101
|
Baskakov IV. Role of sialylation in prion disease pathogenesis and prion structure. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:31-52. [PMID: 32958238 DOI: 10.1016/bs.pmbts.2020.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mammalian prion or PrPSc is a proteinaceous infectious agent that consists of a misfolded, self-replicating state of a sialoglycoprotein called the prion protein or PrPC. Sialylation of the prion protein, a terminal modification of N-linked glycans, was discovered more than 30 years ago, yet the role of sialylation in prion pathogenesis is not well understood. This chapter summarizes current knowledge on the role of sialylation of the prion protein in prion diseases. First, we discuss recent data suggesting that sialylation of PrPSc N-linked glycans determines the fate of prion infection in an organism and control prion lymphotropism. Second, emerging evidence pointing out at the role N-glycans in neuroinflammation are discussed. Thirds, this chapter reviews a mechanism postulating that sialylated N-linked glycans are important players in defining strain-specific structures. A new hypothesis according to which individual strain-specific PrPSc structures govern selection of PrPC sialoglycoforms is discussed. Finally, this chapter explain how N-glycan sialylation control the prion replication and strain interference. In summary, comprehensive review of our knowledge on N-linked glycans and their sialylation provided in this chapter helps to answer important questions of prion biology that have been puzzling for years.
Collapse
Affiliation(s)
- Ilia V Baskakov
- Department of Anatomy and Neurobiology, and Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
102
|
Mank M, Hauner H, Heck AJR, Stahl B. Targeted LC-ESI-MS 2 characterization of human milk oligosaccharide diversity at 6 to 16 weeks post-partum reveals clear staging effects and distinctive milk groups. Anal Bioanal Chem 2020; 412:6887-6907. [PMID: 32794008 PMCID: PMC7496073 DOI: 10.1007/s00216-020-02819-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/03/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022]
Abstract
Many molecular components in human milk (HM), such as human milk oligosaccharides (HMOs), assist in the healthy development of infants. It has been hypothesized that the functional benefits of HM may be highly dependent on the abundance and individual fine structures of contained HMOs and that distinctive HM groups can be defined by their HMO profiles. However, the structural diversity and abundances of individual HMOs may also vary between milk donors and at different stages of lactations. Improvements in efficiency and selectivity of quantitative HMO analysis are essential to further expand our understanding about the impact of HMO variations on healthy early life development. Hence, we applied here a targeted, highly selective, and semi-quantitative LC-ESI-MS2 approach by analyzing 2 × 30 mature human milk samples collected at 6 and 16 weeks post-partum. The analytical approach covered the most abundant HMOs up to hexasaccharides and, for the first time, also assigned blood group A and B tetrasaccharides. Principal component analysis (PCA) was employed and allowed for automatic grouping and assignment of human milk samples to four human milk groups which are related to the maternal Secretor (Se) and Lewis (Le) genotypes. We found that HMO diversity varied significantly between these four HM groups. Variations were driven by HMOs being either dependent or independent of maternal genetic Se and Le status. We found preliminary evidence for an additional HM subgroup within the Se- and Le-positive HM group I. Furthermore, the abundances of 6 distinct HMO structures (including 6'-SL and 3-FL) changed significantly with progression of lactation. Graphical abstract.
Collapse
Affiliation(s)
- Marko Mank
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands.
| | - Hans Hauner
- Else Kröner-Fresenius Center for Nutritional Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81675, Munich, Germany.,Nutritional Medicine Unit, Research Center for Nutrition and Food Sciences (ZIEL), Technische Universität München, Weihenstephaner Berg 1, 85354, Freising, Germany
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584 CH, Utrecht, The Netherlands.,Netherlands Proteomics Center, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Bernd Stahl
- Danone Nutricia Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| |
Collapse
|
103
|
Glanz VY, Myasoedova VA, Grechko AV, Orekhov AN. Trans-sialidase Associated with Atherosclerosis: Defining the Identity of a Key Enzyme Involved in the Pathology. Curr Drug Targets 2020; 20:938-941. [PMID: 30848200 DOI: 10.2174/1389450120666190308111619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/11/2019] [Accepted: 02/27/2019] [Indexed: 01/05/2023]
Abstract
Atherosclerosis is associated with the increased trans-sialidase activity, which can be detected in the blood plasma of atherosclerosis patients. The likely involvement in the disease pathogenesis made this activity an interesting research subject and the enzyme that may perform such activity was isolated and characterized in terms of substrate specificity and enzymatic properties. It was found that the enzyme has distinct optimum pH values, and its activity was enhanced by the presence of Ca2+ ions. Most importantly, the enzyme was able to cause atherogenic modification of lowdensity lipoprotein (LDL) particles in vitro. However, the identity of the discovered enzyme remained to be defined. Currently, sialyltransferases, mainly ST6Gal I, are regarded as major contributors to sialic acid metabolism in human blood. In this mini-review, we discuss the possibility that atherosclerosis- associated trans-sialidase does, in fact, belong to the sialyltransferases family.
Collapse
Affiliation(s)
- Victor Y Glanz
- Department of Genetics, Cytology and Bioengineering, Faculty of Biology and Medicine, Voronezh State University, Voronezh, Russian Federation
| | - Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russian Federation
| | - Andrey V Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 109240 Moscow, Russian Federation
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russian Federation.,Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russian Federation.,Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russian Federation
| |
Collapse
|
104
|
D’Addio M, Frey J, Otto VI. The manifold roles of sialic acid for the biological functions of endothelial glycoproteins. Glycobiology 2020; 30:490-499. [PMID: 32039454 PMCID: PMC7372927 DOI: 10.1093/glycob/cwaa008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Vascular endothelia are covered with a dense glycocalix that is heavily sialylated. Sialylation of vascular glycoconjugates is involved in the regulation of cell-cell interactions, be it among endothelial cells at cell junctions or between endothelial and blood-borne cells. It also plays important roles in modulating the binding of soluble ligands and the signaling by vascular receptors. Here, we provide an overview over the sialylation-function relationships of glycoproteins expressed in the blood and lymphatic vasculature. We first describe cellular interactions in which sialic acid contributes in a stereospecific manner to glycan epitopes recognized by glycan-binding proteins. Our major focus is however on the rarely discussed examples of vascular glycoproteins whose biological functions are modulated by sialylation through other mechanisms.
Collapse
Affiliation(s)
- Marco D’Addio
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Jasmin Frey
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Vivianne I Otto
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Sciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| |
Collapse
|
105
|
Tvaroška I, Selvaraj C, Koča J. Selectins-The Two Dr. Jekyll and Mr. Hyde Faces of Adhesion Molecules-A Review. Molecules 2020; 25:molecules25122835. [PMID: 32575485 PMCID: PMC7355470 DOI: 10.3390/molecules25122835] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Selectins belong to a group of adhesion molecules that fulfill an essential role in immune and inflammatory responses and tissue healing. Selectins are glycoproteins that decode the information carried by glycan structures, and non-covalent interactions of selectins with these glycan structures mediate biological processes. The sialylated and fucosylated tetrasaccharide sLex is an essential glycan recognized by selectins. Several glycosyltransferases are responsible for the biosynthesis of the sLex tetrasaccharide. Selectins are involved in a sequence of interactions of circulated leukocytes with endothelial cells in the blood called the adhesion cascade. Recently, it has become evident that cancer cells utilize a similar adhesion cascade to promote metastases. However, like Dr. Jekyll and Mr. Hyde’s two faces, selectins also contribute to tissue destruction during some infections and inflammatory diseases. The most prominent function of selectins is associated with the initial stage of the leukocyte adhesion cascade, in which selectin binding enables tethering and rolling. The first adhesive event occurs through specific non-covalent interactions between selectins and their ligands, with glycans functioning as an interface between leukocytes or cancer cells and the endothelium. Targeting these interactions remains a principal strategy aimed at developing new therapies for the treatment of immune and inflammatory disorders and cancer. In this review, we will survey the significant contributions to and the current status of the understanding of the structure of selectins and the role of selectins in various biological processes. The potential of selectins and their ligands as therapeutic targets in chronic and acute inflammatory diseases and cancer will also be discussed. We will emphasize the structural characteristic of selectins and the catalytic mechanisms of glycosyltransferases involved in the biosynthesis of glycan recognition determinants. Furthermore, recent achievements in the synthesis of selectin inhibitors will be reviewed with a focus on the various strategies used for the development of glycosyltransferase inhibitors, including substrate analog inhibitors and transition state analog inhibitors, which are based on knowledge of the catalytic mechanism.
Collapse
Affiliation(s)
- Igor Tvaroška
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
- Institute of Chemistry, Slovak Academy of Sciences, 84538 Bratislava, Slovak Republic
- Correspondence: (I.T.); (J.K.); Tel.: +421-948-535-601 (I.T.); +420-731-682-606 (J.K.)
| | - Chandrabose Selvaraj
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
| | - Jaroslav Koča
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, 62500 Brno, Czech Republic
- Correspondence: (I.T.); (J.K.); Tel.: +421-948-535-601 (I.T.); +420-731-682-606 (J.K.)
| |
Collapse
|
106
|
Puigdellívol M, Allendorf DH, Brown GC. Sialylation and Galectin-3 in Microglia-Mediated Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2020; 14:162. [PMID: 32581723 PMCID: PMC7296093 DOI: 10.3389/fncel.2020.00162] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022] Open
Abstract
Microglia are brain macrophages that mediate neuroinflammation and contribute to and protect against neurodegeneration. The terminal sugar residue of all glycoproteins and glycolipids on the surface of mammalian cells is normally sialic acid, and addition of this negatively charged residue is known as “sialylation,” whereas removal by sialidases is known as “desialylation.” High sialylation of the neuronal cell surface inhibits microglial phagocytosis of such neurons, via: (i) activating sialic acid receptors (Siglecs) on microglia that inhibit phagocytosis and (ii) inhibiting binding of opsonins C1q, C3, and galectin-3. Microglial sialylation inhibits inflammatory activation of microglia via: (i) activating Siglec receptors CD22 and CD33 on microglia that inhibit phagocytosis and (ii) inhibiting Toll-like receptor 4 (TLR4), complement receptor 3 (CR3), and other microglial receptors. When activated, microglia release a sialidase activity that desialylates both microglia and neurons, activating the microglia and rendering the neurons susceptible to phagocytosis. Activated microglia also release galectin-3 (Gal-3), which: (i) further activates microglia via binding to TLR4 and TREM2, (ii) binds to desialylated neurons opsonizing them for phagocytosis via Mer tyrosine kinase, and (iii) promotes Aβ aggregation and toxicity in vivo. Gal-3 and desialylation may increase in a variety of brain pathologies. Thus, Gal-3 and sialidases are potential treatment targets to prevent neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - David H Allendorf
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
107
|
Montgomery AP, Dobie C, Szabo R, Hallam L, Ranson M, Yu H, Skropeta D. Design, synthesis and evaluation of carbamate-linked uridyl-based inhibitors of human ST6Gal I. Bioorg Med Chem 2020; 28:115561. [PMID: 32616185 DOI: 10.1016/j.bmc.2020.115561] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/14/2020] [Accepted: 05/16/2020] [Indexed: 12/22/2022]
Abstract
Sialic acid at the terminus of cell surface glycoconjugates is a critical element in cell-cell recognition, receptor binding and immune responses. Sialyltransferases (ST), the enzymes responsible for the biosynthesis of sialylated glycans are highly upregulated in cancer and the resulting hypersialylation of the tumour cell surface correlates strongly with tumour growth, metastasis and drug resistance. Inhibitors of human STs, in particular human ST6Gal I, are thus expected to be valuable chemical tools for the discovery of novel anticancer drugs. Herein, we report on the computationally-guided design and development of uridine-based inhibitors that replace the charged phosphodiester linker of known ST inhibitors with a neutral carbamate to improve pharmacokinetic properties and synthetic accessibility. A series of 24 carbamate-linked uridyl-based compounds were synthesised by coupling aryl and hetaryl α-hydroxyphosphonates with a 5'-amino-5'-deoxyuridine fragment. The inhibitory activities of the newly synthesised compounds against recombinant human ST6Gal I were determined using a luminescent microplate assay, and five promising inhibitors with Ki's ranging from 1 to 20 µM were identified. These results show that carbamate-linked uridyl-based compounds are a potential new class of readily accessible, non-cytotoxic ST inhibitors to be further explored.
Collapse
Affiliation(s)
- Andrew P Montgomery
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Christopher Dobie
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Rémi Szabo
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Laura Hallam
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Marie Ranson
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Haibo Yu
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Danielle Skropeta
- Molecular Horizons and School of Chemistry & Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia; Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.
| |
Collapse
|
108
|
Martínez-Morales PL, Ortiz-Mateos CA, Reyes-Pineda J, Reyes-Vallejo T, Aguilar-Lemarroy A, Jave-Suárez LF, Santos-López G, Reyes-Leyva J, Milflores-Flores L, Vallejo-Ruiz V. Identification and characterization of the V3 promoter of the ST3GAL4 gene. Biosci Trends 2020; 14:144-150. [DOI: 10.5582/bst.2019.01331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Patricia L. Martínez-Morales
- CONACYT- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Oriente, Metepec, Puebla, Mexico
| | | | - Jonatan Reyes-Pineda
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | | | - Adriana Aguilar-Lemarroy
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Occidente, Guadalajara, Jalisco, Mexico
| | - Luis F. Jave-Suárez
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Occidente, Guadalajara, Jalisco, Mexico
| | - Gerardo Santos-López
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Oriente, Laboratorio de Biología Molecular, Metepec, Puebla, Mexico
| | - Julio Reyes-Leyva
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Oriente, Laboratorio de Biología Molecular, Metepec, Puebla, Mexico
| | | | - Verónica Vallejo-Ruiz
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Oriente, Laboratorio de Biología Molecular, Metepec, Puebla, Mexico
| |
Collapse
|
109
|
New Therapeutic Strategies for Osteoarthritis by Targeting Sialic Acid Receptors. Biomolecules 2020; 10:biom10040637. [PMID: 32326143 PMCID: PMC7226619 DOI: 10.3390/biom10040637] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease characterized by articular cartilage degradation and joint degeneration. The articular cartilage is mainly formed by chondrocytes and a collagen-proteoglycan extracellular matrix that contains high levels of glycosylated proteins. It was reported that the shift from glycoproteins containing α-2,6-linked sialic acids to those that contain α-2,3 was associated with the onset of common types of arthritis. However, the pathophysiology of α-2,3-sialylation in cartilage has not been yet elucidated. We show that cartilage from osteoarthritic patients expresses high levels of the α-2,3-sialylated transmembrane mucin receptor, known as podoplanin (PDPN). Additionally, the Maackia amurensis seed lectin (MASL), that can be utilized to target PDPN, attenuates the inflammatory response mediated by NF-kB activation in primary chondrocytes and protects human cartilage breakdown ex vivo and in an animal model of arthritis. These findings reveal that specific lectins targeting α-2,3-sialylated receptors on chondrocytes might effectively inhibit cartilage breakdown. We also present a computational 3D molecular model for this interaction. These findings provide mechanistic information on how a specific lectin could be used as a novel therapy to treat degenerative joint diseases such as osteoarthritis.
Collapse
|
110
|
Ou L, He X, Liu N, Song Y, Li J, Gao L, Huang X, Deng Z, Wang X, Lin S. Sialylation of FGFR1 by ST6Gal‑I overexpression contributes to ovarian cancer cell migration and chemoresistance. Mol Med Rep 2020. [PMID: 32016470 DOI: 10.3892/mmr.2020.10951/html] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Fibroblast growth factor receptors (FGFRs) have been implicated in the malignant transformation and chemoresistance of epithelial ovarian cancer; however, the underlying molecular mechanisms are poorly understood. Increased sialyltransferase activity that enhances protein sialylation is an important post‑translational process promoting cancer progression and malignancy. In the present study, α2,6‑sialyltransferase (ST6Gal‑I) overexpression or knockdown cell lines were developed, and FGFR1 was examined to understand the effect of sialylation on migration and drug resistance, and the underlying mechanisms. It was identified that cells with ST6Gal‑I overexpression had increased cell viability and migratory ability upon serum deprivation. Moreover, ST6Gal‑I overexpression cells had strong resistance to paclitaxel, as demonstrated by low growth inhibition rate and cell apoptosis level. A mechanistic study showed that ST6Gal‑I overexpression induced high α2,6‑sialylation of FGFR1 and increased the expression of phospho‑ERK1/2 and phospho‑focal adhesion kinase. Further study demonstrated that the FGFR1 inhibitor PD173047 reduced cell viability and induced apoptosis; however, ST6Gal‑I overexpression decreased the anticancer effect of PD173047. In addition, ST6Gal‑I overexpression attenuated the effect of Adriamycin on cancer cells. Collectively, these results suggested that FGFR1 sialylation plays an important role in cell migration and drug chemoresistance in ovarian cancer cells.
Collapse
Affiliation(s)
- Lingling Ou
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xiuzhen He
- Chongqing Three Gorges Medical College, Chongqing 404120, P.R. China
| | - Naihua Liu
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yuwei Song
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jinyuan Li
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Lvfen Gao
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xinke Huang
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Zhendong Deng
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaoyu Wang
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Shaoqiang Lin
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
111
|
Ou L, He X, Liu N, Song Y, Li J, Gao L, Huang X, Deng Z, Wang X, Lin S. Sialylation of FGFR1 by ST6Gal‑I overexpression contributes to ovarian cancer cell migration and chemoresistance. Mol Med Rep 2020; 21:1449-1460. [PMID: 32016470 PMCID: PMC7003046 DOI: 10.3892/mmr.2020.10951] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 08/05/2019] [Indexed: 01/01/2023] Open
Abstract
Fibroblast growth factor receptors (FGFRs) have been implicated in the malignant transformation and chemoresistance of epithelial ovarian cancer; however, the underlying molecular mechanisms are poorly understood. Increased sialyltransferase activity that enhances protein sialylation is an important post‑translational process promoting cancer progression and malignancy. In the present study, α2,6‑sialyltransferase (ST6Gal‑I) overexpression or knockdown cell lines were developed, and FGFR1 was examined to understand the effect of sialylation on migration and drug resistance, and the underlying mechanisms. It was identified that cells with ST6Gal‑I overexpression had increased cell viability and migratory ability upon serum deprivation. Moreover, ST6Gal‑I overexpression cells had strong resistance to paclitaxel, as demonstrated by low growth inhibition rate and cell apoptosis level. A mechanistic study showed that ST6Gal‑I overexpression induced high α2,6‑sialylation of FGFR1 and increased the expression of phospho‑ERK1/2 and phospho‑focal adhesion kinase. Further study demonstrated that the FGFR1 inhibitor PD173047 reduced cell viability and induced apoptosis; however, ST6Gal‑I overexpression decreased the anticancer effect of PD173047. In addition, ST6Gal‑I overexpression attenuated the effect of Adriamycin on cancer cells. Collectively, these results suggested that FGFR1 sialylation plays an important role in cell migration and drug chemoresistance in ovarian cancer cells.
Collapse
Affiliation(s)
- Lingling Ou
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xiuzhen He
- Chongqing Three Gorges Medical College, Chongqing 404120, P.R. China
| | - Naihua Liu
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yuwei Song
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jinyuan Li
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Lvfen Gao
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xinke Huang
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Zhendong Deng
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xiaoyu Wang
- Department of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
- Dr Xiaoyu Wang, Department of Stomatology, The First Affiliated Hospital of Jinan University, 613 West Huangpu Avenue, Guangzhou, Guangdong 510632, P.R. China, E-mail:
| | - Shaoqiang Lin
- Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510000, P.R. China
- School of Pharmaceutical Sciences of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
- Correspondence to: Dr Shaoqiang Lin, Integrated Traditional and Western Medicine Research Center of The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nonglinxia Road, Guangzhou, Guangdong 510000, P.R. China, E-mail:
| |
Collapse
|
112
|
Balmaña M, Diniz F, Feijão T, Barrias CC, Mereiter S, Reis CA. Analysis of the Effect of Increased α2,3-Sialylation on RTK Activation in MKN45 Gastric Cancer Spheroids Treated with Crizotinib. Int J Mol Sci 2020; 21:ijms21030722. [PMID: 31979110 PMCID: PMC7037121 DOI: 10.3390/ijms21030722] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/26/2022] Open
Abstract
In the scenario of personalized medicine, targeted therapies are currently the focus of cancer drug development. These drugs can block the growth and spread of tumor cells by interfering with key molecules involved in malignancy, such as receptor tyrosine kinases (RTKs). MET and Recepteur d'Origine Nantais (RON), which are RTKs frequently overactivated in gastric cancer, are glycoprotein receptors whose activation have been shown to be modulated by the cellular glycosylation. In this work, we address the role of sialylation in gastric cancer therapy using an innovative 3D high-throughput cell culture methodology that mimics better the in vivo tumor features. We evaluate the response to targeted treatment of glycoengineered gastric cancer cell models overexpressing the sialyltransferases ST3GAL4 or ST3GAL6 by subjecting 3D spheroids to the tyrosine kinase inhibitor crizotinib. We show here that 3D spheroids of ST3GAL4 or ST3GAL6 overexpressing MKN45 gastric cancer cells are less affected by the inhibitor. In addition, we disclose a potential compensatory pathway via activation of the Insulin Receptor upon crizotinib treatment. Our results suggest that cell sialylation, in addition of being involved in tumor progression, could play a critical role in the response to tyrosine kinase inhibitors in gastric cancer.
Collapse
Affiliation(s)
- Meritxell Balmaña
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Francisca Diniz
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar—ICBAS, University of Porto, 4050-313 Porto, Portugal
| | - Tália Feijão
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Cristina C. Barrias
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar—ICBAS, University of Porto, 4050-313 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, University of Porto, 4200-135 Porto, Portugal
| | - Stefan Mereiter
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Celso A. Reis
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- Institute of Biomedical Sciences of Abel Salazar—ICBAS, University of Porto, 4050-313 Porto, Portugal
- Medical Faculty, University of Porto, 4200-319 Porto, Portugal
- Correspondence: ; Tel.: +351-22-040-88-00 (ext. 6068)
| |
Collapse
|
113
|
Identification of the complete coding cDNAs and expression analysis of B4GALT1, LALBA, ST3GAL5, ST6GAL1 in the colostrum and milk of the Garganica and Maltese goat breeds to reveal possible implications for oligosaccharide biosynthesis. BMC Vet Res 2019; 15:457. [PMID: 31852463 PMCID: PMC6921551 DOI: 10.1186/s12917-019-2206-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Background Milk sialylated oligosaccharides (SOS) play crucial roles in many biological processes. The most abundant free SOS in goat’s milk are 3’sialyllactose (3′-SL), 6’sialyllactose (6′-SL) and disialyllactose (DSL). The production of these molecules is determined genetically by the expression of glycosyltransferases and by the availability of nucleotide sugar substrates, but the precise mechanisms regulating the differential patterns of milk oligosaccharides are not known. We aimed to identify the complete cDNAs of candidate genes implicated in SOS biosynthesis (B4GALT1, LALBA, ST3GAL5, ST6GAL1) and to analyse their expression during lactation in the Garganica and Maltese goat breeds. Moreover, we analysed the colostrum and milk contents of 3′-SL, 6′-SL and disialyllactose (DSL) and the possible correlations between expressed genes and SOS. Results We identified the complete coding cDNAs of B4GALT1 (HQ700335.1), ST3GAL5 (KF055858.2), and ST6GAL1 (HQ709167.1), the single nucleotide polymorphism (SNPs) of these genes and 2 splicing variants of the ST6GAL1 cDNA. RT-qPCR analysis showed that LALBA and ST6GAL1 were the genes with the highest and lowest expression in both breeds, respectively. The interaction effects of the breeds and sampling times were associated with higher levels of B4GALT1 and ST3GAL5 gene expression in Garganica than in Maltese goats at kidding. B4GALT1, LALBA, and ST3GAL5 gene expression changed from kidding to 60 and 120 days in Maltese goats, while in Garganica goats, a difference was observed only for the LALBA gene. Breed and lactation effects were also found for SOS contents. Positive correlations of B4GALT1, LALBA, ST3GAL5, and ST6GAL1 with 3′-SL/6′SL and DSL were found. Conclusions The genetic effect on the oligosaccharide content of milk was previously highlighted in bovines, and this study is the first to investigate this effect in two goat breeds (Garganica and Maltese) during lactation. The genetic variability of candidate genes involved in SOS biosynthesis highlights their potential role in affecting gene expression and ultimately biological function. The investigation of gene regulatory regions as well as the examination of other sialyltransferase genes will be needed to identify the genetic pattern leading to a higher SOS content in the autochtonous Garganica breed and to protect it using a focused breeding strategy.
Collapse
|
114
|
Vergé C, Bouchatal A, Chirat F, Guérardel Y, Maftah A, Petit JM. Involvement of ST6Gal I-mediated α2,6 sialylation in myoblast proliferation and differentiation. FEBS Open Bio 2019; 10:56-69. [PMID: 31622539 PMCID: PMC6943236 DOI: 10.1002/2211-5463.12745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/24/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
Myogenesis is a physiological process which involves the proliferation of myoblasts and their differentiation into multinucleated myotubes, which constitute the future muscle fibers. Commitment of myoblasts to differentiation is regulated by the balance between the myogenic factors Pax7 and MyoD. The formation of myotubes requires the presence of glycans, especially N‐glycans, on the cell surface. We examined here the involvement of α2,6 sialylation during murine myoblastic C2C12 cell differentiation by generating a st6gal1‐knockdown C2C12 cell line; these cells exhibit reduced proliferative potential and precocious differentiation due to the low expression of Pax7. The earlier fusion of st6gal1‐knockdown cells leads to a high fusion index and a drop in reserve cells (Pax7+/MyoD−). In st6gal1‐knockdown cells, the Notch pathway is inactivated; consequently, Pax7 expression is virtually abolished, leading to impairment of the proliferation rate. All these results indicate that the decrease in α2,6 sialylation of N‐glycans favors the differentiation of most cells and provokes a significant loss of reserve cells.
Collapse
Affiliation(s)
- Caroline Vergé
- PEIRENE, EA 7500, Glycosylation and Cell Differentiation, University of Limoges, France
| | - Amel Bouchatal
- PEIRENE, EA 7500, Glycosylation and Cell Differentiation, University of Limoges, France
| | - Frédéric Chirat
- UGSF, UMR 8576, CNRS, University of Lille, Villeneuve d'Ascq, France
| | - Yann Guérardel
- UGSF, UMR 8576, CNRS, University of Lille, Villeneuve d'Ascq, France
| | - Abderrahman Maftah
- PEIRENE, EA 7500, Glycosylation and Cell Differentiation, University of Limoges, France
| | - Jean-Michel Petit
- PEIRENE, EA 7500, Glycosylation and Cell Differentiation, University of Limoges, France
| |
Collapse
|
115
|
Natoni A, Bohara R, Pandit A, O'Dwyer M. Targeted Approaches to Inhibit Sialylation of Multiple Myeloma in the Bone Marrow Microenvironment. Front Bioeng Biotechnol 2019; 7:252. [PMID: 31637237 PMCID: PMC6787837 DOI: 10.3389/fbioe.2019.00252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/17/2019] [Indexed: 11/13/2022] Open
Abstract
Aberrant glycosylation modulates different aspects of tumor biology, and it has long been recognized as a hallmark of cancer. Among the different forms of glycosylation, sialylation, the addition of sialic acid to underlying oligosaccharides, is often dysregulated in cancer. Increased expression of sialylated glycans has been observed in many types of cancer, including multiple myeloma, and often correlates with aggressive metastatic behavior. Myeloma, a cancer of plasma cells, develops in the bone marrow, and colonizes multiple sites of the skeleton including the skull. In myeloma, the bone marrow represents an essential niche where the malignant cells are nurtured by the microenvironment and protected from chemotherapy. Here, we discuss the role of hypersialylation in the metastatic process focusing on multiple myeloma. In particular, we examine how increased sialylation modulates homing of malignant plasma cells into the bone marrow by regulating the activity of molecules important in bone marrow cellular trafficking including selectins and integrins. We also propose that inhibiting sialylation may represent a new therapeutic strategy to overcome bone marrow-mediated chemotherapy resistance and describe different targeted approaches to specifically deliver sialylation inhibitors to the bone marrow microenvironment.
Collapse
Affiliation(s)
- Alessandro Natoni
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| | - Raghvendra Bohara
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Michael O'Dwyer
- Apoptosis Research Centre, School of Medicine, National University of Ireland, Galway, Ireland
| |
Collapse
|
116
|
Huizing M, Yardeni T, Fuentes F, Malicdan MC, Leoyklang P, Volkov A, Dekel B, Brede E, Blake J, Powell A, Chatrathi H, Anikster Y, Carrillo N, Gahl WA, Kopp JB. Rationale and Design for a Phase 1 Study of N-Acetylmannosamine for Primary Glomerular Diseases. Kidney Int Rep 2019; 4:1454-1462. [PMID: 31701055 PMCID: PMC6829193 DOI: 10.1016/j.ekir.2019.06.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Sialic acids are important contributors to the polyanionic component of the glomerular filtration barrier, which regulates permeability selectivity. Pathologic glomerular hyposialylation, associated with podocyte effacement, has been implicated in human and mouse glomerulopathies. Oral treatment with N-acetylmannosamine (ManNAc), the uncharged precursor of sialic acid, ameliorates glomerular pathology in different models of glomerular disease. METHODS Here we explore the sialylation status of kidney biopsies obtained from 27 subjects with various glomerular diseases using lectin histochemistry. RESULTS We identified severe glomerular hyposialylation in 26% of the biopsies. These preliminary findings suggest that this condition may occur relatively frequently and may be a novel target for therapy. We describe the background, rationale, and design of a phase 1 study to test safety, tolerability, and pharmacokinetics of ManNAc in subjects with primary podocyte diseases. CONCLUSION We recently demonstrated that ManNAc was safe and well tolerated in a first-in-human phase 1 study in subjects with UDP-N-acetylglucosamine (GlcNAc) 2-epimerase/ManNAc kinase (GNE) myopathy, a disorder of impaired sialic acid synthesis. Using previous preclinical and clinical data, we propose to test ManNAc therapy for subjects with primary glomerular diseases. Even though the exact mechanisms, affected cell types, and pathologic consequences of glomerular hyposialylation need further study, treatment with this physiological monosaccharide could potentially replace or supplement existing glomerular diseases therapies.
Collapse
Affiliation(s)
- Marjan Huizing
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tal Yardeni
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Federico Fuentes
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - May C.V. Malicdan
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Petcharat Leoyklang
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexander Volkov
- Pediatric Nephrology Unit and Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Benjamin Dekel
- Pediatric Nephrology Unit and Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Emily Brede
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jodi Blake
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Alva Powell
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Harish Chatrathi
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Yair Anikster
- Metabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nuria Carrillo
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - William A. Gahl
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey B. Kopp
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
117
|
Chung CY, Wang Q, Yang S, Chough S, Seo Y, Cipollo JF, Balthasar JP, Betenbaugh MJ. The impact of sialylation linkage-type on the pharmacokinetics of recombinant butyrylcholinesterases. Biotechnol Bioeng 2019; 117:157-166. [PMID: 31544955 DOI: 10.1002/bit.27174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 09/07/2019] [Accepted: 09/13/2019] [Indexed: 11/10/2022]
Abstract
Chinese hamster ovary (CHO) cells typically produce glycoproteins with N-glycans terminating in α-2,3 sialylation. Human cells produce glycoproteins that include α-2,3 and α-2,6 sialic acids. To examine the impact of altering protein sialylation on pharmacokinetic properties, recombinant human butyrylcholinesterase (BChE) was produced in CHO cells by knocking out the α-2,3 sialyltransferase genes followed by overexpression of the α-2,6 sialyltransferase (26BChE) enzyme. The N-glycan composition of 26BChE was compared to BChE with α-2,3 sialylation (23BChE) derived from wild-type CHO cells. Both 23BChE and 26BChE exhibited comparable antennarity distributions with bi-antennary di-sialylated glycans representing the most abundant glycoform. CD-1 mice were intravenously injected with the 23BChE or 26BChE, and residual BChE activities from blood collected at various time points for pharmacokinetic analyses. Although 23BChE contained a slightly lower initial sialylation level compared to 26BChE, the molecule exhibited higher residual activity between 5 and 24 hr postinjection. Pharmacokinetic analyses indicated that 23BChE exhibited an increase in area under the curve and a lower volume of distribution at steady state than that of 26BChE. These findings suggest that the type of sialylation linkage may play a significant role in the pharmacokinetic behavior of a biotherapeutic when tested in in vivo animal models.
Collapse
Affiliation(s)
- Cheng-Yu Chung
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Qiong Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Shuang Yang
- Laboratory for Bacterial Polysaccharides, Division of Bacterial, Parasitic and Allergenic Products (DBPAP), Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Sandra Chough
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Younji Seo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - John F Cipollo
- Laboratory for Bacterial Polysaccharides, Division of Bacterial, Parasitic and Allergenic Products (DBPAP), Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
118
|
Computational analysis of the structure, glycosylation and CMP binding of human ST3GAL sialyltransferases. Carbohydr Res 2019; 486:107823. [PMID: 31557542 DOI: 10.1016/j.carres.2019.107823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 11/24/2022]
Abstract
Sialyltransferases (STs) are the fundamental enzymes which are related to many biological processes such as cell signalling, cellular recognition, cell-cell and host-pathogen interactions and metastasis of cancer. All STs catalyse the terminal sialic acid addition from CMP donor to the glycan units. ST3GAL family is one of the most important STs and divided into the six subfamily in mouse and humans which are ST3Gal I, ST3Gal II, ST3Gal III, ST3Gal IV, ST3Gal V, and ST3Gal VI. The members of the ST3GAL family transfer sialic acid to the terminal galactose residues of glycochains through an α2,3-linkage. There are many reports on the ST3GAL function in mammals but, there is a paucity of information about structure of human ST3GAL family. Herein, we investigated the structure, glycosylation and CMP binding site of human ST3GAL family using computational methods. We found for the first time N-glycosylation positions in ST3Gal IV and VI, mucin type glycosylation in ST3Gal III and O-GlcNAcylation in ST3Gal V and their relation with sialylmotifs. In addition, we predicted CMP binding positions of human ST3GAL enzyme family on three-dimensional structure using molecular docking and first demonstrated the sialylmotifs relation with the CMP binding positions in ST3Gal III-VI subfamilies.
Collapse
|
119
|
Lin CY, Lai HL, Chen HM, Siew JJ, Hsiao CT, Chang HC, Liao KS, Tsai SC, Wu CY, Kitajima K, Sato C, Khoo KH, Chern Y. Functional roles of ST8SIA3-mediated sialylation of striatal dopamine D 2 and adenosine A 2A receptors. Transl Psychiatry 2019; 9:209. [PMID: 31455764 PMCID: PMC6712005 DOI: 10.1038/s41398-019-0529-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 05/22/2019] [Accepted: 06/20/2019] [Indexed: 01/20/2023] Open
Abstract
Sialic acids are typically added to the end of glycoconjugates by sialyltransferases. Among the six ST8 α-N-acetyl-neuraminide α-2,8-sialyltransferases (ST8SIA) existing in adult brains, ST8SIA2 is a schizophrenia-associated gene. However, the in vivo substrates and physiological functions of most sialyltransferases are currently unknown. The ST8SIA3 is enriched in the striatum. Here, we showed that ablation of St8sia3 in mice (St8sia3-KO) led to fewer disialylated and trisialylated terminal glycotopes in the striatum of St8sia3-KO mice. Moreover, the apparent sizes of several striatum-enriched G-protein-coupled receptors (GPCRs) (including the adenosine A2A receptor (A2AR) and dopamine D1/D2 receptors (D1R and D2R)) were smaller in St8sia3-KO mice than in WT mice. A sialidase treatment removed the differences in the sizes of these molecules between St8sia3-KO and WT mice, confirming the involvement of sialylation. Expression of ST8SIA3 in the striatum of St8sia3-KO mice using adeno-associated viruses normalized the sizes of these proteins, demonstrating a direct role of ST8SIA3. The lack of ST8SIA3-mediated sialylation altered the distribution of these proteins in lipid rafts and the interaction between D2R and A2AR. Locomotor activity assays revealed altered pharmacological responses of St8sia3-KO mice to drugs targeting these receptors and verified that a greater population of D2R formed heteromers with A2AR in the striatum of St8sia3-KO mice. Since the A2AR-D2R heteromer is an important drug target for several basal ganglia diseases (such as schizophrenia and Parkinson's disease), the present study not only reveals a crucial role for ST8SIA3 in striatal functions but also provides a new drug target for basal ganglia-related diseases.
Collapse
Affiliation(s)
- Chien-Yu Lin
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsing-Lin Lai
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hui-Mei Chen
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jian-Jing Siew
- 0000 0001 2287 1366grid.28665.3fInstitute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan ,0000 0001 0425 5914grid.260770.4Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Cheng-Te Hsiao
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hua-Chien Chang
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Kuo-Shiang Liao
- 0000 0001 2287 1366grid.28665.3fGenomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shih-Chieh Tsai
- grid.36020.37Department of Research and Development, National Laboratory Animal Center, National Applied Research Laboratories, Taipei and Tainan, Taipei, Taiwan
| | - Chung-Yi Wu
- 0000 0001 2287 1366grid.28665.3fGenomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Ken Kitajima
- 0000 0001 0943 978Xgrid.27476.30Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-860 Japan
| | - Chihiro Sato
- 0000 0001 0943 978Xgrid.27476.30Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-860 Japan
| | - Kay-Hooi Khoo
- 0000 0001 2287 1366grid.28665.3fInstitute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
120
|
Emerging structural insights into glycosyltransferase-mediated synthesis of glycans. Nat Chem Biol 2019; 15:853-864. [PMID: 31427814 DOI: 10.1038/s41589-019-0350-2] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/17/2019] [Indexed: 12/27/2022]
Abstract
Glycans linked to proteins and lipids play key roles in biology; thus, accurate replication of cellular glycans is crucial for maintaining function following cell division. The fact that glycans are not copied from genomic templates suggests that fidelity is provided by the catalytic templates of glycosyltransferases that accurately add sugars to specific locations on growing oligosaccharides. To form new glycosidic bonds, glycosyltransferases bind acceptor substrates and orient a specific hydroxyl group, frequently one of many, for attack of the donor sugar anomeric carbon. Several recent crystal structures of glycosyltransferases with bound acceptor substrates reveal that these enzymes have common core structures that function as scaffolds upon which variable loops are inserted to confer substrate specificity and correctly orient the nucleophilic hydroxyl group. The varied approaches for acceptor binding site assembly suggest an ongoing evolution of these loop regions provides templates for assembly of the diverse glycan structures observed in biology.
Collapse
|
121
|
Janesch B, Saxena H, Sim L, Wakarchuk WW. Comparison of α2,6-sialyltransferases for sialylation of therapeutic proteins. Glycobiology 2019; 29:735-747. [DOI: 10.1093/glycob/cwz050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 11/13/2022] Open
Abstract
AbstractThe development of therapeutic proteins for the treatment of numerous diseases is one of the fastest growing areas of biotechnology. Therapeutic efficacy and serum half-life are particularly important, and these properties rely heavily on the glycosylation state of the protein. Expression systems to produce authentically fully glycosylated therapeutic proteins with appropriate terminal sialic acids are not yet perfected. The in vitro modification of therapeutic proteins by recombinant sialyltransferases offers a promising and elegant strategy to overcome this problem. Thus, the detailed expression and characterization of sialyltransferases for completion of the glycan chains is of great interest to the community. We identified a novel α2,6-sialyltransferase from Helicobacter cetorum and compared it to the human ST6Gal1 and a Photobacterium sp. sialyltransferase using glycoprotein substrates in a 96-well microtiter-plate-based assay. We demonstrated that the recombinant α2,6-sialyltransferase from H. cetorum is an excellent catalyst for modification of N-linked glycans of different therapeutic proteins.
Collapse
Affiliation(s)
- Bettina Janesch
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
- Department of NanoBiotechnology, Institute for Biologically Inspired Materials, NanoGlycobiology Unit, Universität für Bodenkultur Wien, Muthgasse 11, A-1190 Vienna, Austria
| | - Hirak Saxena
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Lyann Sim
- Departments of Chemistry and Biochemistry and Michael Smith Laboratory, University of British Columbia, Vancouver, BC V6T1Z1, Canada
| | - Warren W Wakarchuk
- Department of Chemistry and Biology, Ryerson University, Toronto, ON M5B 2K3, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| |
Collapse
|
122
|
Sosa EA, Moriyama Y, Ding Y, Tejeda-Muñoz N, Colozza G, De Robertis EM. Transcriptome analysis of regeneration during Xenopus laevis experimental twinning. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2019; 63:301-309. [PMID: 31250914 DOI: 10.1387/ijdb.190006ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Animal embryos have the remarkable property of self-organization. Over 125 years ago, Hans Driesch separated the two blastomeres of sea urchin embryos and obtained twins, in what was the foundation of experimental embryology. Since then, embryonic twinning has been obtained experimentally in many animals. In a recent study, we developed bisection methods that generate identical twins reliably from Xenopus blastula embryos. In the present study, we have investigated the transcriptome of regenerating half-embryos after sagittal and dorsal-ventral (D-V) bisections. Individual embryos were operated at midblastula (stage 8) with an eyelash hair and cultured until early gastrula (stage 10.5) or late gastrula (stage 12) and the transcriptome of both halves were analyzed by RNA-seq. Since many genes are activated by wound healing in Xenopus embryos, we resorted to stringent sequence analyses and identified genes up-regulated in identical twins but not in either dorsal or ventral fragments. At early gastrula, cell division-related transcripts such as histones were elevated, whereas at late gastrula, pluripotency genes (such as sox2) and germ layer determination genes (such as eomesodermin, ripply2 and activin receptor ACVRI) were identified. Among the down-regulated transcripts, sizzled, a regulator of Chordin stability, was prominent. These findings are consistent with a model in which cell division is required to heal damage, while maintaining pluripotency to allow formation of the organizer with a displacement of 90 0 from its original site. The extensive transcriptomic data presented here provides a valuable resource for data mining of gene expression during early vertebrate development.
Collapse
Affiliation(s)
- Eric A Sosa
- Howard Hughes Medical Institute, Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
123
|
Matana A, Popović M, Boutin T, Torlak V, Brdar D, Gunjača I, Kolčić I, Boraska Perica V, Punda A, Rudan I, Polašek O, Barbalić M, Hayward C, Zemunik T. Genetic Variants in the ST6GAL1 Gene Are Associated with Thyroglobulin Plasma Level in Healthy Individuals. Thyroid 2019; 29:886-893. [PMID: 30929638 DOI: 10.1089/thy.2018.0661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background: Thyroglobulin (Tg) is a 660 kDa iodoglycoprotein that serves as a scaffold for thyroid hormone synthesis. Although a twin study showed that variability of serum Tg levels has a substantial genetic basis, no genome-wide association study (GWAS) of serum/plasma Tg levels has been performed to date. The aim of this study was to identify genetic variants associated with plasma Tg levels among healthy individuals. Methods: A GWAS was conducted on two Croatian cohorts, and a combined analysis was performed. The analyses included 1094 individuals. A total of 7,597,379 variants, imputed using the 1000 Genomes reference panel, were analyzed for association. GWAS was performed under an additive model, controlling for age, sex, and relatedness within each data set. Combined analysis was conducted using the inverse-variance fixed-effects method. Results: Sixteen variants located on chromosome 3, within the ST6GAL1 gene, reached genome-wide significance. The lead SNP was rs4012172 ( \documentclass{aastex}\usepackage{amsbsy}\usepackage{amsfonts}\usepackage{amssymb}\usepackage{bm}\usepackage{mathrsfs}\usepackage{pifont}\usepackage{stmaryrd}\usepackage{textcomp}\usepackage{portland, xspace}\usepackage{amsmath, amsxtra}\usepackage{upgreek}\pagestyle{empty}\DeclareMathSizes{10}{9}{7}{6}\begin{document} $$p = 1.29 \times {10^{ - 10}}$$ \end{document} ), which explained 3.19% of the variance in Tg levels. ST6GAL1 belongs to the sialyltransferase protein family, which has a fundamental role in the synthesis of specific sialylated structures on various glycoproteins, including Tg. It is known that only immature Tg (poorly sialylated or desialylated) can be transferred to the bloodstream. Conclusions: A highly biologically plausible locus was identified that could have a role in the regulation of plasma Tg levels in healthy individuals.
Collapse
Affiliation(s)
- Antonela Matana
- 1 Department of Medical Biology; School of Medicine; University of Split, Split, Croatia
| | - Marijana Popović
- 1 Department of Medical Biology; School of Medicine; University of Split, Split, Croatia
| | - Thibaud Boutin
- 2 MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine; University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Vesela Torlak
- 3 Department of Nuclear Medicine, University Hospital Split, Split, Croatia
| | - Dubravka Brdar
- 3 Department of Nuclear Medicine, University Hospital Split, Split, Croatia
| | - Ivana Gunjača
- 1 Department of Medical Biology; School of Medicine; University of Split, Split, Croatia
| | - Ivana Kolčić
- 4 Department of Public Health, School of Medicine; University of Split, Split, Croatia
| | - Vesna Boraska Perica
- 1 Department of Medical Biology; School of Medicine; University of Split, Split, Croatia
| | - Ante Punda
- 3 Department of Nuclear Medicine, University Hospital Split, Split, Croatia
| | - Igor Rudan
- 5 Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics; University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Ozren Polašek
- 4 Department of Public Health, School of Medicine; University of Split, Split, Croatia
| | - Maja Barbalić
- 1 Department of Medical Biology; School of Medicine; University of Split, Split, Croatia
| | - Caroline Hayward
- 2 MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine; University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Tatijana Zemunik
- 1 Department of Medical Biology; School of Medicine; University of Split, Split, Croatia
| |
Collapse
|
124
|
Song HW, Yoo G, Bong JH, Kang MJ, Lee SS, Pyun JC. Surface display of sialyltransferase on the outer membrane of Escherichia coli and ClearColi. Enzyme Microb Technol 2019; 128:1-8. [PMID: 31186105 DOI: 10.1016/j.enzmictec.2019.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 01/03/2023]
Abstract
α2,3-Sialyltransferase from Pasteurella multocida (PmST1) is an enzyme that transfers a sialyl group of donor substrates to an acceptor substrate called N-acetyl-d-lactosamine (LacNAc). In this study PmST1 was expressed on the outer membrane of wildtype Escherichia coli (BL21) with lipopolysaccharide (LPS) and ClearColi with no LPS, and then the enzyme activity and expression level of PmST1 were compared. As the first step, the expression levels of PmST1 on the outer membranes of wildtype E. coli (BL21) and ClearColi were compared according to the IPTG induction time, and the absolute amount of surface-displayed PmST1 was calculated using densitometry of SDS-PAGE. As the next step, the influence of LPS on the PmST1 activity was estimated by analyzing Michaelis-Menten plot. The enzyme activity of PmST1 was analyzed by measuring the concentration of CMP, which was a by-product after the transfer of the sialyl group of donor compounds to the acceptor compounds. From a Michaelis-Menten plot, the enzyme activity of the surface-displayed PmST1 and the maximum rate (Vmax) of ClearColi were higher than those of wildtype E. coli (BL21). However, the KM value, which represented the concentration of substrate to reach half the maximum rate (Vmax), was similar for both enzymes. These results represented such a difference in enzyme activity was occurred from the interference of LPS on the mass transport of the donor and acceptor to PmST1 for the sialyl group transfer.
Collapse
Affiliation(s)
- Hyun-Woo Song
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seo-dae-mun-gu, Seoul, 03722, Republic of Korea
| | - Gu Yoo
- School of Chemistry & Institute for Life Sciences, FNES, University of Southampton, Highfield, Southampton, SO17 1BJ, UK
| | - Ji-Hong Bong
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seo-dae-mun-gu, Seoul, 03722, Republic of Korea
| | - Min-Jung Kang
- Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Seung Seo Lee
- School of Chemistry & Institute for Life Sciences, FNES, University of Southampton, Highfield, Southampton, SO17 1BJ, UK.
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-ro, Seo-dae-mun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
125
|
Wang H, Liu L, Cao Q, Mao W, Zhang Y, Qu X, Cai X, Lv Y, Chen H, Xu X, Wang X. Haemophilus parasuis α-2,3-sialyltransferase-mediated lipooligosaccharide sialylation contributes to bacterial pathogenicity. Virulence 2019; 9:1247-1262. [PMID: 30036124 PMCID: PMC6104685 DOI: 10.1080/21505594.2018.1502606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bacterial lipooligosaccharide (LOS) is an important virulence-associated factor, and its sialylation largely confers its ability to mediate cell adhesion, invasion, inflammation, and immune evasion. Here, we investigated the function of the Haemophilus parasuis α-2,3-sialyltransferase gene, lsgB, which determines the terminal sialylation of LOS, by generating a lsgB deletion mutant as well as a complementation strain. Our data indicate a direct effect of lsgB on LOS sialylation and reveal important roles of lsgB in promoting the pathogenicity of H. parasuis, including adhesion to and invasion of porcine cells in vitro, bacterial load and survival in vivo, as well as a contribution to serum resistance. These observations highlight the function of lsgB in mediating LOS sialylation and more importantly its role in H. parasuis infection. These findings provide a more profound understanding of the pathogenic mechanism of this disease-causing bacterium.
Collapse
Affiliation(s)
- Huan Wang
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China
| | - Lu Liu
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China
| | - Qi Cao
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China
| | - Weiting Mao
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China
| | - Yage Zhang
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China
| | - Xinyi Qu
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China
| | - Xuwang Cai
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China.,c Key Laboratory of Development of Veterinary Diagnostic Products , Ministry of Agriculture of the People's Republic of China , Wuhan , China
| | - Yujin Lv
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China.,d College of Veterinary Medicine , Henan University of Animal Husbandry and Economy , Zhengzhou , China
| | - Huanchun Chen
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China.,c Key Laboratory of Development of Veterinary Diagnostic Products , Ministry of Agriculture of the People's Republic of China , Wuhan , China
| | - Xiaojuan Xu
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China.,c Key Laboratory of Development of Veterinary Diagnostic Products , Ministry of Agriculture of the People's Republic of China , Wuhan , China
| | - Xiangru Wang
- a State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine , Huazhong Agricultural University , Wuhan , China.,b Key Laboratory of Preventive Veterinary Medicine in Hubei Province , The Cooperative Innovation Center for Sustainable Pig Production , Wuhan , China.,c Key Laboratory of Development of Veterinary Diagnostic Products , Ministry of Agriculture of the People's Republic of China , Wuhan , China
| |
Collapse
|
126
|
Szczubiał M, Wawrzykowski J, Dąbrowski R, Bochniarz M, Brodzki P, Kankofer M. The effect of pyometra on glycosylation of proteins in the uterine tissues from female dogs. Theriogenology 2019; 131:41-46. [PMID: 30939355 DOI: 10.1016/j.theriogenology.2019.03.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/20/2019] [Accepted: 03/24/2019] [Indexed: 12/26/2022]
Abstract
The main aim of this study was to investigate the effect of pyometra on glycosylation of proteins in the uterine tissues from female dogs, using western blotting with selected lectins (Sambucus nigra agglutinin - SNA and Maackia amurensis agglutinin - MAL II). In addition protein pattern of examined tissues was also evaluated. The study was performed on 10 female dogs undergoing ovariohysterectomy because of pyometra and 10 clinically healthy female dogs, undergoing elective spaying (ovariohysterectomy). Uterine tissue samples of 1 cm2 were taken from the middle region of each uterine horn in both group of animals immediately after ovariohysterectomy. Tissue samples were homogenized and analysed by sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting with SNA and MAL II. SDS-PAGE analysis showed differences between pyometra samples and controls in the amount of obtained protein fractions and the protein content in the individual fractions. Five protein (with a molecular weight of 193.78 kDa, 103.18 kDa, 77.67 kDa, 70.39 kDa, and 53.00 kDa) were found only in the pyometra samples. The remaining fractions differed in intensity of staining, which indicated differ abundance of a given protein. The results of western blotting with SNA and MAL II demonstrated that the pattern obtained from densitometric analysis differs between adequate healthy and pyometra samples with regard to the amount of protein fraction obtained as well as the intensity of staining of particular fraction. The pyometra tissues contained seven SNA-binding proteins (with a molecular weight 189.94 kDa, 165.51 kDa, 100.94 kDa, 59.42 KDa, 41.32 kDa, 35.16 kDa, and 32.6 kDa) that were not in the healthy tissues. Of the nine remaining fractions, six showed significantly higher (P < 0.05) intensity of staining in the healthy uterine tissues. In turn, the MAL II-binding protein with a molecular weight 75.85 kDa, 51.12 kDa, and 49.98 kDa were found only in the pyometra samples. Of the 28 remaining fractions, ten demonstrated significantly higher (P < 0.05), and five fractions had significantly lower (P < 0.05) intensity of staining in the pyometra tissues. The results obtained indicate that proteins in uterine tissues from female dogs with pyometra are differently glycosylated compared to normal uterine tissues. These findings provide the basis for further studies of the possible role of glycosylation in the pathogenesis of canine pyometra.
Collapse
Affiliation(s)
- Marek Szczubiał
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30, 20-612, Lublin, Poland.
| | - Jacek Wawrzykowski
- Department of Animal Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka 12, 20-033, Lublin, Poland
| | - Roman Dąbrowski
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30, 20-612, Lublin, Poland
| | - Mariola Bochniarz
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30, 20-612, Lublin, Poland
| | - Piotr Brodzki
- Department and Clinic of Animal Reproduction, Faculty of Veterinary Medicine, University of Life Sciences, Głeboka 30, 20-612, Lublin, Poland
| | - Marta Kankofer
- Department of Animal Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka 12, 20-033, Lublin, Poland
| |
Collapse
|
127
|
Chinoy ZS, Bodineau C, Favre C, Moremen KW, Durán RV, Friscourt F. Selective Engineering of Linkage-Specific α2,6-N-Linked Sialoproteins Using Sydnone-Modified Sialic Acid Bioorthogonal Reporters. Angew Chem Int Ed Engl 2019; 58:4281-4285. [PMID: 30706985 PMCID: PMC6450558 DOI: 10.1002/anie.201814266] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/12/2019] [Indexed: 02/02/2023]
Abstract
The metabolic oligosaccharide engineering (MOE) strategy using unnatural sialic acids has recently enabled the visualization of the sialome in living systems. However, MOE only reports on global sialylation and dissected information regarding subsets of sialosides is missing. Described here is the synthesis and utilization of sialic acids modified with a sydnone reporter for the metabolic labeling of sialoconjugates. The positioning of the reporter on the sugar significantly altered its metabolic fate. Further in vitro enzymatic assays revealed that the 9-modified neuraminic acid is preferentially accepted by the sialyltransferase ST6Gal-I over ST3Gal-IV, leading to the favored incorporation of the reporter into linkage-specific α2,6-N-linked sialoproteins. This sydnone sugar presents the possibility of investigating the roles of specific sialosides.
Collapse
Affiliation(s)
- Zoeisha S. Chinoy
- Institut Européen de Chimie et Biologie, Université de Bordeaux, 2 rue Robert Escarpit, 33607 Pessac, France
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine, CNRS UMR5287, Bordeaux, France
| | - Clément Bodineau
- Institut Européen de Chimie et Biologie, Université de Bordeaux, 2 rue Robert Escarpit, 33607 Pessac, France
- Institut Bergonié, INSERM U1218, Bordeaux, France
| | - Camille Favre
- Institut Européen de Chimie et Biologie, Université de Bordeaux, 2 rue Robert Escarpit, 33607 Pessac, France
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine, CNRS UMR5287, Bordeaux, France
| | - Kelley W. Moremen
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA USA
| | - Raúl V. Durán
- Institut Européen de Chimie et Biologie, Université de Bordeaux, 2 rue Robert Escarpit, 33607 Pessac, France
- Institut Bergonié, INSERM U1218, Bordeaux, France
- Current address: Centro Andaluz de Biología Molecular y Medicina Regenerativa, Consejo Superior de Investigaciones Científicas - Universidad de Sevilla - Universidad Pablo de Olavide, Avda. Américo Vespucio 24, 41092 Seville, Spain
| | - Frédéric Friscourt
- Institut Européen de Chimie et Biologie, Université de Bordeaux, 2 rue Robert Escarpit, 33607 Pessac, France
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine, CNRS UMR5287, Bordeaux, France
| |
Collapse
|
128
|
Liou LB, Jang SS. α-2,3-Sialyltransferase 1 and neuraminidase-3 from monocytes in patients with rheumatoid arthritis correlate with disease activity measures: A pilot study. J Chin Med Assoc 2019; 82:179-185. [PMID: 30913115 DOI: 10.1097/jcma.0000000000000027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND We decided to study the association of monocyte α-2,3-sialyltransferase 1 (ST3Gal-1), neuraminidase-3 (Neu3), α-2,6-sialyltransferase 1 (ST6Gal-1), and neuraminidase-1 (Neu1) levels with disease activity score 28 (DAS28) in human rheumatoid arthritis (RA), considering that mouse monocytes' sialic acid (SIA) levels relate to their phagocytosis and IgG binding ability. METHODS ST3Gal-1, Neu3, ST6Gal-1, Neu1, α-2,3-SIA, and α-2,6-SIA levels on RA peripheral blood monocytes, T cells, and polymorphonuclear cells were determined by using fluorochrome-conjugated anti-cell-specific marker antibodies and fluorochrome-conjugated anti-enzyme antibodies. Simple correlation and linear regression were used to correlate enzyme levels with DAS28. RESULTS RA monocyte ST3Gal-1 and Neu3 levels correlated with DAS28 in patients having DAS28 >5.1 (r = 0.469, p = 0.002; r = 0.410, p = 0.006, respectively). When multivariable analysis was performed for erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and SIA-related enzyme levels in different cell types as independent variables with DAS28 as a dependent variable, monocyte ST3Gal-1 levels correlated with DAS28 (p = 0.009) but not ESR and CRP in patients having DAS28 >5.1 (both p ≥ 0.292). RA monocyte ST3Gal-1 levels correlated with DAS28 (p = 0.010) and with ESR (p < 0.001) at month 0 when applied to all RA patients including both remission and nonremission groups in multivariable analysis. The latter findings persisted longitudinally at month 3. CONCLUSION Monocyte ST3Gal-1 and Neu3 levels correlated longitudinally with DAS28 by two different methods suggest that monocyte ST3Gal-1 and Neu3 levels may be used as biomarkers to monitor RA disease activity.
Collapse
Affiliation(s)
- Lieh-Bang Liou
- Division of Rheumatology, Allergy, and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, ROC
- Department of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan, ROC
| | - Shr-Shian Jang
- Division of Rheumatology, Allergy, and Immunology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, ROC
| |
Collapse
|
129
|
Selective Engineering of Linkage‐Specific α2,6‐
N
‐Linked Sialoproteins Using Sydnone‐Modified Sialic Acid Bioorthogonal Reporters. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201814266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
130
|
Li J, He J, Zhang C, Chen J, Mao W, Yu C. Dual-type responsive electrochemical biosensor for the detection of α2,6-sialylated glycans based on AuNRs-SA coupled with c-SWCNHs/S-PtNC nanocomposites signal amplification. Biosens Bioelectron 2019; 130:166-173. [PMID: 30735949 DOI: 10.1016/j.bios.2019.01.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/19/2019] [Accepted: 01/22/2019] [Indexed: 02/08/2023]
Abstract
In this study, a dual-type responsive electrochemical biosensor was developed for the quantitative detection of α2,6-sialylated glycans (α2,6-sial-Gs), a potential biomarker of tumors. The gold nanorods (AuNRs), which exhibited great specific surface area, as well as good biocompatibility, was synthesized by the way of seed growth method. Furthermore, a biotin-streptavidin (biotin-SA) system was introduced to improve the immunoreaction efficiency. Accordingly, a label-free biosensor was fabricated based on AuNRs-SA for the quick detection of α2,6-sial-Gs by recording the signal of differential pulse voltammetry (DPV). Furthermore, to expand the ultrasensitive detection of α2,6-sial-Gs, a carboxylated single-walled carbon nanohorns/sulfur-doped platinum nanocluster (c-SWCNHs/S-PtNC) was synthesized for the first time as a novel signal label, which showed an excellent catalytic performance. The usage of c-SWCNHs/S-PtNC could significantly amplify the electrochemical signal recorded by the amperometric i-t curve. Herein, a sandwich type biosensor was constructed by combining the AuNRs-SA on the electrode and c-SWCNHs/S-PtNC (signal amplifier). The label-free biosensor possessed a linear range from 5 ng mL-1 to 5 μg mL-1 with a detection limit of 0.50 ng mL-1, and the sandwich-type biosensor possessed a wide linear range from 1 fg mL-1 to 100 ng mL-1 with a detection limit of 0.69 fg mL-1. Furthermore, the biosensor exhibited excellent recovery and stability, indicating its potential for use in actual samples.
Collapse
Affiliation(s)
- Jia Li
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Junlin He
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Chengli Zhang
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Jun Chen
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Weiran Mao
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China
| | - Chao Yu
- College of Pharmacy, Institute of Life Science and School of Public Health, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
131
|
Novel Zebrafish Mono-α2,8-sialyltransferase (ST8Sia VIII): An Evolutionary Perspective of α2,8-Sialylation. Int J Mol Sci 2019; 20:ijms20030622. [PMID: 30709055 PMCID: PMC6387029 DOI: 10.3390/ijms20030622] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/28/2022] Open
Abstract
The mammalian mono-α2,8-sialyltransferase ST8Sia VI has been shown to catalyze the transfer of a unique sialic acid residues onto core 1 O-glycans leading to the formation of di-sialylated O-glycosylproteins and to a lesser extent to diSia motifs onto glycolipids like GD1a. Previous studies also reported the identification of an orthologue of the ST8SIA6 gene in the zebrafish genome. Trying to get insights into the biosynthesis and function of the oligo-sialylated glycoproteins during zebrafish development, we cloned and studied this fish α2,8-sialyltransferase homologue. In situ hybridization experiments demonstrate that expression of this gene is always detectable during zebrafish development both in the central nervous system and in non-neuronal tissues. Intriguingly, using biochemical approaches and the newly developed in vitro MicroPlate Sialyltransferase Assay (MPSA), we found that the zebrafish recombinant enzyme does not synthetize diSia motifs on glycoproteins or glycolipids as the human homologue does. Using comparative genomics and molecular phylogeny approaches, we show in this work that the human ST8Sia VI orthologue has disappeared in the ray-finned fish and that the homologue described in fish correspond to a new subfamily of α2,8-sialyltransferase named ST8Sia VIII that was not maintained in Chondrichtyes and Sarcopterygii.
Collapse
|
132
|
Wen XY, Tarailo-Graovac M, Brand-Arzamendi K, Willems A, Rakic B, Huijben K, Da Silva A, Pan X, El-Rass S, Ng R, Selby K, Philip AM, Yun J, Ye XC, Ross CJ, Lehman AM, Zijlstra F, Abu Bakar N, Drögemöller B, Moreland J, Wasserman WW, Vallance H, van Scherpenzeel M, Karbassi F, Hoskings M, Engelke U, de Brouwer A, Wevers RA, Pshezhetsky AV, van Karnebeek CD, Lefeber DJ. Sialic acid catabolism by N-acetylneuraminate pyruvate lyase is essential for muscle function. JCI Insight 2018; 3:122373. [PMID: 30568043 DOI: 10.1172/jci.insight.122373] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/14/2018] [Indexed: 11/17/2022] Open
Abstract
Sialic acids are important components of glycoproteins and glycolipids essential for cellular communication, infection, and metastasis. The importance of sialic acid biosynthesis in human physiology is well illustrated by the severe metabolic disorders in this pathway. However, the biological role of sialic acid catabolism in humans remains unclear. Here, we present evidence that sialic acid catabolism is important for heart and skeletal muscle function and development in humans and zebrafish. In two siblings, presenting with sialuria, exercise intolerance/muscle wasting, and cardiac symptoms in the brother, compound heterozygous mutations [chr1:182775324C>T (c.187C>T; p.Arg63Cys) and chr1:182772897A>G (c.133A>G; p.Asn45Asp)] were found in the N-acetylneuraminate pyruvate lyase gene (NPL). In vitro, NPL activity and sialic acid catabolism were affected, with a cell-type-specific reduction of N-acetyl mannosamine (ManNAc). A knockdown of NPL in zebrafish resulted in severe skeletal myopathy and cardiac edema, mimicking the human phenotype. The phenotype was rescued by expression of wild-type human NPL but not by the p.Arg63Cys or p.Asn45Asp mutants. Importantly, the myopathy phenotype in zebrafish embryos was rescued by treatment with the catabolic products of NPL: N-acetyl glucosamine (GlcNAc) and ManNAc; the latter also rescuing the cardiac phenotype. In conclusion, we provide the first report to our knowledge of a human defect in sialic acid catabolism, which implicates an important role of the sialic acid catabolic pathway in mammalian muscle physiology, and suggests opportunities for monosaccharide replacement therapy in human patients.
Collapse
Affiliation(s)
- Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Maja Tarailo-Graovac
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada.,Institute of Physiology and Biochemistry, Faculty of Biology, The University of Belgrade, Belgrade, Serbia.,Departments of Biochemistry, Molecular Biology, and Medical Genetics, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Koroboshka Brand-Arzamendi
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Anke Willems
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bojana Rakic
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Karin Huijben
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Afitz Da Silva
- Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Xuefang Pan
- Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Suzan El-Rass
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Robin Ng
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Katheryn Selby
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Anju Mary Philip
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Junghwa Yun
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - X Cynthia Ye
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Colin J Ross
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Anna M Lehman
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Fokje Zijlstra
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - N Abu Bakar
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Britt Drögemöller
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver Canada
| | - Jacqueline Moreland
- Departments of Biochemistry, Molecular Biology, and Medical Genetics, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Wyeth W Wasserman
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada.,Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Hilary Vallance
- Department of Medical Genetics and Department of Pathology and Laboratory Sciences, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, Canada
| | - Monique van Scherpenzeel
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, Netherlands.,Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Farhad Karbassi
- Zebrafish Centre for Advanced Drug Discovery and Keenan Research Centre for Biomedical science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, Physiology, Laboratory Medicine and Pathobiology and Institute of Medical Science, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Martin Hoskings
- Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver Canada
| | - Udo Engelke
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Arjan de Brouwer
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alexey V Pshezhetsky
- Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Clara Dm van Karnebeek
- Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada.,Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver Canada.,Departments of Pediatrics and Clinical Genetics, Emma Children's Hospital, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Dirk J Lefeber
- Department of Neurology, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, Nijmegen, Netherlands.,Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
133
|
Giovannone N, Antonopoulos A, Liang J, Geddes Sweeney J, Kudelka MR, King SL, Lee GS, Cummings RD, Dell A, Barthel SR, Widlund HR, Haslam SM, Dimitroff CJ. Human B Cell Differentiation Is Characterized by Progressive Remodeling of O-Linked Glycans. Front Immunol 2018; 9:2857. [PMID: 30619255 PMCID: PMC6302748 DOI: 10.3389/fimmu.2018.02857] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/20/2018] [Indexed: 12/16/2022] Open
Abstract
Germinal centers (GC) are microanatomical niches where B cells proliferate, undergo antibody affinity maturation, and differentiate to long-lived memory B cells and antibody-secreting plasma cells. For decades, GC B cells have been defined by their reactivity to the plant lectin peanut agglutinin (PNA), which binds serine/threonine (O-linked) glycans containing the asialylated disaccharide Gal-β1,3-GalNAc-Ser/Thr (also called T-antigen). In T cells, acquisition of PNA binding by activated T cells and thymocytes has been linked with altered tissue homing patterns, cell signaling, and survival. Yet, in GC B cells, the glycobiological basis and significance of PNA binding remains surprisingly unresolved. Here, we investigated the basis for PNA reactivity of GC B cells. We found that GC B cell binding to PNA is associated with downregulation of the α2,3 sialyltransferase, ST3GAL1 (ST3Gal1), and overexpression of ST3Gal1 was sufficient to reverse PNA binding in B cell lines. Moreover, we found that the primary scaffold for PNA-reactive O-glycans in B cells is the B cell receptor-associated receptor-type tyrosine phosphatase CD45, suggesting a role for altered O-glycosylation in antigen receptor signaling. Consistent with similar reports in T cells, ST3Gal1 overexpression in B cells in vitro induced drastic shortening in O-glycans, which we confirmed by both antibody staining and mass spectrometric O-glycomic analysis. Unexpectedly, ST3Gal1-induced changes in O-glycan length also correlated with altered binding of two glycosylation-sensitive CD45 antibodies, RA3-6B2 (more commonly called B220) and MEM55, which (in humans) have previously been reported to favor binding to naïve/GC subsets and memory/plasmablast subsets, respectively. Analysis of primary B cell binding to B220, MEM55, and several plant lectins suggested that B cell differentiation is accompanied by significant loss of O-glycan complexity, including loss of extended Core 2 O-glycans. To our surprise, decreased O-glycan length from naïve to post-GC fates best correlated not with ST3Gal1, but rather downregulation of the Core 2 branching enzyme GCNT1. Thus, our data suggest that O-glycan remodeling is a feature of B cell differentiation, dually regulated by ST3Gal1 and GCNT1, that ultimately results in expression of distinct O-glycosylation states/CD45 glycoforms at each stage of B cell differentiation.
Collapse
Affiliation(s)
- Nicholas Giovannone
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States.,Harvard Medical School, Boston MA, United States
| | | | - Jennifer Liang
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States
| | - Jenna Geddes Sweeney
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States.,Harvard Medical School, Boston MA, United States
| | - Matthew R Kudelka
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States.,Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States
| | - Sandra L King
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States
| | - Gi Soo Lee
- Department of Otology and Laryngology, Harvard Medical School, Boston, MA, United States
| | - Richard D Cummings
- Harvard Medical School, Boston MA, United States.,Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Steven R Barthel
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States
| | - Hans R Widlund
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States.,Harvard Medical School, Boston MA, United States
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Charles J Dimitroff
- Department of Dermatology, Brigham and Women's Hospital, Boston MA, United States.,Harvard Medical School, Boston MA, United States
| |
Collapse
|
134
|
Lübbers J, Rodríguez E, van Kooyk Y. Modulation of Immune Tolerance via Siglec-Sialic Acid Interactions. Front Immunol 2018; 9:2807. [PMID: 30581432 PMCID: PMC6293876 DOI: 10.3389/fimmu.2018.02807] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022] Open
Abstract
One of the key features of the immune system is its extraordinary capacity to discriminate between self and non-self and to respond accordingly. Several molecular interactions allow the induction of acquired immune responses when a foreign antigen is recognized, while others regulate the resolution of inflammation, or the induction of tolerance to self-antigens. Post-translational signatures, such as glycans that are part of proteins (glycoproteins) and lipids (glycolipids) of host cells or pathogens, are increasingly appreciated as key molecules in regulating immunity vs. tolerance. Glycans are sensed by glycan binding receptors expressed on immune cells, such as C-type lectin receptors (CLRs) and Sialic acid binding immunoglobulin type lectins (Siglecs), that respond to specific glycan signatures by triggering tolerogenic or immunogenic signaling pathways. Glycan signatures present on healthy tissue, inflamed and malignant tissue or pathogens provide signals for “self” or “non-self” recognition. In this review we will focus on sialic acids that serve as “self” molecular pattern ligands for Siglecs. We will emphasize on the function of Siglec-expressing mononuclear phagocytes as sensors for sialic acids in tissue homeostasis and describe how the sialic acid-Siglec axis is exploited by tumors and pathogens for the induction of immune tolerance. Furthermore, we highlight how the sialic acid-Siglec axis can be utilized for clinical applications to induce or inhibit immune tolerance.
Collapse
Affiliation(s)
- Joyce Lübbers
- Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Ernesto Rodríguez
- Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| |
Collapse
|
135
|
Yamakawa N, Vanbeselaere J, Chang LY, Yu SY, Ducrocq L, Harduin-Lepers A, Kurata J, Aoki-Kinoshita KF, Sato C, Khoo KH, Kitajima K, Guerardel Y. Systems glycomics of adult zebrafish identifies organ-specific sialylation and glycosylation patterns. Nat Commun 2018; 9:4647. [PMID: 30405127 PMCID: PMC6220181 DOI: 10.1038/s41467-018-06950-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 09/26/2018] [Indexed: 12/31/2022] Open
Abstract
The emergence of zebrafish Danio rerio as a versatile model organism provides the unique opportunity to monitor the functions of glycosylation throughout vertebrate embryogenesis, providing insights into human diseases caused by glycosylation defects. Using a combination of chemical modifications, enzymatic digestion and mass spectrometry analyses, we establish here the precise glycomic profiles of eight individual zebrafish organs and demonstrate that the protein glycosylation and glycosphingolipid expression patterns exhibits exquisite specificity. Concomitant expression screening of a wide array of enzymes involved in the synthesis and transfer of sialic acids shows that the presence of organ-specific sialylation motifs correlates with the localized activity of the corresponding glycan biosynthesis pathways. These findings provide a basis for the rational design of zebrafish lines expressing desired glycosylation profiles.
Collapse
Affiliation(s)
- Nao Yamakawa
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France.,Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
| | - Jorick Vanbeselaere
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France
| | - Lan-Yi Chang
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France.,Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Shin-Yi Yu
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France
| | - Lucie Ducrocq
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France
| | - Anne Harduin-Lepers
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France
| | - Junichi Kurata
- Faculty of Science and Engineering, Soka University, Hachioji, Tokyo, 192-8577, Japan
| | | | - Chihiro Sato
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Ken Kitajima
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
| | - Yann Guerardel
- Université de Lille, CNRS, UMR 8576 - UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F- 59000, Lille, France.
| |
Collapse
|
136
|
Buettner MJ, Shah SR, Saeui CT, Ariss R, Yarema KJ. Improving Immunotherapy Through Glycodesign. Front Immunol 2018; 9:2485. [PMID: 30450094 PMCID: PMC6224361 DOI: 10.3389/fimmu.2018.02485] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/08/2018] [Indexed: 01/04/2023] Open
Abstract
Immunotherapy is revolutionizing health care, with the majority of high impact "drugs" approved in the past decade falling into this category of therapy. Despite considerable success, glycosylation-a key design parameter that ensures safety, optimizes biological response, and influences the pharmacokinetic properties of an immunotherapeutic-has slowed the development of this class of drugs in the past and remains challenging at present. This article describes how optimizing glycosylation through a variety of glycoengineering strategies provides enticing opportunities to not only avoid past pitfalls, but also to substantially improve immunotherapies including antibodies and recombinant proteins, and cell-based therapies. We cover design principles important for early stage pre-clinical development and also discuss how various glycoengineering strategies can augment the biomanufacturing process to ensure the overall effectiveness of immunotherapeutics.
Collapse
Affiliation(s)
- Matthew J Buettner
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Sagar R Shah
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Christopher T Saeui
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States.,Pharmacology/Toxicology Branch I, Division of Clinical Evaluation and Pharmacology/Toxicology, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Bethesda, MD, United States
| | - Ryan Ariss
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| | - Kevin J Yarema
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
137
|
Wu X, Zhao J, Ruan Y, Sun L, Xu C, Jiang H. Sialyltransferase ST3GAL1 promotes cell migration, invasion, and TGF-β1-induced EMT and confers paclitaxel resistance in ovarian cancer. Cell Death Dis 2018; 9:1102. [PMID: 30375371 PMCID: PMC6207573 DOI: 10.1038/s41419-018-1101-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/23/2018] [Accepted: 08/27/2018] [Indexed: 12/14/2022]
Abstract
Sialyltransferases transfer sialic acid to nascent oligosaccharides and are upregulated in cancer. The inhibition of sialyltransferases is emerging as a potential strategy to prevent metastasis in several cancers, including ovarian cancer. ST3GAL1 is a sialyltransferase that catalyzes the transfer of sialic acid from cytidine monophosphate-sialic acid to galactose-containing substrates and is associated with cancer progression and chemoresistance. However, the function of ST3GAL1 in ovarian cancer is uncertain. Herein, we use qRT-PCR, western blotting, and immunohistochemistry to assess the expression of ST3GAL1 in ovarian cancer tissue and cell lines and investigate whether it influences resistance to paclitaxel in vitro and in a mouse xenograft model. We found that ST3GAL1 is upregulated in ovarian cancer tissues and in the ovarian cancer cell lines SKOV-3 and OVCAR3 but downregulated in A2780 ovarian cancer cells. Overexpression of ST3GAL1 in A2780 cells increases cell growth, migration, and invasion whereas ST3GAL1 knockdown in SKOV-3 cells decreases cell growth, migration, and invasion. Furthermore, overexpression of ST3GAL1 increases resistance to paclitaxel while downregulation of ST3GAL1 decreases resistance to paclitaxel in vitro, and overexpression of ST3GAL1 increases tumorigenicity and resistance to paclitaxel in vivo. Transforming growth factor-β1 can increase ST3GAL1 expression and induce ovarian cell epithelial-mesenchymal transition (EMT). However, knockdown of ST3GAL1 inhibits EMT expression. Taken together, our findings have identified a regulatory mechanism involving ST3GAL1 in ovarian cancer. ST3GAL1 may be a promising target for overcoming paclitaxel resistance in ovarian carcinoma.
Collapse
Affiliation(s)
- Xin Wu
- Key Laboratory of Female Reproductive Endocrine Related Diseases; The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Junda Zhao
- First Affiliated Hospital of Xinjiang Medical University, Wulumuqi, 830054, China
| | - Yuanyuan Ruan
- Key Laboratory of Glycoconjugate Research Ministry of Public Health, School of Basic Medical Sciences; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200433, China
| | - Li Sun
- The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Congjian Xu
- Key Laboratory of Female Reproductive Endocrine Related Diseases; The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Hua Jiang
- Key Laboratory of Female Reproductive Endocrine Related Diseases; The Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
138
|
Zhong X, Ma W, Meade CL, Tam AS, Llewellyn E, Cornell R, Cote K, Scarcelli JJ, Marshall JK, Tzvetkova B, Figueroa B, DiNino D, Sievers A, Lee C, Guo J, Mahan E, Francis C, Lam K, D'Antona AM, Zollner R, Zhu HL, Kriz R, Somers W, Lin L. Transient CHO expression platform for robust antibody production and its enhanced N-glycan sialylation on therapeutic glycoproteins. Biotechnol Prog 2018; 35:e2724. [PMID: 30299005 DOI: 10.1002/btpr.2724] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/24/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022]
Abstract
Large-scale transient expression in mammalian cells is a rapid protein production technology often used to shorten overall timelines for biotherapeutics drug discovery. In this study we demonstrate transient expression in a Chinese hamster ovary (CHO) host (ExpiCHO-S™) cell line capable of achieving high recombinant antibody expression titers, comparable to levels obtained using human embryonic kidney (HEK) 293 cells. For some antibodies, ExpiCHO-S™ cells generated protein materials with better titers and improved protein quality characteristics (i.e., less aggregation) than those from HEK293. Green fluorescent protein imaging data indicated that ExpiCHO-S™ displayed a delayed but prolonged transient protein expression process compared to HEK293. When therapeutic glycoproteins containing non-Fc N-linked glycans were expressed in transient ExpiCHO-S™, the glycan pattern was unexpectedly found to have few sialylated N-glycans, in contrast to glycans produced within a stable CHO expression system. To improve N-glycan sialylation in transient ExpiCHO-S™, we co-transfected galactosyltransferase and sialyltransferase genes along with the target genes, as well as supplemented the culture medium with glycan precursors. The authors have demonstrated that co-transfection of glycosyltransferases combined with medium addition of galactose and uridine led to increased sialylation content of N-glycans during transient ExpiCHO-S™ expression. These results have provided a scientific basis for developing a future transient CHO system with N-glycan compositions that are similar to those profiles obtained from stable CHO protein production systems. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 35: e2724, 2019.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Weijun Ma
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Caryl L Meade
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Amy S Tam
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Eliza Llewellyn
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Richard Cornell
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Kaffa Cote
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - John J Scarcelli
- Cell Line Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Jeffrey K Marshall
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Boriana Tzvetkova
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Bruno Figueroa
- Bioprocessing Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Dana DiNino
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts, 01810
| | - Annette Sievers
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Christopher Lee
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Jane Guo
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Evan Mahan
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Christopher Francis
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Khetemenee Lam
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Aaron M D'Antona
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Richard Zollner
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Hongli L Zhu
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Ron Kriz
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Will Somers
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| | - Laura Lin
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, Massachusetts, 02139
| |
Collapse
|
139
|
Fan TC, Yeo HL, Hsu HM, Yu JC, Ho MY, Lin WD, Chang NC, Yu J, Yu AL. Reciprocal feedback regulation of ST3GAL1 and GFRA1 signaling in breast cancer cells. Cancer Lett 2018; 434:184-195. [PMID: 30040982 DOI: 10.1016/j.canlet.2018.07.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023]
Abstract
GFRA1 and RET are overexpressed in estrogen receptor (ER)-positive breast cancers. Binding of GDNF to GFRA1 triggers RET signaling leading to ER phosphorylation and estrogen-independent transcriptional activation of ER-dependent genes. Both GFRA1 and RET are membrane proteins which are N-glycosylated but no O-linked sialylation site on GFRA1 or RET has been reported. We found GFRA1 to be a substrate of ST3GAL1-mediated O-linked sialylation, which is crucial to GDNF-induced signaling in ER-positive breast cancer cells. Silencing ST3GAL1 in breast cancer cells reduced GDNF-induced phosphorylation of RET, AKT and ERα, as well as GDNF-mediated cell proliferation. Moreover, GDNF induced transcription of ST3GAL1, revealing a positive feedback loop regulating ST3GAL1 and GDNF/GFRA1/RET signaling in breast cancers. Finally, we demonstrated ST3GAL1 knockdown augments anti-cancer efficacy of inhibitors of RET and/or ER. Moreover, high expression of ST3GAL1 was associated with poor clinical outcome in patients with late stage breast cancer and high expression of both ST3GAL1 and GFRA1 adversely impacted outcome in those with high grade tumors.
Collapse
Affiliation(s)
- Tan-Chi Fan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hui Ling Yeo
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan; Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Huan-Ming Hsu
- Department of Surgery, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei, Taiwan; Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jyh-Cherng Yu
- Division of General Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Yi Ho
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Wen-Der Lin
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Biochemistry and Molecular Biology, Chang Gung University, Gueishan, Taoyuan, Taiwan
| | - Nai-Chuan Chang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Genomics Research Center, Academia Sinica, Taipei, Taiwan; Department of Pediatrics/Hematology Oncology, University of California in San Diego, San Diego, CA, USA.
| |
Collapse
|
140
|
Pan Y, Hu J, Ma J, Qi X, Zhou H, Miao X, Zheng W, Jia L. MiR-193a-3p and miR-224 mediate renal cell carcinoma progression by targeting alpha-2,3-sialyltransferase IV and the phosphatidylinositol 3 kinase/Akt pathway. Mol Carcinog 2018; 57:1067-1077. [PMID: 29667779 DOI: 10.1002/mc.22826] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 04/04/2018] [Accepted: 04/14/2018] [Indexed: 12/18/2022]
Abstract
Tumor metastasis is a major cause of cancer-related death in renal cell carcinoma (RCC). MicroRNAs (miRNAs) have been widely known to modulate proliferation invasion, metastasis, and apoptosis of cancer cells. In this study, we aimed to investigate the function and novel target of miR-193a-3p and miR-224 in RCC. The levels of miR-193a-3p and miR-224 were significantly increased in RCC tissues and RCC cell lines. Alpha-2,3-Sialyltransferase IV (ST3GalIV) was highly expressed in adjacent nontumor tissues and human normal proximal tubular cell line HK-2 compared to RCC tissues and cell lines. ST3GalIV expression was negatively correlated with miR-193a-3p and miR-224. Further analysis indicated that miR-193a-3p and miR-224 directly targeted ST3GalIV. MiR-193a-3p and miR-224 increased cell proliferation and migration by directly inhibiting ST3GalIV, and this effect was reversed by co-transfection with ST3GalIV in vitro. Overexpression of miR-193a-3p and miR-224 increased RCC cell proliferation in vivo. Furthermore, the phosphatidylinositol 3 kinase (PI3K)/Akt pathway was mediated by miR-193a-3p and miR-224 in RCC cell lines. Collectively, these results suggested that miR-193a-3p and miR-224 played an important role in regulation of RCC by targeting ST3GalIV via PI3K/Akt pathway.
Collapse
Affiliation(s)
- Yue Pan
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Jialei Hu
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Jia Ma
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Xia Qi
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Huimin Zhou
- Department of Microbiology, Dalian Medical University, Dalian, Liaoning Province, China
| | - Xiaoyan Miao
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Wei Zheng
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Li Jia
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
141
|
Yuan Q, He J, Niu Y, Chen J, Zhao Y, Zhang Y, Yu C. Sandwich-type biosensor for the detection of α2,3-sialylated glycans based on fullerene-palladium-platinum alloy and 4-mercaptophenylboronic acid nanoparticle hybrids coupled with Au-methylene blue-MAL signal amplification. Biosens Bioelectron 2018; 102:321-327. [DOI: 10.1016/j.bios.2017.11.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/24/2017] [Accepted: 11/12/2017] [Indexed: 01/05/2023]
|
142
|
Lopez Aguilar A, Meng L, Hou X, Li W, Moremen KW, Wu P. Sialyltransferase-Based Chemoenzymatic Histology for the Detection of N- and O-Glycans. Bioconjug Chem 2018; 29:1231-1239. [PMID: 29569918 DOI: 10.1021/acs.bioconjchem.8b00021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Profiling specific glycans in histopathological samples is hampered by the lack of selective and sensitive tools for their detection. Here, we report on the development of chemoenzymatic histology of membrane polysaccharide (CHoMP)-based methods for the detection of O- and N-linked glycans on tissue sections via the use of sialyltransferases ST3Gal1 and ST6Gal1, respectively. Combining these two methods, we developed tandem labeling and double labeling strategies that permit the detection of unsialylated and sialylated glycans or the detection of O- and N-linked glycans on the same tissue section, respectively. We applied these methods to screen murine tissue specimens, human multiple-organ cancer arrays, and lymphoma and prostate cancer arrays. Using tandem labeling with ST6Gal1 to analyze N-glycans in a prostate cancer array, we found striking differences in expression patterns of both sialylated and unsialylated N-glycans between cancerous and healthy samples. Such differences were also observed between normal tissue from healthy donors and healthy tissue adjacent to tumors. Our double labeling technique identified significant differences in unsialylated O-glycans between B-cell and T-cell lymphomas and between B-cell lymphomas and normal adjacent lymph nodes. Remarkable differences were also detected between adjacent lymph nodes and spleen tissue samples. These new chemoenzymatic histology methods therefore provide valuable tools for the analysis of glycans in clinically relevant tissue samples.
Collapse
Affiliation(s)
- Aime Lopez Aguilar
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Lu Meng
- Complex Carbohydrate Research Center , University of Georgia , Athens , Georgia 30602 , United States
| | - Xiaomeng Hou
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Wei Li
- Department of Oncology , The First Affiliated Hospital of Soochow University , Suzhou 215006 , China
| | - Kelley W Moremen
- Complex Carbohydrate Research Center , University of Georgia , Athens , Georgia 30602 , United States
| | - Peng Wu
- Department of Molecular Medicine , The Scripps Research Institute , La Jolla , California 92037 , United States
| |
Collapse
|
143
|
Wen KC, Sung PL, Hsieh SL, Chou YT, Lee OKS, Wu CW, Wang PH. α2,3-sialyltransferase type I regulates migration and peritoneal dissemination of ovarian cancer cells. Oncotarget 2018; 8:29013-29027. [PMID: 28423672 PMCID: PMC5438708 DOI: 10.18632/oncotarget.15994] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/10/2017] [Indexed: 12/19/2022] Open
Abstract
Epithelial ovarian cancer (EOC) has the highest mortality rate among gynecologic cancers due to advanced stage presentation, peritoneal dissemination, and refractory ascites at diagnosis. We investigated the role of α2,3-sialyltransferase type I (ST3GalI) by analyzing human ovarian cancer datasets and human EOC tissue arrays. We found that high expression of ST3GalI was associated with advanced stage EOC. Transwell migration and cell invasion assays showed that high ST3GalI expression enhanced migration of EOC cells. We also observed that there was a linear relation between ST3GalI expression and epidermal growth factor receptor (EGFR) signaling in EOC patients, and that high ST3GalI expression blocked the effect of EGFR inhibitors. Co-Immunoprecipitation experiments demonstrated that ST3GalI and EGFR were present in the same protein complex. Inhibition of ST3GalI using a competitive inhibitor, Soyasaponin I (SsaI), inhibited tumor cell migration and dissemination in the in vivo mouse model with transplanted MOSEC cells. Further, SsaI synergistically enhanced the anti-tumor effects of EGFR inhibitor on EOC cells. Our study demonstrates that ST3GalI regulates ovarian cancer cell migration and peritoneal dissemination via EGFR signaling. This suggests α2,3-linked sialylation inhibitors in combination with EGFR inhibitors could be effective agents for the treatment of EOC.
Collapse
Affiliation(s)
- Kuo-Chang Wen
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei, Taiwan
| | - Pi-Lin Sung
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Ting Chou
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Oscar Kuang-Sheng Lee
- Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan.,Taipei City Hospital, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Wen Wu
- Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan.,Department of Obstetrics and Gynecology, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
144
|
Liang L, Xu J, Wang M, Xu G, Zhang N, Wang G, Zhao Y. LncRNA HCP5 promotes follicular thyroid carcinoma progression via miRNAs sponge. Cell Death Dis 2018; 9:372. [PMID: 29515098 PMCID: PMC5841368 DOI: 10.1038/s41419-018-0382-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/22/2018] [Accepted: 02/05/2018] [Indexed: 12/21/2022]
Abstract
Long non-coding RNAs (lncRNAs), which are important functional regulators in cancer, have received increased attention in recent years. In this study, next-generation sequencing technology was used to identify aberrantly expressed lncRNAs in follicular thyroid carcinoma (FTC). The long non-coding RNA-HLA complex P5 (HCP5) was found to be overexpressed in FTC. The results of the qPCR analysis were consistent with the sequencing results. In addition, functional experiments showed that overexpression of HCP5 can promote the proliferation, migration, invasiveness and angiogenic ability of FTC cells. Furthermore, according to the sequencing results, HCP5 and alpha-2, 6-sialyltransferase 2 (ST6GAL2) were co-expressed in FTC. We hypothesised that ST6GAL2 may be regulated by HCP5, which would in turn mediate the activity of FTC cells. Through qPCR, immunostaining analyses and functional experiments, we determined that the expression of HCP5 was elevated and was correlated with the levels of ST6GAL2 in FTC tissues and cells. Mechanistic experiments showed that HCP5 functions as a competing endogenous RNA (ceRNA) and acts as a sponge for miR-22-3p, miR-186-5p and miR-216a-5p, which activates ST6GAL2. In summary, our study revealed that HCP5 is a tumour regulator in the development of FTC and that it may contribute to improvement of FTC diagnosis and therapy.
Collapse
Affiliation(s)
- Leilei Liang
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Jingchao Xu
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meng Wang
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Gaoran Xu
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Ning Zhang
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Guangzhi Wang
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China.
| | - Yongfu Zhao
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
145
|
Petit D, Teppa E, Cenci U, Ball S, Harduin-Lepers A. Reconstruction of the sialylation pathway in the ancestor of eukaryotes. Sci Rep 2018; 8:2946. [PMID: 29440651 PMCID: PMC5811610 DOI: 10.1038/s41598-018-20920-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 01/25/2018] [Indexed: 11/18/2022] Open
Abstract
The biosynthesis of sialylated molecules of crucial relevance for eukaryotic cell life is achieved by sialyltransferases (ST) of the CAZy family GT29. These enzymes are widespread in the Deuterostoma lineages and more rarely described in Protostoma, Viridiplantae and various protist lineages raising the question of their presence in the Last eukaryotes Common Ancestor (LECA). If so, it is expected that the main enzymes associated with sialic acids metabolism are also present in protists. We conducted phylogenomic and protein sequence analyses to gain insights into the origin and ancient evolution of ST and sialic acid pathway in eukaryotes, Bacteria and Archaea. Our study uncovered the unreported occurrence of bacterial GT29 ST and evidenced the existence of 2 ST groups in the LECA, likely originating from the endosymbiotic event that generated mitochondria. Furthermore, distribution of the major actors of the sialic acid pathway in the different eukaryotic phyla indicated that these were already present in the LECA, which could also access to this essential monosaccharide either endogenously or via a sialin/sialidase uptake mechanism involving vesicles. This pathway was lost in several basal eukaryotic lineages including Archaeplastida despite the presence of two different ST groups likely assigned to other functions.
Collapse
Affiliation(s)
- Daniel Petit
- Université de Limoges, Laboratoire Pereine 123, av. A. Thomas, 87060, Limoges Cedex, France
| | - Elin Teppa
- Bioinformatics Unit, Fundación Instituto Leloir -IIBBA CONICET, Av. Patricias Argentinas 435, C1405BWE, Buenos Aires, Argentina
| | - Ugo Cenci
- University of Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F 59000, Lille, France
- UGSF, Bât. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France
| | - Steven Ball
- University of Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F 59000, Lille, France
- UGSF, Bât. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France
| | - Anne Harduin-Lepers
- University of Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F 59000, Lille, France.
- UGSF, Bât. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France.
| |
Collapse
|
146
|
Maso K, Grigoletto A, Pasut G. Transglutaminase and Sialyltransferase Enzymatic Approaches for Polymer Conjugation to Proteins. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 112:123-142. [PMID: 29680235 DOI: 10.1016/bs.apcsb.2018.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Proteins hold a central role in medicine and biology, also confirmed by the several therapeutic applications based on biologic drugs. Such therapies are of great relevance thanks to high potency and safety of proteins. Nevertheless, many proteins as therapeutics might present issues like fast kidney clearance, rapid enzymatic degradation, or immunogenicity. Such defects implicate frequent administrations or administrations at high doses of the therapeutics, thus yielding or exacerbating potential side effects. A successful technology for improving the clinical profiles of proteins is the conjugation of polymers to the protein surface. The design of a protein-polymer conjugate presents critical aspects that determine the efficacy and safety of the final product. The control over stoichiometry and conjugation site is a strict criterion on which researchers have been intensively focused during the years, in order to obtain homogeneous and batch-to-batch reproducible products. An innovative site-specific conjugation strategy relies on the use of enzymes as tools to mediate polymer conjugation. Enzymatic approaches are attractive because they allow site-selective polymer conjugation at specific protein amino acids. In these reactions, the polymer is a substrate analog that replaces the native substrate. Furthermore, enzymes can count other advantages such as high yields of conversion and physiological conditions of reaction. This chapter provides a meaningful description of protein-polymer conjugation through transglutaminase-mediated and sialyltransferase-mediated enzymatic strategies, reporting the mechanism of action and some relevant examples.
Collapse
Affiliation(s)
| | | | - Gianfranco Pasut
- University of Padua, Padua, Italy; Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| |
Collapse
|
147
|
Mariño-Crespo Ó, Fernández-Briera A, Gil-Martín E. Identification of proteins with the CDw75 epitope in human colorectal cancer. Oncol Lett 2018; 15:580-587. [PMID: 29391890 DOI: 10.3892/ol.2017.7336] [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: 11/07/2016] [Accepted: 07/28/2017] [Indexed: 11/05/2022] Open
Abstract
The CDw75 epitope is an α(2,6) sialylated antigen overexpressed in colorectal cancer (CRC), where its expression correlates with the progression of the disease. The CDw75 epitope is located mainly in N-glycoproteins, whose identity remains unknown. The aim of the present study was to identify proteins with the CDw75 epitope as a strategy to deepen the understanding of molecular pathogenesis of CRC and to identify novel biomarkers for this disease. For this purpose, a two-dimensional electrophoresis approach was employed. Protein spots in the gels were matched to the corresponding CDw75 positive spots in the immunoblotted polyvinylidene difluoride membranes, and further identification of the protein species was performed by mass spectrometry. Additionally, one-dimensional western blotting experiments were performed to verify the expression of these candidate proteins in the colorectal tissue and their coincidence in molecular mass with the CDw75-positive bands. The findings of the present study indicate that haptoglobin and the keratins 8 (K8) and 18 (K18) are proteins with the CDw75 epitope in the colorectal tissue from CRC patients and also suggest novel functions and cellular locations for these proteins in the colorectal tissue and in relation to CRC.
Collapse
Affiliation(s)
- Óscar Mariño-Crespo
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, 36310 Vigo, Spain
| | - Almudena Fernández-Briera
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, 36310 Vigo, Spain
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, 36310 Vigo, Spain
| |
Collapse
|
148
|
Silva M, Videira PA, Sackstein R. E-Selectin Ligands in the Human Mononuclear Phagocyte System: Implications for Infection, Inflammation, and Immunotherapy. Front Immunol 2018; 8:1878. [PMID: 29403469 PMCID: PMC5780348 DOI: 10.3389/fimmu.2017.01878] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022] Open
Abstract
The mononuclear phagocyte system comprises a network of circulating monocytes and dendritic cells (DCs), and “histiocytes” (tissue-resident macrophages and DCs) that are derived in part from blood-borne monocytes and DCs. The capacity of circulating monocytes and DCs to function as the body’s first-line defense against offending pathogens greatly depends on their ability to egress the bloodstream and infiltrate inflammatory sites. Extravasation involves a sequence of coordinated molecular events and is initiated by E-selectin-mediated deceleration of the circulating leukocytes onto microvascular endothelial cells of the target tissue. E-selectin is inducibly expressed by cytokines (tumor necrosis factor-α and IL-1β) on inflamed endothelium, and binds to sialofucosylated glycan determinants displayed on protein and lipid scaffolds of blood cells. Efficient extravasation of circulating monocytes and DCs to inflamed tissues is crucial in facilitating an effective immune response, but also fuels the immunopathology of several inflammatory disorders. Thus, insights into the structural and functional properties of the E-selectin ligands expressed by different monocyte and DC populations is key to understanding the biology of protective immunity and the pathobiology of several acute and chronic inflammatory diseases. This review will address the role of E-selectin in recruitment of human circulating monocytes and DCs to sites of tissue injury/inflammation, the structural biology of the E-selectin ligands expressed by these cells, and the molecular effectors that shape E-selectin ligand cell-specific display. In addition, therapeutic approaches targeting E-selectin receptor/ligand interactions, which can be used to boost host defense or, conversely, to dampen pathological inflammatory conditions, will also be discussed.
Collapse
Affiliation(s)
- Mariana Silva
- Department of Dermatology, Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA, United States
| | - Paula A Videira
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisboa, Portugal.,Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Robert Sackstein
- Department of Dermatology, Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Program of Excellence in Glycosciences, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
149
|
Sieve I, Münster-Kühnel AK, Hilfiker-Kleiner D. Regulation and function of endothelial glycocalyx layer in vascular diseases. Vascul Pharmacol 2018; 100:26-33. [DOI: 10.1016/j.vph.2017.09.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 12/23/2022]
|
150
|
Jia L, Luo S, Ren X, Li Y, Hu J, Liu B, Zhao L, Shan Y, Zhou H. miR-182 and miR-135b Mediate the Tumorigenesis and Invasiveness of Colorectal Cancer Cells via Targeting ST6GALNAC2 and PI3K/AKT Pathway. Dig Dis Sci 2017; 62:3447-3459. [PMID: 29030743 DOI: 10.1007/s10620-017-4755-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/07/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Metastasis is a leading cause of cancer-related death including colorectal cancer (CRC). MicroRNAs are known to regulate cancer pathways and to be expressed aberrantly in cancer. Aberrant sialylation is closely associated with malignant phenotype of tumor cells, including invasiveness and metastasis. AIM This study aimed to investigate the association of miR-182 and miR-135b with proliferation and invasion by targeting sialyltransferase ST6GALNAC2 in CRC cells and explore the potential molecular mechanism. METHODS We measured the levels of miR-182, miR-135b, and ST6GALNAC2 in a series of CRC cell lines and tissues using real-time PCR. Bioinformatics analysis and luciferase reporter assay were performed to test the direct binding of miR-182 and miR-135b to the target gene ST6GALNAC2. We also analyzed the possible role of miR-182/-135b on colony formation, wound healing, invasion, and tube formation. RESULTS The expression of miR-182 and miR-135b was higher in tumor tissues compared to adjacent noncancerous tissues of CRC patients, as well as up-regulated in SW620 cells than in SW480 cells with different metastatic potential. By applying bioinformatics analysis and luciferase reporter assay, we identified ST6GALNAC2 as the direct target of miR-182/-135b. Furthermore, miR-182/-135b inhibited significantly ST6GALNAC2 expression, and consistently, ST6GALNAC2 mediated migration, adhesion, invasion, proliferation, and tumor angiogenesis in CRC cell lines. Additionally, PI3K/AKT signaling pathway was regulated by miR-182/135b, which was partially blocked by altered level of ST6GALNAC2 in CRC. CONCLUSIONS The miR-182/-135b/ST6GALNAC2/PI3K/AKT axis may serve as a predictive biomarker and a potential therapeutic target in CRC treatment.
Collapse
Affiliation(s)
- Li Jia
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China.
| | - Shihua Luo
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
- Department of Traumatology, Shanghai Ruijin Hospital, Jiaotong University, Shanghai, 200025, China
| | - Xiang Ren
- College of Stomatology, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Yang Li
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Jialei Hu
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Bing Liu
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Lifen Zhao
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Yujia Shan
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| | - Huimin Zhou
- College of Laboratory Medicine, Dalian Medical University, Dalian, 116044, Liaoning Province, China
| |
Collapse
|