Skip to main content

Advertisement

Log in

Breaking the Glyco-Code of HIV Persistence and Immunopathogenesis

  • HIV Pathogenesis and Treatment (AL Landay and NS Utay, Section Editors)
  • Published:
Current HIV/AIDS Reports Aims and scope Submit manuscript

Abstract

Purpose of review

Glycoimmunology is an emerging field focused on understanding how immune responses are mediated by glycans (carbohydrates) and their interaction with glycan-binding proteins called lectins. How glycans influence immunological functions is increasingly well understood. In a parallel way, in the HIV field, it is increasingly understood how the host immune system controls HIV persistence and immunopathogenesis. However, what has mostly been overlooked, despite its potential for therapeutic applications, is the role that the host glycosylation machinery plays in modulating the persistence and immunopathogenesis of HIV. Here, we will survey four areas in which the links between glycan-lectin interactions and immunology and between immunology and HIV are well described. For each area, we will describe these links and then delineate the opportunities for the HIV field in investigating potential interactions between glycoimmunology and HIV persistence/immunopathogenesis.

Recent findings

Recent studies show that the human glycome (the repertoire of human glycan structures) plays critical roles in driving or modulating several cellular processes and immunological functions that are central to maintaining HIV infection.

Summary

Understanding the links between glycoimmunology and HIV infection may create a new paradigm for discovering novel glycan-based therapies that can lead to eradication, functional cure, or improved tolerance of lifelong infection.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, Spina CA, et al. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science. 1997;278(5341):1291–5.

    Article  CAS  PubMed  Google Scholar 

  2. Wandeler G, Johnson LF, Egger M. Trends in life expectancy of HIV-positive adults on antiretroviral therapy across the globe: comparisons with general population. Current opinion in HIV and AIDS. 2016;11(5):492–500. https://doi.org/10.1097/COH.0000000000000298.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Rodger AJ, Lodwick R, Schechter M, Deeks S, Amin J, Gilson R, et al. Mortality in well controlled HIV in the continuous antiretroviral therapy arms of the SMART and ESPRIT trials compared with the general population. Aids. 2013;27(6):973–9. https://doi.org/10.1097/QAD.0b013e32835cae9c.

    Article  CAS  PubMed  Google Scholar 

  4. Graf EH, Pace MJ, Peterson BA, Lynch LJ, Chukwulebe SB, Mexas AM, et al. Gag-positive reservoir cells are susceptible to HIV-specific cytotoxic T lymphocyte mediated clearance. PLoS One. 2013;8(8):e71879. https://doi.org/10.1371/journal.pone.0071879.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Shan L, Deng K, Shroff NS, Durand CM, Rabi SA, Yang HC, et al. Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity. 2012;36(3):491–501. https://doi.org/10.1016/j.immuni.2012.01.014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. • Abdel-Mohsen M, Chavez L, Tandon R, Chew GM, Deng X, Danesh A, et al. Human galectin-9 is a potent mediator of HIV transcription and reactivation. PLoS Pathog. 2016;12(6):e1005677. https://doi.org/10.1371/journal.ppat.1005677 This paper suggested that host glycan-lectin interactions mediate signals that define HIV transcriptional state, as it demonstrated that the human glycan-binding protein galectin-9 regulates HIV transcription.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Abdel-Mohsen M, Wang C, Strain MC, Lada SM, Deng X, Cockerham LR, et al. Select host restriction factors are associated with HIV persistence during antiretroviral therapy. Aids. 2015;29(4):411–20. https://doi.org/10.1097/QAD.0000000000000572.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Brown TT, Cole SR, Li X, Kingsley LA, Palella FJ, Riddler SA, et al. Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study. Arch Intern Med. 2005;165(10):1179–84. https://doi.org/10.1001/archinte.165.10.1179.

    Article  PubMed  Google Scholar 

  9. Seaberg EC, Munoz A, Lu M, Detels R, Margolick JB, Riddler SA, et al. Association between highly active antiretroviral therapy and hypertension in a large cohort of men followed from 1984 to 2003. Aids. 2005;19(9):953–60.

    Article  PubMed  Google Scholar 

  10. Kendall CE, Wong J, Taljaard M, Glazier RH, Hogg W, Younger J, et al. A cross-sectional, population-based study measuring comorbidity among people living with HIV in Ontario. BMC Public Health. 2014;14:161. https://doi.org/10.1186/1471-2458-14-161.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Serrano-Villar S, Perez-Elias MJ, Dronda F, Casado JL, Moreno A, Royuela A, et al. Increased risk of serious non-AIDS-related events in HIV-infected subjects on antiretroviral therapy associated with a low CD4/CD8 ratio. PloS one. 2014;9(1):e85798. https://doi.org/10.1371/journal.pone.0085798.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hasse B, Ledergerber B, Furrer H, Battegay M, Hirschel B, Cavassini M, et al. Morbidity and aging in HIV-infected persons: the Swiss HIV cohort study. Clin Infect Dis. 2011;53(11):1130–9. https://doi.org/10.1093/cid/cir626.

    Article  PubMed  Google Scholar 

  13. Pumpradit W, Ananworanich J, Lolak S, Shikuma C, Paul R, Siangphoe U, et al. Neurocognitive impairment and psychiatric comorbidity in well-controlled human immunodeficiency virus-infected Thais from the 2NN Cohort Study. J Neurovirol. 2010;16(1):76–82. https://doi.org/10.3109/13550280903493914.

    Article  PubMed  Google Scholar 

  14. Guaraldi G, Orlando G, Zona S, Menozzi M, Carli F, Garlassi E, et al. Premature age-related comorbidities among HIV-infected persons compared with the general population. Clin Infect Dis. 2011;53(11):1120–6. https://doi.org/10.1093/cid/cir627.

    Article  PubMed  Google Scholar 

  15. Negin J, Martiniuk A, Cumming RG, Naidoo N, Phaswana-Mafuya N, Madurai L, et al. Prevalence of HIV and chronic comorbidities among older adults. Aids. 2012;26(Suppl 1):S55–63. https://doi.org/10.1097/QAD.0b013e3283558459.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Lo YC, Chen MY, Sheng WH, Hsieh SM, Sun HY, Liu WC, et al. Risk factors for incident diabetes mellitus among HIV-infected patients receiving combination antiretroviral therapy in Taiwan: a case-control study. HIV Med. 2009;10(5):302–9. https://doi.org/10.1111/j.1468-1293.2008.00687.x.

    Article  CAS  PubMed  Google Scholar 

  17. Moremen KW, Tiemeyer M, Nairn AV. Vertebrate protein glycosylation: diversity, synthesis and function. Nature reviews Molecular cell biology. 2012;13(7):448–62. https://doi.org/10.1038/nrm3383.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Dalziel M, Crispin M, Scanlan CN, Zitzmann N, Dwek RA. Emerging principles for the therapeutic exploitation of glycosylation. Science. 2014;343(6166):1235681. https://doi.org/10.1126/science.1235681.

    Article  CAS  PubMed  Google Scholar 

  19. Marth JD. A unified vision of the building blocks of life. Nat Cell Biol. 2008;10(9):1015–6. https://doi.org/10.1038/ncb0908-1015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Varki A. Evolutionary forces shaping the Golgi glycosylation machinery: why cell surface glycans are universal to living cells. Cold Spring Harb Perspect Biol. 2011;3(6). https://doi.org/10.1101/cshperspect.a005462.

  21. Barrera C, Espejo R, Reyes VE. Differential glycosylation of MHC class II molecules on gastric epithelial cells: implications in local immune responses. Human immunology. 2002;63(5):384–93.

    Article  CAS  PubMed  Google Scholar 

  22. de Freitas Junior JC, Silva Bdu R, de Souza WF, de Araujo WM, Abdelhay ES, Morgado-Diaz JA. Inhibition of N-linked glycosylation by tunicamycin induces E-cadherin-mediated cell-cell adhesion and inhibits cell proliferation in undifferentiated human colon cancer cells. Cancer Chemother Pharmacol. 2011;68(1):227–38. https://doi.org/10.1007/s00280-010-1477-8.

    Article  CAS  PubMed  Google Scholar 

  23. Dwek RA, Butters TD, Platt FM, Zitzmann N. Targeting glycosylation as a therapeutic approach. Nat Rev Drug Discov. 2002;1(1):65–75. https://doi.org/10.1038/nrd708.

    Article  CAS  PubMed  Google Scholar 

  24. Walt D, Aoki-Kinoshita KF, Bendiak B, Bertozzi CR, Boons GJ, Darvill A, et al. Transforming glycoscience: a roadmap for the future. Washington: Nacional Academies Press; 2012.

    Google Scholar 

  25. Lanteri M, Giordanengo V, Hiraoka N, Fuzibet JG, Auberger P, Fukuda M, et al. Altered T cell surface glycosylation in HIV-1 infection results in increased susceptibility to galectin-1-induced cell death. Glycobiology. 2003;13(12):909–18. https://doi.org/10.1093/glycob/cwg110.

    Article  CAS  PubMed  Google Scholar 

  26. Ardman B, Sikorski MA, Settles M, Staunton DE. Human immunodeficiency virus type 1-infected individuals make autoantibodies that bind to CD43 on normal thymic lymphocytes. J Exp Med. 1990;172(4):1151–8.

    Article  CAS  PubMed  Google Scholar 

  27. Giordanengo V, Limouse M, Desroys du Roure L, Cottalorda J, Doglio A, Passeron A, et al. Autoantibodies directed against CD43 molecules with an altered glycosylation status on human immunodeficiency virus type 1 (HIV-1)-infected CEM cells are found in all HIV-1+ individuals. Blood. 1995;86(6):2302–11.

    CAS  PubMed  Google Scholar 

  28. Horiya S, MacPherson IS, Krauss IJ. Recent strategies targeting HIV glycans in vaccine design. Nature chemical biology. 2014;10(12):990–9. https://doi.org/10.1038/nchembio.1685.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ward AB, Wilson IA. The HIV-1 envelope glycoprotein structure: nailing down a moving target. Immunological reviews. 2017;275(1):21–32. https://doi.org/10.1111/imr.12507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Arnold JN, Saldova R, Galligan MC, Murphy TB, Mimura-Kimura Y, Telford JE, et al. Novel glycan biomarkers for the detection of lung cancer. Journal of proteome research. 2011;10(4):1755–64. https://doi.org/10.1021/pr101034t.

    Article  CAS  PubMed  Google Scholar 

  31. Saldova R, Royle L, Radcliffe CM, Abd Hamid UM, Evans R, Arnold JN, et al. Ovarian cancer is associated with changes in glycosylation in both acute-phase proteins and IgG. Glycobiology. 2007;17(12):1344–56. https://doi.org/10.1093/glycob/cwm100.

    Article  CAS  PubMed  Google Scholar 

  32. Gornik O, Lauc G. Glycosylation of serum proteins in inflammatory diseases. Disease markers. 2008;25(4-5):267–78.

    Article  CAS  PubMed  Google Scholar 

  33. Gornik O, Royle L, Harvey DJ, Radcliffe CM, Saldova R, Dwek RA, et al. Changes of serum glycans during sepsis and acute pancreatitis. Glycobiology. 2007;17(12):1321–32. https://doi.org/10.1093/glycob/cwm106.

    Article  CAS  PubMed  Google Scholar 

  34. Keser T, Gornik I, Vuckovic F, Selak N, Pavic T, Lukic E, et al. Increased plasma N-glycome complexity is associated with higher risk of type 2 diabetes. Diabetologia. 2017;60(12):2352–60. https://doi.org/10.1007/s00125-017-4426-9.

    Article  CAS  PubMed  Google Scholar 

  35. Knezevic A, Gornik O, Polasek O, Pucic M, Redzic I, Novokmet M, et al. Effects of aging, body mass index, plasma lipid profiles, and smoking on human plasma N-glycans. Glycobiology. 2010;20(8):959–69. https://doi.org/10.1093/glycob/cwq051.

    Article  CAS  PubMed  Google Scholar 

  36. Stanta JL, Saldova R, Struwe WB, Byrne JC, Leweke FM, Rothermund M, et al. Identification of N-glycosylation changes in the CSF and serum in patients with schizophrenia. Journal of proteome research. 2010;9(9):4476–89. https://doi.org/10.1021/pr1002356.

    Article  CAS  PubMed  Google Scholar 

  37. Alonzi DS, Su YH, Butters TD. Urinary glycan markers for disease. Biochem Soc Trans. 2011;39(1):393–8. https://doi.org/10.1042/BST0390393.

    Article  CAS  PubMed  Google Scholar 

  38. Gizaw ST, Ohashi T, Tanaka M, Hinou H, Nishimura S. Glycoblotting method allows for rapid and efficient glycome profiling of human Alzheimer’s disease brain, serum and cerebrospinal fluid towards potential biomarker discovery. Biochimica et biophysica acta. 2016;1860(8):1716–27. https://doi.org/10.1016/j.bbagen.2016.03.009.

    Article  CAS  PubMed  Google Scholar 

  39. Pucic M, Knezevic A, Vidic J, Adamczyk B, Novokmet M, Polasek O, et al. High throughput isolation and glycosylation analysis of IgG-variability and heritability of the IgG glycome in three isolated human populations. Mol Cell Proteomics. 2011;10(10):M111 010090. https://doi.org/10.1074/mcp.M111.010090.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Goede V, Fischer K, Busch R, Engelke A, Eichhorst B, Wendtner CM, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. The New England journal of medicine. 2014;370(12):1101–10. https://doi.org/10.1056/NEJMoa1313984.

    Article  CAS  PubMed  Google Scholar 

  41. Junttila TT, Parsons K, Olsson C, Lu Y, Xin Y, Theriault J, et al. Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer. Cancer research. 2010;70(11):4481–9. https://doi.org/10.1158/0008-5472.CAN-09-3704.

    Article  CAS  PubMed  Google Scholar 

  42. Sondermann P, Szymkowski DE. Harnessing Fc receptor biology in the design of therapeutic antibodies. Current opinion in immunology. 2016;40:78–87. https://doi.org/10.1016/j.coi.2016.03.005.

    Article  CAS  PubMed  Google Scholar 

  43. Dunn-Walters D, Boursier L, Spencer J. Effect of somatic hypermutation on potential N-glycosylation sites in human immunoglobulin heavy chain variable regions. Molecular immunology. 2000;37(3-4):107–13.

    Article  CAS  PubMed  Google Scholar 

  44. Anumula KR. Quantitative glycan profiling of normal human plasma derived immunoglobulin and its fragments Fab and Fc. J Immunol Methods. 2012;382(1-2):167–76. https://doi.org/10.1016/j.jim.2012.05.022.

    Article  CAS  PubMed  Google Scholar 

  45. Arnold JN, Wormald MR, Sim RB, Rudd PM, Dwek RA. The impact of glycosylation on the biological function and structure of human immunoglobulins. Annual review of immunology. 2007;25:21–50. https://doi.org/10.1146/annurev.immunol.25.022106.141702.

    Article  CAS  PubMed  Google Scholar 

  46. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69(Suppl 1):S4–9. https://doi.org/10.1093/gerona/glu057.

    Article  PubMed  Google Scholar 

  47. Nasi M, De Biasi S, Gibellini L, Bianchini E, Pecorini S, Bacca V, et al. Ageing and inflammation in patients with HIV infection. Clin Exp Immunol. 2017;187(1):44–52. https://doi.org/10.1111/cei.12814.

    Article  CAS  PubMed  Google Scholar 

  48. Negredo E, Back D, Blanco JR, Blanco J, Erlandson KM, Garolera M, et al. Aging in HIV-infected subjects: a new scenario and a new view. BioMed research international. 2017;2017:5897298. https://doi.org/10.1155/2017/5897298.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Minciullo PL, Catalano A, Mandraffino G, Casciaro M, Crucitti A, Maltese G, et al. Inflammaging and anti-inflammaging: the role of cytokines in extreme longevity. Arch Immunol Ther Exp (Warsz). 2016;64(2):111–26. https://doi.org/10.1007/s00005-015-0377-3.

    Article  CAS  PubMed  Google Scholar 

  50. Deeks SG. HIV infection, inflammation, immunosenescence, and aging. Annu Rev Med. 2011;62:141–55. https://doi.org/10.1146/annurev-med-042909-093756.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, Lane HC, et al. Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med. 2008;5(10):e203. https://doi.org/10.1371/journal.pmed.0050203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. The Journal of infectious diseases. 2011;203(6):780–90. https://doi.org/10.1093/infdis/jiq118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Musselwhite LW, Sheikh V, Norton TD, Rupert A, Porter BO, Penzak SR, et al. Markers of endothelial dysfunction, coagulation and tissue fibrosis independently predict venous thromboembolism in HIV. Aids. 2011;25(6):787–95. https://doi.org/10.1097/QAD.0b013e3283453fcb.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Brown JB, Conner C, Nichols GA. Secondary failure of metformin monotherapy in clinical practice. Diabetes care. 2010;33(3):501–6. https://doi.org/10.2337/dc09-1749.

    Article  CAS  PubMed  Google Scholar 

  55. Burdo TH, Orzechowski K, Knight HL, Miller AD, Williams K. Dorsal root ganglia damage in SIV-infected rhesus macaques: an animal model of HIV-induced sensory neuropathy. The American journal of pathology. 2012;180(4):1362–9. https://doi.org/10.1016/j.ajpath.2011.12.016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Erlandson KM, Allshouse AA, Jankowski CM, Lee EJ, Rufner KM, Palmer BE, et al. Association of functional impairment with inflammation and immune activation in HIV type 1-infected adults receiving effective antiretroviral therapy. The Journal of infectious diseases. 2013;208(2):249–59. https://doi.org/10.1093/infdis/jit147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Naeger DM, Martin JN, Sinclair E, Hunt PW, Bangsberg DR, Hecht F, et al. Cytomegalovirus-specific T cells persist at very high levels during long-term antiretroviral treatment of HIV disease. PloS one. 2010;5(1):e8886. https://doi.org/10.1371/journal.pone.0008886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Schacker TW, Nguyen PL, Beilman GJ, Wolinsky S, Larson M, Reilly C, et al. Collagen deposition in HIV-1 infected lymphatic tissues and T cell homeostasis. The Journal of clinical investigation. 2002;110(8):1133–9. https://doi.org/10.1172/JCI16413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Weber R, Ruppik M, Rickenbach M, Spoerri A, Furrer H, Battegay M, et al. Decreasing mortality and changing patterns of causes of death in the Swiss HIV cohort study. HIV Med. 2013;14(4):195–207. https://doi.org/10.1111/j.1468-1293.2012.01051.x.

    Article  CAS  PubMed  Google Scholar 

  60. Borges AH, O'Connor JL, Phillips AN, Ronsholt FF, Pett S, Vjecha MJ, et al. Factors associated with plasma IL-6 levels during HIV infection. The Journal of infectious diseases. 2015;212(4):585–95. https://doi.org/10.1093/infdis/jiv123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Freiberg MS, Bebu I, Tracy R, So-Armah K, Okulicz J, Ganesan A, et al. D-Dimer levels before HIV seroconversion remain elevated even after viral suppression and are associated with an increased risk of non-AIDS events. PloS one. 2016;11(4):e0152588. https://doi.org/10.1371/journal.pone.0152588.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kamat A, Misra V, Cassol E, Ancuta P, Yan Z, Li C, et al. A plasma biomarker signature of immune activation in HIV patients on antiretroviral therapy. PloS one. 2012;7(2):e30881. https://doi.org/10.1371/journal.pone.0030881.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Nixon DE, Landay AL. Biomarkers of immune dysfunction in HIV. Curr Opin HIV AIDS. 2010;5(6):498–503. https://doi.org/10.1097/COH.0b013e32833ed6f4.

    Article  PubMed  PubMed Central  Google Scholar 

  64. So-Armah KA, Tate JP, Chang CH, Butt AA, Gerschenson M, Gibert CL, et al. Do biomarkers of inflammation, monocyte activation, and altered coagulation explain excess mortality between HIV infected and uninfected people? Journal of acquired immune deficiency syndromes. 2016;72(2):206–13. https://doi.org/10.1097/QAI.0000000000000954.

    Article  CAS  PubMed  Google Scholar 

  65. Anthony RM, Nimmerjahn F, Ashline DJ, Reinhold VN, Paulson JC, Ravetch JV. Recapitulation of IVIG anti-inflammatory activity with a recombinant IgG Fc. Science. 2008;320(5874):373–6. https://doi.org/10.1126/science.1154315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kaneko Y, Nimmerjahn F, Ravetch JV. Anti-inflammatory activity of immunoglobulin G resulting from Fc sialylation. Science. 2006;313(5787):670–3. https://doi.org/10.1126/science.1129594.

    Article  CAS  PubMed  Google Scholar 

  67. Washburn N, Schwab I, Ortiz D, Bhatnagar N, Lansing JC, Medeiros A, et al. Controlled tetra-Fc sialylation of IVIg results in a drug candidate with consistent enhanced anti-inflammatory activity. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(11):E1297–306. https://doi.org/10.1073/pnas.1422481112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Bohm S, Schwab I, Lux A, Nimmerjahn F. The role of sialic acid as a modulator of the anti-inflammatory activity of IgG. Semin Immunopathol. 2012;34(3):443–53. https://doi.org/10.1007/s00281-012-0308-x.

    Article  CAS  PubMed  Google Scholar 

  69. Sondermann P, Pincetic A, Maamary J, Lammens K, Ravetch JV. General mechanism for modulating immunoglobulin effector function. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(24):9868–72. https://doi.org/10.1073/pnas.1307864110.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Ahmed AA, Giddens J, Pincetic A, Lomino JV, Ravetch JV, Wang LX, et al. Structural characterization of anti-inflammatory immunoglobulin G Fc proteins. J Mol Biol. 2014;426(18):3166–79. https://doi.org/10.1016/j.jmb.2014.07.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Yu X, Vasiljevic S, Mitchell DA, Crispin M, Scanlan CN. Dissecting the molecular mechanism of IVIg therapy: the interaction between serum IgG and DC-SIGN is independent of antibody glycoform or Fc domain. J Mol Biol. 2013;425(8):1253–8. https://doi.org/10.1016/j.jmb.2013.02.006.

    Article  CAS  PubMed  Google Scholar 

  72. Varki A, Gagneux P. Multifarious roles of sialic acids in immunity. Annals of the New York Academy of Sciences. 2012;1253:16–36. https://doi.org/10.1111/j.1749-6632.2012.06517.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Kim YH, Min KH, Wang Z, Kim J, Jacobson O, Huang P, et al. Development of sialic acid-coated nanoparticles for targeting cancer and efficient evasion of the immune system. Theranostics. 2017;7(4):962–73. https://doi.org/10.7150/thno.19061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. • Spence S, Greene MK, Fay F, Hams E, Saunders SP, Hamid U, et al. Targeting Siglecs with a sialic acid-decorated nanoparticle abrogates inflammation. Sci Transl Med. 2015;7(303):303ra140. https://doi.org/10.1126/scitranslmed.aab3459 This paper highlighted the therapeutic potential of enhancing sialic-acid-siglec anti-inflammatory binding in reducing systemic inflammation.

    Article  CAS  PubMed  Google Scholar 

  75. Anthony RM, Ravetch JV. A novel role for the IgG Fc glycan: the anti-inflammatory activity of sialylated IgG Fcs. J Clin Immunol. 2010;30(Suppl 1):S9–14. https://doi.org/10.1007/s10875-010-9405-6.

    Article  CAS  PubMed  Google Scholar 

  76. Anthony RM, Wermeling F, Karlsson MC, Ravetch JV. Identification of a receptor required for the anti-inflammatory activity of IVIG. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(50):19571–8. https://doi.org/10.1073/pnas.0810163105.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Kristic J, Vuckovic F, Menni C, Klaric L, Keser T, Beceheli I, et al. Glycans are a novel biomarker of chronological and biological ages. J Gerontol A Biol Sci Med Sci. 2014;69(7):779–89. https://doi.org/10.1093/gerona/glt190.

    Article  CAS  PubMed  Google Scholar 

  78. Yu X, Wang Y, Kristic J, Dong J, Chu X, Ge S, et al. Profiling IgG N-glycans as potential biomarker of chronological and biological ages: a community-based study in a Han Chinese population. Medicine (Baltimore). 2016;95(28):e4112. https://doi.org/10.1097/MD.0000000000004112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Trbojevic Akmacic I, Ventham NT, Theodoratou E, Vuckovic F, Kennedy NA, Kristic J, et al. Inflammatory bowel disease associates with proinflammatory potential of the immunoglobulin G glycome. Inflamm Bowel Dis. 2015;21(6):1237–47. https://doi.org/10.1097/MIB.0000000000000372.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Vučković F, Krištić J, Gudelj I, Artacho MT, Keser T, Pezer M, et al. Systemic lupus erythematosus associates with the decreased immunosuppressive potential of the IgG glycome. Arthritis & Rheumatology. 2015;67(11):2978–89. https://doi.org/10.1002/art.39273.

    Article  CAS  Google Scholar 

  81. Vuckovic F, Theodoratou E, Thaci K, Timofeeva M, Vojta A, Stambuk J, et al. IgG glycome in colorectal cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2016;22(12):3078–86. https://doi.org/10.1158/1078-0432.CCR-15-1867.

    Article  CAS  Google Scholar 

  82. Lemmers RFH, Vilaj M, Urda D, Agakov F, Simurina M, Klaric L, et al. IgG glycan patterns are associated with type 2 diabetes in independent European populations. Biochimica et biophysica acta. 2017;1861(9):2240–9. https://doi.org/10.1016/j.bbagen.2017.06.020.

    Article  CAS  PubMed  Google Scholar 

  83. An HJ, Lebrilla CB. A glycomics approach to the discovery of potential cancer biomarkers. Methods in molecular biology. 2010;600:199–213. https://doi.org/10.1007/978-1-60761-454-8_14.

    Article  CAS  PubMed  Google Scholar 

  84. Willerson JT, Ridker PM. Inflammation as a cardiovascular risk factor. Circulation. 2004;109(21 Suppl 1):II2–10. https://doi.org/10.1161/01.CIR.0000129535.04194.38.

    Article  PubMed  Google Scholar 

  85. Akinkuolie AO, Buring JE, Ridker PM, Mora S. A novel protein glycan biomarker and future cardiovascular disease events. Journal of the American Heart Association. 2014;3(5):e001221. https://doi.org/10.1161/JAHA.114.001221.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ercan A, Cui J, Chatterton DE, Deane KD, Hazen MM, Brintnell W, et al. Aberrant IgG galactosylation precedes disease onset, correlates with disease activity, and is prevalent in autoantibodies in rheumatoid arthritis. Arthritis and rheumatism. 2010;62(8):2239–48. https://doi.org/10.1002/art.27533.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Rombouts Y, Ewing E, van de Stadt LA, Selman MH, Trouw LA, Deelder AM, et al. Anti-citrullinated protein antibodies acquire a pro-inflammatory Fc glycosylation phenotype prior to the onset of rheumatoid arthritis. Annals of the rheumatic diseases. 2013. https://doi.org/10.1136/annrheumdis-2013-203565.

  88. • Vadrevu SK, Trbojevic-Akmacic I, Kossenkov AV, Colomb F, Giron LB, Anzurez A, et al. Frontline science: plasma and immunoglobulin G galactosylation associate with HIV persistence during antiretroviral therapy. Journal of leukocyte biology. 2018. https://doi.org/10.1002/JLB.3HI1217-500R This paper reported that levels of certain plasma and antibody glycomic alterations, in particular, non-fucosylated galactosylated glycomic traits, negatively correlate with levels of cell-associated HIV DNA and RNA in ART-suppressed individuals. These glycomic traits have been previously associated with higher plasma-mediated innate immune functions (ADCC/ADCP).

  89. Moore JS, Wu X, Kulhavy R, Tomana M, Novak J, Moldoveanu Z, et al. Increased levels of galactose-deficient IgG in sera of HIV-1-infected individuals. Aids. 2005;19(4):381–9.

    Article  CAS  PubMed  Google Scholar 

  90. Karsten CM, Pandey MK, Figge J, Kilchenstein R, Taylor PR, Rosas M, et al. Anti-inflammatory activity of IgG1 mediated by Fc galactosylation and association of FcgammaRIIB and dectin-1. Nature medicine. 2012;18(9):1401–6. https://doi.org/10.1038/nm.2862.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Banda NK, Wood AK, Takahashi K, Levitt B, Rudd PM, Royle L, et al. Initiation of the alternative pathway of murine complement by immune complexes is dependent on N-glycans in IgG antibodies. Arthritis and rheumatism. 2008;58(10):3081–9. https://doi.org/10.1002/art.23865.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Malhotra R, Wormald MR, Rudd PM, Fischer PB, Dwek RA, Sim RB. Glycosylation changes of IgG associated with rheumatoid arthritis can activate complement via the mannose-binding protein. Nature medicine. 1995;1(3):237–43.

    Article  CAS  PubMed  Google Scholar 

  93. Parekh RB, Dwek RA, Sutton BJ, Fernandes DL, Leung A, Stanworth D, et al. Association of rheumatoid arthritis and primary osteoarthritis with changes in the glycosylation pattern of total serum IgG. Nature. 1985;316(6027):452–7.

    Article  CAS  PubMed  Google Scholar 

  94. Tomana M, Schrohenloher RE, Koopman WJ, Alarcon GS, Paul WA. Abnormal glycosylation of serum IgG from patients with chronic inflammatory diseases. Arthritis and rheumatism. 1988;31(3):333–8.

    Article  CAS  PubMed  Google Scholar 

  95. Oswald DM, Sim ES, Baker C, Farhan O, Debanne SM, Morris NJ, et al. Plasma glycomics predict cardiovascular disease in patients with ART-controlled HIV infections. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2018:fj201800923R. https://doi.org/10.1096/fj.201800923R.

  96. Alpert MD, Harvey JD, Lauer WA, Reeves RK, Piatak M Jr, Carville A, et al. ADCC develops over time during persistent infection with live-attenuated SIV and is associated with complete protection against SIV(mac)251 challenge. PLoS pathogens. 2012;8(8):e1002890. https://doi.org/10.1371/journal.ppat.1002890.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Baum LL, Cassutt KJ, Knigge K, Khattri R, Margolick J, Rinaldo C, et al. HIV-1 gp120-specific antibody-dependent cell-mediated cytotoxicity correlates with rate of disease progression. Journal of immunology. 1996;157(5):2168–73.

    CAS  Google Scholar 

  98. Bruel T, Guivel-Benhassine F, Amraoui S, Malbec M, Richard L, Bourdic K, et al. Elimination of HIV-1-infected cells by broadly neutralizing antibodies. Nature communications. 2016;7:10844. https://doi.org/10.1038/ncomms10844.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Chung AW, Navis M, Isitman G, Wren L, Silvers J, Amin J, et al. Activation of NK cells by ADCC antibodies and HIV disease progression. Journal of acquired immune deficiency syndromes. 2011;58(2):127–31. https://doi.org/10.1097/QAI.0b013e31822c62b9.

    Article  CAS  PubMed  Google Scholar 

  100. Isitman G, Lisovsky I, Tremblay-McLean A, Kovacs C, Harris M, Routy JP, et al. Antibody-dependent cellular cytotoxicity activity of effector cells from HIV-infected elite and viral controllers. AIDS research and human retroviruses. 2016;32(10-11):1079–88. https://doi.org/10.1089/AID.2016.0157.

    Article  CAS  PubMed  Google Scholar 

  101. Lee WS, Kent SJ. Anti-HIV-1 antibody-dependent cellular cytotoxicity: is there more to antibodies than neutralization? Curr Opin HIV AIDS. 2018;13(2):160–6. https://doi.org/10.1097/COH.0000000000000439.

    Article  CAS  PubMed  Google Scholar 

  102. Madhavi V, Ana-Sosa-Batiz FE, Jegaskanda S, Center RJ, Winnall WR, Parsons MS, et al. Antibody-dependent effector functions against HIV decline in subjects receiving antiretroviral therapy. The Journal of infectious diseases. 2015;211(4):529–38. https://doi.org/10.1093/infdis/jiu486.

    Article  CAS  PubMed  Google Scholar 

  103. Halper-Stromberg A, Nussenzweig MC. Towards HIV-1 remission: potential roles for broadly neutralizing antibodies. J Clin Invest. 2016;126(2):415–23. https://doi.org/10.1172/JCI80561.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Bhiman JN, Lynch RM. Broadly neutralizing antibodies as treatment: effects on virus and immune system. Curr HIV/AIDS Rep. 2017;14(2):54–62. https://doi.org/10.1007/s11904-017-0352-1.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Masuda K, Kubota T, Kaneko E, Iida S, Wakitani M, Kobayashi-Natsume Y, et al. Enhanced binding affinity for FcgammaRIIIa of fucose-negative antibody is sufficient to induce maximal antibody-dependent cellular cytotoxicity. Molecular immunology. 2007;44(12):3122–31. https://doi.org/10.1016/j.molimm.2007.02.005.

    Article  CAS  PubMed  Google Scholar 

  106. Thomann M, Reckermann K, Reusch D, Prasser J, Tejada ML. Fc-galactosylation modulates antibody-dependent cellular cytotoxicity of therapeutic antibodies. Molecular immunology. 2016;73:69–75. https://doi.org/10.1016/j.molimm.2016.03.002.

    Article  CAS  PubMed  Google Scholar 

  107. Heyl KA, Karsten CM, Slevogt H. Galectin-3 binds highly galactosylated IgG1 and is crucial for the IgG1 complex mediated inhibition of C5aReceptor induced immune responses. Biochemical and biophysical research communications. 2016;479(1):86–90. https://doi.org/10.1016/j.bbrc.2016.09.038.

    Article  CAS  PubMed  Google Scholar 

  108. Chung AW, Crispin M, Pritchard L, Robinson H, Gorny MK, Yu X, et al. Identification of antibody glycosylation structures that predict monoclonal antibody Fc-effector function. Aids. 2014;28(17):2523–30. https://doi.org/10.1097/QAD.0000000000000444.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Shinkawa T, Nakamura K, Yamane N, Shoji-Hosaka E, Kanda Y, Sakurada M, et al. The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. The Journal of biological chemistry. 2003;278(5):3466–73. https://doi.org/10.1074/jbc.M210665200.

    Article  CAS  PubMed  Google Scholar 

  110. Ackerman ME, Crispin M, Yu X, Baruah K, Boesch AW, Harvey DJ, et al. Natural variation in Fc glycosylation of HIV-specific antibodies impacts antiviral activity. J Clin Invest. 2013;123(5):2183–92. https://doi.org/10.1172/JCI65708.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Lorenzo-Redondo R, Fryer HR, Bedford T, Kim EY, Archer J, Pond SLK, et al. Persistent HIV-1 replication maintains the tissue reservoir during therapy. Nature. 2016;530(7588):51–6. https://doi.org/10.1038/nature16933.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Rosenbloom DIS, Hill AL, Laskey SB, Siliciano RF. Re-evaluating evolution in the HIV reservoir. Nature. 2017;551(7681):E6–9. https://doi.org/10.1038/nature24634.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Panico M, Bouche L, Binet D, O'Connor MJ, Rahman D, Pang PC, et al. Mapping the complete glycoproteome of virion-derived HIV-1 gp120 provides insights into broadly neutralizing antibody binding. Scientific reports. 2016;6:32956. https://doi.org/10.1038/srep32956.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Puryear WB, Yu X, Ramirez NP, Reinhard BM, Gummuluru S. HIV-1 incorporation of host-cell-derived glycosphingolipid GM3 allows for capture by mature dendritic cells. Proc Natl Acad Sci U S A. 2012;109(19):7475–80. https://doi.org/10.1073/pnas.1201104109.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Geijtenbeek TB, Kwon DS, Torensma R, van Vliet SJ, van Duijnhoven GC, Middel J, et al. DC-SIGN, a dendritic cell-specific HIV-1-binding protein that enhances trans-infection of T cells. Cell. 2000;100(5):587–97.

    Article  CAS  PubMed  Google Scholar 

  116. Rappocciolo G, Piazza P, Fuller CL, Reinhart TA, Watkins SC, Rowe DT, et al. DC-SIGN on B lymphocytes is required for transmission of HIV-1 to T lymphocytes. PLoS Pathog. 2006;2(7):e70. https://doi.org/10.1371/journal.ppat.0020070.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Turville SG, Cameron PU, Handley A, Lin G, Pohlmann S, Doms RW, et al. Diversity of receptors binding HIV on dendritic cell subsets. Nat Immunol. 2002;3(10):975–83. https://doi.org/10.1038/ni841.

    Article  CAS  PubMed  Google Scholar 

  118. de Witte L, Nabatov A, Pion M, Fluitsma D, de Jong MA, de Gruijl T, et al. Langerin is a natural barrier to HIV-1 transmission by Langerhans cells. Nat Med. 2007;13(3):367–71. https://doi.org/10.1038/nm1541.

    Article  CAS  PubMed  Google Scholar 

  119. Mayr L, Su B, Moog C. Langerhans cells: the ‘yin and yang’ of HIV restriction and transmission. Trends Microbiol. 2017;25(3):170–2. https://doi.org/10.1016/j.tim.2017.01.009.

    Article  CAS  PubMed  Google Scholar 

  120. Pena-Cruz V, Agosto LM, Akiyama H, Olson A, Moreau Y, Larrieux JR, et al. HIV-1 replicates and persists in vaginal epithelial dendritic cells. J Clin Invest. 2018;128(8):3439–44. https://doi.org/10.1172/JCI98943.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Saifuddin M, Hart ML, Gewurz H, Zhang Y, Spear GT. Interaction of mannose-binding lectin with primary isolates of human immunodeficiency virus type 1. J Gen Virol. 2000;81(Pt 4):949–55. https://doi.org/10.1099/0022-1317-81-4-949.

    Article  CAS  PubMed  Google Scholar 

  122. Ying H, Ji X, Hart ML, Gupta K, Saifuddin M, Zariffard MR, et al. Interaction of mannose-binding lectin with HIV type 1 is sufficient for virus opsonization but not neutralization. AIDS Res Hum Retroviruses. 2004;20(3):327–35. https://doi.org/10.1089/088922204322996563.

    Article  CAS  PubMed  Google Scholar 

  123. Dzwonek A, Novelli V, Bajaj-Elliott M, Turner M, Clapson M, Klein N. Mannose-binding lectin in susceptibility and progression of HIV-1 infection in children. Antivir Ther. 2006;11(4):499–505.

    CAS  PubMed  Google Scholar 

  124. Zinyama-Gutsire RB, Chasela C, Madsen HO, Rusakaniko S, Kallestrup P, Christiansen M, et al. Role of mannose-binding lectin deficiency in HIV-1 and schistosoma infections in a rural adult population in Zimbabwe. PLoS One. 2015;10(4):e0122659. https://doi.org/10.1371/journal.pone.0122659.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Vallinoto AC, Freitas FB, Guirelli I, Machado LF, Azevedo VN, Cayres-Vallinoto I, et al. Characterization of mannose-binding lectin plasma levels and genetic polymorphisms in HIV-1-infected individuals. Rev Soc Bras Med Tro. 2011;44(1):1–3.

    Article  Google Scholar 

  126. Mehta-D'souza P, Klopocki AG, Oganesyan V, Terzyan S, Mather T, Li Z, et al. Glycan bound to the selectin low affinity state engages Glu-88 to stabilize the high affinity state under force. J Biol Chem. 2017;292(6):2510–8. https://doi.org/10.1074/jbc.M116.767186.

    Article  CAS  PubMed  Google Scholar 

  127. Lambert AA, Gilbert C, Richard M, Beaulieu AD, Tremblay MJ. The C-type lectin surface receptor DCIR acts as a new attachment factor for HIV-1 in dendritic cells and contributes to trans- and cis-infection pathways. Blood. 2008;112(4):1299–307. https://doi.org/10.1182/blood-2008-01-136473.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Pohlmann S, Soilleux EJ, Baribaud F, Leslie GJ, Morris LS, Trowsdale J, et al. DC-SIGNR, a DC-SIGN homologue expressed in endothelial cells, binds to human and simian immunodeficiency viruses and activates infection in trans. Proc Natl Acad Sci U S A. 2001;98(5):2670–5. https://doi.org/10.1073/pnas.051631398.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Madsen J, Gaiha GD, Palaniyar N, Dong T, Mitchell DA, Clark HW. Surfactant protein D modulates HIV infection of both T-cells and dendritic cells. PLoS One. 2013;8(3):e59047. https://doi.org/10.1371/journal.pone.0059047.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Lambert AA, Imbeault M, Gilbert C, Tremblay MJ. HIV-1 induces DCIR expression in CD4+ T cells. PLoS Pathog. 2010;6(11):e1001188. https://doi.org/10.1371/journal.ppat.1001188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Pandit H, Gopal S, Sonawani A, Yadav AK, Qaseem AS, Warke H, et al. Surfactant protein D inhibits HIV-1 infection of target cells via interference with gp120-CD4 interaction and modulates pro-inflammatory cytokine production. PLoS One. 2014;9(7):e102395. https://doi.org/10.1371/journal.pone.0102395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Izquierdo-Useros N, Lorizate M, Puertas MC, Rodriguez-Plata MT, Zangger N, Erikson E, et al. Siglec-1 is a novel dendritic cell receptor that mediates HIV-1 trans-infection through recognition of viral membrane gangliosides. PLoS Biol. 2012;10(12):e1001448. https://doi.org/10.1371/journal.pbio.1001448.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Zou Z, Chastain A, Moir S, Ford J, Trandem K, Martinelli E, et al. Siglecs facilitate HIV-1 infection of macrophages through adhesion with viral sialic acids. PLoS One. 2011;6(9):e24559. https://doi.org/10.1371/journal.pone.0024559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Martinez-Picado J, McLaren PJ, Erkizia I, Martin MP, Benet S, Rotger M, et al. Identification of Siglec-1 null individuals infected with HIV-1. Nat Commun. 2016;7:12412. https://doi.org/10.1038/ncomms12412.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Varchetta S, Lusso P, Hudspeth K, Mikulak J, Mele D, Paolucci S, et al. Sialic acid-binding Ig-like lectin-7 interacts with HIV-1 gp120 and facilitates infection of CD4pos T cells and macrophages. Retrovirology. 2013;10:154. https://doi.org/10.1186/1742-4690-10-154.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Ouellet M, Mercier S, Pelletier I, Bounou S, Roy J, Hirabayashi J, et al. Galectin-1 acts as a soluble host factor that promotes HIV-1 infectivity through stabilization of virus attachment to host cells. J Immunol. 2005;174(7):4120–6.

    Article  CAS  PubMed  Google Scholar 

  137. Mendez-Huergo SP, Blidner AG, Rabinovich GA. Galectins: emerging regulatory checkpoints linking tumor immunity and angiogenesis. Curr Opin Immunol. 2017;45:8–15. https://doi.org/10.1016/j.coi.2016.12.003.

    Article  CAS  PubMed  Google Scholar 

  138. Zhuo Y, Bellis SL. Emerging role of alpha2,6-sialic acid as a negative regulator of galectin binding and function. The Journal of biological chemistry. 2011;286(8):5935–41. https://doi.org/10.1074/jbc.R110.191429.

    Article  CAS  PubMed  Google Scholar 

  139. Barondes SH, Cooper DN, Gitt MA, Leffler H. Galectins. Structure and function of a large family of animal lectins. J Biol Chem. 1994;269(33):20807–10.

    CAS  PubMed  Google Scholar 

  140. Gordon-Alonso M, Hirsch T, Wildmann C, van der Bruggen P. Galectin-3 captures interferon-gamma in the tumor matrix reducing chemokine gradient production and T-cell tumor infiltration. Nature communications. 2017;8(1):793. https://doi.org/10.1038/s41467-017-00925-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. •• Smith LK, Boukhaled GM, Condotta SA, Mazouz S, Guthmiller JJ, Vijay R, et al. Interleukin-10 directly inhibits CD8(+) T cell function by enhancing N-glycan branching to decrease antigen sensitivity. Immunity. 2018;48(2):299–312 e5. https://doi.org/10.1016/j.immuni.2018.01.006 This paper showed that, during chronic viral infection, IL-10 promotes the production of highly branched N -glycans on the surface of CD8 T cells. These N -glycans bind to galectins, and this binding significantly impairs T cell function and promotes viral persistence.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Anthony RM, Kobayashi T, Wermeling F, Ravetch JV. Intravenous gammaglobulin suppresses inflammation through a novel T(H)2 pathway. Nature. 2011;475(7354):110–3. https://doi.org/10.1038/nature10134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Byrne B, Donohoe GG, O'Kennedy R. Sialic acids: carbohydrate moieties that influence the biological and physical properties of biopharmaceutical proteins and living cells. Drug discovery today. 2007;12(7-8):319–26. https://doi.org/10.1016/j.drudis.2007.02.010.

    Article  CAS  PubMed  Google Scholar 

  144. Jandus C, Simon HU, von Gunten S. Targeting siglecs--a novel pharmacological strategy for immuno- and glycotherapy. Biochemical pharmacology. 2011;82(4):323–32. https://doi.org/10.1016/j.bcp.2011.05.018.

    Article  CAS  PubMed  Google Scholar 

  145. Pillai S, Netravali IA, Cariappa A, Mattoo H. Siglecs and immune regulation. Annual review of immunology. 2012;30:357–92. https://doi.org/10.1146/annurev-immunol-020711-075018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Rabinovich GA, Rubinstein N, Toscano MA. Role of galectins in inflammatory and immunomodulatory processes. Biochimica et biophysica acta. 2002;1572(2-3):274–84.

    Article  CAS  PubMed  Google Scholar 

  147. Toscano MA, Bianco GA, Ilarregui JM, Croci DO, Correale J, Hernandez JD, et al. Differential glycosylation of TH1, TH2 and TH-17 effector cells selectively regulates susceptibility to cell death. Nature immunology. 2007;8(8):825–34. https://doi.org/10.1038/ni1482.

    Article  CAS  PubMed  Google Scholar 

  148. Farzi B, Young D, Scrimgeour J, Cetinkaya C. Mechanical properties of P-selectin PSGL-1 bonds. Colloids and surfaces B, Biointerfaces. 2018;173:529–38. https://doi.org/10.1016/j.colsurfb.2018.10.017.

    Article  CAS  PubMed  Google Scholar 

  149. Ley K. The role of selectins in inflammation and disease. Trends Mol Med. 2003;9(6):263–8.

    Article  CAS  PubMed  Google Scholar 

  150. Ley K, Kansas GS. Selectins in T-cell recruitment to non-lymphoid tissues and sites of inflammation. Nature reviews Immunology. 2004;4(5):325–35. https://doi.org/10.1038/nri1351.

    Article  CAS  PubMed  Google Scholar 

  151. Catalfamo M, Wilhelm C, Tcheung L, Proschan M, Friesen T, Park JH, et al. CD4 and CD8 T cell immune activation during chronic HIV infection: roles of homeostasis, HIV, type I IFN, and IL-7. Journal of immunology. 2011;186(4):2106–16. https://doi.org/10.4049/jimmunol.1002000.

    Article  CAS  Google Scholar 

  152. Chavez L, Calvanese V, Verdin E. HIV latency is established directly and early in both resting and activated primary CD4 T cells. PLoS pathogens. 2015;11(6):e1004955. https://doi.org/10.1371/journal.ppat.1004955.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Doitsh G, Galloway NL, Geng X, Yang Z, Monroe KM, Zepeda O, et al. Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature. 2014;505(7484):509–14. https://doi.org/10.1038/nature12940.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Freeman ML, Mudd JC, Shive CL, Younes SA, Panigrahi S, Sieg SF, et al. CD8 T-cell expansion and inflammation linked to CMV coinfection in ART-treated HIV infection. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2016;62(3):392–6. https://doi.org/10.1093/cid/civ840.

    Article  CAS  Google Scholar 

  155. Khaitan A, Unutmaz D. Revisiting immune exhaustion during HIV infection. Curr HIV/AIDS Rep. 2011;8(1):4–11. https://doi.org/10.1007/s11904-010-0066-0.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Kwon KJ, Siliciano RF. HIV persistence: clonal expansion of cells in the latent reservoir. The Journal of clinical investigation. 2017;127(7):2536–8. https://doi.org/10.1172/JCI95329.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Murray AJ, Kwon KJ, Farber DL, Siliciano RF. The latent reservoir for HIV-1: how immunologic memory and clonal expansion contribute to HIV-1 persistence. Journal of immunology. 2016;197(2):407–17. https://doi.org/10.4049/jimmunol.1600343.

    Article  CAS  Google Scholar 

  158. Tandon R, Chew GM, Byron MM, Borrow P, Niki T, Hirashima M, et al. Galectin-9 is rapidly released during acute HIV-1 infection and remains sustained at high levels despite viral suppression even in elite controllers. AIDS Res Hum Retroviruses. 2014;30(7):654–64. https://doi.org/10.1089/AID.2014.0004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Lhuillier C, Barjon C, Niki T, Gelin A, Praz F, Morales O, et al. Impact of Exogenous galectin-9 on human T cells: contribution of the T cell receptor complex to antigen-independent activation but not to apoptosis induction. The Journal of biological chemistry. 2015;290(27):16797–811. https://doi.org/10.1074/jbc.M115.661272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Elahi S, Niki T, Hirashima M, Horton H. Galectin-9 binding to Tim-3 renders activated human CD4+ T cells less susceptible to HIV-1 infection. Blood. 2012;119(18):4192–204. https://doi.org/10.1182/blood-2011-11-389585.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Schaefer K, Webb NE, Pang M, Hernandez-Davies JE, Lee KP, Gonzalez P, et al. Galectin-9 binds to O-glycans on protein disulfide isomerase. Glycobiology. 2017;27(9):878–87. https://doi.org/10.1093/glycob/cwx065.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Wu C, Thalhamer T, Franca RF, Xiao S, Wang C, Hotta C, et al. Galectin-9-CD44 interaction enhances stability and function of adaptive regulatory T cells. Immunity. 2014;41(2):270–82. https://doi.org/10.1016/j.immuni.2014.06.011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Kouo T, Huang L, Pucsek AB, Cao M, Solt S, Armstrong T, et al. Galectin-3 shapes antitumor immune responses by suppressing CD8+ T cells via LAG-3 and inhibiting expansion of plasmacytoid dendritic cells. Cancer Immunol Res. 2015;3(4):412–23. https://doi.org/10.1158/2326-6066.CIR-14-0150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. • Li CW, Lim SO, Chung EM, Kim YS, Park AH, Yao J, et al. Eradication of triple-negative breast cancer cells by targeting glycosylated PD-L1. Cancer Cell. 2018;33(2):187–201 e10. https://doi.org/10.1016/j.ccell.2018.01.009 This paper showed that the glycosylation of PD-L1 plays a critical role in immunosuppression by enhancing interactions with PD-1, and that targeting the glycosylation of immune inhibitory receptors is an effective strategy to promote the function of the immune system.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Li CW, Lim SO, Xia W, Lee HH, Chan LC, Kuo CW, et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nature communications. 2016;7:12632. https://doi.org/10.1038/ncomms12632.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. • Okada M, Chikuma S, Kondo T, Hibino S, Machiyama H, Yokosuka T, et al. Blockage of Core fucosylation reduces cell-surface expression of PD-1 and promotes anti-tumor immune responses of T cells. Cell reports. 2017;20(5):1017–28. https://doi.org/10.1016/j.celrep.2017.07.027 This paper showed that loss of core fucosylation reduces cell-surface expression of PD-1 and enhances both T cell activation and T cell anti-tumor immune responses.

    Article  CAS  PubMed  Google Scholar 

  167. Alter G, Altfeld M. NK cells in HIV-1 infection: evidence for their role in the control of HIV-1 infection. J Intern Med. 2009;265(1):29–42. https://doi.org/10.1111/j.1365-2796.2008.02045.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Bostik P, Takahashi Y, Mayne AE, Ansari AA. Innate immune natural killer cells and their role in HIV and SIV infection. HIV Ther. 2010;4(4):483–504. https://doi.org/10.2217/HIV.10.28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Brunetta E, Fogli M, Varchetta S, Bozzo L, Hudspeth KL, Marcenaro E, et al. The decreased expression of Siglec-7 represents an early marker of dysfunctional natural killer-cell subsets associated with high levels of HIV-1 viremia. Blood. 2009;114(18):3822–30. https://doi.org/10.1182/blood-2009-06-226332.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Hudak JE, Canham SM, Bertozzi CR. Glycocalyx engineering reveals a siglec-based mechanism for NK cell immunoevasion. Nature chemical biology. 2014;10(1):69–75. https://doi.org/10.1038/nchembio.1388.

    Article  CAS  PubMed  Google Scholar 

  171. • Xiao H, Woods EC, Vukojicic P, Bertozzi CR. Precision glycocalyx editing as a strategy for cancer immunotherapy. Proc Natl Acad Sci U S A. 2016;113(37):10304–9. https://doi.org/10.1073/pnas.1608069113 This paper showed that anti-HER2-sialidase conjugate reduces the inhibitory interaction between sialic-acid and siglec, and activate anti-tumor immunity. These results highlight the promising value of glycocalyx editing in enhancing immunological responses.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Golden-Mason L, McMahan RH, Strong M, Reisdorph R, Mahaffey S, Palmer BE, et al. Galectin-9 functionally impairs natural killer cells in humans and mice. J Virol. 2013;87(9):4835–45. https://doi.org/10.1128/JVI.01085-12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Tsuboi S, Sutoh M, Hatakeyama S, Hiraoka N, Habuchi T, Horikawa Y, et al. A novel strategy for evasion of NK cell immunity by tumours expressing core2 O-glycans. The EMBO journal. 2011;30(15):3173–85. https://doi.org/10.1038/emboj.2011.215.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Wang W, Guo H, Geng J, Zheng X, Wei H, Sun R, et al. Tumor-released galectin-3, a soluble inhibitory ligand of human NKp30, plays an important role in tumor escape from NK cell attack. The Journal of biological chemistry. 2014;289(48):33311–9. https://doi.org/10.1074/jbc.M114.603464.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O'Shea MA, et al. Evidence for HIV-associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV-infected viremic individuals. J Exp Med. 2008;205(8):1797–805. https://doi.org/10.1084/jem.20072683.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Kardava L, Moir S, Wang W, Ho J, Buckner CM, Posada JG, et al. Attenuation of HIV-associated human B cell exhaustion by siRNA downregulation of inhibitory receptors. J Clin Invest. 2011;121(7):2614–24. https://doi.org/10.1172/JCI45685.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Bonzi J, Bornet O, Betzi S, Kasper BT, Mahal LK, Mancini SJ, et al. Pre-B cell receptor binding to galectin-1 modifies galectin-1/carbohydrate affinity to modulate specific galectin-1/glycan lattice interactions. Nat Commun. 2015;6:6194. https://doi.org/10.1038/ncomms7194.

    Article  CAS  PubMed  Google Scholar 

  178. Elantak L, Espeli M, Boned A, Bornet O, Bonzi J, Gauthier L, et al. Structural basis for galectin-1-dependent pre-B cell receptor (pre-BCR) activation. J Biol Chem. 2012;287(53):44703–13. https://doi.org/10.1074/jbc.M112.395152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Gauthier L, Rossi B, Roux F, Termine E, Schiff C. Galectin-1 is a stromal cell ligand of the pre-B cell receptor (BCR) implicated in synapse formation between pre-B and stromal cells and in pre-BCR triggering. Proc Natl Acad Sci U S A. 2002;99(20):13014–9. https://doi.org/10.1073/pnas.202323999.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Cao A, Alluqmani N, Buhari FHM, Wasim L, Smith LK, Quaile AT, et al. Galectin-9 binds IgM-BCR to regulate B cell signaling. Nature communications. 2018;9(1):3288. https://doi.org/10.1038/s41467-018-05771-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Giovannone N, Liang J, Antonopoulos A, Geddes Sweeney J, King SL, Pochebit SM, et al. Galectin-9 suppresses B cell receptor signaling and is regulated by I-branching of N-glycans. Nat Commun. 2018;9(1):3287. https://doi.org/10.1038/s41467-018-05770-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Macatangay BJ, Landay AL, Rinaldo CR. MDSC: a new player in HIV immunopathogenesis. AIDS. 2012;26(12):1567–9. https://doi.org/10.1097/QAD.0b013e328355e682.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Vollbrecht T, Stirner R, Tufman A, Roider J, Huber RM, Bogner JR, et al. Chronic progressive HIV-1 infection is associated with elevated levels of myeloid-derived suppressor cells. AIDS. 2012;26(12):F31–7. https://doi.org/10.1097/QAD.0b013e328354b43f.

    Article  CAS  PubMed  Google Scholar 

  184. Zhang Y, Zhang M, Li X, Tang Z, He L, Lv K. Expansion of CD11b(+)Ly-6C(+) myeloid-derived suppressor cells (MDSCs) driven by galectin-9 attenuates CVB3-induced myocarditis. Mol Immunol. 2017;83:62–71. https://doi.org/10.1016/j.molimm.2017.01.013.

    Article  CAS  PubMed  Google Scholar 

  185. Pantaleo G, Graziosi C, Demarest JF, Butini L, Montroni M, Fox CH, et al. HIV infection is active and progressive in lymphoid tissue during the clinically latent stage of disease. Nature. 1993;362(6418):355–8. https://doi.org/10.1038/362355a0.

    Article  CAS  PubMed  Google Scholar 

  186. Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beilman GJ, et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med. 2004;200(6):749–59. https://doi.org/10.1084/jem.20040874.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Niessen CM. Tight junctions/adherens junctions: basic structure and function. The Journal of investigative dermatology. 2007;127(11):2525–32. https://doi.org/10.1038/sj.jid.5700865.

    Article  CAS  PubMed  Google Scholar 

  188. Turner JR. Intestinal mucosal barrier function in health and disease. Nature reviews Immunology. 2009;9(11):799–809. https://doi.org/10.1038/nri2653.

    Article  CAS  PubMed  Google Scholar 

  189. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365–71. https://doi.org/10.1038/nm1511.

    Article  CAS  PubMed  Google Scholar 

  190. Chan W, Sviridov D, Dart AM. HIV, atherosclerosis and inflammation: implications for treatment. Journal of HIV therapy. 2009;14(3):61–8.

    PubMed  Google Scholar 

  191. Deeks SG. Immune dysfunction, inflammation, and accelerated aging in patients on antiretroviral therapy. Topics in HIV medicine : a publication of the International AIDS Society, USA. 2009;17(4):118–23.

    Google Scholar 

  192. Klatt NR, Chomont N, Douek DC, Deeks SG. Immune activation and HIV persistence: implications for curative approaches to HIV infection. Immunological reviews. 2013;254(1):326–42. https://doi.org/10.1111/imr.12065.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Smith D. HIV as an inflammatory disease. Journal of HIV therapy. 2009;14(3):50–1.

    PubMed  Google Scholar 

  194. Vyboh K, Jenabian MA, Mehraj V, Routy JP. HIV and the gut microbiota, partners in crime: breaking the vicious cycle to unearth new therapeutic targets. Journal of immunology research. 2015;2015:614127. https://doi.org/10.1155/2015/614127.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Brenchley JM, Douek DC. Microbial translocation across the GI tract. Annual review of immunology. 2012;30:149–73. https://doi.org/10.1146/annurev-immunol-020711-075001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nature medicine. 2006;12(12):1365–71. https://doi.org/10.1038/nm1511.

    Article  CAS  PubMed  Google Scholar 

  197. Rodger AJ, Fox Z, Lundgren JD, Kuller LH, Boesecke C, Gey D, et al. Activation and coagulation biomarkers are independent predictors of the development of opportunistic disease in patients with HIV infection. The Journal of infectious diseases. 2009;200(6):973–83. https://doi.org/10.1086/605447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. McKinnon LR, Nyanga B, Kim CJ, Izulla P, Kwatampora J, Kimani M, et al. Early HIV-1 infection is associated with reduced frequencies of cervical Th17 cells. Journal of acquired immune deficiency syndromes. 2015;68(1):6–12. https://doi.org/10.1097/QAI.0000000000000389.

    Article  CAS  PubMed  Google Scholar 

  199. Schuetz A, Deleage C, Sereti I, Rerknimitr R, Phanuphak N, Phuang-Ngern Y, et al. Initiation of ART during early acute HIV infection preserves mucosal Th17 function and reverses HIV-related immune activation. PLoS pathogens. 2014;10(12):e1004543. https://doi.org/10.1371/journal.ppat.1004543.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A, Hogan C, et al. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004;200(6):761–70. https://doi.org/10.1084/jem.20041196.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Brenchley JM, Douek DC. HIV infection and the gastrointestinal immune system. Mucosal Immunol. 2008;1(1):23–30. https://doi.org/10.1038/mi.2007.1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Macal M, Sankaran S, Chun TW, Reay E, Flamm J, Prindiville TJ, et al. Effective CD4+ T-cell restoration in gut-associated lymphoid tissue of HIV-infected patients is associated with enhanced Th17 cells and polyfunctional HIV-specific T-cell responses. Mucosal Immunol. 2008;1(6):475–88. https://doi.org/10.1038/mi.2008.35.

    Article  CAS  PubMed  Google Scholar 

  203. Goto Y, Obata T, Kunisawa J, Sato S, Ivanov II, Lamichhane A, et al. Innate lymphoid cells regulate intestinal epithelial cell glycosylation. Science. 2014;345(6202):1254009. https://doi.org/10.1126/science.1254009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Pham TA, Clare S, Goulding D, Arasteh JM, Stares MD, Browne HP, et al. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization resistance to an opportunistic pathogen. Cell Host Microbe. 2014;16(4):504–16. https://doi.org/10.1016/j.chom.2014.08.017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Terahara K, Nochi T, Yoshida M, Takahashi Y, Goto Y, Hatai H, et al. Distinct fucosylation of M cells and epithelial cells by Fut1 and Fut2, respectively, in response to intestinal environmental stress. Biochemical and biophysical research communications. 2011;404(3):822–8. https://doi.org/10.1016/j.bbrc.2010.12.067.

    Article  CAS  PubMed  Google Scholar 

  206. Marcobal A, Barboza M, Sonnenburg ED, Pudlo N, Martens EC, Desai P, et al. Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways. Cell host & microbe. 2011;10(5):507–14. https://doi.org/10.1016/j.chom.2011.10.007.

    Article  CAS  Google Scholar 

  207. Bergstrom K, Fu J, Johansson ME, Liu X, Gao N, Wu Q, et al. Core 1- and 3-derived O-glycans collectively maintain the colonic mucus barrier and protect against spontaneous colitis in mice. Mucosal immunology. 2016. https://doi.org/10.1038/mi.2016.45.

  208. Nishida A, Nagahama K, Imaeda H, Ogawa A, Lau CW, Kobayashi T, et al. Inducible colitis-associated glycome capable of stimulating the proliferation of memory CD4+ T cells. The Journal of experimental medicine. 2012;209(13):2383–94. https://doi.org/10.1084/jem.20112631.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Srikrishna G, Turovskaya O, Shaikh R, Newlin R, Foell D, Murch S, et al. Carboxylated glycans mediate colitis through activation of NF-kappa B. Journal of immunology. 2005;175(8):5412–22.

    Article  CAS  Google Scholar 

  210. Fu J, Wei B, Wen T, Johansson ME, Liu X, Bradford E, et al. Loss of intestinal core 1-derived O-glycans causes spontaneous colitis in mice. The Journal of clinical investigation. 2011;121(4):1657–66. https://doi.org/10.1172/JCI45538.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Goto Y, Uematsu S, Kiyono H. Epithelial glycosylation in gut homeostasis and inflammation. Nat Immunol. 2016;17(11):1244–51. https://doi.org/10.1038/ni.3587.

    Article  CAS  PubMed  Google Scholar 

  212. Pickard JM, Chervonsky AV. Intestinal fucose as a mediator of host-microbe symbiosis. J Immunol. 2015;194(12):5588–93. https://doi.org/10.4049/jimmunol.1500395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Comstock LE, Kasper DL. Bacterial glycans: key mediators of diverse host immune responses. Cell. 2006;126(5):847–50. https://doi.org/10.1016/j.cell.2006.08.021.

    Article  CAS  PubMed  Google Scholar 

  214. Becker DJ, Lowe JB. Fucose: biosynthesis and biological function in mammals. Glycobiology. 2003;13(7):41R–53R. https://doi.org/10.1093/glycob/cwg054.

    Article  CAS  PubMed  Google Scholar 

  215. Domino SE, Hiraiwa N, Lowe JB. Molecular cloning, chromosomal assignment and tissue-specific expression of a murine alpha(1,2)fucosyltransferase expressed in thymic and epididymal epithelial cells. The Biochemical journal. 1997;327(Pt 1):105–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Kaisho T, Takeuchi O, Kawai T, Hoshino K, Akira S. Endotoxin-induced maturation of MyD88-deficient dendritic cells. Journal of immunology. 2001;166(9):5688–94.

    Article  CAS  Google Scholar 

  217. Pickard JM, Maurice CF, Kinnebrew MA, Abt MC, Schenten D, Golovkina TV, et al. Rapid fucosylation of intestinal epithelium sustains host-commensal symbiosis in sickness. Nature. 2014;514(7524):638–41. https://doi.org/10.1038/nature13823.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Kelly RJ, Rouquier S, Giorgi D, Lennon GG, Lowe JB. Sequence and expression of a candidate for the human Secretor blood group alpha(1,2)fucosyltransferase gene (FUT2). Homozygosity for an enzyme-inactivating nonsense mutation commonly correlates with the non-secretor phenotype. J Biol Chem. 1995;270(9):4640–9.

    Article  CAS  PubMed  Google Scholar 

  219. Koda Y, Soejima M, Liu Y, Kimura H. Molecular basis for secretor type alpha(1,2)-fucosyltransferase gene deficiency in a Japanese population: a fusion gene generated by unequal crossover responsible for the enzyme deficiency. Am J Hum Genet. 1996;59(2):343–50.

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Aghdassi AA, Weiss FU, Mayerle J, Lerch MM, Simon P. Genetic susceptibility factors for alcohol-induced chronic pancreatitis. Pancreatology. 2015;15(4 Suppl):S23–31. https://doi.org/10.1016/j.pan.2015.05.476.

    Article  CAS  PubMed  Google Scholar 

  221. Folseraas T, Melum E, Rausch P, Juran BD, Ellinghaus E, Shiryaev A, et al. Extended analysis of a genome-wide association study in primary sclerosing cholangitis detects multiple novel risk loci. J Hepatol. 2012;57(2):366–75. https://doi.org/10.1016/j.jhep.2012.03.031.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, Ahmad T, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci. Nat Genet. 2010;42(12):1118–25. https://doi.org/10.1038/ng.717.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Ishitoya S, Yamamoto S, Mitsumori K, Ogawa O, Terai A. Non-secretor status is associated with female acute uncomplicated pyelonephritis. BJU Int. 2002;89(9):851–4.

    Article  CAS  PubMed  Google Scholar 

  224. Svensson L, Petersson A, Henry SM. Secretor genotyping for A385T, G428A, C571T, C628T, 685delTGG, G849A, and other mutations from a single PCR. Transfusion. 2000;40(7):856–60.

    Article  CAS  PubMed  Google Scholar 

  225. Yu LC, Yang YH, Broadberry RE, Chen YH, Chan YS, Lin M. Correlation of a missense mutation in the human Secretor alpha 1,2-fucosyltransferase gene with the Lewis(a+b+) phenotype: a potential molecular basis for the weak Secretor allele (Sew). Biochem J. 1995;312(Pt 2):329–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Blackwell CC, Jonsdottir K, Hanson M, Todd WT, Chaudhuri AK, Mathew B, et al. Non-secretion of ABO antigens predisposing to infection by Neisseria meningitidis and Streptococcus pneumoniae. Lancet. 1986;2(8501):284–5.

    Article  CAS  PubMed  Google Scholar 

  227. Blackwell CC, Jonsdottir K, Hanson MF, Weir DM. Non-secretion of ABO blood group antigens predisposing to infection by Haemophilus influenzae. Lancet. 1986;2(8508):687.

    Article  CAS  PubMed  Google Scholar 

  228. Magalhaes A, Gomes J, Ismail MN, Haslam SM, Mendes N, Osorio H, et al. Fut2-null mice display an altered glycosylation profile and impaired BabA-mediated Helicobacter pylori adhesion to gastric mucosa. Glycobiology. 2009;19(12):1525–36. https://doi.org/10.1093/glycob/cwp131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Magalhaes A, Reis CA. Helicobacter pylori adhesion to gastric epithelial cells is mediated by glycan receptors. Braz J Med Biol Res. 2010;43(7):611–8.

    Article  CAS  PubMed  Google Scholar 

  230. Azevedo M, Eriksson S, Mendes N, Serpa J, Figueiredo C, Resende LP, et al. Infection by Helicobacter pylori expressing the BabA adhesin is influenced by the secretor phenotype. J Pathol. 2008;215(3):308–16. https://doi.org/10.1002/path.2363.

    Article  CAS  PubMed  Google Scholar 

  231. Boren T, Falk P, Roth KA, Larson G, Normark S. Attachment of Helicobacter pylori to human gastric epithelium mediated by blood group antigens. Science. 1993;262(5141):1892–5.

    Article  CAS  PubMed  Google Scholar 

  232. Lewis ZT, Totten SM, Smilowitz JT, Popovic M, Parker E, Lemay DG, et al. Maternal fucosyltransferase 2 status affects the gut bifidobacterial communities of breastfed infants. Microbiome. 2015;3:13. https://doi.org/10.1186/s40168-015-0071-z.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Lindesmith L, Moe C, Marionneau S, Ruvoen N, Jiang X, Lindblad L, et al. Human susceptibility and resistance to Norwalk virus infection. Nat Med. 2003;9(5):548–53. https://doi.org/10.1038/nm860.

    Article  CAS  PubMed  Google Scholar 

  234. Rayes A, Morrow AL, Payton LR, Lake KE, Lane A, Davies SM. A genetic modifier of the gut microbiome influences the risk of graft-versus-host disease and bacteremia after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2016;22(3):418–22. https://doi.org/10.1016/j.bbmt.2015.11.017.

    Article  PubMed  Google Scholar 

  235. Smilowitz JT, O’Sullivan A, Barile D, German JB, Lonnerdal B, Slupsky CM. The human milk metabolome reveals diverse oligosaccharide profiles. J Nutr. 2013;143(11):1709–18. https://doi.org/10.3945/jn.113.178772.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nature reviews Cancer. 2015;15(9):540–55. https://doi.org/10.1038/nrc3982.

    Article  CAS  PubMed  Google Scholar 

  237. Gubbels JA, Felder M, Horibata S, Belisle JA, Kapur A, Holden H, et al. MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells. Mol Cancer. 2010;9:11. https://doi.org/10.1186/1476-4598-9-11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. RodrIguez E, Schetters STT, van Kooyk Y. The tumour glyco-code as a novel immune checkpoint for immunotherapy. Nat Rev Immunol. 2018;18(3):204–11. https://doi.org/10.1038/nri.2018.3.

    Article  CAS  PubMed  Google Scholar 

  239. Danishefsky SJ, Shue YK, Chang MN, Wong CH. Development of Globo-H cancer vaccine. Accounts of chemical research. 2015;48(3):643–52. https://doi.org/10.1021/ar5004187.

    Article  CAS  PubMed  Google Scholar 

  240. Huang YL, Hung JT, Cheung SK, Lee HY, Chu KC, Li ST, et al. Carbohydrate-based vaccines with a glycolipid adjuvant for breast cancer. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(7):2517–22. https://doi.org/10.1073/pnas.1222649110.

    Article  PubMed  PubMed Central  Google Scholar 

  241. Bull C, Boltje TJ, Wassink M, de Graaf AM, van Delft FL, den Brok MH, et al. Targeting aberrant sialylation in cancer cells using a fluorinated sialic acid analog impairs adhesion, migration, and in vivo tumor growth. Mol Cancer Ther. 2013;12(10):1935–46. https://doi.org/10.1158/1535-7163.MCT-13-0279.

    Article  CAS  PubMed  Google Scholar 

  242. Dube DH, Bertozzi CR. Glycans in cancer and inflammation--potential for therapeutics and diagnostics. Nat Rev Drug Discov. 2005;4(6):477–88. https://doi.org/10.1038/nrd1751.

    Article  CAS  PubMed  Google Scholar 

  243. Jennewein MF, Alter G. The immunoregulatory roles of antibody glycosylation. Trends Immunol. 2017;38(5):358–72. https://doi.org/10.1016/j.it.2017.02.004.

    Article  CAS  PubMed  Google Scholar 

  244. Lisacek F, Mariethoz J, Alocci D, Rudd PM, Abrahams JL, Campbell MP, et al. Databases and associated tools for glycomics and glycoproteomics. Methods Mol Biol. 2017;1503:235–64. https://doi.org/10.1007/978-1-4939-6493-2_18.

    Article  CAS  PubMed  Google Scholar 

  245. Miles D, Papazisis K. Rationale for the clinical development of STn-KLH (Theratope) and anti-MUC-1 vaccines in breast cancer. Clin Breast Cancer. 2003;3(Suppl 4):S134–8.

    Article  PubMed  Google Scholar 

  246. Miles D, Roche H, Martin M, Perren TJ, Cameron DA, Glaspy J, et al. Phase III multicenter clinical trial of the sialyl-TN (STn)-keyhole limpet hemocyanin (KLH) vaccine for metastatic breast cancer. Oncologist. 2011;16(8):1092–100. https://doi.org/10.1634/theoncologist.2010-0307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Posey AD Jr, Schwab RD, Boesteanu AC, Steentoft C, Mandel U, Engels B, et al. Engineered CAR T cells targeting the cancer-associated Tn-glycoform of the membrane mucin MUC1 control adenocarcinoma. Immunity. 2016;44(6):1444–54. https://doi.org/10.1016/j.immuni.2016.05.014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Tacken PJ, de Vries IJ, Torensma R, Figdor CG. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol. 2007;7(10):790–802. https://doi.org/10.1038/nri2173.

    Article  CAS  PubMed  Google Scholar 

  249. Unger WW, Mayer CT, Engels S, Hesse C, Perdicchio M, Puttur F, et al. Antigen targeting to dendritic cells combined with transient regulatory T cell inhibition results in long-term tumor regression. Oncoimmunology. 2015;4(8):e970462. https://doi.org/10.4161/21624011.2014.970462.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The authors would like to thank Marli Markovitz for providing the artwork and graphic design (https://marlimark13.wixsite.com/marlim) and to Rachel E. Locke, Ph.D, who provided critical comments and editing.

Funding

MA-M is funded by the following grants: NIH (R21 AI129636, R21 NS106970, and P30CA010815-49S2), W.W. Smith Charitable Trust (grant # A1701), Campbell Foundation award, Penn Center for AIDS Research (P30 AI 045008), and The Foundation for AIDS Research (amfAR) Impact grant # 109840-65-RGRL. MA-M is a member of the investigation team of the NIH-funded BEAT-HIV Martin Delaney Collaboratory to cure HIV-1 infection (1UM1Al126620). Additionally, GL is supported by funding from the European Structural and Investments funds for projects “New generation of high throughput glycoanalytical services” (contract # KK.01.2.1.01.0003) and “Croatian National Centre of Research Excellence in Personalized Healthcare” (contract # KK.01.1.1.01.0010).

Author information

Authors and Affiliations

Authors

Contributions

MA-M conceived and designed the review. All the authors wrote and edited the review.

Corresponding author

Correspondence to Mohamed Abdel-Mohsen.

Ethics declarations

Conflict of Interest

GL declares he is a founder and owner of Genos Ltd, a biotech company, that specializes in glycan analysis and has several patents in the field. IT-A is an employee of Genos Ltd. Other authors have no competing interests.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on HIV Pathogenesis and Treatment

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Colomb, F., Giron, L.B., Trbojevic-Akmacic, I. et al. Breaking the Glyco-Code of HIV Persistence and Immunopathogenesis. Curr HIV/AIDS Rep 16, 151–168 (2019). https://doi.org/10.1007/s11904-019-00433-w

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11904-019-00433-w

Keywords

Navigation