome, after Neu1 hydrolyzes the terminal sialic acid residue from glycoconjugates, the sialylated glycoconjugate is transported by the sialin transporter encoded by the SLC17A5 gene. CMAH, cytidine monophospho-N-acetylneuraminic acid hydroxylase; CMAS, CMP-N-acetylneuraminate synthetase; CMP, cytidine-5′-monophosphate; CMP-Sia-Tr, CMP sialic acid transporter in the Golgi apparatus, encoded by the SLC35A1 gene; CTP, cytidine-5′-triphosphate; ManNAc, N-acetylmannosamine; ManNAc-6-P, N-acetylmannosamine 6-phosphate; ManNAc-9-P, N-acetylmannosamine 9-phosphate; Neu1, neuraminidase 1; Neu4, neuraminidase 4; PEP, phosphoenolpyruvate; PPi, pyrophosphate; Sia, sialic acid; Sialin, sialic acid transporter in the lysosome; UDP-GlcNac, uridine diphosphate N-acetylglucosamine.
Figure 3. Sialic acid metabolism in eukaryote cells. (1) Sialic acid molecules found in the cytosol of eukaryotic cells are obtained from exogenous sources, such as glycolipids or glycoproteins. (2) Sialic acids can be produced from cytosolic UDP-GlcNAc molecules, biosynthesized from glucose in the hexosamine pathway. Finally, in the lysosome, after Neu1 hydrolyzes the terminal sialic acid residue from glycoconjugates, the sialylated glycoconjugate is transported by the sialin transporter encoded by the SLC17A5 gene. CMAH, cytidine monophospho-N-acetylneuraminic acid hydroxylase; CMAS, CMP-N-acetylneuraminate synthetase; CMP, cytidine-5′-monophosphate; CMP-Sia-Tr, CMP sialic acid transporter in the Golgi apparatus, encoded by the SLC35A1 gene; CTP, cytidine-5′-triphosphate; ManNAc, N-acetylmannosamine; ManNAc-6-P, N-acetylmannosamine 6-phosphate; ManNAc-9-P, N-acetylmannosamine 9-phosphate; Neu1, neuraminidase 1; Neu4, neuraminidase 4; PEP, phosphoenolpyruvate; PPi, pyrophosphate; Sia, sialic acid; Sialin, sialic acid transporter in the lysosome; UDP-GlcNac, uridine diphosphate N-acetylglucosamine.
Biologics 05 00010 g003
Figure 4. De novo pathways for the synthesis of KDN in animal cells. Man-6-P, a C2 epimer of glucose 6-phosphate, is a key precursor monosaccharide for KDN. The biosynthesis of KDN from Man-6-P follows comparable enzyme reaction steps to those of Neu5Ac from ManNAc-6-P. Notably, unlike other free monosaccharides, mannose is efficiently taken up by different cell types using a transporter that is unresponsive to glucose. CMP, cytidine-5′-monophosphate; CMP-KDN, cytidine 5′-(3-deoxy-D-glycero-D-galacto-2-nonulosonic phosphate); CMP-KDN-synthetase, CMP-3-deoxy-d-glycero-d-galacto-nonulosonic acid synthetase; CTP, cytidine triphosphate; KDN-9-P, KDN-9-phosphate; Man-6-P, mannose-6-phosphate; Pi, inorganic phosphate; PPi, pyrophosphate.
Figure 4. De novo pathways for the synthesis of KDN in animal cells. Man-6-P, a C2 epimer of glucose 6-phosphate, is a key precursor monosaccharide for KDN. The biosynthesis of KDN from Man-6-P follows comparable enzyme reaction steps to those of Neu5Ac from ManNAc-6-P. Notably, unlike other free monosaccharides, mannose is efficiently taken up by different cell types using a transporter that is unresponsive to glucose. CMP, cytidine-5′-monophosphate; CMP-KDN, cytidine 5′-(3-deoxy-D-glycero-D-galacto-2-nonulosonic phosphate); CMP-KDN-synthetase, CMP-3-deoxy-d-glycero-d-galacto-nonulosonic acid synthetase; CTP, cytidine triphosphate; KDN-9-P, KDN-9-phosphate; Man-6-P, mannose-6-phosphate; Pi, inorganic phosphate; PPi, pyrophosphate.
Biologics 05 00010 g004
Figure 5. Biosynthesis of N-acetylneuraminic acid in prokaryotes. In the bacterium E. coli, glucose is metabolized through a series of enzymatic reactions. The process begins when glucose enters the cell and is phosphorylated by hexokinase to form glucose-6-phosphate. The next step involves an isomerization reaction catalyzed by the enzyme glucose-6-phosphate isomerase (PGI), which converts glucose-6-phosphate into fructose-6-phosphate by a rearrangement. The pathway proceeds through several more steps, including the cleavage of fructose-6-phosphate, followed by a series of four enzyme-catalyzed reactions to generate a nucleotide sugar called UDP-N-acetylglucosamine (UDP-GlcNAc). Then, UDP-GlcNAc is converted to ManNAc, catalyzed by NeuC, a hydrolyzing UDP-GlcNAc epimerase. Neu5Ac is produced via a condensation reaction of ManNAc with phosphoenol pyruvate (PEP), which is catalyzed by NeuB (sialic acid synthase). The Neu5Ac is primed by NeuA (Sialic acid O-acetylesterase) and CTP to form CMP-Neu5Ac. CMP: cytidine 5′-monophosphate. CTP: cytidine triphosphate; CMP-Neu5Ac: cytidine-5′-monophospho-N-acetylneuraminic acid; UDP-GlcNAc: uridine diphosphate N-acetylglucosamine.
Figure 5. Biosynthesis of N-acetylneuraminic acid in prokaryotes. In the bacterium E. coli, glucose is metabolized through a series of enzymatic reactions. The process begins when glucose enters the cell and is phosphorylated by hexokinase to form glucose-6-phosphate. The next step involves an isomerization reaction catalyzed by the enzyme glucose-6-phosphate isomerase (PGI), which converts glucose-6-phosphate into fructose-6-phosphate by a rearrangement. The pathway proceeds through several more steps, including the cleavage of fructose-6-phosphate, followed by a series of four enzyme-catalyzed reactions to generate a nucleotide sugar called UDP-N-acetylglucosamine (UDP-GlcNAc). Then, UDP-GlcNAc is converted to ManNAc, catalyzed by NeuC, a hydrolyzing UDP-GlcNAc epimerase. Neu5Ac is produced via a condensation reaction of ManNAc with phosphoenol pyruvate (PEP), which is catalyzed by NeuB (sialic acid synthase). The Neu5Ac is primed by NeuA (Sialic acid O-acetylesterase) and CTP to form CMP-Neu5Ac. CMP: cytidine 5′-monophosphate. CTP: cytidine triphosphate; CMP-Neu5Ac: cytidine-5′-monophospho-N-acetylneuraminic acid; UDP-GlcNAc: uridine diphosphate N-acetylglucosamine.
Biologics 05 00010 g005
Figure 6. Sialylated glycans in cancer. Tumor cells have increased levels of sialylated glycans on their cell surface, including sialyl LewisA (SLeA), sialyl LewisX (SLeX), and sialyl-Tn (sTn). GD2, di-sialoganglioside.
Figure 6. Sialylated glycans in cancer. Tumor cells have increased levels of sialylated glycans on their cell surface, including sialyl LewisA (SLeA), sialyl LewisX (SLeX), and sialyl-Tn (sTn). GD2, di-sialoganglioside.
Biologics 05 00010 g006
Table 2. Sialic acid concentrations in human cancer, in different tumor cells.
Table 2. Sialic acid concentrations in human cancer, in different tumor cells.
CancerNeu5AcNeu5GcKDNReference
Endometrial cancer (µg/g)6.99 × 105 ± 3.05 × 1021.3 × 104 ± 5 × 103-[411]
Osteosarcoma cells (µg/mL)53.617.50.36[412]
Prostate cancer (µg/100 μg protein)3.09 × 10−5–6.04 × 10−2--[413]
Pancreatic cancer (µg/L)9.27 × 10−3–3.7 × 10−3--[151]
Throat cancer (µg/g)850.032[36]
Malignant mesothelioma (µg per 106 cells)Culture mediaCell membrane extractsCulture mediaCell membrane extracts-[414]
STAV-FCS5.05Traces35.7720.37
STAV-AB10.85ND13.6619.23
Wester11.53ND18.6525.3
Adenocarcinoma cells (µg per 106 cells)
Wart6Traces16.5121.12
Osteosarcoma cells (µg per 106 cells)3.97-2.754.48-[415]
KDN, 2-keto-3-deoxy-D-glycero-D-galacto-nononic acid; Neu5Ac, N-acetylneuraminic acid; Neu5Gc, N-glycolylneuraminic acid; ND, not detected; STAV-AB, cancer cell line, specifically, malignant mesothelioma; STAV-FCS, cancer cell sub-line, specifically, the sarcomatoid phenotype of mesothelioma.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guerrero-Flores, G.N.; Butler, F.M.; Martinez Marignac, V.L.; Zhang, G.; Pacheco, F.J.; Boskovic, D.S. Sialic Acids in Health and Disease. Biologics 2025, 5, 10. https://doi.org/10.3390/biologics5020010

AMA Style

Guerrero-Flores GN, Butler FM, Martinez Marignac VL, Zhang G, Pacheco FJ, Boskovic DS. Sialic Acids in Health and Disease. Biologics. 2025; 5(2):10. https://doi.org/10.3390/biologics5020010

Chicago/Turabian Style

Guerrero-Flores, Gerardo N., Fayth M. Butler, Veronica L. Martinez Marignac, Guangyu Zhang, Fabio J. Pacheco, and Danilo S. Boskovic. 2025. "Sialic Acids in Health and Disease" Biologics 5, no. 2: 10. https://doi.org/10.3390/biologics5020010

APA Style

Guerrero-Flores, G. N., Butler, F. M., Martinez Marignac, V. L., Zhang, G., Pacheco, F. J., & Boskovic, D. S. (2025). Sialic Acids in Health and Disease. Biologics, 5(2), 10. https://doi.org/10.3390/biologics5020010

Article Metrics

Back to TopTop