1
|
Barboza BR, Macedo-da-Silva J, Silva LAMT, Gomes VDM, Santos DM, Marques-Neto AM, Mule SN, Angeli CB, Borsoi J, Moraes CB, Moutinho-Melo C, Mühlenhoff M, Colli W, Marie SKN, Pereira LDV, Alves MJM, Palmisano G. ST8Sia2 polysialyltransferase protects against infection by Trypanosoma cruzi. PLoS Negl Trop Dis 2024; 18:e0012454. [PMID: 39321148 PMCID: PMC11466412 DOI: 10.1371/journal.pntd.0012454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 10/10/2024] [Accepted: 08/13/2024] [Indexed: 09/27/2024] Open
Abstract
Glycosylation is one of the most structurally and functionally diverse co- and post-translational modifications in a cell. Addition and removal of glycans, especially to proteins and lipids, characterize this process which has important implications in several biological processes. In mammals, the repeated enzymatic addition of a sialic acid unit to underlying sialic acids (Sia) by polysialyltransferases, including ST8Sia2, leads to the formation of a sugar polymer called polysialic acid (polySia). The functional relevance of polySia has been extensively demonstrated in the nervous system. However, the role of polysialylation in infection is still poorly explored. Previous reports have shown that Trypanosoma cruzi (T. cruzi), a flagellated parasite that causes Chagas disease (CD), changes host sialylation of glycoproteins. To understand the role of host polySia during T. cruzi infection, we used a combination of in silico and experimental tools. We observed that T. cruzi reduces both the expression of the ST8Sia2 and the polysialylation of target substrates. We also found that chemical and genetic inhibition of host ST8Sia2 increased the parasite load in mammalian cells. We found that modulating host polysialylation may induce oxidative stress, creating a microenvironment that favors T. cruzi survival and infection. These findings suggest a novel approach to interfere with parasite infections through modulation of host polysialylation.
Collapse
Affiliation(s)
- Bruno Rafael Barboza
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Vinícius de Morais Gomes
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Deivid Martins Santos
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Antônio Moreira Marques-Neto
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Simon Ngao Mule
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Claudia Blanes Angeli
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Juliana Borsoi
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Carolina Borsoi Moraes
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cristiane Moutinho-Melo
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Laboratory of Immunological and Antitumor Analysis, Department of Antibiotics, Bioscience Center, and Keizo Asami Immunopathology Laboratory, Federal University of Pernambuco, Recife, Brazil
| | - Martina Mühlenhoff
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Walter Colli
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Suely Kazue Nagashi Marie
- Laboratory of Molecular and Cellular Biology (LIM 15), Department of Neurology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Lygia da Veiga Pereira
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | - Maria Julia Manso Alves
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
2
|
Guo H, Mi P. Polymer-drug and polymer-protein conjugated nanocarriers: Design, drug delivery, imaging, therapy, and clinical applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1988. [PMID: 39109479 DOI: 10.1002/wnan.1988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 01/06/2025]
Abstract
Polymer-drug conjugates and polymer-protein conjugates have been pivotal in the realm of drug delivery systems for over half a century. These polymeric drugs are characterized by the conjugation of therapeutic molecules or functional moieties to polymers, enabling a range of benefits including extended circulation times, targeted delivery, controlled release, and decreased immunogenicity. This review delves into recent advancements and challenges in the clinical translations and preclinical studies of polymer-drug conjugates and polymer-protein conjugates. The design principles and functionalization strategies crucial for the development of these polymeric drugs were explored followed by the review of structural properties and characteristics of various polymer carriers. This review also identifies significant obstacles in the clinical translation of polymer-drug conjugates and provides insights into the directions for their future development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Haochen Guo
- Department of Radiology, Huaxi MR Research Center (HMRRC), and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Babulic JL, Kofsky JM, Boddington ME, Kim Y, Leblanc EV, Cook MG, Garnier CR, Emberley-Korkmaz S, Colpitts CC, Capicciotti CJ. One-Step Selective Labeling of Native Cell Surface Sialoglycans by Exogenous α2,8-Sialylation. ACS Chem Biol 2023; 18:2418-2429. [PMID: 37934063 DOI: 10.1021/acschembio.3c00475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Exo-enzymatic glycan labeling strategies have emerged as versatile tools for efficient and selective installation of terminal glyco-motifs onto live cell surfaces. Through employing specific enzymes and nucleotide-sugar probes, cells can be equipped with defined glyco-epitopes for modulating cell function or selective visualization and enrichment of glycoconjugates. Here, we identifyCampylobacter jejunisialyltransferase Cst-II I53S as a tool for cell surface glycan modification, expanding the exo-enzymatic labeling toolkit to include installation of α2,8-disialyl epitopes. Labeling with Cst-II was achieved with biotin- and azide-tagged CMP-Neu5Ac derivatives on a model glycoprotein and native sialylated cell surface glycans across a panel of cell lines. The introduction of modified Neu5Ac derivatives onto cells by Cst-II was also retained on the surface for 6 h. By examining the specificity of Cst-II on cell surfaces, it was revealed that the α2,8-sialyltransferase primarily labeled N-glycans, with O-glycans labeled to a lesser extent, and there was an apparent preference for α2,3-linked sialosides on cells. This approach thus broadens the scope of tools for selective exo-enzymatic labeling of native sialylated glycans and is highly amenable for the construction of cell-based arrays.
Collapse
Affiliation(s)
- Jonathan L Babulic
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Joshua M Kofsky
- Department of Chemistry, Queen's University, Kingston K7L 3N6, Canada
| | - Marie E Boddington
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Youjin Kim
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Emmanuelle V Leblanc
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Madeleine G Cook
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Cole R Garnier
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Sophie Emberley-Korkmaz
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
| | - Chantelle J Capicciotti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston K7L 3N6, Canada
- Department of Chemistry, Queen's University, Kingston K7L 3N6, Canada
- Department of Surgery, Queen's University, Kingston K7L 3N6, Canada
| |
Collapse
|
4
|
Tschang M, Kumar S, Young W, Schachner M, Theis T. Small Organic Compounds Mimicking the Effector Domain of Myristoylated Alanine-Rich C-Kinase Substrate Stimulate Female-Specific Neurite Outgrowth. Int J Mol Sci 2023; 24:14271. [PMID: 37762575 PMCID: PMC10532424 DOI: 10.3390/ijms241814271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/15/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Myristoylated alanine-rich C-kinase substrate (MARCKS) is a critical member of a signaling cascade that influences disease-relevant neural functions such as neural growth and plasticity. The effector domain (ED) of MARCKS interacts with the extracellular glycan polysialic acid (PSA) through the cell membrane to stimulate neurite outgrowth in cell culture. We have shown that a synthetic ED peptide improves functional recovery after spinal cord injury in female but not male mice. However, peptides themselves are unstable in therapeutic applications, so we investigated more pharmacologically relevant small organic compounds that mimic the ED peptide to maximize therapeutic potential. Using competition ELISAs, we screened small organic compound libraries to identify molecules that structurally and functionally mimic the ED peptide of MARCKS. Since we had shown sex-specific effects of MARCKS on spinal cord injury recovery, we assayed neuronal viability as well as neurite outgrowth from cultured cerebellar granule cells of female and male mice separately. We found that epigallocatechin, amiodarone, sertraline, tegaserod, and nonyloxytryptamine bind to a monoclonal antibody against the ED peptide, and compounds stimulate neurite outgrowth in cultured cerebellar granule cells of female mice only. Therefore, a search for compounds that act in males appears warranted.
Collapse
Affiliation(s)
- Monica Tschang
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.T.); (W.Y.)
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08844, USA;
| | - Wise Young
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.T.); (W.Y.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.T.); (W.Y.)
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (M.T.); (W.Y.)
| |
Collapse
|
5
|
Shinde P, Kiepas A, Zhang L, Sudhir S, Konstantopoulos K, Stamatos NM. Polysialylation controls immune function of myeloid cells in murine model of pneumococcal pneumonia. Cell Rep 2023; 42:112648. [PMID: 37339052 PMCID: PMC10592499 DOI: 10.1016/j.celrep.2023.112648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/13/2023] [Accepted: 05/31/2023] [Indexed: 06/22/2023] Open
Abstract
Polysialic acid (polySia) is a post-translational modification of a select group of cell-surface proteins that guides cellular interactions. As the overall impact of changes in expression of this glycan on leukocytes during infection is not known, we evaluate the immune response of polySia-deficient ST8SiaIV-/- mice infected with Streptococcus pneumoniae (Spn). Compared with wild-type (WT) mice, ST8SiaIV-/- mice are less susceptible to infection and clear Spn from airways faster, with alveolar macrophages demonstrating greater viability and phagocytic activity. Leukocyte pulmonary recruitment, paradoxically, is diminished in infected ST8SiaIV-/- mice, corroborated by adoptive cell transfer, microfluidic migration experiments, and intravital microscopy, and possibly explained by dysregulated ERK1/2 signaling. PolySia is progressively lost from neutrophils and monocytes migrating from bone marrow to alveoli in Spn-infected WT mice, consistent with changing cellular functions. These data highlight multidimensional effects of polySia on leukocytes during an immune response and suggest therapeutic interventions for optimizing immunity.
Collapse
Affiliation(s)
- Prajakta Shinde
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lei Zhang
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shreya Sudhir
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nicholas M Stamatos
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
6
|
Gretenkort L, Thiesler H, Hildebrandt H. Neuroimmunomodulatory properties of polysialic acid. Glycoconj J 2023; 40:277-294. [PMID: 37171513 DOI: 10.1007/s10719-023-10120-z] [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: 11/15/2022] [Revised: 11/16/2022] [Accepted: 05/02/2023] [Indexed: 05/13/2023]
Abstract
Polymeric sialic acid (polysialic acid, polySia) is a remarkable posttranslational modification of only few select proteins. The major, and most prominent polySia protein carrier is the neural cell adhesion molecule NCAM. Here, the key functions of polySia are to regulate interactions of NCAM and to balance cellular interactions in brain development and plasticity. During recent years, however, increasing evidence points towards a role of polySia in the modulation of immune responses. These immunomodulatory functions can be mediated by polySia on proteins other than NCAM, presented either on the cell surface or released into the extracellular space. This perspective review summarizes our current knowledge and addresses major open questions on polySia and polySia receptors in modulating innate immune responses in the brain.
Collapse
Affiliation(s)
- Lina Gretenkort
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| |
Collapse
|
7
|
Zhang Q, Li S, He L, Feng X. A brief review of polysialic acid-based drug delivery systems. Int J Biol Macromol 2023; 230:123151. [PMID: 36610578 DOI: 10.1016/j.ijbiomac.2023.123151] [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: 09/13/2022] [Revised: 12/27/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
Polysialic acid (PSA) is a straight-chain homoglycan linked by N-acetylneuraminic acid monomers via α-2, 8- or α-2, 9-glycosidic bonds. As a negatively charged non-glycosaminoglycan, PSA has the remarkable characteristics of non-immunogenicity and biodegradation. Although different in class, PSA is similar to poly(ethylene glycol), and was originally used to increase the stability of the delivery system in circulation to prolong the half-life. As research continues, PSA's application potential in the pharmaceutical field becomes increasingly prominent. It can be used as a biomaterial for protein polysialylation and tissue engineering, and it can be used alone or with other materials to develop multifunctional drug delivery systems. In this article, the results of the bioproduction and biofunction of PSA are introduced, the common strategies for chemical modification of PSA are summarized, and the application progress of PSA-based drug delivery systems is reviewed.
Collapse
Affiliation(s)
- Qixiong Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Shanshan Li
- College of Pharmacy, Southwest Minzu University, Chengdu 610000, China
| | - Lin He
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xueting Feng
- College of Pharmacy, Southwest Minzu University, Chengdu 610000, China
| |
Collapse
|
8
|
"Glyco-sulfo barcodes" regulate chemokine receptor function. Cell Mol Life Sci 2023; 80:55. [PMID: 36729338 PMCID: PMC9894980 DOI: 10.1007/s00018-023-04697-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/16/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023]
Abstract
Chemokine ligands and receptors regulate the directional migration of leukocytes. Post-translational modifications of chemokine receptors including O-glycosylation and tyrosine sulfation have been reported to regulate ligand binding and resulting signaling. Through in silico analyses, we determined potential conserved O-glycosylation and sulfation sites on human and murine CC chemokine receptors. Glyco-engineered CHO cell lines were used to measure the impact of O-glycosylation on CC chemokine receptor CCR5, while mutation of tyrosine residues and treatment with sodium chlorate were performed to determine the effect of tyrosine sulfation. Changing the glycosylation or tyrosine sulfation on CCR5 reduced the receptor signaling by the more positively charged CCL5 and CCL8 more profoundly compared to the less charged CCL3. The loss of negatively charged sialic acids resulted only in a minor effect on CCL3-induced signal transduction. The enzymes GalNAc-T1 and GalNAc-T11 were shown to be involved in the process of chemokine receptor O-glycosylation. These results indicate that O-glycosylation and tyrosine sulfation are involved in the fine-tuning and recognition of chemokine interactions with CCR5 and the resulting signaling.
Collapse
|
9
|
Fliniaux I, Marchand G, Molinaro C, Decloquement M, Martoriati A, Marin M, Bodart JF, Harduin-Lepers A, Cailliau K. Diversity of sialic acids and sialoglycoproteins in gametes and at fertilization. Front Cell Dev Biol 2022; 10:982931. [PMID: 36340022 PMCID: PMC9630641 DOI: 10.3389/fcell.2022.982931] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/10/2022] [Indexed: 09/22/2023] Open
Abstract
Sialic acids are a family of 9-carbon monosaccharides with particular physicochemical properties. They modulate the biological functions of the molecules that carry them and are involved in several steps of the reproductive process. Sialoglycoproteins participate in the balance between species recognition and specificity, and the mechanisms of these aspects remain an issue in gametes formation and binding in metazoan reproduction. Sialoglycoproteins form a specific coat at the gametes surface and specific polysialylated chains are present on marine species oocytes. Spermatozoa are submitted to critical sialic acid changes in the female reproductive tract facilitating their migration, their survival through the modulation of the female innate immune response, and the final oocyte-binding event. To decipher the role of sialic acids in gametes and at fertilization, the dynamical changes of enzymes involved in their synthesis and removal have to be further considered.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Katia Cailliau
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| |
Collapse
|
10
|
Thiesler H, Küçükerden M, Gretenkort L, Röckle I, Hildebrandt H. News and Views on Polysialic Acid: From Tumor Progression and Brain Development to Psychiatric Disorders, Neurodegeneration, Myelin Repair and Immunomodulation. Front Cell Dev Biol 2022; 10:871757. [PMID: 35617589 PMCID: PMC9013797 DOI: 10.3389/fcell.2022.871757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/08/2022] [Indexed: 12/15/2022] Open
Abstract
Polysialic acid (polySia) is a sugar homopolymer consisting of at least eight glycosidically linked sialic acid units. It is a posttranslational modification of a limited number of proteins with the neural cell adhesion molecule NCAM being the most prominent. As extensively reviewed before, polySia-NCAM is crucial for brain development and synaptic plasticity but also modulates tumor growth and malignancy. Functions of polySia have been attributed to its polyanionic character, its spatial expansion into the extracellular space, and its modulation of NCAM interactions. In this mini-review, we first summarize briefly, how the modulation of NCAM functions by polySia impacts tumor cell growth and leads to malformations during brain development of polySia-deficient mice, with a focus on how the latter may be linked to altered behaviors in the mouse model and to neurodevelopmental predispositions to psychiatric disorders. We then elaborate on the implications of polySia functions in hippocampal plasticity, learning and memory of mice in light of recently described polySia changes related to altered neurogenesis in the aging human brain and in neurodegenerative disease. Furthermore, we highlight recent progress that extends the range of polySia functions across diverse fields of neurobiology such as cortical interneuron development and connectivity, myelination and myelin repair, or the regulation of microglia activity. We discuss possible common and distinct mechanisms that may underlie these seemingly divergent roles of polySia, and provide prospects for new therapeutic approaches building on our improved understanding of polySia functions.
Collapse
Affiliation(s)
| | | | | | | | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Villanueva-Cabello TM, Gutiérrez-Valenzuela LD, Salinas-Marín R, López-Guerrero DV, Martínez-Duncker I. Polysialic Acid in the Immune System. Front Immunol 2022; 12:823637. [PMID: 35222358 PMCID: PMC8873093 DOI: 10.3389/fimmu.2021.823637] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 12/28/2021] [Indexed: 01/01/2023] Open
Abstract
Polysialic acid (polySia) is a highly regulated polymer of sialic acid (Sia) with such potent biophysical characteristics that when expressed drastically influences the interaction properties of cells. Although much of what is known of polySia in mammals has been elucidated from the study of its role in the central nervous system (CNS), polySia is also expressed in other tissues, including the immune system where it presents dynamic changes during differentiation, maturation, and activation of different types of immune cells of the innate and adaptive response, being involved in key regulatory mechanisms. At least six polySia protein carriers (CCR7, ESL-1, NCAM, NRP2, ST8Sia 2, and ST8Sia 4) are expressed in different types of immune cells, but there is still much to be explored in regard not only to the regulatory mechanisms that determine their expression and the structure of polySia chains but also to the identification of the cis- and trans- ligands of polySia that establish signaling networks. This review summarizes the current knowledge on polySia in the immune system, addressing its biosynthesis, its tools for identification and structural characterization, and its functional roles and therapeutic implications.
Collapse
Affiliation(s)
- Tania M. Villanueva-Cabello
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Lya D. Gutiérrez-Valenzuela
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Roberta Salinas-Marín
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | | | - Iván Martínez-Duncker
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
- *Correspondence: Iván Martínez-Duncker,
| |
Collapse
|
12
|
Rivero O, Alhama-Riba J, Ku HP, Fischer M, Ortega G, Álmos P, Diouf D, van den Hove D, Lesch KP. Haploinsufficiency of the Attention-Deficit/Hyperactivity Disorder Risk Gene St3gal3 in Mice Causes Alterations in Cognition and Expression of Genes Involved in Myelination and Sialylation. Front Genet 2021; 12:688488. [PMID: 34650588 PMCID: PMC8505805 DOI: 10.3389/fgene.2021.688488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022] Open
Abstract
Genome wide association meta-analysis identified ST3GAL3, a gene encoding the beta-galactosidase-alpha-2,3-sialyltransferase-III, as a risk gene for attention-deficit/hyperactivity disorder (ADHD). Although loss-of-function mutations in ST3GAL3 are implicated in non-syndromic autosomal recessive intellectual disability (NSARID) and West syndrome, the impact of ST3GAL3 haploinsufficiency on brain function and the pathophysiology of neurodevelopmental disorders (NDDs), such as ADHD, is unknown. Since St3gal3 null mutant mice display severe developmental delay and neurological deficits, we investigated the effects of partial inactivation of St3gal3 in heterozygous (HET) knockout (St3gal3±) mice on behavior as well as expression of markers linked to myelination processes and sialylation pathways. Our results reveal that male St3gal3 HET mice display cognitive deficits, while female HET animals show increased activity, as well as increased cognitive control, compared to their wildtype littermates. In addition, we observed subtle alterations in the expression of several markers implicated in oligodendrogenesis, myelin formation, and protein sialylation as well as cell adhesion/synaptic target glycoproteins of ST3GAL3 in a brain region- and/or sex-specific manner. Taken together, our findings indicate that haploinsufficiency of ST3GAL3 results in a sex-dependent alteration of cognition, behavior and markers of brain plasticity.
Collapse
Affiliation(s)
- Olga Rivero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.,Biomedical Network Research Centre on Mental Health (CIBERSAM), Valencia, Spain
| | - Judit Alhama-Riba
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Hsing-Ping Ku
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Matthias Fischer
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Gabriela Ortega
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Péter Álmos
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - David Diouf
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Daniel van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
13
|
Jarahian M, Marofi F, Maashi MS, Ghaebi M, Khezri A, Berger MR. Re-Expression of Poly/Oligo-Sialylated Adhesion Molecules on the Surface of Tumor Cells Disrupts Their Interaction with Immune-Effector Cells and Contributes to Pathophysiological Immune Escape. Cancers (Basel) 2021; 13:5203. [PMID: 34680351 PMCID: PMC8534074 DOI: 10.3390/cancers13205203] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
Glycans linked to surface proteins are the most complex biological macromolecules that play an active role in various cellular mechanisms. This diversity is the basis of cell-cell interaction and communication, cell growth, cell migration, as well as co-stimulatory or inhibitory signaling. Our review describes the importance of neuraminic acid and its derivatives as recognition elements, which are located at the outermost positions of carbohydrate chains linked to specific glycoproteins or glycolipids. Tumor cells, especially from solid tumors, mask themselves by re-expression of hypersialylated neural cell adhesion molecule (NCAM), neuropilin-2 (NRP-2), or synaptic cell adhesion molecule 1 (SynCAM 1) in order to protect themselves against the cytotoxic attack of the also highly sialylated immune effector cells. More particularly, we focus on α-2,8-linked polysialic acid chains, which characterize carrier glycoproteins such as NCAM, NRP-2, or SynCam-1. This characteristic property correlates with an aggressive clinical phenotype and endows them with multiple roles in biological processes that underlie all steps of cancer progression, including regulation of cell-cell and/or cell-extracellular matrix interactions, as well as increased proliferation, migration, reduced apoptosis rate of tumor cells, angiogenesis, and metastasis. Specifically, re-expression of poly/oligo-sialylated adhesion molecules on the surface of tumor cells disrupts their interaction with immune-effector cells and contributes to pathophysiological immune escape. Further, sialylated glycoproteins induce immunoregulatory cytokines and growth factors through interactions with sialic acid-binding immunoglobulin-like lectins. We describe the processes, which modulate the interaction between sialylated carrier glycoproteins and their ligands, and illustrate that sialic acids could be targets of novel therapeutic strategies for treatment of cancer and immune diseases.
Collapse
Affiliation(s)
- Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran;
| | - Marwah Suliman Maashi
- Stem Cells and Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah 11211, Saudi Arabia;
| | - Mahnaz Ghaebi
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan 4513956184, Iran;
| | - Abdolrahman Khezri
- Department of Biotechnology, Inland Norway University of Applied Sciences, 2418 Hamar, Norway;
| | - Martin R. Berger
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| |
Collapse
|
14
|
Delgado C, Bu L, Zhang J, Liu FY, Sall J, Liang FX, Furley AJ, Fishman GI. Neural cell adhesion molecule is required for ventricular conduction system development. Development 2021; 148:269045. [PMID: 34100064 PMCID: PMC8217711 DOI: 10.1242/dev.199431] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/26/2021] [Indexed: 11/23/2022]
Abstract
The most distal portion of the ventricular conduction system (VCS) contains cardiac Purkinje cells (PCs), which are essential for synchronous activation of the ventricular myocardium. Contactin-2 (CNTN2), a member of the immunoglobulin superfamily of cell adhesion molecules (IgSF-CAMs), was previously identified as a marker of the VCS. Through differential transcriptional profiling, we discovered two additional highly enriched IgSF-CAMs in the VCS: NCAM-1 and ALCAM. Immunofluorescence staining showed dynamic expression patterns for each IgSF-CAM during embryonic and early postnatal stages, but ultimately all three proteins became highly enriched in mature PCs. Mice deficient in NCAM-1, but not CNTN2 or ALCAM, exhibited defects in PC gene expression and VCS patterning, as well as cardiac conduction disease. Moreover, using ST8sia2 and ST8sia4 knockout mice, we show that inhibition of post-translational modification of NCAM-1 by polysialic acid leads to disrupted trafficking of sarcolemmal intercalated disc proteins to junctional membranes and abnormal expansion of the extracellular space between apposing PCs. Taken together, our data provide insights into the complex developmental biology of the ventricular conduction system. Summary: The cell adhesion molecule NCAM-1 and its post-translational modification by polysialylation are required for normal formation and function of the specialized ventricular conduction system.
Collapse
Affiliation(s)
- Camila Delgado
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, NY 10016, USA
| | - Lei Bu
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, NY 10016, USA
| | - Jie Zhang
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, NY 10016, USA
| | - Fang-Yu Liu
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, NY 10016, USA
| | - Joseph Sall
- Microscopy Laboratory, Division of Advanced Research Technologies, NYU Langone Health, NY 10016, USA
| | - Feng-Xia Liang
- Microscopy Laboratory, Division of Advanced Research Technologies, NYU Langone Health, NY 10016, USA
| | - Andrew J Furley
- Department of Biomedical Science, The University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Glenn I Fishman
- Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, NY 10016, USA
| |
Collapse
|
15
|
Schmitt-Ulms G, Mehrabian M, Williams D, Ehsani S. The IDIP framework for assessing protein function and its application to the prion protein. Biol Rev Camb Philos Soc 2021; 96:1907-1932. [PMID: 33960099 DOI: 10.1111/brv.12731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 01/06/2023]
Abstract
The quest to determine the function of a protein can represent a profound challenge. Although this task is the mandate of countless research groups, a general framework for how it can be approached is conspicuously lacking. Moreover, even expectations for when the function of a protein can be considered to be 'known' are not well defined. In this review, we begin by introducing concepts pertinent to the challenge of protein function assignments. We then propose a framework for inferring a protein's function from four data categories: 'inheritance', 'distribution', 'interactions' and 'phenotypes' (IDIP). We document that the functions of proteins emerge at the intersection of inferences drawn from these data categories and emphasise the benefit of considering them in an evolutionary context. We then apply this approach to the cellular prion protein (PrPC ), well known for its central role in prion diseases, whose function continues to be considered elusive by many investigators. We document that available data converge on the conclusion that the function of the prion protein is to control a critical post-translational modification of the neural cell adhesion molecule in the context of epithelial-to-mesenchymal transition and related plasticity programmes. Finally, we argue that this proposed function of PrPC has already passed the test of time and is concordant with the IDIP framework in a way that other functions considered for this protein fail to achieve. We anticipate that the IDIP framework and the concepts analysed herein will aid the investigation of other proteins whose primary functional assignments have thus far been intractable.
Collapse
Affiliation(s)
- Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 0S8, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | | | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Sepehr Ehsani
- Theoretical and Philosophical Biology, Department of Philosophy, University College London, Bloomsbury, London, WC1E 6BT, U.K.,Ronin Institute for Independent Scholarship, Montclair, NJ, 07043, U.S.A
| |
Collapse
|
16
|
Oommen AM, Cunningham S, O'Súilleabháin PS, Hughes BM, Joshi L. An integrative network analysis framework for identifying molecular functions in complex disorders examining major depressive disorder as a test case. Sci Rep 2021; 11:9645. [PMID: 33958659 PMCID: PMC8102631 DOI: 10.1038/s41598-021-89040-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/14/2021] [Indexed: 12/02/2022] Open
Abstract
In addition to the psychological depressive phenotype, major depressive disorder (MDD) patients are also associated with underlying immune dysregulation that correlates with metabolic syndrome prevalent in depressive patients. A robust integrative analysis of biological pathways underlying the dysregulated neural connectivity and systemic inflammatory response will provide implications in the development of effective strategies for the diagnosis, management and the alleviation of associated comorbidities. In the current study, focusing on MDD, we explored an integrative network analysis methodology to analyze transcriptomic data combined with the meta-analysis of biomarker data available throughout public databases and published scientific peer-reviewed articles. Detailed gene set enrichment analysis and complex protein–protein, gene regulatory and biochemical pathway analysis has been undertaken to identify the functional significance and potential biomarker utility of differentially regulated genes, proteins and metabolite markers. This integrative analysis method provides insights into the molecular mechanisms along with key glycosylation dysregulation underlying altered neutrophil-platelet activation and dysregulated neuronal survival maintenance and synaptic functioning. Highlighting the significant gap that exists in the current literature, the network analysis framework proposed reduces the impact of data gaps and permits the identification of key molecular signatures underlying complex disorders with multiple etiologies such as within MDD and presents multiple treatment options to address their molecular dysfunction.
Collapse
Affiliation(s)
- Anup Mammen Oommen
- Advanced Glycoscience Research Cluster (AGRC), National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Stephen Cunningham
- Advanced Glycoscience Research Cluster (AGRC), National University of Ireland Galway, Galway, Ireland. .,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.
| | - Páraic S O'Súilleabháin
- Department of Psychology, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Brian M Hughes
- School of Psychology, National University of Ireland Galway, Galway, Ireland
| | - Lokesh Joshi
- Advanced Glycoscience Research Cluster (AGRC), National University of Ireland Galway, Galway, Ireland. .,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
17
|
Li Z, Lang Y, Liu L, Bunyatov MI, Sarmiento AI, de Groot RJ, Boons GJ. Synthetic O-acetylated sialosides facilitate functional receptor identification for human respiratory viruses. Nat Chem 2021; 13:496-503. [PMID: 33753916 DOI: 10.1038/s41557-021-00655-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 02/08/2021] [Indexed: 01/31/2023]
Abstract
The transmission of viruses from animal reservoirs to humans poses major threats to public health. Preparedness for future zoonotic outbreaks requires a fundamental understanding of how viruses of animal origin have adapted to binding to a cell surface component and/or receptor of the new host. Here we report on the specificities of human and animal viruses that engage with O-acetylated sialic acid, which include betacoronaviruses, toroviruses and influenza C and D viruses. Key to these studies was the development of a chemoenzymatic methodology that can provide almost any sialate-acetylation pattern. A collection of O-acetylated sialoglycans was printed as a microarray for the determination of receptor specificity. These studies showed host-specific patterns of receptor recognition and revealed that three distinct human respiratory viruses uniquely bind 9-O-acetylated α2,8-linked disialoside. Immunofluorescence and cell entry studies support that such a glycotope as part of a ganglioside is a functional receptor for human coronaviruses.
Collapse
Affiliation(s)
- Zeshi Li
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Yifei Lang
- Virology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Mehman I Bunyatov
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Angelic Isaza Sarmiento
- Virology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Raoul J de Groot
- Virology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands. .,Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA. .,Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands. .,Chemistry Department, University of Georgia, Athens, GA, USA.
| |
Collapse
|
18
|
Fu W, Yu G, Liang J, Fan P, Dong K, Zhang B, Chen X, Zhu H, Chu L. miR-144-5p and miR-451a Inhibit the Growth of Cholangiocarcinoma Cells Through Decreasing the Expression of ST8SIA4. Front Oncol 2021; 10:563486. [PMID: 33520692 PMCID: PMC7841262 DOI: 10.3389/fonc.2020.563486] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidences indicate that non-coding RNAs play crucial roles in the progression of an extensive range of carcinomas. This study aimed to investigate the action mechanism of miR-144-5p and miR-451a in cholangiocarcinoma. We found that miR-144-5p and miR-451a were significantly decreased in cholangiocarcinoma patient samples compared to the adjacent normal bile duct samples. The downregulation of these two miRNAs was correlated with a more advanced disease state of cholangiocarcinoma patients. Overexpression of miR-144-5p and miR-451a suppressed the proliferation, invasion and migration of cholangiocarcinoma cells in vitro and inhibited xenograft tumor growth. Knockdown of these two miRNAs had the opposite effects. miR-144-5p and miR-451a regulated the expression of ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 4 (ST8SIA4), and presented a correlation with ST8SIA4 in patient samples. Overexpression of ST8SIA4 promoted the proliferation, invasion and migration of cholangiocarcinoma cells, and the changes were reversed by upregulating the expression of miR-144-5p and miR-451a. Our findings indicated that miR-144-5p and miR-451a displayed a tumor suppressor role through decreasing the expression of ST8SIA4 in cholangiocarcinoma.
Collapse
Affiliation(s)
- Wan Fu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangcai Yu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Fan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Keshuai Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Giacometti LL, Huang F, Hamilton BS, Barker JM. Brain region-dependent alterations in polysialic acid immunoreactivity across the estrous cycle in mice. Horm Behav 2020; 126:104851. [PMID: 32941849 PMCID: PMC7725886 DOI: 10.1016/j.yhbeh.2020.104851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/13/2020] [Accepted: 09/03/2020] [Indexed: 11/27/2022]
Abstract
N-glycosylation is a posttranslational modification that plays significant roles in regulating protein function. One form of N-glycosylation, polysialylation, has been implicated in many processes including learning and memory, addiction, and neurodegenerative disease. Polysialylation appears to be modulated by the estrous cycle in the hypothalamus in rat, but this has not been assessed in other brain regions. To determine if polysialylation was similarly estrous phase-dependent in other neuroanatomical structures, the percent area of polysialic acid (PSA) immunoreactivity in subregions of the medial prefrontal cortex, hippocampus, and nucleus accumbens was assessed in each of the four phases in adult female mice. In this study, we found that PSA immunoreactivity fluctuated across the estrous cycle in a subregion-specific manner. In the prefrontal cortex, PSA immunoreactivity was significantly lower in proestrus phase compared to estrus in the prelimbic cortex, but did not differ across the estrous cycle in the infralimbic cortex. In the hippocampus, PSA immunoreactivity was significantly increased in proestrus compared to metestrus in the CA1 and CA2 and compared to diestrus in CA3, but remain unchanged in the dentate gyrus. PSA immunoreactivity did not vary across the estrous cycle in the nucleus accumbens core or shell. These findings may have implications for estrous cycle-dependent alterations in behavior.
Collapse
Affiliation(s)
- Laura L Giacometti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Fangyi Huang
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Brianna S Hamilton
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America
| | - Jacqueline M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, United States of America.
| |
Collapse
|
20
|
Polysialic acid and Siglec-E orchestrate negative feedback regulation of microglia activation. Cell Mol Life Sci 2020; 78:1637-1653. [PMID: 32725371 PMCID: PMC7904730 DOI: 10.1007/s00018-020-03601-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 06/23/2020] [Accepted: 07/17/2020] [Indexed: 11/09/2022]
Abstract
Polysialic acid (polySia) emerges as a novel regulator of microglia activity. We recently identified polysialylated proteins in the Golgi compartment of murine microglia that are released in response to inflammatory stimulation. Since exogenously added polySia is able to attenuate the inflammatory response, we proposed that the release of polysialylated proteins constitutes a mechanism for negative feedback regulation of microglia activation. Here, we demonstrate that translocation of polySia from the Golgi to the cell surface can be induced by calcium depletion of the Golgi compartment and that polysialylated proteins are continuously released for at least 24 h after the onset of inflammatory stimulation. The latter was unexpected, because polySia signals detected by immunocytochemistry are rapidly depleted. However, it indicates that the amount of released polySia is much higher than anticipated based on immunostaining. This may be crucial for microglial responses during traumatic brain injury (TBI), as we detected polySia signals in activated microglia around a stab wound in the adult mouse brain. In BV2 microglia, the putative polySia receptor Siglec-E is internalized during lipopolysaccharide (LPS)-induced activation and in response to polySia exposure, indicating interaction. Correspondingly, CRISPR/Cas9-mediated Siglec-E knockout prevents inhibition of pro inflammatory activation by exogenously added polySia and leads to a strong increase of the LPS response. A comparable increase of LPS-induced activation has been observed in microglia with abolished polySia synthesis. Together, these results indicate that the release of the microglia-intrinsic polySia pool, as implicated in TBI, inhibits the inflammatory response by acting as a trans-activating ligand of Siglec-E.
Collapse
|
21
|
Neurons, Glia, Extracellular Matrix and Neurovascular Unit: A Systems Biology Approach to the Complexity of Synaptic Plasticity in Health and Disease. Int J Mol Sci 2020; 21:ijms21041539. [PMID: 32102370 PMCID: PMC7073232 DOI: 10.3390/ijms21041539] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023] Open
Abstract
The synaptic cleft has been vastly investigated in the last decades, leading to a novel and fascinating model of the functional and structural modifications linked to synaptic transmission and brain processing. The classic neurocentric model encompassing the neuronal pre- and post-synaptic terminals partly explains the fine-tuned plastic modifications under both pathological and physiological circumstances. Recent experimental evidence has incontrovertibly added oligodendrocytes, astrocytes, and microglia as pivotal elements for synapse formation and remodeling (tripartite synapse) in both the developing and adult brain. Moreover, synaptic plasticity and its pathological counterpart (maladaptive plasticity) have shown a deep connection with other molecular elements of the extracellular matrix (ECM), once considered as a mere extracellular structural scaffold altogether with the cellular glue (i.e., glia). The ECM adds another level of complexity to the modern model of the synapse, particularly, for the long-term plasticity and circuit maintenance. This model, called tetrapartite synapse, can be further implemented by including the neurovascular unit (NVU) and the immune system. Although they were considered so far as tightly separated from the central nervous system (CNS) plasticity, at least in physiological conditions, recent evidence endorsed these elements as structural and paramount actors in synaptic plasticity. This scenario is, as far as speculations and evidence have shown, a consistent model for both adaptive and maladaptive plasticity. However, a comprehensive understanding of brain processes and circuitry complexity is still lacking. Here we propose that a better interpretation of the CNS complexity can be granted by a systems biology approach through the construction of predictive molecular models that enable to enlighten the regulatory logic of the complex molecular networks underlying brain function in health and disease, thus opening the way to more effective treatments.
Collapse
|
22
|
Zhou GP, Liao SM, Chen D, Huang RB. The Cooperative Effect between Polybasic Region (PBR) and Polysialyltransferase Domain (PSTD) within Tumor-Target Polysialyltranseferase ST8Sia II. Curr Top Med Chem 2020; 19:2831-2841. [PMID: 31755393 DOI: 10.2174/1568026619666191121145924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/16/2019] [Accepted: 10/20/2019] [Indexed: 12/29/2022]
Abstract
ST8Sia II (STX) is a highly homologous mammalian polysialyltransferase (polyST), which is a validated tumor-target in the treatment of cancer metastasis reliant on tumor cell polysialylation. PolyST catalyzes the synthesis of α2,8-polysialic acid (polySia) glycans by carrying out the activated CMP-Neu5Ac (Sia) to N- and O-linked oligosaccharide chains on acceptor glycoproteins. In this review article, we summarized the recent studies about intrinsic correlation of two polybasic domains, Polysialyltransferase domain (PSTD) and Polybasic region (PBR) within ST8Sia II molecule, and suggested that the critical amino acid residues within the PSTD and PBR motifs of ST8Sia II for polysialylation of Neural cell adhesion molecules (NCAM) are related to ST8Sia II activity. In addition, the conformational changes of the PSTD domain due to point mutations in the PBR or PSTD domain verified an intramolecular interaction between the PBR and the PSTD. These findings have been incorporated into Zhou's NCAM polysialylation/cell migration model, which will provide new perspectives on drug research and development related to the tumor-target ST8Sia II.
Collapse
Affiliation(s)
- Guo-Ping Zhou
- National Engineering Research Center for Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Key Laboratory of Bio-refinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning, 530007, China.,Gordon Life Science Institute, NC 27804, United States
| | - Si-Ming Liao
- National Engineering Research Center for Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Key Laboratory of Bio-refinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning, 530007, China
| | - Dong Chen
- National Engineering Research Center for Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Key Laboratory of Bio-refinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning, 530007, China
| | - Ri-Bo Huang
- National Engineering Research Center for Non-Food Biorefinery, State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Key Laboratory of Bio-refinery, Guangxi Academy of Sciences, 98 Daling Road, Nanning, 530007, China
| |
Collapse
|
23
|
Janesch B, Baumann L, Mark A, Thompson N, Rahmani S, Sim L, Withers SG, Wakarchuk WW. Directed evolution of bacterial polysialyltransferases. Glycobiology 2020; 29:588-598. [PMID: 30976781 DOI: 10.1093/glycob/cwz021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/15/2019] [Accepted: 03/18/2019] [Indexed: 01/30/2023] Open
Abstract
Polysialyltransferases (polySTs) are glycosyltransferases that synthesize polymers of sialic acid found in vertebrates and some bacterial pathogens. Bacterial polySTs have utility in the modification of therapeutic proteins to improve serum half-life, and the potential for tissue engineering. PolySTs are membrane-associated proteins and as recombinant proteins suffer from inherently low solubility, low expression levels and poor thermal stability. To improve their physicochemical and biochemical properties, we applied a directed evolution approach using a FACS-based ultrahigh-throughput assay as a simple, robust and reliable screening method. We were able to enrich a large mutant library and, in combination with plate-based high-throughput secondary screening, we discovered mutants with increased enzymatic activity and improved stability compared to the wildtype enzyme. This work presents a powerful strategy for the screening of directed evolution libraries of bacterial polySTs to identify better catalysts for in vitro polysialylation of therapeutics.
Collapse
Affiliation(s)
- Bettina Janesch
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Lars Baumann
- Departments of Chemistry and Biochemistry, and Michael Smith Laboratory, University of British Columbia, Vancouver, BC, Canada
| | - Alison Mark
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Nicole Thompson
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Sadia Rahmani
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| | - Lyann Sim
- Departments of Chemistry and Biochemistry, and Michael Smith Laboratory, University of British Columbia, Vancouver, BC, Canada
| | - Stephen G Withers
- Departments of Chemistry and Biochemistry, and Michael Smith Laboratory, University of British Columbia, Vancouver, BC, Canada
| | - Warren W Wakarchuk
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
| |
Collapse
|
24
|
Zhou X, Yang G, Guan F. Biological Functions and Analytical Strategies of Sialic Acids in Tumor. Cells 2020; 9:E273. [PMID: 31979120 PMCID: PMC7072699 DOI: 10.3390/cells9020273] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/21/2022] Open
Abstract
Sialic acids, a subset of nine carbon acidic sugars, often exist as the terminal sugars of glycans on either glycoproteins or glycolipids on the cell surface. Sialic acids play important roles in many physiological and pathological processes via carbohydrate-protein interactions, including cell-cell communication, bacterial and viral infections. In particular, hypersialylation in tumors, as well as their roles in tumor growth and metastasis, have been widely described. Recent studies have indicated that the aberrant sialylation is a vital way for tumor cells to escape immune surveillance and keep malignance. In this article, we outline the present state of knowledge on the metabolic pathway of human sialic acids, the function of hypersialylation in tumors, as well as the recent labeling and analytical techniques for sialic acids. It is expected to offer a brief introduction of sialic acid metabolism and provide advanced analytical strategies in sialic acid studies.
Collapse
Affiliation(s)
- Xiaoman Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Ganglong Yang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Feng Guan
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, College of Life Science, Northwest University, Xi’an 710069, China
| |
Collapse
|
25
|
Kranaster P, Karreman C, Dold JEGA, Krebs A, Funke M, Holzer AK, Klima S, Nyffeler J, Helfrich S, Wittmann V, Leist M. Time and space-resolved quantification of plasma membrane sialylation for measurements of cell function and neurotoxicity. Arch Toxicol 2019; 94:449-467. [PMID: 31828357 DOI: 10.1007/s00204-019-02642-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/02/2019] [Indexed: 12/28/2022]
Abstract
While there are many methods to quantify the synthesis, localization, and pool sizes of proteins and DNA during physiological responses and toxicological stress, only few approaches allow following the fate of carbohydrates. One of them is metabolic glycoengineering (MGE), which makes use of chemically modified sugars (CMS) that enter the cellular biosynthesis pathways leading to glycoproteins and glycolipids. The CMS can subsequently be coupled (via bio-orthogonal chemical reactions) to tags that are quantifiable by microscopic imaging. We asked here, whether MGE can be used in a quantitative and time-resolved way to study neuronal glycoprotein synthesis and its impairment. We focused on the detection of sialic acid (Sia), by feeding human neurons the biosynthetic precursor N-acetyl-mannosamine, modified by an azide tag. Using this system, we identified non-toxic conditions that allowed live cell labeling with high spatial and temporal resolution, as well as the quantification of cell surface Sia. Using combinations of immunostaining, chromatography, and western blotting, we quantified the percentage of cellular label incorporation and effects on glycoproteins such as polysialylated neural cell adhesion molecule. A specific imaging algorithm was used to quantify Sia incorporation into neuronal projections, as potential measure of complex cell function in toxicological studies. When various toxicants were studied, we identified a subgroup (mitochondrial respiration inhibitors) that affected neurite glycan levels several hours before any other viability parameter was affected. The MGE-based neurotoxicity assay, thus allowed the identification of subtle impairments of neurochemical function with very high sensitivity.
Collapse
Affiliation(s)
- Petra Kranaster
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, 78457, Konstanz, Germany
| | - Christiaan Karreman
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Jeremias E G A Dold
- Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, 78457, Konstanz, Germany.,Department of Chemistry, University of Konstanz, 78457, Konstanz, Germany
| | - Alice Krebs
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, 78457, Konstanz, Germany
| | - Melina Funke
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Anna-Katharina Holzer
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany
| | - Stefanie Klima
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany.,Kooperatives Promotionskolleg (KPK) InViTe, University of Konstanz, 78457, Konstanz, Germany
| | - Johanna Nyffeler
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany.,Environmental Protection Agency, Durham, NC, USA
| | - Stefan Helfrich
- The Bioimaging Center, University of Konstanz, 78457, Konstanz, Germany.,KNIME GmbH, 78467, Konstanz, Germany
| | - Valentin Wittmann
- Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, 78457, Konstanz, Germany.,Department of Chemistry, University of Konstanz, 78457, Konstanz, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457, Konstanz, Germany. .,Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, 78457, Konstanz, Germany.
| |
Collapse
|
26
|
Emerging Roles of Synapse Organizers in the Regulation of Critical Periods. Neural Plast 2019; 2019:1538137. [PMID: 31565044 PMCID: PMC6745111 DOI: 10.1155/2019/1538137] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 01/10/2023] Open
Abstract
Experience remodels cortical connectivity during developmental windows called critical periods. Experience-dependent regulation of synaptic strength during these periods establishes circuit functions that are stabilized as critical period plasticity wanes. These processes have been extensively studied in the developing visual cortex, where critical period opening and closure are orchestrated by the assembly, maturation, and strengthening of distinct synapse types. The synaptic specificity of these processes points towards the involvement of distinct molecular pathways. Attractive candidates are pre- and postsynaptic transmembrane proteins that form adhesive complexes across the synaptic cleft. These synapse-organizing proteins control synapse development and maintenance and modulate structural and functional properties of synapses. Recent evidence suggests that they have pivotal roles in the onset and closure of the critical period for vision. In this review, we describe roles of synapse-organizing adhesion molecules in the regulation of visual critical period plasticity and we discuss the potential they offer to restore circuit functions in amblyopia and other neurodevelopmental disorders.
Collapse
|
27
|
Fewou SN, Röckle I, Hildebrandt H, Eckhardt M. Transgenic overexpression of polysialyltransferase ST8SiaIV under the control of a neuron-specific promoter does not affect brain development but impairs exploratory behavior. Glycobiology 2019; 29:657-668. [PMID: 31147692 PMCID: PMC6704368 DOI: 10.1093/glycob/cwz040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/15/2019] [Accepted: 05/21/2019] [Indexed: 11/12/2022] Open
Abstract
A large body of the literature has demonstrated that the polysialic acid (polySia) modification of the neural cell adhesion molecule (NCAM) is a key regulator of cellular interactions during brain development, maintenance and plasticity. To properly fulfill these functions, polySia concentration has to be carefully controlled. This is done by the regulation of the expression of the two polySia-synthesizing enzymes ST8SiaII and ST8SiaIV. From this point of view we and others have demonstrated that downregulation of ST8SiaIV during oligodendrocyte differentiation is a prerequisite for efficient myelin formation and maintenance. Here, we addressed the question whether the prevention of polySia downregulation in neurons affects brain and particularly myelin development and functioning. For this purpose, we developed transgenic (tg) mouse lines overexpressing the polysialyltransferase ST8SiaIV in neurons. tg expression of ST8SiaIV prevented the postnatal downregulation of polySia, and most of the polySias in the forebrain and brain stem of adult tg mice were associated with NCAM-140 and NCAM-180 isoforms. Structural examination of the brain revealed no overt abnormalities of axons and myelin. In addition, ultrastructural and western blot analyses indicated normal myelin development. However, behavioral studies revealed reduced rearing activity, a measure for exploratory behavior, while parameters of motor activity were not affected in tg mice. Taken together, these results suggest that a persisting presence of polySia in neurons has no major effect on brain structure, myelination and myelin maintenance, but causes mild behavioral changes.
Collapse
Affiliation(s)
- Simon Ngamli Fewou
- Institut für Biochemie und Molekularbiologie, Universität Bonn, Bonn, Germany.,Institut für Klinische Biochemie, Medizinische Hochschule Hannover, Hannover, Germany.,Faculty of Health Sciences, Université des Montagnes, Bangangte, Cameroon
| | - Iris Röckle
- Institut für Klinische Biochemie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Herbert Hildebrandt
- Institut für Klinische Biochemie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Matthias Eckhardt
- Institut für Biochemie und Molekularbiologie, Universität Bonn, Bonn, Germany
| |
Collapse
|
28
|
Guo X, Elkashef SM, Loadman PM, Patterson LH, Falconer RA. Recent advances in the analysis of polysialic acid from complex biological systems. Carbohydr Polym 2019; 224:115145. [PMID: 31472857 DOI: 10.1016/j.carbpol.2019.115145] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/25/2022]
Abstract
Polysialic acid (polySia) is a unique, well-characterised carbohydrate polymer highly-expressed on the cell surface of neurons in the early stages of mammalian brain development. Post-embryogenesis, it is also re-expressed in a number of tumours of neuroendocrine origin. It plays important roles in modulating cell-cell, and cell-matrix adhesion and migration, tumour invasion and metastasis. Techniques for structural and quantitative characterisation of polySia from tumours and cancer cells are thus essential in exploring the relationship between polySia expression levels and structural and functional changes associated with cancer progression and metastasis. A variety of techniques have been developed to structurally and quantitatively analyse polySia in clinical tissues and other biological samples. In this review, analytical approaches used for the determination of polySia in biological matrices in the past 20 years are discussed, with a particular focus on chemical approaches, and quantitative analysis.
Collapse
Affiliation(s)
- Xiaoxiao Guo
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Sara M Elkashef
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Paul M Loadman
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Laurence H Patterson
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom
| | - Robert A Falconer
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, United Kingdom.
| |
Collapse
|
29
|
Li F, Ding J. Sialylation is involved in cell fate decision during development, reprogramming and cancer progression. Protein Cell 2019; 10:550-565. [PMID: 30478534 PMCID: PMC6626595 DOI: 10.1007/s13238-018-0597-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/31/2018] [Indexed: 01/01/2023] Open
Abstract
Sialylation, or the covalent addition of sialic acid to the terminal end of glycoproteins, is a biologically important modification that is involved in embryonic development, neurodevelopment, reprogramming, oncogenesis and immune responses. In this review, we have given a comprehensive overview of the current literature on the involvement of sialylation in cell fate decision during development, reprogramming and cancer progression. Sialylation is essential for early embryonic development and the deletion of UDP-GlcNAc 2-epimerase, a rate-limiting enzyme in sialic acid biosynthesis, is embryonically lethal. Furthermore, the sialyltransferase ST6GAL1 is required for somatic cell reprogramming, and its downregulation is associated with decreased reprogramming efficiency. In addition, sialylation levels and patterns are altered during cancer progression, indicating the potential of sialylated molecules as cancer biomarkers. Taken together, the current evidences demonstrate that sialylation is involved in crucial cell fate decision.
Collapse
Affiliation(s)
- Fenjie Li
- Program in Stem Cell and Regenerative Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Department of Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Junjun Ding
- Program in Stem Cell and Regenerative Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Department of Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
30
|
Yang C, Zhang P, Fang W, Chen Y, Zhang N, Qiao Z, Troy FA, Wang B. Molecular Mechanisms Underlying How Sialyllactose Intervention Promotes Intestinal Maturity by Upregulating GDNF Through a CREB-Dependent Pathway in Neonatal Piglets. Mol Neurobiol 2019; 56:7994-8007. [PMID: 31161424 DOI: 10.1007/s12035-019-1628-9] [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: 11/08/2018] [Accepted: 04/26/2019] [Indexed: 12/23/2022]
Abstract
Sialylated milk oligosaccharides (SMOs) have a multifunctional health benefit, yet the molecular details underlying their potential role in modulating intestinal maturation remains unknown. To test the hypothesis that sialyllactose (SL) may mediate intestinal maturation and function through controlling neuronal function, studies were carried out where the diet of postnatal piglets was supplemented with a mixture of 3'- and 6'-sialyllactose from postnatal day 3 to 38. Gene transcription pathways regulating enteric nervous system function, polysialic acid (polySia) synthesis, and cell proliferation were quantified. Our new findings show that SL intervention: (1) upregulated the level of gene and protein expression of the glial-derived neurotrophic factor (GDNF) in the ileum; (2) upregulated phosphorylation of the cAMP responsive element-binding protein (CREB), the downstream target of GDNF signaling pathway; (3) promoted cell proliferation based on an increase in the number and density of Ki-67 positive cells in the crypts; (4) increased the crypt width in the ileum by 10%, while gene markers for the functional cells were not affected; (5) upregulated mRNA expression level of ST8Sia IV, a key polysialyltransferase responsible for synthesis of polySia-NCAM; (6) reduced the incidence and severity of diarrhea. These results show that SL promotes intestinal maturation in neonatal piglets by upregulating GDNF, synthesis of polySia and CREB-interactive pathway.
Collapse
Affiliation(s)
- Changwei Yang
- Medical College of Xiamen University, Xiamen City, 361005, China.,School of Public Health & Management, Wenzhou Medical University, Wenzhou, 325035, China
| | - Panwang Zhang
- Medical College of Xiamen University, Xiamen City, 361005, China
| | - Wang Fang
- Medical College of Xiamen University, Xiamen City, 361005, China
| | - Yue Chen
- Medical College of Xiamen University, Xiamen City, 361005, China
| | - Nai Zhang
- Medical College of Xiamen University, Xiamen City, 361005, China
| | - Zhiliang Qiao
- Medical College of Xiamen University, Xiamen City, 361005, China
| | - Frederic A Troy
- Medical College of Xiamen University, Xiamen City, 361005, China. .,Department of Biochemistry & Molecular Medicine, University of California School of Medicine, Davis, CA, 95616, USA.
| | - Bing Wang
- Medical College of Xiamen University, Xiamen City, 361005, China. .,Graham Centre for Agricultural Innovation, School of Animal & Veterinary Science, Charles Sturt University, Wagga Wagga 2678, Australia.
| |
Collapse
|
31
|
Villanueva-Cabello TM, Gutiérrez-Valenzuela LD, López-Guerrero DV, Cruz-Muñoz ME, Mora-Montes HM, Martínez-Duncker I. Polysialic acid is expressed in human naïve CD4+ T cells and is involved in modulating activation. Glycobiology 2019; 29:557-564. [DOI: 10.1093/glycob/cwz032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 03/28/2019] [Accepted: 04/13/2019] [Indexed: 02/01/2023] Open
Affiliation(s)
- Tania M Villanueva-Cabello
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mor, México
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor, México
| | - Lya D Gutiérrez-Valenzuela
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mor, México
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor, México
| | - Delia V López-Guerrero
- Facultad de Nutrición, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mor, México
| | - Mario E Cruz-Muñoz
- Laboratorio de Inmunología Molecular, Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mor, México
| | | | - Iván Martínez-Duncker
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mor, México
| |
Collapse
|
32
|
Semenyuk P, Muronetz V. Protein Interaction with Charged Macromolecules: From Model Polymers to Unfolded Proteins and Post-Translational Modifications. Int J Mol Sci 2019; 20:E1252. [PMID: 30871103 PMCID: PMC6429204 DOI: 10.3390/ijms20051252] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 12/18/2022] Open
Abstract
Interaction of proteins with charged macromolecules is involved in many processes in cells. Firstly, there are many naturally occurred charged polymers such as DNA and RNA, polyphosphates, sulfated glycosaminoglycans, etc., as well as pronouncedly charged proteins such as histones or actin. Electrostatic interactions are also important for "generic" proteins, which are not generally considered as polyanions or polycations. Finally, protein behavior can be altered due to post-translational modifications such as phosphorylation, sulfation, and glycation, which change a local charge of the protein region. Herein we review molecular modeling for the investigation of such interactions, from model polyanions and polycations to unfolded proteins. We will show that electrostatic interactions are ubiquitous, and molecular dynamics simulations provide an outstanding opportunity to look inside binding and reveal the contribution of electrostatic interactions. Since a molecular dynamics simulation is only a model, we will comprehensively consider its relationship with the experimental data.
Collapse
Affiliation(s)
- Pavel Semenyuk
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia.
| | - Vladimir Muronetz
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234 Moscow, Russia.
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia.
| |
Collapse
|
33
|
Kühnle A, Veelken R, Galuska CE, Saftenberger M, Verleih M, Schuppe HC, Rudloff S, Kunz C, Galuska SP. Polysialic acid interacts with lactoferrin and supports its activity to inhibit the release of neutrophil extracellular traps. Carbohydr Polym 2019; 208:32-41. [DOI: 10.1016/j.carbpol.2018.12.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 01/13/2023]
|
34
|
Curto Y, Alcaide J, Röckle I, Hildebrandt H, Nacher J. Effects of the Genetic Depletion of Polysialyltransferases on the Structure and Connectivity of Interneurons in the Adult Prefrontal Cortex. Front Neuroanat 2019; 13:6. [PMID: 30787870 PMCID: PMC6372547 DOI: 10.3389/fnana.2019.00006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/21/2019] [Indexed: 12/29/2022] Open
Abstract
Polysialic acid (polySia) is a complex sugar that in the nervous system appears mainly as a posttranslational modification of the neural cell adhesion molecule (NCAM). PolySia plays important roles during brain development, but also in its plasticity during adulthood. Two polysialyltransferases (polyST), ST8SIA2 and ST8SIA4, are involved in the synthesis and attachment of polySia. Both polyST are relevant for developmental migration of cortical interneurons and their establishment in the prefrontal cortex (PFC). In contrast, only ST8SIA4 appears to be important for the structural plasticity of a subpopulation of cortical interneurons in the adult. Interestingly, ST8SIA2 and NCAM are candidate genes for schizophrenia, a disorder in which interneuronal circuits are altered. However, there is still no data on the effects of polyST depletion on the dendritic structure or the connectivity of cortical interneurons. Here, we studied the contribution of each polyST on these parameters in the medial PFC (mPFC) of polyST knock-out mice with GAD67-GFP-labeled interneurons. Genetic depletion of ST8SIA4, but not ST8SIA2, resulted in a decrease in the complexity of the dendritic arbor of interneurons. In contrast, ablation of either of the two polyST induced a decrease in the density of parvalbumin (PV) expressing perisomatic puncta on pyramidal neurons. Thus, the depletion of each polyST results in similar impairments of not only developmental migration but also efferent synaptic connectivity of interneurons. In contrast, the loss of ST8SIA4 has a unique effect on dendritic structure, hence on afferent connectivity, suggesting differential and independent contributions of each polyST to neuritogenesis and synaptogenesis.
Collapse
Affiliation(s)
- Yasmina Curto
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, Spain
| | - Julia Alcaide
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, Spain
| | - Iris Röckle
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Juan Nacher
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM): Spanish National Network for Research in Mental Health, Madrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
35
|
Volkers G, Lizak C, Niesser J, Rosell FI, Preidl J, Gnanapragassam VS, Horstkorte R, Rademann J, Strynadka NCJ. Structural Basis for Binding of Fluorescent CMP-Neu5Ac Mimetics to Enzymes of the Human ST8Sia Family. ACS Chem Biol 2018; 13:2320-2328. [PMID: 30015474 DOI: 10.1021/acschembio.8b00478] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Polysialyltransferases synthesize polysialic acid on cell surface-expressed glycoconjugates, which is crucial for developing processes and signaling pathways in eukaryotes. Recent advances in cancer research have rendered polysialyltransferases important drug targets because polysialic acid contributes to cancer cell progression, metastasis, and treatment of resistant tumors. To aid the development of high-throughput screening assays for polysialyltransferase inhibitors, we demonstrate that a previously developed class of fluorescent CMP-sialic acid mimetics for sialyltransferases has nanomolar affinities for oligo- and polysialyltransferases and can be used for the rapid screening of new polysialyltransferase inhibitors. We demonstrate that these CMP-Neu5Ac mimetics inhibit polysialylation in vitro and perform cell culture experiments, where we observe reduced polysialylation of NCAM. Furthermore, we describe the structural basis of CMP-Neu5Ac mimetics binding to the human oligosialyltransferase ST8SiaIII and extrapolate why their affinity is high for human polysialyltransferases. Our results show that this novel class of compounds is a promising tool for the development of potent and selective drugs against polysialyltransferase activity.
Collapse
Affiliation(s)
- Gesa Volkers
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Christian Lizak
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Jürgen Niesser
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Frederico I. Rosell
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Johannes Preidl
- Institute of Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany
| | - Vinayaga S. Gnanapragassam
- Institute for Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Hollystraße 1, D-06114 Halle (Saale), Germany
| | - Ruediger Horstkorte
- Institute for Physiological Chemistry, Martin-Luther-University Halle-Wittenberg, Hollystraße 1, D-06114 Halle (Saale), Germany
| | - Jörg Rademann
- Institute of Pharmacy, Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany
| | - Natalie C. J. Strynadka
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
36
|
Immormino RM, Lauzier DC, Nakano H, Hernandez ML, Alexis NE, Ghio AJ, Tilley SL, Doerschuk CM, Peden DB, Cook DN, Moran TP. Neuropilin-2 regulates airway inflammatory responses to inhaled lipopolysaccharide. Am J Physiol Lung Cell Mol Physiol 2018; 315:L202-L211. [PMID: 29671604 PMCID: PMC6139664 DOI: 10.1152/ajplung.00067.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Neuropilins are multifunctional receptors that play important roles in immune regulation. Neuropilin-2 (NRP2) is expressed in the lungs, but whether it regulates airway immune responses is unknown. Here, we report that Nrp2 is weakly expressed by alveolar macrophages (AMs) in the steady state but is dramatically upregulated following in vivo lipopolysaccharide (LPS) inhalation. Ex vivo treatment of human AMs with LPS also increased NRP2 mRNA expression and cell-surface display of NRP2 protein. LPS-induced Nrp2 expression in AMs was dependent upon the myeloid differentiation primary response 88 signaling pathway and the transcription factor NF-κB. In addition to upregulating display of NRP2 on the cell membrane, inhaled LPS also triggered AMs to release soluble NRP2 into the airways. Finally, myeloid-specific ablation of NRP2 resulted in increased expression of the chemokine (C-C motif) ligand 2 ( Ccl2) in the lungs and prolonged leukocyte infiltration in the airways following LPS inhalation. These findings suggest that NRP2 expression by AMs regulates LPS-induced inflammatory cell recruitment to the airways and reveal a novel role for NRP2 during innate immune responses in the lungs.
Collapse
Affiliation(s)
- Robert M Immormino
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina , Chapel Hill, North Carolina
| | - David C Lauzier
- Department of Pediatrics, University of North Carolina , Chapel Hill, North Carolina
| | - Hideki Nakano
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park, North Carolina
| | - Michelle L Hernandez
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina , Chapel Hill, North Carolina.,Department of Pediatrics, University of North Carolina , Chapel Hill, North Carolina
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina , Chapel Hill, North Carolina.,Department of Pediatrics, University of North Carolina , Chapel Hill, North Carolina
| | - Andrew J Ghio
- National Health and Environmental Effects Research Laboratory, Environmental Protection Agency , Chapel Hill, North Carolina
| | - Stephen L Tilley
- Department of Medicine, University of North Carolina , Chapel Hill, North Carolina
| | - Claire M Doerschuk
- Department of Medicine, University of North Carolina , Chapel Hill, North Carolina
| | - David B Peden
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina , Chapel Hill, North Carolina.,Department of Pediatrics, University of North Carolina , Chapel Hill, North Carolina
| | - Donald N Cook
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park, North Carolina
| | - Timothy P Moran
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina , Chapel Hill, North Carolina.,Department of Pediatrics, University of North Carolina , Chapel Hill, North Carolina
| |
Collapse
|
37
|
Keys TG, Wetter M, Hang I, Rutschmann C, Russo S, Mally M, Steffen M, Zuppiger M, Müller F, Schneider J, Faridmoayer A, Lin CW, Aebi M. A biosynthetic route for polysialylating proteins in Escherichia coli. Metab Eng 2017; 44:293-301. [PMID: 29101090 DOI: 10.1016/j.ymben.2017.10.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/12/2017] [Accepted: 10/27/2017] [Indexed: 01/08/2023]
Abstract
Polysialic acid (polySia) is a posttranslational modification found on only a handful of proteins in the central nervous and immune systems. The addition of polySia to therapeutic proteins improves pharmacokinetics and reduces immunogenicity. To date, polysialylation of therapeutic proteins has only been achieved in vitro by chemical or chemoenzymatic strategies. In this work, we develop a biosynthetic pathway for site-specific polysialylation of recombinant proteins in the cytoplasm of Escherichia coli. The pathway takes advantage of a bacterial cytoplasmic polypeptide-glycosyltransferase to establish a site-specific primer on the target protein. The glucose primer is extended by glycosyltransferases derived from lipooligosaccharide, lipopolysaccharide and capsular polysaccharide biosynthesis from different bacterial species to synthesize long chain polySia. We demonstrate the new biosynthetic route by modifying green fluorescent proteins and a therapeutic DARPin (designed ankyrin repeat protein).
Collapse
Affiliation(s)
- Timothy G Keys
- Institute of Microbiology, Department of Biology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | | | - Ivan Hang
- Institute of Microbiology, Department of Biology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | | | | | | | | | | | | | | | | | - Chia-Wei Lin
- Institute of Microbiology, Department of Biology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Markus Aebi
- Institute of Microbiology, Department of Biology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland.
| |
Collapse
|
38
|
Loers G, Astafiev S, Hapiak Y, Saini V, Mishra B, Gul S, Kaur G, Schachner M, Theis T. The polysialic acid mimetics idarubicin and irinotecan stimulate neuronal survival and neurite outgrowth and signal via protein kinase C. J Neurochem 2017; 142:392-406. [PMID: 28542923 PMCID: PMC5539918 DOI: 10.1111/jnc.14076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 02/05/2023]
Abstract
Polysialic acid (PSA) is a large, negatively charged, linear homopolymer of alpha2-8-linked sialic acid residues. It is generated by two polysialyltransferases and attached to N- and/or O-linked glycans, and its main carrier is the neural cell adhesion molecule (NCAM). PSA controls the development and regeneration of the nervous system by enhancing cell migration, axon pathfinding, synaptic targeting, synaptic plasticity, by regulating the differentiation of progenitor cells and by modulating cell-cell and cell-matrix adhesions. In the adult, PSA plays a role in the immune system, and PSA mimetics promote functional recovery after nervous system injury. In search for novel small molecule mimetics of PSA that are applicable for therapy, we identified idarubicin, an antineoplastic anthracycline, and irinotecan, an antineoplastic agent of the topoisomerase I inhibitor class, as PSA mimetics using a competition enzyme-linked immunosorbent assay. Idarubicin and irinotecan compete with the PSA-mimicking peptide and colominic acid, the bacterial analog of PSA, for binding to the PSA-specific monoclonal antibody 735. Idarubicin and irinotecan stimulate neurite outgrowth and survival of cultured cerebellar neurons after oxidative stress via protein kinase C and Erk1/2 in a similar manner as colominic acid, whereas Fyn, casein kinase II and the phosphatase and tensin homolog are only involved in idarubicin and irinotecan-stimulated neurite outgrowth. These novel results show that the structure and function of PSA can be mimicked by the small organic compounds irinotecan and idarubicin which trigger the same signaling cascades as PSA, thus introducing the possibility of retargeting these drugs to treat nervous system injuries.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Steven Astafiev
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Yuliya Hapiak
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Vedangana Saini
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Bibhudatta Mishra
- Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, D-20251 Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology Screening Port (Fraunhofer-IME SP), Schnackenburgalle114, D-22525 Hamburg, Germany
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, GT Road, 143005 Amritsar, India
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China
- To whom correspondence should be addressed: Melitta Schachner, Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA; phone: +1-732-445-1780; fax: +1-732-445-2063; ; or Melitta Schachner, Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong 515041, China; phone: + 86 754 8890 0276; fax: + 86 754 8890 0236;
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| |
Collapse
|
39
|
X-ray crystallographic structure of a bacterial polysialyltransferase provides insight into the biosynthesis of capsular polysialic acid. Sci Rep 2017; 7:5842. [PMID: 28724897 PMCID: PMC5517516 DOI: 10.1038/s41598-017-05627-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/31/2017] [Indexed: 12/21/2022] Open
Abstract
Polysialic acid (polySia) is a homopolymeric saccharide that is associated with some neuroinvasive pathogens and is found on selective cell types in their eukaryotic host. The presence of a polySia capsule on these bacterial pathogens helps with resistance to phagocytosis, cationic microbial peptides and bactericidal antibody production. The biosynthesis of bacterial polySia is catalysed by a single polysialyltransferase (PST) transferring sialic acid from a nucleotide-activated donor to a lipid-linked acceptor oligosaccharide. Here we present the X-ray structure of the bacterial PST from Mannheimia haemolytica serotype A2, thereby defining the architecture of this class of enzymes representing the GT38 family. The structure reveals a prominent electropositive groove between the two Rossmann-like domains forming the GT-B fold that is suitable for binding of polySia chain products. Complex structures of PST with a sugar donor analogue and an acceptor mimetic combined with kinetic studies of PST active site mutants provide insight into the principles of substrate binding and catalysis. Our results are the basis for a molecular understanding of polySia biosynthesis in bacteria and might assist the production of polysialylated therapeutic reagents and the development of novel antibiotics.
Collapse
|
40
|
Okerblom J, Varki A. Biochemical, Cellular, Physiological, and Pathological Consequences of Human Loss of N-Glycolylneuraminic Acid. Chembiochem 2017; 18:1155-1171. [PMID: 28423240 DOI: 10.1002/cbic.201700077] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Indexed: 12/15/2022]
Abstract
About 2-3 million years ago, Alu-mediated deletion of a critical exon in the CMAH gene became fixed in the hominin lineage ancestral to humans, possibly through a stepwise process of selection by pathogen targeting of the CMAH product (the sialic acid Neu5Gc), followed by reproductive isolation through female anti-Neu5Gc antibodies. Loss of CMAH has occurred independently in some other lineages, but is functionally intact in Old World primates, including our closest relatives, the chimpanzee. Although the biophysical and biochemical ramifications of losing tens of millions of Neu5Gc hydroxy groups at most cell surfaces remains poorly understood, we do know that there are multiscale effects functionally relevant to both sides of the host-pathogen interface. Hominin CMAH loss might also contribute to understanding human evolution, at the time when our ancestors were starting to use stone tools, increasing their consumption of meat, and possibly hunting. Comparisons with chimpanzees within ethical and practical limitations have revealed some consequences of human CMAH loss, but more has been learned by using a mouse model with a human-like Cmah inactivation. For example, such mice can develop antibodies against Neu5Gc that could affect inflammatory processes like cancer progression in the face of Neu5Gc metabolic incorporation from red meats, display a hyper-reactive immune system, a human-like tendency for delayed wound healing, late-onset hearing loss, insulin resistance, susceptibility to muscular dystrophy pathologies, and increased sensitivity to multiple human-adapted pathogens involving sialic acids. Further studies in such mice could provide a model for other human-specific processes and pathologies involving sialic acid biology that have yet to be explored.
Collapse
Affiliation(s)
- Jonathan Okerblom
- Biomedical Sciences Graduate Program, University of California in San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0687, USA
| | - Ajit Varki
- Glycobiology Research and Training Center, GRTC) and, Center for Academic Research and Training in Anthropogeny, CARTA), Departments of Medicine and Cellular and Molecular Medicine, University of California in San Diego, La Jolla, CA, 92093-0687, USA
| |
Collapse
|
41
|
Chen Y, Ren H, Zhang N, Troy FA, Wang B. Biochemical Characterization and Analyses of Polysialic-Acid-Associated Carrier Proteins and Genes in Piglets during Neonatal Development. Chembiochem 2017; 18:1270-1278. [PMID: 28444921 DOI: 10.1002/cbic.201700029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Indexed: 01/22/2023]
Abstract
Polysialic acid plays a key role in cancer metastasis and neurodevelopment. Our aim was to determine the developmental gene-expression profiles for the two polysialyltransferases ST8Sia II and ST8Sia IV, neural cell-adhesion molecules (NCAMs), SynCAM 1, neuropilin-2 (NRP2) and their polysialylated cognate glycans in different regions of the piglet brain during postnatal development. Our findings show that: 1) the cellular levels of mRNA coding for ST8Sia II and ST8Sia IV, NCAMs, SynCAM 1, NRP2 and polySia are age-dependent and cell-type-specific during neonatal brain development, 2) there was a lack of correlation between abundance level of mRNA coding for ST8Sia II and ST8Sia IV and the abundance level of the post-translation expression of polySia in all nine brain regions, 3) expression levels of polySia did not correlate with the levels of the carrier proteins NCAM-140, SynCAM 1 and NRP2 in nine brain regions, and 4) the cellular abundance of ST8Sia II and ST8Sia IV in nine subregions of piglet brain is regulated at the level of translation/post-translation, and not at the level of transcription. Collectively, our findings suggest that neuronal and glial cells within different regions of the brain have different transcriptional programs that can direct cell division at different rates based on the activity levels of ST8Sia II and ST8Sia IV and the level of their carrier proteins during neurodevelopment.
Collapse
Affiliation(s)
- Yue Chen
- Medical College of Xiamen University, Xiamen City, 361005, China
| | - He Ren
- Medical College of Xiamen University, Xiamen City, 361005, China
| | - Nai Zhang
- Medical College of Xiamen University, Xiamen City, 361005, China
| | - Frederic A Troy
- Medical College of Xiamen University, Xiamen City, 361005, China.,Department of Biochemistry and Molecular Medicine, University of California School of Medicine, Davis, CA, 95616, USA
| | - Bing Wang
- Medical College of Xiamen University, Xiamen City, 361005, China.,School of Animal and Veterinary Science, Charles Sturt University, Locked Bag, Boorooma Street, Wagga Wagga, NSW, 2678, Australia
| |
Collapse
|
42
|
Somplatzki S, Mühlenhoff M, Kröger A, Gerardy-Schahn R, Böldicke T. Intrabodies against the Polysialyltransferases ST8SiaII and ST8SiaIV inhibit Polysialylation of NCAM in rhabdomyosarcoma tumor cells. BMC Biotechnol 2017; 17:42. [PMID: 28499450 PMCID: PMC5429572 DOI: 10.1186/s12896-017-0360-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 04/19/2017] [Indexed: 01/05/2023] Open
Abstract
Background Polysialic acid (polySia) is a carbohydrate modification of the neural cell adhesion molecule (NCAM), which is implicated in neural differentiation and plays an important role in tumor development and metastasis. Polysialylation of NCAM is mediated by two Golgi-resident polysialyltransferases (polyST) ST8SiaII and ST8SiaIV. Intracellular antibodies (intrabodies; IB) expressed inside the ER and retaining proteins passing the ER such as cell surface receptors or secretory proteins provide an efficient means of protein knockdown. To inhibit the function of ST8SiaII and ST8SiaIV specific ER IBs were generated starting from two corresponding hybridoma clones. Both IBs αST8SiaII-IB and αST8SiaIV-IB were constructed in the scFv format and their functions characterized in vitro and in vivo. Results IBs directed against the polySTs prevented the translocation of the enzymes from the ER to the Golgi-apparatus. Co-immunoprecipitation of ST8SiaII and ST8SiaIV with the corresponding IBs confirmed the intracellular interaction with their cognate antigens. In CHO cells overexpressing ST8SiaII and ST8SiaIV, respectively, the transfection with αST8SiaII-IB or αST8SiaIV-IB inhibited significantly the cell surface expression of polysialylated NCAM. Furthermore stable expression of ST8SiaII-IB, ST8SiaIV-IB and luciferase in the rhabdomyosarcoma cell line TE671 reduced cell surface expression of polySia and delayed tumor growth if cells were xenografted into C57BL/6 J RAG-2 mice. Conclusion Data obtained strongly indicate that αST8SiaII-IB and αST8SiaIV-IB are promising experimental tools to analyze the individual role of the two enzymes during brain development and during migration and proliferation of tumor cells. Electronic supplementary material The online version of this article (doi:10.1186/s12896-017-0360-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefan Somplatzki
- Helmholtz Centre for Infection Research, Structural and Functional Protein Research, Inhoffenstraße 7, D-38124, Braunschweig, Germany
| | - Martina Mühlenhoff
- Institute of Cellular Chemistry, Hannover Medical School, D-30625, Hannover, Germany
| | - Andrea Kröger
- Helmholtz Centre for Infection Research, Group Innate Immunity and Infection, Inhoffenstraße 7, D-38124, Braunschweig, Germany
| | - Rita Gerardy-Schahn
- Institute of Cellular Chemistry, Hannover Medical School, D-30625, Hannover, Germany
| | - Thomas Böldicke
- Helmholtz Centre for Infection Research, Structural and Functional Protein Research, Inhoffenstraße 7, D-38124, Braunschweig, Germany.
| |
Collapse
|
43
|
Galuska CE, Lütteke T, Galuska SP. Is Polysialylated NCAM Not Only a Regulator during Brain Development But also during the Formation of Other Organs? BIOLOGY 2017; 6:biology6020027. [PMID: 28448440 PMCID: PMC5485474 DOI: 10.3390/biology6020027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 11/16/2022]
Abstract
In mammals several cell adhesion molecules are involved during the pre- and postnatal development of all organ systems. A very prominent member of this family is the neural cell adhesion molecule (NCAM). Interestingly, NCAM can be a target for a special form of posttranslational modification: polysialylation. Whereas nearly all extracellular proteins bear mono-sialic acid residues, only a very small group can be polysialylated. Polysialic acid is a highly negatively-charged sugar polymer and can comprise more than 90 sialic acid residues in postnatal mouse brains increasing dramatically the hydrodynamic radius of their carriers. Thus, adhesion and communication processes on cell surfaces are strongly influenced allowing, e.g., the migration of neuronal progenitor cells. In the developing brain the essential role of polysialylated NCAM has been demonstrated in many studies. In comparison to the neuronal system, however, during the formation of other organs the impact of the polysialylated form of NCAM is not well characterized and the number of studies is limited so far. This review summarizes these observations and discusses possible roles of polysialylated NCAM during the development of organs other than the brain.
Collapse
Affiliation(s)
- Christina E Galuska
- Department of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | - Thomas Lütteke
- ITech Progress GmbH, Donnersbergweg 4, 67059 Ludwigshafen, Germany.
| | - Sebastian P Galuska
- Department of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| |
Collapse
|
44
|
Röhrig CH, Choi SSH, Baldwin N. The nutritional role of free sialic acid, a human milk monosaccharide, and its application as a functional food ingredient. Crit Rev Food Sci Nutr 2016; 57:1017-1038. [DOI: 10.1080/10408398.2015.1040113] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
| | - Sharon S. H. Choi
- Intertek Scientific & Regulatory Consultancy, Mississauga, Ontario, Canada
| | - Nigel Baldwin
- Intertek Scientific & Regulatory Consultancy, Hampshire, United Kingdom
| |
Collapse
|
45
|
Gong L, Zhou X, Yang J, Jiang Y, Yang H. Effects of the regulation of polysialyltransferase ST8SiaII on the invasiveness and metastasis of small cell lung cancer cells. Oncol Rep 2016; 37:131-138. [PMID: 28004110 DOI: 10.3892/or.2016.5279] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/30/2016] [Indexed: 11/05/2022] Open
Abstract
Invasiveness and metastasis may seriously affect the prognosis of small cell lung cancer (SCLC). In the present study, we analyzed the effects and inherent mechanisms of action of polysialic acid-modified neural cell adhesion molecule (NCAM) on the invasive and metastatic potential of SCLC. Gene transfection and short hairpin RNA (shRNA) interference were used to enhance or inhibit, respectively, the expression of polysialyltransferase ST8SiaII in the SCLC cell line H446. We studied in vitro positive or negative changes in the invasive and metastatic potential of the SCLC cells as well as the changes in expression of genes related to signaling molecules and metastasis. When ST8SiaII expression was enhanced, the in vitro transmembrane invasion (P<0.01) and migration (P<0.01) abilities of the SCLC cells markedly increased. Phosphorylation levels of fibroblast growth factor receptor 1 (FGFR1), extracellular signal-related kinase 1/2 (ERK1/2), and matrix metalloproteinase-9 (MMP-9) in the SCLC cells were also significantly increased. In contrast, when ST8SiaII expression was inhibited, the transmembrane invasion (P<0.01) and migration (P<0.01) of the SCLC cells as well as expression of the above signaling molecules were suppressed. Polysialic acid-modified NCAM on the surface of SCLC cells is closely related to the metastatic potential of these cells; regulation of ST8SiaII may thus affect the invasiveness and metastasis of SCLC, and these processes may be associated with phosphorylation of FGFR1, ERK1/2 or MMP-9.
Collapse
Affiliation(s)
- Liang Gong
- Department of Respiratory Diseases, Southwest Hospital of the Third Military Medical University, Shapingba, Chongqing 400038, P.R. China
| | - Xiangdong Zhou
- Department of Respiratory Diseases, Southwest Hospital of the Third Military Medical University, Shapingba, Chongqing 400038, P.R. China
| | - Jingxiang Yang
- Department of Respiratory Diseases, Southwest Hospital of the Third Military Medical University, Shapingba, Chongqing 400038, P.R. China
| | - Yongyuan Jiang
- Department of Respiratory Diseases, Southwest Hospital of the Third Military Medical University, Shapingba, Chongqing 400038, P.R. China
| | - Heping Yang
- Department of Respiratory Diseases, Southwest Hospital of the Third Military Medical University, Shapingba, Chongqing 400038, P.R. China
| |
Collapse
|
46
|
Mehrabian M, Hildebrandt H, Schmitt-Ulms G. NCAM1 Polysialylation: The Prion Protein's Elusive Reason for Being? ASN Neuro 2016; 8:8/6/1759091416679074. [PMID: 27879349 PMCID: PMC5122176 DOI: 10.1177/1759091416679074] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/08/2016] [Accepted: 10/02/2016] [Indexed: 01/06/2023] Open
Abstract
Much confusion surrounds the physiological function of the cellular prion protein (PrPC). It is, however, anticipated that knowledge of its function will shed light on its contribution to neurodegenerative diseases and suggest ways to interfere with the cellular toxicity central to them. Consequently, efforts to elucidate its function have been all but exhaustive. Building on earlier work that uncovered the evolutionary descent of the prion founder gene from an ancestral ZIP zinc transporter, we recently investigated a possible role of PrPC in a morphogenetic program referred to as epithelial-to-mesenchymal transition (EMT). By capitalizing on PrPC knockout cell clones in a mammalian cell model of EMT and using a comparative proteomics discovery strategy, neural cell adhesion molecule-1 emerged as a protein whose upregulation during EMT was perturbed in PrPC knockout cells. Follow-up work led us to observe that PrPC regulates the polysialylation of the neural cell adhesion molecule NCAM1 in cells undergoing morphogenetic reprogramming. In addition to governing cellular migration, polysialylation modulates several other cellular plasticity programs PrPC has been phenotypically linked to. These include neurogenesis in the subventricular zone, controlled mossy fiber sprouting and trimming in the hippocampal formation, hematopoietic stem cell renewal, myelin repair and maintenance, integrity of the circadian rhythm, and glutamatergic signaling. This review revisits this body of literature and attempts to present it in light of this novel contextual framework. When approached in this manner, a coherent model of PrPC acting as a regulator of polysialylation during specific cell and tissue morphogenesis events comes into focus.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Herbert Hildebrandt
- Institute for Cellular Chemistry, Hannover Medical School, Hannover, Germany
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Szabo R, Skropeta D. Advancement of Sialyltransferase Inhibitors: Therapeutic Challenges and Opportunities. Med Res Rev 2016; 37:219-270. [DOI: 10.1002/med.21407] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/14/2016] [Accepted: 08/03/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Rémi Szabo
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
| | - Danielle Skropeta
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
- Centre for Medical & Molecular Bioscience; University of Wollongong; Wollongong NSW 2522 Australia
| |
Collapse
|
48
|
Ray GJ, Siekmann J, Scheinecker R, Zhang Z, Gerasimov MV, Szabo CM, Kosma P. Reaction of Oxidized Polysialic Acid and a Diaminooxy Linker: Characterization and Process Optimization Using Nuclear Magnetic Resonance Spectroscopy. Bioconjug Chem 2016; 27:2071-80. [PMID: 27506297 DOI: 10.1021/acs.bioconjchem.6b00336] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- G. Joseph Ray
- Baxter International Inc., 25212
West Illinois Route 120, Round Lake, Illinois 60073, United States
| | - Jürgen Siekmann
- Baxalta Innovations GmbH, now part of Shire, Industriestraße 67, Vienna, A-1221, Austria
| | - Richard Scheinecker
- Baxalta Innovations GmbH, now part of Shire, Industriestraße 67, Vienna, A-1221, Austria
| | - Zhenqing Zhang
- Soochow University, College of Pharmaceutical Sciences, 199 Ren’ai Road, Suzhou, Jiangsu 215123, China
| | - Mikhail V. Gerasimov
- Baxter International Inc., 25212
West Illinois Route 120, Round Lake, Illinois 60073, United States
| | - Christina M. Szabo
- Baxter International Inc., 25212
West Illinois Route 120, Round Lake, Illinois 60073, United States
| | - Paul Kosma
- University of Natural Resources and Life Sciences, Department of Chemistry, Muthgasse 18, Vienna, A-1190, Austria
| |
Collapse
|
49
|
Hoja-Łukowicz D, Przybyło M, Duda M, Pocheć E, Bubka M. On the trail of the glycan codes stored in cancer-related cell adhesion proteins. Biochim Biophys Acta Gen Subj 2016; 1861:3237-3257. [PMID: 27565356 DOI: 10.1016/j.bbagen.2016.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/22/2016] [Accepted: 08/14/2016] [Indexed: 12/14/2022]
Abstract
Changes in the profile of protein glycosylation are a hallmark of ongoing neoplastic transformation. A unique set of tumor-associated carbohydrate antigens expressed on the surface of malignant cells may serve as powerful diagnostic and therapeutic targets. Cell-surface proteins with altered glycosylation affect the growth, proliferation and survival of those cells, and contribute to their acquisition of the ability to migrate and invade. They may also facilitate tumor-induced immunosuppression and the formation of distant metastases. Deciphering the information encoded in these particular glycan portions of glycoconjugates may shed light on the mechanisms of cancer progression and metastasis. A majority of the related review papers have focused on overall changes in the patterns of cell-surface glycans in various cancers, without pinpointing the molecular carriers of these glycan structures. The present review highlights the ways in which particular tumor-associated glycan(s) coupled with a given membrane-bound protein influence neoplastic cell behavior during the development and progression of cancer. We focus on altered glycosylated cell-adhesion molecules belonging to the cadherin, integrin and immunoglobulin-like superfamilies, examined in the context of molecular interactions.
Collapse
Affiliation(s)
- Dorota Hoja-Łukowicz
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Małgorzata Duda
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Ewa Pocheć
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Monika Bubka
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| |
Collapse
|
50
|
Wratil PR, Horstkorte R, Reutter W. Metabolic Glycoengineering with N-Acyl Side Chain Modified Mannosamines. Angew Chem Int Ed Engl 2016; 55:9482-512. [PMID: 27435524 DOI: 10.1002/anie.201601123] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Indexed: 12/14/2022]
Abstract
In metabolic glycoengineering (MGE), cells or animals are treated with unnatural derivatives of monosaccharides. After entering the cytosol, these sugar analogues are metabolized and subsequently expressed on newly synthesized glycoconjugates. The feasibility of MGE was first discovered for sialylated glycans, by using N-acyl-modified mannosamines as precursor molecules for unnatural sialic acids. Prerequisite is the promiscuity of the enzymes of the Roseman-Warren biosynthetic pathway. These enzymes were shown to tolerate specific modifications of the N-acyl side chain of mannosamine analogues, for example, elongation by one or more methylene groups (aliphatic modifications) or by insertion of reactive groups (bioorthogonal modifications). Unnatural sialic acids are incorporated into glycoconjugates of cells and organs. MGE has intriguing biological consequences for treated cells (aliphatic MGE) and offers the opportunity to visualize the topography and dynamics of sialylated glycans in vitro, ex vivo, and in vivo (bioorthogonal MGE).
Collapse
Affiliation(s)
- Paul R Wratil
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité-Universitätsmedizin Berlin, Arnimallee 22, 14195, Berlin, Germany.
| | - Rüdiger Horstkorte
- Institut für Physiologische Chemie, Martin-Luther-Universität Halle-Wittenberg, Hollystrasse 1, 06114, Halle, Germany.
| | - Werner Reutter
- Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Charité-Universitätsmedizin Berlin, Arnimallee 22, 14195, Berlin, Germany
| |
Collapse
|