Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.045
Filter
1.
Glycobiology ; 34(10)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39244665

ABSTRACT

Lipooligosaccharides are the most abundant cell surface glycoconjugates on the outer membrane of Gram-negative bacteria. They play important roles in host-microbe interactions. Certain Gram-negative pathogenic bacteria cap their lipooligosaccharides with the sialic acid, N-acetylneuraminic acid (Neu5Ac), to mimic host glycans that among others protects these bacteria from recognition by the hosts immune system. This process of molecular mimicry is not fully understood and remains under investigated. To explore the functional role of sialic acid-capped lipooligosaccharides at the molecular level, it is important to have tools readily available for the detection and manipulation of both Neu5Ac on glycoconjugates and the involved sialyltransferases, preferably in live bacteria. We and others have shown that the native sialyltransferases of some Gram-negative bacteria can incorporate extracellular unnatural sialic acid nucleotides onto their lipooligosaccharides. We here report on the expanded use of native bacterial sialyltransferases to incorporate neuraminic acids analogs with a reporter group into the lipooligosaccharides of a variety of Gram-negative bacteria. We show that this approach offers a quick strategy to screen bacteria for the expression of functional sialyltransferases and the ability to use exogenous CMP-Neu5Ac to decorate their glycoconjugates. For selected bacteria we also show this strategy complements two other glycoengineering techniques, Metabolic Oligosaccharide Engineering and Selective Exo-Enzymatic Labeling, and that together they provide tools to modify, label, detect and visualize sialylation of bacterial lipooligosaccharides.


Subject(s)
Lipopolysaccharides , Sialyltransferases , Sialyltransferases/metabolism , Sialyltransferases/genetics , Sialyltransferases/chemistry , Lipopolysaccharides/metabolism , Lipopolysaccharides/chemistry , Neuraminic Acids/metabolism , Neuraminic Acids/chemistry , Gram-Negative Bacteria/metabolism , N-Acetylneuraminic Acid/metabolism , N-Acetylneuraminic Acid/chemistry
2.
Nat Commun ; 15(1): 7735, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232002

ABSTRACT

Breastfeeding provides many health benefits, but its impact on respiratory health remains unclear. This study addresses the complex and dynamic nature of the mother-milk-infant triad by investigating maternal genomic factors regulating human milk oligosaccharides (HMOs), and their associations with respiratory health among human milk-fed infants. Nineteen HMOs are quantified from 980 mothers of the CHILD Cohort Study. Genome-wide association studies identify HMO-associated loci on chromosome 19p13.3 and 19q13.33 (lowest P = 2.4e-118), spanning several fucosyltransferase (FUT) genes. We identify novel associations on chromosome 3q27.3 for 6'-sialyllactose (P = 2.2e-9) in the sialyltransferase (ST6GAL1) gene. These, plus additional associations on chromosomes 7q21.32, 7q31.32 and 13q33.3, are replicated in the independent INSPIRE Cohort. Moreover, gene-environment interaction analyses suggest that fucosylated HMOs may modulate overall risk of recurrent wheeze among preschoolers with variable genetic risk scores (P < 0.01). Thus, we report novel genetic factors associated with HMOs, some of which may protect the respiratory health of children.


Subject(s)
Genome-Wide Association Study , Milk, Human , Oligosaccharides , Sialyltransferases , Humans , Milk, Human/chemistry , Milk, Human/metabolism , Female , Oligosaccharides/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism , Infant , Male , Child, Preschool , Fucosyltransferases/genetics , Breast Feeding , Respiratory Sounds/genetics , Gene-Environment Interaction , Polymorphism, Single Nucleotide , Adult , Cohort Studies , Mothers , Child , Chromosomes, Human, Pair 3/genetics , Lactose/analogs & derivatives
3.
PLoS One ; 19(9): e0311212, 2024.
Article in English | MEDLINE | ID: mdl-39348343

ABSTRACT

Colorectal cancer (CRC) has become a significant global health concern and ranks among the leading causes of morbidity and mortality worldwide. Due to its malignant nature, current immunotherapeutic treatments are used to tackle this issue. However, not all patients respond positively to treatment, thereby limiting clinical effectiveness and requiring the identification of novel therapeutic targets to optimise current strategies. The putative ligand of Siglec-15, Sialyl-Tn (STn), is associated with tumour progression and is synthesised by the sialyltransferases ST6GALNAC1 and ST6GALNAC2. However, the deregulation of both sialyltransferases within the literature remain limited, and the involvement of microRNAs (miRNAs) in STn production require further elucidation. Here, we identified miRNAs involved in the regulation of ST6GALNAC1 via a computational approach and further analysis of miRNA binding sites were determined. In silico tools predicted miR-21, miR-30e and miR-26b to regulate the ST6GALNAC1 gene, all of which had shown significant upregulated expression in the tumour cohort. Moreover, each miRNA displayed a high binding affinity towards the seed region of ST6GALNAC1. Additionally, enrichment analysis outlined pathways associated with several cancer hallmarks, including epithelial to mesenchymal transition (EMT) and MYC targets associated with tumour progression. Furthermore, our in silico findings demonstrated that the ST6GALNAC1 expression profile was significantly downregulated in CRC tumours, and its low expression correlated with poor survival outcomes when compared with patient survival data. In comparison to its counterpart, there were no significant differences in the expression of ST6GALNAC2 between normal and malignant tissues, which was further evidenced in our immunohistochemistry analysis. Immunohistochemistry staining highlighted significantly higher expression was more prevalent in normal human tissues with regard to ST6GALNAC1. In conclusion, the integrated in silico analysis highlighted that STn production is not reliant on deregulated sialyltransferase expression in CRC, and ST6GALNAC1 expression is regulated by several oncomirs. We proposed the involvement of other sialyltransferases in the production of the STn antigen and CRC progression via the Siglec-15/Sia axis.


Subject(s)
Colorectal Neoplasms , Disease Progression , Gene Expression Regulation, Neoplastic , MicroRNAs , Sialyltransferases , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Male , Female , Cell Line, Tumor , Clinical Relevance , beta-D-Galactoside alpha 2-6-Sialyltransferase , Antigens, Tumor-Associated, Carbohydrate , Antigens, CD
4.
Int J Biol Macromol ; 278(Pt 4): 135196, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39256125

ABSTRACT

In neurological diseases, the regulation of autophagy plays a crucial role in their pathology, particularly the relationship between autophagy and hepatic encephalopathy (HE) which merits detailed investigation. Glycosphingolipids are abundant and broadly functional in the nervous system and are closely associated with autophagy. However, the specific link and mechanisms between glycosphingolipids and autophagy in HE remain unclear. This study aims to explore the impact of glycosphingolipid changes on the autophagy in HE and its potential mechanisms. Utilizing lectin microarrays, we observed elevated expression levels of α2-3 sialylated glycosphingolipid in the brain tissue of HBV transgenic mice and ammonia-induced astrocyte models, suggesting that the increase in α2-3 sialylated glycosphingolipid is related to HE. Further research revealed that the increased expression of α2-3 sialylated glycosphingolipid, mediated by ST3GAL2, affects autophagy by regulating the autophagy initiation complex Vps34-Beclin-1. In summary, our research not only comprehensively reveals the changes in brain glycosphingolipid during HBV-related HE but also elucidates the interactions and regulatory mechanisms between α2-3 sialylated glycosphingolipid and autophagy. This study provides a new perspective on understanding the pathogenesis of HE and offers novel theories and targets for future research and treatment strategies.


Subject(s)
Autophagy , Glycosphingolipids , Hepatic Encephalopathy , Sialyltransferases , Animals , Hepatic Encephalopathy/metabolism , Hepatic Encephalopathy/pathology , Mice , Glycosphingolipids/metabolism , Sialyltransferases/metabolism , Sialyltransferases/genetics , Mice, Transgenic , Brain/metabolism , Brain/pathology , Humans , beta-Galactoside alpha-2,3-Sialyltransferase , Astrocytes/metabolism , Male
5.
Carbohydr Polym ; 343: 122471, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39174097

ABSTRACT

Sialylation, a crucial post-translational modification of glycoconjugates, entails the attachment of sialic acid (SA) to the terminal glycans of glycoproteins and glycolipids through a tightly regulated enzymatic process involving various enzymes. This review offers a comprehensive exploration of sialylation within the gut, encompassing its involvement in mucosal protection and its impact on disease progression. The sialylation of mucins and epithelial glycoproteins contributes to the integrity of the intestinal mucosal barrier. Furthermore, sialylation regulates immune responses in the gut, shaping interactions among immune cells, as well as their activation and tolerance. Additionally, the gut microbiota and gut-brain axis communication are involved in the role of sialylation in intestinal health. Altered sialylation patterns have been implicated in various intestinal diseases, including inflammatory bowel disease (IBD), colorectal cancer (CRC), and other intestinal disorders. Emerging research underscores sialylation as a promising avenue for diagnostic, prognostic, and therapeutic interventions in intestinal diseases. Potential strategies such as sialic acid supplementation, inhibition of sialidases, immunotherapy targeting sialylated antigens, and modulation of sialyltransferases have been utilized in the treatment of intestinal diseases. Future research directions will focus on elucidating the molecular mechanisms underlying sialylation alterations, identifying sialylation-based biomarkers, and developing targeted interventions for precision medicine approaches.


Subject(s)
Intestinal Mucosa , N-Acetylneuraminic Acid , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Animals , N-Acetylneuraminic Acid/metabolism , Gastrointestinal Microbiome , Sialyltransferases/metabolism , Mucins/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/immunology
6.
J Biotechnol ; 394: 24-33, 2024 Nov 10.
Article in English | MEDLINE | ID: mdl-39103019

ABSTRACT

The production of therapeutic glycoproteins is primarily expensive due to the necessity of culturing mammalian cells. These systems often require complex and costly culture media and typically yield low amounts of protein. Leishmania tarentolae, a non-pathogenic protozoan to mammals, has emerged as a cost-effective alternative system for heterologous glycoprotein expression due to its suitability for large-scale production using low-cost culture media, and its ability to perform mammalian-like post-translational modifications, including glycosylation. Nevertheless, differences in the carbohydrate residues at the end of N-glycan chains are observed in Leishmania compared to mammalian cells due to the absence of biosynthetic enzymes in Leishmania that are required for the incorporation of terminal sialic acid. In this study, a genetically optimized L. tarentolae cell line was engineered for the production of recombinant interferon-ß (IFN-ß) featuring a complete mammalian N-glycosylation profile. Genomic and metabolomic analyses revealed that heterologous expression of the sialyltransferase enzyme and cultivation in a medium containing sialic acid were sufficient to generate mammalian-like protein N-glycosylation. N-glycan mass spectrometry analysis demonstrated a glycosylation pattern compatible with the incorporation of sialic acid into the glycan structure. In vitro IFN-ß activity indicated that the expressed protein exhibited reduced inflammatory effects compared to IFN-beta produced by other platforms, such as bacteria, non-optimized L. tarentolae, and mammalian cells.


Subject(s)
Interferon-beta , Leishmania , Recombinant Proteins , Sialyltransferases , Glycosylation , Leishmania/genetics , Leishmania/metabolism , Leishmania/enzymology , Humans , Interferon-beta/metabolism , Interferon-beta/genetics , Sialyltransferases/metabolism , Sialyltransferases/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Animals , Polysaccharides/metabolism , Mice
7.
Cell Death Differ ; 31(10): 1302-1317, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39164456

ABSTRACT

The existence of heterogeneity has plunged cancer treatment into a challenging dilemma. We profiled malignant epithelial cells from 5 gastric adenocarcinoma patients through single-cell sequencing (scRNA-seq) analysis, demonstrating the heterogeneity of gastric adenocarcinoma (GA), and identified the CCKBR+ stem cell-like cancer cells associated poorly differentiated and worse prognosis. We further conducted targeted analysis using single-cell transcriptome libraries, including 40 samples, to confirm these screening results. In addition, we revealed that FOXOs are involved in the progression and development of CCKBR+ gastric adenocarcinoma. Inhibited the expression of FOXOs and disrupting cancer cell stemness reduce the CCKBR+ GA organoid formation and impede tumor progression. Mechanically, CUT&Tag sequencing and Lectin pulldown revealed that FOXOs can activate ST3GAL3/4/5 as well as ST6GALNAC6, promoting elevated sialyation levels in CCKBR+ tumor cells. This FOXO-sialyltransferase axis contributes to the maintenance of homeostasis and the growth of CCKBR+ tumor cells. This insight provides novel perspectives for developing targeted therapeutic strategies aimed at the treating CCKBR associated gastric cancer.


Subject(s)
Forkhead Transcription Factors , Stomach Neoplasms , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Humans , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Receptor, Cholecystokinin B/metabolism , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/antagonists & inhibitors , Cell Line, Tumor , Sialyltransferases/metabolism , Sialyltransferases/genetics , Animals , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Adenocarcinoma/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/genetics , Mice, Nude
8.
J Med Chem ; 67(16): 14586-14608, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39136957

ABSTRACT

In the aftermath of the COVID-19 pandemic, opportunities to modulate biological pathways common to the lifecycles of viruses need to be carefully considered. N-linked glycosylation in humans is mediated exclusively by the oligosaccharyltransferase complex and is frequently hijacked by viruses to facilitate infection. As such, STT3A/B, the catalytic domain of the OST complex, became an intriguing drug target with broad-spectrum antiviral potential. However, due to the critical role N-linked glycosylation plays in a number of fundamental human processes, the toxicological ramifications of STT3A/B inhibition required attention commensurate to that given to antiviral efficacy. Herein, we describe how known STT3A/B inhibitor NGI-1 inspired the discovery of superior tool compounds which were evaluated in in vitro efficacy and translational safety (e.g., CNS, cardiovascular, liver) studies. The described learnings will appeal to those interested in the therapeutic utility of modulating N-linked glycosylation as well as the broader scientific community.


Subject(s)
Antiviral Agents , Membrane Proteins , SARS-CoV-2 , Sialyltransferases , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Humans , Animals , SARS-CoV-2/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Sialyltransferases/antagonists & inhibitors , Sialyltransferases/metabolism , Drug Discovery , COVID-19 Drug Treatment , Glycosylation , Rats , Hexosyltransferases
9.
Carbohydr Res ; 544: 109242, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39167930

ABSTRACT

Human glycans are capped with sialic acids and these nine-carbon sugars mediate many of the biological functions and interactions of glycans. Structurally diverse sialic acid caps mark human cells as self and they form the ligands for the Siglec immune receptors and other glycan-binding proteins. Sialic acids enable host interactions with the human microbiome and many human pathogens utilize sialic acids to infect host cells. Alterations in sialic acid-carrying glycans, sialoglycans, can be found in every major human disease including inflammatory conditions and cancer. Twenty sialyltransferase family members in the Golgi apparatus of human cells transfer sialic acids to distinct glycans and glycoconjugates. Sialyltransferases catalyze specific reactions to form unique sialoglycans or they have shared functions where multiple family members generate the same sialoglycan product. Moreover, some sialyltransferases compete for the same glycan substrate, but create different sialic acid caps. The redundant and competing functions make it difficult to understand the individual roles of the human sialyltransferases in biology and to reveal the specific contributions to pathobiological processes. Recent insights hint towards the existence of biosynthetic rules formed by the individual functions of sialyltransferases, their interactions, and cues from the local Golgi environment that coordinate sialoglycan biosynthesis. In this review, we discuss the current structural and functional understanding of the human sialyltransferase family and we review recent technological advances that enable the dissection of individual sialyltransferase activities.


Subject(s)
Sialyltransferases , Humans , Sialyltransferases/metabolism , Polysaccharides/metabolism , Polysaccharides/chemistry
10.
Clin Neuropathol ; 43(4): 113-126, 2024.
Article in English | MEDLINE | ID: mdl-39120072

ABSTRACT

Glioma is the most common brain tumor, accounting for a large majority of cancer-related deaths. ß-galactoside α2, 6 sialyltranferase 1 (ST6Gal1), the primary enzyme responsible for the conjugation of α2, 6 sialic acids to protein and lipid targets, is strongly associated with the occurrence and development of several brain tumor types. Still, the expression, targets, and functions of ST6Gal1 in glioma patients remain undetermined. As sialylation of the Ig-like cell adhesion family molecules have prominent roles in the latter's regulation in other biological contexts, we screened for members that have potential to be regulated by ST6Gal1 in silico and examined co-expressed protein modules using data derived from the Cancer Genome Atlas (TCGA) database, and we identified neural cell adhesion molecule (NCAM1) as a major ST6Gal1-interacting target. Bioinformatic binding analysis confirmed the interaction of ST6Gal1 and NCAM1. Immunohistochemistry was then used to evaluate post-operative samples from 156 patients with gliomas. ST6Gal1 and NCAM1 were co-expressed in gliomas, and their expression correlated significantly (p = 0.002) by univariate analysis. Our study also found that the expression levels of both ST6Gal1 and NCAM1 corresponded negatively with glioma grade, isocitrate dehydrogenase (IDH) mutation, and proliferation index (Ki67). Consistently, Kaplan-Meier survival curves showed that lower ST6Gal1 and NCAM1 protein levels are linked to unfavorable outcomes in glioma patients (p = 0.018 and p < 0.001, respectively). Our data indicate that ST6Gal1 may participate in the inhibition of oncogenesis and malignant progression via interacting with and targeting NCAM1 in glioma, thus presenting a novel strategy for intervention.


Subject(s)
Brain Neoplasms , Glioma , Sialyltransferases , Humans , Glioma/pathology , Glioma/genetics , Glioma/metabolism , Sialyltransferases/genetics , Sialyltransferases/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Male , Female , Adult , Middle Aged , Antigens, CD/metabolism , CD56 Antigen/metabolism , Aged , Biomarkers, Tumor/metabolism , beta-D-Galactoside alpha 2-6-Sialyltransferase
11.
Int Immunopharmacol ; 138: 112569, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38959540

ABSTRACT

BACKGROUND: Bladder cancer (BLCA) is one of the top ten most common cancers in the world. Aberrant sialylation is a common feature in tumorigenesis and tumor immunity. This study seeks to explore the potential impact of sialyltransferase ST3Gal5 on BLCA. METHODS: Initially, glycosyltransferase-related DEGs (GRDEGs) were identified using multiple bioinformatics approaches in TCGA-BLCA cohort and validated using GEO databases. Clinical prognosis integration facilitated the determination of ST3Gal5 as an independent prognostic factor in BLCA, employing univariate and multivariate Cox regression analyses. Immune cell infiltration was assessed via CIBERSORT and ssGSEA analyses, while HLA and immune checkpoint genes' levels, along with drug sensitivity, were evaluated in low- and high-ST3Gal5 groups. The TIDE and IPS scores were used to gauge the immune checkpoint blockade (ICB) response. Furthermore, functional experiments, both in vivo and in vitro, were conducted to elucidate the biological roles of ST3Gal5. RESULTS: In agreement with bioinformatics findings, ST3Gal5 expression was down-regulated in BLCA tissues and cells, correlating with poorer prognostic outcomes. The StromalScore, ImmuneScore, and ESTIMATEScore were significantly elevated in low-ST3Gal5 group. Moreover, the levels of HLA and immune checkpoint genes were upregulated in low-ST3Gal5 group. Down-regulated ST3Gal5 promoted the proliferation, migration, and invasion of BLCA cells in vivo and in vitro. CONCLUSION: Our findings demonstrated that low ST3Gal5 level promoted tumorigenesis and progression of BLCA, implying its potential as a predictive biomarker and therapeutic target.


Subject(s)
Computational Biology , Gene Expression Regulation, Neoplastic , Sialyltransferases , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/mortality , Sialyltransferases/genetics , Sialyltransferases/metabolism , Humans , Computational Biology/methods , Animals , Cell Line, Tumor , Prognosis , Cell Proliferation , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , beta-Galactoside alpha-2,3-Sialyltransferase , Mice , Male , Female , Cell Movement , Mice, Nude
12.
Front Endocrinol (Lausanne) ; 15: 1289653, 2024.
Article in English | MEDLINE | ID: mdl-38978616

ABSTRACT

Background: Type 1 (T1D) and type 2 (T2D) diabetes lead to an aberrant metabolism of sialoglycoconjugates and elevated free serum sialic acid (FSSA) level. The present study evaluated sialidase and sialyltranferase activities in serum and some organs relevant to diabetes at early and late stages of T1D and T2D. Methods: Sialic acid level with sialidase and sialyltransferase activities were monitored in the serum, liver, pancreas, skeletal muscle and kidney of diabetic animals at early and late stages of the diseases. Results: The FSSA and activity of sialidase in the serum were significantly increased at late stage of both T1D and T2D while sialic acid level in the liver was significantly decreased in the early and late stages of T1D and T2D, respectively. Furthermore, the activity of sialidase was significantly elevated in most of the diabetes-relevant organs while the activity of sialyltransferase remained largely unchanged. A multiple regression analysis revealed the contribution of the liver to the FSSA while pancreas and kidney contributed to the activity of sialidase in the serum. Conclusions: We concluded that the release of hepatic sialic acid in addition to pancreatic and renal sialidase might (in)directly contribute to the increased FSSA during both types of diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , N-Acetylneuraminic Acid , Neuraminidase , Sialyltransferases , Animals , Neuraminidase/metabolism , Sialyltransferases/metabolism , N-Acetylneuraminic Acid/metabolism , Diabetes Mellitus, Type 2/metabolism , Rats , Male , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/blood , Liver/metabolism , Liver/enzymology , Rats, Wistar , Pancreas/metabolism , Pancreas/enzymology , Kidney/metabolism , Muscle, Skeletal/metabolism
13.
BMC Musculoskelet Disord ; 25(1): 565, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033138

ABSTRACT

INTRODUCTION: Growth plate damage in long bones often results in progressive skeletal growth imbalance and deformity, leading to significant physical problems. Gangliosides, key glycosphingolipids in cartilage, are notably abundant in articular cartilage and regulate chondrocyte homeostasis. This suggests their significant roles in regulating growth plate cartilage repair. METHODS: Chondrocytes from 3 to 5 day-old C57BL/6 mice underwent glycoblotting and mass spectrometry. Based on the results of the glycoblotting analysis, we employed GD3 synthase knockout mice (GD3-/-), which lack b-series gangliosides. In 3-week-old mice, physeal injuries were induced in the left tibiae, with right tibiae sham operated. Tibiae were analyzed at 5 weeks postoperatively for length and micro-CT for growth plate height and bone volume at injury sites. Tibial shortening ratio and bone mineral density were measured by micro-CT. RESULTS: Glycoblotting analysis indicated that b-series gangliosides were the most prevalent in physeal chondrocytes among ganglioside series. At 3 weeks, GD3-/- exhibited reduced tibial shortening (14.7 ± 0.2 mm) compared to WT (15.0 ± 0.1 mm, P = 0.03). By 5 weeks, the tibial lengths in GD3-/- (16.0 ± 0.4 mm) closely aligned with sham-operated lengths (P = 0.70). Micro-CT showed delayed physeal bridge formation in GD3-/-, with bone volume measuring 168.9 ± 5.8 HU at 3 weeks (WT: 180.2 ± 3.2 HU, P = 0.09), but normalizing by 5 weeks. CONCLUSION: This study highlights that GD3 synthase knockout mice inhibit physeal bridge formation after growth plate injury, proposing a new non-invasive approach for treating skeletal growth disorders.


Subject(s)
Chondrocytes , Gangliosides , Growth Plate , Mice, Inbred C57BL , Mice, Knockout , Animals , Growth Plate/pathology , Growth Plate/metabolism , Gangliosides/metabolism , Chondrocytes/metabolism , Mice , Leg Length Inequality , Tibia/diagnostic imaging , Tibia/pathology , Tibia/metabolism , Tibia/growth & development , X-Ray Microtomography , Sialyltransferases/deficiency , Sialyltransferases/genetics , Sialyltransferases/metabolism , Disease Models, Animal
14.
Glycoconj J ; 41(3): 175-183, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38958800

ABSTRACT

Altered glycosylation is a common feature of cancer cells. Some subsets of glycans are found to be frequently enriched on the tumor cell surface and implicated in different tumor phenotypes. Among these, changes in sialylation have long been associated with metastatic cell behaviors such as invasion and enhanced cell survival. Sialylation typically exists in three prominent linkages: α2,3, α2,6, and α2,8, catalyzed by a group of sialyltransferases. The aberrant expression of all three linkages has been related to cancer progression. The increased α2,6 sialylation on N-glycans catalyzed by ß-galactoside α2,6 sialyltransferase 1 (ST6Gal1) is frequently observed in many cancers. In contrast, functions of α2,3 sialylation on N-glycans catalyzed by at least three ß-galactoside α2,3-sialyltransferases, ST3Gal3, ST3Gal4, and ST3Gal6 remain elusive due to a possibility of compensating for one another. In this minireview, we briefly describe functions of sialylation and recent findings that different α2,3 sialyltransferases specifically modify target proteins, as well as sialylation regulatory mechanisms vis a complex formation among integrin α3ß1, Golgi phosphoprotein 3 (GOLPH3), phosphatidylinositol 4-kinase IIα (PI4KIIα), focal adhesion kinase (FAK) and sialyltransferase, which suggests a new concept for the regulation of glycosylation in cell biology.


Subject(s)
Polysaccharides , Sialyltransferases , Humans , Sialyltransferases/metabolism , Polysaccharides/metabolism , Animals , Glycosylation , Neoplasms/metabolism
15.
Sci Rep ; 14(1): 16568, 2024 07 17.
Article in English | MEDLINE | ID: mdl-39019950

ABSTRACT

Mucus stasis is a pathologic hallmark of muco-obstructive diseases, including cystic fibrosis (CF). Mucins, the principal component of mucus, are extensively modified with hydroxyl (O)-linked glycans, which are largely terminated by sialic acid. Sialic acid is a negatively charged monosaccharide and contributes to the biochemical/biophysical properties of mucins. Reports suggest that mucin sialylation may be altered in CF; however, the consequences of reduced sialylation on mucus clearance have not been fully determined. Here, we investigated the consequences of reduced sialylation on the charge state and conformation of the most prominent airway mucin, MUC5B, and defined the functional consequences of reduced sialylation on mucociliary transport (MCT). Reduced sialylation contributed to a lower charged MUC5B form and decreased polymer expansion. The inhibition of total mucin sialylation de novo impaired MCT in primary human bronchial epithelial cells and rat airways, and specific α-2,3 sialylation blockade was sufficient to recapitulate these findings. Finally, we show that ST3 beta-galactoside alpha-2,3-sialyltransferase (ST3Gal1) expression is downregulated in CF and partially restored by correcting CFTR via Elexacaftor/Tezacaftor/Ivacaftor treatment. Overall, this study demonstrates the importance of mucin sialylation in mucus clearance and identifies decreased sialylation by ST3Gal1 as a possible therapeutic target in CF and potentially other muco-obstructive diseases.


Subject(s)
Mucin-5B , Mucus , Humans , Animals , Mucin-5B/metabolism , Rats , Mucus/metabolism , Sialyltransferases/metabolism , N-Acetylneuraminic Acid/metabolism , Mucociliary Clearance , Respiratory Mucosa/metabolism , Cystic Fibrosis/metabolism , Mucins/metabolism , Epithelial Cells/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Bronchi/metabolism
16.
Nanoscale ; 16(31): 14932-14939, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39046038

ABSTRACT

6'-Sialyllactose (6'-SL), the most abundant sialylated human milk oligosaccharide, has attracted attention for its potential application in supplementary infant formulas. Herein, we report a facile strategy to construct a cascade bioreactor for the enzymatic synthesis of 6'-SL by co-immobilizing an enzymatic module consisting of CMP-sialic acid synthase and α-2,6-sialyltransferase into hierarchically porous MIL-53 (HP-MIL-53). The as-prepared HP-MIL-53 showed high enzyme immobilization capacity, reaching 226 mg g-1. Furthermore, the co-immobilized enzymes exhibited higher initial catalytic efficiency, and thermal, pH and storage stability than the free ones. Finally, the 6'-SL yield remained >80% after 13 cycles of use. We expect that HP-MIL-53 would have potential industrial applications in the enzymatic modular synthesis of 6'-SL and other glycans.


Subject(s)
Enzymes, Immobilized , Sialyltransferases , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Sialyltransferases/metabolism , Porosity , Humans , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Oligosaccharides/biosynthesis , N-Acylneuraminate Cytidylyltransferase/metabolism , N-Acylneuraminate Cytidylyltransferase/chemistry , Bioreactors , Milk, Human/chemistry , Milk, Human/metabolism , Lactose/chemistry , Lactose/analogs & derivatives , Lactose/metabolism , Hydrogen-Ion Concentration , beta-D-Galactoside alpha 2-6-Sialyltransferase
17.
Int J Biol Macromol ; 276(Pt 2): 133959, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39029847

ABSTRACT

Escherichia coli F18 (E. coli F18) is the main cause of bacterial diarrhea in piglets. Previous transcriptome reported that ST3GAL1 was associated to E. coli F18 infection. However, its role in mediating the resistance to E. coli F18 remains elusive. Here, we revealed that the downregulation of ST3GAL1 expression contributed to the enhancement of E. coli F18 resistance in IPEC-J2 cells. Bisulfite sequencing identified 26 methylated CpG sites in the ST3GAL1 core promoter. Among these, the ST3GAL1 mRNA levels significantly correlated with methylation levels of the mC-8 site in the specificity protein 1 (SP1) transcription factor (P < 0.01). Interestingly, ST3GAL1 expression may enhances the immune response by activating TLRs signaling, meanwhile decreases the production of the E. coli F18 receptor by inhibiting glycosphingolipid biosynthesis signaling, thereby leading to enhance the resistance to E. coli F18 infection. Besides, low ST3GAL1 expression may increase E. coli resistance by reducing sialylation. Together, these results support the status of ST3GAL1 as a viable target for efforts to modulate E. coli F18 susceptibility, offering a theoretical foundation for the use of this gene as a key biomarker for molecular breeding to improve porcine disease resistance.


Subject(s)
Escherichia coli Infections , Escherichia coli , Sialyltransferases , Animals , Cell Line , CpG Islands , Disease Susceptibility , DNA Methylation , Escherichia coli Infections/genetics , Escherichia coli Infections/veterinary , Promoter Regions, Genetic , Sialyltransferases/genetics , Sialyltransferases/metabolism , Swine , Swine Diseases/genetics , Swine Diseases/microbiology
18.
Mol Cell Proteomics ; 23(9): 100821, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39069074

ABSTRACT

Intrahepatic cholangiocarcinoma (iCCA) has a poor prognosis, and elucidation of the molecular mechanisms underlying iCCA malignancy is of great significance. Glycosylation, an important post-translational modification, is closely associated with tumor progression. Altered glycosylation, including aberrant sialylation resulting from abnormal expression of sialyltransferases (STs) and neuraminidases (NEUs), is a significant feature of cancer cells. However, there is limited information on the roles of STs and NEUs in iCCA malignancy. Here, utilizing our proteogenomic resources from a cohort of 262 patients with iCCA, we identified ST3GAL1 as a prognostically relevant molecule in iCCA. Moreover, overexpression of ST3GAL1 promoted proliferation, migration, and invasion and inhibited apoptosis of iCCA cells in vitro. Through proteomic analyses, we identified the downstream pathway potentially regulated by ST3GAL1, which was the NF-κB signaling pathway, and further demonstrated that this pathway was positively correlated with malignancy in iCCA cells. Notably, glycoproteomics showed that O-glycosylation was changed in iCCA cells with high ST3GAL1 expression. Importantly, the altered O-glycopeptides underscored the potential utility of O-glycosylation profiling as a discriminatory marker for iCCA cells with ST3GAL1 overexpression. Additionally, miR-320b was identified as a post-transcriptional regulator of ST3GAL1, capable of suppressing ST3GAL1 expression and then reducing the proliferation, migration, and invasion abilities of iCCA cell lines. Taken together, these results suggest ST3GAL1 could serve as a promising therapeutic target for iCCA.


Subject(s)
Bile Duct Neoplasms , Cell Movement , Cell Proliferation , Cholangiocarcinoma , Sialyltransferases , Humans , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Sialyltransferases/metabolism , Sialyltransferases/genetics , Cell Line, Tumor , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/genetics , Glycosylation , Male , Female , Apoptosis , Phenotype , Gene Expression Regulation, Neoplastic , Proteomics/methods , Middle Aged , Prognosis , Signal Transduction , Neoplasm Invasiveness , NF-kappa B/metabolism , beta-Galactoside alpha-2,3-Sialyltransferase
19.
Neurobiol Dis ; 199: 106564, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38876323

ABSTRACT

Biallelic variants in the SPG11 gene account for the most common form of autosomal recessive hereditary spastic paraplegia characterized by motor and cognitive impairment, with currently no therapeutic option. We previously observed in a Spg11 knockout mouse that neurodegeneration is associated with accumulation of gangliosides in lysosomes. To test whether a substrate reduction therapy could be a therapeutic option, we downregulated the key enzyme involved in ganglioside biosynthesis using an AAV-PHP.eB viral vector expressing a miRNA targeting St3gal5. Downregulation of St3gal5 in Spg11 knockout mice prevented the accumulation of gangliosides, delayed the onset of motor and cognitive symptoms, and prevented the upregulation of serum levels of neurofilament light chain, a biomarker widely used in neurodegenerative diseases. Importantly, similar results were observed when Spg11 knockout mice were administrated venglustat, a pharmacological inhibitor of glucosylceramide synthase expected to decrease ganglioside synthesis. Downregulation of St3gal5 or venglustat administration in Spg11 knockout mice strongly decreased the formation of axonal spheroids, previously associated with impaired trafficking. Venglustat had similar effect on cultured human SPG11 neurons. In conclusion, this work identifies the first disease-modifying therapeutic strategy in SPG11, and provides data supporting its relevance for therapeutic testing in SPG11 patients.


Subject(s)
Gangliosides , Mice, Knockout , Spastic Paraplegia, Hereditary , Animals , Humans , Mice , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/genetics , Gangliosides/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Mice, Inbred C57BL , Neurofilament Proteins , Neurons/metabolism , Proteins/genetics , Proteins/metabolism , Sialyltransferases/genetics , Sialyltransferases/deficiency , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
20.
Glycobiology ; 34(7)2024 05 26.
Article in English | MEDLINE | ID: mdl-38873803

ABSTRACT

Dendritic cells (DCs) are central for the initiation and regulation of appropriate immune responses. While several studies suggest important regulatory roles of sialoglycans in DC biology, our understanding is still inadequate primarily due to a lack of appropriate models. Previous approaches based on enzymatic- or metabolic-glycoengineering and primary cell isolation from genetically modified mice have limitations related to specificity, stability, and species differences. This study addresses these challenges by introducing a workflow to genetically glycoengineer the human DC precursor cell line MUTZ-3, described to differentiate and maturate into fully functional dendritic cells, using CRISPR-Cas9, thereby providing and validating the first isogenic cell model for investigating glycan alteration on human DC differentiation, maturation, and activity. By knocking out (KO) the ST6GAL1 gene, we generated isogenic cells devoid of ST6GAL1-mediated α(2,6)-linked sialylation, allowing for a comprehensive investigation into its impact on DC function. Glycan profiling using lectin binding assay and functional studies revealed that ST6GAL1 KO increased the expression of important antigen presenting and co-stimulatory surface receptors and a specifically increased activation of allogenic human CD4 + T cells. Additionally, ST6GAL1 KO induces significant changes in surface marker expression and cytokine response to TNFα-induced maturation, and it affects migration and the endocytic capacity. These results indicate that genetic glycoengineering of the isogenic MUTZ-3 cellular model offers a valuable tool to study how specific glycan structures influence human DC biology, contributing to our understanding of glycoimmunology.


Subject(s)
CD4-Positive T-Lymphocytes , Dendritic Cells , Sialic Acids , Sialyltransferases , Tumor Necrosis Factor-alpha , Humans , Dendritic Cells/metabolism , Dendritic Cells/immunology , Sialyltransferases/genetics , Sialyltransferases/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Sialic Acids/metabolism , CRISPR-Cas Systems , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Line , Cell Differentiation , beta-D-Galactoside alpha 2-6-Sialyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL