1
|
Tarantino A, Ciconte G, Melgari D, Frosio A, Ghiroldi A, Piccoli M, Villa M, Creo P, Calamaio S, Castoldi V, Coviello S, Micaglio E, Cirillo F, Locati ET, Negro G, Boccellino A, Mastrocinque F, Ćalović Ž, Ricagno S, Leocani L, Vicedomini G, Santinelli V, Rivolta I, Anastasia L, Pappone C. NaV1.5 autoantibodies in Brugada syndrome: pathogenetic implications. Eur Heart J 2024; 45:4336-4348. [PMID: 39078224 PMCID: PMC11491155 DOI: 10.1093/eurheartj/ehae480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/22/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND AND AIMS Patients suffering from Brugada syndrome (BrS) are predisposed to life-threatening cardiac arrhythmias. Diagnosis is challenging due to the elusive electrocardiographic (ECG) signature that often requires unconventional ECG lead placement and drug challenges to be detected. Although NaV1.5 sodium channel dysfunction is a recognized pathophysiological mechanism in BrS, only 25% of patients have detectable SCN5A variants. Given the emerging role of autoimmunity in cardiac ion channel function, this study explores the presence and potential impact of anti-NaV1.5 autoantibodies in BrS patients. METHODS Using engineered HEK293A cells expressing recombinant NaV1.5 protein, plasma from 50 BrS patients and 50 controls was screened for anti-NaV1.5 autoantibodies via western blot, with specificity confirmed by immunoprecipitation and immunofluorescence. The impact of these autoantibodies on sodium current density and their pathophysiological effects were assessed in cellular models and through plasma injection in wild-type mice. RESULTS Anti-NaV1.5 autoantibodies were detected in 90% of BrS patients vs. 6% of controls, yielding a diagnostic area under the curve of .92, with 94% specificity and 90% sensitivity. These findings were consistent across varying patient demographics and independent of SCN5A mutation status. Electrophysiological studies demonstrated a significant reduction specifically in sodium current density. Notably, mice injected with BrS plasma showed Brugada-like ECG abnormalities, supporting the pathogenic role of these autoantibodies. CONCLUSIONS The study demonstrates the presence of anti-NaV1.5 autoantibodies in the majority of BrS patients, suggesting an immunopathogenic component of the syndrome beyond genetic predispositions. These autoantibodies, which could serve as additional diagnostic markers, also prompt reconsideration of the underlying mechanisms of BrS, as evidenced by their role in inducing the ECG signature of the syndrome in wild-type mice. These findings encourage a more comprehensive diagnostic approach and point to new avenues for therapeutic research.
Collapse
Affiliation(s)
- Adriana Tarantino
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- School of Medicine, University Vita-Salute San Raffaele, Via Olgettina, 58, 20132 Milan, Italy
| | - Giuseppe Ciconte
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- School of Medicine, University Vita-Salute San Raffaele, Via Olgettina, 58, 20132 Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Dario Melgari
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Anthony Frosio
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Andrea Ghiroldi
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Marco Piccoli
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Marco Villa
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Pasquale Creo
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Serena Calamaio
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Valerio Castoldi
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS Ospedale San Raffaele, Via Olgettina, 58, 20132 Milan, Italy
| | - Simona Coviello
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Emanuele Micaglio
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Federica Cirillo
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Emanuela Teresina Locati
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Gabriele Negro
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Antonio Boccellino
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Flavio Mastrocinque
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Žarko Ćalović
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Stefano Ricagno
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Letizia Leocani
- School of Medicine, University Vita-Salute San Raffaele, Via Olgettina, 58, 20132 Milan, Italy
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS Ospedale San Raffaele, Via Olgettina, 58, 20132 Milan, Italy
| | - Gabriele Vicedomini
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Vincenzo Santinelli
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| | - Ilaria Rivolta
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore, 48, 20900 Monza, Italy
| | - Luigi Anastasia
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- School of Medicine, University Vita-Salute San Raffaele, Via Olgettina, 58, 20132 Milan, Italy
| | - Carlo Pappone
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
- School of Medicine, University Vita-Salute San Raffaele, Via Olgettina, 58, 20132 Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan, 2, 20097 San Donato Milanese, Milan, Italy
| |
Collapse
|
2
|
Chen X, Su W, Chen J, Ouyang P, Gong J. ST3GAL4 promotes tumorigenesis in breast cancer by enhancing aerobic glycolysis. Hum Cell 2024; 38:1. [PMID: 39422756 DOI: 10.1007/s13577-024-01137-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024]
Abstract
Sialyltransferases are enzymes that play a crucial role in regulating cancer progression by modifying glycoproteins through sialylation. In particular, the ST3 beta-galactoside alpha-2,3-sialyltransferase 4 (ST3GAL4) enzyme is known to be upregulated in breast cancer, but its specific biological functions have not been fully understood. This study aimed to investigate the impact and mechanisms of ST3GAL4 on aerobic glycolysis in breast cancer. We examined ST3GAL4 expression in tumor tissue samples and breast cancer cell lines and also manipulated ST3GAL4 expression in breast cancer cells using lentivirus transduction. The study evaluated cellular processes such as cell viability, cell cycle progression, and aerobic glycolysis by measuring parameters like extracellular acidification rate, glucose uptake, lactate production, and lactate dehydrogenase A (LDHA) expression. We found that ST3GAL4 expression was consistently increased in tumor tissues and breast cancer cell lines. High ST3GAL4 expression was associated with a poor prognosis for patients with breast cancer. Inhibiting ST3GAL4 expression decreased cell viability, disrupted cell cycle progression, and reduced aerobic glycolysis and LDHA expression. Furthermore, suppressing ST3GAL4 expression in animal models reduced tumor growth and cell proliferation. Conversely, overexpressing ST3GAL4 promoted cell viability and cell cycle progression, but these effects were reversed when an inhibitor of aerobic glycolysis was used. The study provided evidence in cells and animal models that ST3GAL4 promotes tumorigenesis in breast cancer by enhancing aerobic glycolysis. These findings suggest that targeting ST3GAL4 may be a potential strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China
- Department of Breast Medicine, Foshan Women and Children Hospital, Foshan, 528000, China
| | - Weijie Su
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, China
| | - Jiewen Chen
- Department of Breast Medicine, Foshan Women and Children Hospital, Foshan, 528000, China
| | - Peng Ouyang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China
| | - Jin Gong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China.
| |
Collapse
|
3
|
Mohamed KA, Kruf S, Büll C. Putting a cap on the glycome: Dissecting human sialyltransferase functions. Carbohydr Res 2024; 544:109242. [PMID: 39167930 DOI: 10.1016/j.carres.2024.109242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/24/2024] [Accepted: 08/13/2024] [Indexed: 08/23/2024]
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.
Collapse
Affiliation(s)
- Khadra A Mohamed
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands
| | - Stijn Kruf
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands
| | - Christian Büll
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525AJ, Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Yagi H, Tateo S, Saito T, Ohta Y, Nishi E, Obitsu S, Suzuki T, Seetaha S, Hellec C, Nakano A, Tojima T, Kato K. Deciphering the sub-Golgi localization of glycosyltransferases via 3D super-resolution imaging. Cell Struct Funct 2024; 49:47-55. [PMID: 38987202 DOI: 10.1247/csf.24008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024] Open
Abstract
The Golgi apparatus, a crucial organelle involved in protein processing, including glycosylation, exhibits complex sub-structures, i.e., cis-, medial, and trans-cisternae. This study investigated the distribution of glycosyltransferases within the Golgi apparatus of mammalian cells via 3D super-resolution imaging. Focusing on human glycosyltransferases involved in N-glycan modification, we found that even enzymes presumed to coexist in the same Golgi compartment exhibit nuanced variations in localization. By artificially making their N-terminal regions [composed of a cytoplasmic, transmembrane, and stem segment (CTS)] identical, it was possible to enhance the degree of their colocalization, suggesting the decisive role of this region in determining the sub-Golgi localization of enzymes. Ultimately, this study reveals the molecular codes within CTS regions as key determinants of glycosyltransferase localization, providing insights into precise control over the positioning of glycosyltransferases, and consequently, the interactions between glycosyltransferases and substrate glycoproteins as cargoes in the secretory pathway. This study advances our understanding of Golgi organization and opens avenues for programming the glycosylation of proteins for clinical applications.Key words: Golgi apparatus, glycosyltransferase, 3D super-resolution imaging, N-glycosylation.
Collapse
Affiliation(s)
- Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University
- Exploratory Research Center on Life and Living Systems (ExCELLS)
| | - Seigo Tateo
- Exploratory Research Center on Life and Living Systems (ExCELLS)
- Institute for Molecular Science, National Institutes of Natural Sciences
| | - Taiki Saito
- Exploratory Research Center on Life and Living Systems (ExCELLS)
- Institute for Molecular Science, National Institutes of Natural Sciences
| | - Yusaku Ohta
- Exploratory Research Center on Life and Living Systems (ExCELLS)
| | - Emiko Nishi
- Exploratory Research Center on Life and Living Systems (ExCELLS)
| | - Saemi Obitsu
- Exploratory Research Center on Life and Living Systems (ExCELLS)
| | - Tatsuya Suzuki
- Exploratory Research Center on Life and Living Systems (ExCELLS)
- Institute for Molecular Science, National Institutes of Natural Sciences
| | - Supaphorn Seetaha
- Exploratory Research Center on Life and Living Systems (ExCELLS)
- Institute for Molecular Science, National Institutes of Natural Sciences
- Department of Biochemistry, Faculty of Science, Kasetsart University
| | - Charles Hellec
- Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics
| | - Takuro Tojima
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics
| | - Koichi Kato
- Graduate School of Pharmaceutical Sciences, Nagoya City University
- Exploratory Research Center on Life and Living Systems (ExCELLS)
- Institute for Molecular Science, National Institutes of Natural Sciences
| |
Collapse
|
5
|
Tokoro Y, Nagae M, Nakano M, Harduin-Lepers A, Kizuka Y. LacdiNAc synthase B4GALNT3 has a unique PA14 domain and suppresses N-glycan capping. J Biol Chem 2024; 300:107450. [PMID: 38844136 PMCID: PMC11254600 DOI: 10.1016/j.jbc.2024.107450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Structural variation of N-glycans is essential for the regulation of glycoprotein functions. GalNAcβ1-4GlcNAc (LacdiNAc or LDN), a unique subterminal glycan structure synthesized by B4GALNT3 or B4GALNT4, is involved in the clearance of N-glycoproteins from the blood and maintenance of cell stemness. Such regulation of glycoprotein functions by LDN is largely different from that by the dominant subterminal structure, N-acetyllactosamine (Galβ1-4GlcNAc, LacNAc). However, the mechanisms by which B4GALNT activity is regulated and how LDN plays different roles from LacNAc remain unclear. Here, we found that B4GALNT3 and four have unique domain organization containing a noncatalytic PA14 domain, which is a putative glycan-binding module. A mutant lacking this domain dramatically decreases the activity toward various substrates, such as N-glycan, O-GalNAc glycan, and glycoproteins, indicating that this domain is essential for enzyme activity and forms part of the catalytic region. In addition, to clarify the mechanism underlying the functional differences between LDN and LacNAc, we examined the effects of LDN on the maturation of N-glycans, focusing on the related glycosyltransferases upstream and downstream of B4GALNT. We revealed that, unlike LacNAc synthesis, prior formation of bisecting GlcNAc in N-glycan almost completely inhibits LDN synthesis by B4GALNT3. Moreover, the presence of LDN negatively impacted the actions of many glycosyltransferases for terminal modifications, including sialylation, fucosylation, and human natural killer-1 synthesis. These findings demonstrate that LDN has significant impacts on N-glycan maturation in a completely different way from LacNAc, which could contribute to obtaining a comprehensive overview of the system regulating complex N-glycan biosynthesis.
Collapse
Affiliation(s)
- Yuko Tokoro
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Masamichi Nagae
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Laboratory of Molecular Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Suita, Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashihiroshima, Japan
| | - Anne Harduin-Lepers
- University of Lille, CNRS, UMR 8576 -UGSF- Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Yasuhiko Kizuka
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan.
| |
Collapse
|
6
|
Gu J, Isaji T. Specific sialylation of N-glycans and its novel regulatory mechanism. Glycoconj J 2024; 41:175-183. [PMID: 38958800 PMCID: PMC11329402 DOI: 10.1007/s10719-024-10157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
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 β-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 β-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 α3β1, 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.
Collapse
Affiliation(s)
- Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan.
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| |
Collapse
|
7
|
Isaji T, Gu J. Novel regulatory mechanisms of N-glycan sialylation: Implication of integrin and focal adhesion kinase in the regulation. Biochim Biophys Acta Gen Subj 2024; 1868:130617. [PMID: 38614280 DOI: 10.1016/j.bbagen.2024.130617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Sialylation of glycoproteins, including integrins, is crucial in various cancers and diseases such as immune disorders. These modifications significantly impact cellular functions and are associated with cancer progression. Sialylation, catalyzed by specific sialyltransferases (STs), has traditionally been considered to be regulated at the mRNA level. SCOPE OF REVIEW Recent research has expanded our understanding of sialylation, revealing ST activity changes beyond mRNA level variations. This includes insights into COPI vesicle formation and Golgi apparatus maintenance and identifying specific target proteins of STs that are not predictable through recombinant enzyme assays. MAJOR CONCLUSIONS This review summarizes that Golgi-associated pathways largely influence the regulation of STs. GOLPH3, GORAB, PI4K, and FAK have become critical elements in sialylation regulation. Some STs have been revealed to possess specificity for specific target proteins, suggesting the presence of additional, enzyme-specific regulatory mechanisms. GENERAL SIGNIFICANCE This study enhances our understanding of the molecular interplay in sialylation regulation, mainly focusing on the role of integrin and FAK. It proposes a bidirectional system where sialylations might influence integrins and vice versa. The diversity of STs and their specific linkages offer new perspectives in cancer research, potentially broadening our understanding of cellular mechanisms and opening avenues for new therapeutic approaches in targeting sialylation pathways.
Collapse
Affiliation(s)
- Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan.
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558, Japan.
| |
Collapse
|
8
|
Li X, Xiao Y, Li P, Zhu Y, Guo Y, Bian H, Li Z. Sialyltransferase ST3GAL6 silencing reduces α2,3-sialylated glycans to regulate autophagy by decreasing HSPB8-BAG3 in the brain with hepatic encephalopathy. J Zhejiang Univ Sci B 2024; 25:485-498. [PMID: 38910494 PMCID: PMC11199091 DOI: 10.1631/jzus.b2300917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 02/20/2024] [Indexed: 05/23/2024]
Abstract
End-stage liver diseases, such as cirrhosis and liver cancer caused by hepatitis B, are often combined with hepatic encephalopathy (HE); ammonia poisoning is posited as one of its main pathogenesis mechanisms. Ammonia is closely related to autophagy, but the molecular mechanism of ammonia's regulatory effect on autophagy in HE remains unclear. Sialylation is an essential form of glycosylation. In the nervous system, abnormal sialylation affects various physiological processes, such as neural development and synapse formation. ST3 β-galactoside α2,3-sialyltransferase 6 (ST3GAL6) is one of the significant glycosyltransferases responsible for adding α2,3-linked sialic acid to substrates and generating glycan structures. We found that the expression of ST3GAL6 was upregulated in the brains of mice with HE and in astrocytes after ammonia induction, and the expression levels of α2,3-sialylated glycans and autophagy-related proteins microtubule-associated protein light chain 3 (LC3) and Beclin-1 were upregulated in ammonia-induced astrocytes. These findings suggest that ST3GAL6 is related to autophagy in HE. Therefore, we aimed to determine the regulatory relationship between ST3GAL6 and autophagy. We found that silencing ST3GAL6 and blocking or degrading α2,3-sialylated glycans by way of Maackia amurensis lectin-II (MAL-II) and neuraminidase can inhibit autophagy. In addition, silencing the expression of ST3GAL6 can downregulate the expression of heat shock protein β8 (HSPB8) and Bcl2-associated athanogene 3 (BAG3). Notably, the overexpression of HSPB8 partially restored the reduced autophagy levels caused by silencing ST3GAL6 expression. Our results indicate that ST3GAL6 regulates autophagy through the HSPB8-BAG3 complex.
Collapse
Affiliation(s)
- Xiaocheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yaqing Xiao
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Pengfei Li
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Yayun Zhu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yonghong Guo
- The Infectious Disease Department, Gongli Hospital, Pudong New Area, Shanghai 200135, China. ,
| | - Huijie Bian
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an 710032, China. ,
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an 710069, China.
| |
Collapse
|
9
|
Kofsky JM, Babulic JL, Boddington ME, De León González FV, Capicciotti CJ. Glycosyltransferases as versatile tools to study the biology of glycans. Glycobiology 2023; 33:888-910. [PMID: 37956415 DOI: 10.1093/glycob/cwad092] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/15/2023] Open
Abstract
All cells are decorated with complex carbohydrate structures called glycans that serve as ligands for glycan-binding proteins (GBPs) to mediate a wide range of biological processes. Understanding the specific functions of glycans is key to advancing an understanding of human health and disease. However, the lack of convenient and accessible tools to study glycan-based interactions has been a defining challenge in glycobiology. Thus, the development of chemical and biochemical strategies to address these limitations has been a rapidly growing area of research. In this review, we describe the use of glycosyltransferases (GTs) as versatile tools to facilitate a greater understanding of the biological roles of glycans. We highlight key examples of how GTs have streamlined the preparation of well-defined complex glycan structures through chemoenzymatic synthesis, with an emphasis on synthetic strategies allowing for site- and branch-specific display of glyco-epitopes. We also describe how GTs have facilitated expansion of glyco-engineering strategies, on both glycoproteins and cell surfaces. Coupled with advancements in bioorthogonal chemistry, GTs have enabled selective glyco-epitope editing of glycoproteins and cells, selective glycan subclass labeling, and the introduction of novel biomolecule functionalities onto cells, including defined oligosaccharides, antibodies, and other proteins. Collectively, these approaches have contributed great insight into the fundamental biological roles of glycans and are enabling their application in drug development and cellular therapies, leaving the field poised for rapid expansion.
Collapse
Affiliation(s)
- Joshua M Kofsky
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, ON K7L 3N6, Canada
| | - Jonathan L Babulic
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L 2V7, Canada
| | - Marie E Boddington
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L 2V7, Canada
| | | | - Chantelle J Capicciotti
- Department of Chemistry, Queen's University, 90 Bader Lane, Kingston, ON K7L 3N6, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart Street, Kingston, ON K7L 2V7, Canada
- Department of Surgery, Queen's University, 76 Stuart Street, Kingston, ON K7L 2V7, Canada
| |
Collapse
|
10
|
Huang HW, Shivatare VS, Tseng TH, Wong CH. Cell-based production of Fc-GlcNAc and Fc-alpha-2,6 sialyl glycan enriched antibody with improved effector functions through glycosylation pathway engineering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572280. [PMID: 38187613 PMCID: PMC10769250 DOI: 10.1101/2023.12.18.572280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Glycosylation of antibody plays an important role in Fc-mediated killing of tumor cells and virus-infected cells through effector functions such as antibody-dependent cellular cytotoxicity (ADCC), antibody dependent cell-mediated phagocytosis (ADCP) and vaccinal effect. Previous studies showed that therapeutical humanized antibodies with α2-6 sialyl complex type (SCT) glycan attached to Fc-Asn297 exhibited optimal binding to the Fc receptors on effector cells associated with ADCC, ADCP and vaccinal effect. However, the production of antibodies with homogeneous Fc-SCT needs multiple in vitro enzymatic and purification steps. In this study, we report two different approaches to shorten the processes to produce SCT-enriched antibodies. First, we expressed a bacterial endoglycosidase in GNT1-KO EXPI293 cells to trim all N -glycans to mono-GlcNAc glycoforms for in vitro transglycosylation to generate homogeneous SCT antibody. Second, we engineered the glycosylation pathway of HEK293 cells through knockout of the undesired glycosyltransferases and expression of the desired glycosyltransferases to produce SCT enriched antibodies with similar binding affinity to Fc receptors and ADCC activity to homogenous SCT antibody.
Collapse
|
11
|
Tian J, Zhu Q, Huang X, Li Y. A new sandwich-type electrochemiluminescence sensor based on HPSNs-NH2@Au NPs and AuPdPt NPs for determination of α(2,3)-sial-Gs. Mikrochim Acta 2023; 190:420. [PMID: 37770767 DOI: 10.1007/s00604-023-06000-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/13/2023] [Indexed: 09/30/2023]
Abstract
A novel sandwich-type "on-off" electrochemiluminescence (ECL) biosensor for the determination of α(2,3)-sial-Gs was designed. Specifically, amino-functionalized porous silica nanoparticles (HPSNs-NH2) were first prepared and then decorated with gold nanoparticles (Au NPs) to form HPSNs-NH2@Au NP nanocomposite, which exhibited a strong ability to enhance ECL intensity with K2S2O8 as co-reactant (signal-on) and could immobilize the target-specific binding molecules of maackia amurensis lectin (MAL). Additionally, AuPdPt trimetallic nanoparticles were prepared to serve as a quenched ECL signal indicator (signal-off) with the ability of capturing the target non-specific binding molecules of 3-aminophenylboronic acid (APBA) to form a signal label. The sandwich-type ECL biosensor was constructed based on the structure of MAL-α(2,3)-sial-Gs-APBA and achieved a determination toward α(2,3)-sial-Gs with a wide linear range from 1 fg mL-1 to 10 ng mL-1 and a low detection limit of 0.5 fg mL-1. Furthermore, the proposed ECL biosensor showed satisfactory selectivity, stability, and reproducibility for α(2,3)-sial-Gs determination.
Collapse
Affiliation(s)
- Jiangman Tian
- Department of Pharmacy, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402160, PR China
| | - Qihao Zhu
- Department of Pharmacy, Yongchuan Hospital of Traditional Chinese Medicine, Chongqing, 402160, PR China
| | - Xiaojing Huang
- Central Laboratory, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402160, PR China
| | - Yuan Li
- Central Laboratory, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402160, PR China.
| |
Collapse
|
12
|
Sun Y, Isaji T, Oyama Y, Xu X, Liu J, Hanamatsu H, Yokota I, Miura N, Furukawa JI, Fukuda T, Gu J. Focal-adhesion kinase regulates the sialylation of N-glycans via the PI4KIIα-PI4P pathway. J Biol Chem 2023; 299:105051. [PMID: 37451482 PMCID: PMC10406863 DOI: 10.1016/j.jbc.2023.105051] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
Sialylation is a terminal glycosylated modification of glycoproteins that regulates critical biological events such as cell adhesion and immune response. Our previous study showed that integrin α3β1 plays a crucial role in regulating the sialylation of N-glycans. However, the underlying mechanism for the regulation remains unclear. This study investigated how sialylation is affected by focal adhesion kinase (FAK), which is a critical downstream signal molecule of integrin β1. We established a stable FAK knockout (KO) cell line using the CRISPR/Cas9 system in HeLa cells. The results obtained from lectin blot, flow cytometric analysis, and MS showed that the sialylation levels were significantly decreased in the KO cells compared with that in wild-type (WT) cells. Moreover, phosphatidylinositol 4-phosphate (PI4P) expression levels were also reduced in the KO cells due to a decrease in the stability of phosphatidylinositol 4-kinase-IIα (PI4KIIα). Notably, the decreased levels of sialylation, PI4P, and the complex formation between GOLPH3 and ST3GAL4 or ST6GAL1, which are the main sialyltransferases for modification of N-glycans, were significantly restored by the re-expression of FAK. Furthermore, the decreased sialylation and phosphorylation of Akt and cell migration caused by FAK deficiency all were restored by overexpressing PI4KIIα, which suggests that PI4KIIα is one of the downstream molecules of FAK. These findings indicate that FAK regulates sialylation via the PI4P synthesis pathway and a novel mechanism is suggested for the integrin-FAK-PI4KIIα-GOLPH3-ST axis modulation of sialylation in N-glycans.
Collapse
Affiliation(s)
- Yuhan Sun
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| | - Yoshiyuki Oyama
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Xing Xu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Jianwei Liu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Hisatoshi Hanamatsu
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ikuko Yokota
- Division of Glyco-Systems Biology, Institute for Glyco-Core Research, Tokai National Higher Education and Research System, Nagoya, Japan
| | - Nobuaki Miura
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Jun-Ichi Furukawa
- Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Division of Glyco-Systems Biology, Institute for Glyco-Core Research, Tokai National Higher Education and Research System, Nagoya, Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| |
Collapse
|
13
|
Dragan M, Chen Z, Li Y, Le J, Sun P, Haensel D, Sureshchandra S, Pham A, Lu E, Pham KT, Verlande A, Vu R, Gutierrez G, Li W, Jang C, Masri S, Dai X. Ovol1/2 loss-induced epidermal defects elicit skin immune activation and alter global metabolism. EMBO Rep 2023; 24:e56214. [PMID: 37249012 PMCID: PMC10328084 DOI: 10.15252/embr.202256214] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
Skin epidermis constitutes the outer permeability barrier that protects the body from dehydration, heat loss, and myriad external assaults. Mechanisms that maintain barrier integrity in constantly challenged adult skin and how epidermal dysregulation shapes the local immune microenvironment and whole-body metabolism remain poorly understood. Here, we demonstrate that inducible and simultaneous ablation of transcription factor-encoding Ovol1 and Ovol2 in adult epidermis results in barrier dysregulation through impacting epithelial-mesenchymal plasticity and inflammatory gene expression. We find that aberrant skin immune activation then ensues, featuring Langerhans cell mobilization and T cell responses, and leading to elevated levels of secreted inflammatory factors in circulation. Finally, we identify failure to gain body weight and accumulate body fat as long-term consequences of epidermal-specific Ovol1/2 loss and show that these global metabolic changes along with the skin barrier/immune defects are partially rescued by immunosuppressant dexamethasone. Collectively, our study reveals key regulators of adult barrier maintenance and suggests a causal connection between epidermal dysregulation and whole-body metabolism that is in part mediated through aberrant immune activation.
Collapse
Affiliation(s)
- Morgan Dragan
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
| | - Zeyu Chen
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- Present address:
Department of Dermatology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
- Present address:
Institute of PsoriasisTongji University School of MedicineShanghaiChina
| | - Yumei Li
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Johnny Le
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Peng Sun
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Daniel Haensel
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- Present address:
Program in Epithelial BiologyStanford University School of MedicineStanfordCAUSA
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Anh Pham
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Eddie Lu
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Katherine Thanh Pham
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Amandine Verlande
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Remy Vu
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
| | - Guadalupe Gutierrez
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Wei Li
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Cholsoon Jang
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Selma Masri
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Xing Dai
- Department of Biological Chemistry, School of MedicineUniversity of CaliforniaIrvineCAUSA
- The NSF‐Simons Center for Multiscale Cell Fate ResearchUniversity of CaliforniaIrvineCAUSA
- Department of Dermatology, School of MedicineUniversity of CaliforniaIrvineCAUSA
| |
Collapse
|
14
|
Marín-Quílez A, Díaz-Ajenjo L, Di Buduo CA, Zamora-Cánovas A, Lozano ML, Benito R, González-Porras JR, Balduini A, Rivera J, Bastida JM. Inherited Thrombocytopenia Caused by Variants in Crucial Genes for Glycosylation. Int J Mol Sci 2023; 24:5109. [PMID: 36982178 PMCID: PMC10049517 DOI: 10.3390/ijms24065109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/13/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
Protein glycosylation, including sialylation, involves complex and frequent post-translational modifications, which play a critical role in different biological processes. The conjugation of carbohydrate residues to specific molecules and receptors is critical for normal hematopoiesis, as it favors the proliferation and clearance of hematopoietic precursors. Through this mechanism, the circulating platelet count is controlled by the appropriate platelet production by megakaryocytes, and the kinetics of platelet clearance. Platelets have a half-life in blood ranging from 8 to 11 days, after which they lose the final sialic acid and are recognized by receptors in the liver and eliminated from the bloodstream. This favors the transduction of thrombopoietin, which induces megakaryopoiesis to produce new platelets. More than two hundred enzymes are responsible for proper glycosylation and sialylation. In recent years, novel disorders of glycosylation caused by molecular variants in multiple genes have been described. The phenotype of the patients with genetic alterations in GNE, SLC35A1, GALE and B4GALT is consistent with syndromic manifestations, severe inherited thrombocytopenia, and hemorrhagic complications.
Collapse
Affiliation(s)
- Ana Marín-Quílez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-U765, 30003 Murcia, Spain
| | - Lorena Díaz-Ajenjo
- IBSAL, CIC, IBMCC, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain
| | | | - Ana Zamora-Cánovas
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-U765, 30003 Murcia, Spain
| | - María Luisa Lozano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-U765, 30003 Murcia, Spain
| | - Rocío Benito
- IBSAL, CIC, IBMCC, Universidad de Salamanca-CSIC, 37007 Salamanca, Spain
| | - José Ramón González-Porras
- Department of Hematology, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), 37007 Salamanca, Spain
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - José Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-U765, 30003 Murcia, Spain
| | - José María Bastida
- Department of Hematology, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), 37007 Salamanca, Spain
| |
Collapse
|
15
|
Madunić K, Luijkx YMCA, Mayboroda OA, Janssen GMC, van Veelen PA, Strijbis K, Wennekes T, Lageveen-Kammeijer GSM, Wuhrer M. O-Glycomic and Proteomic Signatures of Spontaneous and Butyrate-Stimulated Colorectal Cancer Cell Line Differentiation. Mol Cell Proteomics 2023; 22:100501. [PMID: 36669592 PMCID: PMC9999233 DOI: 10.1016/j.mcpro.2023.100501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Gut microbiota of the gastrointestinal tract provide health benefits to the human host via bacterial metabolites. Bacterial butyrate has beneficial effects on intestinal homeostasis and is the preferred energy source of intestinal epithelial cells, capable of inducing differentiation. It was previously observed that changes in the expression of specific proteins as well as protein glycosylation occur with differentiation. In this study, specific mucin O-glycans were identified that mark butyrate-induced epithelial differentiation of the intestinal cell line CaCo-2 (Cancer Coli-2), by applying porous graphitized carbon nano-liquid chromatography with electrospray ionization tandem mass spectrometry. Moreover, a quantitative proteomic approach was used to decipher changes in the cell proteome. It was found that the fully differentiated butyrate-stimulated cells are characterized by a higher expression of sialylated O-glycan structures, whereas fucosylation is downregulated with differentiation. By performing an integrative approach, we generated hypotheses about the origin of the observed O-glycome changes. These insights pave the way for future endeavors to study the dynamic O-glycosylation patterns in the gut, either produced via cellular biosynthesis or through the action of bacterial glycosidases as well as the functional role of these patterns in homeostasis and dysbiosis at the gut-microbiota interface.
Collapse
Affiliation(s)
- K Madunić
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands
| | - Y M C A Luijkx
- Department Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands; Department Biomolecular Health Sciences, Utrecht University, Utrecht, The Netherlands
| | - O A Mayboroda
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands
| | - G M C Janssen
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands
| | - P A van Veelen
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands
| | - K Strijbis
- Department Biomolecular Health Sciences, Utrecht University, Utrecht, The Netherlands
| | - T Wennekes
- Department Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | | | - M Wuhrer
- Center for Proteomics and Metabolomics, Leiden University, The Netherlands.
| |
Collapse
|
16
|
Song W, Liang C, Sun Y, Morii S, Yomogida S, Isaji T, Fukuda T, Hang Q, Hara A, Nakano M, Gu J. Expression of GnT-III decreases chemoresistance via negatively regulating P-glycoprotein expression: Involvement of the TNFR2-NF-κB signaling pathway. J Biol Chem 2023; 299:103051. [PMID: 36813234 PMCID: PMC10033316 DOI: 10.1016/j.jbc.2023.103051] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
The phenomenon of multidrug resistance (MDR) is called chemoresistance with respect to the treatment of cancer, and it continues to be a major challenge. The role of N-glycosylation in chemoresistance, however, remains poorly understood. Here, we established a traditional model for adriamycin resistance in K562 cells, which are also known as K562/adriamycin-resistant (ADR) cells. Lectin blot, mass spectrometry, and RT-PCR analysis showed that the expression levels of N-acetylglucosaminyltransferase III (GnT-III) mRNA and its products, bisected N-glycans, are significantly decreased in K562/ADR cells, compared with the levels in parent K562 cells. By contrast, the expression levels of both P-glycoprotein (P-gp) and its intracellular key regulator, NF-κB signaling, are significantly increased in K562/ADR cells. These upregulations were sufficiently suppressed by the overexpression of GnT-III in K562/ADR cells. We found that the expression of GnT-III consistently decreased chemoresistance for doxorubicin and dasatinib, as well as activation of the NF-κB pathway by tumor necrosis factor (TNF) α, which binds to two structurally distinct glycoproteins, TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2), on the cell surface. Interestingly, our immunoprecipitation analysis revealed that only TNFR2, but not TNFR1, contains bisected N-glycans. The lack of GnT-III strongly induced TNFR2's autotrimerization without ligand stimulation, which was rescued by the overexpression of GnT-III in K562/ADR cells. Furthermore, the deficiency of TNFR2 suppressed P-gp expression while it increased GnT-III expression. Taken together, these results clearly show that GnT-III negatively regulates chemoresistance via the suppression of P-gp expression, which is regulated by the TNFR2-NF/κB signaling pathway.
Collapse
Affiliation(s)
- Wanli Song
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Caixia Liang
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Yuhan Sun
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Sayaka Morii
- Graduate School of Integrated Sciences for Life, Hiroshima University, Sendai, Miyagi, Japan
| | - Shin Yomogida
- Division of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Qinglei Hang
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Akiyoshi Hara
- Division of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi Japan
| | - Miyako Nakano
- Graduate School of Integrated Sciences for Life, Hiroshima University, Sendai, Miyagi, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| |
Collapse
|
17
|
Wiertelak W, Olczak M, Maszczak-Seneczko D. An interaction between SLC35A1 and ST3Gal4 is differentially affected by CDG-causing mutations in the SLC35A1 gene. Biochem Biophys Res Commun 2022; 635:46-51. [DOI: 10.1016/j.bbrc.2022.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022]
|
18
|
ST3GAL3 Promotes the Inflammatory Response of Fibroblast-Like Synoviocytes in Rheumatoid Arthritis by Activating the TLR9/MyD88 Pathway. Mediators Inflamm 2022; 2022:4258742. [DOI: 10.1155/2022/4258742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/09/2022] [Accepted: 10/28/2022] [Indexed: 11/12/2022] Open
Abstract
This study is aimed at investigating the role of β-galactoside-α2,3-sialyltransferase III (ST3GAL3) in fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA), as well as its potential mechanism of action. The Gene Expression Omnibus (GEO) database and gene set enrichment analysis (GSEA) were used to analyse the expression of ST3GAL3 and the enrichment signalling pathways associated with ST3GAL3 in RA. The effects of ST3GAL3 on tumour necrosis factor- (TNF-) α and interleukin- (IL-) 1β-treated MH7A cells were determined using methyl thiazolyl tetrazolium (MTT), transwell, and enzyme-linked immunosorbent assays (ELISA). The expression of proliferation-associated proteins and Toll-like receptor (TLR) pathway-enriched proteins was analysed using western blotting. As a main result, ST3GAL3 was screened as an overlapping upregulated gene from GSE101193 and GSE94519 datasets. ST3GAL3 expression in MH7A cells significantly increased with increasing treatment time with TNF-α or IL-1β. TLR9/myeloid differentiation primary response protein 88 (MyD88) is a downstream activation pathway of ST3GAL3. ST3GAL3 overexpression promoted MH7A cell proliferation and migration. Additionally, ST3GAL3 overexpression upregulated the expression of proliferation-associated proteins (cyclinD, cyclinE, and proliferating cell nuclear antigen) and TLR pathway enrichment factors (TLR9 and MyD88) and increased the production of matrix metallopeptidase (MMP) 1, MMP3, interleukin- (IL-) 6, and IL-8, whereas si-ST3GAL3 had the opposite effect. The addition of TLR9 agonists (CpG 2216 and CpG 2006) reversed the effects of si-ST3GAL3 on MH7A cell proliferation, migration, and inflammation. TLR9-specific siRNA reversed the effects of ST3GAL3 overexpression on MH7A cell proliferation, migration, and inflammation. In conclusion, ST3GAL3 is likely involved in RA pathogenesis by activating the TLR9/MyD88 pathway.
Collapse
|
19
|
Wen R, Zhao H, Zhang D, Chiu CL, Brooks JD. Sialylated glycoproteins as biomarkers and drivers of progression in prostate cancer. Carbohydr Res 2022; 519:108598. [PMID: 35691122 DOI: 10.1016/j.carres.2022.108598] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/20/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023]
Abstract
Sialic acids have been implicated in cancer initiation, progression, and immune evasion in diverse human malignancies. Sialylation of terminal glycans on cell surface and secreted glycoproteins is a long-recognized feature of cancer cells. Recently, immune checkpoint inhibitor immunotherapy has tremendously improved the outcomes of patients with various cancers. However, available immunotherapy approaches have had limited efficacy in metastatic castration-resistant prostate cancer. Sialic acid modified glycoproteins in prostate cancers and their interaction with Siglec receptors on tumor infiltrating immune cells might underlie immunosuppressive signaling in prostate cancer. Here, we summarize the function of sialic acids and relevant glycosynthetic enzymes in cancer initiation and progression. We also discuss the possible uses of sialic acids as biomarkers in prostate cancer and the potential methods for targeting Siglec-sialic acid interactions for prostate cancer treatment.
Collapse
Affiliation(s)
- Ru Wen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Dalin Zhang
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chun-Lung Chiu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
20
|
Li J, Long Y, Sun J, Wu J, He X, Wang S, Wang X, Miao X, Huang R, Yan J. Comprehensive landscape of the ST3GAL family reveals the significance of ST3GAL6-AS1/ST3GAL6 axis on EGFR signaling in lung adenocarcinoma cell invasion. Front Cell Dev Biol 2022; 10:931132. [PMID: 36092699 PMCID: PMC9462654 DOI: 10.3389/fcell.2022.931132] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/20/2022] [Indexed: 11/20/2022] Open
Abstract
Sialylation aberration has been implicated in lung cancer development by altering signaling pathways. Hence, it is urgent to identify key sialyltransferases in the development of lung adenocarcinoma (LUAD), which is a common malignant subtype of non-small cell lung cancer. Herein, by systematically investigating the expression levels of ST3GAL family members in several public databases, we consistently found the frequent downregulation of ST3GAL6 in LUAD samples. Its downregulation is significantly negatively associated with stage, and significantly reduced in proximal-proliferative molecular subtype and predicts poor clinical outcomes. By protein–protein interaction network analysis and validation, we found that ST3GAL6 deficiency promotes LUAD cell invasiveness with the activated EGFR/MAPK signaling, accompanied by the elevated expression levels of matrix metalloproteinases 2 and 9, which can be partially reversed by EGFR inhibitor, gefitinib. Additionally, the ST3GAL6 level was positively regulated by ST3GAL6-AS1, an antisense long non-coding RNA to its host gene. The downregulation of ST3GAL6-AS1 also heralds a worse prognosis in LUAD patients and promotes LUAD cell invasiveness, recapitulating the function of its host gene, ST3GAL6. Altogether, ST3GAL6-AS1-regulated ST3GAL6 is a frequently downregulated sialyltransferase in LUAD patients and negatively regulates EGFR signaling, which can serve as a promising independent prognostic marker in LUAD patients.
Collapse
Affiliation(s)
- Jiaxuan Li
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Long
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jingya Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiajun Wu
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Xiao He
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Simei Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiongbiao Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiayi Miao
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Xiayi Miao, ; Ruimin Huang, ; Jun Yan,
| | - Ruimin Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Xiayi Miao, ; Ruimin Huang, ; Jun Yan,
| | - Jun Yan
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- *Correspondence: Xiayi Miao, ; Ruimin Huang, ; Jun Yan,
| |
Collapse
|
21
|
Pu C, Biyuan, Xu K, Zhao Y. Glycosylation and its research progress in endometrial cancer. Clin Transl Oncol 2022; 24:1865-1880. [PMID: 35752750 PMCID: PMC9418304 DOI: 10.1007/s12094-022-02858-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/10/2022] [Indexed: 12/12/2022]
Abstract
Endometrial cancer (EC) is one of the most common tumors in the female reproductive system, which seriously threatens women's health, particularly in developed countries. 13% of the patients with EC have a poor prognosis due to recurrence and metastasis. Therefore, identifying good predictive biomarkers and therapeutic targets is critical to enable the early detection of metastasis and improve the prognosis. For decades, extensive studies had focused on glycans and glycoproteins in the progression of cancer. The types of glycans that are covalently attached to the polypeptide backbone, usually via nitrogen or oxygen linkages, are known as N‑glycans or O‑glycans, respectively. The degree of protein glycosylation and the aberrant changes in the carbohydrate structures have been implicated in the extent of tumorigenesis and reported to play a critical role in regulating tumor invasion, metabolism, and immunity. This review summarizes the essential biological role of glycosylation in EC, with a focus on the recent advances in glycomics and glycosylation markers, highlighting their implications in the diagnosis and treatment of EC.
Collapse
Affiliation(s)
- Congli Pu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Biyuan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kai Xu
- Department of Otorhinolaryngology Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yingchao Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
22
|
Tondepu C, Karumbaiah L. Glycomaterials to Investigate the Functional Role of Aberrant Glycosylation in Glioblastoma. Adv Healthc Mater 2022; 11:e2101956. [PMID: 34878733 PMCID: PMC9048137 DOI: 10.1002/adhm.202101956] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/30/2021] [Indexed: 02/03/2023]
Abstract
Glioblastoma (GBM) is a stage IV astrocytoma that carries a dismal survival rate of ≈10 months postdiagnosis and treatment. The highly invasive capacity of GBM and its ability to escape therapeutic challenges are key factors contributing to the poor overall survival rate. While current treatments aim to target the cancer cell itself, they fail to consider the significant role that the GBM tumor microenvironment (TME) plays in promoting tumor progression and therapeutic resistance. The GBM tumor glycocalyx and glycan-rich extracellular matrix (ECM), which are important constituents of the TME have received little attention as therapeutic targets. A wide array of aberrantly modified glycans in the GBM TME mediate tumor growth, invasion, therapeutic resistance, and immunosuppression. Here, an overview of the landscape of aberrant glycan modifications in GBM is provided, and the design and utility of 3D glycomaterials are discussed as a tool to evaluate glycan-mediated GBM progression and therapeutic efficacy. The development of alternative strategies to target glycans in the TME can potentially unveil broader mechanisms of restricting tumor growth and enhancing the efficacy of tumor-targeting therapeutics.
Collapse
Affiliation(s)
- Chaitanya Tondepu
- Regenerative Bioscience Science Center, University of Georgia, Athens, GA, 30602, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Science Center, University of Georgia, Athens, GA, 30602, USA
- Division of Neuroscience, Biomedical & Translational Sciences Institute, University of Georgia, Athens, GA, 30602, USA
- Edgar L. Rhodes Center for ADS, College of Agriculture and Environmental Sciences, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
23
|
Upregulation of sialyltransferases ST3Gal1 and ST6Gal1 promotes stabilization of erythrocyte mass and recovery of anemia in Trypanosoma brucei brucei-infected pigs. Res Vet Sci 2022; 145:102-108. [DOI: 10.1016/j.rvsc.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/14/2022] [Accepted: 02/04/2022] [Indexed: 11/23/2022]
|
24
|
Glycomic Analysis Reveals That Sialyltransferase Inhibition Is Involved in the Antiviral Effects of Arbidol. J Virol 2022; 96:e0214121. [PMID: 35044216 PMCID: PMC8941891 DOI: 10.1128/jvi.02141-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Due to the high mutation rate of influenza virus and the rapid increase of drug resistance, it is imperative to discover host-targeting antiviral agents with broad-spectrum antiviral activity. Considering the discrepancy between the urgent demand of antiviral drugs during an influenza pandemic and the long-term process of drug discovery and development, it is feasible to explore host-based antiviral agents and strategies from antiviral drugs on the market. In the current study, the antiviral mechanism of arbidol (ARB), a broad-spectrum antiviral drug with potent activity at early stages of viral replication, was investigated from the aspect of hemagglutinin (HA) receptors of host cells. N-glycans that act as the potential binding receptors of HA on 16-human bronchial epithelial (16-HBE) cells were comprehensively profiled for the first time by using an in-depth glycomic approach based on TiO2-PGC chip-Q-TOF MS. Their relative levels upon the treatment of ARB and virus were carefully examined by employing an ultra-high sensitive qualitative method based on Chip LC-QQQ MS, showing that ARB treatment led to significant and extensive decrease of sialic acid (SA)-linked N-glycans (SA receptors), and thereby impaired the virus utilization on SA receptors for rolling and entry. The SA-decreasing effect of ARB was demonstrated to result from its inhibitory effect on sialyltransferases (ST), ST3GAL4 and ST6GAL1 of 16-HBE cells. Silence of STs, natural ST inhibitors, as well as sialidase treatment of 16-HBE cells, resulted in similar potent antiviral activity, whereas ST-inducing agent led to the diminished antiviral effect of ARB. These observations collectively suggesting the involvement of ST inhibition in the antiviral effect of ARB. IMPORTANCE This study revealed, for the first time, that ST inhibition and the resulted destruction of SA receptors of host cells may be an underlying mechanism for the antiviral activity of ARB. ST inhibition has been proposed as a novel host-targeting antiviral approach recently and several compounds are currently under exploration. ARB is the first antiviral drug on the market that was found to possess ST inhibiting function. This will provide crucial evidence for the clinical usages of ARB, such as in combination with neuraminidase (NA) inhibitors to exert optimized antiviral effect, etc. More importantly, as an agent that can inhibit the expression of STs, ARB can serve as a novel lead compound for the discovery and development of host-targeting antiviral drugs.
Collapse
|
25
|
Zhong X, D’Antona AM, Scarcelli JJ, Rouse JC. New Opportunities in Glycan Engineering for Therapeutic Proteins. Antibodies (Basel) 2022; 11:5. [PMID: 35076453 PMCID: PMC8788452 DOI: 10.3390/antib11010005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/22/2021] [Accepted: 12/31/2021] [Indexed: 11/17/2022] Open
Abstract
Glycans as sugar polymers are important metabolic, structural, and physiological regulators for cellular and biological functions. They are often classified as critical quality attributes to antibodies and recombinant fusion proteins, given their impacts on the efficacy and safety of biologics drugs. Recent reports on the conjugates of N-acetyl-galactosamine and mannose-6-phosphate for lysosomal degradation, Fab glycans for antibody diversification, as well as sialylation therapeutic modulations and O-linked applications, have been fueling the continued interest in glycoengineering. The current advancements of the human glycome and the development of a comprehensive network in glycosylation pathways have presented new opportunities in designing next-generation therapeutic proteins.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, 610 Main Street, Cambridge, MA 02139, USA;
| | - Aaron M. D’Antona
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, 610 Main Street, Cambridge, MA 02139, USA;
| | - John J. Scarcelli
- BioProcess R&D, Biotherapeutics Pharmaceutical Sciences, Medicinal Sciences, Pfizer Worldwide R&D, 1 Burtt Road, Andover, MA 01810, USA;
| | - Jason C. Rouse
- Analytical R&D, Biotherapeutics Pharmaceutical Sciences, Medicinal Sciences, Pfizer Worldwide R&D, 1 Burtt Road, Andover, MA 01810, USA;
| |
Collapse
|
26
|
Gao Z, Xu M, Yue S, Shan H, Xia J, Jiang J, Yang S. Abnormal sialylation and fucosylation of saliva glycoproteins: Characteristics of lung cancer-specific biomarkers. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 3:100079. [PMID: 35005612 PMCID: PMC8718573 DOI: 10.1016/j.crphar.2021.100079] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 10/31/2021] [Accepted: 12/15/2021] [Indexed: 12/23/2022] Open
Abstract
Dysregulated surface glycoproteins play an important role in tumor cell proliferation and progression. Abnormal glycosylation of these glycoproteins may activate tumor signal transduction and lead to tumor development. The tumor microenvironment alters its molecular composition, some of which regulate protein glycosylation biosynthesis. The glycosylation of saliva proteins in lung cancer patients is different from healthy controls, in which the glycans of cancer patients are highly sialylated and hyperfucosylated. Most studies have shown that O-glycans from cancer are truncated O-glycans, while N-glycans contain fucoses and sialic acids. Because glycosylation analysis is challenging, there are few reports on how glycosylation of saliva proteins is related to the occurrence or progression of lung cancer. In this review, we discussed glycoenzymes involved in protein glycosylation, their changes in tumor microenvironment, potential tumor biomarkers present in body fluids, and abnormal glycosylation of saliva or lung glycoproteins. We further explored the effect of glycosylation changes on tumor signal transduction, and emphasized the role of receptor tyrosine kinases in tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Ziyuan Gao
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
- Department of Respiratory and Critical Care Medicine, Dushu Lake Hospital to Soochow University, Suzhou, Jiangsu, 215125, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University; Suzhou Jiangsu, 215006, China
| | - Mingming Xu
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Shuang Yue
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Huang Shan
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| | - Jun Xia
- Department of Clinical Laboratory Center, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Junhong Jiang
- Department of Respiratory and Critical Care Medicine, Dushu Lake Hospital to Soochow University, Suzhou, Jiangsu, 215125, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Soochow University; Suzhou Jiangsu, 215006, China
| | - Shuang Yang
- Center for Clinical Mass Spectrometry, School of Pharmaceutical Sciences, Soochow University, Jiangsu, 215123, China
| |
Collapse
|
27
|
Pisa E, Martire A, Chiodi V, Traversa A, Caputo V, Hauser J, Macrì S. Exposure to 3'Sialyllactose-Poor Milk during Lactation Impairs Cognitive Capabilities in Adulthood. Nutrients 2021; 13:nu13124191. [PMID: 34959743 PMCID: PMC8707534 DOI: 10.3390/nu13124191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 11/17/2022] Open
Abstract
Breast milk exerts pivotal regulatory functions early in development whereby it contributes to the maturation of brain and associated cognitive functions. However, the specific components of maternal milk mediating this process have remained elusive. Sialylated human milk oligosaccharides (HMOs) represent likely candidates since they constitute the principal neonatal dietary source of sialic acid, which is crucial for brain development and neuronal patterning. We hypothesize that the selective neonatal lactational deprivation of a specific sialylated HMOs, sialyl(alpha2,3)lactose (3′SL), may impair cognitive capabilities (attention, cognitive flexibility, and memory) in adulthood in a preclinical model. To operationalize this hypothesis, we cross-fostered wild-type (WT) mouse pups to B6.129-St3gal4tm1.1Jxm/J dams, knock-out (KO) for the gene synthesizing 3′SL, thereby providing milk with approximately 80% 3′SL content reduction. We thus exposed lactating WT pups to a selective reduction of 3′SL and investigated multiple cognitive domains (including memory and attention) in adulthood. Furthermore, to account for the underlying electrophysiological correlates, we investigated hippocampal long-term potentiation (LTP). Neonatal access to 3′SL-poor milk resulted in decreased attention, spatial and working memory, and altered LTP compared to the control group. These results support the hypothesis that early-life dietary sialylated HMOs exert a long-lasting role in the development of cognitive functions.
Collapse
Affiliation(s)
- Edoardo Pisa
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Alberto Martire
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.); (V.C.)
| | - Valentina Chiodi
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.); (V.C.)
| | - Alice Traversa
- Laboratory of Clinical Genomics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Viviana Caputo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Jonas Hauser
- Brain Health, Nestlé Institute of Health Science, Nestlé Research, Société des Produits Nestlé SA, 1000 Lausanne, Switzerland
- Correspondence: (J.H.); (S.M.); Tel.: +41-21-785-8933 (J.H.); +39-06-4990-6829 (S.M.); Fax: +39-06-4957-821 (S.M.)
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
- Correspondence: (J.H.); (S.M.); Tel.: +41-21-785-8933 (J.H.); +39-06-4990-6829 (S.M.); Fax: +39-06-4957-821 (S.M.)
| |
Collapse
|
28
|
Simultaneous analysis of serum α2,3-linked sialylation and core-type fucosylation of prostate-specific antigen for the detection of high-grade prostate cancer. Br J Cancer 2021; 126:764-770. [PMID: 34802050 DOI: 10.1038/s41416-021-01637-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Altered prostate-specific antigen (PSA) glycosylation patterns can be useful biomarkers in detecting high-grade prostate cancer (HGPC). The microfluidic immunoassay system can analyse α2,3-linked sialylated PSA (α2,3-Sia-PSA) and α1,6-linked fucosylated PSA (α1,6-Fuc-PSA) using different lectins, Mackkia amurensis agglutinin and Pholiota squarrosa lectin, respectively. Here, we investigated the diagnostic value of simultaneous analysis of α2,3-Sia-PSA and α1,6-Fuc-PSA for the detection of HGPC. METHODS Men with serum PSA levels of 4-20 ng/mL who underwent prostate biopsy were included. The model to predict HGPC (Gleason grade ≥2) was constructed by multivariate logistic regression analysis, in combination with α2,3-Sia-PSA and α1,6-Fuc-PSA (SF index). RESULTS In the development cohort (n = 150), the SF index showed good discrimination for HGPC (area under the receiver-operating curve (AUC) 0.842; 95% confidence interval (CI) 0.782-0.903), compared to the single PSA test (AUC 0.632, 95% CI 0.543-0.721), α2,3-Sia-PSA (AUC 0.711, 95% CI 0.629-0.793) and α1,6-Fuc-PSA (AUC 0.738, 95% CI 0.657-0.819). Decision-curve analysis showed the superior benefit of the SF index. In the validation cohort (n = 57), the SF index showed good discrimination for HGPC (AUC 0.769, 95% CI 0.643-0.895). CONCLUSIONS The SF index could differentiate HGPC, providing useful information for decision making for prostate biopsy in men with abnormal PSA levels.
Collapse
|
29
|
Berghuis AY, Pijnenborg JFA, Boltje TJ, Pijnenborg JMA. Sialic acids in gynecological cancer development and progression: Impact on diagnosis and treatment. Int J Cancer 2021; 150:678-687. [PMID: 34741527 PMCID: PMC9299683 DOI: 10.1002/ijc.33866] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 11/19/2022]
Abstract
Gynecological cancers are in the top 10 of most common cancers in women. Survival and outcome are strongly related to the stage at diagnosis. Therefore, early diagnosis is essential in reducing morbidity and mortality. The high mortality rate of gynecological cancers can mainly be attributed to ovarian cancer (OC). OC is commonly diagnosed at an advanced stage due to a lack of proper screening tools allowing early detection. Endometrial cancer (EC) on the contrary, is mostly diagnosed at an early stage and has, in general, better outcomes. The incidence of nonendometrioid EC has increased in the last decade, displaying a shared tumor biology with OC and consequently significantly worse outcome. New approaches allowing detection of gynecological cancers in an early stage are therefore desired. Recent studies on cancer biology have shown the relevance of altered glycosylation in the occurrence and progression of cancer. The aberrant expression of sialic acid, a specific carbohydrate terminating glycoproteins and glycolipids on the cell‐surface, is frequently correlated with malignancy. We aimed to determine the current understanding of sialic acid function in different gynecological cancers to identify the gaps in knowledge and its potential use for new diagnostic and therapeutic avenues. Therefore we performed a review on current literature focusing on studies where sialylation was linked to gynecological cancers. The identified studies showed elevated levels of sialic acid in serum, tissue and sialylated antigens in most patients with gynecological cancers, underlining its potential for diagnosis.
What's new?
Recent studies have shown the relevance of altered glycosylation in the occurrence and progression of cancer. In this review, the authors found elevated levels of sialic acid in serum and tissue and high levels of sialylated antigens in most patients with gynaecological cancers, underlining the potential of sialic acid for diagnosis. Elevated levels of sialylation were related with tumour growth, poor differentiation, inhibition of apoptosis, and chemoresistance. Taken together, the studies suggest that sialylation levels could be used to discriminate healthy and benign samples from cancer samples and even early and advanced stages in ovarian, cervical, and endometrial cancer.
Collapse
Affiliation(s)
- Anna Y Berghuis
- Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Johan F A Pijnenborg
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Thomas J Boltje
- Cluster for Molecular Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Johanna M A Pijnenborg
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, The Netherlands.,Radboud Institute of Health Science, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Wu ZL, Ertelt JM. Fluorescent glycan fingerprinting of SARS2 spike proteins. Sci Rep 2021; 11:20428. [PMID: 34650101 PMCID: PMC8516889 DOI: 10.1038/s41598-021-98919-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 09/15/2021] [Indexed: 11/08/2022] Open
Abstract
Glycosylation is the most common post-translational modification and has myriad of biological functions. However, glycan analysis has always been a challenge. Here, we would like to present new techniques for glycan fingerprinting based on enzymatic fluorescent labeling and gel electrophoresis. The method is illustrated on SARS2 spike (S) glycoproteins. SARS2, a novel coronavirus and the causative agent of the COVID-19 pandemic, has had significant social and economic impacts since the end of 2019. To obtain the N-glycan fingerprint of an S protein, glycans released from the protein are first labeled through enzymatic incorporation of fluorophore-conjugated sialic acid or fucose, then separated by SDS-PAGE, and finally visualized with a fluorescent imager. To identify the labeled glycans of a fingerprint, glycan standards and glycan ladders are enzymatically generated and run alongside the samples as references. By comparing the mobility of a labeled glycan to that of a glycan standard, the identity of glycans maybe determined. O-glycans can also be fingerprinted. Due to the lack of an enzyme for broad O-glycan release, O-glycans on the S protein can be labeled with fluorescent sialic acid and digested with trypsin to obtain labeled glycan peptides that are then separated by gel electrophoresis. Glycan fingerprinting could serve as a quick method for globally assessing the glycosylation of a specific glycoprotein.
Collapse
Affiliation(s)
- Zhengliang L Wu
- Bio-techne, R&D Systems, Inc., 614 McKinley Place N.E., Minneapolis, MN, 55413, USA.
| | - James M Ertelt
- Bio-techne, R&D Systems, Inc., 614 McKinley Place N.E., Minneapolis, MN, 55413, USA
| |
Collapse
|
31
|
Serra G, Memo L, Antona V, Corsello G, Favero V, Lago P, Giuffrè M. Jacobsen syndrome and neonatal bleeding: report on two unrelated patients. Ital J Pediatr 2021; 47:147. [PMID: 34210338 PMCID: PMC8252210 DOI: 10.1186/s13052-021-01108-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/11/2021] [Indexed: 11/10/2022] Open
Abstract
Introduction In 1973, Petrea Jacobsen described the first patient showing dysmorphic features, developmental delay and congenital heart disease (atrial and ventricular septal defect) associated to a 11q deletion, inherited from the father. Since then, more than 200 patients have been reported, and the chromosomal critical region responsible for this contiguous gene disorder has been identified. Patients’ presentation We report on two unrelated newborns observed in Italy affected by Jacobsen syndrome (JBS, also known as 11q23 deletion). Both patients presented prenatal and postnatal bleeding, growth and developmental delay, craniofacial dysmorphisms, multiple congenital anomalies, and pancytopenia of variable degree. Array comparative genomic hybridization (aCGH) identified a terminal deletion at 11q24.1-q25 of 12.5 Mb and 11 Mb, in Patient 1 and 2, respectively. Fluorescent in situ hybridization (FISH) analysis of the parents documented a de novo origin of the deletion for Patient 1; parents of Patient 2 refused further genetic investigations. Conclusions Present newborns show the full phenotype of JBS including thrombocytopenia, according to their wide 11q deletion size. Bleeding was particularly severe in one of them, leading to a cerebral hemorrhage. Our report highlights the relevance of early diagnosis, genetic counselling and careful management and follow-up of JBS patients, which may avoid severe clinical consequences and lower the mortality risk. It may provide further insights and a better characterization of JBS, suggesting new elements of the genotype-phenotype correlations.
Collapse
Affiliation(s)
- Gregorio Serra
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy.
| | - Luigi Memo
- Clinical Genetics Outpatient Service, Neonatology and Neonatal Intensive Care Unit, San Bortolo Hospital, Vicenza, Italy
| | - Vincenzo Antona
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Giovanni Corsello
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Valentina Favero
- Neonatal Intensive Care Unit, Ca' Foncello Hospital, Treviso, Italy
| | - Paola Lago
- Neonatal Intensive Care Unit, Ca' Foncello Hospital, Treviso, Italy
| | - Mario Giuffrè
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| |
Collapse
|
32
|
Nguyen NTB, Lin J, Tay SJ, Mariati, Yeo J, Nguyen-Khuong T, Yang Y. Multiplexed engineering glycosyltransferase genes in CHO cells via targeted integration for producing antibodies with diverse complex-type N-glycans. Sci Rep 2021; 11:12969. [PMID: 34155258 PMCID: PMC8217518 DOI: 10.1038/s41598-021-92320-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/09/2021] [Indexed: 02/05/2023] Open
Abstract
Therapeutic antibodies are decorated with complex-type N-glycans that significantly affect their biodistribution and bioactivity. The N-glycan structures on antibodies are incompletely processed in wild-type CHO cells due to their limited glycosylation capacity. To improve N-glycan processing, glycosyltransferase genes have been traditionally overexpressed in CHO cells to engineer the cellular N-glycosylation pathway by using random integration, which is often associated with large clonal variations in gene expression levels. In order to minimize the clonal variations, we used recombinase-mediated-cassette-exchange (RMCE) technology to overexpress a panel of 42 human glycosyltransferase genes to screen their impact on antibody N-linked glycosylation. The bottlenecks in the N-glycosylation pathway were identified and then released by overexpressing single or multiple critical genes. Overexpressing B4GalT1 gene alone in the CHO cells produced antibodies with more than 80% galactosylated bi-antennary N-glycans. Combinatorial overexpression of B4GalT1 and ST6Gal1 produced antibodies containing more than 70% sialylated bi-antennary N-glycans. In addition, antibodies with various tri-antennary N-glycans were obtained for the first time by overexpressing MGAT5 alone or in combination with B4GalT1 and ST6Gal1. The various N-glycan structures and the method for producing them in this work provide opportunities to study the glycan structure-and-function and develop novel recombinant antibodies for addressing different therapeutic applications.
Collapse
Affiliation(s)
- Ngan T. B. Nguyen
- grid.452198.30000 0004 0485 9218Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jianer Lin
- grid.452198.30000 0004 0485 9218Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Shi Jie Tay
- grid.452198.30000 0004 0485 9218Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mariati
- grid.452198.30000 0004 0485 9218Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jessna Yeo
- grid.452198.30000 0004 0485 9218Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Terry Nguyen-Khuong
- grid.452198.30000 0004 0485 9218Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yuansheng Yang
- grid.452198.30000 0004 0485 9218Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
33
|
CMAS and ST3GAL4 Play an Important Role in the Adsorption of Influenza Virus by Affecting the Synthesis of Sialic Acid Receptors. Int J Mol Sci 2021; 22:ijms22116081. [PMID: 34200006 PMCID: PMC8200212 DOI: 10.3390/ijms22116081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Influenza A viruses (IAVs) initiate infection by attaching Hemagglutinin (HA) on the viral envelope to sialic acid (SA) receptors on the cell surface. Importantly, HA of human IAVs has a higher affinity for α-2,6-linked SA receptors, and avian strains prefer α-2,3-linked SA receptors, whereas swine strains have a strong affinity for both SA receptors. Host gene CMAS and ST3GAL4 were found to be essential for IAV attachment and entry. Loss of CMAS and ST3GAL4 hindered the synthesis of sialic acid receptors, which in turn prevented the adsorption of IAV. Further, the knockout of CMAS had an effect on the adsorption of swine, avian and human IAVs. However, ST3GAL4 knockout prevented the adsorption of swine and avian IAV and the impact on avian IAV was more distinct, whereas it had no effect on the adsorption of human IAV. Collectively, our findings demonstrate that knocking out CMAS and ST3GAL4 negatively regulated IAV replication by inhibiting the synthesis of SA receptors, which also provides new insights into the production of gene-edited animals in the future.
Collapse
|
34
|
Role of Glycans on Key Cell Surface Receptors That Regulate Cell Proliferation and Cell Death. Cells 2021; 10:cells10051252. [PMID: 34069424 PMCID: PMC8159107 DOI: 10.3390/cells10051252] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Cells undergo proliferation and apoptosis, migration and differentiation via a number of cell surface receptors, most of which are heavily glycosylated. This review discusses receptor glycosylation and the known roles of glycans on the functions of receptors expressed in diverse cell types. We included growth factor receptors that have an intracellular tyrosine kinase domain, growth factor receptors that have a serine/threonine kinase domain, and cell-death-inducing receptors. N- and O-glycans have a wide range of functions including roles in receptor conformation, ligand binding, oligomerization, and activation of signaling cascades. A better understanding of these functions will enable control of cell survival and cell death in diseases such as cancer and in immune responses.
Collapse
|
35
|
Rab11-mediated post-Golgi transport of the sialyltransferase ST3GAL4 suggests a new mechanism for regulating glycosylation. J Biol Chem 2021; 296:100354. [PMID: 33524390 PMCID: PMC7949161 DOI: 10.1016/j.jbc.2021.100354] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Glycosylation, the most common posttranslational modification of proteins, is a stepwise process that relies on tight regulation of subcellular glycosyltransferase location to control the addition of each monosaccharide. Glycosyltransferases primarily reside and function in the endoplasmic reticulum (ER) and the Golgi apparatus; whether and how they traffic beyond the Golgi, how this trafficking is controlled, and how it impacts glycosylation remain unclear. Our previous work identified a connection between N-glycosylation and Rab11, a key player in the post-Golgi transport that connects recycling endosomes and other compartments. To learn more about the specific role of Rab11, we knocked down Rab11 in HeLa cells. Our findings indicate that Rab11 knockdown results in a dramatic enhancement in the sialylation of N-glycans. Structural analyses of glycans using lectins and LC-MS revealed that α2,3-sialylation is selectively enhanced, suggesting that an α2,3-sialyltransferase that catalyzes the sialyation of glycoproteins is activated or upregulated as the result of Rab11 knockdown. ST3GAL4 is the major α2,3-sialyltransferase that acts on N-glycans; we demonstrated that the localization of ST3GAL4, but not the levels of its mRNA, protein, or donor substrate, was altered by Rab11 depletion. In knockdown cells, ST3GAL4 is densely distributed in the trans-Golgi network, compared with the wider distribution in the Golgi and in other peripheral puncta in control cells, whereas the α2,6-sialyltransferase ST6GAL1 is predominantly localized to the Golgi regardless of Rab11 knockdown. This indicates that Rab11 may negatively regulate α2,3-sialylation by transporting ST3GAL4 to post-Golgi compartments (PGCs), which is a novel mechanism of glycosyltransferase regulation.
Collapse
|
36
|
Wu Y, Chen X, Dong W, Xu Z, Jian Y, Xu C, Zhang L, Wei A, Yu X, Wang S, Wang Y, Liu G, Sun X, Wang S. ST3Gal IV Mediates the Growth and Proliferation of Cervical Cancer Cells In Vitro and In Vivo Via the Notch/p21/CDKs Pathway. Front Oncol 2021; 10:540332. [PMID: 33598419 PMCID: PMC7882721 DOI: 10.3389/fonc.2020.540332] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022] Open
Abstract
ST3Gal IV is one of the principal sialyltransferases responsible for the biosynthesis of α2, 3-sialic acid to the termini N-glycans or O-glycans of glycoproteins and glycolipids. It has been reported that ST3Gal IV expression is associated with gastric carcinoma, pancreatic adenocarcinoma and breast cancer. While the expression and functions of ST3Gal IV in cervical cancer are still poorly understood. In this study, we found that ST3Gal IV was downregulated in human cervical cancer tissues compared to normal cervix tissues, and ST3Gal IV expression was negatively associated with the pathological grade of cervical cancer. ST3Gal IV upregulation inhibited the growth and proliferation of cervical cancer HeLa and SiHa cells in vitro and in vivo. Furthermore, ST3Gal IV overexpression enhanced the expression of several Notch pathway components such as Jagged1, Notch1, Hes1 and Hey1, while cell cycle protein expression like Cyclin D1, Cyclin E1, CDK2 and CDK4 were decreased. These results indicate that expression of ST3Gal IV is reduced in cervical cancer and plays a negative role in cell proliferation via Notch/p21/CDKs signaling pathway. Thus, sialyltransferase ST3Gal IV might be a target for the diagnosis and therapy of cervical cancer.
Collapse
Affiliation(s)
- Yinshuang Wu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Xixi Chen
- Department of Biological Sciences, School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| | - Weijie Dong
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Zhongyang Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Yuli Jian
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Chunyan Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Anwen Wei
- Department of Gynaecology, Jiaxing University Affiliated Women and Children Hospital, Jiaxing, China
| | - Xiao Yu
- Department of Pathology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Shidan Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Yue Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Gang Liu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Xiaoxin Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Institute of Glycobiology, Dalian Medical University, Dalian, China
| |
Collapse
|
37
|
Noor SI, Hoffmann M, Rinis N, Bartels MF, Winterhalter PR, Hoelscher C, Hennig R, Himmelreich N, Thiel C, Ruppert T, Rapp E, Strahl S. Glycosyltransferase POMGNT1 deficiency strengthens N-cadherin-mediated cell-cell adhesion. J Biol Chem 2021; 296:100433. [PMID: 33610554 PMCID: PMC7994789 DOI: 10.1016/j.jbc.2021.100433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Defects in protein O-mannosylation lead to severe congenital muscular dystrophies collectively known as α-dystroglycanopathy. A hallmark of these diseases is the loss of the O-mannose-bound matriglycan on α-dystroglycan, which reduces cell adhesion to the extracellular matrix. Mutations in protein O-mannose β1,2-N-acetylglucosaminyltransferase 1 (POMGNT1), which is crucial for the elongation of O-mannosyl glycans, have mainly been associated with muscle-eye-brain (MEB) disease. In addition to defects in cell-extracellular matrix adhesion, aberrant cell-cell adhesion has occasionally been observed in response to defects in POMGNT1. However, specific molecular consequences of POMGNT1 deficiency on cell-cell adhesion are largely unknown. We used POMGNT1 knockout HEK293T cells and fibroblasts from an MEB patient to gain deeper insight into the molecular changes in POMGNT1 deficiency. Biochemical and molecular biological techniques combined with proteomics, glycoproteomics, and glycomics revealed that a lack of POMGNT1 activity strengthens cell-cell adhesion. We demonstrate that the altered intrinsic adhesion properties are due to an increased abundance of N-cadherin (N-Cdh). In addition, site-specific changes in the N-glycan structures in the extracellular domain of N-Cdh were detected, which positively impact on homotypic interactions. Moreover, in POMGNT1-deficient cells, ERK1/2 and p38 signaling pathways are activated and transcriptional changes that are comparable with the epithelial-mesenchymal transition (EMT) are triggered, defining a possible molecular mechanism underlying the observed phenotype. Our study indicates that changes in cadherin-mediated cell-cell adhesion and other EMT-related processes may contribute to the complex clinical symptoms of MEB or α-dystroglycanopathy in general and suggests that the impact of changes in O-mannosylation on N-glycosylation has been underestimated.
Collapse
Affiliation(s)
- Sina Ibne Noor
- Centre for Organismal Studies (COS), Glycobiology, Heidelberg University, Heidelberg, Germany
| | - Marcus Hoffmann
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany
| | - Natalie Rinis
- Centre for Organismal Studies (COS), Glycobiology, Heidelberg University, Heidelberg, Germany
| | - Markus F Bartels
- Centre for Organismal Studies (COS), Glycobiology, Heidelberg University, Heidelberg, Germany
| | - Patrick R Winterhalter
- Centre for Organismal Studies (COS), Glycobiology, Heidelberg University, Heidelberg, Germany
| | - Christina Hoelscher
- Centre for Organismal Studies (COS), Glycobiology, Heidelberg University, Heidelberg, Germany
| | - René Hennig
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany; glyXera GmbH, Magdeburg, Germany
| | - Nastassja Himmelreich
- Center for Child and Adolescent Medicine, Department Pediatrics I, University of Heidelberg, Heidelberg, Germany
| | - Christian Thiel
- Center for Child and Adolescent Medicine, Department Pediatrics I, University of Heidelberg, Heidelberg, Germany
| | - Thomas Ruppert
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Germany; glyXera GmbH, Magdeburg, Germany
| | - Sabine Strahl
- Centre for Organismal Studies (COS), Glycobiology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
38
|
Dobie C, Skropeta D. Insights into the role of sialylation in cancer progression and metastasis. Br J Cancer 2020; 124:76-90. [PMID: 33144696 PMCID: PMC7782833 DOI: 10.1038/s41416-020-01126-7] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/11/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
Upregulation of sialyltransferases—the enzymes responsible for the addition of sialic acid to growing glycoconjugate chains—and the resultant hypersialylation of up to 40–60% of tumour cell surfaces are established hallmarks of several cancers, including lung, breast, ovarian, pancreatic and prostate cancer. Hypersialylation promotes tumour metastasis by several routes, including enhancing immune evasion and tumour cell survival, and stimulating tumour invasion and migration. The critical role of enzymes that regulate sialic acid in tumour cell growth and metastasis points towards targeting sialylation as a potential new anti-metastatic cancer treatment strategy. Herein, we explore insights into the mechanisms by which hypersialylation plays a role in promoting metastasis, and explore the current state of sialyltransferase inhibitor development.
Collapse
Affiliation(s)
- Christopher Dobie
- School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine & Health; and Molecular Horizons, University of Wollongong, NSW, 2522, Wollongong, Australia
| | - Danielle Skropeta
- School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine & Health; and Molecular Horizons, University of Wollongong, NSW, 2522, Wollongong, Australia. .,Illawarra Health & Medical Research Institute, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
39
|
Han J, Pan Y, Qin W, Gu Y, Xu X, Zhao R, Sha J, Zhang R, Gu J, Ren S. Quantitation of sex-specific serum N-glycome changes in expression level during mouse aging based on Bionic Glycome method. Exp Gerontol 2020; 141:111098. [PMID: 33010330 DOI: 10.1016/j.exger.2020.111098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/27/2020] [Accepted: 09/22/2020] [Indexed: 12/24/2022]
Abstract
Studying the changes of serum N-glycome during mouse aging is beneficial to explore the molecular basis behind the alterations reported in human. However, such studies remainscarce and lack some information such as sialylation due to the method limitation. Here, we introduced Bionic Glycome method to quantify the serum N-glycome changes during C57BL/6 mouse aging (from the pubertal period to the old age stage). This technique enabled reliable and comprehensive quantitation of the expression level changes of more than 20 N-glycans in mouse serum at 12 time points in both genders for the first time, involving the analysis of sialic acid and its different linkages. The results demonstrated that the expression level of total glycans increased from middle age to old age. Interestingly, sex-specific N-glycome profiles and alterations were observed. Female mice showed higher level of serum fucosylation and lower level of serum afucosylation than male mice (fucosylation: p < 1.0E-6; afucosylation: p < 1.0E-6). Obviously, higher increase of serum fucosylation level was found in female mice than in male mice from middle age to old age. In addition, the opposite alterations of the afucosylated glycans with α2,3-linked sialic acid and those only with α2,6-linked sialic acid were observed at old age in male mice. These findings suggested that N-glycome could be a valuable target for investigating aging and possible contributors to aging.
Collapse
Affiliation(s)
- Jing Han
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yiqing Pan
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wenjun Qin
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Yong Gu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoyan Xu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Ran Zhao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200090, China
| | - Jichen Sha
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Rongrong Zhang
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jianxin Gu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shifang Ren
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
40
|
Dalangood S, Zhu Z, Ma Z, Li J, Zeng Q, Yan Y, Shen B, Yan J, Huang R. Identification of glycogene-type and validation of ST3GAL6 as a biomarker predicts clinical outcome and cancer cell invasion in urinary bladder cancer. Theranostics 2020; 10:10078-10091. [PMID: 32929335 PMCID: PMC7481430 DOI: 10.7150/thno.48711] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Urinary bladder cancer (UBC) is one of the most common causes of morbidity and mortality worldwide characterized by a high risk of invasion and metastasis; however, the molecular classification biomarkers and underlying molecular mechanisms for UBC patient stratification on clinical outcome need to be investigated. Methods: A systematic transcriptomic analysis of 185 glycogenes in the public UBC datasets with survival information and clinicopathological parameters were performed using unsupervised hierarchical clustering. The gene signature for glycogene-type classification was identified using Limma package in R language, and correlated to 8 known molecular features by Gene Set Variation Analysis (GSVA). The clinical relevance and function of a glycogene was characterized by immunohistochemistry in UBC patient samples, and quantitative RT-PCR, Western blotting, promoter activity, MAL II blotting, immunofluorescence staining, wound healing, and transwell assays in UBC cells. Results: A 14-glycogene signature for glycogene-type classification was identified. Among them, ST3GAL6, a glycotransferase to transfer sialic acid to 3'-hydroxyl group of a galactose residue, showed a significant negative association with the subtype with luminal feature in UBC patients (n=2,130 in total). Increased ST3GAL6 was positively correlated to tumor stage, grade, and survival in UBCs from public datasets or our cohort (n=52). Transcription factor GATA3, a luminal-specific marker for UBC, was further identified as a direct upstream regulator of ST3GAL6 to negatively regulate its transactivation. ST3GAL6 depletion decreased MAL II level, cell invasion and migration in 5637 and J82 UBC cells. ST3GAL6 could reverse the effects of GATA3 on global sialylation and cell invasion in SW780 cells. Conclusions: Herein, we successfully identified a novel 14-gene signature for glycogene-type classification of UBC patients. ST3GAL6 gene, from this signature, was demonstrated as a potential biomarker for poor outcomes and cell invasion in UBCs.
Collapse
Affiliation(s)
- Sumiya Dalangood
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing 210061, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhen Zhu
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Zhihui Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaxuan Li
- MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Qinghe Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yilin Yan
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Bing Shen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Jun Yan
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- Model Animal Research Center of Nanjing University, Nanjing 210061, China
| | - Ruimin Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
41
|
Gu Y, Han J, Liu X, Pan Y, Xu X, Sha J, Ren S, Gu J. Dynamic alterations in serum IgG N-glycan profiles in the development of colitis-associated colon Cancer in mouse model. Biochim Biophys Acta Gen Subj 2020; 1864:129668. [PMID: 32553689 DOI: 10.1016/j.bbagen.2020.129668] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Alternative glycosylation of serum IgG has been shown to be closely associated with colorectal cancer (CRC). Currently, a dynamic study which can not only minimize the influence of genetic background, environment and other interfering factors during cancer development, but also focus on investigating carcinogenic characteristics of IgG glycan is lacking. METHODS Serum IgG N-glycans were characterized at four stages of CRC development by ultra-performance liquid chromatography in a typical colitis-related CRC mouse model induced by azoxymethane-dextran sodium sulfate. Furthermore, the expression of related glycosyltransferases in splenic B lymphocytes at the corresponding time was also assessed. RESULTS The relative abundance of seven IgG glycans, which can be classified as monoantennary, core fucose, sialic acid, galactose and bisecting, was changed during tumor growth. The abundance of some glycans was altered during the first stage of cancer induction. Correspondingly, the expression of glycosyltransferases in splenic B lymphocytes and different tissues in cancer groups was also decreased compared to that in controls. CONCLUSIONS This study represents the comprehensive analysis of IgG glycosylation in the dynamic process of colitis-associated CRC. To our knowledge, this is the first report that the expression of glycosyltransferases in mouse splenic B lymphocytes is consistent or inconsistent with the alterations of IgG N-glycans, and the variation tendency is tissue nonspecific. GENERAL SIGNIFICANCE Providing a novel approach to identify the IgG glycans related to the development of CRC and laying a foundation for research on structure and function of glycans using mouse.
Collapse
Affiliation(s)
- Yong Gu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jing Han
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xin Liu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yiqing Pan
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoyan Xu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jichen Sha
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shifang Ren
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Jianxin Gu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|