Review Topical Sections

Glucuronoxylomannan: the salient polysaccharide in cryptococcal immunity

  • Received: 14 March 2022 Revised: 21 May 2022 Accepted: 15 June 2022 Published: 23 June 2022
  • Cryptococcal meningitis (CM) is a dominant cause of morbidity and mortality among patients with human immunodeficiency virus/ acquired immune deficiency syndrome (HIV/AIDS) caused by Cryptococcus neoformans and Cryptococcus gattii species complex. The complex is composed of closely related members, yet with diverse epidemiology, pathogenesis, and drug-resistant pattern. Cell-mediated immunity is the strongest pillar in immunity to cryptococcosis, further worsening HIV/AIDS patients' scenario. Antifungal resistance and immune evasion again tilt the host-parasite balance in favor of the fungal pathogen. In this regard, researchers are actively challenged to discover immunotherapy and vaccine for CM, to produce specific treatment and prevention that will address CM conventional therapeutics failure. As the major capsular polysaccharide of the Cryptococcus, which is tightly linked to pathogenicity, immunogenicity, and immune evasion, the glucuronoxylomannan (GXM) is cardinally targeted for vaccine and immunotherapy development. Further, the amount of GXM shed in body fluids correlates with the disease severity. Herein, we reviewed the literature with the journey so far in line with GXM as the salient immunological target on cryptococcosis.

    Citation: Mansur Aliyu, Ali Akbar Saboor-Yaraghi, Sadegh Khodavaisy, Behrouz Robat-Jazi, Muhammad Ibrahim Getso. Glucuronoxylomannan: the salient polysaccharide in cryptococcal immunity[J]. AIMS Allergy and Immunology, 2022, 6(2): 71-89. doi: 10.3934/Allergy.2022008

    Related Papers:

  • Cryptococcal meningitis (CM) is a dominant cause of morbidity and mortality among patients with human immunodeficiency virus/ acquired immune deficiency syndrome (HIV/AIDS) caused by Cryptococcus neoformans and Cryptococcus gattii species complex. The complex is composed of closely related members, yet with diverse epidemiology, pathogenesis, and drug-resistant pattern. Cell-mediated immunity is the strongest pillar in immunity to cryptococcosis, further worsening HIV/AIDS patients' scenario. Antifungal resistance and immune evasion again tilt the host-parasite balance in favor of the fungal pathogen. In this regard, researchers are actively challenged to discover immunotherapy and vaccine for CM, to produce specific treatment and prevention that will address CM conventional therapeutics failure. As the major capsular polysaccharide of the Cryptococcus, which is tightly linked to pathogenicity, immunogenicity, and immune evasion, the glucuronoxylomannan (GXM) is cardinally targeted for vaccine and immunotherapy development. Further, the amount of GXM shed in body fluids correlates with the disease severity. Herein, we reviewed the literature with the journey so far in line with GXM as the salient immunological target on cryptococcosis.


    Abbreviations

    188Re

    rhenium-188

    213Bi

    bismuth-213

    CALAS

    cryptococcal antigen latex agglutination system

    C-IRIS

    cryptococcosis-associated immune reconstitution inflammatory syndrome

    CM

    cryptococcal meningitis

    CNPS

    C. neoformans polysaccharide capsule

    CNS

    central nervous system

    CrAg

    cryptococcal capsular antigen

    CTBA

    cetyltrimethylammonium bromide

    DCs

    dendritic cells

    EIA

    enzyme immunoassay

    GalXM

    galactoxylomannan

    GXM

    glucuronoxylomannan

    GXMGal

    glucuronoxylomanogalactan

    GXMR-CAR

    GXM-specific chimeric antigen receptor

    GXM-TT

    GXM conjugated to tetanus toxoid

    HIV/AIDs

    human immunodeficiency virus/acquired immune deficiency syndrome

    HLA

    human leukocyte antigen

    ICP

    intracranial pressure

    IFN-γ

    interferon-gamma

    IL

    interleukin

    IMMY

    immuno-mycologics

    LFA

    lateral flow assay

    MHC

    major histocompatibility complex

    MPL

    monophosphoryl lipid A

    NETS

    neutrophil extracellular traps

    NLRP3

    nod-like receptors (NLR) family pyrin domain containing 3

    P13-BSA

    P13 conjugated to bovine serum albumin

    P13-TT

    P13 conjugated to tetanus toxoid

    pAPC

    professional antigen presenting cells

    RANTES

    regulated upon activation, normal T cell expressed and secreted

    ROS

    reactive oxygen species

    SCID

    severe combined immunodeficiency

    SRBCs

    sheep red blood cells

    T11TS

    T11 Target structure

    TLRs

    toll-like receptors

    TNF-α

    tumour necrosis factor-alpha

    TT

    tetanus toxoid

    加载中


    Authors' contributions



    MA conceived and designed the manuscript outline, MA and AAS search and review the literature, and MA drew Figures 16. MA, AAS, SK, and MIG wrote the manuscript. BRJ and MIG critically revised the manuscript, and AAs and SK supervised all the processes. All authors reviewed and approved the final version of the manuscript for submission.

    Conflict of interest



    All authors declare no conflicts of interest in this paper.

    [1] Cogliati M (2013) Global molecular epidemiology of Cryptococcus neoformans and Cryptococcus gattii: an atlas of the molecular types. Scientifica 2013: 675213. https://doi.org/10.1155/2013/675213
    [2] Li Y, Zou M, Yin J, et al. (2020) Microbiological, epidemiological, and clinical characteristics of patients with cryptococcal meningitis at a tertiary hospital in China: a 6-year retrospective analysis. Front Microbiol 11: 1837. https://doi.org/10.3389/fmicb.2020.01837
    [3] Taha M, Tartor Y, Ibrahim S, et al. (2020) Molecular typing and susceptibility profile of Cryptococcus neoformans and Cryptococcus gattii species complex: an updated review. J Anim Health Prod 9: 17-26. https://doi.org/10.17582/journal.jahp/2020/9.s1.17.26
    [4] Francisco EC, de Jong AW, Hagen F (2021) Cryptococcosis and Cryptococcus. Mycopathologia 186: 729-731. https://doi.org/10.1007/s11046-021-00577-7
    [5] Kwon-Chung KJ, Bennett JE, Wickes BL, et al. (2017) The case for adopting the “species complex” nomenclature for the etiologic agents of Cryptococcosis. mSphere 2: e00357-16. https://doi.org/10.1128/mSphere.00357-16
    [6] Maziarz EK, Perfect JR (2016) Cryptococcosis. Infect Dis Clin N Am 30: 179-206. https://doi.org/10.1016/j.idc.2015.10.006
    [7] Cassim N, Schnippel K, Coetzee LM, et al. (2017) Establishing a cost-per-result of laboratory-based, reflex cryptococcal antigenaemia screening (CrAg) in HIV+ patients with CD4 counts less than 100 cells/µL using a Lateral Flow Assay (LFA) at a typical busy CD4 laboratory in South Africa. PLoS One 12: e0171675. https://doi.org/10.1371/journal.pone.0171675
    [8] Rivet-Danon D, Guitard J, Grenouillet F, et al. (2015) Rapid diagnosis of cryptococcosis using an antigen detection immunochromatographic test. J Infection 70: 499-503. https://doi.org/10.1016/j.jinf.2014.12.017
    [9] Illnait M, Vilaseca JC, Fernández CM, et al. (2001) Enzyme-linked immunosorbent assay for detection and quantification of Cryptococcus neoformans antigen. Mem Inst Oswaldo Cruz 96: 241-245. https://doi.org/10.1590/S0074-02762001000200018
    [10] Rajasingham R, Meya DB, Boulware DR (2012) Integrating cryptococcal antigen screening and pre-emptive treatment into routine HIV care. J Acquir Immune Defic Syndr 59: 85-91. https://doi.org/10.1097/QAI.0b013e31824c837e
    [11] Márquez M (2019) Lateral flow assay: a world of possibilities for the diagnostic. Austin Clin Microbiol 3: 1010.
    [12] Xu Y, Xia W, Ni F (2020) False-negative serum cryptococcal antigen lateral flow immunoassay result for a patient with disseminated cryptococcal disease. Infect Drug Resist 13: 2877-2881. https://doi.org/10.2147/IDR.S265784
    [13] Wake RM, Jarvis JN, Harrison TS, et al. (2018) Brief report: point of care cryptococcal antigen screening: pipetting finger-prick blood improves performance of immunomycologics lateral flow assay. JAIDS J Acquired Immune Defic Syndr 78: 574-578. https://doi.org/10.1097/QAI.0000000000001721
    [14] Kwizera R, Omali D, Tadeo K, et al. (2021) Evaluation of the dynamiker cryptococcal antigen lateral flow assay for the diagnosis of HIV-associated cryptococcosis. J Clin Microbiol 59: e02421-20. https://doi.org/10.1128/JCM.02421-20
    [15] Tenforde MW, Boyer-Chammard T, Muthoga C, et al. (2020) Diagnostic accuracy of the Biosynex CryptoPS cryptococcal antigen semiquantitative lateral flow assay in patients with advanced HIV disease. J Clin Microbiol 59: e02307-02320. https://doi.org/10.1128/JCM.02307-20
    [16] Mpoza E, Mukaremera L, Kundura DA, et al. (2018) Evaluation of a point-of-care immunoassay test kit “StrongStep” for cryptococcal antigen detection. PLoS One 13: e0190652. https://doi.org/10.1371/journal.pone.0190652
    [17] Rajasingham R, Wake RM, Beyene T, et al. (2019) Cryptococcal meningitis diagnostics and screening in the era of point-of-care laboratory testing. J Clin Microbiol 57: e01238-18. https://doi.org/10.1128/JCM.01238-18
    [18] Reagan KL, McHardy I, Thompson III GR, et al. (2019) Evaluation of the clinical performance of 2 point-of-care cryptococcal antigen tests in dogs and cats. J Vet Intern Med 33: 2082-2089. https://doi.org/10.1111/jvim.15599
    [19] Skipper C, Tadeo K, Martyn E, et al. (2020) Evaluation of serum cryptococcal antigen testing using two novel semiquantitative lateral flow assays in persons with cryptococcal antigenemia. J Clin Microbiol 58: e02046-19. https://doi.org/10.1128/JCM.02046-19
    [20] Noguera MC, Escandón P, Rodríguez J, et al. (2021) Comparison of two commercial tests (Immy vs. Dynamiker) for cryptococcal capsular antigen. Rev Soc Bras Med Trop 54: e0307-2021.
    [21] Shi D, Haas PJ, Boekhout T, et al. (2021) Neglecting genetic diversity hinders timely diagnosis of Cryptococcus infections. J Clin Microbiol 59: e02837-20. https://doi.org/10.1128/JCM.02837-20
    [22] Casadevall A, Coelho C, Cordero RJ, et al. (2019) The capsule of Cryptococcus neoformans. Virulence 10: 822-831. https://doi.org/10.1080/21505594.2018.1431087
    [23] Patil SA, Katyayani S, Arvind N (2012) Significance of antibody detection in the diagnosis of cryptococcal meningitis. J Immunoassay Immunochem 33: 140-148. https://doi.org/10.1080/15321819.2011.606862
    [24] Frases S, Nimrichter L, Viana NB, et al. (2008) Cryptococcus neoformans capsular polysaccharide and exopolysaccharide fractions manifest physical, chemical, and antigenic differences. Eukaryotic Cell 7: 319-327. https://doi.org/10.1128/EC.00378-07
    [25] Camacho E, Casadevall A (2018) Cryptococcal traits mediating adherence to biotic and abiotic surfaces. J Fungi 4: 88. https://doi.org/10.3390/jof4030088
    [26] Ulrich S, Ebel F (2020) Monoclonal antibodies as tools to combat fungal infections. J Fungi 6: 22. https://doi.org/10.3390/jof6010022
    [27] Guazzelli L, Crawford CJ, Ulc R, et al. (2020) A synthetic glycan array containing Cryptococcus neoformans glucuronoxylomannan capsular polysaccharide fragments allows the mapping of protective epitopes. Chem Sci 11: 9209-9217. https://doi.org/10.1039/D0SC01249A
    [28] Devi S, Schneerson R, Egan W, et al. (1991) Cryptococcus neoformans serotype A glucuronoxylomannan-protein conjugate vaccines: synthesis, characterization, and immunogenicity. Infect Immun 59: 3700-3707. https://doi.org/10.1128/iai.59.10.3700-3707.1991
    [29] Cherniak R, Reiss E, Slodki ME, et al. (1980) Structure and antigenic activity of the capsular polysaccharide of Cryptococcus neoformans serotype A. Mol Immunol 17: 1025-1032. https://doi.org/10.1016/0161-5890(80)90096-6
    [30] Urai M, Kaneko Y, Ueno K, et al. (2016) Evasion of innate immune responses by the highly virulent Cryptococcus gattii by altering capsule glucuronoxylomannan structure. Front Cell Infect Microbiol 5: 101. https://doi.org/10.3389/fcimb.2015.00101
    [31] Cherniak R, Valafar H, Morris LC, et al. (1998) Cryptococcus neoformans chemotyping by quantitative analysis of 1H nuclear magnetic resonance spectra of glucuronoxylomannans with a computer-simulated artificial neural network. Clin Diagn Lab Immunol 5: 146-159. https://doi.org/10.1128/CDLI.5.2.146-159.1998
    [32] Kuttel MM, Casadevall A, Oscarson S (2020) Cryptococcus neoformans capsular GXM conformation and epitope presentation: a molecular modelling study. Molecules 25: 2651. https://doi.org/10.3390/molecules25112651
    [33] McFadden DC, Fries BC, Wang F, et al. (2007) Capsule structural heterogeneity and antigenic variation in Cryptococcus neoformans. Eukaryot Cell 6: 1464-1473. https://doi.org/10.1128/EC.00162-07
    [34] Ueno K, Yanagihara N, Shimizu K, et al. (2020) Vaccines and protective immune memory against cryptococcosis. Biol Pharm Bull 43: 230-239. https://doi.org/10.1248/bpb.b19-00841
    [35] Diamond RD, Bennett JE (1974) Prognostic factors in cryptococcal meningitis: a study in 111 cases. Ann Intern Med 80: 176-181. https://doi.org/10.7326/0003-4819-80-2-176
    [36] Sato K, Kawakami K (2017) Recognition of Cryptococcus neoformans by pattern recognition receptors and its role in host defense to this infection. Med Mycol J 58: J83-J90. https://doi.org/10.3314/mmj.17.011
    [37] Perera N, Yang FL, Chern J, et al. (2018) Carboxylic and O-acetyl moieties are essential for the immunostimulatory activity of glucuronoxylomannan: A novel TLR4 specific immunostimulator from Auricularia auricula-judae. Chem Commun 54: 6995-6998. https://doi.org/10.1039/C7CC09927D
    [38] Monari C, Bistoni F, Casadevall A, et al. (2005) Glucuronoxylomannan, a microbial compound, regulates expression of costimulatory molecules and production of cytokines in macrophages. J Infect Dis 191: 127-137. https://doi.org/10.1086/426511
    [39] de Oliveira HC, Trevijano-Contador N, Garcia-Rodas R (2019) Cryptococcal pathogenicity and morphogenesis. Curr Fungal Infect Rep 13: 67-76. https://doi.org/10.1007/s12281-019-00340-y
    [40] Elsegeiny W, Marr KA, Williamson PR (2018) Immunology of cryptococcal infections: developing a rational approach to patient therapy. Front Immunol 9: 651. https://doi.org/10.3389/fimmu.2018.00651
    [41] Mohamed SH, Nyazika TK, Ssebambulidde K, et al. (2022) Fungal CNS Infections in Africa: The neuroimmunology of cryptococcal meningitis. Front Immunol 13: 804674. https://doi.org/10.3389/fimmu.2022.804674
    [42] Hardison SE, Herrera G, Young ML, et al. (2012) Protective immunity against pulmonary cryptococcosis is associated with STAT1-mediated classical macrophage activation. J Immunol 189: 4060-4068. https://doi.org/10.4049/jimmunol.1103455
    [43] Rohatgi S, Pirofski L (2015) Host immunity to Cryptococcus neoformans. Future Microbiol 10: 565-581. https://doi.org/10.2217/fmb.14.132
    [44] Wozniak KL, Young ML, Wormley FL (2011) Protective immunity against experimental pulmonary cryptococcosis in T cell-depleted mice. Clin Vaccine Immunol 18: 717-723. https://doi.org/10.1128/CVI.00036-11
    [45] Meya DB, Okurut S, Zziwa G, et al. (2019) HIV-associated Cryptococcal immune reconstitution inflammatory syndrome is associated with aberrant t cell function and increased cytokine responses. J Fungi 5: 42. https://doi.org/10.3390/jof5020042
    [46] Zhou Q, Murphy WJ (2006) Immune response and immunotherapy to Cryptococcus infections. Immunol Res 35: 191-208. https://doi.org/10.1385/IR:35:3:191
    [47] Rocha JD, Nascimento MT, Decote-Ricardo D, et al. (2015) Capsular polysaccharides from Cryptococcus neoformans modulate production of neutrophil extracellular traps (NETs) by human neutrophils. Sci Rep 5: 8008. https://doi.org/10.1038/srep08008
    [48] Scriven JE, Graham LM, Schutz C, et al. (2016) A glucuronoxylomannan-associated immune signature, characterized by monocyte deactivation and an increased interleukin 10 level, is a predictor of death in cryptococcal meningitis. J Infect Dis 213: 1725-1734. https://doi.org/10.1093/infdis/jiw007
    [49] Van Dyke MCC, Wormley FL (2018) A call to arms: quest for a cryptococcal vaccine. Trends Microbiol 26: 436-446. https://doi.org/10.1016/j.tim.2017.10.002
    [50] Yoon HA, Kuniholm MH, Nakouzi A, et al. (2016) Association of decreased cryptococcal antibody levels with cryptococcosis-associated immune reconstitution inflammatory syndrome. Open Forum Infect Dis 3: 899. https://doi.org/10.1093/ofid/ofw194.64
    [51] Yoon HA, Nakouzi A, Chang CC, et al. (2019) Association between plasma antibody responses and risk for cryptococcus-associated immune reconstitution inflammatory syndrome. J Infect Dis 219: 420-428. https://doi.org/10.1093/infdis/jiy447
    [52] Maitta RW, Datta K, Chang Q, et al. (2004) Protective and nonprotective human immunoglobulin M monoclonal antibodies to Cryptococcus neoformans glucuronoxylomannan manifest different specificities and gene use profiles. Infect Immun 72: 4810-4818. https://doi.org/10.1128/IAI.72.8.4810-4818.2004
    [53] Maitta RW, Datta K, Pirofski LA (2004) Efficacy of immune sera from human immunoglobulin transgenic mice immunized with a peptide mimotope of Cryptococcus neoformans glucuronoxylomannan. Vaccine 22: 4062-4068. https://doi.org/10.1016/j.vaccine.2004.03.060
    [54] Albuquerque PC, Fonseca FL, Dutra FF, et al. (2014) Cryptococcus neoformans glucuronoxylomannan fractions of different molecular masses are functionally distinct. Mycoses 57: 53. https://doi.org/10.2217/fmb.13.163
    [55] Boulware DR, von Hohenberg M, Rolfes MA, et al. (2016) Human immune response varies by the degree of relative cryptococcal antigen shedding. Open Forum Infect Dis 3: ofv194. https://doi.org/10.1093/ofid/ofv194
    [56] Ana Caroline C, Rella A, Normile T, et al. (2019) Cryptococcus neoformans glucuronoxylomannan and sterylglucoside are required for host protection in an animal vaccination model. Mbio 10: e02909-18. https://doi.org/10.1128/mBio.02909-18
    [57] Decote-Ricardo D, LaRocque-de-Freitas IF, Rocha JDB, et al. (2019) Immunomodulatory role of capsular polysaccharides constituents of Cryptococcus neoformans. Front Med 6: 129. https://doi.org/10.3389/fmed.2019.00129
    [58] Vecchiarelli A, Pericolini E, Gabrielli E, et al. (2011) Cryptococcus neoformans galactoxylomannan is a potent negative immunomodulator, inspiring new approaches in anti-inflammatory immunotherapy. Immunotherapy 3: 997-1005. https://doi.org/10.2217/imt.11.86
    [59] Chuck SL, Sande MA (1989) Infections with Cryptococcus neoformans in the acquired immunodeficiency syndrome. N Engl J Med 321: 794-799. https://doi.org/10.1056/NEJM198909213211205
    [60] Silva E, Silva M, Paula C, et al. (2016) Effect of GXM (glucuronoxylomannan) on the inflammatory response in lung infection caused by Cryptococcus neoformans (serotype A) in immunodeficient murine model (BALB/c-SCID). J Med Microbiol Diagn 5: 4-6.
    [61] Robertson EJ, Najjuka G, Rolfes MA, et al. (2014) Cryptococcus neoformans ex vivo capsule size is associated with intracranial pressure and host immune response in HIV-associated cryptococcal meningitis. J Infect Dis 209: 74-82. https://doi.org/10.1093/infdis/jit435
    [62] Vecchiarelli A, Pericolini E, Gabrielli E, et al. (2013) Elucidating the immunological function of the Cryptococcus neoformans capsule. Future Microbiol 8: 1107-1116. https://doi.org/10.2217/fmb.13.84
    [63] Denham ST, Verma S, Reynolds RC, et al. (2018) Regulated release of cryptococcal polysaccharide drives virulence and suppresses immune cell infiltration into the central nervous system. Infect Immun 86: e00662-17. https://doi.org/10.1128/IAI.00662-17
    [64] Gassiep I, Aye C, Armstrong M, et al. (2018) Correlation between serum cryptococcal antigen titre and meningitis in immunocompetent patients. J Med Microbiol 67: 1515-1518. https://doi.org/10.1099/jmm.0.000830
    [65] Nalintya E, Kiggundu R, Meya D (2016) Evolution of cryptococcal antigen testing: what is new?. Curr Fungal Infect Rep 10: 62-67. https://doi.org/10.1007/s12281-016-0256-3
    [66] Casadevall A (2016) New Insights in to polysaccharide capsule structure and antibody-function from Cryptococcus neoformans. Glycobiology 26: 1386.
    [67] Nami S, Mohammadi R, Vakili M, et al. (2019) Fungal vaccines, mechanism of actions and immunology: A comprehensive review. Biomed Pharmacother 109: 333-344. https://doi.org/10.1016/j.biopha.2018.10.075
    [68] Dromer F, Salamero J, Contrepois A (1987) Production, characterization, and antibody specificity of a mouse monoclonal antibody reactive with Cryptococcus neoformans capsular polysaccharide. Infect Immun 55: 742-748. https://doi.org/10.1128/iai.55.3.742-748.1987
    [69] Arturo Casadevall, Matthew Scharff, Mukherjee J (2002) Antibodies to polysaccharide of C. neoformans. U.S. Patent Application, US20030103977A1 .
    [70] Hancock RE, Nijnik A, Philpott DJ (2012) Modulating immunity as a therapy for bacterial infections. Nat Rev Microbiol 10: 243-254. https://doi.org/10.1038/nrmicro2745
    [71] Bowen A, Wear MP, Cordero RJB, et al. (2017) A monoclonal antibody to Cryptococcus neoformans glucuronoxylomannan manifests hydrolytic activity for both peptides and polysaccharides. J Biol Chem 292: 417-434. https://doi.org/10.1074/jbc.M116.767582
    [72] Kumaresan P, Da Silva T, Laskowski T (2020) Glucuronoxylomannan in the Cryptococcus species capsule as a target for CAR+ T-cell therapy. J Immunol 204: 231. https://doi.org/10.1101/715045
    [73] Tang T, Cheng X, Truong B, et al. (2021) Molecular basis and therapeutic implications of CD40/CD40L immune checkpoint. Pharmacol Therapeut 219: 107709. https://doi.org/10.1016/j.pharmthera.2020.107709
    [74] Antachopoulos C, Walsh TJ (2012) Immunotherapy of Cryptococcus infections. Clin Microbiol Infec 18: 126-133. https://doi.org/10.1111/j.1469-0691.2011.03741.x
    [75] Shah M, Garg G, Dadachova E (2015) Preclinical testing of radiopharmaceuticals for novel applications in HIV, bacterial and fungal infectious diseases. Q J Nucl Med Mol Imaging 59: 317-326.
    [76] Helal M, Dadachova E (2018) Radioimmunotherapy as a novel approach in HIV, bacterial, and fungal infectious diseases. Cancer Biother Radio 33: 330-335. https://doi.org/10.1089/cbr.2018.2481
    [77] Jones TH, McClelland EE, McFeeters H, et al. (2017) Novel antifungal activity for the lectin scytovirin: Inhibition of Cryptococcus neoformans and Cryptococcus gattii. Front Microbiol 8: 755. https://doi.org/10.3389/fmicb.2017.00755
    [78] Banerjee S, Khajanchi S, Chaudhuri S (2015) A mathematical model to elucidate brain tumor abrogation by immunotherapy with T11 target structure. PLoS One 10: e0123611. https://doi.org/10.1371/journal.pone.0123611
    [79] Hazra I, Sk Md OF, Datta A, et al. (2019) T11TS immunotherapy augments microglial and lymphocyte protective immune responses against Cryptococcus neoformans in the brain. Scand J Immunol 89: e12733. https://doi.org/10.1111/sji.12733
    [80] Omar F, Hazra I, Mondal S, et al. (2020) T11TS immunotherapy potentiates the repressed calcineurin-NFAT signalling pathway of T cells in Cryptococcus neoformans infected rats: a cue towards T-cell activation for antifungal immunity. J Appl Microbiol 129: 753-767. https://doi.org/10.1111/jam.14631
    [81] Omar F, Hazra I, Datta A, et al. (2020) Regulation of key molecules of immunological synapse by T11TS immunotherapy abrogates Cryptococcus neoformans infection in rats. Mol Immunol 122: 207-221. https://doi.org/10.1016/j.molimm.2020.04.021
    [82] Murphy JW, Schafer F, Casadevall A, et al. (1998) Antigen-induced protective and nonprotective cell-mediated immune components against. Cryptococcus neoformans. Infect Immun 66: 2632-2639. https://doi.org/10.1128/IAI.66.6.2632-2639.1998
    [83] Rathore SS, Sathiyamoorthy J, Lalitha C, et al. (2022) A holistic review on Cryptococcus neoformans. Microb Pathogenesis 166: 105521. https://doi.org/10.1016/j.micpath.2022.105521
    [84] Devi SJ (1996) Preclinical efficacy of a glucuronoxylomannan-tetanus toxoid conjugate vaccine of Cryptococcus neoformans in a murine model. Vaccine 14: 841-844. https://doi.org/10.1016/0264-410X(95)00256-Z
    [85] Fleuridor R, Lees A, Pirofski L (2001) A cryptococcal capsular polysaccharide mimotope prolongs the survival of mice with Cryptococcus neoformans infection. J Immunol 166: 1087-1096. https://doi.org/10.4049/jimmunol.166.2.1087
    [86] Datta K, Lees A, Pirofski L (2008) Therapeutic efficacy of a conjugate vaccine containing a peptide mimotope of cryptococcal capsular polysaccharide glucuronoxylomannan. Clin Vaccine Immunol 15: 1176-1187. https://doi.org/10.1128/CVI.00130-08
    [87] Ueno K, Urai M, Ohkouchi K, et al. (2016) Dendritic cell-based vaccine against fungal infection. Vaccine Des 1403: 537-549. https://doi.org/10.1007/978-1-4939-3387-7_30
    [88] Ueno K, Urai M, Sadamoto S, et al. (2019) A dendritic cell-based systemic vaccine induces long-lived lung-resident memory Th17 cells and ameliorates pulmonary mycosis. Mucosal Immunol 12: 265-276. https://doi.org/10.1038/s41385-018-0094-4
  • Reader Comments
  • © 2022 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1507) PDF downloads(137) Cited by(0)

Article outline

Figures and Tables

Figures(6)  /  Tables(1)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog