Review Topical Sections

Glucuronoxylomannan: the salient polysaccharide in cryptococcal immunity

  • 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:

    [1] Darrell O. Ricke . Etiology model for many vaccination adverse reactions, including SARS-CoV-2 spike vaccines. AIMS Allergy and Immunology, 2022, 6(4): 200-215. doi: 10.3934/Allergy.2022015
    [2] Laiba Noor, Syeda Momna Ishtiaq, Farhat Batool, Muhammad Imran Arshad . Age-dependent trends in adverse reactions of SARS-CoV-2 vaccines: A narrative review. AIMS Allergy and Immunology, 2024, 8(3): 146-166. doi: 10.3934/Allergy.2024008
    [3] Daniel Danladi Gaiya, Jonathan Danladi Gaiya, Richard Auta, Aliyu Muhammad, Bege Jonathan, Stella Kuyet Udu, Ekpa Emmanuel, Amina Shehu Bature . The journey so far with SARS-CoV-2 variants: Pathogenesis, immunity and treatments. AIMS Allergy and Immunology, 2023, 7(4): 222-250. doi: 10.3934/Allergy.2023016
    [4] Ken S. Rosenthal, Daniel H. Zimmerman . J-LEAPS vaccines elicit antigen specific Th1 responses by promoting maturation of type 1 dendritic cells (DC1). AIMS Allergy and Immunology, 2017, 1(2): 89-100. doi: 10.3934/Allergy.2017.2.89
    [5] Yu. A. Desheva, A. S. Mamontov, P. G. Nazarov . Contribution of antibody-dependent enhancement to the pathogenesis of coronavirus infections. AIMS Allergy and Immunology, 2020, 4(3): 50-59. doi: 10.3934/Allergy.2020005
    [6] Michael D. Caponegro, Jeremy Tetsuo Miyauchi, Stella E. Tsirka . Contributions of immune cell populations in the maintenance, progression, and therapeutic modalities of glioma. AIMS Allergy and Immunology, 2018, 2(1): 24-44. doi: 10.3934/Allergy.2018.1.24
    [7] Darrell O. Ricke . Rare dizziness, syncope, loss of consciousness, seizure, and risk of falling after vaccination. AIMS Allergy and Immunology, 2023, 7(2): 164-175. doi: 10.3934/Allergy.2023011
    [8] Arosh S. Perera Molligoda Arachchige . Aging and immunity: Unraveling the complex relationship. AIMS Allergy and Immunology, 2023, 7(3): 213-221. doi: 10.3934/Allergy.2023015
    [9] Robert Cote, Laura Lynn Eggink, J. Kenneth Hoober . CLEC receptors, endocytosis and calcium signaling. AIMS Allergy and Immunology, 2017, 1(4): 207-231. doi: 10.3934/Allergy.2017.4.207
    [10] Waleed M. Hussein, Phil M. Choi, Cheng Zhang, Emma Sierecki, Wayne Johnston, Zhongfan Jia, Michael J. Monteiro, Mariusz Skwarczynski, Yann Gambin, Istvan Toth . Investigating the affinity of poly tert-butyl acrylate toward Toll-Like Receptor 2. AIMS Allergy and Immunology, 2018, 2(3): 141-147. doi: 10.3934/Allergy.2018.3.141
  • 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

    The Cryptococcus neoformans and Cryptococcus gattii species complexes are the common causative agents of life-threatening cryptococcal meningitis (CM) among the immunocompromised, especially human immunodeficiency virus/ acquired immune deficiency syndrome (HIV/AIDS) patients, other immunosuppressed patients and to a small extent immunocompetent individuals [1],[2]. If available, timely diagnosis, specific treatment (immunotherapy), and vaccine can help convert the fatality recorded against CM in human populations. After many molecular studies, the C. neoformans/C. gattii species complex was divided into seven species haploids and four inter-species diploid/aneuploid hybrids. The species exhibit varying epidemiology, pathogenicity, and chemotherapeutic susceptibility, but all C. neoformans/C. gattii species complex members cause pneumonia and meningoencephalitis [3]. Generally, in this review, we use C. neoformans in the write-up and figures to refer to C. neoformans/C. gattii species complex.

    Classification of the C. neoformans/C. gattii species complex summarized in Table 1 below were as follows, under C. neoformans we have C. neoformans sensu stricto formally serotype A (VNI/AFLP1 and VNII/VNB/AFLP1A, VNII/AFLP1B) and C. deneoformans (VNIV/AFLP2) previously serotype D. While the C. gattii had five species under it namely, C. gattii sensu lato (VGI/AFLP4: serotype B), C. bacillisporus (VGIII/AFLP5: serotype B & C), C. deuterogattii (VGII/AFLP6: serotype B), C. tetragattii (VGIV/AFLP7: serotype C), and C. decagattii (VGIV and VGIIIc/AFLP10: serotype B). Further, the diploid/aneuploid species hybrids between C. neoformans and C. gattii are C. neoformans × C. deneoformans hybrid (AFLP3/VNIII:serotype AD), C. deneoformans × C. gattii hybrid (AFLP8:serotype DB), C. neoformans × C. gattii hybrid (AFLP9:serotype AB) and C. neoformans × C. deuterogattii hybrid (AFLP11:serotype AB) [3][5].

    Table 1.  Classification of the C. neoformans/C. gattii species complex.
    Parent specie(s) Species Serotype(s)
    C. neoformans C. neoformans sensu stricto (VNI/AFLP1, VNII/VNB/AFLP1A, VNII/AFLP1B) A
    C. deneoformans (VNIV/AFLP2) D
    C. gattii C. gattii sensu lato (VGI/AFLP4) B
    C. bacillisporus (VGIII/AFLP5) B and C
    C. deuterogattii (VGII/AFLP6) B
    C. tetragattii (VGIV/AFLP7) C
    C. decagattii (VGIV and VGIIIc/AFLP10) B
    Hybrid (diploid/aneuploidy) C. neoformans and C. gattii C. neoformans × C. deneoformans hybrid (AFLP3/VNIII) AD
    C. deneoformans × C. gattii hybrid (AFLP8) DB
    C. neoformans × C. gattii hybrid (AFLP9) AB
    C. neoformans × C. deuterogattii hybrid (AFLP11) AB

     | Show Table
    DownLoad: CSV

    The respiratory tract is the primary route of entry for Cryptococcus; the lungs and central nervous system (CNS) are the common sites of the invasion, causing a spectrum of infections from simple colonization to systemic/invasive fungemia and meningitis [6]. Figure 1 below outlines the pathogenesis of CM. Cryptococcal screening is recommended in HIV patients with a CD4 count of 100 cells/µL or less [7]. Conventionally, cryptococcosis is diagnosed in the laboratory using Indian ink staining, this procedure is retarded by reduced sensitivity. The fungal culture is considered the gold standard, however, a definitive diagnosis can be reached in one to two weeks. Serological detection of fungal capsular polysaccharides plays a significant role in CM diagnosis [8]. Enzyme immunoassay (EIA) and latex agglutination assays have been employed successfully in targeting the cryptococcal capsular antigen in body fluids to diagnose cryptococcosis [8],[9].

    Figure 1.  Pathogenesis of cryptococcal meningitis. A schematic representation of the pathogenesis of C. neoformans from transmission to the evolution of cryptococcal meningitis.

    The immunochromatographic dipstick assay detects the cryptococcal antigen, principally glucuronoxylomannan (GXM), in serum by the fourth week of infection before meningism [10]. The cryptococcal capsular antigen (CrAg) lateral flow assay (LFA) being more sensitive than the CrAg latex detects 1 ng of CrAg/mL, while the latter detects 19 ng of CrAg/mL of the sample. The CrAg LFA employs simple technology that is convenient for mass production at affordable cost, for rapid testing, and for easy interpretation on a portable device for testing many parameters in a sample. Contrastingly, CrAg LFA has some setbacks as the chemical reporters can fail and the test is inappropriate for assays requiring high analytical sensitivity and reproducibility [11]. A study reported a false-negative serum CrAg LFA in a patient with disseminated cryptococcosis and positive C. neoformans blood culture result [12].

    The cryptococcal antigen latex agglutination system (CALAS) is a quantitative/semiquantitative assay that detects cryptococcal polysaccharide capsular antigens. Despite CALAS accuracy in the diagnosis of CM using CSF, it had been substantially replaced by CrAg LFA due to the former's unfavorable requirement of laboratory expertise, long incubation, and interpretation dexterity. The FDA-approved Immuno-Mycologics (IMMY) CrAg LFA is considered superior among CrAg LFA platforms, however, needle prick-sample proved less sensitive compared to blood collected with a pipette and read after 20 minutes [13].

    Other CrAg LFA available in the market include Dynamiker [14], CryptoPS (Biosynex) [15], StrongStep (Liming Bio) [16], and FungiXpert cryptococcal capsular polysaccharide K-Set (Genobio) [17]. Scientists have reported extensively on the properties of these CrAg LFA tests and their comparison with IMMY or CALAS [15],[16],[18][20]. Recently, researchers compared the four CrAg LFA certified in Europe and reported that IMMY and FungiXpert LFAs were able to detect all the species in the C. neoformans/gattii complex, however, Dynamiker and CryptoPS failed to diagnose C. tetragattii and C. bacillisporus, also C. deuterogattii is a blind spot on CrptoPS. Importantly C. tetragattii is prevalent in Subsaharan Africa and the Indo-Pakistani subcontinent; accounting for one-fifth of CM among HIV/AIDs patients [21].

    The C. neoformans polysaccharide capsule (CNPS); principal virulent factor, salient serological target, and critical immune evasion factor that is shed in culture and body fluids is composed of GXM (90%), galactoxylomannan (GalXM)—recently reviewed as GXMGal, and mannoproteins [22]. The intact capsular and the secreted GXM are both utilized for cryptococcal studies [23]. However, the duo possesses distinct chemical, physical, and antigenic properties [24]. The GXM is common to all the species in the Cryptococcus neoformans/gattii complex [25].

    In the early 1990s, advancement was recorded in the identification of the GXM part of CNPS, which is specific for C. neoformans as such murine monoclonal antibodies were produced and characterized, which showed the upper hand in the diagnosis. Moreover, studies to uncover the role of anti-GXM for possible immunotherapy of C. neoformans commenced in pari-passu [26]. The cryptococcal GXM, unlike proteinaceous antigens, provoked an inadequate immune response. Previous studies coupled GXM to carrier proteins and use adjuvants to boost humoral immune response [27],[28].

    The component carbohydrate residues of GXM are mannose, xylose, and glucuronic acid in decreasing order. The mannan moieties provide GXM backbone through their lineal linkage to each other with α-(1→3) bond, while two xylose molecules occupy positions β-(1→2) and β-(1→4), and a glucuronic acid moiety takes up position β-(1→2) of the mannose molecule forming the octasaccharide structure of GXM as shown in Figure 2 below [29]. The GXM mannose backbone is substantially acylated at position 6 [30]. The GXM polysaccharide expressed on C. neoformans exercises six motifs triad, present in different motif combinations in any given GXM; a complexity superior to bacterial single oligosaccharide motif [31].

    Figure 2.  A schematic diagram of GXM: Mannose (purple hexagons) form the backbone structure of GXM linked to each other by α-(1→3) bond also linked to xylose (green pentagon) through β-(1→2) and β-(1→4) or glucuronic acid (orange hexagon) through β-(1→2) bonding. GXM: Glucuronoxylomannan.

    The C. neoformans sensu stricto (serotype A) dominant triad has a six-residue repeat unit bearing two xylose side chains at β-(1→2) [32]. Glucuronic acid position confers structural variability of GXM having different motifs within the same isogenic C. neoformans phenotypes, yielding considerable structural and antigenic variations [33]. GXM provides the framework for the serotype classification of Cryptococcus, based on structural diversification of the GXM molecule. As each GXM possesses six differing triads (motifs) of oligosaccharides that account for its diversity making the molecule highly heterogeneous [32].

    The cryptococcal GXM is tolerogenic, poorly immunogenic, and provoked T cell-independent humoral immune response in humans and experimental animals [28],[34]. Only 40% of CM patients produce detectable antibodies against cryptococcal polysaccharide capsular antigens [35]. The dendritic cells (DCs) recognize the C. neoformans GXM using toll-like receptors (TLR)-2 and TLR4, leading to the fungal agent's phagocytosis with no role in cytotoxicity. Bedsides, the protective immune response follows recognition of GXM from ingested fungus by nod-like receptors (NLR) family pyrin domain containing 3 (NLRP3) that promote expression of IL-1β [36], as depicted in Figure 3b below. TLR4 is essential for GXM recognition and subsequent activation of macrophages [37].

    Other pattern recognition receptors utilized by monocyte-derived macrophages to recognize GXM for subsequent uptake include CD14, CD18, and FcγRII, in addition to TLR2 and TLR4. Exposure of these macrophages to GXM demonstrates the immune system's downregulation through decreased expression of MHC I and MHC II and increased secretion of IL-10 which is counterproductive to cryptococcal immunity, as shown in Figure 3a below [38]. Phagocytic cells: macrophages and neutrophils form the first-line innate defense against cryptococcosis [39].

    Figure 3.  The double edge sword immune response to GXM. (a) Immunity response to C. neoformans through PAMPs like CD14, CD18, FCγII (CD32), TLR2 and TLR4 on pAPC usually resulted in an immune evasive response, where the pAPC will secrete IL-10, an anti-inflammatory cytokine that favors cryptococcal growth. (b) While the proinflammatory response from the pAPC usually results from the recognition of fungal antigen by NLPR3 within the cytosol and subsequent secretion of IL-1β, a proinflammatory cytokine that favors cryptococcal clearance. Interleukin (IL), nod-like receptor (NLR) family pyrin domain containing 3 (NLRP3), pathogen-associated molecular pattern (PAMPs), professional antigen-presenting cell (pAPC), toll-like receptor (TLR).

    The proinflammatory Th1 subtypes of CD4+ T cells that produce IFNγ as its signature cytokine was shown to be a key player in immunity to cryptococcosis, through its ability to polarize macrophages to M1 phenotype: fungicidal [40],[41]. This fact was further supported by a higher prevalence of CM among HIV/AIDs patients with severely low CD4 cell count [41],[42], a similar phenomenon was observed in idiopathic CD4 lymphocytopenia [43]. CD4+ T cells are essential for cryptococcal clearance, a property that was shown to be transferred in the murine study, in addition to macrophages and neutrophils attraction [43]. In a murine study, it was reported that the presence of CD4+ or CD8+ T cells is indispensable for cryptococcal immunity [44]. Another study found IL-17A secreted by tissue-resident memory T cells to be a requisite towards immunity to cryptococcal infection in the study mice [34]. To further showcase the significance of Th1 and Th17 CD4+ T cells, scientists reported consistently low levels of IFNγ and IL-17 secreting cells among HIV/AIDs patients with CM complicated by immune reconstitution inflammatory syndrome (IRIS) [45]. IFNγ, IL-17A and TNFα production by CD4+ T cells function in immune cells chemotaxis and granuloma formation at the local site of cryptococcal infection; hence IL-17A(−/−) mice failed to form granuloma after dendritic cell vaccination for cryptococcal infection [34]. In sum, cell-mediated immunity through Th1 and M1 macrophages, in addition to humoral immunity with associated complement activation are salient towards cryptococcal elimination [46].

    In an in vitro study, Rocha and coworkers reported that C. neoformans GXM blocks the release of neutrophil extracellular traps (NETs) and inhibits the generation of reactive oxygen species (ROS) needed for fungal killing by human neutrophils [47]. O-acetyl groups present in GXM are essential to immune evasion by the Cryptococcus [30]. Increased serum levels of GXM in patients with CM were found to be associated with monocyte anergy: reduced monocytes expression of HLA-DR and TNF-α, reduce Th1 response, increase expression of IL-6 and IP-10, and increase neutrophils count. Cumulatively, labeled as a mortal immune signature in CM patients [48]. Cell-mediated immunity through the expression of Th1 cytokines is a prerequisite to anti-cryptococcal immunity, while CD8+ T cells provide anti-cryptococcal immunity in the absence of CD4+ T cells [49]. GXM Immune evasion to phagocytes was outlined in Figure 4 below.

    Figure 4.  GXM Immune evasion. Phagocytes namely monocytes, neutrophils, and macrophages provide a defense to C. neoformans through engulfment of C. neoformans, but the microbe devises evasive strategies to counter lasting immunity from these cells through downregulation of proinflammatory responses, and influencing upregulation of anti-inflammatory response as shown for each cell type in the above diagram. DCs, not shown in the diagram, are superior to the above-mentioned cells in immunity to cryptococcosis. CXCL10: C-X-C motif chemokine ligand 10; DCs: dendritic cells; GXM: glucuronoxylomannan; HLA: human leukocyte antigen; IFN-γ: interferon-gamma; IL: interleukin; MHC: major histocompatibility complex; NETS: neutrophil extracellular traps; ROS: reactive oxygen species; TNF-α: tumor necrosis factor-alpha.

    Scientists found a direct association between a low serum GXM IgM and the development of cryptococcosis-associated immune reconstitution inflammatory syndrome (C-IRIS) among patients with CM. Further, GXM-specific IgM deficiency was shown to be associated with disseminated cryptococcosis among the study mice [50]. No association was found between C-IRIS and B cell populations [51]. Mice immunization with GXM conjugated to protein carrier yields different monoclonal antibodies (mAbs), some with protective properties, others with non-protective specificity [52]. GXM-specific IgM was shown to confer protective immunity to cryptococcosis [53].

    The C. neoformans express GXM of varying molecular masses that were recognized by murine bone marrow-derived macrophages, but have a differing effect on the expression of IL-10, IL-6, TNF-α, and regulated upon activation, normal T cell expressed and secreted (RANTES:CCL5) [54]. The CSF secretion of GXM is positively correlated with levels of IL-6, IL-7, IL-8, and TNF-α expression among patients with CM [55]. Extracellular vesicles containing GXM and sterylglucosides were shown to protect against secondary cryptococcosis [56]. C. neoformans display varying cell morphology types during infection to evade the host's immune system [39].

    A complex of GXM and GaLXM: glucuronoxylomanogalactan (GXMGal) released during infection evades the immune system through apoptosis induction on immune cells, thereby conferring fungal survival within the host [57],[58]. Hence, the characteristic cytopenia observed in CM, further compounds immunosuppression among the patients [59]. GXM is antiphagocytic and resists respiratory burst stress [22],[57]. Excessive production of GXM was directly related to decreasing survival of severe combined immunodeficiency (SCID) mice models infected with C. neoformans [60].

    A clinical study reported a direct relationship between C. neoformans that produced large capsules in vivo with raised intracranial pressure (ICP), inadequate CNS immune response, reduced cryptococcal clearance, dwindle expression of pro-inflammatory cytokines, and low leukocyte count among patients with CM [61]. The GXM possesses several immunomodulatory effects during cryptococcal infections, which include increasing Fas Ligand expression, subduing LPS-induced signaling, blocking T cell proliferation, immune cells apoptosis induction, and interference of antigen presentation by professional antigen-presenting cells (pAPCs), cumulatively favoring cryptococcal survival [62]. Furthermore, GXM disrupts the healthy balance of pro-inflammatory to anti-inflammatory cytokines, facilitating anti-inflammatory cytokines expression in the presence of Cryptococcus pathogen [63].

    GXM is an essential component of Cryptococcus that is required for survival, pathogenicity, and immune evasion. GXM is continuously shed in the serum and CSF during infection. The body fluid level of GXM correlates directly with a fungal burden in CM [64]. As a formidable cryptococcal biomarker, scientists target GXM to diagnose cryptococcosis in the last four decades. Of note, low immunogenicity inherent to polysaccharide antigens seems to stand in the way of developing anti-GXM for immunoassays. So, Cryptococcal serological diagnostic immunoassays evolve in specificity from targeting the whole CNPS, narrowing it down to GXM [65]. Others developed polyclonal or monoclonal antibodies by coupling GXM to carrier proteins, to boost the polysaccharide's immune response [34],[66],[67].

    Scientists describe a monoclonal antibody of IgG1 subclass to CNPS after immunizing mice with extracted soluble capsular polysaccharide of C. neoformans serotype A, obtained through ultrafiltration of grown culture. The discovered IgG1 cross-reacts mildly with other C. neoformans serotypes and Trichosporon beigelii. The anti-CNPS also successfully recognized the GXM of C. neoformans [68]. Devi and colleagues discover a superior monoclonal antibody response secondary to immunization of laboratory mice with a vaccine they synthesize from conjugation of +GXM to tetanus toxoid (TT) [28]. The same study's highest antibody response for both IgG and IgM in the mice was recorded when the initial immunization dose was followed by monophosphoryl lipid A (MPL) adjuvant injection on the second day [28]. In another study, the GXM was precipitated with cetyltrimethylammonium bromide (CTBA) and conjugated to TT using adipic acid as a spacer; the vaccine formed was then used for intraperitoneal immunization of Balb/c mice yielding monoclonal antibodies specific for all the four serotypes of C. neoformans [69].

    The emergence of widespread antibiotic resistance has pushed the scientific world to search for alternative therapies for infectious diseases, which are affordable and effective, especially those that stimulate appropriate host responses [70]. The well-studied anti-GXM (18B7) was shown to hydrolyze the fungal oligosaccharide and increase GXM release from Cryptococcus [71], a therapeutic potential that needed to be elaborated. Interestingly, another study uncovered a mechanism by which GXM activates macrophages through TLR4 signaling to secrete pro-inflammatory cytokines providing protective immunity to the host [37].

    Researchers reported promising preliminary studies of CD8+T cells expressing GXM-specific chimeric antigen receptor (GXM-CAR) as a form of immunotherapeutic modality. Treatment of immunocompromised NSG mice infected with C. neoformans yielded a significantly lower fungal burden than the control [72]. The CD40L expressed by activated T cells interacts with CD40 molecules expressed by myeloid cells for successful co-stimulation that is tightly linked to NF-κβ activation: proinflammatory response. Hence, CD40L/CD40 immune checkpoint provides an interesting target for immunotherapy. [73]. The mutation of CD40L in humans and CD40L(−/−) among mice both leads to disseminated cryptococcosis, emphasizing the critical role of Th1 cells in cryptococcal immunity. Again combination therapy of CD40L and IL-2 in systemic murine cryptococcosis reveals extended survival of the study mice compared to the control [46]. Recombinant IFNγ therapy in a clinical study yielded a promising result, however, limited by side effects, and as such advocated as an adjuvant for systemic cryptococcal therapy [46],[74].

    Radiolabeled anti-GXM therapy shows promising results [75]. Radioimmunotherapy was conducted using 18B7 mAb labeled with alpha emitter bismuth-213 (213Bi) or a powerful beta emitter rhenium-188 (188Re) to target GXM in a mice model of C. neoformans infection. 18B7-213Bi and 18B7-188Re radiolabeled mAbs extend mice survival by 60% and 40% on the eleventh week of therapy, respectively [76]. Scytovirin, a lectin from cyanobacteria, has activity against mannose molecules found on microbial agents; has antiviral and antifungal effects. Scytovirin proves promising in inhibiting C. neoformans with lesser effect than C. gattii [77].

    A glycopeptide isolated from sheep red blood cells (SRBCs) membrane termed T11TS was a CD2 receptor agonist, promoting T cell proliferation, activation, and survival [78]. In a rat model of CM, T11TS therapy facilitates fungal recognition through upregulated TLRs expression, increases antigen presentation by microglial cells, and promotes proinflammatory cytokines secretion, in sum promoting fungal immunity [79]. T11TS immunotherapy in a rodent model of C. neoformans infection was shown to induce nuclear retention of NFAT, a mechanism that causes upregulation of IL-2 expression [80]. Similarly, T11TS counters molecular mechanisms of C. neoformans immune evasion and facilitates immune recognition and effector immune response against the microbe, which cumulatively, favor C. neoformans clearance from the study rats [81]. Figure 5 summarizes various immunotherapeutics that target GXM discussed above.

    Figure 5.  GXM as an immunotherapy target. GXM, the major polysaccharide in the C. neoformans capsule is a molecular target for immunotherapy. From the top, T11TS treatment upregulates TLR on microglial cells and serves as a growth factor on helper T cells facilitating a proinflammatory response to C. neoformans. A monoclonal antibody to GXM (18B7) facilitates the release of GXM from the C. neoformans capsule through hydrolysis of oligosaccharide constituent of GXM, on the left. The GXM as a polysaccharide activates macrophages by signaling through TLR4 that yields a proinflammatory response to target the C. neoformans, on the right. Cytotoxic T cells expressing GXMR-CAR specifically improve GXM recognition on C. neoformans. On the bottom of the diagram Radiolabeled anti-GXM: 213Bi, beta emitter, and 188Re, an alpha emitter both target GXM delivering the cytotoxic payload to the C. neoformans. GXM: glucuronoxylomannan; GXMR-CAR: GXM-specific chimeric antigen receptor; 213Bi: bismuth-213; 188Re: rhenium-188; T11TS: T11 target structure; TLR: toll-like receptor.

    Researchers respond exponentially to the dire need to develop a vaccine for cryptococcosis, considering the morbidity and mortality associated with the systemic disease, which worsened with the advent of HIV infection. The cryptococcal vaccine is intended to protect individuals prone to severe cryptococcosis due to genetic predisposition or compromised immune system. Initial cryptococcal vaccines explore the immunization modality from the first principle perspective: using live attenuated and heat-killed Cryptococcus protocols [49]. A preclinical immunization study was conducted to compare the immune response between heat-killed C. neoformans and culture filtrate of C. neoformans. The culture filtrate immunization gives way to the robust immune system that is CD4+ T cell-dependent, associated with copious secretion of IFN-γ and IL-2, optimal fungal clearance, and increased mice survival. However, no anti-GXM was discovered in the studied mice sera. Of note, the heat-killed vaccine displayed an indifferent immune response [82]. Th17 polarization is a prerequisite to vaccine activity against cryptococcal pathogens [83]. Further, the lack of IFNγ, IL-17, and TNFα cytokines production in the study mice cumulatively lead to cryptococcal vaccine failure [34], which signifies the role of CD4+ T cells in cryptococcal immunity.

    GXM conjugated to tetanus toxoid (GXM-TT) used to immunize mice in the presence of monophosphoryl lipid A (MPL) as an adjuvant, recorded promising protection against systemic cryptococcosis through antibodies induction [84]. Researchers studied GXM mimotope's immunogenicity: P13 conjugated to either TT (P13-TT) or bovine serum albumin (P13-BSA) adjuvanted with aluminum hydroxide. Both vaccines yielded IgM and IgG to GXM and P13 that confer mice survival with decreased serum levels of GXM than controls [85]. Another study of P13 on chronically infected mice prolongs the study animals' survival through immunomodulation by IL-10 expression. However, the protective immune response recorded depends on the conjugate protein type, mice strain, and infection route [86].

    Scientists cultured bone marrow DCs with a heat-killed clinical isolate of C. gattii species complex (C. bacillisporus:VGIII/AFLP5) and transferred them to mice, which were later infected with the clinical isolate. They reported prominent Th1 and Th17 cytokines expression that lowered the fungal load, decreased the lung GXM levels, improved the associated pathology, and increased mice survival [87]. Pulsed DC vaccines of C. gattii induce IL-17A secreting resident memory Th17 cells in the lung, which also potentiate the activity of neutrophils and giant immune cells in the study animals. The increased mice survival and the decreased fungal levels are deficient in IL-17A knock-out mice [88]. Other cryptococcal antigenic compounds (mannoprotein and MP98), synthetic cryptococcal proteins, GXM peptide mimetic, and fungal extract are being extensively studied to provide potent and safe vaccines [49] not covered in this review. Besides, with ongoing studies, it is hoped that there will be light at the end of the tunnel in the treatment and prevention of CM. Figure 6 below summarized the salient role played by GXM in immunity, pathogenicity, and vaccine development in cryptococcal infection.

    Figure 6.  GXM is the cornerstone polysaccharide on the C. neoformans cell wall, responsible for downregulating the immune system by evading phagocytes, thereby inducing monocyte anergy, neutropenia, and facilitating M2 macrophages phenotype that are anti-inflammatory. Yet, the development of immunotherapy and vaccine both target GXM for a better outcome. GXM: glucoronoloxylomannan; GXMR-CAR: GXM-specific chimeric antigen receptor; 213Bi: bismuth-213; 188Re: rhenium-188; TT: tetanus toxoid; P13-TT: P13 conjugated to TT; P13-BSA: P13 conjugated to bovine serum albumin; DC: dendritic cell.

    The global morbidity and mortality indices associated with CM, especially among HIV/AIDs patients, further complicated by widespread antifungal resistance, translate into the pressing need for immunotherapy and vaccine to curtail this severe infection's menace. Of note, C. neoformans evolves escape mechanisms to evade the immune system and persist in a strong or weak immune response. Both immunotherapy and vaccine development are still ongoing, and substantial literature targets GXM as a promising biomarker.



    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(2272) PDF downloads(148) Cited by(0)

Figures and Tables

Figures(6)  /  Tables(1)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog