Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 282
1.
Microbiol Spectr ; 9(3): e0039921, 2021 12 22.
Article En | MEDLINE | ID: mdl-34878295

Escherichia coli K1 causes bacteremia and meningitis in human neonates. The K1 capsule, an α2,8-linked polysialic acid (PSA) homopolymer, is its essential virulence factor. PSA is usually partially modified by O-acetyl groups. It is known that O-acetylation alters the antigenicity of PSA, but its impact on the interactions between E. coli K1 and host cells is unclear. In this study, a phase variant was obtained by passage of E. coli K1 parent strain, which expressed a capsule with 44% O-acetylation whereas the capsule of the parent strain has only 3%. The variant strain showed significantly reduced adherence and invasion to macrophage-like cells in comparison to the parent strain. Furthermore, we found that O-acetylation of PSA enhanced the modulation of trafficking of E. coli-containing vacuoles (ECV), enabling them to avoid fusing with lysosomes in these cells. Intriguingly, by using quartz crystal microbalance, we demonstrated that the PSA purified from the parent strain interacted with human sialic acid-binding immunoglobulin-like lectins (Siglecs), including Siglec-5, Siglec-7, Siglec-11, and Siglec-14. However, O-acetylated PSA from the variant interacted much less and also suppressed the production of Siglec-mediated proinflammatory cytokines. The adherence of the parent strain to human macrophage-like cells was significantly blocked by monoclonal antibodies against Siglec-11 and Siglec-14. Furthermore, the variant strain caused increased bacteremia and higher lethality in neonatal mice compared to the parent strain. These data elucidate that O-acetylation of K1 capsule enables E. coli to escape from Siglec-mediated innate immunity and lysosomal degradation; therefore, it is a strategy used by E. coli K1 to regulate its virulence. IMPORTANCE Escherichia coli K1 is a leading cause of neonatal meningitis. The mortality and morbidity of this disease remain significantly high despite antibiotic therapy. One major limitation on advances in prevention and therapy for meningitis is an incomplete understanding of its pathogenesis. E. coli K1 is surrounded by PSA, which is observed to have high-frequency variation of O-acetyl modification. Here, we present an in-depth study of the function of O-acetylation in PSA at each stage of host-pathogen interaction. We found that a high level of O-acetylation significantly interfered with Siglec-mediated bacterial adherence to macrophage-like cells, and blunted the proinflammatory response. Furthermore, the O-acetylation of PSA modulated the trafficking of ECVs to prevent them from fusing with lysosomes, enabling them to escape degradation by lysozymes within these cells. Elucidating how subtle modification of the capsule enhances bacterial defenses against host innate immunity will enable the future development of effective drugs or vaccines against infection by E. coli K1.


Bacterial Capsules/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Sialic Acids/immunology , Acetylation , Animals , Escherichia coli/genetics , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Lysosomes/immunology , Lysosomes/microbiology , Male , Mice , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Vacuoles/immunology , Vacuoles/microbiology
2.
Int J Mol Sci ; 22(7)2021 Mar 26.
Article En | MEDLINE | ID: mdl-33810246

Autoimmune disease development depends on multiple factors, including genetic and environmental. Abnormalities such as sialylation levels and/or quality have been recently highlighted. The adjunction of sialic acid at the terminal end of glycoproteins and glycolipids is essential for distinguishing between self and non-self-antigens and the control of pro- or anti-inflammatory immune reactions. In autoimmunity, hyposialylation is responsible for chronic inflammation, the anarchic activation of the immune system and organ lesions. A detailed characterization of this mechanism is a key element for improving the understanding of these diseases and the development of innovative therapies. This review focuses on the impact of sialylation in autoimmunity in order to determine future treatments based on the regulation of hyposialylation.


Autoantibodies/metabolism , Autoimmune Diseases/immunology , Protein Processing, Post-Translational , Sialic Acids/metabolism , Animals , Autoantibodies/immunology , Autoimmune Diseases/therapy , Humans , Immunophenotyping/methods , Precision Medicine/methods , Sialic Acids/immunology
3.
Eur J Immunol ; 51(6): 1412-1422, 2021 06.
Article En | MEDLINE | ID: mdl-33576494

Heterologous polyclonal antibodies might represent an alternative to the use of convalescent plasma or monoclonal antibodies (mAbs) in coronavirus disease (COVID-19) by targeting multiple antigen epitopes. However, heterologous antibodies trigger human natural xenogeneic antibody responses particularly directed against animal-type carbohydrates, mainly the N-glycolyl form of the neuraminic acid (Neu5Gc) and the α1,3-galactose, potentially leading to serum sickness or allergy. Here, we immunized cytidine monophosphate-N-acetylneuraminic acid hydroxylase and α1,3-galactosyl-transferase (GGTA1) double KO pigs with the Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike receptor binding domain to produce glyco-humanized polyclonal neutralizing antibodies lacking Neu5Gc and α1,3-galactose epitopes. Animals rapidly developed a hyperimmune response with anti-SARS-CoV-2 end-titers binding dilutions over one to a million and end-titers neutralizing dilutions of 1:10 000. The IgG fraction purified and formulated following clinical Good Manufacturing Practices, named XAV-19, neutralized spike/angiotensin converting enzyme-2 interaction at a concentration <1 µg/mL, and inhibited infection of human cells by SARS-CoV-2 in cytopathic assays. We also found that pig GH-pAb Fc domains fail to interact with human Fc receptors, thereby avoiding macrophage-dependent exacerbated inflammatory responses and a possible antibody-dependent enhancement. These data and the accumulating safety advantages of using GH-pAbs in humans warrant clinical assessment of XAV-19 against COVID-19.


Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/therapy , SARS-CoV-2/immunology , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/genetics , Antibodies, Viral/pharmacology , COVID-19/genetics , Galactosyltransferases/deficiency , Galactosyltransferases/immunology , HEK293 Cells , Humans , Immunization, Passive , SARS-CoV-2/genetics , Sialic Acids/genetics , Sialic Acids/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Swine , COVID-19 Serotherapy
4.
mBio ; 12(1)2021 01 19.
Article En | MEDLINE | ID: mdl-33468699

Surface expression of the common vertebrate sialic acid (Sia) N-acetylneuraminic acid (Neu5Ac) by commensal and pathogenic microbes appears structurally to represent "molecular mimicry" of host sialoglycans, facilitating multiple mechanisms of host immune evasion. In contrast, ketodeoxynonulosonic acid (Kdn) is a more ancestral Sia also present in prokaryotic glycoconjugates that are structurally quite distinct from vertebrate sialoglycans. We detected human antibodies against Kdn-terminated glycans, and sialoglycan microarray studies found these anti-Kdn antibodies to be directed against Kdn-sialoglycans structurally similar to those on human cell surface Neu5Ac-sialoglycans. Anti-Kdn-glycan antibodies appear during infancy in a pattern similar to those generated following incorporation of the nonhuman Sia N-glycolylneuraminic acid (Neu5Gc) onto the surface of nontypeable Haemophilus influenzae (NTHi), a human commensal and opportunistic pathogen. NTHi grown in the presence of free Kdn took up and incorporated the Sia into its lipooligosaccharide (LOS). Surface display of the Kdn within NTHi LOS blunted several virulence attributes of the pathogen, including Neu5Ac-mediated resistance to complement and whole blood killing, complement C3 deposition, IgM binding, and engagement of Siglec-9. Upper airway administration of Kdn reduced NTHi infection in human-like Cmah null (Neu5Gc-deficient) mice that express a Neu5Ac-rich sialome. We propose a mechanism for the induction of anti-Kdn antibodies in humans, suggesting that Kdn could be a natural and/or therapeutic "Trojan horse" that impairs colonization and virulence phenotypes of free Neu5Ac-assimilating human pathogens.IMPORTANCE All cells in vertebrates are coated with a dense array of glycans often capped with sugars called sialic acids. Sialic acids have many functions, including serving as a signal for recognition of "self" cells by the immune system, thereby guiding an appropriate immune response against foreign "nonself" and/or damaged cells. Several pathogenic bacteria have evolved mechanisms to cloak themselves with sialic acids and evade immune responses. Here we explore a type of sialic acid called "Kdn" (ketodeoxynonulosonic acid) that has not received much attention in the past and compare and contrast how it interacts with the immune system. Our results show potential for the use of Kdn as a natural intervention against pathogenic bacteria that take up and coat themselves with external sialic acid from the environment.


Antigens, CD/immunology , Haemophilus Infections/immunology , Haemophilus influenzae/immunology , Host-Pathogen Interactions/immunology , N-Acetylneuraminic Acid/chemistry , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Sialic Acids/immunology , Animals , Antibodies/chemistry , Antibodies/metabolism , Antigens, CD/metabolism , Biological Transport , Complement C3/immunology , Complement C3/metabolism , Female , Glycoconjugates/chemistry , Glycoconjugates/immunology , Haemophilus Infections/genetics , Haemophilus Infections/microbiology , Haemophilus influenzae/chemistry , Host-Pathogen Interactions/genetics , Humans , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Mice , Mice, Inbred C57BL , Molecular Mimicry/genetics , Molecular Mimicry/immunology , N-Acetylneuraminic Acid/immunology , Protein Binding , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sialic Acids/chemistry , Sugar Acids/chemistry , Sugar Acids/immunology
5.
Cell Mol Life Sci ; 78(4): 1637-1653, 2021 Feb.
Article En | MEDLINE | ID: mdl-32725371

Polysialic acid (polySia) emerges as a novel regulator of microglia activity. We recently identified polysialylated proteins in the Golgi compartment of murine microglia that are released in response to inflammatory stimulation. Since exogenously added polySia is able to attenuate the inflammatory response, we proposed that the release of polysialylated proteins constitutes a mechanism for negative feedback regulation of microglia activation. Here, we demonstrate that translocation of polySia from the Golgi to the cell surface can be induced by calcium depletion of the Golgi compartment and that polysialylated proteins are continuously released for at least 24 h after the onset of inflammatory stimulation. The latter was unexpected, because polySia signals detected by immunocytochemistry are rapidly depleted. However, it indicates that the amount of released polySia is much higher than anticipated based on immunostaining. This may be crucial for microglial responses during traumatic brain injury (TBI), as we detected polySia signals in activated microglia around a stab wound in the adult mouse brain. In BV2 microglia, the putative polySia receptor Siglec-E is internalized during lipopolysaccharide (LPS)-induced activation and in response to polySia exposure, indicating interaction. Correspondingly, CRISPR/Cas9-mediated Siglec-E knockout prevents inhibition of pro inflammatory activation by exogenously added polySia and leads to a strong increase of the LPS response. A comparable increase of LPS-induced activation has been observed in microglia with abolished polySia synthesis. Together, these results indicate that the release of the microglia-intrinsic polySia pool, as implicated in TBI, inhibits the inflammatory response by acting as a trans-activating ligand of Siglec-E.


Inflammation/genetics , Microglia/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Sialic Acids/genetics , Animals , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , CRISPR-Cas Systems/genetics , Cells, Cultured , Feedback, Physiological/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/genetics , Humans , Inflammation/immunology , Inflammation/pathology , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Microglia/immunology , Microglia/pathology , Phagocytosis/drug effects , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Sialic Acids/immunology
6.
J Cereb Blood Flow Metab ; 41(5): 1039-1057, 2021 05.
Article En | MEDLINE | ID: mdl-32703109

Neonatal hypoxia-ischemia (nHI) disrupts hippocampal GABAergic development leading to memory deficits in mice. Polysialic-acid neural-cell adhesion molecule (PSA-NCAM) developmentally declines to trigger GABAergic maturation. We hypothesized that nHI changes PSA-NCAM abundance and cellular distribution, impairing GABAergic development, and marking nascent neurodegeneration. Cell degeneration, atrophy, and PSA-NCAM immunoreactivity (IR) were measured in CA1 of nHI-injured C57BL6 mice related to: (i) cellular subtype markers; (ii) GAD65/67 and synatophysin (SYP), pre-synaptic markers; (iii) phospho-Ser396Tau, cytoskeletal marker; and (iv) GAP43, axonalregeneration marker. PSA-NCAM IR was minimal in CA1 of shams at P11. After nHI, PSA-NCAM IR was increased in injured pyramidal cells (PCs), minimal in parvalbumin (PV)+INs, and absent in glia. PSA-NCAM IR correlated with injury severity and became prominent in perikaryal cytoplasm at P18. GAD65/67 and SYP IRs only weakly related to PSA-NCAM after nHI. Injured phospho-Ser396Tau+ PCs and PV+INs variably co-expressed PSA-NCAM at P40. While PCs with cytoplasmic marginalized PSA-NCAM had increased perisomatic GAP43, those with perikaryal cytoplasmic PSA-NCAM had minimal GAP43. PSA-NCAM increased in serum of nHI-injured mice. Increased PSA-NCAM is likely a generic acute response to nHI brain injury. PSA-NCAM aberrant cellular localization may aggravate neuronal degeneration. The significance of PSA-NCAM as a biomarker of recovery from nHI and nascent neurodegeneration needs further study.


Brain Injuries/metabolism , Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neurodegenerative Diseases/metabolism , Sialic Acids/metabolism , Animals , Brain Injuries/pathology , CA1 Region, Hippocampal/metabolism , Female , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Glutamate Decarboxylase/metabolism , Hypoxia-Ischemia, Brain/complications , Injury Severity Score , Male , Memory Disorders/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , Neural Cell Adhesion Molecule L1/immunology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Sialic Acids/immunology , Synaptophysin/metabolism , tau Proteins/metabolism
7.
Int J Mol Sci ; 21(15)2020 Jul 31.
Article En | MEDLINE | ID: mdl-32752058

Sialic acids (Sias) are the most abundant terminal sugar residues of glycoproteins and glycolipids on the surface of mammalian cells. The nervous tissue is the organ with the highest expression level of Sias. The 'sialylation' of glycoconjugates is performed via sialyltransferases, whereas 'desialylation' is done by sialidases or is a possible consequence of oxidative damage. Sialic acid residues on the neural cell surfaces inhibit complement and microglial activation, as well as phagocytosis of the underlying structures, via binding to (i) complement factor H (CFH) or (ii) sialic acid-binding immunoglobulin-like lectin (SIGLEC) receptors. In contrast, activated microglial cells show sialidase activity that desialylates both microglia and neurons, and further stimulates innate immunity via microglia and complement activation. The desialylation conveys neurons to become susceptible to phagocytosis, as well as triggers a microglial phagocytosis-associated oxidative burst and inflammation. Dysfunctions of the 'Sia-SIGLEC' and/or 'Sia-complement' axes often lead to neurological diseases. Thus, Sias on glycoconjugates of the intact glycocalyx and its desialylation are major regulators of neuroinflammation.


Immunity, Innate/genetics , Nerve Tissue/metabolism , Sialic Acids/genetics , Sialyltransferases/genetics , Glycoconjugates/genetics , Glycoconjugates/immunology , Humans , Macrophages , Microglia/immunology , Microglia/metabolism , Nerve Tissue/immunology , Neurons/metabolism , Neurons/pathology , Phagocytosis/genetics , Sialic Acids/immunology , Sialic Acids/metabolism , Sialyltransferases/immunology
8.
Anal Biochem ; 600: 113743, 2020 07 01.
Article En | MEDLINE | ID: mdl-32325083

Male and female immune systems are strikingly different and yet little is known about sex differences in immune glycans, though glycans play central roles in regulating the immune response. Polysialic acid (polySia) occurs on the majority of leukocytes and is a potent immunomodulatory glycan which enables cell migration and serves as an immune checkpoint. Due to widespread influence of polySia on the immune system, we aimed to characterize its levels in serum, its presence on specific proteins, and differences in the amounts of polySia in male and female serum. However, polySia is difficult to quantify and detect on specific proteins, which makes it challenging to elucidate the molecular details of polySia function. We developed a sandwich ELISA that allows for the quantification of polySia as well as specific polysialylated proteins in complex mixtures without any pretreatment or harsh conditions. The assay is quick, linear, and robust under a wide variety of conditions and gave a limit of detection of approximately 0.2 ng polySia per mL of serum. We then quantified polySia and polysialylated CD56 in human and mouse serum. These studies strongly support our hypothesis of differences in glycosylation between the sexes as significantly less polySia was observed in female samples than in male samples.


Enzyme-Linked Immunosorbent Assay , Sialic Acids/blood , Animals , Female , Healthy Volunteers , Humans , Male , Mice , Mice, Inbred C57BL , Sex Characteristics , Sialic Acids/immunology
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165692, 2020 05 01.
Article En | MEDLINE | ID: mdl-31972227

Many important pathogen-host interactions rely on highly specific carbohydrate binding events. In the case of the protozoan Trypanosoma cruzi, the causative agent of Chagas disease, glycointeractions involving sialic acid (SA) residues are pivotal for parasite infectivity, escape from immune surveillance and pathogenesis. Though unable to synthesize SA de novo, T. cruzi displays a unique trans-Sialidase (TS) enzyme, which is able to cleave terminal SA residues from host donor glycoconjugates and transfer them onto parasite surface mucins, thus generating protective/adhesive structures. In addition, this parasite sheds TS into the bloodstream, as a way of modifying the surface SA signature, and thereby the signaling/functional properties of mammalian host target cells on its own advantage. Here, we discuss the pathogenic aspects of T. cruzi TS: its molecular adaptations, the multiplicity of interactions in which it is involved during infections, and the array of novel and appealing targets for intervention in Chagas disease provided by TS-remodeled sialoglycophenotypes.


Chagas Disease/immunology , Glycoproteins/metabolism , Host-Parasite Interactions/immunology , Neuraminidase/metabolism , Polysaccharides/immunology , Protozoan Proteins/metabolism , Trypanosoma cruzi/pathogenicity , Animals , Chagas Disease/parasitology , Glycoproteins/immunology , Humans , Immune Evasion , Neuraminidase/immunology , Polysaccharides/chemistry , Polysaccharides/metabolism , Protozoan Proteins/immunology , Sialic Acids/chemistry , Sialic Acids/immunology , Sialic Acids/metabolism , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/immunology , Virulence Factors/immunology , Virulence Factors/metabolism
10.
Front Immunol ; 10: 2360, 2019.
Article En | MEDLINE | ID: mdl-31649671

Altered sialylation is generally maintained by a fine balance between sialidases and sialyltransferases, which plays an essential role during disease pathogenesis. TLR4 is a membrane-bound highly sialylated glycoprotein predominantly having α2,3-linked sialic acids. It is one of the most important client molecules in the anti-leishmanial innate immune arm. Here, we initiated a comprehensive study on the modulation of TLR4 sialylation in Leishmania donovani (L. d)-infected macrophages by a mammalian sialidase/neuraminidase-1 (Neu1) having substrate specificity toward α2,3-linked sialic acids. We observed reduced membrane-associated Neu1 with its decreased enzyme activity in infected macrophages. Moreover, we demonstrated reduced association of Neu1 with TLR4 leading to enhanced sialylation of TLR4 in these infected cells. Conversely, Neu1 over expression exhibited enhanced association of TLR4 with Neu1 leading to reduced sialylation which possibly linked to increased association of TLR4 with its downstream adaptor protein, MyD88. This, in turn, activated downstream MAP kinase signaling pathway, with enhanced nuclear translocation of NFκB that resulted in increased genetic and protein levels expression of Th1 cytokines and effector molecule nitric oxide secretion which ultimately leads to reduced parasite burden in macrophages. This was further validated by Neu1 silencing in infected macrophages which reversed such a situation. Such events strongly confirm the importance of Neu1 in modulation of TLR4 sialylation during parasite infection resulting in impairment of innate immune response. Furthermore, decreased membrane-bound Neu1 in infected macrophages could be attributed to its reduced tyrosine-phosphorylation as well as diminished association with cathepsin A. Both these phenomenon possibly play significant roles in inhibiting translocation of the sialidase from cytosol to membrane. Taken together, our study first time demonstrated impaired translocation of cytosolic Neu1 to the membrane of L. donovani-infected macrophages due to impaired phosphorylation of this enzyme. This novel finding establishes a link between enhanced α2,3-linked sialic acids on TLR4 and reduced membrane-bound Neu1 which plays a significant role for inhibiting downstream signaling to establish successful infection in the host cells.


Leishmania donovani/immunology , Leishmaniasis, Visceral , MAP Kinase Signaling System/immunology , Macrophages , Neuraminidase/immunology , Toll-Like Receptor 4/immunology , Animals , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/pathology , Macrophages/immunology , Macrophages/parasitology , Macrophages/pathology , Mesocricetus , Mice , Mice, Inbred BALB C , Sialic Acids/immunology
11.
PLoS Pathog ; 15(6): e1007896, 2019 06.
Article En | MEDLINE | ID: mdl-31233555

Streptococcus gordonii and Streptococcus sanguinis are primary colonizers of the tooth surface. Although generally non-pathogenic in the oral environment, they are a frequent cause of infective endocarditis. Both streptococcal species express a serine-rich repeat surface adhesin that mediates attachment to sialylated glycans on mucin-like glycoproteins, but the specific sialoglycan structures recognized can vary from strain to strain. Previous studies have shown that sialoglycan binding is clearly important for aortic valve infections caused by some S. gordonii, but this process did not contribute to the virulence of a strain of S. sanguinis. However, these streptococci can bind to different subsets of sialoglycan structures. Here we generated isogenic strains of S. gordonii that differ only in the type and range of sialoglycan structures to which they adhere and examined whether this rendered them more or less virulent in a rat model of endocarditis. The findings indicate that the recognition of specific sialoglycans can either enhance or diminish pathogenicity. Binding to sialyllactosamine reduces the initial colonization of mechanically-damaged aortic valves, whereas binding to the closely-related trisaccharide sialyl T-antigen promotes higher bacterial densities in valve tissue 72 hours later. A surprising finding was that the initial attachment of streptococci to aortic valves was inversely proportional to the affinity of each strain for platelets, suggesting that binding to platelets circulating in the blood may divert bacteria away from the endocardial surface. Importantly, we found that human and rat platelet GPIbα (the major receptor for S. gordonii and S. sanguinis on platelets) display similar O-glycan structures, comprised mainly of a di-sialylated core 2 hexasaccharide, although the rat GPIbα has a more heterogenous composition of modified sialic acids. The combined results suggest that streptococcal interaction with a minor O-glycan on GPIbα may be more important than the over-all affinity for GPIbα for pathogenic effects.


Endocarditis, Bacterial/immunology , Glycoproteins/immunology , Sialic Acids/immunology , Streptococcal Infections/immunology , Streptococcus gordonii/immunology , Streptococcus sanguis/immunology , Animals , Disease Models, Animal , Endocarditis, Bacterial/pathology , Female , Humans , Male , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Streptococcal Infections/pathology , Streptococcus gordonii/pathogenicity , Streptococcus sanguis/pathogenicity
12.
PLoS Pathog ; 15(6): e1007871, 2019 06.
Article En | MEDLINE | ID: mdl-31226171

Infection of host cells by Toxoplasma gondii is an active process, which is regulated by secretion of microneme (MICs) and rhoptry proteins (ROPs and RONs) from specialized organelles in the apical pole of the parasite. MIC1, MIC4 and MIC6 assemble into an adhesin complex secreted on the parasite surface that functions to promote infection competency. MIC1 and MIC4 are known to bind terminal sialic acid residues and galactose residues, respectively and to induce IL-12 production from splenocytes. Here we show that rMIC1- and rMIC4-stimulated dendritic cells and macrophages produce proinflammatory cytokines, and they do so by engaging TLR2 and TLR4. This process depends on sugar recognition, since point mutations in the carbohydrate-recognition domains (CRD) of rMIC1 and rMIC4 inhibit innate immune cells activation. HEK cells transfected with TLR2 glycomutants were selectively unresponsive to MICs. Following in vitro infection, parasites lacking MIC1 or MIC4, as well as expressing MIC proteins with point mutations in their CRD, failed to induce wild-type (WT) levels of IL-12 secretion by innate immune cells. However, only MIC1 was shown to impact systemic levels of IL-12 and IFN-γ in vivo. Together, our data show that MIC1 and MIC4 interact physically with TLR2 and TLR4 N-glycans to trigger IL-12 responses, and MIC1 is playing a significant role in vivo by altering T. gondii infection competency and murine pathogenesis.


Cell Adhesion Molecules/immunology , Dendritic Cells/immunology , Immunity, Innate , Macrophages/immunology , Protozoan Proteins/immunology , Sialic Acids/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Interleukin-12/immunology , Mice , Mice, Knockout , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Toxoplasmosis, Animal/genetics
13.
Front Immunol ; 10: 807, 2019.
Article En | MEDLINE | ID: mdl-31057542

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.


Autoantibodies/immunology , Immune System Diseases , Mixed Function Oxygenases , N-Acetylneuraminic Acid/immunology , Sialic Acids/immunology , Animals , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/immunology , N-Acetylneuraminic Acid/genetics , Sialic Acids/genetics
14.
Glycobiology ; 29(7): 557-564, 2019 07 01.
Article En | MEDLINE | ID: mdl-30989215

The activation of human naïve CD4+ T cells, responsible for orchestrating the immune response, has been reported to cause increased de novo sialylation and overexpression of the genes coding for polysialyltransferases ST8SiaII and ST8SiaIV, suggesting the potential of CD4+ T cells to synthesize polysialic acid (PSA), a type of glycosylation not previously described in these cells. PSA has been found as a post-translational modification in a limited number of mammalian proteins, having a very relevant role in modulating interactions due to its characteristic biophysical properties. In this work, we confirm that human CD4+ T cells express both polysialyltransferases and synthesize PSA, as assessed with the anti-PSA monoclonal antibody (mAb) 12E3. The expression of PSA in resting cells was found restricted to a cell subpopulation (PSA+), that after anti-CD3/anti-CD28 mAbs mediated activation, increased in percentage and mean fluorescence intensity (MFI) expression. Additionally, through ST8SIAII and ST8SIAIV-silencing and by measuring the mRNA of IL-2, IL-2R and IFN-γ, we show that PSA is involved in modulating the activation response of CD4+ T cells.


CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Sialic Acids/biosynthesis , CD4-Positive T-Lymphocytes/cytology , Glycosylation , Humans , Sialic Acids/immunology , Sialyltransferases/metabolism
15.
Front Immunol ; 10: 3024, 2019.
Article En | MEDLINE | ID: mdl-31998308

Select residues in the biantennary sugar moiety attached to the fragment crystallizable of immunoglobulin G (IgG) antibodies can modulate IgG effector functions. Thus, afucosylated IgG glycovariants have enhanced cytotoxic activity, whereas IgG glycovariants rich in terminal sialic acid residues can trigger anti-inflammatory effects. More recent evidence suggests that terminal α2,6 linked sialic acids can be attached to antibodies post IgG secretion. These findings raise concerns for the use of therapeutic antibodies as they may change their glycosylation status in the patient and hence affect their activity. To investigate to what extent B cell extrinsic sialylation processes modify therapeutic IgG preparations in vivo, we analyzed changes in human intravenous IgG (IVIg) sialylation upon injection in mice deficient in B cells or in mice lacking the sialyltransferase 1, which catalyzes the addition of α2,6 linked sialic acid residues. By performing a time course of IgG glycan analysis with HILIC-UPLC-FLR (plus MS) and xCGE-LIF our study suggests that therapeutic IgG glycosylation is stable upon injection in vivo. Only a very small fraction of IgG molecules acquired sialic acid structures predominantly in the Fab- but not the Fc-portion upon injection in vivo, suggesting that therapeutic antibody glycosylation will remain stable upon injection in vivo.


B-Lymphocytes/immunology , Immunoglobulin G/immunology , Inflammation/immunology , Polysaccharides/immunology , Animals , Glycosylation , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulins, Intravenous/immunology , Mice , Mice, Inbred C57BL , Sialic Acids/immunology
16.
Front Immunol ; 9: 2276, 2018.
Article En | MEDLINE | ID: mdl-30333834

B cells express various inhibitory co-receptors including CD22, CD72, and Siglec-G. These receptors contain immunoreceptor tyrosine-based inhibition motifs (ITIMs) in the cytoplasmic region. Although many of the inhibitory co-receptors negatively regulate BCR signaling by activating SH2-containing protein tyrosine phosphatase 1 (SHP-1), different inhibitory co-receptors have distinct functional properties. CD22, Siglec-G, and CD72 preferentially regulate tonic signaling in conventional B cells, B-1 cell homeostasis, and development of lupus-like disease, respectively. CD72 recognizes RNA-related lupus self-antigen Sm/RNP as a ligand. This ligand recognition recruits CD72 to BCR in Sm/RNP-reactive B cells thereby suppressing production of anti-Sm/RNP autoantibody involved in the pathogenesis of lupus. In contrast, Siglec-G recognizes α2,3 as well as α2,6 sialic acids whereas CD22 recognizes α2,6 sialic acid alone. Because glycoproteins including BCR are dominantly glycosylated with α2,3 sialic acids in B-1 cells, Siglec-G but not CD22 recruits BCR as a ligand specifically in B-1 cells, and regulates B-1 cell homeostasis by suppressing BCR signaling in B-1 cells. Thus, recognition of distinct ligands determines functional properties of different inhibitory B cell co-receptors.


Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Autoimmunity , B-Lymphocyte Subsets/immunology , Homeostasis/immunology , Receptors, Antigen, B-Cell/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Animals , B-Lymphocyte Subsets/cytology , Humans , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Sialic Acids/immunology , Signal Transduction/immunology
17.
Xenotransplantation ; 25(6): e12424, 2018 11.
Article En | MEDLINE | ID: mdl-29932472

All living cells are covered with a dense "sugar-coat" of carbohydrate chains (glycans) conjugated to proteins and lipids. The cell surface glycome is determined by a non-template driven process related to the collection of enzymes that assemble glycans in a sequential manner. In mammals, many of these glycans are topped with sialic acids (Sia), a large family of acidic sugars. The "Sialome" is highly diverse owing to various Sia types, linkage to underlying glycans, range of carriers, and complex spatial organization. Presented at the front of cells, Sia play a major role in immunity and recognition of "self" versus "non-self," largely mediated by the siglecs family of Sia-binding host receptors. Albeit many mammalian pathogens have evolved to hijack this recognition system to avoid host immune attack, presenting a fascinating host-pathogen evolutionary arms race. Similarly, cancer cells exploit Sia for their own survival and propagation. As part of this ongoing fitness, humans lost the ability to synthesize the Sia type N-glycolylneuraminic acid (Neu5Gc), in contrast to other mammals. While this loss had provided an advantage against certain pathogens, humans are continuously exposed to Neu5Gc through mammalian-derived diet (eg, red meat), consequently generating a complex immune response against it. Circulating anti-Neu5Gc antibodies together with Neu5Gc on some human tissues mediate chronic inflammation "xenosialitis" that exacerbate various human diseases (eg, cancer and atherosclerosis). Similarly, Neu5Gc-containing xenografts are exposed to human anti-Neu5Gc antibodies with implications to sustainability. This review aimed to provide a glimpse into the evolution of Sia and their implications to xenotransplantation.


Heterografts/drug effects , Polysaccharides/immunology , Sialic Acids/pharmacology , Transplantation, Heterologous , Animals , Heterografts/immunology , Humans , Immunoglobulins/immunology , Sialic Acids/chemistry , Sialic Acids/immunology , Transplantation, Heterologous/methods
18.
Semin Immunol ; 39: 102-110, 2018 10.
Article En | MEDLINE | ID: mdl-29903548

Antibodies are antigen recognizing immunoglobulins with an amazingly diverse repertoire in the antigen specific domain. The diversity of the antibody response is further increased by modifications such as somatic recombination and hypermutation. Furthermore, variation in the isotype and post-translational modifications such as Fc glycosylation further increase diversity of the effector functions. In particular variations in the glycan structures contribute significantly to the functional capacities of the antibodies. This is of particular interest given the dynamic nature of these modifications that is strongly influenced by the inflammatory environment. Intriguingly, the glycan profile of antibodies has been unravelled in great detail in inflammatory (auto)immune diseases but received only limited attention in the area of infectious diseases and vaccination. Here, we reviewed the current knowledge on immunoglobulin glycosylation and specifically focussed on studies in the field of infectious diseases and vaccination against infectious diseases, an area with a lot of interesting opportunities.


Aging/immunology , Antibodies/metabolism , HIV Infections/prevention & control , Influenza, Human/prevention & control , Protein Processing, Post-Translational , Viral Vaccines/administration & dosage , Antibodies/chemistry , Antibodies/genetics , Antigens/genetics , Antigens/immunology , Antigens/metabolism , Carbohydrate Sequence , Fucose/immunology , Fucose/metabolism , Galactose/immunology , Galactose/metabolism , Glycosylation , HIV Infections/immunology , HIV Infections/virology , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Inflammation , Influenza, Human/immunology , Influenza, Human/virology , Sialic Acids/immunology , Sialic Acids/metabolism , Vaccination , Viral Vaccines/immunology
19.
Cell Immunol ; 333: 58-64, 2018 11.
Article En | MEDLINE | ID: mdl-29685495

ST6Gal1 is a critical sialyltransferase enzyme that controls the addition of α2,6-linked sialic acids to the termini of glycans. Attachment of sialic acids to glycoproteins as a posttranslational modification influences cellular responses, and is a well-known modifier of immune cell behavior. ST6Gal1 activity impacts processes such as: effector functions of immunoglobulin G via Fc sialylation, hematopoietic capacity by hematopoietic stem and progenitor cell surface sialylation, and lymphocyte activation thresholds though CD22 engagement and inhibition of galectins. This review summarizes recent studies that suggest α2,6 sialylation by ST6Gal1 has an immunoregulatory effect on immune reactions.


Immunoglobulin G/immunology , Immunologic Factors/immunology , Leukocytes/immunology , Sialic Acids/immunology , Sialyltransferases/immunology , Animals , Humans , Lymphocyte Activation/immunology , Polysaccharides/immunology , Protein Processing, Post-Translational/immunology
20.
Mol Oral Microbiol ; 33(2): 155-167, 2018 04.
Article En | MEDLINE | ID: mdl-29235255

The oral pathogen Tannerella forsythia possesses a unique surface (S-) layer with a complex O-glycan containing a bacterial sialic acid mimic in the form of either pseudaminic acid or legionaminic acid at its terminal position. We hypothesize that different T. forsythia strains employ these stereoisomeric sugar acids for interacting with the immune system and resident host tissues in the periodontium. Here, we show how T. forsythia strains ATCC 43037 and UB4 displaying pseudaminic acid and legionaminic acid, respectively, and selected cell surface mutants of these strains modulate the immune response in monocytes and human oral keratinocytes (HOK) using a multiplex immunoassay. When challenged with T. forsythia, monocytes secrete proinflammatory cytokines, chemokines and vascular endothelial growth factor (VEGF) with the release of interleukin-1ß (IL-1ß) and IL-7 being differentially regulated by the two T. forsythia wild-type strains. Truncation of the bacteria's O-glycan leads to significant reduction of IL-1ß and regulates macrophage inflammatory protein-1. HOK infected with T. forsythia produce IL-1Ra, chemokines and VEGF. Although the two wild-type strains elicit preferential immune responses for IL-8, both truncation of the O-glycan and deletion of the S-layer result in significantly increased release of IL-8, granulocyte-macrophage colony-stimulating factor and monocyte chemoattractant protein-1. Through immunofluorescence and confocal laser scanning microscopy of infected HOK we additionally show that T. forsythia is highly invasive and tends to localize to the perinuclear region. This indicates, that the T. forsythia S-layer and attached sugars, particularly pseudaminic acid in ATCC 43037, contribute to dampening the response of epithelial tissues to initial infection and hence play a pivotal role in orchestrating the bacterium's virulence.


Cell Membrane/immunology , Cell Membrane/metabolism , Keratinocytes/immunology , Monocytes/immunology , Periodontal Diseases/immunology , Tannerella forsythia/immunology , Tannerella forsythia/pathogenicity , Cell Membrane/chemistry , Cell Membrane/genetics , Chemokines/metabolism , Cytokines/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/metabolism , Interleukin-7/metabolism , Interleukin-8/metabolism , Keratinocytes/metabolism , Keratinocytes/microbiology , Macrophage Inflammatory Proteins , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Monocytes/metabolism , Mutation , N-Acetylneuraminic Acid/immunology , Polysaccharides/immunology , Sialic Acids/immunology , Sugar Acids/immunology , Tannerella forsythia/genetics , Vascular Endothelial Growth Factor A/metabolism , Virulence
...