Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.725
Filter
1.
AAPS PharmSciTech ; 25(5): 125, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834759

ABSTRACT

DOX liposomes have better therapeutic effects and lower toxic side effects. The targeting ability of liposomes is one of the key factors affecting the therapeutic effect of DOX liposomes. This study developed two types of targeted liposomes. Sialic acid (SA)-modified liposomes were designed to target the highly expressed Siglec-1 receptor on tumor-associated macrophages surface. Phosphatidylserine (PS)-modified liposomes were designed to promote phagocytosis by monocyte-derived macrophages through PS apoptotic signaling. In order to assess and compare the therapeutic potential of different targeted pathways in the context of anti-tumor treatment, we compared four phosphatidylserine membrane materials (DOPS, DSPS, DPPS and DMPS) and found that liposomes prepared using DOPS as material could significantly improve the uptake ability of RAW264.7 cells for DOX liposomes. On this basis, normal DOX liposomes (CL-DOX) and SA-modified DOX liposomes (SAL-DOX), PS-modified DOX liposomes (PS-CL-DOX), SA and PS co-modified DOX liposomes (PS-SAL-DOX) were prepared. The anti-tumor cells function of each liposome on S180 and RAW264.7 in vitro was investigated, and it was found that SA on the surface of liposomes can increase the inhibitory effect. In vivo efficacy results exhibited that SAL-DOX and PS-CL-DOX were superior to other groups in terms of ability to inhibit tumor growth and tumor inhibition index, among which SAL-DOX had the best anti-tumor effect. Moreover, SAL-DOX group mice had high expression of IFN-γ as well as IL-12 factors, which could significantly inhibit mice tumor growth, improve the immune microenvironment of the tumor site, and have excellent targeted delivery potential.


Subject(s)
Doxorubicin , Liposomes , N-Acetylneuraminic Acid , Phosphatidylserines , Tumor-Associated Macrophages , Animals , Mice , N-Acetylneuraminic Acid/chemistry , RAW 264.7 Cells , Phosphatidylserines/metabolism , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Phagocytosis/drug effects , Drug Delivery Systems/methods , Apoptosis/drug effects
2.
Nat Commun ; 15(1): 4386, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782953

ABSTRACT

Sialin, a member of the solute carrier 17 (SLC17) transporter family, is unique in its ability to transport not only sialic acid using a pH-driven mechanism, but also transport mono and diacidic neurotransmitters, such as glutamate and N-acetylaspartylglutamate (NAAG), into synaptic vesicles via a membrane potential-driven mechanism. While most transporters utilize one of these mechanisms, the structural basis of how Sialin transports substrates using both remains unclear. Here, we present the cryogenic electron-microscopy structures of human Sialin: apo cytosol-open, apo lumen-open, NAAG-bound, and inhibitor-bound. Our structures show that a positively charged cytosol-open vestibule accommodates either NAAG or the Sialin inhibitor Fmoc-Leu-OH, while its luminal cavity potentially binds sialic acid. Moreover, functional analyses along with molecular dynamics simulations identify key residues in binding sialic acid and NAAG. Thus, our findings uncover the essential conformational states in NAAG and sialic acid transport, demonstrating a working model of SLC17 transporters.


Subject(s)
Cryoelectron Microscopy , Molecular Dynamics Simulation , Humans , N-Acetylneuraminic Acid/metabolism , N-Acetylneuraminic Acid/chemistry , Lysosomes/metabolism , HEK293 Cells , Protein Conformation , Organic Anion Transporters/metabolism , Organic Anion Transporters/chemistry , Organic Anion Transporters/antagonists & inhibitors , Dipeptides/chemistry , Dipeptides/metabolism , Dipeptides/pharmacology , Symporters
3.
Int J Biol Macromol ; 269(Pt 1): 132022, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38697414

ABSTRACT

Edible bird's nest (EBN) is made up of sialylated-mucin glycoprotein with various health benefits due to its high antioxidative activity. However, as a macromolecule with distinct charged sialic acid and amino acids, fractions with different charges would have varied physicochemical properties and antioxidant activity, which have not been studied. Therefore, this study aimed to fractionate and purify the enzymatic hydrolysed of cleaned EBN (EBNhc) and EBN by-product (EBNhbyp) through anion exchange chromatography (AEC), and determine their molecular weights, physicochemical properties, and antioxidative activities. Overall, 26 fractionates were collected from enzymatic hydrolysate by AEC, which were classified into 5 fractions. It was found that the positively charged fraction of EBNhc (CF 1) and EBNhbyp (DF 1) showed the significantly highest (p < 0.05) soluble protein contents (22.86 and 18.40 mg/g), total peptide contents (511.13 and 800.47 mg/g) and ferric reducing antioxidant power (17.44 and 6.96 mg/g) among the fractionates. In conclusion, a positively charged fraction (CF 1 and DF 1) showed more desired physicochemical properties and antioxidative activities. This research suggests the potential of AEC fractionation as a technology to purify EBN and produce positively charged EBN fractionates with antioxidative potential that could be applied as food components to provide health benefits.


Subject(s)
Antioxidants , Birds , Glycoproteins , Animals , Chromatography, Ion Exchange/methods , Antioxidants/chemistry , Antioxidants/isolation & purification , Antioxidants/pharmacology , Glycoproteins/chemistry , Glycoproteins/isolation & purification , Hydrolysis , Molecular Weight , N-Acetylneuraminic Acid/chemistry , Chemical Fractionation/methods
4.
Immunohorizons ; 8(5): 384-396, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38809232

ABSTRACT

The mammalian Siglec receptor sialoadhesin (Siglec1, CD169) confers innate immunity against the encapsulated pathogen group B Streptococcus (GBS). Newborn lung macrophages have lower expression levels of sialoadhesin at birth compared with the postnatal period, increasing their susceptibility to GBS infection. In this study, we investigate the mechanisms regulating sialoadhesin expression in the newborn mouse lung. In both neonatal and adult mice, GBS lung infection reduced Siglec1 expression, potentially delaying acquisition of immunity in neonates. Suppression of Siglec1 expression required interactions between sialic acid on the GBS capsule and the inhibitory host receptor Siglec-E. The Siglec1 gene contains multiple STAT binding motifs, which could regulate expression of sialoadhesin downstream of innate immune signals. Although GBS infection reduced STAT1 expression in the lungs of wild-type newborn mice, we observed increased numbers of STAT1+ cells in Siglece-/- lungs. To test if innate immune activation could increase sialoadhesin at birth, we first demonstrated that treatment of neonatal lung macrophages ex vivo with inflammatory activators increased sialoadhesin expression. However, overcoming the low sialoadhesin expression at birth using in vivo prenatal exposures or treatments with inflammatory stimuli were not successful. The suppression of sialoadhesin expression by GBS-Siglec-E engagement may therefore contribute to disease pathogenesis in newborns and represent a challenging but potentially appealing therapeutic opportunity to augment immunity at birth.


Subject(s)
Animals, Newborn , Mice, Knockout , N-Acetylneuraminic Acid , STAT1 Transcription Factor , Sialic Acid Binding Ig-like Lectin 1 , Streptococcal Infections , Streptococcus agalactiae , Animals , Mice , Streptococcus agalactiae/immunology , N-Acetylneuraminic Acid/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Immunity, Innate , Mice, Inbred C57BL , Lung/immunology , Lung/microbiology , Lung/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Female , Macrophages/immunology , Macrophages/metabolism , Lectins/metabolism , Lectins/genetics , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Antigens, CD/metabolism , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte
5.
Anal Methods ; 16(22): 3475-3485, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38780482

ABSTRACT

Sialylation is an important modification of proteins, related to protein life and bioactivity. However, the evaluation of sialylation is only based on the average molecular composition by peptide mapping and glycan profiling because sialylated proteins are usually too heterogeneous to obtain good quality mass spectra by conventional intact mass analysis methods. In this study, a simple strong cation exchange-mass spectroscopy (SCX-MS) method was developed for intact mass analysis of sialylated glycoproteins. The developed SCX-MS method provided good separation for sialylated glycoproteins and had an inherent characteristic of native MS. Thus, the intact mass analysis of highly heterogeneous glycoprotein, which cannot be obtained by reversed-phase liquid chromatography (RPLC)-MS and size exclusion chromatography (SEC)-MS methods, can be well analyzed using the current SCX-MS method. First, the method was developed and optimized using the etanercept monomer. Conditions including MS parameters, flow rate, and gradient were investigated. Then, the developed method was used to analyze a new recombinant vaccine, protein 1. Similar to the etanercept monomer, the intact molecular information of protein 1, which cannot be obtained by RPLC-MS and SEC-MS, can be achieved using SCX-MS. Combined with information obtained on peptide mapping and glycan profiles obtained by LC-MS, the new vaccine was well characterized. Finally, the SCX-MS method was used to quickly evaluate the batch-to-batch reproducibility of protein 1. It was much faster than peptide mapping and glycan profiling methods and can provide information complementary to these strategies. It should be useful for many applications where speed and comprehensive characterization are required, such as recombinant sialylated vaccines and fusion proteins.


Subject(s)
Glycoproteins , Mass Spectrometry , Glycoproteins/chemistry , Glycoproteins/analysis , Mass Spectrometry/methods , Chromatography, Ion Exchange/methods , Etanercept/chemistry , Glycosylation , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/analysis , Humans , Animals , Cations/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/analysis
6.
J Am Chem Soc ; 146(22): 15366-15375, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38768956

ABSTRACT

Inspired by the specificity of α-(2,9)-sialyl epitopes in bacterial capsular polysaccharides (CPS), a doubly fluorinated disaccharide has been validated as a vaccine lead against Neisseria meningitidis serogroups C and/or B. Emulating the importance of fluorine in drug discovery, this molecular editing approach serves a multitude of purposes, which range from controlling α-selective chemical sialylation to mitigating competing elimination. Conjugation of the disialoside with two carrier proteins (CRM197 and PorA) enabled a semisynthetic vaccine to be generated; this was then investigated in six groups of six mice. The individual levels of antibodies formed were compared and classified as highly glycan-specific and protective. All glycoconjugates induced a stable and long-term IgG response and binding to the native CPS epitope was achieved. The generated antibodies were protective against MenC and/or MenB; this was validated in vitro by SBA and OPKA assays. By merging the fluorinated glycan epitope of MenC with an outer cell membrane protein of MenB, a bivalent vaccine against both serogroups was created. It is envisaged that validation of this synthetic, fluorinated disialoside bioisostere as a potent antigen will open new therapeutic avenues.


Subject(s)
Halogenation , Animals , Mice , N-Acetylneuraminic Acid/chemistry , Meningococcal Vaccines/immunology , Meningococcal Vaccines/chemistry , Neisseria meningitidis, Serogroup B/immunology , Neisseria meningitidis, Serogroup B/chemistry , Meningitis, Meningococcal/prevention & control , Meningitis, Meningococcal/immunology
7.
Commun Biol ; 7(1): 430, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594506

ABSTRACT

Despite recent advances in cancer immunotherapy, pancreatic ductal adenocarcinoma (PDAC) remains unresponsive due to an immunosuppressive tumor microenvironment, which is characterized by the abundance of cancer-associated fibroblasts (CAFs). Once identified, CAF-mediated immune inhibitory mechanisms could be exploited for cancer immunotherapy. Siglec receptors are increasingly recognized as immune checkpoints, and their ligands, sialic acids, are known to be overexpressed by cancer cells. Here, we unveil a previously unrecognized role of sialic acid-containing glycans on PDAC CAFs as crucial modulators of myeloid cells. Using multiplex immunohistochemistry and transcriptomics, we show that PDAC stroma is enriched in sialic acid-containing glycans compared to tumor cells and normal fibroblasts, and characterized by ST3GAL4 expression. We demonstrate that sialic acids on CAF cell lines serve as ligands for Siglec-7, -9, -10 and -15, distinct from the ligands on tumor cells, and that these receptors are found on myeloid cells in the stroma of PDAC biopsies. Furthermore, we show that CAFs drive the differentiation of monocytes to immunosuppressive tumor-associated macrophages in vitro, and that CAF sialylation plays a dominant role in this process compared to tumor cell sialylation. Collectively, our findings unravel sialic acids as a mechanism of CAF-mediated immunomodulation, which may provide targets for immunotherapy in PDAC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , N-Acetylneuraminic Acid/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/metabolism , Macrophages/metabolism , Polysaccharides/metabolism , Tumor Microenvironment
8.
Br J Dermatol ; 190(5): e51, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38630930
9.
Biochem Biophys Res Commun ; 710: 149881, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38583233

ABSTRACT

Maackia amurensis lectins serve as research and botanical agents that bind to sialic residues on proteins. For example, M. amurensis seed lectin (MASL) targets the sialic acid modified podoplanin (PDPN) receptor to suppress arthritic chondrocyte inflammation, and inhibit tumor cell growth and motility. However, M. amurensis lectin nomenclature and composition are not clearly defined. Here, we sought to definitively characterize MASL and its effects on tumor cell behavior. We utilized SDS-PAGE and LC-MS/MS to find that M. amurensis lectins can be divided into two groups. MASL is a member of one group which is composed of subunits that form dimers, evidently mediated by a cysteine residue in the carboxy region of the protein. In contrast to MASL, members of the other group do not dimerize under nonreducing conditions. These data also indicate that MASL is composed of 4 isoforms with an identical amino acid sequence, but unique glycosylation sites. We also produced a novel recombinant soluble human PDPN receptor (shPDPN) with 17 threonine residues glycosylated with sialic acid moieties with potential to act as a ligand trap that inhibits OSCC cell growth and motility. In addition, we report here that MASL targets PDPN with very strong binding kinetics in the nanomolar range. Moreover, we confirm that MASL can inhibit the growth and motility of human oral squamous cell carcinoma (OSCC) cells that express the PDPN receptor. Taken together, these data characterize M. amurensis lectins into two major groups based on their intrinsic properties, clarify the composition of MASL and its subunit isoform sequence and glycosylation sites, define sialic acid modifications on the PDPN receptor and its ability to act as a ligand trap, quantitate MASL binding to PDPN with KD in the nanomolar range, and verify the ability of MASL to serve as a potential anticancer agent.


Subject(s)
Antineoplastic Agents , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck , N-Acetylneuraminic Acid/metabolism , Maackia/chemistry , Maackia/metabolism , Mouth Neoplasms/pathology , Chromatography, Liquid , Ligands , Tandem Mass Spectrometry , Lectins/pharmacology , Antineoplastic Agents/pharmacology , Sequence Analysis , Cell Movement
10.
Cell Mol Biol Lett ; 29(1): 46, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561669

ABSTRACT

BACKGROUND: Small extracellular vesicles (sEV) are closely associated with the development and metastasis of many types of mammalian cancer. Glycoconjugates are highly expressed on sEV and play important roles in sEV biogenesis and their interaction with other cells. However, the study on vesicular glycoconjugates are far behind proteins and nucleic acids. Especially, the functions of sialic acids which are the terminal components of glycoconjugates, are poorly understood in sEV. METHODS: Sialic acid levels on sEV from plasma and bladder cancer cells were determined by ELISA and lectin blotting. Effects of sialylation on sEV uptake were determined by flow cytometry. Vesicular glycoproteins bearing sialic acids responsible for sEV uptake was identified by proteomics and density gradient centrifugation, and their site-specific sialylation functions were assayed by N-glycosylation site mutation. Effects of integrin ß1 bearing sialic acids on the pro-metastatic function of sEV in vivo were explored using Balb/c nu/nu mice. RESULTS: (1) Increased sialic acid levels were observed in sEV from malignant bladder cancer cells. (2) Elimination of sialic acids on sEV impaired sEV uptake by recipient cells. (3) Vesicular integrin ß1 bearing sialic acids was identified to play a key role in sEV uptake. (4) Desialylation of the hybrid domain of vesicular integrin ß1 inhibited its binding to matrix fibronectin, and reduced sEV entry into recipient cells. (5) Sialylation on integrin ß1 affected pro-metastatic function of sEV in Balb/c nu/nu mice. CONCLUSIONS: Taken together, our findings indicate important functional roles of sialic acids in sEV uptake and reprogramming plasticity of surrounding normal epithelial cells.


Subject(s)
Extracellular Vesicles , Urinary Bladder Neoplasms , Animals , Mice , Extracellular Vesicles/metabolism , Glycoconjugates , Integrin beta1/metabolism , Mammals , N-Acetylneuraminic Acid/metabolism , Sialic Acids/metabolism
11.
Biochem Pharmacol ; 223: 116199, 2024 May.
Article in English | MEDLINE | ID: mdl-38604256

ABSTRACT

GNEM (GNE Myopathy) is a rare neuromuscular disease caused due to biallelic mutations in sialic acid biosynthetic GNE enzyme (UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine Kinase). Recently direct or indirect role of GNE in other cellular functions have been elucidated. Hyposialylation of IGF-1R leads to apoptosis due to mitochondrial dysfunction while hyposialylation of ß1 integrin receptor leads to altered F-actin assembly, disrupted cytoskeletal organization and slow cell migration. Other cellular defects in presence of GNE mutation include altered ER redox state and chaperone expression such as HSP70 or PrdxIV. Currently, there is no cure to treat GNEM. Possible therapeutic trials focus on supplementation with sialic acid, ManNAc, sialyllactose and gene therapy that slows the disease progression. In the present study, we analyzed the effect of small molecules like BGP-15 (HSP70 modulator), IGF-1 (IGF-1R ligand) and CGA (cofilin activator) on cellular phenotypes of GNE heterozygous knock out L6 rat skeletal muscle cell line (SKM­GNEHz). Treatment with BGP-15 improved GNE epimerase activity by 40 % and reduced ER stress by 45 % for SKM­GNEHz. Treatment with IGF-1 improved epimerase activity by 37.5 %, F-actin assembly by 100 %, cell migration upto 36 % (36 h) and atrophy by 0.44-fold for SKM­GNEHz. Treatment with CGA recovered epimerase activity by 49 %, F-actin assembly by 132 % and cell migration upto 41 % (24 h) in SKM­GNEHz. Our study shows that treatment with these small effector molecules reduces the detrimental phenotype observed in SKM­GNEHz, thereby, providing insights into potential therapeutic targets for GNEM.


Subject(s)
Distal Myopathies , N-Acetylneuraminic Acid , Oximes , Piperidines , Animals , Rats , Actins/genetics , Distal Myopathies/drug therapy , Distal Myopathies/genetics , Insulin-Like Growth Factor I , Mutation , N-Acetylneuraminic Acid/genetics , N-Acetylneuraminic Acid/metabolism , Oximes/pharmacology , Piperidines/pharmacology , Racemases and Epimerases/genetics
12.
Carbohydr Res ; 539: 109123, 2024 May.
Article in English | MEDLINE | ID: mdl-38669826

ABSTRACT

Sialic acid, the terminal structure of cell surface glycans, has essential functions in regulating immune response, cell-to-cell communication, and cell adhesion. More importantly, an increased level of sialic acid, termed hypersialylation, has emerged as a commonly observed phenotype in cancer. Therefore, targeting sialic acid ligands (sialoglycans) and their receptors (Siglecs) may provide a new therapeutic approach for cancer immunotherapy. We highlight the complexity of the sialic acid metabolism and its involvement in malignant transformation within individual cancer subtypes. In this review, we focus on the dysregulation of sialylation, the intricate nature of sialic acid synthesis, and clinical perspective. We aim to provide a brief insight into the mechanism of hypersialylation and how our understanding of these processes can be leveraged for the development of novel therapeutics.


Subject(s)
N-Acetylneuraminic Acid , Neoplasms , Humans , Neoplasms/metabolism , Neoplasms/pathology , N-Acetylneuraminic Acid/metabolism , N-Acetylneuraminic Acid/chemistry , Animals
13.
mBio ; 15(5): e0011924, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38587424

ABSTRACT

Gonorrhea, caused by the bacterium Neisseria gonorrhoeae (Gc), is characterized by neutrophilic influx to infection sites. Gc has developed mechanisms to resist killing by neutrophils that include modifications to its surface lipooligosaccharide (LOS). One such LOS modification is sialylation: Gc sialylates its terminal LOS sugars with cytidine-5'-monophosphate-N-acetylneuraminic acid, which is scavenged from the host using LOS sialyltransferase (Lst) since Gc cannot make its sialic acid. Sialylation enables sensitive strains of Gc to resist complement-mediated killing in a serum-dependent manner. However, little is known about the contribution of sialylation to complement-independent, direct Gc-neutrophil interactions. In the absence of complement, we found sialylated Gc expressing opacity-associated (Opa) proteins decreased the oxidative burst and granule exocytosis from primary human neutrophils. In addition, sialylated Opa+ Gc survived better than vehicle treated or Δlst Gc when challenged with neutrophils. However, Gc sialylation did not significantly affect Opa-dependent association with or internalization of Gc by neutrophils. Previous studies have implicated sialic acid-binding immunoglobulin-type lectins (Siglecs) in modulating neutrophil interactions with sialylated Gc. Blocking neutrophil Siglecs with antibodies that bind to their extracellular domains eliminated the ability of sialylated Opa+ Gc to suppress the oxidative burst and resist neutrophil killing. These findings highlight a new role for sialylation in Gc evasion of human innate immunity, with implications for the development of vaccines and therapeutics for gonorrhea. IMPORTANCE: Neisseria gonorrhoeae, the bacterium that causes gonorrhea, is an urgent global health concern due to increasing infection rates, widespread antibiotic resistance, and its ability to thwart protective immune responses. The mechanisms by which Gc subverts protective immune responses remain poorly characterized. One way N. gonorrhoeae evades human immunity is by adding sialic acid that is scavenged from the host onto its lipooligosaccharide, using the sialyltransferase Lst. Here, we found that sialylation enhances N. gonorrhoeae survival from neutrophil assault and inhibits neutrophil activation, independently of the complement system. Our results implicate bacterial binding of sialic acid-binding lectins (Siglecs) on the neutrophil surface, which dampens neutrophil antimicrobial responses. This work identifies a new role for sialylation in protecting N. gonorrhoeae from cellular innate immunity, which can be targeted to enhance the human immune response in gonorrhea.


Subject(s)
Gonorrhea , N-Acetylneuraminic Acid , Neisseria gonorrhoeae , Neutrophil Activation , Neutrophils , Sialic Acid Binding Immunoglobulin-like Lectins , Neisseria gonorrhoeae/immunology , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/metabolism , Humans , N-Acetylneuraminic Acid/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Gonorrhea/immunology , Gonorrhea/microbiology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Lipopolysaccharides/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Respiratory Burst , Host-Pathogen Interactions/immunology , Immune Evasion
14.
Gut Microbes ; 16(1): 2334967, 2024.
Article in English | MEDLINE | ID: mdl-38630006

ABSTRACT

Human milk oligosaccharides (HMOs) are vital milk carbohydrates that help promote the microbiota-dependent growth and immunity of infants. Sialic acid (SA) is a crucial component of sialylated milk oligosaccharides (S-MOs); however, the effects of SA supplementation in lactating mothers on S-MO biosynthesis and their breastfed infants are unknown. Probiotic intervention during pregnancy or lactation demonstrates promise for modulating the milk glycobiome. Here, we evaluated whether SA and a probiotic (Pro) mixture could increase S-MO synthesis in lactating mothers and promote the microbiota development of their breastfed neonates. The results showed that SA+Pro intervention modulated the gut microbiota and 6'-SL contents in milk of maternal rats more than the SA intervention, which promoted Lactobacillus reuteri colonization in neonates and immune development. Deficient 6'-SL in the maternal rat milk of St6gal1 knockouts (St6gal1-/-) disturbed intestinal microbial structures in their offspring, thereby impeding immune tolerance development. SA+Pro intervention in lactating St6gal1± rats compromised the allergic responses of neonates by promoting 6'-SL synthesis and the neonatal gut microbiota. Our findings from human mammary epithelial cells (MCF-10A) indicated that the GPR41-PI3K-Akt-PPAR pathway helped regulate 6'-SL synthesis in mammary glands after SA+Pro intervention through the gut - breast axis. We further validated our findings using a human-cohort study, confirming that providing SA+Pro to lactating Chinese mothers increased S-MO contents in their breast milk and promoted gut Bifidobacterium spp. and Lactobacillus spp. colonization in infants, which may help enhance immune responses. Collectively, our findings may help alter the routine supplementation practices of lactating mothers to modulate milk HMOs and promote the development of early-life gut microbiota and immunity.


Subject(s)
Gastrointestinal Microbiome , N-Acetylneuraminic Acid , Female , Infant , Pregnancy , Humans , Animals , Rats , Lactation , Cohort Studies , Phosphatidylinositol 3-Kinases , Milk, Human , Immunity
15.
Langmuir ; 40(13): 7067-7077, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38518180

ABSTRACT

The importance of sialic acid on cell functions has been recently unveiled, and consequently, great attention has been paid to its interaction with tumor cells. In this line of research, we have realized phosphorene nanosheets functionalized with sialic acid molecules for biological applications with no need for another linker molecule. The formation of phosphorene sheets is feasible by using hydrogen plasma treatment and conversion of amorphous phosphorus on silicon substrates into highly crystalline nanosheets. Through immersion of these freshly prepared nanosheets into an aqueous solution containing sialic acid molecules, the formation of chemical binding between biomolecules and P atoms is initiated to form a carpet-like coverage. We have studied these structures by using Raman spectroscopy, electron microscopy, FTIR-ATR spectroscopy, and X-ray photoelectron spectroscopy. While XPS supports the passivation of sialic-activated phosphorene nanosheets (SAP) against oxidation in air or aqueous solutions, the FTIR analysis corroborates the evolution of P-O-C and P-C bonds between such biomolecules and the sheet surface. Moreover, the high-resolution TEM images demonstrate a considerable reduction in the lattice spacing from 0.32 nm for pristine phosphorene to 0.30 nm. Similarly, Raman spectroscopy depicts a shift in A2g in-plane vibrations, owing to the evolution of stress in the passivated sheets. To investigate their biocompatibility, we examined the toxicity of these bioactivated structures and observed no or little sign of toxicity. For the latter evaluation, we exploited MTT, flow cytometry, and animal models for in vivo investigations.


Subject(s)
N-Acetylneuraminic Acid , Water , Animals , Oxidation-Reduction
16.
Molecules ; 29(6)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38542909

ABSTRACT

N-glycolylneuraminic acid (Neu5Gc), a sialic acid predominantly found in the non-neurohumoral fluids of hind-mouthed animals, is incapable of synthesizing Neu5Gc due to a deletion in the CMAH exon of the gene encoding human CMP-Neu5Gc hydroxylase. But consumption of animal-derived foods that contain Neu5Gc, such as red meat, can instigate an immune response in humans, as Neu5Gc is recognized as a foreign substance by the human immune system. This recognition leads to the production of anti-Neu5Gc antibodies, subsequently resulting in chronic inflammation. When Neu5Gc is consumed excessively or frequently, it may contribute to the development of heart disease and cancer. This makes Neu5Gc, an endogenous pathogenic factor derived from red meat, a new hot topic in red meat safety research. In this study, aptamers obtained by the magnetic bead SELEX technique were subjected to homology and secondary structure prediction analysis as well as affinity determination. The result indicated that the aptamer 2B.N2A9 exhibited a robust binding affinity, with an affinity constant (Ka) of 1.87 × 108 L/mol. This aptamer demonstrated optimal binding specificity within a pH range of 5.4 to 7.4. Molecular docking analysis further revealed that aptamer 2B.N2A9 formed stable binding interactions with the target Neu5Gc at specific sites, namely G-14, C-15, G-13, G-58, G-60, and C-59. An Enzyme-Linked Oligonucleotide Sorbent Assay (ELOSA) methodology was established to detect the endogenous pathogenic factor Neu5Gc present in red meat. This method demonstrated a limit of detection (LOD) of 0.71 ng/mL, along with an average recovery rate of 92.23%. The aptamer obtained in this study exhibited favorable binding properties to Neu5Gc. The assay was relatively convenient and demonstrated good sensitivity. Further investigation into the distribution of Neu5Gc in various red meats is of public health significance and scientific potential. A practical detection method should be provided to guide red meat diets and ensure the nutrition and safety of meat products.


Subject(s)
N-Acetylneuraminic Acid , Red Meat , Animals , Humans , Molecular Docking Simulation , Inflammation , Oligonucleotides
17.
J Am Chem Soc ; 146(12): 8780-8786, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38497732

ABSTRACT

The small RNAs on living cell membranes were recently found to be N-glycosylated and terminated with sialic acids, although the glycosylation sites and potential functions remain unclear. Herein, we designed a second-generation hierarchical coding strategy (HieCo 2) for in situ visualization of cell surface RNA-specific sialylation. After covalently binding DNA codes to sialic acids and then binding a DNA code to a target RNA via sequence specificity, cascade decoding processes were performed with subsequent signal amplification that enabled sensitive in situ visualization of low-abundance Y5 RNA-specific sialic acids on living cell membranes. The proposed strategy unveils the number of glycosylation sites on a single RNA and reveals the binding preference of glycosylated RNAs to different sialic acid binding-immunoglobulin lectin-type receptors, demonstrating a new route for exploration of the glycosylated RNA-related biological and pathological processes.


Subject(s)
RNA , Sialic Acids , Glycosylation , RNA/metabolism , Cell Membrane/metabolism , Sialic Acids/metabolism , DNA/metabolism , N-Acetylneuraminic Acid/metabolism
18.
Microb Pathog ; 190: 106628, 2024 May.
Article in English | MEDLINE | ID: mdl-38508422

ABSTRACT

Rotavirus infections in suckling and weaning piglets cause severe dehydration and death, resulting in significant economic losses in the pig breeding industry. With the continuous emergence of porcine rotavirus (PoRV) variants and poor vaccine cross-protection among various genotypes, there is an urgent need to develop alternative strategies such as seeking effective antiviral products from nature, microbial metabolites and virus-host protein interaction. Sialidases play a crucial role in various physiopathological processes and offer a promising target for developing antivirus drugs. However, the effect of bacterial-derived sialidases on the infection of PoRVs remains largely unknown. Herein, we investigated the impact of bacterial-derived sialidases (sialidase Cp and Vc) on PoRV strain OSU(Group A) infection, using differentiated epithelial monkey kidney cells (MA104) as a model. Our results indicated that the pretreatment of MA104 with exogenous sialidases effectively suppressed PoRV OSU in a concentration-dependent manner. Notably, even at a concentration of 0.01 µU/mL, sialidases significantly inhibited the virus (MOI = 0.01). Meanwhile, we found that sialidase Vc pretreatment sharply reduced the binding rate of PoRV OSU. Last, we demonstrated that PoRV OSU might recognize α-2,3-linked sialic acid as the primary attachment factor in MA104. Our findings provide new insights into the underlying mechanism of PoRV OSU infections, shedding lights on the development of alternative antivirus approaches based on bacteria-virus interaction.


Subject(s)
Neuraminidase , Rotavirus Infections , Rotavirus , Virus Replication , Animals , Neuraminidase/metabolism , Neuraminidase/genetics , Rotavirus/drug effects , Rotavirus/physiology , Swine , Virus Replication/drug effects , Cell Line , Epithelial Cells/virology , Epithelial Cells/microbiology , Virus Attachment/drug effects , N-Acetylneuraminic Acid/metabolism , N-Acetylneuraminic Acid/pharmacology , Antiviral Agents/pharmacology , Haplorhini , Swine Diseases/virology , Swine Diseases/microbiology
19.
Langmuir ; 40(14): 7471-7478, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38554266

ABSTRACT

Neuraminidases (NA) are sialic acid-cleaving enzymes that are used by both bacteria and viruses. These enzymes have sialoside structure-related binding and cleaving preferences. Differentiating between these enzymes requires using a large array of hard-to-access sialosides. In this work, we used electrochemical impedimetric biosensing to differentiate among several pathogene-related NAs. We used a limited set of sialosides and tailored the surface properties. Various sialosides were grafted on two different surfaces with unique properties. Electrografting on glassy carbon electrodes provided low-density sialoside-functionalized surfaces with a hydrophobic submonolayer. A two-step assembly on gold electrodes provided a denser sialoside layer on a negatively charged submonolayer. The synthesis of each sialoside required dozens of laborious steps. Utilizing the unique protein-electrode interaction modes resulted in richer biodata without increasing the synthetic load. These principles allowed for profiling NAs and determining the efficacy of various antiviral inhibitors.


Subject(s)
Biosensing Techniques , Sialic Acids , Sialic Acids/chemistry , Neuraminidase/chemistry , Neuraminidase/metabolism , N-Acetylneuraminic Acid/chemistry , Bacteria
20.
Microbiology (Reading) ; 170(3)2024 03.
Article in English | MEDLINE | ID: mdl-38488830

ABSTRACT

Sialic acid (Sia) transporters are critical to the capacity of host-associated bacteria to utilise Sia for growth and/or cell surface modification. While N-acetyl-neuraminic acid (Neu5Ac)-specific transporters have been studied extensively, little is known on transporters dedicated to anhydro-Sia forms such as 2,7-anhydro-Neu5Ac (2,7-AN) or 2,3-dehydro-2-deoxy-Neu5Ac (Neu5Ac2en). Here, we used a Sia-transport-null strain of Escherichia coli to investigate the function of members of anhydro-Sia transporter families previously identified by computational studies. First, we showed that the transporter NanG, from the Glycoside-Pentoside-Hexuronide:cation symporter family, is a specific 2,7-AN transporter, and identified by mutagenesis a crucial functional residue within the putative substrate-binding site. We then demonstrated that NanX transporters, of the Major Facilitator Superfamily, also only transport 2,7-AN and not Neu5Ac2en nor Neu5Ac. Finally, we provided evidence that SiaX transporters, of the Sodium-Solute Symporter superfamily, are promiscuous Neu5Ac/Neu5Ac2en transporters able to acquire either substrate equally well. The characterisation of anhydro-Sia transporters expands our current understanding of prokaryotic Sia metabolism within host-associated microbial communities.


Subject(s)
N-Acetylneuraminic Acid , N-Acetylneuraminic Acid/analogs & derivatives , Organic Anion Transporters , Symporters , N-Acetylneuraminic Acid/chemistry , Symporters/genetics , Symporters/metabolism , Bacteria/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...