1
|
Ide D, Gorelik A, Illes K, Nagar B. Structural Analysis of Mammalian Sialic Acid Esterase. J Mol Biol 2024; 436:168801. [PMID: 39321866 DOI: 10.1016/j.jmb.2024.168801] [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: 06/12/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Sialic acid esterase (SIAE) catalyzes the removal of O-acetyl groups from sialic acids found on cell surface glycoproteins to regulate cellular processes such as B cell receptor signalling and apoptosis. Loss-of-function mutations in SIAE are associated with several common autoimmune diseases including Crohn's, ulcerative colitis, and arthritis. To gain a better understanding of the function and regulation of this protein, we determined crystal structures of SIAE from three mammalian homologs, including an acetate bound structure. The structures reveal that the catalytic domain adopts the fold of the SGNH hydrolase superfamily. The active site is composed of a catalytic dyad, as opposed to the previously reported catalytic triad. Attempts to determine a substrate-bound structure yielded only the hydrolyzed product acetate in the active site. Rigid docking of complete substrates followed by molecular dynamics simulations revealed that the active site does not form specific interactions with substrates, rather it appears to be broadly specific to accept sialoglycans with diverse modifications. Based on the acetate bound structure, a catalytic mechanism is proposed. Structural mapping of disease mutations reveals that most are located on the surface of the enzyme and would only cause minor disruptions to the protein fold, suggesting that these mutations likely affect binding to other factors. These results improve our understanding of SIAE biology and may aid in the development of therapies for autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Danilo Ide
- Department of Biochemistry and Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montreal, QC H3G 0B1, Canada
| | - Alexei Gorelik
- Department of Biochemistry and Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montreal, QC H3G 0B1, Canada
| | - Katalin Illes
- Department of Biochemistry and Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montreal, QC H3G 0B1, Canada
| | - Bhushan Nagar
- Department of Biochemistry and Centre de Recherche en Biologie Structurale (CRBS), McGill University, Montreal, QC H3G 0B1, Canada.
| |
Collapse
|
2
|
Patel MA, Fraser DD, Daley M, Cepinskas G, Veraldi N, Grazioli S. The plasma proteome differentiates the multisystem inflammatory syndrome in children (MIS-C) from children with SARS-CoV-2 negative sepsis. Mol Med 2024; 30:51. [PMID: 38632526 PMCID: PMC11022403 DOI: 10.1186/s10020-024-00806-x] [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: 08/11/2023] [Accepted: 03/09/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND The Multi-System Inflammatory Syndrome in Children (MIS-C) can develop several weeks after SARS-CoV-2 infection and requires a distinct treatment protocol. Distinguishing MIS-C from SARS-CoV-2 negative sepsis (SCNS) patients is important to quickly institute the correct therapies. We performed targeted proteomics and machine learning analysis to identify novel plasma proteins of MIS-C for early disease recognition. METHODS A case-control study comparing the expression of 2,870 unique blood proteins in MIS-C versus SCNS patients, measured using proximity extension assays. The 2,870 proteins were reduced in number with either feature selection alone or with a prior COMBAT-Seq batch effect adjustment. The leading proteins were correlated with demographic and clinical variables. Organ system and cell type expression patterns were analyzed with Natural Language Processing (NLP). RESULTS The cohorts were well-balanced for age and sex. Of the 2,870 unique blood proteins, 58 proteins were identified with feature selection (FDR-adjusted P < 0.005, P < 0.0001; accuracy = 0.96, AUC = 1.00, F1 = 0.95), and 15 proteins were identified with a COMBAT-Seq batch effect adjusted feature selection (FDR-adjusted P < 0.05, P < 0.0001; accuracy = 0.92, AUC = 1.00, F1 = 0.89). All of the latter 15 proteins were present in the former 58-protein model. Several proteins were correlated with illness severity scores, length of stay, and interventions (LTA4H, PTN, PPBP, and EGF; P < 0.001). NLP analysis highlighted the multi-system nature of MIS-C, with the 58-protein set expressed in all organ systems; the highest levels of expression were found in the digestive system. The cell types most involved included leukocytes not yet determined, lymphocytes, macrophages, and platelets. CONCLUSIONS The plasma proteome of MIS-C patients was distinct from that of SCNS. The key proteins demonstrated expression in all organ systems and most cell types. The unique proteomic signature identified in MIS-C patients could aid future diagnostic and therapeutic advancements, as well as predict hospital length of stays, interventions, and mortality risks.
Collapse
Affiliation(s)
- Maitray A Patel
- Epidemiology and Biostatistics, Western University, N6A 3K7, London, ON, Canada
| | - Douglas D Fraser
- Lawson Health Research Institute, N6C 2R5, London, ON, Canada.
- Children's Health Research Institute, N6C 4V3, London, ON, Canada.
- Pediatrics, Western University, N6A 3K7, London, ON, Canada.
- Clinical Neurological Sciences, Western University, N6A 3K7, London, ON, Canada.
- Physiology & Pharmacology, Western University, N6A 3K7, London, ON, Canada.
- London Health Sciences Centre, Room C2-C82, 800 Commissioners Road East, N6A 5W9, London, ON, Canada.
| | - Mark Daley
- Epidemiology and Biostatistics, Western University, N6A 3K7, London, ON, Canada
- Computer Science, Western University, N6A 3K7, London, ON, Canada
| | - Gediminas Cepinskas
- Lawson Health Research Institute, N6C 2R5, London, ON, Canada
- Medical Biophysics, Western University, N6A 3K7, London, ON, Canada
| | - Noemi Veraldi
- Department of Pediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Serge Grazioli
- Department of Pediatrics, Gynaecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Neonatal and Pediatric Intensive Care, Department of Child, Woman, and Adolescent Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
3
|
Tsubata T. The ligand interactions of B cell Siglecs are involved in the prevention of autoimmunity to sialylated self-antigens and in the quality control of signaling-competent B cells. Int Immunol 2023; 35:461-473. [PMID: 37504378 DOI: 10.1093/intimm/dxad030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are a family of membrane molecules that recognize sialic acid. Most of them are inhibitory receptors that inhibit immune-cell activation by recognizing sialic acid as a self-motif. Human B cells express CD22 (also known as Siglec-2), Siglec-5, Siglec-6 and Siglec-10 whereas mouse B cells express CD22 and Siglec-G (ortholog of human Siglec-10). Siglecs recognize both sialylated molecules expressed on the same cell (cis-ligands) and those expressed by other cells (trans-ligands). In Guillain-Barré syndrome (GBS), antibody production to gangliosides (which are sialic acid-containing glycolipids) expressed by neurons plays a pathogenic role. A Siglec-10 variant deficient in recognition of gangliosides is genetically associated with GBS, suggesting that Siglec-10 induces self-tolerance to gangliosides by recognizing gangliosides as trans-ligands. Recognition of the BCR as a cis-ligand by Siglec-G and CD22 suppresses BCR signaling in B-1 cells and conventional B cells, respectively. This signal suppression prevents excess expansion of B-1 cells and is involved in the quality control of signaling-competent B cells by setting a threshold for tonic signaling during B cell development. CD22 recognizes other cis-ligands including CD22 and β7 integrin. Interaction of CD22 with other CD22 molecules induces CD22 clustering that suppresses CD22-mediated signal inhibition upon BCR ligation, and interaction with β7 integrin maintains its function in the gut-homing of B cells. Taken together, interactions of B cell Siglecs with multiple trans- and cis-ligands play important roles in B cell homeostasis and immune responses.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Pathology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-8310, Japan
| |
Collapse
|
4
|
Vaill M, Kawanishi K, Varki N, Gagneux P, Varki A. Comparative physiological anthropogeny: exploring molecular underpinnings of distinctly human phenotypes. Physiol Rev 2023; 103:2171-2229. [PMID: 36603157 PMCID: PMC10151058 DOI: 10.1152/physrev.00040.2021] [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: 11/05/2021] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Anthropogeny is a classic term encompassing transdisciplinary investigations of the origins of the human species. Comparative anthropogeny is a systematic comparison of humans and other living nonhuman hominids (so-called "great apes"), aiming to identify distinctly human features in health and disease, with the overall goal of explaining human origins. We begin with a historical perspective, briefly describing how the field progressed from the earliest evolutionary insights to the current emphasis on in-depth molecular and genomic investigations of "human-specific" biology and an increased appreciation for cultural impacts on human biology. While many such genetic differences between humans and other hominids have been revealed over the last two decades, this information remains insufficient to explain the most distinctive phenotypic traits distinguishing humans from other living hominids. Here we undertake a complementary approach of "comparative physiological anthropogeny," along the lines of the preclinical medical curriculum, i.e., beginning with anatomy and considering each physiological system and in each case considering genetic and molecular components that are relevant. What is ultimately needed is a systematic comparative approach at all levels from molecular to physiological to sociocultural, building networks of related information, drawing inferences, and generating testable hypotheses. The concluding section will touch on distinctive considerations in the study of human evolution, including the importance of gene-culture interactions.
Collapse
Affiliation(s)
- Michael Vaill
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
| | - Kunio Kawanishi
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Department of Experimental Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Nissi Varki
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Pascal Gagneux
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Ajit Varki
- Center for Academic Research and Training in Anthropogeny, University of California, San Diego, La Jolla, California
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
5
|
Fosse JH, Andresen AMS, Ploss FB, Weli SC, Heffernan IA, Sapkota S, Lundgård K, Kuiper RV, Solhaug A, Falk K. The infectious salmon anemia virus esterase prunes erythrocyte surfaces in infected Atlantic salmon and exposes terminal sialic acids to lectin recognition. Front Immunol 2023; 14:1158077. [PMID: 37180109 PMCID: PMC10167051 DOI: 10.3389/fimmu.2023.1158077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023] Open
Abstract
Many sialic acid-binding viruses express a receptor-destroying enzyme (RDE) that removes the virus-targeted receptor and limits viral interactions with the host cell surface. Despite a growing appreciation of how the viral RDE promotes viral fitness, little is known about its direct effects on the host. Infectious salmon anemia virus (ISAV) attaches to 4-O-acetylated sialic acids on Atlantic salmon epithelial, endothelial, and red blood cell surfaces. ISAV receptor binding and destruction are effectuated by the same molecule, the haemagglutinin esterase (HE). We recently discovered a global loss of vascular 4-O-acetylated sialic acids in ISAV-infected fish. The loss correlated with the expression of viral proteins, giving rise to the hypothesis that it was mediated by the HE. Here, we report that the ISAV receptor is also progressively lost from circulating erythrocytes in infected fish. Furthermore, salmon erythrocytes exposed to ISAV ex vivo lost their capacity to bind new ISAV particles. The loss of ISAV binding was not associated with receptor saturation. Moreover, upon loss of the ISAV receptor, erythrocyte surfaces became more available to the lectin wheat germ agglutinin, suggesting a potential to alter interactions with endogenous lectins of similar specificity. The pruning of erythrocyte surfaces was inhibited by an antibody that prevented ISAV attachment. Furthermore, recombinant HE, but not an esterase-silenced mutant, was sufficient to induce the observed surface modulation. This links the ISAV-induced erythrocyte modulation to the hydrolytic activity of the HE and shows that the observed effects are not mediated by endogenous esterases. Our findings are the first to directly link a viral RDE to extensive cell surface modulation in infected individuals. This raises the questions of whether other sialic acid-binding viruses that express RDEs affect host cells to a similar extent, and if such RDE-mediated cell surface modulation influences host biological functions with relevance to viral disease.
Collapse
|
6
|
Kibler A, Seifert M, Budeus B. Age-related changes of the human splenic marginal zone B cell compartment. Immunol Lett 2023; 256-257:59-65. [PMID: 37044264 DOI: 10.1016/j.imlet.2023.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/24/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023]
Abstract
In this review, we will summarize the growing body of knowledge on the age-related changes of human splenic B cell composition and molecular evidence of immune maturation and discuss the contribution of these changes on splenic protective function. From birth on, the splenic marginal zone (sMZ) contains a specialized B cell subpopulation, which recruits and archives memory B cells from immune responses throughout the organism. The quality of sMZ B cell responses is augmented by germinal center (GC)-dependent maturation of memory B cells during childhood, however, in old age, these mechanisms likely contribute to waning of splenic protective function.
Collapse
Affiliation(s)
- Artur Kibler
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| | - Marc Seifert
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany; Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, Düsseldorf, Germany.
| | - Bettina Budeus
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
7
|
Zhang Y, Cui D, Huang M, Zheng Y, Zheng B, Chen L, Chen Q. NONO regulates B-cell development and B-cell receptor signaling. FASEB J 2023; 37:e22862. [PMID: 36906291 DOI: 10.1096/fj.202201909rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/13/2023]
Abstract
The paraspeckle protein NONO is a multifunctional nuclear protein participating in the regulation of transcriptional regulation, mRNA splicing and DNA repair. However, whether NONO plays a role in lymphopoiesis is not known. In this study, we generated mice with global deletion of NONO and bone marrow (BM) chimeric mice in which NONO is deleted in all of mature B cells. We found that the global deletion of NONO in mice did not affect T-cell development but impaired early B-cell development in BM at pro- to pre-B-cell transition stage and B-cell maturation in the spleen. Studies of BM chimeric mice demonstrated that the impaired B-cell development in NONO-deficient mice is B-cell-intrinsic. NONO-deficient B cells displayed normal BCR-induced cell proliferation but increased BCR-induced cell apoptosis. Moreover, we found that NONO deficiency impaired BCR-induced activation of ERK, AKT, and NF-κB pathways in B cells, and altered BCR-induced gene expression profile. Thus, NONO plays a critical role in B-cell development and BCR-induced B-cell activation.
Collapse
Affiliation(s)
- Yongguang Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, China
| | - Dongya Cui
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, China
| | - Miaohui Huang
- Department of Reproductive Medicine, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, China
| | - Yongwei Zheng
- Guangzhou Bio-Gene Technology Co., Ltd, Guangzhou, China
| | - Baijiao Zheng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, China
| | - Liling Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, China
| |
Collapse
|
8
|
Aamelfot M, Fosse JH, Viljugrein H, Ploss FB, Benestad SL, McBeath A, Christiansen DH, Garver K, Falk K. Destruction of the vascular viral receptor in infectious salmon anaemia provides in vivo evidence of homologous attachment interference. PLoS Pathog 2022; 18:e1010905. [PMID: 36240255 PMCID: PMC9621750 DOI: 10.1371/journal.ppat.1010905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/31/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022] Open
Abstract
Viral interference is a process where infection with one virus prevents a subsequent infection with the same or a different virus. This is believed to limit superinfection, promote viral genome stability, and protect the host from overwhelming infection. Mechanisms of viral interference have been extensively studied in plants, but remain poorly understood in vertebrates. We demonstrate that infection with infectious salmon anaemia virus (ISAV) strongly reduces homologous viral attachment to the Atlantic salmon, Salmo salar L. vascular surface. A generalised loss of ISAV binding was observed after infection with both high-virulent and low-virulent ISAV isolates, but with different kinetics. The loss of ISAV binding was accompanied by an increased susceptibility to sialidase, suggesting a loss of the vascular 4-O-sialyl-acetylation that mediates ISAV attachment and simultaneously protects the sialic acid from cleavage. Moreover, the ISAV binding capacity of cultured cells dramatically declined 3 days after ISAV infection, accompanied by reduced cellular permissiveness to infection with a second antigenically distinct isolate. In contrast, neither infection with infectious haematopoietic necrosis virus nor stimulation with the viral mimetic poly I:C restricted subsequent cellular ISAV attachment, revealing an ISAV-specific mechanism rather than a general cellular antiviral response. Our study demonstrates homologous ISAV attachment interference by de-acetylation of sialic acids on the vascular surface. This is the first time the kinetics of viral receptor destruction have been mapped throughout the full course of an infection, and the first report of homologous attachment interference by the loss of a vascular viral receptor. Little is known about the biological functions of vascular O-sialyl-acetylation. Our findings raise the question of whether this vascular surface modulation could be linked to the breakdown of central vascular functions that characterises infectious salmon anaemia. Viral interference, also referred to as superinfection exclusion, is a process that supports viral genome integrity and protects the host from overwhelming infection. Here, we demonstrate that infection of Atlantic salmon with infectious salmon anaemia virus (ISAV) results in the destruction of the viral vascular surface receptor, thus preventing virus attachment. We also observed that the loss of viral receptor strongly restricted the extent of a second ISAV infection in cultured cells, suggesting viral interference. To our knowledge, this is the first time the kinetics of viral receptor destruction has been explored in an infected host. This is important, because we know little of how such responses develop in animals and humans. Our study therefore improves the general understanding of how viral infections progress. Finally, our findings raise the question of whether modulation of the vascular surface by ISAV and other viruses may contribute to the pathogenesis of viral disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kyle Garver
- Fisheries and Oceans Canada Pacific Biological Station, Nanaimo, British Columbia, Canada
| | - Knut Falk
- Norwegian Veterinary Institute, Ås, Norway
- * E-mail:
| |
Collapse
|
9
|
Anderson AC, Stangherlin S, Pimentel KN, Weadge JT, Clarke AJ. The SGNH hydrolase family: a template for carbohydrate diversity. Glycobiology 2022; 32:826-848. [PMID: 35871440 PMCID: PMC9487903 DOI: 10.1093/glycob/cwac045] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/20/2022] [Accepted: 07/05/2022] [Indexed: 11/14/2022] Open
Abstract
The substitution and de-substitution of carbohydrate materials are important steps in the biosynthesis and/or breakdown of a wide variety of biologically important polymers. The SGNH hydrolase superfamily is a group of related and well-studied proteins with a highly conserved catalytic fold and mechanism composed of 16 member families. SGNH hydrolases can be found in vertebrates, plants, fungi, bacteria, and archaea, and play a variety of important biological roles related to biomass conversion, pathogenesis, and cell signaling. The SGNH hydrolase superfamily is chiefly composed of a diverse range of carbohydrate-modifying enzymes, including but not limited to the carbohydrate esterase families 2, 3, 6, 12 and 17 under the carbohydrate-active enzyme classification system and database (CAZy.org). In this review, we summarize the structural and functional features that delineate these subfamilies of SGNH hydrolases, and which generate the wide variety of substrate preferences and enzymatic activities observed of these proteins to date.
Collapse
Affiliation(s)
- Alexander C Anderson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph N1G2W1, Canada
| | - Stefen Stangherlin
- Department of Chemistry & Biochemistry, Wilfrid Laurier University, Waterloo N2L3C5, Canada
| | - Kyle N Pimentel
- Department of Molecular and Cellular Biology, University of Guelph, Guelph N1G2W1, Canada
| | - Joel T Weadge
- Department of Biology, Wilfrid Laurier University, Waterloo N2L3C5, Canada
| | - Anthony J Clarke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph N1G2W1, Canada
- Department of Chemistry & Biochemistry, Wilfrid Laurier University, Waterloo N2L3C5, Canada
| |
Collapse
|
10
|
Srivastava S, Verhagen A, Sasmal A, Wasik BR, Diaz S, Yu H, Bensing BA, Khan N, Khedri Z, Secrest P, Sullam P, Varki N, Chen X, Parrish CR, Varki A. Development and applications of sialoglycan-recognizing probes (SGRPs) with defined specificities: exploring the dynamic mammalian sialoglycome. Glycobiology 2022; 32:1116-1136. [PMID: 35926090 PMCID: PMC9680117 DOI: 10.1093/glycob/cwac050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/20/2022] [Accepted: 07/14/2022] [Indexed: 01/07/2023] Open
Abstract
Glycans that are abundantly displayed on vertebrate cell surface and secreted molecules are often capped with terminal sialic acids (Sias). These diverse 9-carbon-backbone monosaccharides are involved in numerous intrinsic biological processes. They also interact with commensals and pathogens, while undergoing dynamic changes in time and space, often influenced by environmental conditions. However, most of this sialoglycan complexity and variation remains poorly characterized by conventional techniques, which often tend to destroy or overlook crucial aspects of Sia diversity and/or fail to elucidate native structures in biological systems, i.e. in the intact sialome. To date, in situ detection and analysis of sialoglycans has largely relied on the use of plant lectins, sialidases, or antibodies, whose preferences (with certain exceptions) are limited and/or uncertain. We took advantage of naturally evolved microbial molecules (bacterial adhesins, toxin subunits, and viral hemagglutinin-esterases) that recognize sialoglycans with defined specificity to delineate 9 classes of sialoglycan recognizing probes (SGRPs: SGRP1-SGRP9) that can be used to explore mammalian sialome changes in a simple and systematic manner, using techniques common in most laboratories. SGRP candidates with specificity defined by sialoglycan microarray studies were engineered as tagged probes, each with a corresponding nonbinding mutant probe as a simple and reliable negative control. The optimized panel of SGRPs can be used in methods commonly available in most bioscience labs, such as ELISA, western blot, flow cytometry, and histochemistry. To demonstrate the utility of this approach, we provide examples of sialoglycome differences in tissues from C57BL/6 wild-type mice and human-like Cmah-/- mice.
Collapse
Affiliation(s)
- Saurabh Srivastava
- Department of Cellular and Molecular Medicine, School of Medicine, University of California at San Diego, San Diego, CA, USA,Glycobiology Research and Training Center, University of California at San Diego, San Diego, CA, USA
| | - Andrea Verhagen
- Department of Cellular and Molecular Medicine, School of Medicine, University of California at San Diego, San Diego, CA, USA,Glycobiology Research and Training Center, University of California at San Diego, San Diego, CA, USA
| | - Aniruddha Sasmal
- Department of Cellular and Molecular Medicine, School of Medicine, University of California at San Diego, San Diego, CA, USA,Glycobiology Research and Training Center, University of California at San Diego, San Diego, CA, USA
| | - Brian R Wasik
- College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Sandra Diaz
- Department of Cellular and Molecular Medicine, School of Medicine, University of California at San Diego, San Diego, CA, USA,Glycobiology Research and Training Center, University of California at San Diego, San Diego, CA, USA
| | - Hai Yu
- Department of Chemistry, University of California at Davis, Davis, CA, USA
| | - Barbara A Bensing
- Department of Medicine, University of California, San Francisco, CA, USA,VA Medical Center, San Francisco, CA, USA
| | - Naazneen Khan
- Department of Cellular and Molecular Medicine, School of Medicine, University of California at San Diego, San Diego, CA, USA,Glycobiology Research and Training Center, University of California at San Diego, San Diego, CA, USA
| | - Zahra Khedri
- Department of Cellular and Molecular Medicine, School of Medicine, University of California at San Diego, San Diego, CA, USA,Glycobiology Research and Training Center, University of California at San Diego, San Diego, CA, USA
| | - Patrick Secrest
- Department of Cellular and Molecular Medicine, School of Medicine, University of California at San Diego, San Diego, CA, USA,Glycobiology Research and Training Center, University of California at San Diego, San Diego, CA, USA
| | - Paul Sullam
- Department of Medicine, University of California, San Francisco, CA, USA,VA Medical Center, San Francisco, CA, USA
| | - Nissi Varki
- Department of Cellular and Molecular Medicine, School of Medicine, University of California at San Diego, San Diego, CA, USA,Glycobiology Research and Training Center, University of California at San Diego, San Diego, CA, USA
| | - Xi Chen
- Department of Chemistry, University of California at Davis, Davis, CA, USA
| | - Colin R Parrish
- College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Ajit Varki
- Corresponding author: UCSD School of Medicine, La Jolla, CA 92093-0687, USA.
| |
Collapse
|
11
|
Ertunc N, Phitak T, Wu D, Fujita H, Hane M, Sato C, Kitajima K. Sulfation of sialic acid is ubiquitous and essential for vertebrate development. Sci Rep 2022; 12:12496. [PMID: 35864127 PMCID: PMC9304399 DOI: 10.1038/s41598-022-15143-4] [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: 03/28/2022] [Accepted: 06/20/2022] [Indexed: 11/09/2022] Open
Abstract
Glycosylation of proteins and lipids occurs in vertebrates, usually terminating with sialylation, which regulates the physicochemical and biological properties of these glycoconjugates. Although less commonly known, sialic acid residues also undergo various modifications, such as acetylation, methylation, and sulfation. However, except for acetylation, the enzymes or functions of the other modification processes are unknown. To the best of our knowledge, this study is the first to demonstrate the ubiquitous occurrence of sulfated sialic acids and two genes encoding the sialate: O-sulfotransferases 1 and 2 in vertebrates. These two enzymes showed about 50% amino acid sequence identity, and appeared to be complementary to each other in acceptor substrate preferences. Gene targeting experiments showed that the deficiency of these genes was lethal for medaka fish during young fry development and accompanied by different phenotypes. Thus, the sulfation of sialic acids is essential for the vertebrate development.
Collapse
Affiliation(s)
- Nursah Ertunc
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Molecular Cell Biology, Faculty of Medical Technology, Graduate School of Health Sciences, Fujita Health University, 1-98 Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Thanyaluck Phitak
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Biochemistry Department, Faculty of Medicine, Chiangmai University, Chiangmai, 50200, Thailand
| | - Di Wu
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Hiroshi Fujita
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan
| | - Masaya Hane
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Chihiro Sato
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan.,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Ken Kitajima
- Bioscience and Biotechnology Center, and Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan. .,Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan.
| |
Collapse
|
12
|
Go S, Sato C, Hane M, Go S, Kitajima K. Implication of N-glycolylneuraminic acid in regulation of cell adhesiveness of C2C12 myoblast cells during differentiation into myotube cells. Glycoconj J 2022; 39:619-631. [PMID: 35639196 DOI: 10.1007/s10719-022-10049-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022]
Abstract
A transition of sialic acid (Sia) species on GM3 ganglioside from N-acetylneuraminic acid (Neu5Ac) to N-glycolylneuraminic acid (Neu5Gc) takes place in mouse C2C12 myoblast cells during their differentiation into myotube cells. However, the meaning of this Sia transition remains unclear. This study thus aims to gain a functional insight into this phenomenon. The following lines of evidence show that the increased de novo synthesis of Neu5Gc residues in differentiating myoblast cells promotes adhesiveness of the cells, which is beneficial for promotion of differentiation. First, the Sia transition occurred even in the C2C12 cells cultured in serum-free medium, indicating that it happens through de novo synthesis of Neu5Gc. Second, GM3(Neu5Gc) was localized in myoblast cells, but not in myotube cells, and related to expression of the CMP-Neu5Ac hydroxylase (CMAH) gene. Notably, expression of CMAH precedes myotube formation not only in differentiating C2C12 cells, but also in mouse developing embryos. Since the myoblast cells were attached on the dish surface more strongly than the myotube cells, expression of GM3(Neu5Gc) may be related to the surface attachment of the myoblast cells. Third, exogenous Neu5Gc, but not Neu5Ac, promoted differentiation of C2C12 cells, thus increasing the number of cells committed to fuse with each other. Fourth, the CMAH-transfected C2C12 cells were attached on the gelatin-coated surface much more rapidly than the mock-cells, suggesting that the expression of CMAH promotes cell adhesiveness through the expression of Neu5Gc.
Collapse
Affiliation(s)
- Shiori Go
- Graduate School of Bioagricultural Sciences and Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Chihiro Sato
- Graduate School of Bioagricultural Sciences and Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Masaya Hane
- Graduate School of Bioagricultural Sciences and Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Shinji Go
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Ken Kitajima
- Graduate School of Bioagricultural Sciences and Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan.
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan.
| |
Collapse
|
13
|
Bufi R, Korstanje R. The impact of genetic background on mouse models of kidney disease. Kidney Int 2022; 102:38-44. [DOI: 10.1016/j.kint.2022.03.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022]
|
14
|
Li Z, Unione L, Liu L, Lang Y, de Vries RP, de Groot RJ, Boons GJ. Synthetic O-Acetylated Sialosides and their Acetamido-deoxy Analogues as Probes for Coronaviral Hemagglutinin-esterase Recognition. J Am Chem Soc 2022; 144:424-435. [PMID: 34967208 DOI: 10.1021/jacs.1c10329] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
O-Acetylation is a common modification of sialic acids that can occur at carbons 4-, 7-, 8-, and/or 9. Acetylated sialosides are employed as receptors by several betacoronaviruses and toroviruses, and by influenza C and D viruses. The molecular basis by which these viruses recognize specific O-acetylated sialosides is poorly understood, and it is unknown how viruses have evolved to recognize specific O-acetylated sialosides expressed by their host. Here, we describe a chemoenzymatic approach that can readily provide sialoglycan analogues in which acetyl esters at C4 and/or C7 are replaced by stabilizing acetamide moieties. The analogues and their natural counterparts were used to examine the ligand requirements of the lectin domain of coronaviral hemagglutinin-esterases (HEs). It revealed that HEs from viruses targeting different host species exhibit different requirements for O-acetylation. It also showed that ester-to-amide perturbation results in decreased or loss of binding. STD NMR and molecular modeling of the complexes of the HE of BCoV with the acetamido analogues and natural counterparts revealed that binding is governed by the complementarity between the acetyl moieties of the sialosides and the hydrophobic patches of the lectin. The precise spatial arrangement of these elements is important, and an ester-to-amide perturbation results in substantial loss of binding. Molecular Dynamics simulations with HEs from coronaviruses infecting other species indicate that these viruses have adapted their HE specificity by the incorporation of hydrophobic or hydrophilic elements to modulate acetyl ester recognition.
Collapse
Affiliation(s)
- Zeshi Li
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Luca Unione
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Yifei Lang
- Virology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, The Netherlands
| | - Robert P de Vries
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
| | - Raoul J de Groot
- Virology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, The Netherlands
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CG, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht 3584, The Netherlands
- Chemistry Department, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
15
|
Visser EA, Moons SJ, Timmermans SBPE, de Jong H, Boltje TJ, Büll C. Sialic acid O-acetylation: From biosynthesis to roles in health and disease. J Biol Chem 2021; 297:100906. [PMID: 34157283 PMCID: PMC8319020 DOI: 10.1016/j.jbc.2021.100906] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Sialic acids are nine-carbon sugars that frequently cap glycans at the cell surface in cells of vertebrates as well as cells of certain types of invertebrates and bacteria. The nine-carbon backbone of sialic acids can undergo extensive enzymatic modification in nature and O-acetylation at the C-4/7/8/9 position in particular is widely observed. In recent years, the detection and analysis of O-acetylated sialic acids have advanced, and sialic acid-specific O-acetyltransferases (SOATs) and O-acetylesterases (SIAEs) that add and remove O-acetyl groups, respectively, have been identified and characterized in mammalian cells, invertebrates, bacteria, and viruses. These advances now allow us to draw a more complete picture of the biosynthetic pathway of the diverse O-acetylated sialic acids to drive the generation of genetically and biochemically engineered model cell lines and organisms with altered expression of O-acetylated sialic acids for dissection of their roles in glycoprotein stability, development, and immune recognition, as well as discovery of novel functions. Furthermore, a growing number of studies associate sialic acid O-acetylation with cancer, autoimmunity, and infection, providing rationale for the development of selective probes and inhibitors of SOATs and SIAEs. Here, we discuss the current insights into the biosynthesis and biological functions of O-acetylated sialic acids and review the evidence linking this modification to disease. Furthermore, we discuss emerging strategies for the design, synthesis, and potential application of unnatural O-acetylated sialic acids and inhibitors of SOATs and SIAEs that may enable therapeutic targeting of this versatile sialic acid modification.
Collapse
Affiliation(s)
- Eline A Visser
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Sam J Moons
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Suzanne B P E Timmermans
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Heleen de Jong
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Thomas J Boltje
- Institute for Molecules and Materials, Department of Synthetic Organic Chemistry, Radboud University Nijmegen, Nijmegen, the Netherlands.
| | - Christian Büll
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Hubrecht Institute, Utrecht, the Netherlands.
| |
Collapse
|
16
|
Matsuda Y, Watanabe T, Li XK. Approaches for Controlling Antibody-Mediated Allograft Rejection Through Targeting B Cells. Front Immunol 2021; 12:682334. [PMID: 34276669 PMCID: PMC8282180 DOI: 10.3389/fimmu.2021.682334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/17/2021] [Indexed: 01/14/2023] Open
Abstract
Both acute and chronic antibody-mediated allograft rejection (AMR), which are directly mediated by B cells, remain difficult to treat. Long-lived plasma cells (LLPCs) in bone marrow (BM) play a crucial role in the production of the antibodies that induce AMR. However, LLPCs survive through a T cell-independent mechanism and resist conventional immunosuppressive therapy. Desensitization therapy is therefore performed, although it is accompanied by severe side effects and the pathological condition may be at an irreversible stage when these antibodies, which induce AMR development, are detected in the serum. In other words, AMR control requires the development of a diagnostic method that predicts its onset before LLPC differentiation and enables therapeutic intervention and the establishment of humoral immune monitoring methods providing more detailed information, including individual differences in the susceptibility to immunosuppressive agents and the pathological conditions. In this study, we reviewed recent studies related to the direct or indirect involvement of immunocompetent cells in the differentiation of naïve-B cells into LLPCs, the limitations of conventional methods, and the possible development of novel control methods in the context of AMR. This information will significantly contribute to the development of clinical applications for AMR and improve the prognosis of patients who undergo organ transplantation.
Collapse
Affiliation(s)
- Yoshiko Matsuda
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
17
|
Schinke CD, Bird JT, Qu P, Yaccoby S, Lyzogubov VV, Shelton R, Ling W, Boyle EM, Deshpande S, Byrum SD, Washam C, Mackintosh S, Stephens O, Thanendrarajan S, Zangari M, Shaughnessy J, Zhan F, Barlogie B, van Rhee F, Walker BA. PHF19 inhibition as a therapeutic target in multiple myeloma. Curr Res Transl Med 2021; 69:103290. [PMID: 33894670 DOI: 10.1016/j.retram.2021.103290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/16/2021] [Accepted: 04/06/2021] [Indexed: 02/01/2023]
Abstract
Epigenetic deregulation is increasingly recognized as a contributing pathological factor in multiple myeloma (MM). In particular tri-methylation of H3 lysine 27 (H3K27me3), which is catalyzed by PHD finger protein 19 (PHF19), a subunit of the Polycomb Repressive Complex 2 (PRC2), has recently shown to be a crucial mediator of MM tumorigenicity. Overexpression of PHF19 in MM has been associated with worse clinical outcome. Yet, while there is mounting evidence that PHF19 overexpression plays a crucial role in MM carcinogenesis downstream mechanisms remain to be elucidated. In the current study we use a functional knock down (KD) of PHF19 to investigate the biological role of PHF19 and show that PHF19KD leads to decreased tumor growth in vitro and in vivo. Expression of major cancer players such as bcl2, myc and EGR1 were decreased upon PHF19KD further underscoring the role of PHF19 in MM biology. Additionally, our results highlighted the prognostic impact of PHF19 overexpression, which was significantly associated with worse survival. Overall, our study underscores the premise that targeting the PHF19-PRC2 complex would open up avenues for novel MM therapies.
Collapse
Affiliation(s)
- Carolina D Schinke
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| | - Jordan T Bird
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Pingping Qu
- Cancer Research and Biostatistics, Seattle, WA, United States
| | - Shmuel Yaccoby
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Valeriy V Lyzogubov
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Randal Shelton
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Wen Ling
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Eileen M Boyle
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, United States
| | - Sharyu Deshpande
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Stephanie D Byrum
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Charity Washam
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Samuel Mackintosh
- College of Medicine, Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Owen Stephens
- The College of Medicine, Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sharmilan Thanendrarajan
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Maurizio Zangari
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - John Shaughnessy
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Fenghuang Zhan
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Bart Barlogie
- Division of Hematology, The Mount Sinai Hospital, New York, NY, Sinai, USA
| | - Frits van Rhee
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Brian A Walker
- Division of Hematology Oncology, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
18
|
Irons EE, Punch PR, Lau JTY. Blood-Borne ST6GAL1 Regulates Immunoglobulin Production in B Cells. Front Immunol 2020; 11:617. [PMID: 32391003 PMCID: PMC7190976 DOI: 10.3389/fimmu.2020.00617] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Humoral immunity is an effective but metabolically expensive defense mechanism. It is unclear whether systemic cues exist to communicate the dynamic need for antigen presentation and immunoglobulin production. Here, we report a novel role for the liver-produced, acute phase reactant ST6GAL1 in IgG production. B cell expression of ST6GAL1, a sialyltransferase mediating the attachment of α2,6-linked sialic acids on N-glycans, is classically implicated in the dysregulated B cell development and immunoglobulin levels of St6gal1-deficient mice. However, the blood-borne pool of ST6GAL1, upregulated during systemic inflammation, can also extrinsically modify leukocyte cell surfaces. We show that B cell independent, extracellular ST6GAL1 enhances B cell IgG production and increases blood IgG titers. B cells of mice lacking the hepatocyte specific St6gal1 promoter have reduced sialylation of cell surface CD22 and CD45 and produce less IgG upon stimulation. Sialylation of B cells by extracellular ST6GAL1 boosts expression of IgM, IgD, and CD86, proliferation, and IgG production in vitro. In vivo, elevation of blood ST6GAL1 enhances B cell development and systemic IgG in a CD22-dependent manner. Our data point to a function of an extracellular glycosyltransferase in promoting humoral immunity. Manipulation of systemic ST6GAL1 may represent an effective therapeutic approach for humoral insufficiency.
Collapse
Affiliation(s)
- Eric E Irons
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, NY, United States
| | - Patrick R Punch
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, NY, United States
| | - Joseph T Y Lau
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
19
|
Abstract
Sialic acid-binding immunoglobulin-type lectins (Siglecs) are expressed on the majority of white blood cells of the immune system and play critical roles in immune cell signaling. Through recognition of sialic acid-containing glycans as ligands, they help the immune system distinguish between self and nonself. Because of their restricted cell type expression and roles as checkpoints in immune cell responses in human diseases such as cancer, asthma, allergy, neurodegeneration, and autoimmune diseases they have gained attention as targets for therapeutic interventions. In this review we describe the Siglec family, its roles in regulation of immune cell signaling, current efforts to define its roles in disease processes, and approaches to target Siglecs for treatment of human disease.
Collapse
Affiliation(s)
- Shiteng Duan
- Departments of Molecular Medicine, and Immunology and Microbiology, Scripps Research, La Jolla, California 92037, USA;
| | - James C Paulson
- Departments of Molecular Medicine, and Immunology and Microbiology, Scripps Research, La Jolla, California 92037, USA;
| |
Collapse
|
20
|
Barnard KN, Wasik BR, LaClair JR, Buchholz DW, Weichert WS, Alford-Lawrence BK, Aguilar HC, Parrish CR. Expression of 9- O- and 7,9- O-Acetyl Modified Sialic Acid in Cells and Their Effects on Influenza Viruses. mBio 2019; 10:e02490-19. [PMID: 31796537 PMCID: PMC6890989 DOI: 10.1128/mbio.02490-19] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022] Open
Abstract
Sialic acids (Sia) are widely displayed on the surfaces of cells and tissues. Sia come in a variety of chemically modified forms, including those with acetyl modifications at the C-7, C-8, and C-9 positions. Here, we analyzed the distribution and amounts of these acetyl modifications in different human and canine cells. Since Sia or their variant forms are receptors for influenza A, B, C, and D viruses, we examined the effects of these modifications on virus infections. We confirmed that 9-O-acetyl and 7,9-O-acetyl modified Sia are widely but variably expressed across cell lines from both humans and canines. Although they were expressed on the cell surfaces of canine MDCK cell lines, they were located primarily within the Golgi compartment of human HEK-293 and A549 cells. The O-acetyl modified Sia were expressed at low levels of 1 to 2% of total Sia in these cell lines. We knocked out and overexpressed the sialate O-acetyltransferase gene (CasD1) and knocked out the sialate O-acetylesterase gene (SIAE) using CRISPR/Cas9 editing. Knocking out CasD1 removed 7,9-O- and 9-O-acetyl Sia expression, confirming previous reports. However, overexpression of CasD1 and knockout of SIAE gave only modest increases in 9-O-acetyl levels in cells and no change in 7,9-O-acetyl levels, indicating that there are complex regulations of these modifications. These modifications were essential for influenza C and D infection but had no obvious effect on influenza A and B infection.IMPORTANCE Sialic acids are key glycans that are involved in many different normal cellular functions, as well as being receptors for many pathogens. However, Sia come in diverse chemically modified forms. Here, we examined and manipulated the expression of 7,9-O- and 9-O-acetyl modified Sia on cells commonly used in influenza virus and other research by engineering the enzymes that produce or remove the acetyl groups.
Collapse
Affiliation(s)
- Karen N Barnard
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Brian R Wasik
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Justin R LaClair
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - David W Buchholz
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Wendy S Weichert
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Brynn K Alford-Lawrence
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Hector C Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Colin R Parrish
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
21
|
Netravali IA, Cariappa A, Yates K, Haining WN, Bertocchi A, Allard-Chamard H, Rosenberg I, Pillai S. 9-O-acetyl sialic acid levels identify committed progenitors of plasmacytoid dendritic cells. Glycobiology 2019; 29:861-875. [PMID: 31411667 DOI: 10.1093/glycob/cwz062] [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: 03/22/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 11/12/2022] Open
Abstract
The origins of plasmacytoid dendritic cells (pDCs) have long been controversial and progenitors exclusively committed to this lineage have not been described. We show here that the fate of hematopoietic progenitors is determined in part by their surface levels of 9-O-acetyl sialic acid. Pro-pDCs were identified as lineage negative 9-O-acetyl sialic acid low progenitors that lack myeloid and lymphoid potential but differentiate into pre-pDCs. The latter cells are also lineage negative, 9-O-acetyl sialic acid low cells but are exclusively committed to the pDC lineage. Levels of 9-O-acetyl sialic acid provide a distinct way to define progenitors and thus facilitate the study of hematopoietic differentiation.
Collapse
Affiliation(s)
- Ilka A Netravali
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Annaiah Cariappa
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Kathleen Yates
- Dana-Farber Cancer Institute, Pediatric Oncology, Harvard Medical School, Boston, MA 02115, USA
| | - W Nicholas Haining
- Dana-Farber Cancer Institute, Pediatric Oncology, Harvard Medical School, Boston, MA 02115, USA
| | - Alice Bertocchi
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Hugues Allard-Chamard
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.,Division of Rheumatology, Faculté de Médecine et des Sciences de la Santé de l', Université de Sherbrooke et Centre de Recherche Clinique Étienne-Le Bel, Sherbrooke, Québec, Canada, J1K 2R1
| | - Ian Rosenberg
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
22
|
Dhar C, Sasmal A, Varki A. From "Serum Sickness" to "Xenosialitis": Past, Present, and Future Significance of the Non-human Sialic Acid Neu5Gc. Front Immunol 2019; 10:807. [PMID: 31057542 PMCID: PMC6481270 DOI: 10.3389/fimmu.2019.00807] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/26/2019] [Indexed: 01/01/2023] Open
Abstract
The description of "serum sickness" more than a century ago in humans transfused with animal sera eventually led to identification of a class of human antibodies directed against glycans terminating in the common mammalian sialic acid N-Glycolylneuraminic acid (Neu5Gc), hereafter called "Neu5Gc-glycans." The detection of such glycans in malignant and fetal human tissues initially raised the possibility that it was an oncofetal antigen. However, "serum sickness" antibodies were also noted in various human disease states. These findings spurred further research on Neu5Gc, and the discovery that it is not synthesized in the human body due to a human-lineage specific genetic mutation in the enzyme CMAH. However, with more sensitive techniques Neu5Gc-glycans were detected in smaller quantities on certain human cell types, particularly epithelia and endothelia. The likely explanation is metabolic incorporation of Neu5Gc from dietary sources, especially red meat of mammalian origin. This incorporated Neu5Gc on glycans appears to be the first example of a "xeno-autoantigen," against which varying levels of "xeno-autoantibodies" are present in all humans. The resulting chronic inflammation or "xenosialitis" may have important implications in human health and disease, especially in conditions known to be aggravated by consumption of red meat. In this review, we will cover the early history of the discovery of "serum sickness" antibodies, the subsequent recognition that they were partly directed against Neu5Gc-glycans, the discovery of the genetic defect eliminating Neu5Gc production in humans, and the later recognition that this was not an oncofetal antigen but the first example of a "xeno-autoantigen." Further, we will present comments about implications for disease risks associated with red meat consumption such as cancer and atherosclerosis. We will also mention the potential utility of these anti-Neu5Gc-glycan antibodies in cancer immunotherapy and provide some suggestions and perspectives for the future. Other reviews in this special issue cover many other aspects of this unusual pathological process, for which there appears to be no other described precedent.
Collapse
Affiliation(s)
- Chirag Dhar
- Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, United States
| | - Aniruddha Sasmal
- Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, United States
| | - Ajit Varki
- Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
23
|
Mahajan VS, Alsufyani F, Mattoo H, Rosenberg I, Pillai S. Alterations in sialic-acid O-acetylation glycoforms during murine erythrocyte development. Glycobiology 2019; 29:222-228. [PMID: 30597004 PMCID: PMC6381321 DOI: 10.1093/glycob/cwy110] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/12/2018] [Accepted: 12/28/2018] [Indexed: 11/16/2022] Open
Abstract
We used Casd1-deficient mice to confirm that this enzyme is responsible for 9-O-acetylation of sialic acids in vivo. We observed a complete loss of 9-O-acetylation of sialic acid on the surface of myeloid, erythroid and CD4+ T cells in Casd1-deficient mice. Although 9-O-acetylation of sialic acids on multiple hematopoietic lineages was lost, there were no obvious defects in hematopoiesis. Interestingly, erythrocytes from Casd1-deficient mice also lost reactivity to TER-119, a rat monoclonal antibody that is widely used to mark the murine erythroid lineage. The sialic acid glyco-epitope recognized by TER-119 on erythrocytes was sensitive to the sialic acid O-acetyl esterase activity of the hemagglutinin-esterase from bovine coronavirus but not to the corresponding enzyme from the influenza C virus. During erythrocyte development, TER-119+ Ery-A and Ery-B cells could be stained by catalytically inactive bovine coronavirus hemagglutinin-esterase but not by the inactive influenza C hemagglutinin-esterase, while TER-119+ Ery-C cells and mature erythrocytes were recognized by both virolectins. Although the structure of the sialoglycoconjugate recognized by TER-119 was not chemically demonstrated, its selective binding to virolectins suggests that it may be comprised of a 7,9-di-O-acetyl form of sialic acid. As erythrocytes mature, the surfaces of Ery-C cells and mature erythrocytes also acquire an additional distinct CASD1-dependent 9-O-acetyl sialic acid moiety that can be recognized by virolectins from both influenza C and bovine coronavirus.
Collapse
Affiliation(s)
- Vinay S Mahajan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital, Department of Pathology, Boston, MA, USA
| | | | - Hamid Mattoo
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Ian Rosenberg
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Clark EA, Giltiay NV. CD22: A Regulator of Innate and Adaptive B Cell Responses and Autoimmunity. Front Immunol 2018; 9:2235. [PMID: 30323814 PMCID: PMC6173129 DOI: 10.3389/fimmu.2018.02235] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022] Open
Abstract
CD22 (Siglec 2) is a receptor predominantly restricted to B cells. It was initially characterized over 30 years ago and named “CD22” in 1984 at the 2nd International workshop in Boston (1). Several excellent reviews have detailed CD22 functions, CD22-regulated signaling pathways and B cell subsets regulated by CD22 or Siglec G (2–4). This review is an attempt to highlight recent and possibly forgotten findings. We also describe the role of CD22 in autoimmunity and the great potential for CD22-based immunotherapeutics for the treatment of autoimmune diseases such as systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- Edward A Clark
- Department of Immunology, University of Washington, Seattle, WA, United States.,Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Natalia V Giltiay
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
25
|
Bhambri A, Dhaunta N, Patel SS, Hardikar M, Bhatt A, Srikakulam N, Shridhar S, Vellarikkal S, Pandey R, Jayarajan R, Verma A, Kumar V, Gautam P, Khanna Y, Khan JA, Fromm B, Peterson KJ, Scaria V, Sivasubbu S, Pillai B. Large scale changes in the transcriptome of Eisenia fetida during regeneration. PLoS One 2018; 13:e0204234. [PMID: 30260966 PMCID: PMC6160089 DOI: 10.1371/journal.pone.0204234] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022] Open
Abstract
Earthworms show a wide spectrum of regenerative potential with certain species like Eisenia fetida capable of regenerating more than two-thirds of their body while other closely related species, such as Paranais litoralis seem to have lost this ability. Earthworms belong to the phylum Annelida, in which the genomes of the marine oligochaete Capitella telata and the freshwater leech Helobdella robusta have been sequenced and studied. Herein, we report the transcriptomic changes in Eisenia fetida (Indian isolate) during regeneration. Following injury, E. fetida regenerates the posterior segments in a time spanning several weeks. We analyzed gene expression changes both in the newly regenerating cells and in the adjacent tissue, at early (15days post amputation), intermediate (20days post amputation) and late (30 days post amputation) by RNAseq based de novo assembly and comparison of transcriptomes. We also generated a draft genome sequence of this terrestrial red worm using short reads and mate-pair reads. An in-depth analysis of the miRNome of the worm showed that many miRNA gene families have undergone extensive duplications. Sox4, a master regulator of TGF-beta mediated epithelial-mesenchymal transition was induced in the newly regenerated tissue. Genes for several proteins such as sialidases and neurotrophins were identified amongst the differentially expressed transcripts. The regeneration of the ventral nerve cord was also accompanied by the induction of nerve growth factor and neurofilament genes. We identified 315 novel differentially expressed transcripts in the transcriptome, that have no homolog in any other species. Surprisingly, 82% of these novel differentially expressed transcripts showed poor potential for coding proteins, suggesting that novel ncRNAs may play a critical role in regeneration of earthworm.
Collapse
Affiliation(s)
- Aksheev Bhambri
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
- Academy of Scientific & Innovative Research (AcSIR), Mathura Road, Delhi, India
| | - Neeraj Dhaunta
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Surendra Singh Patel
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
- Academy of Scientific & Innovative Research (AcSIR), Mathura Road, Delhi, India
| | - Mitali Hardikar
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Abhishek Bhatt
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Nagesh Srikakulam
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Shruti Shridhar
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Shamsudheen Vellarikkal
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
- Academy of Scientific & Innovative Research (AcSIR), Mathura Road, Delhi, India
| | - Rajesh Pandey
- CSIR Ayurgenomics Unit - TRISUTRA, CSIR-IGIB, New Delhi, India
| | - Rijith Jayarajan
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Ankit Verma
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Vikram Kumar
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Pradeep Gautam
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | - Yukti Khanna
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
| | | | - Bastian Fromm
- Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Kevin J. Peterson
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Vinod Scaria
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
- Academy of Scientific & Innovative Research (AcSIR), Mathura Road, Delhi, India
| | - Sridhar Sivasubbu
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
- Academy of Scientific & Innovative Research (AcSIR), Mathura Road, Delhi, India
| | - Beena Pillai
- CSIR – Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, India
- Academy of Scientific & Innovative Research (AcSIR), Mathura Road, Delhi, India
- * E-mail:
| |
Collapse
|
26
|
Yam-Puc JC, Zhang L, Zhang Y, Toellner KM. Role of B-cell receptors for B-cell development and antigen-induced differentiation. F1000Res 2018; 7:429. [PMID: 30090624 PMCID: PMC5893946 DOI: 10.12688/f1000research.13567.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
B-cell development is characterized by a number of tightly regulated selection processes. Signals through the B-cell receptor (BCR) guide and are required for B-cell maturation, survival, and fate decision. Here, we review the role of the BCR during B-cell development, leading to the emergence of B1, marginal zone, and peripheral follicular B cells. Furthermore, we discuss BCR-derived signals on activated B cells that lead to germinal center and plasma cell differentiation.
Collapse
Affiliation(s)
- Juan Carlos Yam-Puc
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Lingling Zhang
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Yang Zhang
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Kai-Michael Toellner
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
27
|
Ereño-Orbea J, Sicard T, Cui H, Mazhab-Jafari MT, Benlekbir S, Guarné A, Rubinstein JL, Julien JP. Molecular basis of human CD22 function and therapeutic targeting. Nat Commun 2017; 8:764. [PMID: 28970495 PMCID: PMC5624926 DOI: 10.1038/s41467-017-00836-6] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022] Open
Abstract
CD22 maintains a baseline level of B-cell inhibition to keep humoral immunity in check. As a B-cell-restricted antigen, CD22 is targeted in therapies against dysregulated B cells that cause autoimmune diseases and blood cancers. Here we report the crystal structure of human CD22 at 2.1 Å resolution, which reveals that specificity for α2-6 sialic acid ligands is dictated by a pre-formed β-hairpin as a unique mode of recognition across sialic acid-binding immunoglobulin-type lectins. The CD22 ectodomain adopts an extended conformation that facilitates concomitant CD22 nanocluster formation on B cells and binding to trans ligands to avert autoimmunity in mammals. We structurally delineate the CD22 site targeted by the therapeutic antibody epratuzumab at 3.1 Å resolution and determine a critical role for CD22 N-linked glycosylation in antibody engagement. Our studies provide molecular insights into mechanisms governing B-cell inhibition and valuable clues for the design of immune modulators in B-cell dysfunction.The B-cell-specific co-receptor CD22 is a therapeutic target for depleting dysregulated B cells. Here the authors structurally characterize the ectodomain of CD22 and present its crystal structure with the bound therapeutic antibody epratuzumab, which gives insights into the mechanism of inhibition of B-cell activation.
Collapse
Affiliation(s)
- June Ereño-Orbea
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
| | - Taylor Sicard
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada, M5S 1A8
| | - Hong Cui
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
| | - Mohammad T Mazhab-Jafari
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
| | - Samir Benlekbir
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
| | - Alba Guarné
- Department of Biochemistry and Biomedical Science, McMaster University, Hamilton, ON, Canada, L8S 4L8
| | - John L Rubinstein
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada, M5S 1A8
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada, M5G 1L7
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada, M5G 0A4.
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada, M5S 1A8.
- Department of Immunology, University of Toronto, Toronto, ON, Canada, M5S 1A8.
| |
Collapse
|
28
|
Wasik BR, Barnard KN, Ossiboff RJ, Khedri Z, Feng KH, Yu H, Chen X, Perez DR, Varki A, Parrish CR. Distribution of O-Acetylated Sialic Acids among Target Host Tissues for Influenza Virus. mSphere 2017; 2:e00379-16. [PMID: 28904995 PMCID: PMC5588038 DOI: 10.1128/msphere.00379-16] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 08/17/2017] [Indexed: 12/30/2022] Open
Abstract
Sialic acids (Sias) are important glycans displayed on the cells and tissues of many different animals and are frequent targets for binding and modification by pathogens, including influenza viruses. Influenza virus hemagglutinins bind Sias during the infection of their normal hosts, while the encoded neuraminidases and/or esterases remove or modify the Sia to allow virion release or to prevent rebinding. Sias naturally occur in a variety of modified forms, and modified Sias can alter influenza virus host tropisms through their altered interactions with the viral glycoproteins. However, the distribution of modified Sia forms and their effects on pathogen-host interactions are still poorly understood. Here we used probes developed from viral Sia-binding proteins to detect O-acetylated (4-O-acetyl, 9-O-acetyl, and 7,9-O-acetyl) Sias displayed on the tissues of some natural or experimental hosts for influenza viruses. These modified Sias showed highly variable displays between the hosts and tissues examined. The 9-O-acetyl (and 7,9-) modified Sia forms were found on cells and tissues of many hosts, including mice, humans, ferrets, guinea pigs, pigs, horses, dogs, as well as in those of ducks and embryonated chicken egg tissues and membranes, although in variable amounts. The 4-O-acetyl Sias were found in the respiratory tissues of fewer animals, being primarily displayed in the horse and guinea pig, but were not detected in humans or pigs. The results suggest that these Sia variants may influence virus tropisms by altering and selecting their cell interactions. IMPORTANCE Sialic acids (Sias) are key glycans that control or modulate many normal cell and tissue functions while also interacting with a variety of pathogens, including many different viruses. Sias are naturally displayed in a variety of different forms, with modifications at several positions that can alter their functional interactions with pathogens. In addition, Sias are often modified or removed by enzymes such as host or pathogen esterases or sialidases (neuraminidases), and Sia modifications can alter those enzymatic activities to impact pathogen infections. Sia chemical diversity in different hosts and tissues likely alters the pathogen-host interactions and influences the outcome of infection. Here we explored the display of 4-O-acetyl, 9-O-acetyl, and 7,9-O-acetyl modified Sia forms in some target tissues for influenza virus infection in mice, humans, birds, guinea pigs, ferrets, swine, horses, and dogs, which encompass many natural and laboratory hosts of those viruses.
Collapse
Affiliation(s)
- Brian R. Wasik
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Karen N. Barnard
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Robert J. Ossiboff
- Department of Population Medicine and Diagnostic Sciences, Animal Health Diagnostic Center, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Zahra Khedri
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Kurtis H. Feng
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Hai Yu
- Department of Chemistry, University of California, Davis, Davis, California, USA
| | - Xi Chen
- Department of Chemistry, University of California, Davis, Davis, California, USA
| | - Daniel R. Perez
- Department of Population Health, Poultry Diagnostic and Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Ajit Varki
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California, USA
| | - Colin R. Parrish
- Department of Microbiology and Immunology, Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
29
|
Ravasio V, Damiati E, Zizioli D, Orizio F, Giacopuzzi E, Manzoni M, Bresciani R, Borsani G, Monti E. Genomic and biochemical characterization of sialic acid acetylesterase (siae) in zebrafish. Glycobiology 2017; 27:938-946. [DOI: 10.1093/glycob/cwx068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/21/2017] [Indexed: 01/08/2023] Open
|
30
|
SIAE Rare Variants in Juvenile Idiopathic Arthritis and Primary Antibody Deficiencies. J Immunol Res 2017; 2017:1514294. [PMID: 28900629 PMCID: PMC5576406 DOI: 10.1155/2017/1514294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/18/2017] [Indexed: 01/23/2023] Open
Abstract
Sialic acid acetylesterase (SIAE) deficiency was suggested to lower the levels of ligands for sialic acid-binding immunoglobulin-like receptors, decreasing the threshold for B-cell activation. In humans, studies of rare heterozygous loss-of-function mutations in SIAE gene in common autoimmune diseases, including juvenile idiopathic arthritis (JIA), yielded inconsistent results. Considering the distinct pathogenesis of the two main subtypes of JIA, autoinflammatory systemic (sJIA) and autoimmune oligo/polyarticular (aJIA), and a predisposition to autoimmunity displayed by patients and families with primary antibody deficiencies (PADs), the aim of our study was to analyze whether SIAE rare variants are associated with both the phenotype of JIA and the autoimmunity risk in families with PADs. A cohort of 69 patients with JIA, 117 healthy children, 54 patients, and family members with PADs were enrolled in the study. Three novel SIAE variants (p.Q343P, p.Y495X, and c.1320+33T>C) were found only in patients with aJIA but interestingly also in their healthy relatives without autoimmunity, while none of PAD patients or their relatives carried SIAE defects. Our results show that SIAE rare variants are not causative of autoimmunity as single defects.
Collapse
|
31
|
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: 52] [Impact Index Per Article: 7.4] [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
|
32
|
|
33
|
Calzas C, Taillardet M, Fourati IS, Roy D, Gottschalk M, Soudeyns H, Defrance T, Segura M. Evaluation of the Immunomodulatory Properties of Streptococcus suis and Group B Streptococcus Capsular Polysaccharides on the Humoral Response. Pathogens 2017; 6:pathogens6020016. [PMID: 28425925 PMCID: PMC5488650 DOI: 10.3390/pathogens6020016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/15/2017] [Accepted: 04/17/2017] [Indexed: 12/02/2022] Open
Abstract
Streptococcus suis and group B Streptococcus (GBS) are encapsulated streptococci causing septicemia and meningitis. Antibodies (Abs) against capsular polysaccharides (CPSs) have a crucial protective role, but the structure/composition of the CPS, including the presence of sialic acid, may interfere with the generation of anti-CPS Ab responses. We investigated the features of the CPS-specific Ab response directed against S. suis serotypes 2 and 14 and GBS serotypes III and V after infection or immunization with purified native or desialylated CPSs in mice. Whereas S. suis-infected mice developed a very low/undetectable CPS-specific IgM response, significant anti-CPS IgM titers were measured in GBS-infected animals (especially for type III GBS). No isotype switching was detected in S. suis- or GBS-infected mice. While the expression of sialic acid was essential for the immunogenicity of purified GBS type III CPS, this sugar was not responsible for the inability of purified S. suis types 2, 14 and GBS type V CPSs to induce a specific Ab response. Thus, other biochemical criteria unrelated to the presence of sialic acid may be responsible for the inaptitude of the host immune system to mount an effective response against certain S. suis and GBS CPS types.
Collapse
Affiliation(s)
- Cynthia Calzas
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte St., Saint-Hyacinthe, QC J2S 2M2, Canada.
| | - Morgan Taillardet
- CIRI, INSERM, U1111, CNRS UMR5308, University of Lyon 1, 21 Avenue Tony Garnier, 69007 Lyon, France.
| | - Insaf Salem Fourati
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, University of Montreal, C.P. 6128, Succ. Centre-ville, Montreal, QC H3C 3J7, Canada.
| | - David Roy
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte St., Saint-Hyacinthe, QC J2S 2M2, Canada.
| | - Marcelo Gottschalk
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte St., Saint-Hyacinthe, QC J2S 2M2, Canada.
| | - Hugo Soudeyns
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, University of Montreal, C.P. 6128, Succ. Centre-ville, Montreal, QC H3C 3J7, Canada.
| | - Thierry Defrance
- CIRI, INSERM, U1111, CNRS UMR5308, University of Lyon 1, 21 Avenue Tony Garnier, 69007 Lyon, France.
| | - Mariela Segura
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Department of Pathology and Microbiology, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte St., Saint-Hyacinthe, QC J2S 2M2, Canada.
| |
Collapse
|
34
|
Oleksyn D, Zhao J, Vosoughi A, Zhao JC, Misra R, Pentland AP, Ryan D, Anolik J, Ritchlin C, Looney J, Anandarajah AP, Schwartz G, Calvi LM, Georger M, Mohan C, Sanz I, Chen L. PKK deficiency in B cells prevents lupus development in Sle lupus mice. Immunol Lett 2017; 185:1-11. [PMID: 28274793 DOI: 10.1016/j.imlet.2017.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of autoantibodies that can result in damage to multiple organs. It is well documented that B cells play a critical role in the development of the disease. We previously showed that protein kinase C associated kinase (PKK) is required for B1 cell development as well as for the survival of recirculating mature B cells and B-lymphoma cells. Here, we investigated the role of PKK in lupus development in a lupus mouse model. We demonstrate that the conditional deletion of PKK in B cells prevents lupus development in Sle1Sle3 mice. The loss of PKK in Sle mice resulted in the amelioration of multiple classical lupus-associated phenotypes and histologic features of lupus nephritis, including marked reduction in the levels of serum autoantibodies, proteinuria, spleen size, peritoneal B-1 cell population and the number of activated CD4 T cells. In addition, the abundance of autoreactive plasma cells normally seen in Sle lupus mice was also significantly decreased in the PKK-deficient Sle mice. Sle B cells deficient in PKK display defective proliferation responses to BCR and LPS stimulation. Consistently, B cell receptor-mediated NF-κB activation, which is required for the survival of activated B cells, was impaired in the PKK-deficient B cells. Taken together, our work uncovers a critical role of PKK in lupus development and suggests that targeting the PKK-mediated pathway may represent a promising therapeutic strategy for lupus treatment.
Collapse
Affiliation(s)
- D Oleksyn
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Zhao
- Department of Biomedical Genetics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A Vosoughi
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J C Zhao
- Department of Biology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - R Misra
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A P Pentland
- Department of Dermatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - D Ryan
- Department of Pathology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Anolik
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - C Ritchlin
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - J Looney
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - A P Anandarajah
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - G Schwartz
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - L M Calvi
- Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - M Georger
- Department of Medicine, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - C Mohan
- Department Biomedical Engineering, University of Houston, Houston, TX 77204, United States
| | - I Sanz
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States
| | - L Chen
- Division of Allergy/Immunology and Rheumatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States; Department of Dermatology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, United States.
| |
Collapse
|
35
|
Abstract
An important underlying mechanism that contributes to autoimmunity is the loss of inhibitory signaling in the immune system. Sialic acid-recognizing Ig superfamily lectins or Siglecs are a family of cell surface proteins largely expressed in hematopoietic cells. The majority of Siglecs are inhibitory receptors expressed in immune cells that bind to sialic acid-containing ligands and recruit SH2-domain-containing tyrosine phosphatases to their cytoplasmic tails. They deliver inhibitory signals that can contribute to the constraining of immune cells, and thus protect the host from autoimmunity. The inhibitory functions of CD22/Siglec-2 and Siglec-G and their contributions to tolerance and autoimmunity, primarily in the B lymphocyte context, are considered in some detail in this review. The relevance to autoimmunity and unregulated inflammation of modified sialic acids, enzymes that modify sialic acid, and other sialic acid-binding proteins are also reviewed.
Collapse
Affiliation(s)
- Vinay S Mahajan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Departments of Medicine and Pathology, Harvard Medical School, Boston, MA, USA.,Deaprtment of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.,Departments of Medicine and Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
36
|
Blazina Š, Ihan A, Lovrečić L, Hovnik T. 11q terminal deletion and combined immunodeficiency (Jacobsen syndrome): Case report and literature review on immunodeficiency in Jacobsen syndrome. Am J Med Genet A 2016; 170:3237-3240. [PMID: 27605496 DOI: 10.1002/ajmg.a.37859] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/01/2016] [Indexed: 11/08/2022]
Abstract
Antibody deficiency is common finding in patients with Jacobsen syndrome (JS). In addition, there have been few reports of T-cell defects in this condition, possibly because most of the reported patients have not been specifically evaluated for T-cell function. In this article, we present a child with an 11q deletion and combined immunodeficiency and we perform a literature overview on immunodeficiency in JS. Our patient presented with recurrent bacterial and prolonged viral infections involving the respiratory system, as well as other classic features of the syndrome. In addition to low IgM, IgG4, and B-cells, also low recent thymic emigrants, helper and naïve T-cells were found. We propose that patients with Jacobsen syndrome need thorough immunological evaluations as T-cell dysfunction might be more prevalent than previously reported. Patients with infections consistent with T-cell defects should be classified as having combined immunodeficiency. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Štefan Blazina
- Department of Allergy, Rheumatology and Clinical Immunology, Children's Hospital Ljubljana, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Luca Lovrečić
- Division of Obstetrics and Gynecology, Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Tinka Hovnik
- Unit of Special Laboratory Diagnostics, Children's Hospital Ljubljana, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
37
|
Wasik BR, Barnard KN, Parrish CR. Effects of Sialic Acid Modifications on Virus Binding and Infection. Trends Microbiol 2016; 24:991-1001. [PMID: 27491885 PMCID: PMC5123965 DOI: 10.1016/j.tim.2016.07.005] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/29/2022]
Abstract
Sialic acids (Sias) are abundantly displayed on the surfaces of vertebrate cells, and particularly on all mucosal surfaces. Sias interact with microbes of many types, and are the targets of specific recognition by many different viruses. They may mediate virus binding and infection of cells, or alternatively can act as decoy receptors that bind virions and block virus infection. These nine-carbon backbone monosaccharides naturally occur in many different modified forms, and are attached to underlying glycans through varied linkages, creating significant diversity in the pathogen receptor forms. Here we review the current knowledge regarding the distribution of modified Sias in different vertebrate hosts, tissues, and cells, their effects on viral pathogens where those have been examined, and outline unresolved questions. Sialic acids (Sias) are components of cell-surface glycoproteins and glycolipids, as well as secreted glycoproteins and milk oligosaccharides. Sias play important roles in cell signaling, development, and host–pathogen interactions. Cellular enzymes can modify Sias, yet how modifications vary between tissues and hosts has not been fully elucidated. Many viruses use Sias as receptors, with different modifications aiding or inhibiting virus infection. How modified Sias influence viral protein evolution and determine host/tissue tropism are poorly understood, and are important areas of research. New advances in molecular glycobiology using pathogen proteins to detect varied forms allows for improved study of modified Sias that have otherwise proven difficult to isolate. This opens new avenues of inquiry for virology, as well as host interactions with bacterial and eukaryotic pathogens.
Collapse
Affiliation(s)
- Brian R Wasik
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | - Karen N Barnard
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Colin R Parrish
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
38
|
Mahajan VS, Demissie E, Mattoo H, Viswanadham V, Varki A, Morris R, Pillai S. Striking Immune Phenotypes in Gene-Targeted Mice Are Driven by a Copy-Number Variant Originating from a Commercially Available C57BL/6 Strain. Cell Rep 2016; 15:1901-9. [PMID: 27210752 DOI: 10.1016/j.celrep.2016.04.080] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/28/2016] [Accepted: 04/21/2016] [Indexed: 12/21/2022] Open
Abstract
We describe a homozygous copy-number variant that disrupts the function of Dock2 in a commercially available C57BL/6 mouse strain that is widely used for backcrossing. This Dock2 allele was presumed to have spontaneously arisen in a colony of Irf5 knockout mice. We discovered that this allele has actually been inadvertently backcrossed into multiple mutant mouse lines, including two engineered to be deficient in Siae and Cmah. This particular commercially obtained subline of C57BL/6 mice also exhibits several striking immune phenotypes that have been previously described in the context of Dock2 deficiency. Inadvertent backcrossing of a number of gene-targeted mice into this background has complicated the interpretation of several immunological studies. In light of these findings, published studies involving immune or hematopoietic phenotypes in which these C57BL/6 mice have been used as controls, as experimental animals, or for backcrossing will need to be reinterpreted.
Collapse
Affiliation(s)
- Vinay S Mahajan
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA
| | - Ezana Demissie
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA
| | - Hamid Mattoo
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA
| | - Vinay Viswanadham
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA
| | - Ajit Varki
- Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert Morris
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, 400 Technology Square, Cambridge, MA 02139, USA.
| |
Collapse
|
39
|
Chen L, Oleksyn D, Pulvino M, Sanz I, Ryan D, Ryan C, Lin CS, Poligone B, Pentland AP, Ritchlin C, Zhao J. A critical role for the protein kinase PKK in the maintenance of recirculating mature B cells and the development of B1 cells. Immunol Lett 2016; 172:67-78. [PMID: 26921474 DOI: 10.1016/j.imlet.2016.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 02/16/2016] [Accepted: 02/21/2016] [Indexed: 01/10/2023]
Abstract
Protein kinase C associated kinase (PKK) regulates NF-κB activation and is required for the survival of certain lymphoma cells. Mice lacking PKK die soon after birth, and previous studies suggest that the role of PKK in B cell development might be context dependent. We have generated a mouse strain harboring conditional null alleles for PKK and a Cre-recombinase transgene under the control of the endogenous CD19 promoter. In the present study, we show that knockout of PKK in B cells results in the reduction of long-lived recirculating mature B cell population in lymph nodes and bone marrow as well as a decrease in peritoneal B1 cells, while PKK deficiency has no apparent effect on early B cell development in bone marrow or the development of follicular and marginal zone B cells in the spleen. In addition, we demonstrate that PKK-deficient B cells display defective proliferation and survival responses to stimulation of B cell receptor (BCR), which may underlie the reduction of recirculating mature B cells in PKK mutant mice. Consistently, BCR-mediated NF-κB activation, known to be required for the survival of activated but not resting B cells, is attenuated in PKK-deficient B cells. Thus, our results reveal a critical role of PKK in the maintenance of recirculating mature B cells as well as the development of B1 cells in mice.
Collapse
Affiliation(s)
- Luojing Chen
- Division of Allergy/Immunology and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States; Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States.
| | - David Oleksyn
- Division of Allergy/Immunology and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States; Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States
| | - Mary Pulvino
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States
| | - Ignacio Sanz
- Division of Allergy/Immunology and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States
| | - Daniel Ryan
- Department of Pathology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States
| | - Charlotte Ryan
- Department of Pathology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States
| | - Chyuan-Sheng Lin
- Department of Pathology and Cell Biology & Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, United States
| | - Brian Poligone
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States
| | - Alice P Pentland
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States
| | - Christopher Ritchlin
- Division of Allergy/Immunology and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States
| | - Jiyong Zhao
- Department of Biomedical Genetics, University of Rochester Medical Center, 601 Elmwood Ave. Rochester, NY 14642, United States.
| |
Collapse
|
40
|
Macauley MS, Kawasaki N, Peng W, Wang SH, He Y, Arlian BM, McBride R, Kannagi R, Khoo KH, Paulson JC. Unmasking of CD22 Co-receptor on Germinal Center B-cells Occurs by Alternative Mechanisms in Mouse and Man. J Biol Chem 2015; 290:30066-77. [PMID: 26507663 DOI: 10.1074/jbc.m115.691337] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Indexed: 11/06/2022] Open
Abstract
CD22 is an inhibitory B-cell co-receptor whose function is modulated by sialic acid (Sia)-bearing glycan ligands. Glycan remodeling in the germinal center (GC) alters CD22 ligands, with as yet no ascribed biological consequence. Here, we show in both mice and humans that loss of high affinity ligands on GC B-cells unmasks the binding site of CD22 relative to naive and memory B-cells, promoting recognition of trans ligands. The conserved modulation of CD22 ligands on GC B-cells is striking because high affinity glycan ligands of CD22 are species-specific. In both species, the high affinity ligand is based on the sequence Siaα2-6Galβ1-4GlcNAc, which terminates N-glycans. The human ligand has N-acetylneuraminic acid (Neu5Ac) as the sialic acid, and the high affinity ligand on naive B-cells contains 6-O-sulfate on the GlcNAc. On human GC B-cells, this sulfate modification is lost, giving rise to lower affinity CD22 ligands. Ligands of CD22 on naive murine B-cells do not contain the 6-O-sulfate modification. Instead, the high affinity ligand for mouse CD22 has N-glycolylneuraminic acid (Neu5Gc) as the sialic acid, which is replaced on GC B-cells with Neu5Ac. Human naive and memory B-cells express sulfated glycans as high affinity CD22 ligands, which are lost on GC B-cells. In mice, Neu5Gc-containing glycans serve as high affinity CD22 ligands that are replaced by Neu5Ac-containing glycans on GC B-cells. Our results demonstrate that loss of high affinity CD22 ligands on GC B-cells occurs in both mice and humans through alternative mechanisms, unmasking CD22 relative to naive and memory B-cells.
Collapse
Affiliation(s)
| | | | | | | | - Yuan He
- Cell and Molecular Biology, and
| | | | | | - Reiji Kannagi
- Biomedical Science, Academia Sinica, Taipei 115, Taiwan
| | | | - James C Paulson
- From the Departments of Chemical Physiology, Cell and Molecular Biology, and Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California 92037 and
| |
Collapse
|
41
|
Defaus S, Gupta P, Andreu D, Gutiérrez-Gallego R. Mammalian protein glycosylation--structure versus function. Analyst 2015; 139:2944-67. [PMID: 24779027 DOI: 10.1039/c3an02245e] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Carbohydrates fulfil many common as well as extremely important functions in nature. They show a variety of molecular displays--e.g., free mono-, oligo-, and polysaccharides, glycolipids, proteoglycans, glycoproteins, etc.--with particular roles and localizations in living organisms. Structure-specific peculiarities are so many and diverse that it becomes virtually impossible to cover them all from an analytical perspective. Hence this manuscript, focused on mammalian glycosylation, rather than a complete list of analytical descriptors or recognized functions for carbohydrate structures, comprehensively reviews three central issues in current glycoscience, namely (i) structural analysis of glycoprotein glycans, covering both classical and novel approaches for teasing out the structural puzzle as well as potential pitfalls of these processes; (ii) an overview of functions attributed to carbohydrates, covering from monosaccharide to complex, well-defined epitopes and full glycans, including post-glycosylational modifications, and (iii) recent technical advances allowing structural identification of glycoprotein glycans with simultaneous assignation of biological functions.
Collapse
Affiliation(s)
- S Defaus
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona Biomedical Research Park, 08003 Barcelona, Spain.
| | | | | | | |
Collapse
|
42
|
Stöckmann H, Duke RM, Millán Martín S, Rudd PM. Ultrahigh throughput, ultrafiltration-based n-glycomics platform for ultraperformance liquid chromatography (ULTRA(3)). Anal Chem 2015; 87:8316-22. [PMID: 26183862 DOI: 10.1021/acs.analchem.5b01463] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Accurate, reproducible, and fast quantification of N-glycans is crucial not only for the development and quality control of modern glycosylated biopharmaceuticals, but also in clinical biomarker discovery. Several methods exist for fluorescent labeling of N-glycans and subsequent chromatographic separation and quantification. However, the methods can be complex, lengthy, and expensive. Here we report an automated ultrafiltration-based N-glycoanalytical workflow combined with a glycan labeling strategy that is based on the reaction of glycosylamines with fluorescent carbamate. The entire protocol is quick, simple, and cost-effective and requires less than 1 μg of protein per sample. As many as 768 affinity purified IgG glycoprotein samples can be prepared in a single run with a liquid handling platform.
Collapse
Affiliation(s)
- Henning Stöckmann
- NIBRT GlycoScience Group, NIBRT - The National Institute for Bioprocessing Research and Training, Fosters Avenue, Mount Merrion, Blackrock, Co. Dublin, Ireland
| | - Rebecca M Duke
- NIBRT GlycoScience Group, NIBRT - The National Institute for Bioprocessing Research and Training, Fosters Avenue, Mount Merrion, Blackrock, Co. Dublin, Ireland
| | - Silvia Millán Martín
- NIBRT GlycoScience Group, NIBRT - The National Institute for Bioprocessing Research and Training, Fosters Avenue, Mount Merrion, Blackrock, Co. Dublin, Ireland
| | - Pauline M Rudd
- NIBRT GlycoScience Group, NIBRT - The National Institute for Bioprocessing Research and Training, Fosters Avenue, Mount Merrion, Blackrock, Co. Dublin, Ireland
| |
Collapse
|
43
|
Baumann AMT, Bakkers MJG, Buettner FFR, Hartmann M, Grove M, Langereis MA, de Groot RJ, Mühlenhoff M. 9-O-Acetylation of sialic acids is catalysed by CASD1 via a covalent acetyl-enzyme intermediate. Nat Commun 2015; 6:7673. [PMID: 26169044 PMCID: PMC4510713 DOI: 10.1038/ncomms8673] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/01/2015] [Indexed: 12/13/2022] Open
Abstract
Sialic acids, terminal sugars of glycoproteins and glycolipids, play important roles in development, cellular recognition processes and host–pathogen interactions. A common modification of sialic acids is 9-O-acetylation, which has been implicated in sialoglycan recognition, ganglioside biology, and the survival and drug resistance of acute lymphoblastic leukaemia cells. Despite many functional implications, the molecular basis of 9-O-acetylation has remained elusive thus far. Following cellular approaches, including selective gene knockout by CRISPR/Cas genome editing, we here show that CASD1—a previously identified human candidate gene—is essential for sialic acid 9-O-acetylation. In vitro assays with the purified N-terminal luminal domain of CASD1 demonstrate transfer of acetyl groups from acetyl-coenzyme A to CMP-activated sialic acid and formation of a covalent acetyl-enzyme intermediate. Our study provides direct evidence that CASD1 is a sialate O-acetyltransferase and serves as key enzyme in the biosynthesis of 9-O-acetylated sialoglycans. 9-O-Acetylation is one of the most common modifications of sialic acids, implicated in sialoglycan recognition and ganglioside biology. Here, the authors show that the key enzyme for the biosynthesis of 9-O-acetylated sialoglycans is CASD1, which uses CMP-activated sialic acid as acceptor substrate.![]()
Collapse
Affiliation(s)
- Anna-Maria T Baumann
- Institute of Cellular Chemistry, Hannover Medical School, D-30623 Hannover, Germany
| | - Mark J G Bakkers
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Falk F R Buettner
- Institute of Cellular Chemistry, Hannover Medical School, D-30623 Hannover, Germany
| | - Maike Hartmann
- Institute of Cellular Chemistry, Hannover Medical School, D-30623 Hannover, Germany
| | - Melanie Grove
- Institute of Cellular Chemistry, Hannover Medical School, D-30623 Hannover, Germany
| | - Martijn A Langereis
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Raoul J de Groot
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Martina Mühlenhoff
- Institute of Cellular Chemistry, Hannover Medical School, D-30623 Hannover, Germany
| |
Collapse
|
44
|
Langereis MA, Bakkers MJG, Deng L, Padler-Karavani V, Vervoort SJ, Hulswit RJG, van Vliet ALW, Gerwig GJ, de Poot SAH, Boot W, van Ederen AM, Heesters BA, van der Loos CM, van Kuppeveld FJM, Yu H, Huizinga EG, Chen X, Varki A, Kamerling JP, de Groot RJ. Complexity and Diversity of the Mammalian Sialome Revealed by Nidovirus Virolectins. Cell Rep 2015; 11:1966-78. [PMID: 26095364 PMCID: PMC5292239 DOI: 10.1016/j.celrep.2015.05.044] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 05/01/2015] [Accepted: 05/22/2015] [Indexed: 12/23/2022] Open
Abstract
Sialic acids (Sias), 9-carbon-backbone sugars, are among the most complex and versatile molecules of life. As terminal residues of glycans on proteins and lipids, Sias are key elements of glycotopes of both cellular and microbial lectins and thus act as important molecular tags in cell recognition and signaling events. Their functions in such interactions can be regulated by post-synthetic modifications, the most common of which is differential Sia-O-acetylation (O-Ac-Sias). The biology of O-Ac-Sias remains mostly unexplored, largely because of limitations associated with their specific in situ detection. Here, we show that dual-function hemagglutinin-esterase envelope proteins of nidoviruses distinguish between a variety of closely related O-Ac-Sias. By using soluble forms of hemagglutinin-esterases as lectins and sialate-O-acetylesterases, we demonstrate differential expression of distinct O-Ac-sialoglycan populations in an organ-, tissue- and cell-specific fashion. Our findings indicate that programmed Sia-O-acetylation/de-O-acetylation may be critical to key aspects of cell development, homeostasis, and/or function. Virolectins detect and distinguish between closely related O-Ac-Sias in situ O-Ac-sialoglycans occur in nature in a diversity not appreciated so far O-Ac-Sias are differentially expressed in a species-, tissue-, and cell-specific fashion There is extensive cell-to-cell variability in O-Ac-Sia expression in vivo and in vitro
Collapse
Affiliation(s)
- Martijn A Langereis
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Mark J G Bakkers
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Lingquan Deng
- Glycobiology Research and Training Center, Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Vered Padler-Karavani
- Glycobiology Research and Training Center, Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Stephin J Vervoort
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Ruben J G Hulswit
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Arno L W van Vliet
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Gerrit J Gerwig
- Bio-Organic Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Stefanie A H de Poot
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Willemijn Boot
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Anne Marie van Ederen
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Balthasar A Heesters
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Chris M van der Loos
- Department of Cardiovascular Pathology, Free University Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Frank J M van Kuppeveld
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands
| | - Hai Yu
- Department of Chemistry, University of California, Davis, Davis, CA 95616, USA
| | - Eric G Huizinga
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Xi Chen
- Department of Chemistry, University of California, Davis, Davis, CA 95616, USA
| | - Ajit Varki
- Glycobiology Research and Training Center, Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0687, USA
| | - Johannis P Kamerling
- Bio-Organic Chemistry, Bijvoet Center for Biomolecular Research, Faculty of Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Raoul J de Groot
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, the Netherlands.
| |
Collapse
|
45
|
Orizio F, Damiati E, Giacopuzzi E, Benaglia G, Pianta S, Schauer R, Schwartz-Albiez R, Borsani G, Bresciani R, Monti E. Human sialic acid acetyl esterase: Towards a better understanding of a puzzling enzyme. Glycobiology 2015; 25:992-1006. [DOI: 10.1093/glycob/cwv034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/17/2015] [Indexed: 01/09/2023] Open
|
46
|
Müller J, Lunz B, Schwab I, Acs A, Nimmerjahn F, Daniel C, Nitschke L. Siglec-G Deficiency Leads to Autoimmunity in Aging C57BL/6 Mice. THE JOURNAL OF IMMUNOLOGY 2015; 195:51-60. [PMID: 25987743 DOI: 10.4049/jimmunol.1403139] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/20/2015] [Indexed: 01/06/2023]
Abstract
Siglec-G, a member of the sialic acid-binding Ig-like lectin (Siglec) family, is expressed on B cell and dendritic cell surfaces. It acts as an inhibitory coreceptor and modulates B cell activation, especially on B1 cells, as Siglec-G-deficient mice show mainly a B1 cell-restricted phenotype resulting in increased B1 cell numbers. Although higher B1 cell numbers are discussed to be associated with autoimmunity, loss of Siglec-G does not result in autoimmune disease in BALB/c mice. However, there is evidence from Siglec-G × CD22 double-deficient mice and Siglec-G(-/-) mice on an autoimmune-prone MRL/lpr background that Siglec-G is important to maintain tolerance in B cells. In this study, we analyzed the role of Siglec-G in induction and maintenance of B cell tolerance on C57BL/6 background and in the FcγRIIb-deficient background. We find that aging Siglec-G-deficient and Siglec-G × FcγRIIb double-deficient mice develop an autoimmune phenotype with elevated autoantibody levels and mild glomerulonephritis. Aging Siglec-G-deficient mice have elevated numbers of plasma cells and germinal center B cells, as well as a higher number of activated CD4 T cells, which likely all contribute to autoantibody production. Additional loss of the inhibitory receptor FcγRIIb in Siglec-G(-/-) mice does not result in exacerbation of disease. These results indicate that Siglec-G is important to maintain tolerance in B cells and prevent autoimmunity.
Collapse
Affiliation(s)
- Jennifer Müller
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany; and
| | - Benjamin Lunz
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany; and
| | - Inessa Schwab
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany; and
| | - Andreas Acs
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany; and
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany; and
| | - Christoph Daniel
- Department of Nephropathology, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany; and
| |
Collapse
|
47
|
Beck-Engeser GB, Winkelmann R, Wheeler ML, Shansab M, Yu P, Wünsche S, Walchhütter A, Metzner M, Vettermann C, Eilat D, DeFranco A, Jäck HM, Wabl M. APOBEC3 enzymes restrict marginal zone B cells. Eur J Immunol 2015; 45:695-704. [PMID: 25501566 PMCID: PMC4972502 DOI: 10.1002/eji.201445218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 10/31/2014] [Accepted: 12/09/2014] [Indexed: 12/16/2022]
Abstract
In general, a long-lasting immune response to viruses is achieved when they are infectious and replication competent. In the mouse, the neutralizing antibody response to Friend murine leukemia virus is contributed by an allelic form of the enzyme Apobec3 (abbreviated A3). This is counterintuitive because A3 directly controls viremia before the onset of adaptive antiviral immune responses. It suggests that A3 also affects the antibody response directly. Here, we studied the relative size of cell populations of the adaptive immune system as a function of A3 activity. We created a transgenic mouse that expresses all seven human A3 enzymes and compared it to WT and mouse A3-deficient mice. A3 enzymes decreased the number of marginal zone B cells, but not the number of follicular B or T cells. When mouse A3 was knocked out, the retroelement hitchhiker-1 and sialyl transferases encoded by genes close to it were overexpressed three and two orders of magnitude, respectively. We suggest that A3 shifts the balance, from the fast antibody response mediated by marginal zone B cells with little affinity maturation, to a more sustained germinal center B-cell response, which drives affinity maturation and, thereby, a better neutralizing response.
Collapse
Affiliation(s)
| | - Rebecca Winkelmann
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger-Center, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthew L. Wheeler
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Maryam Shansab
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Philipp Yu
- Institut für Immunologie, Philipps-Universität Marburg, Marburg, Germany
| | - Sarah Wünsche
- Institut für Immunologie, Philipps-Universität Marburg, Marburg, Germany
| | - Anja Walchhütter
- Institut für Immunologie, Philipps-Universität Marburg, Marburg, Germany
| | - Mirjam Metzner
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger-Center, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Vettermann
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger-Center, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Dan Eilat
- Department of Medicine, Hadassah University Hospital and The Hebrew University Faculty of Medicine, Jerusalem, Israel
| | - Anthony DeFranco
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger-Center, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Wabl
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| |
Collapse
|
48
|
Mooster JL, Le Bras S, Massaad MJ, Jabara H, Yoon J, Galand C, Heesters BA, Burton OT, Mattoo H, Manis J, Geha RS. Defective lymphoid organogenesis underlies the immune deficiency caused by a heterozygous S32I mutation in IκBα. ACTA ACUST UNITED AC 2015; 212:185-202. [PMID: 25601653 PMCID: PMC4322042 DOI: 10.1084/jem.20140979] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Mooster et al. created a knock-in mouse harboring a mutation (S32I) in IκBα that has been identified in a patient with ectodermal dysplasia with immunodeficiency. The mice are characterized by defective architectural cell function; they lack lymph nodes, Peyer’s patches, splenic marginal zones, and follicular DCs and fail to develop germinal centers. These features have not been previously recognized in patients. Patients with ectodermal dysplasia with immunodeficiency (ED-ID) caused by mutations in the inhibitor of NF-κB α (IκBα) are susceptible to severe recurrent infections, despite normal T and B cell numbers and intact in vitro lymphocyte function. Moreover, the outcome of hematopoietic stem cell transplantation (HSCT) in these patients is poor despite good engraftment. Mice heterozygous for the IκBα S32I mutation found in patients exhibited typical features of ED-ID. Strikingly, the mice lacked lymph nodes, Peyer’s patches, splenic marginal zones, and follicular dendritic cells and failed to develop contact hypersensitivity (CHS) or form germinal centers (GCs), all features not previously recognized in patients and typical of defective noncanonical NF-κB signaling. Lymphotoxin β receptor (LTβR)–driven induction of chemokines and adhesion molecules mediated by both canonical and noncanonical NF-κB pathways was impaired, and levels of p100 were markedly diminished in the mutant. IκBα mutant→Rag2−/−, but not WT→IκBα mutant, bone marrow chimeras formed proper lymphoid organs and developed CHS and GCs. Defective architectural cell function explains the immunodeficiency and poor outcome of HSCT in patients with IκBα deficiency and suggests that correction of this niche is critical for reconstituting their immune function.
Collapse
Affiliation(s)
- Jana L Mooster
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Severine Le Bras
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Michel J Massaad
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Haifa Jabara
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Juhan Yoon
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Claire Galand
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Balthasar A Heesters
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, Netherlands
| | - Oliver T Burton
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Hamid Mattoo
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114
| | - John Manis
- Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Raif S Geha
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
49
|
Abstract
The sialic acids N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc) differ by a single oxygen atom and are widely found at the terminal position of glycans on vertebrate cell surfaces. In animals capable of synthesizing Neu5Gc, most tissues and cell types express both sialic acids, in proportions that vary between species. However, it has long been noted that Neu5Gc is consistently expressed at trace to absent levels in the brains of all vertebrates studied to date. Although several reports have claimed to find low levels of Neu5Gc-containing glycans in neural tissue, no study definitively excludes the possibility of contamination with glycans from non-neural cell types. This distribution of a molecule - prominently but variably expressed in extraneural tissues but very low or absent in the brain - is, to our knowledge, unique. The evolutionarily conserved brain-specific suppression of Neu5Gc may indicate that its presence is toxic to this organ; however, no studies to date have directly addressed this very interesting question. Here we provide a historical background to this issue and discuss potential mechanisms causing the suppression of Neu5Gc expression in brain tissue, as well as mechanisms by which Neu5Gc may exert the presumed toxicity. Finally, we discuss future approaches towards understanding the mechanisms and implications of this unusual finding.
Collapse
Affiliation(s)
- Leela R L Davies
- Glycobiology Research and Training Center, Center for Academic Research and Training in Anthropogeny, Biomedical Sciences Graduate Program, Departments of Medicine and Cellular and Molecular Medicine, University of California at San Diego, 9500 Gilman Dr., MC 0687, La Jolla, CA, 92093-0687, USA
| | | |
Collapse
|
50
|
Gerardy-Schahn R, Delannoy P, von Itzstein M. Advanced Technologies in Sialic Acid and Sialoglycoconjugate Analysis. Top Curr Chem (Cham) 2015; 367:75-103. [PMID: 26017094 PMCID: PMC7122537 DOI: 10.1007/128_2013_458] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Although the structural diversity of sialic acid (Sia) is rapidly expanding, understanding of its biological significance has lagged behind. Advanced technologies to detect and probe diverse structures of Sia are absolutely necessary not only to understand further biological significance but also to pursue medicinal and industrial applications. Here we describe analytical methods for detection of Sia that have recently been developed or improved, with a special focus on 9-O-acetylated N-acetylneuraminic acid (Neu5,9Ac), N-glycolylneuraminic acid (Neu5Gc), deaminoneuraminic acid (Kdn), O-sulfated Sia (SiaS), and di-, oligo-, and polysialic acid (diSia/oligoSia/polySia) in glycoproteins and glycolipids. Much more attention has been paid to these Sia and sialoglycoconjugates during the last decade, in terms of regulation of the immune system, neural development and function, tumorigenesis, and aging.
Collapse
Affiliation(s)
| | - Philippe Delannoy
- Lille University of Science and Technology, Villeneuve d'Ascq Cedex, France
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Southport, Queensland Australia
| |
Collapse
|