Abstract
Cryptococcosis is caused by the fungal genus Cryptococcus. Cryptococcosis, predominantly meningoencephalitis, emerged with the HIV pandemic, primarily afflicting HIV-infected patients with profound T-cell deficiency. Where in use, combination antiretroviral therapy has markedly reduced the incidence of and risk for disease, but cryptococcosis continues to afflict those without access to therapy, particularly in sub-Saharan Africa and Asia. However, cryptococcosis also occurs in solid organ transplant recipients and patients with other immunodeficiencies as well as those with no known immunodeficiency. This article reviews innate and adaptive immune responses to C. neoformans, with an emphasis on recent studies on the role of B cells, natural IgM and Fc gamma receptor polymorphisms in resistance to cryptococcosis.
Keywords: acquired immunity, antibodies, B cells, cryptococcosis, Cryptococcus neoformans, Fc receptors, host response, innate immunity, macrophages, polymorphism, T cells
Cryptococcal disease, or cryptococcosis, is caused by a basidiomycetous yeast belonging to the genus Cryptococcus. This genus is unique among pathogenic fungi in having a polysaccharide capsule. Although the Cryptococcus genus contains many species, the majority of human infections are caused by two species: Cryptococcus neoformans and Cryptococcus gattii [1]. These species are further categorized based on the antigenic specificity of their capsular polysaccharide, with each variety constituting a separate serotype. C. neoformans is comprised of three varieties, C. neoformans var. grubii (serotype A), C. neoformans var. neoformans (serotype D) and a hybrid (serotype AD). C. gattii is comprised of two serotypes, B and C. C. neoformans predominantly causes disease in individuals with immune impairment, most commonly those with HIV/AIDS and CD4 T cells less than 50 cells/μl [2,3], whereas C. gattii predominantly causes disease in individuals without consistent or unifying immune defects [4]. However, the pathogenesis of C. gattii is an area of intense investigation as it is thought that as yet unidentified immune defects could enhance susceptibility to C. gattii-related cryptococcosis. This review will focus on the species C. neoformans.
Ecology
C. neoformans is an environmental microbe that is found in soil. Historically, the majority of human cases of cryptococcosis outside of Australia were attributed to C. neoformans var. grubii and var. neoformans [5]. However, a significant minority of cases in the world are due to C. gattii [6], which recent data show is an emerging cause of cryptococcosis in North America [7]. C. neoformans is a free-living microbe that does not require a host to reproduce or survive. As such, it is honed for survival, with its key virulence factors, including the polysaccharide capsule and cell wall associated melanin serving as protection from environmental assaults ranging from predators such as ameobae, UV irradiation and temperature [8–10]. Thus, C. neoformans is an accidental human pathogen. For a review of this topic, see reference [11].
Epidemiology
Cryptococcosis emerged as a global epidemic in patients with HIV/AIDS in the 1980s. Prior to the HIV/AIDS pandemic, there were fewer than 200 cases of cryptococcosis in the literature [12]. The occurrence of disease in HIV-infected individuals with profound CD4 T-cell deficiency highlights the central role of T-cell-mediated immunity in immunity to C. neoformans. However, loss of CD4 T cells alone is not sufficient for disease to occur. The use of fluconazole therapy in patients with profound CD4 T-cell deficiency and the introduction of combination antiretroviral therapy (cART) in the mid-1990s led to a marked decrease in cryptococcosis in those with access to cART [13,14]. However, cryptococcosis remains a catastrophe in under-resourced settings where cART is not available, such as sub-Saharan Africa and parts of Asia [15,16]. In 2009, Park et al. estimated the global burden of the disease to be close to 950,000 cases with approximately 625,000 deaths annually [16]. Cryptococcal meningitis also causes significant morbidity and mortality in the USA [17]. Compounding HIV-associated cryptococcosis-attributable morbidity and mortality is the immune reconstitution inflammatory syndrome (IRIS). There are two types of C. neoformans-associated IRIS; paradoxical IRIS, which is a recurrence of cryptococcosis that occurs after the initiation of cART, and unmasking IRIS, which occurs soon after the initiation of cART in patients with no prior diagnosis of cryptococcosis [18]. A recent study found that delaying the start of cART in patients presenting with HIV-associated cryptococcosis was associated with a significant reduction in mortality [19]. The occurrence of IRIS in HIV-associated cryptococcosis as well as in solid organ transplant recipients [20] reveals that excessive as well as insufficient inflammatory responses can result in disease, underscoring that the outcome of host–C. neoformans interaction is determined by the immune status of the patient. This fits with the paradigm put forth in the Damage response framework, whereby host damage and disease can stem from either insufficient or excessive immune responses [21].
Other forms of immunodeficiency, including that due to drugs used to prevent organ rejection also pose a risk for cryptococcosis [22], which occurs in approximately 2.8% of solid organ transplant recipients [23]. Some biologics, such as the TNF-α inhibitor adalimumab have also been linked to an increased risk for cryptococcosis [24]. Others at increased risk for cryptococcosis are pregnant women [25], and those with X-linked immunodeficiency [26], liver disease [27], idiopathic CD4 T-cell deficiency [28] and apparently immune competent individuals [29,30]. Presence of anti-GM-CSF autoantibodies was also associated with some cases of cryptococcal meningitis in immunocompetent individuals [31]. Hence, conditions associated with cryptococcosis range from profound CD4 T-cell deficiency to none that can be identified.
Pathogenesis of human Cryptococcosis
Infection with C. neoformans occurs by inhalation of desiccated yeast cells or spores from the environment in early childhood, most likely at the time of acquisition of other encapsulated microbes [32]. This event is not thought to be associated with clinical manifestations, although an association between childhood asthma and serological evidence of cryptococcal infection has been noted [33]. Based on serological surveys of immunocompetent and immunocompromised adults and children, cryptococcal infection is common [32,34,35]. However, disease is rare. In most, infection leads to a state of latency, most likely in the lungs, where the yeast resides in granulomata usually without evidence of clinical disease. However, in some, predominantly those with underlying immune impairment, the state of latency transitions to a state of disease as the fungal burden rises [11,36]. Although reactivation is a major cause of disease due to C. neoformans, particularly in those with immune impairment, disease can also follow primary acquisition [22]. Cryptococcosis is most commonly a disseminated disease characterized by meningoencephalitis and/or fungemia, but pneumonia and skin lesions can also occur.
HIV-associated cryptococcosis can be heralded by identification of cryptococcal antigen (CrAg) in the blood. Monitoring serum CrAg levels in HIV-infected individuals with CD4 T-cell levels less than 100 cells/μl in resource-limited settings in regions of high HIV/AIDS prevalence has been extremely successful and cost-effective in identifying high-risk patients who are candidates for fluconazole treatment in sub-Saharan Africa [37–40]. The success of this approach underscores the importance of rapid, point-of-care diagnostic tests [41]. CrAg screening and fluconazole treatment are a goal of the Global Action Fund for Fungal Infections (GAFFI [42]).
Cryptococcal virulence
The central determinant of virulence for C. neoformans is its polysaccharide capsule. There is abundant experimental evidence that the capsule is required for cryptococcal virulence in immunologically normal hosts [43,44]. The capsule is comprised primarily of glucuronoxylomannan (GXM), which can have many deleterious effects on the host response including inhibition of phagocytosis [45,46]. Fungal containment is crucial for host resistance to cryptococcosis. Thus, impairment of the function of macrophages or phagocytes by GXM or other cryptococcal virulence factors, such as melanin, enhances cryptococcal virulence. However, C. neoformans is also able to replicate intracellularly and can escape the intracellular state without being killed or killing the host cell [47,48]. The latter, which could contribute to intracellular or extracellular dissemination, is one of several mechanisms that have been implicated in how C. neoformans enters the bloodstream and invades the CNS. For a review of cryptococcal virulence, see reference [49].
Host response to C. neoformans
Fungal diseases in animals and humans are relatively rare. In fact, although there are more than 1.5 million fungal species, only 300 cause human disease [50]. One determinant of the latter is that many fungal species cannot survive at mammalian thermal temperatures [51]. Importantly, C. neoformans and other fungi that cause human mycotic disease are able to survive at mammalian temperatures. Nonetheless, most of these fungi are very rare causes of disease in immunologically intact individuals. This is exemplified by the fact that cryptococcosis was rare before the HIV/AIDS pandemic and frequent use of transplant drugs that induce immune suppression. Similarly, candidiasis was rare before the use of broad-spectrum antibiotics and intravenous catheters. Hence, the rise in fungal diseases in the last quarter of the 20th century paralleled the emergence of an expanded population of patients with immune impairment. The degree to which cryptococcosis and HIV-associated immune deficiency are intertwined is illustrated by a report in the 1950s of cryptococcosis in young people in sub-Saharan Africa, the area where the HIV/AIDS pandemic is thought to have begun [52].
The recognition that normal host defense is sufficient to confer resistance to cryptococcosis in most individuals led to intense study of the immune response to C. neoformans. This research led to a better understanding of factors that predispose to cryptococcosis and has informed strategies to devise new drugs, vaccines and immunotherapy. The immune response to C. neoformans is reviewed below. This is an enormous topic and we have tried to cite critical literature, but it is not possible to cite every study. With respect to our goal of reviewing immunity to C. neoformans, we have endeavored to provide an overview of the innate and acquired immune response, while focusing in more detail on new discoveries on the contribution of B cells, natural antibody and genetic polymorphisms to host defense against C. neoformans.
Innate immune & cellular responses to C. neoformans
The first line of defense against C. neoformans is the surface barrier(s) of the innate immune system (e.g., skin, nasal mucosa). In addition, human serum and saliva demonstrate anticryptococcal activity [53,54].
Complement
Complement is a vital component of the innate immune response and a key mediator of phagocytosis of C. neoformans [55,56]. Complement-deficient animals are more susceptible to C. neoformans infection than complement sufficient animals [57] and complement components can be depleted in patients with cryptococcal fungemia [58]. The cryptococcal capsule activates the complement system, mainly through the alternative pathway, leading to C3 deposition on the fungal capsule [56]. Species differences have been shown to influence the site of C3 deposition on the cryptococcal capsule [59]. It was also observed that specific anticapsular antibodies could promote C. neoformans activation of the classical complement pathway [59,60]. Also, mannose-binding lectin (MBL) can bind to the C. neoformans cell wall and activate complement via the lectin pathway [61].
Phagocytosis
Containment of C. neoformans by phagocytic cells, including macrophages, dendritic cells and neutrophils, is crucial for natural host defense [46,62–63]. Once the fungus is inhaled, it travels to the lungs where it encounters various phagocytic effector cells. Phagocytosis can be mediated by complement receptors as well as Fc receptors. Complement-mediated phagocytosis of C. neoformans occurs via recognition of complement-opsonized yeast by complement receptors (CRs) CR1, CR3 and CR4 [64]. Fc-γ receptors on macrophages/neutrophils/dendritic cells can bind and mediate phagocytosis of antibody- opsonized C. neoformans [65], although phagocytosis of nonopsonized C. neoformans can also occur [66]. Additionally, mannose receptors on macrophages and dendritic cells can bind cryptococcal mannoproteins and mediate phagocytosis of C. neoformans [67,68].
When C. neoformans reaches the brain, resident microglial cells act as the primary phagocytic cells. Microglial cells express Toll-like receptors (TLRs), which can identify pathogen-associated molecular patterns. TLR2, TLR4 and TLR9 have been shown to bind and interact with zymosan (yeast β-glucan), GXM and fungal DNA, respectively [69–71]. Redlich et al. demonstrated that stimulation of microglial cells by these TLR agonists enhanced C. neoformans phagocytosis [72].
Macrophages
Alveolar macrophages (AMs) internalize C. neoformans in the lungs where they can link innate and adaptive immunity, but C. neoformans can also survive and proliferate intracellularly [73]. In mice, depletion of AMs delays cryptococcal dissemination and improves survival, suggesting that AMs promote fungal growth and dissemination [74]. However, depletion is detrimental in rats [63], highlighting the importance of species differences in host–C. neoformans interaction. C. neoformans can also escape from host macrophages through nonlytic exocytosis [48,75]. It can also be taken up by and survive in amoebae [8], underscoring its ability to resist destruction by environmental predators. C. neoformans has been shown to be able to disseminate to the brain inside of monocytes, providing experimental support for a Trojan horse model to explain fungal invasion of CNS [76]. Nonetheless, monocytes and macrophages have a major role in controlling C. neoformans in the lungs [77]. Macrophages can promote Th1-like responses that induce fungal clearance, serve as APCs to T lymphocytes [78], and secrete cytokines that skew CD4 T cells toward Th1/ Th2 or Th17 pathways [79]. AMs can enhance or control C. neoformans pathogenesis depending on the immune status of the host [80]. The host response to C. neoformans in the lungs correlates with macrophage polarization [81], whereby M1 (classically activated) macrophages lead to Th1 responses (mainly IFN-γ dominated) and M2 (alternatively activated) macrophages lead to Th2 responses. M1 macrophages are more efficient fungicidal cells than M2 macrophages. Changes in the cytokine environment can influence macrophage polarization, with M1 macrophages enhancing host defense against C. neoformans [81].
Dendritic cells
Dendritic cells (DCs) phagocytose C. neoformans via complement or antibody-mediated opsonization, leading to fungal internalization and killing [68,82]. DCs also induce T-cell responses following stimulation of pattern recognition receptors (e.g., TLR4 and TLR9) and secrete cytokines that are crucial for immunity to C. neoformans, including IL-12 and IL-23 [83]. In vivo, DCs internalize C. neoformans in the lungs of mice [84], and their lysosomal components can mediate fungal killing [85].
Polymorphonuclear cells
Neutrophils can enhance granuloma formation, by containing and killing C. neoformans in the lungs by oxidative and nonoxidative mechanisms [86–88]. They also contain antimicrobial peptides (defensins) that are cytotoxic to C. neoformans [89]. However, early depletion of neutrophils in vivo was protective against C. neoformans in a murine pulmonary infection model in which there was less inflammatory damage [90]. On the other hand, in a systemic infection model, survival of neutrophil-depleted mice was comparable to that of neutrophil sufficient mice, underscoring that regulation of the inflammatory response in the lungs is crucial for protection [90].
Eosinophils were associated with a lack of protection against C. neoformans in pulmonary infection model in which their presence was associated with excessive lung inflammation [91]. C57BL/6 mice develop eosinophilic pneumonia in response to pulmonary cryptococcal infection [92] and eosinophilia has also been observed in human cryptococcosis [93].
Natural killer cells
Natural killer (NK) cells bind and inhibit C. neoformans growth in vitro [94] and induce fungal clearance in mice [95]. Human NK cells are able to kill C. neoformans [96] with direct killing being mediated by perforin [97,98]. A recent study showed that in HIV-infected patients, reduced expression of an NK-cell receptor that mediates direct fungal recognition, (NKp30), led to significantly reduced anticryptococcal cytotoxicity [99].
NK T cells
NK T cells (NKT cells) play an important role in the development of Th1 responses and host resistance to C. neoformans. In mice, NKT cells increased in the lungs after intratracheal infection with C. neoformans, and the chemokine MCP-1 was implicated in NKT-cell migration and accumulation [100]. Compared with NKT-cell-deficient mice, activation of NKT cells with a synthetic glycolipid (α-galactosylceramide) resulted in increased IFN-γ production and improved host defense [101]. Along with IFN-γ, NKT cells also secrete IL-4, suggesting that this subset regulates both Th1- and Th2-mediated immune responses [102].
Adaptive immunity
CD4 T cells
CD4 T cells are a crucial component of cell-mediated immunity to C. neoformans in mice [103,104], as they mediate fungal clearance [105,106] and confer protection upon adoptive transfer to naïve mice [103,107]. CD4 T cells also play a dominant role in recruiting macrophages and granulocytes to the lungs in pulmonary cryptococcal infection [108]. Cryptococcal mannoproteins stimulate a protective CD4 T-cell response to C. neoformans, with glycosylation of mannoproteins being essential for an optimal response [109]. However, the specific epitopes recognized by T cells have not been definitively identified.
In humans, CD4 T-cell deficiency is a major predisposing factor for cryptococcosis [110,111], whereby a CD4 T-cell count less than 100 cells/μl and detectable serum CrAg portend high risk for HIV-associated cryptococcosis [37,112]. Cryptococcosis has also been reported in idiopathic CD4 lymphocytopenia, which is characterized by reduced levels of CD4 T cells without evidence of HIV infection [28,113].
CD8 T cells
CD8 T cells also play an important role in the host immune response to C. neoformans [114]. CD8 T cells mediate killing of C. neoformans [115], whereby killing requires direct cell contact thought to be mediated by granulysin. In vivo depletion of murine CD8 T cells reduced survival in a lethal cryptococcal infection model [116]. CD8 T cells also limit growth and survival of C. neoformans in macrophages by means of IFN-γ production independent of CD4 T cells [117]. For a complete review on this topic, see reference [114].
Gamma delta (γδ) T cells
γδ T cells are known to regulate Th1-Th2 responses to C. neoformans. They secrete anti-inflammatory Th2 cytokines to balance exaggerated Th1 response caused by NKT cells. Depletion of γδ T cells resulted in increased IFN-γ synthesis and promoted cryptococcal clearance via Th1-mediated responses in lungs of mice [118]. Of note, increased production of IL-17A by γδ T cells was observed in neutrophil- depleted mice during pulmonary cryptococcal infection, suggesting that γδ T cells can induce protective response to C. neoformans in the absence of traditional adaptive immune response [119].
T-cell-derived cytokines
Th1-type CD4 T cells orchestrate host immunity to C. neoformans [103]. Th1-type responses are characterized by production of IL-2, IL-12, IFN-γ and TNF-α. IL-12, IFN-γ and TNF-α, protect against cryptococcosis in experimental models [120,121]. C. neoformans also induces secretion of other pro-inflammatory cytokines, including Type-I IFN (IFN-I), IL-1β and IL-6 from innate immune cells [122]. IL-1β and IL-6 induce the development of T-helper Th17 cells in the presence of IL-23. IL-17 and IL-22 are the major cytokines secreted by Th17 cells. While Th17 immunity was required for vaccine-mediated protection against C. neoformans in mice [123], Th17-mediated responses were not required to protect naïve mice [124]. Of note, IL-17A produced by neutrophils rather than T cells, was implicated in optimal vaccine-mediated protection against C. neoformans in mice [125]. TNF-α production is required for the development of protective T-cell immunity to C. neoformans in mice [120]. TNF-α is crucial for induction of IL-12 and IFN-γ in the lungs following C. neoformans infection, which then promotes Th1-cell-mediated immunity [121].
Analysis of C. neoformans-specific CD4 T-cell responses in patients with HIV-associated cryptococcal meningitis revealed that the presence of an IFN-γ/TNF-α CD4 T-cell response correlated with survival [126]. In other studies, fungal clearance was positively associated with cerebrospinal fluid (CSF) IFN-γ levels, which were in turn positively correlated with the CD4 T-cell count [127,128]. The latter provided the rationale for IFN-γ as adjunctive therapy for HIV-associated cryptococcosis. The addition of a short course of IFN-γ to standard treatment increased the rate of cryptococcal clearance from the cerebrospinal fluid [129]. The importance of IFN-γ in resistance to cryptococcosis has been demonstrated experimentally with a C. neoformans- based vaccine that produces IFN-γ [130]. Thus, adjunctive IFN-γ and vaccines that induce tissue-specific IFN-γ production hold promise as immunotherapeutic interventions for cryptococcosis.
The role of B cells in immunity to C. neoformans
Numerous studies have now shown that B cells play a crucial role in protection against experimental cryptococcosis [131–133]. Prior to these reports, very few studies directly examined the role of B cells in host defense against C. neoformans. An early study utilizing an intravenous infection model reported no difference in C. neoformans lethality in B-cell-depleted and B-cell-sufficient mice [134]. However, another study linked B cells with resistance to C. neoformans in SCID mice [135], whereby SCID recipients of T cells from B-cell-deficient mice failed to express the adoptive immunity seen in recipients of T cells from B-cell-sufficient mice. Of note, B cells were the predominant cell type in the lungs of C. neoformans-infected A/JCr mice [136] and cryptococcal infection was more lethal and associated with more pulmonary immunopathology and inflammation in B-cell-deficient (uMT mice) than B-cell-sufficient mice [137], linking B cells to control of lung inflammation.
Mature B cells are subdivided into B-1 and B-2 cells. B-1 cells can be further classified into two populations based on expression of CD5, CD5+ B-1a cells and CD5− B-1b cells, whereas conventional B-2 cells are composed of follicular and marginal zone B-cell subsets. B-1 cells enhance resistance to and prevent dissemination of C. neoformans in several different mouse models. X-linked immunodeficient (XID) mice, which lack B-1 cells and natural IgM, were more susceptible to intravenous infection with C. neoformans than CBA/Ca control mice [138]. In pulmonary infection models, B-1a cells were associated with fungal containment during the early immune response [132], and XID mice, which lack B-1a cells, are less able to contain C. neoformans in the lungs than control mice and develop more fungal dissemination to the brain [133]. This phenotype is associated with a virtual absence of serum IgM (see below), reduced yeast uptake by macrophages, an aberrant tissue inflammatory response and enlargement of yeast cells in the lungs [133]. These studies implicate B-1a cells and IgM in cryptococcosis, with the caveat that XID mice have other defects including T-cell and NK-cell immunodeficiency. In C. neoformans infected C57BL/6 mice, CD5+B-1a cells exhibited more binding to C. neoformans than B-1b and B-2 cells and directly mediated lung and brain fungal clearance during early pulmonary cryptococcal infection [132]. Reconstitution of B-1 cells in B-1-cell-depleted mice increased AM phagocytosis of C. neoformans and reduced lung and brain fungal burdens. Several reports have demonstrated that B-1-cell-derived mono-nuclear phagocytes have fungicidal activity against C. neoformans [139,140].
In humans, IgM memory B cells are considered the main homolog of mouse CD5+ B-1 cells. These cells, which are identified by their expression of CD27 and IgM, produce naturally occurring IgM that binds conserved microbial determinants and carbohydrates [141]. In one study, peripheral blood IgM memory B-cell levels were lower in HIV-infected individuals with a history of cryptococcosis than in those with no history of cryptococcosis and were predictive of cryptococcal disease status [142]. In this study, a predisease cohort had CD4 T-cell levels greater than 400 cells/μl, suggesting that IgM memory B-cell levels could hold promise as a biomarker of risk for human cryptococcosis.
Natural antibodies to C. neoformans
IgM memory B cells produce ‘naturally occurring’ IgM that binds conserved microbial determinants [143]. Because natural IgM is produced in the absence of antigen stimulation, it is a part of the innate immune system and considered to provide ready-made pathogen defense. In mice, IgM deficiency was associated with increased susceptibility to pulmonary C. neoformans infection and reduced AM phagocytosis of C. neoformans, which was restored by reconstitution with natural mouse (nonimmune) IgM [144]. B-1 cells from C. neoformans-infected mice secreted C. neoformans-binding IgM and depletion of these cells resulted in reduced AM phagocytosis and increased fungal dissemination to the brain [132]. These findings support the hypothesis that B-1 cells enhance innate antifungal immunity via natural IgM, which promotes fungal containment in the lungs [132,133]. This is consistent with a report that an antibody to laminarin, a fungal cell wall determinant recognized by natural IgM, bound to and was protective against C. neoformans in mice [145,146].
The natural antibody response to C. neoformans in humans has been studied in detail. IgM and IgG that bind cryptococcal polysaccharides (capsule and cell wall) are present in normal human serum, although they are generally not opsonic for macrophage phagocytosis of C. neoformans [147,148]. Serum GXM-binding antibodies are virtually ubiquitous in adults and can be detected early in childhood [32,34–35,149]. Like other capsular polysaccharides, IgG2 is the predominant GXM-binding IgG subclass. Notably, IgG2 is decreased in individuals with HIV/AIDS [34,35]. HIV-infected individuals have higher levels of GXM-IgG1 and lower levels of IgG2 than HIV-uninfected individuals [94].
A number of studies have shown that levels of GXM-binding IgM are lower in HIV-infected than HIV-uninfected individuals [34,142,150–151], including one in which levels were lower in HIV-infected individuals who developed cryptococcosis than those who did not [142]. GXM-binding IgM levels were also lower in HIV-uninfected solid organ transplant recipients who developed cryptococcosis post-transplant than those who did not [152]. Given that IgM memory B cells are a major source of serum IgM, reduced levels of GXM-binding IgM could stem from a loss of IgM-producing B cells. Consistent with this hypothesis, IgM memory B cells are depleted in HIV/AIDS [153,154]. In fact, loss of these cells begins early in HIV infection before severe CD4 T-cell deficiency is manifest and unlike other B-cell subsets, IgM memory B cells are not fully reconstituted by cART [155]. The effect of drugs used in solid organ transplant recipients on B-cell and antibody levels has not been studied, but mycophenolate, prednisone and cyclophosphamide each depleted B-1a cells in mice [156]. Whether or not there is a loss of IgM memory B cells in solid organ transplant recipients and apparently normal individuals with cryptococcosis requires investigation.
Adaptive antibody response to C. neoformans
The acquired antibody response to various cryptococcal antigens, namely, GXM capsule, cell wall polysaccharides and cryptococcal proteins has been studied extensively. The antibody response to GXM is characteristic of antibody responses to other encapsulated microbes, such as Streptococcus pneumoniae. As T-independent type 2 antigens, capsular polysaccharides elicit restricted antibody responses notable for the use of a limited number of antibody variable region genes and an absence of class switching and recall or memory responses. GXM is considered a rational vaccine candidate for cryptococcosis, but similar to bacterial capsular polysaccharide-based vaccines, it was necessary to convert it to a T-dependent antigen by conjugating it to a protein carrier to enhance its immunogenicity. An investigational GXM-tetanus toxoid (GXM-TT) [157] vaccine was promising in preclinical trials [158], but was not developed further due to nonscientific reasons [159]. Extensive work with GXM-TT in mice led to a paradigm shift in our understanding of antibody immunity to capsular polysaccharides stemming from the observations that GXM elicited protective as well as a nonprotective antibodies and that antibody action was a function of idiotype, isotype and specificity. This is reviewed in [160].
When the antibody response to GXM-TT was examined in human volunteers the GXM response was predominantly IgG2, which was also the main mediator of C. neoformans phagocytosis by human mononuclear cells [161]. A human monoclonal antibody derived from a GXM-TT vaccinated donor protected mice against experimental cryptococcosis [162] as did human monoclonal IgMs produced from GXM-diphtheria toxoid (GXM-DT) vaccinated Xeno-mouse™ mice [163] and a peptide mimotope- vaccine-derived from a protective human monoclonal antibody [164].
Studies of GXM-TT (and DT) elicited mouse and human monoclonal antibodies revealed that they are derived from a restricted B-cell repertoire [165,166]. The immunoglobulin VH3 family genes encoded the human anti-GXM antibodies, and this family shares structural homologies with the VH5 (7183) family genes used for encoding mouse anti-GXM antibodies [163,167]. VH3-expressing B cells dominate the human response to bacterial polysaccharide antigens and are depleted in HIV infection. Thus, it has been hypothesized that a decrease in VH3-encoded antibodies could contribute to susceptibility for cryptococcosis [168].
A mouse monoclonal GXM IgG1 showed promise as adjunctive therapy for cryptococcosis in HIV-infected patients in a Phase I trial [169,170], but was not advanced further due to a lack of resources. Nonetheless, there is extensive preclinical data on this antibody, including its efficacy as radioimmunotherapy [171].
Genetic susceptibility to cryptococcal disease
Given that not all HIV-infected individuals with CD4 T-cell deficiency develop cryptococcosis and cryptococcosis occurs in HIV-uninfected patients without CD4 T-cell deficiency, other risk factors are under investigation. The idea that genetic factors could impact susceptibility seems plausible because polymorphisms can affect the expression of a multitude of host response genes. However, to date, only a few studies have addressed this possibility [172].
MBL polymorphisms
MBL is a circulating C-type lectin that plays an important role in innate immunity as a first line of pathogen defense. It selectively recognizes the pattern of glycans displayed on certain microbial surfaces, leading to opsonization and subsequent activation of the lectin pathway of the complement system. Complement activation results in further opsonization and induction of inflammatory reactions. Human MBL is encoded by a single gene (MBL2), encoding six common single-nucleotide polymorphisms (SNPs). These polymorphisms have a major effect on serum MBL levels. MBL deficiency caused by polymorphisms in the MBL2 gene was associated with increased susceptibility to cryptococcosis in HIV-uninfected Chinese patients [173].
Fc-γ receptor polymorphisms
Fc-γ receptors (FCGR) are present on certain immune cells (macrophages, monocytes, neutrophils, natural killer cells, B cells and mast cells), and bind IgGs connecting the humoral response to cellular effector mechanisms. FCGRs contribute to regulation of a multitude of immune and inflammatory responses [174]. FCGR polymorphisms are associated with certain autoimmune diseases as well as increased susceptibility to certain infections [175,176], including cryptococcosis [94,177,178].
FCGR2A 131H/R polymorphism involves the substitution of arginine (R) to histidine (H) at the 131 amino acid position in the ligand binding domain of the receptor. FCGR2A is unique in its ability to bind human IgG2 and is crucial for clearance of encapsulated pathogens. As noted above, IgG2 is the main subclass of antibodies to microbial polysaccharides [175]. The FCGR2A 131H allele displays higher IgG (including IgG2) binding than the 131R allele. FCGR2A 131H-expressing effector cells have a higher capacity for phagocytosis [179,180], as exemplified by a study showing that monocytes from FCGR2A 131HH donors internalized immune complexes more efficiently than monocytes from 131RR donors [181]. Meletiadis et al. reported an association of FCGR2A 131R with cryptococcosis in HIV-uninfected patients, including solid organ transplant recipients [177]. Although this study did not examine IgG2 levels, it speculated that increased susceptibility to cryptococcosis in individuals with FCGR2A 131RR could be explained by inefficient phagocytosis of IgG2-opsonized C. neoformans [177,182]. However, FCGR2A 131H/R polymorphism was not associated with risk for cryptococcosis in an HIV-infected cohort that had lower levels of IgG2 than HIV-uninfected participants [94]. Given previous data showing that IgG2 is the main isotype responsible for phagocytosis of C. neoformans in immune sera [161], it is possible that FCGR2A 131R confers risk in those with normal levels of IgG2 [94]. This requires further study. Of note, FCGR2A 131R was not associated with cryptococcosis in an HIV-uninfected Chinese cohort [178].
FCGR3A 158F/V polymorphism results in substitution of phenylalanine (F) for valine (V) at amino acid position 158. Compared with FF homozygous donors, FCGR3A expressed on NK cells and monocytes in VV homozygotes bound more IgG1 and IgG3 despite identical levels of receptor expression [183]. The FCGR3A 158V polymorphism was first reported to be associated with risk for cryptococcosis among HIV-uninfected patients by Meletiadis et al. [177]. Recently, an association between FCGR3A 158V and risk for cryptococcosis was also reported in HIV-infected individuals [94]. This study also showed that the FCGR3A 158V allele exhibited more binding of C. neoformans–Ig complexes and when expressed by NK cells, it induced more antibody-dependent cellular cytotoxicity (ADCC) against C. neoformans-infected monocytes than FCGR3A 158F-expressing cells [94]. These findings provide a possible mechanistic explanation for how the 158V polymorphism could enhance C. neoformans virulence. It could increase phagocyte cargo (due to increased binding and uptake of C. neoformans–immune complexes), thereby increasing fungal burden, and/or it could increase immune activation via ADCC, thereby leading to host damage and fungal dissemination [94,172]. Again of note, FCGR3A 158F/V polymorphism was not associated with cryptococcosis in the aforementioned Chinese cohort [178]. Given that the cohorts in which this polymorphism was associated with cryptococcosis (Meletiadis et al. [177] and Rohatgi et al. [94]) were both predominantly Caucasian, further studies are needed to determine whether it affects risk in other racial groups.
FCGR2B is the only inhibitory receptor to suppress downstream events such as cellular proliferation, phagocytosis and inflammatory cytokine release. In addition to its expression on B cells, FCGR2B is expressed on macrophages, neutrophils and mast cells, but not on T or NK cells. FCGR2B receptors contain a polymorphism at position 232, in which an isoleucine (I) is replaced by a threonine (T). Receptors encoded by FCGR2B 232T lack inhibitory activity, as they are unable to interact with activating receptors. FCGRR2B 232T/T genotype was associated with increased risk for SLE and protection against malaria [184]. In a case-control genetic association study in the aforementioned study Chinese cohort, FCGR2B 232I/I was associated with cryptococcal meningitis [178].
Although more work and studies in larger, more diverse cohorts must be done, the discovery of SNPs of FCGRs that are associated with risk for cryptococcosis suggests that these polymorphisms, which are likely to contribute to dysregulation of inflammatory responses to C. neoformans, could serve as potential biomarkers of risk for disease and enable the identification of those who might benefit from prophylaxis.
Conclusion & future perspective
C. neoformans is ubiquitous in the environment and exposure is very common, yet clinically apparent disease is rare, except in patients with immune impairment. HIV/AIDS is the most common predisposing condition for disseminated cryptococcal disease, but disease also occurs in patients with other types of immunodeficiency, including that due to the immunosuppressive drugs used in solid organ transplantation. However, some patients with cryptococcosis have no known underlying condition.
Many different innate and acquired immune constituents and functions, including B cells, T cells, macrophages, cytokines and phagocytosis contribute to host defense against C. neoformans, but the loss or absence of a single constituent, (e.g., CD4 T cells), appears to be insufficient to cause human cryptococcosis. Recent studies reveal associations between FCGR polymorphisms and HIV-associated and HIV-unassociated cryptococcosis and roles for B cells, natural and C. neoformans-specific antibodies in resistance to disease. Current data suggest that B- and T-cell deficiency, absence of natural IgM, reductions in tissue-specific IFN-γ and FCGR polymorphisms could all promote C. neoformans growth and dissemination. However, more studies are needed to validate this hypothesis.
New insights into susceptibility and resistance to cryptococcosis are likely to inform the development of novel agents to treat and prevent disease. However, a major challenge over the next 5–10 years will be to ensure the availability of cART and antifungal agents in all areas of the world, particularly those where HIV/ AIDS remains epidemic, such as sub-Saharan Africa and parts of Asia. Fluconazole therapy and initiation of cART can reduce the burden of HIV-associated cryptococcosis, but access to these agents and point-of-care rapid diagnostics, which are priorities for GAFFI [42], are needed to conquer cryptococcosis in under-resourced settings. Triumph over cryptococcosis in the resourced world will require clinical biomarkers that can identify high-risk patients who would benefit from antifungal prophylaxis, such as those with acquired and primary immunodeficiencies. Although levels of IgM memory B cells, GXM-IgM and FCGR polymorphisms have been associated with risk for disease in some studies, much work remains to be done to identify and validate biomarkers as clinical tools.
Given that the immune status of the patient is the main determinant of the outcome of host–C. neoformans interaction, therapies that augment host immunity are logical, particularly for patients with known defects. Immunotherapy, such as passive antibody-based therapy and prophylaxis, vaccines and selected mediators such as IFN-γ, are potential modalities for treating and preventing cryptococcosis. Development of a vaccine is a high priority for the cryptococcal field. Given that C. neoformans is ubiquitous in the environment, yet disease is rare, the ideal vaccine for C. neoformans would prevent reactivation as well as acute disease. However, because cryptococcosis may occur in the setting of either insufficient or excessive immune responses [21], more than one type of vaccine might be required. The vast amount of knowledge about the host response to C. neoformans gained over the past two decades is poised to accelerate vaccine development. In this regard, a number of vaccine candidates and platforms have been developed over the past two decades [185]. Although the most promising vaccines developed so far are GXM-protein conjugates and whole cell vaccines that enhance IFN-γ production, vaccines that bolster innate immune responses and leverage the immunogenicity of cell wall determinants are in development [186]. Over the past two and a half decades, immense progress has been made in our understanding of immunity to C. neoformans. However, the challenge to the field will be to obtain sufficient resources to translate this knowledge to identify biomarkers of disease risk and treat and prevent cryptococcosis with development and clinical trials of therapies and vaccines that bolster immunity to C. neoformans.
Executive summary.
Background
Cryptococcosis occurs predominantly in immunocompromised patients, most commonly those with HIV/AIDS.
Cryptococcus neoformans (variety neoformans and variety grubii) and Cryptococcus gattii cause majority of disease in humans.
Ecology
Cryptococcal species are ubiquitous environmental microbes, differing in geographic distribution.
Epidemiology
Incidence of HIV-associated cryptococcosis has been reduced by combination antiretroviral therapy (cART) but remains a threat to those not on cART, especially in underresourced areas.
Cryptococcosis also occurs in solid organ transplant recipients, and in people with no apparent immune defects.
Pathogenesis
C. neoformans is acquired by inhalation followed by a state of latency in the lungs.
Cryptococcosis occurs during latency breakdown in the setting of immune deficiency.
HIV-associated cryptococcosis is heralded by CD4 T-cell counts less than 100 cells/μl and detectable serum cryptococcal antigen (CrAg).
Cryptococcal virulence
The central virulence factor of C. neoformans is its polysaccharide capsule.
Other virulence determinants include capacity to grow at mammalian temperatures as well as intracellular replication.
Host response to C. neoformans
Innate and acquired immune mechanisms contribute to resistance to cryptococcosis.
CD4 T cells and cytokines enhance phagocytosis and cryptococcal containment.
CD8 T cells enhance cryptococcal killing.
Macrophage phagocytosis promotes cryptococcal containment, but C. neoformans can replicate in macrophages.
B cells enhance resistance to C. neoformans in experimental models.
Natural IgM promotes containment of C. neoformans in murine lungs, preventing dissemination to brain.
HIV-associated cryptococcosis was linked to reduced levels of IgM memory B cells and lower levels of IgM.
Genetic susceptibility to cryptococcosis
MBL and Fc-γ receptor polymorphisms have been associated with cryptococcosis.
Genetic factors influence susceptibility and resistance to C. neoformans.
Conclusion & future perspective
Victory against cryptococcosis will require access to antifungal drugs, cART and administration of fluconazole based on pre-emptive CrAg screening of patients with CD4 T-cell count less than 100 cells/μl.
Clinical biomarkers are needed to assess risk for cryptococcosis in high-risk populations and to enable antifungal prophylaxis and therapy.
Novel adjunctive immunotherapies, (monoclonal antibodies and/or IFN-γ) and immunomodulators should be explored.
Need for vaccine development which can prevent acute as well as reactivated disease.
Acknowledgments
The authors thank Wendy Szymczak and Arturo Casadevall for reviewing this manuscript.
Footnotes
For reprint orders, please contact: reprints@futuremedicine.com
Financial & competing interests disclosure
L Pirofski is supported by NIH grant R01AI097096. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as:
• of interest;
•• of considerable interest
- 1.Kwon-Chung KJ, Fraser JA, Doering TL, et al. Cryptococcus neoformans and Cryptococcus gattii, the etiologic agents of Cryptococcosis. Cold Spring Harb Perspect Med. 2014;4(7):1–27. doi: 10.1101/cshperspect.a019760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Andama AO, Den Boon S, Meya D, et al. Prevalence and outcomes of cryptococcal antigenemia in HIV-seropositive patients hospitalized for suspected tuberculosis in Uganda. J Acquir Immune Defic Syndr. 2013;63(2):189–194. doi: 10.1097/QAI.0b013e3182926f95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jarvis JN, Harrison TS, Govender N, et al. Routine cryptococcal antigen screening for HIV-infected patients with low CD4+ T-lymphocyte counts – time to implement in South Africa? S Afr Med J. 2011;101(4):232–234. doi: 10.7196/samj.4752. [DOI] [PubMed] [Google Scholar]
- 4.Sorrell TC. Cryptococcus neoformans variety gattii. Med Mycol. 2001;39(2):155–168. [PubMed] [Google Scholar]
- 5.Lin X, Heitman J. The biology of the Cryptococcus neoformans species complex. Annu Rev Microbiol. 2006;60:69–105. doi: 10.1146/annurev.micro.60.080805.142102. [DOI] [PubMed] [Google Scholar]
- 6.Litvintseva AP, Thakur R, Reller LB, Mitchell TG. Prevalence of clinical isolates of Cryptococcus gattii serotype C among patients with AIDS in sub-Saharan Africa. J Infect Dis. 2005;192(5):888–892. doi: 10.1086/432486. [DOI] [PubMed] [Google Scholar]
- 7.Byrnes EJ, 3rd, Li W, Lewit Y, et al. Emergence and pathogenicity of highly virulent Cryptococcus gattii genotypes in the northwest United States. PLoS Pathog. 2010;6(4):e1000850. doi: 10.1371/journal.ppat.1000850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8•.Steenbergen JN, Shuman HA, Casadevall A. Cryptococcus neoformans interactions with amoebae suggest an explanation for its virulence and intracellular pathogenic strategy in macrophages. Proc Natl Acad Sci USA. 2001;98(26):15245–15250. doi: 10.1073/pnas.261418798. Provides a mechanistic explanation for the ability of Cryptococcus neoformans to survive intracellularly by establishing parallels between amoeba and mammalian macrophages. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wang Y, Casadevall A. Decreased susceptibility of melanized Cryptococcus neoformans to UV light. Appl Environ Microbiol. 1994;60(10):3864–3866. doi: 10.1128/aem.60.10.3864-3866.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Rosas AL, Casadevall A. Melanization affects susceptibility of Cryptococcus neoformans to heat and cold. FEMS Microbiol Lett. 1997;153(2):265–272. doi: 10.1111/j.1574-6968.1997.tb12584.x. [DOI] [PubMed] [Google Scholar]
- 11.Casadevall A, Pirofski LA. Accidental virulence, cryptic pathogenesis, martians, lost hosts, and the pathogenicity of environmental microbes. Eukaryot Cell. 2007;6(12):2169–2174. doi: 10.1128/EC.00308-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hajjeh RA, Brandt ME, Pinner RW. Emergence of cryptococcal disease: epidemiologic perspectives 100 years after its discovery. Epidemiol Rev. 1995;17(2):303–320. doi: 10.1093/oxfordjournals.epirev.a036195. [DOI] [PubMed] [Google Scholar]
- 13.Bicanic T, Harrison TS. Cryptococcal meningitis. Br Med Bull. 2004;72:99–118. doi: 10.1093/bmb/ldh043. [DOI] [PubMed] [Google Scholar]
- 14.Aberg JA, Price RW, Heeren DM, Bredt B. A pilot study of the discontinuation of antifungal therapy for disseminated cryptococcal disease in patients with acquired immunodeficiency syndrome, following immunologic response to antiretroviral therapy. J Infect Dis. 2002;185(8):1179–1182. doi: 10.1086/339680. [DOI] [PubMed] [Google Scholar]
- 15.Jackson A, Van Der Horst C. New insights in the prevention, diagnosis, and treatment of cryptococcal meningitis. Curr HIV/AIDS Rep. 2012;9(3):267–277. doi: 10.1007/s11904-012-0127-7. [DOI] [PubMed] [Google Scholar]
- 16••.Park BJ, Wannemuehler KA, Marston BJ, Govender N, Pappas PG, Chiller TM. Estimation of the current global burden of cryptococcal meningitis among persons living with HIV/AIDS. AIDS. 2009;23(4):525–530. doi: 10.1097/QAD.0b013e328322ffac. Provided the first estimate of the global prevalence of cryptococcal disease and showed that disease was associated with a higher rate of mortality than tuberculosis in sub-Saharan Africa. [DOI] [PubMed] [Google Scholar]
- 17.Pyrgos V, Seitz AE, Steiner CA, Prevots DR, Williamson PR. Epidemiology of cryptococcal meningitis in the US: 1997–2009. PLoS ONE. 2013;8(2):e56269. doi: 10.1371/journal.pone.0056269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18•.Haddow LJ, Colebunders R, Meintjes G, et al. Cryptococcal immune reconstitution inflammatory syndrome in HIV-1-infected individuals: literature review and proposed clinical case definitions. Lancet Infect Dis. 2010;10(11):791–802. doi: 10.1016/S1473-3099(10)70170-5. Provides a comprehensive review of the immune reconstitution inflammatory syndrome in HIV-infected patients with C. neoformans. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Boulware DR, Meya DB, Muzoora C, et al. Timing of antiretroviral therapy after diagnosis of cryptococcal meningitis. N Engl J Med. 2014;370(26):2487–2498. doi: 10.1056/NEJMoa1312884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20•.Singh N, Lortholary O, Alexander BD, et al. An immune reconstitution syndrome-like illness associated with Cryptococcus neoformans infection in organ transplant recipients. Clin Infect Dis. 2005;40(12):1756–1761. doi: 10.1086/430606. Describes the occurrence of immune reconstitution inflammatory syndrome in solid organ transplant recipients. [DOI] [PubMed] [Google Scholar]
- 21••.Casadevall A, Pirofski LA. The damage-response framework of microbial pathogenesis. Nat Rev Microbiol. 2003;1(1):17–24. doi: 10.1038/nrmicro732. Reviews the damage-response framework – a unified theory of microbial pathogenesis that integrates the role of the host and microbe in the outcome of host–microbe interaction. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Singh N, Dromer F, Perfect JR, Lortholary O. Cryptococcosis in solid organ transplant recipients: current state of the science. Clin Infect Dis. 2008;47(10):1321–1327. doi: 10.1086/592690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Singh N, Forrest G. Cryptococcosis in solid organ transplant recipients. Am J Transplant. 2009;9(Suppl 4):S192–S198. doi: 10.1111/j.1600-6143.2009.02911.x. [DOI] [PubMed] [Google Scholar]
- 24.Horcajada JP, Pena JL, Martinez-Taboada VM, et al. Invasive Cryptococcosis and adalimumab treatment. Emerg Infect Dis. 2007;13(6):953–955. doi: 10.3201/eid1306.070154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ely EW, Peacock JE, Jr, Haponik EF, Washburn RG. Cryptococcal pneumonia complicating pregnancy. Medicine (Baltimore) 1998;77(3):153–167. doi: 10.1097/00005792-199805000-00001. [DOI] [PubMed] [Google Scholar]
- 26.Winkelstein JA, Marino MC, Ochs H, et al. The X-linked hyper-IgM syndrome: clinical and immunologic features of 79 patients. Medicine (Baltimore) 2003;82(6):373–384. doi: 10.1097/01.md.0000100046.06009.b0. [DOI] [PubMed] [Google Scholar]
- 27.Singh N, Husain S, De Vera M, Gayowski T, Cacciarelli TV. Cryptococcus neoformans infection in patients with cirrhosis, including liver transplant candidates. Medicine (Baltimore) 2004;83(3):188–192. doi: 10.1097/01.md.0000126760.45299.69. [DOI] [PubMed] [Google Scholar]
- 28.Netea MG, Brouwer AE, Hoogendoorn EH, et al. Two patients with cryptococcal meningitis and idiopathic CD4 lymphopenia: defective cytokine production and reversal by recombinant interferon- gamma therapy. Clin Infect Dis. 2004;39(9):e83–e87. doi: 10.1086/425121. [DOI] [PubMed] [Google Scholar]
- 29.Mitchell DH, Sorrell TC, Allworth AM, et al. Cryptococcal disease of the CNS in immunocompetent hosts: influence of cryptococcal variety on clinical manifestations and outcome. Clin Infect Dis. 1995;20(3):611–616. doi: 10.1093/clinids/20.3.611. [DOI] [PubMed] [Google Scholar]
- 30.Lui G, Lee N, Ip M, et al. Cryptococcosis in apparently immunocompetent patients. QJM. 2006;99(3):143–151. doi: 10.1093/qjmed/hcl014. [DOI] [PubMed] [Google Scholar]
- 31.Rosen LB, Freeman AF, Yang LM, et al. Anti-GM-CSF autoantibodies in patients with cryptococcal meningitis. J Immunol. 2013;190(8):3959–3966. doi: 10.4049/jimmunol.1202526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32•.Goldman DL, Khine H, Abadi J, et al. Serologic evidence for Cryptococcus neoformans infection in early childhood. Pediatrics. 2001;107(5):E66. doi: 10.1542/peds.107.5.e66. Provides serological evidence to show that infection with C. neoformans occurs in early childhood. [DOI] [PubMed] [Google Scholar]
- 33.Goldman DL, Li X, Tsirilakis K, Andrade C, Casadevall A, Vicencio AG. Increased chitinase expression and fungal-specific antibodies in the bronchoalveolar lavage fluid of asthmatic children. Clin Exp Allergy. 2012;42(4):523–530. doi: 10.1111/j.1365-2222.2011.03886.x. [DOI] [PubMed] [Google Scholar]
- 34.Deshaw M, Pirofski LA. Antibodies to the Cryptococcus neoformans capsular glucuronoxylomannan are ubiquitous in serum from HIV+ and HIV- individuals. Clin Exp Immunol. 1995;99(3):425–432. doi: 10.1111/j.1365-2249.1995.tb05568.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Abadi J, Pirofski L. Antibodies reactive with the cryptococcal capsular polysaccharide glucuronoxylomannan are present in sera from children with and without human immunodeficiency virus infection. J Infect Dis. 1999;180(3):915–919. doi: 10.1086/314953. [DOI] [PubMed] [Google Scholar]
- 36•.Garcia-Hermoso D, Janbon G, Dromer F. Epidemiological evidence for dormant Cryptococcus neoformans infection. J Clin Microbiol. 1999;37(10):3204–3209. doi: 10.1128/jcm.37.10.3204-3209.1999. Provided evidence to support the concept that C. neoformans can be acquired long before infection and that disease can subsequently occur in the setting of immune defects. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Jarvis JN, Lawn SD, Vogt M, Bangani N, Wood R, Harrison TS. Screening for cryptococcal antigenemia in patients accessing an antiretroviral treatment program in South Africa. Clin Infect Dis. 2009;48(7):856–862. doi: 10.1086/597262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38••.Meya DB, Manabe YC, Castelnuovo B, et al. Cost–effectiveness of serum cryptococcal antigen screening to prevent deaths among HIV-infected persons with a CD4+ cell count < or = 100 cells/microL who start HIV therapy in resource-limited settings. Clin Infect Dis. 2010;51(4):448–455. doi: 10.1086/655143. Showed that screening HIV-infected patients with less than 100 CD4 T cells for serum cryptococcal antigen to identify those who should receive fluconazole therapy was cost-effective and prevented deaths in a resource-limited setting. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Patel S, Shin GY, Wijewardana I, et al. The prevalence of cryptococcal antigenemia in newly diagnosed HIV patients in a Southwest London cohort. J Infect. 2013;66(1):75–79. doi: 10.1016/j.jinf.2012.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rajasingham R, Rolfes MA, Birkenkamp KE, Meya DB, Boulware DR. Cryptococcal meningitis treatment strategies in resource-limited settings: a cost-effectiveness analysis. PLoS Med. 2012;9(9):e1001316. doi: 10.1371/journal.pmed.1001316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jarvis JN, Percival A, Bauman S, et al. Evaluation of a novel point-of-care cryptococcal antigen test on serum, plasma, and urine from patients with HIV-associated cryptococcal meningitis. Clin Infect Dis. 2011;53(10):1019–1023. doi: 10.1093/cid/cir613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Global Action Fund for Fungal Infections (GAFFI) www.gaffi.org.
- 43.Olszewski MA, Zhang Y, Huffnagle GB. Mechanisms of cryptococcal virulence and persistence. Future Microbiol. 2010;5(8):1269–1288. doi: 10.2217/fmb.10.93. [DOI] [PubMed] [Google Scholar]
- 44.Vecchiarelli A, Pericolini E, Gabrielli E, et al. Elucidating the immunological function of the Cryptococcus neoformans capsule. Future Microbiol. 2013;8(9):1107–1116. doi: 10.2217/fmb.13.84. [DOI] [PubMed] [Google Scholar]
- 45.Zaragoza O, Rodrigues ML, De Jesus M, Frases S, Dadachova E, Casadevall A. The capsule of the fungal pathogen Cryptococcus neoformans. Adv Appl Microbiol. 2009;68:133–216. doi: 10.1016/S0065-2164(09)01204-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Del Poeta M. Role of phagocytosis in the virulence of Cryptococcus neoformans. Eukaryot Cell. 2004;3(5):1067–1075. doi: 10.1128/EC.3.5.1067-1075.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Feldmesser M, Tucker S, Casadevall A. Intracellular parasitism of macrophages by Cryptococcus neoformans. Trends Microbiol. 2001;9(6):273–278. doi: 10.1016/s0966-842x(01)02035-2. [DOI] [PubMed] [Google Scholar]
- 48.Alvarez M, Casadevall A. Phagosome extrusion and host-cell survival after Cryptococcus neoformans phagocytosis by macrophages. Curr Biol. 2006;16(21):2161–2165. doi: 10.1016/j.cub.2006.09.061. [DOI] [PubMed] [Google Scholar]
- 49.Ma H, May RC. Virulence in Cryptococcus species. Adv Appl Microbiol. 2009;67:131–190. doi: 10.1016/S0065-2164(08)01005-8. [DOI] [PubMed] [Google Scholar]
- 50.Garcia-Solache MA, Casadevall A. Global warming will bring new fungal diseases for mammals. mBio. 2010;1(1):e00061–10. doi: 10.1128/mBio.00061-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Casadevall A. Fungi and the rise of mammals. PLoS Pathog. 2012;8(8):e1002808. doi: 10.1371/journal.ppat.1002808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Molez JF. The historical question of acquired immunodeficiency syndrome in the 1960s in the Congo River basin area in relation to cryptococcal meningitis. Am J Trop Med Hyg. 1998;58(3):273–276. doi: 10.4269/ajtmh.1998.58.273. [DOI] [PubMed] [Google Scholar]
- 53.Baum GL, Artis D. Growth inhibition of Cryptococcus neoformans by cell free human serum. Am J Med Sci. 1961;241:613–616. doi: 10.1097/00000441-196105000-00009. [DOI] [PubMed] [Google Scholar]
- 54.Igel HJ, Bolande RP. Humoral defense mechanisms in cryptococcosis: substances in normal human serum, saliva, and cerebrospinal fluid affecting the growth of Cryptococcus neoformans. J Infect Dis. 1966;116(1):75–83. doi: 10.1093/infdis/116.1.75. [DOI] [PubMed] [Google Scholar]
- 55.Zaragoza O, Taborda CP, Casadevall A. The efficacy of complement-mediated phagocytosis of Cryptococcus neoformans is dependent on the location of C3 in the polysaccharide capsule and involves both direct and indirect C3-mediated interactions. Eur J Immunol. 2003;33(7):1957–1967. doi: 10.1002/eji.200323848. [DOI] [PubMed] [Google Scholar]
- 56.Diamond RD, May JE, Kane MA, Frank MM, Bennett JE. The role of the classical and alternate complement pathways in host defenses against Cryptococcus neoformans infection. J Immunol. 1974;112(6):2260–2270. [PubMed] [Google Scholar]
- 57.Rhodes JC, Wicker LS, Urba WJ. Genetic control of susceptibility to Cryptococcus neoformans in mice. Infect Immun. 1980;29(2):494–499. doi: 10.1128/iai.29.2.494-499.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Macher AM, Bennett JE, Gadek JE, Frank MM. Complement depletion in cryptococcal sepsis. J Immunol. 1978;120(5):1686–1690. [PubMed] [Google Scholar]
- 59.Gates MA, Kozel TR. Differential localization of complement component 3 within the capsular matrix of Cryptococcus neoformans. Infect Immun. 2006;74(6):3096–3106. doi: 10.1128/IAI.01213-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Pirofski LA. Of mice and men, revisited: new insights into an ancient molecule from studies of complement activation by Cryptococcus neoformans. Infect Immun. 2006;74(6):3079–3084. doi: 10.1128/IAI.00431-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Van Asbeck EC, Hoepelman AI, Scharringa J, Herpers BL, Verhoef J. Mannose binding lectin plays a crucial role in innate immunity against yeast by enhanced complement activation and enhanced uptake of polymorphonuclear cells. BMC Microbiol. 2008;8:229. doi: 10.1186/1471-2180-8-229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mansour MK, Levitz SM. Interactions of fungi with phagocytes. Curr Opin Microbiol. 2002;5(4):359–365. doi: 10.1016/s1369-5274(02)00342-9. [DOI] [PubMed] [Google Scholar]
- 63.Shao X, Mednick A, Alvarez M, Van Rooijen N, Casadevall A, Goldman DL. An innate immune system cell is a major determinant of species-related susceptibility differences to fungal pneumonia. J Immunol. 2005;175(5):3244–3251. doi: 10.4049/jimmunol.175.5.3244. [DOI] [PubMed] [Google Scholar]
- 64.Levitz SM, Tabuni A. Binding of Cryptococcus neoformans by human cultured macrophages. Requirements for multiple complement receptors and actin. J Clin Invest. 1991;87(2):528–535. doi: 10.1172/JCI115027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Griffin FM., Jr Roles of macrophage Fc and C3b receptors in phagocytosis of immunologically coated Cryptococcus neoformans. Proc Natl Acad Sci USA. 1981;78(6):3853–3857. doi: 10.1073/pnas.78.6.3853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Guerra CR, Seabra SH, De Souza W, Rozental S. Cryptococcus neoformans is internalized by receptor-mediated or ‘triggered’ phagocytosis, dependent on actin recruitment. PLoS ONE. 2014;9(2):e89250. doi: 10.1371/journal.pone.0089250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Dan JM, Kelly RM, Lee CK, Levitz SM. Role of the mannose receptor in a murine model of Cryptococcus neoformans infection. Infect Immun. 2008;76(6):2362–2367. doi: 10.1128/IAI.00095-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Syme RM, Spurrell JC, Amankwah EK, Green FH, Mody CH. Primary dendritic cells phagocytose Cryptococcus neoformans via mannose receptors and Fcgamma receptor II for presentation to T lymphocytes. Infect Immun. 2002;70(11):5972–5981. doi: 10.1128/IAI.70.11.5972-5981.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ozinsky A, Underhill DM, Fontenot JD, et al. The repertoire for pattern recognition of pathogens by the innate immune system is defined by cooperation between toll-like receptors. Proc Natl Acad Sci USA. 2000;97(25):13766–13771. doi: 10.1073/pnas.250476497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Shoham S, Huang C, Chen JM, Golenbock DT, Levitz SM. Toll-like receptor 4 mediates intracellular signaling without TNF-alpha release in response to Cryptococcus neoformans polysaccharide capsule. J Immunol. 2001;166(7):4620–4626. doi: 10.4049/jimmunol.166.7.4620. [DOI] [PubMed] [Google Scholar]
- 71.Nakamura K, Miyazato A, Xiao G, et al. Deoxynucleic acids from Cryptococcus neoformans activate myeloid dendritic cells via a TLR9-dependent pathway. J Immunol. 2008;180(6):4067–4074. doi: 10.4049/jimmunol.180.6.4067. [DOI] [PubMed] [Google Scholar]
- 72.Redlich S, Ribes S, Schutze S, Eiffert H, Nau R. Toll-like receptor stimulation increases phagocytosis of Cryptococcus neoformans by microglial cells. J Neuroinflammation. 2013;10:71. doi: 10.1186/1742-2094-10-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Tucker SC, Casadevall A. Replication of Cryptococcus neoformans in macrophages is accompanied by phagosomal permeabilization and accumulation of vesicles containing polysaccharide in the cytoplasm. Proc Natl Acad Sci USA. 2002;99(5):3165–3170. doi: 10.1073/pnas.052702799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kechichian TB, Shea J, Del Poeta M. Depletion of alveolar macrophages decreases the dissemination of a glucosylceramide-deficient mutant of Cryptococcus neoformans in immunodeficient mice. Infect Immun. 2007;75(10):4792–4798. doi: 10.1128/IAI.00587-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Nicola AM, Robertson EJ, Albuquerque P, da Derengowski LS, Casadevall A. Nonlytic exocytosis of Cryptococcus neoformans from macrophages occurs in vivo and is influenced by phagosomal pH. mBio. 2011;2(4):e00167–11. doi: 10.1128/mBio.00167-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Charlier C, Nielsen K, Daou S, Brigitte M, Chretien F, Dromer F. Evidence of a role for monocytes in dissemination and brain invasion by Cryptococcus neoformans. Infect Immun. 2009;77(1):120–127. doi: 10.1128/IAI.01065-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Baba T. Electron microscopic cytochemical analysis of hepatic granuloma induced by Cryptococcus neoformans. Mycopathologia. 1988;104(1):37–46. doi: 10.1007/BF00437922. [DOI] [PubMed] [Google Scholar]
- 78.Vecchiarelli A, Dottorini M, Pietrella D, et al. Role of human alveolar macrophages as antigen-presenting cells in Cryptococcus neoformans infection. Am J Respir Cell Mol Biol. 1994;11(2):130–137. doi: 10.1165/ajrcmb.11.2.8049074. [DOI] [PubMed] [Google Scholar]
- 79.Voelz K, Lammas DA, May RC. Cytokine signaling regulates the outcome of intracellular macrophage parasitism by Cryptococcus neoformans. Infect Immun. 2009;77(8):3450–3457. doi: 10.1128/IAI.00297-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Mcquiston TJ, Williamson PR. Paradoxical roles of alveolar macrophages in the host response to Cryptococcus neoformans. J Infect Chemother. 2012;18(1):1–9. doi: 10.1007/s10156-011-0306-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Davis MJ, Tsang TM, Qiu Y, et al. Macrophage M1/M2 polarization dynamically adapts to changes in cytokine microenvironments in Cryptococcus neoformans infection. mBio. 2013;4(3):e00264–13. doi: 10.1128/mBio.00264-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kelly RM, Chen J, Yauch LE, Levitz SM. Opsonic requirements for dendritic cell-mediated responses to Cryptococcus neoformans. Infect Immun. 2005;73(1):592–598. doi: 10.1128/IAI.73.1.592-598.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Siegemund S, Alber G. Cryptococcus neoformans activates bone marrow-derived conventional dendritic cells rather than plasmacytoid dendritic cells and down-regulates macrophages. FEMS Immunol Med Microbiol. 2008;52(3):417–427. doi: 10.1111/j.1574-695X.2008.00391.x. [DOI] [PubMed] [Google Scholar]
- 84.Wozniak KL, Vyas JM, Levitz SM. In vivo role of dendritic cells in a murine model of pulmonary cryptococcosis. Infect Immun. 2006;74(7):3817–3824. doi: 10.1128/IAI.00317-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wozniak KL, Levitz SM. Cryptococcus neoformans enters the endolysosomal pathway of dendritic cells and is killed by lysosomal components. Infect Immun. 2008;76(10):4764–4771. doi: 10.1128/IAI.00660-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Mambula SS, Simons ER, Hastey R, Selsted ME, Levitz SM. Human neutrophil-mediated nonoxidative antifungal activity against Cryptococcus neoformans. Infect Immun. 2000;68(11):6257–6264. doi: 10.1128/iai.68.11.6257-6264.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kozel TR, Pfrommer GS, Redelman D. Activated neutrophils exhibit enhanced phagocytosis of Cryptococcus neoformans opsonized with normal human serum. Clin Exp Immunol. 1987;70(1):238–246. [PMC free article] [PubMed] [Google Scholar]
- 88.Miller MF, Mitchell TG. Killing of Cryptococcus neoformans strains by human neutrophils and monocytes. Infect Immun. 1991;59(1):24–28. doi: 10.1128/iai.59.1.24-28.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Alcouloumre MS, Ghannoum MA, Ibrahim AS, Selsted ME, Edwards JE., Jr Fungicidal properties of defensin NP-1 and activity against Cryptococcus neoformans in vitro. Antimicrob Agents Chemother. 1993;37(12):2628–2632. doi: 10.1128/aac.37.12.2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90•.Mednick AJ, Feldmesser M, Rivera J, Casadevall A. Neutropenia alters lung cytokine production in mice and reduces their susceptibility to pulmonary cryptococcosis. Eur J Immunol. 2003;33(6):1744–1753. doi: 10.1002/eji.200323626. Revealed that neutrophils can play a detrimental role in experimental cryptococcosis by dysregulating the lung cytokine response. [DOI] [PubMed] [Google Scholar]
- 91.Holmer SM, Evans KS, Asfaw YG, et al. Impact of surfactant protein D, interleukin-5, and eosinophilia on Cryptococcosis. Infect Immun. 2014;82(2):683–693. doi: 10.1128/IAI.00855-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Feldmesser M, Casadevall A, Kress Y, Spira G, Orlofsky A. Eosinophil-Cryptococcus neoformans interactions in vivo and in vitro. Infect Immun. 1997;65(5):1899–1907. doi: 10.1128/iai.65.5.1899-1907.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yamaguchi H, Komase Y, Ikehara M, Yamamoto T, Shinagawa T. Disseminated cryptococcal infection with eosinophilia in a healthy person. J Infect Chemother. 2008;14(4):319–324. doi: 10.1007/s10156-008-0618-z. [DOI] [PubMed] [Google Scholar]
- 94••.Rohatgi S, Gohil S, Kuniholm MH, et al. Fc gamma receptor 3A polymorphism and risk for HIV-associated cryptococcal disease. mBio. 2013;4(5):e00573–13. doi: 10.1128/mBio.00573-13. Showed that the same high affinity FcRIII polymorphism that was associated with risk for cyrptococcosis was also identified in HIV-infected patients with cryptococcosis and provided a possible mechanistic explanation for why it might enhance cryptococcal pathogenesis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Schmidt S, Zimmermann SY, Tramsen L, Koehl U, Lehrnbecher T. Natural killer cells and antifungal host response. Clin Vaccine Immunol. 2013;20(4):452–458. doi: 10.1128/CVI.00606-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Levitz SM, Dupont MP. Phenotypic and functional characterization of human lymphocytes activated by interleukin-2 to directly inhibit growth of Cryptococcus neoformans in vitro. J Clin Invest. 1993;91(4):1490–1498. doi: 10.1172/JCI116354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Ma LL, Wang CL, Neely GG, Epelman S, Krensky AM, Mody CH. NK cells use perforin rather than granulysin for anticryptococcal activity. J Immunol. 2004;173(5):3357–3365. doi: 10.4049/jimmunol.173.5.3357. [DOI] [PubMed] [Google Scholar]
- 98.Marr KJ, Jones GJ, Zheng C, et al. Cryptococcus neoformans directly stimulates perforin production and rearms NK cells for enhanced anticryptococcal microbicidal activity. Infect Immun. 2009;77(6):2436–2446. doi: 10.1128/IAI.01232-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Li SS, Kyei SK, Timm-Mccann M, et al. The NK receptor NKp30 mediates direct fungal recognition and killing and is diminished in NK cells from HIV-infected patients. Cell Host Microbe. 2013;14(4):387–397. doi: 10.1016/j.chom.2013.09.007. [DOI] [PubMed] [Google Scholar]
- 100.Kawakami K, Kinjo Y, Uezu K, et al. Monocyte chemoattractant protein-1-dependent increase of V alpha 14 NKT cells in lungs and their roles in Th1 response and host defense in cryptococcal infection. J Immunol. 2001;167(11):6525–6532. doi: 10.4049/jimmunol.167.11.6525. [DOI] [PubMed] [Google Scholar]
- 101.Kawakami K, Kinjo Y, Yara S, et al. Activation of Valpha14(+) natural killer T cells by alpha-galactosylceramide results in development of Th1 response and local host resistance in mice infected with Cryptococcus neoformans. Infect Immun. 2001;69(1):213–220. doi: 10.1128/IAI.69.1.213-220.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Taniguchi M, Harada M, Kojo S, Nakayama T, Wakao H. The regulatory role of Valpha14 NKT cells in innate and acquired immune response. Annu Rev Immunol. 2003;21:483–513. doi: 10.1146/annurev.immunol.21.120601.141057. [DOI] [PubMed] [Google Scholar]
- 103.Huffnagle GB, Yates JL, Lipscomb MF. T cell-mediated immunity in the lung: a Cryptococcus neoformans pulmonary infection model using SCID and athymic nude mice. Infect Immun. 1991;59(4):1423–1433. doi: 10.1128/iai.59.4.1423-1433.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Mody CH, Lipscomb MF, Street NE, Toews GB. Depletion of CD4+ (L3T4+) lymphocytes in vivo impairs murine host defense to Cryptococcus neoformans. J Immunol. 1990;144(4):1472–1477. [PubMed] [Google Scholar]
- 105.Hill JO, Aguirre KM. CD4+ T cell-dependent acquired state of immunity that protects the brain against Cryptococcus neoformans. J Immunol. 1994;152(5):2344–2350. [PubMed] [Google Scholar]
- 106.Buchanan KL, Doyle HA. Requirement for CD4(+) T lymphocytes in host resistance against Cryptococcus neoformans in the central nervous system of immunized mice. Infect Immun. 2000;68(2):456–462. doi: 10.1128/iai.68.2.456-462.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lim TS, Murphy JW. Transfer of immunity to cryptococcosis by T-enriched splenic lymphocytes from Cryptococcus neoformans-sensitized mice. Infect Immun. 1980;30(1):5–11. doi: 10.1128/iai.30.1.5-11.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Huffnagle GB, Lipscomb MF, Lovchik JA, Hoag KA, Street NE. The role of CD4+ and CD8+ T cells in the protective inflammatory response to a pulmonary cryptococcal infection. J Leukoc Biol. 1994;55(1):35–42. doi: 10.1002/jlb.55.1.35. [DOI] [PubMed] [Google Scholar]
- 109.Specht CA, Nong S, Dan JM, Lee CK, Levitz SM. Contribution of glycosylation to T cell responses stimulated by recombinant Cryptococcus neoformans mannoprotein. J Infect Dis. 2007;196(5):796–800. doi: 10.1086/520536. [DOI] [PubMed] [Google Scholar]
- 110.Crowe SM, Carlin JB, Stewart KI, Lucas CR, Hoy JF. Predictive value of CD4 lymphocyte numbers for the development of opportunistic infections and malignancies in HIV-infected persons. J Acquir Immune Defic Syndr. 1991;4(8):770–776. [PubMed] [Google Scholar]
- 111.Jarvis JN, Harrison TS. HIV-associated cryptococcal meningitis. AIDS. 2007;21(16):2119–2129. doi: 10.1097/QAD.0b013e3282a4a64d. [DOI] [PubMed] [Google Scholar]
- 112.Lortholary O, Poizat G, Zeller V, et al. Long-term outcome of AIDS-associated cryptococcosis in the era of combination antiretroviral therapy. AIDS. 2006;20(17):2183–2191. doi: 10.1097/01.aids.0000252060.80704.68. [DOI] [PubMed] [Google Scholar]
- 113.Zonios DI, Falloon J, Huang CY, Chaitt D, Bennett JE. Cryptococcosis and idiopathic CD4 lymphocytopenia. Medicine (Baltimore) 2007;86(2):78–92. doi: 10.1097/md.0b013e31803b52f5. [DOI] [PubMed] [Google Scholar]
- 114.Van De Veerdonk FL, Netea MG. T-cell subsets and antifungal host defenses. Curr Fungal Infect Rep. 2010;4(4):238–243. doi: 10.1007/s12281-010-0034-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Ma LL, Spurrell JC, Wang JF, et al. CD8 T cell-mediated killing of Cryptococcus neoformans requires granulysin and is dependent on CD4 T cells and IL-15. J Immunol. 2002;169(10):5787–5795. doi: 10.4049/jimmunol.169.10.5787. [DOI] [PubMed] [Google Scholar]
- 116.Mody CH, Chen GH, Jackson C, Curtis JL, Toews GB. In vivo depletion of murine CD8 positive T cells impairs survival during infection with a highly virulent strain of Cryptococcus neoformans. Mycopathologia. 1994;125(1):7–17. doi: 10.1007/BF01103969. [DOI] [PubMed] [Google Scholar]
- 117.Lindell DM, Moore TA, Mcdonald RA, Toews GB, Huffnagle GB. Generation of antifungal effector CD8+ T cells in the absence of CD4+ T cells during Cryptococcus neoformans infection. J Immunol. 2005;174(12):7920–7928. doi: 10.4049/jimmunol.174.12.7920. [DOI] [PubMed] [Google Scholar]
- 118.Uezu K, Kawakami K, Miyagi K, et al. Accumulation of gammadelta T cells in the lungs and their regulatory roles in Th1 response and host defense against pulmonary infection with Cryptococcus neoformans. J Immunol. 2004;172(12):7629–7634. doi: 10.4049/jimmunol.172.12.7629. [DOI] [PubMed] [Google Scholar]
- 119.Wozniak KL, Kolls JK, Wormley FL., Jr Depletion of neutrophils in a protective model of pulmonary cryptococcosis results in increased IL-17A production by gammadelta T cells. BMC Immunol. 2012;13:65. doi: 10.1186/1471-2172-13-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Huffnagle GB, Toews GB, Burdick MD, et al. Afferent phase production of TNF-alpha is required for the development of protective T cell immunity to Cryptococcus neoformans. J Immunol. 1996;157(10):4529–4536. [PubMed] [Google Scholar]
- 121.Herring AC, Lee J, McDonald RA, Toews GB, Huffnagle GB. Induction of interleukin-12 and gamma interferon requires tumor necrosis factor alpha for protective T1-cell-mediated immunity to pulmonary Cryptococcus neoformans infection. Infect Immun. 2002;70(6):2959–2964. doi: 10.1128/IAI.70.6.2959-2964.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Retini C, Vecchiarelli A, Monari C, Tascini C, Bistoni F, Kozel TR. Capsular polysaccharide of Cryptococcus neoformans induces proinflammatory cytokine release by human neutrophils. Infect Immun. 1996;64(8):2897–2903. doi: 10.1128/iai.64.8.2897-2903.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hole CR, Wormley FL., Jr Vaccine and immunotherapeutic approaches for the prevention of cryptococcosis: lessons learned from animal models. Front Microbiol. 2012;3:291. doi: 10.3389/fmicb.2012.00291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Szymczak WA, Sellers RS, Pirofski LA. IL-23 dampens the allergic response to Cryptococcus neoformans through IL-17-independent and -dependent mechanisms. Am J Pathol. 2012;180(4):1547–1559. doi: 10.1016/j.ajpath.2011.12.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Wozniak KL, Hardison SE, Kolls JK, Wormley FL. Role of IL-17A on resolution of pulmonary C. neoformans infection. PLoS ONE. 2011;6(2):e17204. doi: 10.1371/journal.pone.0017204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126••.Jarvis JN, Casazza JP, Stone HH, et al. The phenotype of the Cryptococcus-specific CD4+ memory T-cell response is associated with disease severity and outcome in HIV-associated cryptococcal meningitis. J Infect Dis. 2013;207(12):1817–1828. doi: 10.1093/infdis/jit099. Found that presence of IFN-γ/TNF-α-producing cryptococcal antigen-specific peripheral CD4+ T-cell response correlated with survival in patients with HIV-associated cryptococcal meningitis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Bicanic T, Muzoora C, Brouwer AE, et al. Independent association between rate of clearance of infection and clinical outcome of HIV-associated cryptococcal meningitis. analysis of a combined cohort of 262 patients. Clin Infect Dis. 2009;49(5):702–709. doi: 10.1086/604716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Siddiqui AA, Brouwer AE, Wuthiekanun V, et al. IFN-gamma at the site of infection determines rate of clearance of infection in cryptococcal meningitis. J Immunol. 2005;174(3):1746–1750. doi: 10.4049/jimmunol.174.3.1746. [DOI] [PubMed] [Google Scholar]
- 129•.Jarvis JN, Meintjes G, Rebe K, et al. Adjunctive interferon-gamma immunotherapy for the treatment of HIV-associated cryptococcal meningitis: a randomized controlled trial. AIDS. 2012;26(9):1105–1113. doi: 10.1097/QAD.0b013e3283536a93. Showed that addition of IFN-γ to standard treatment significantly increased the rate of clearance of cryptococcal infection from cerebrospinal fluid and was not associated with any adverse events. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130•.Wormley FL, Jr, Perfect JR, Steele C, Cox GM. Protection against cryptococcosis by using a murine gamma interferon-producing Cryptococcus neoformans strain. Infect Immun. 2007;75(3):1453–1462. doi: 10.1128/IAI.00274-06. Established the importance of IFN-γ at the site of infection in resistance to cryptococcosis using a C. neoformans strain that produced murine IFN-γ. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Subramaniam KS, Datta K, Marks MS, Pirofski LA. Improved survival of mice deficient in secretory immunoglobulin M following systemic infection with Cryptococcus neoformans. Infect Immun. 2010;78(1):441–452. doi: 10.1128/IAI.00506-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132•.Rohatgi S, Pirofski LA. Molecular characterization of the early B cell response to pulmonary Cryptococcus neoformans infection. J Immunol. 2012;189(12):5820–5830. doi: 10.4049/jimmunol.1201514. Established that B1 cells are an indispensable component of the early immune response to C. neoformans and that do so by enhancing the phagocytic ability of alveolar macrophages. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Szymczak WA, Davis MJ, Lundy SK, Dufaud C, Olszewski M, Pirofski LA. X-linked immunodeficient mice exhibit enhanced susceptibility to Cryptococcus neoformans Infection. mBio. 2013;4(4):e00265–13. doi: 10.1128/mBio.00265-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Monga DP, Kumar R, Mohapatra LN, Malaviya AN. Experimental cryptococcosis in normal and B-cell-deficient mice. Infect Immun. 1979;26(1):1–3. doi: 10.1128/iai.26.1.1-3.1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Aguirre KM, Johnson LL. A role for B cells in resistance to Cryptococcus neoformans in mice. Infect Immun. 1997;65(2):525–530. doi: 10.1128/iai.65.2.525-530.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136•.Feldmesser M, Mednick A, Casadevall A. Antibody-mediated protection in murine Cryptococcus neoformans infection is associated with pleotrophic effects on cytokine and leukocyte responses. Infect Immun. 2002;70(3):1571–1580. doi: 10.1128/IAI.70.3.1571-1580.2002. Demonstrated that the protective efficacy of mouse monoclonal antibodies to glucuronoxylomannan was associated with downregulation of inflammatory cytokine responses, reducing host damage. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137•.Rivera J, Zaragoza O, Casadevall A. Antibody-mediated protection against Cryptococcus neoformans pulmonary infection is dependent on B cells. Infect Immun. 2005;73(2):1141–1150. doi: 10.1128/IAI.73.2.1141-1150.2005. Showed that B-cell-deficient mice have deregulated, excessive inflammatory response to C. neoformans that abrogates antibody-mediated protection. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Marquis G, Montplaisir S, Pelletier M, Mousseau S, Auger P. Genetic resistance to murine cryptococcosis: increased susceptibility in the CBA/N XID mutant strain of mice. Infect Immun. 1985;47(1):282–287. doi: 10.1128/iai.47.1.282-287.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Almeida SR, Aroeira LS, Frymuller E, et al. Mouse B-1 cell-derived mononuclear phagocyte, a novel cellular component of acute non-specific inflammatory exudate. Int Immunol. 2001;13(9):1193–1201. doi: 10.1093/intimm/13.9.1193. [DOI] [PubMed] [Google Scholar]
- 140•.Ghosn EE, Russo M, Almeida SR. Nitric oxide-dependent killing of Cryptococcus neoformans by B-1-derived mononuclear phagocyte. J Leukoc Biol. 2006;80(1):36–44. doi: 10.1189/jlb.1005603. Showed that B1-derived phagocytic cells could internalize and kill C. neoformans in vitro. [DOI] [PubMed] [Google Scholar]
- 141.Gronwall C, Vas J, Silverman GJ. Protective Roles of Natural IgM Antibodies. Front Immunol. 2012;3:66. doi: 10.3389/fimmu.2012.00066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Subramaniam K, Metzger B, Hanau LH, et al. IgM(+) memory B cell expression predicts HIV-associated cryptococcosis status. J Infect Dis. 2009;200(2):244–251. doi: 10.1086/599318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Capolunghi F, Rosado MM, Sinibaldi M, Aranburu A, Carsetti R. Why do we need IgM memory B cells? Immunol Lett. 2013;152(2):114–120. doi: 10.1016/j.imlet.2013.04.007. [DOI] [PubMed] [Google Scholar]
- 144•.Subramaniam KS, Datta K, Quintero E, Manix C, Marks MS, Pirofski LA. The absence of serum IgM enhances the susceptibility of mice to pulmonary challenge with Cryptococcus neoformans. J Immunol. 2010;184(10):5755–5767. doi: 10.4049/jimmunol.0901638. Showed that C. neoformans dissemination to the brain was increased in IgM-deficient mice and that alveolar macrophages from these mice had an impaired ability to phagocytose C. neoformans, which was reconstituted by naive mouse IgM. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Rachini A, Pietrella D, Lupo P, et al. An anti-beta-glucan monoclonal antibody inhibits growth and capsule formation of Cryptococcus neoformans in vitro and exerts therapeutic, anticryptococcal activity in vivo. Infect Immun. 2007;75(11):5085–5094. doi: 10.1128/IAI.00278-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Torosantucci A, Chiani P, Bromuro C, et al. Protection by anti-beta-glucan antibodies is associated with restricted beta-1,3 glucan binding specificity and inhibition of fungal growth and adherence. PLoS ONE. 2009;4(4):e5392. doi: 10.1371/journal.pone.0005392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Houpt DC, Pfrommer GS, Young BJ, Larson TA, Kozel TR. Occurrences, immunoglobulin classes, and biological activities of antibodies in normal human serum that are reactive with Cryptococcus neoformans glucuronoxylomannan. Infect Immun. 1994;62(7):2857–2864. doi: 10.1128/iai.62.7.2857-2864.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Keller RG, Pfrommer GS, Kozel TR. Occurrences, specificities, and functions of ubiquitous antibodies in human serum that are reactive with the Cryptococcus neoformans cell wall. Infect Immun. 1994;62(1):215–220. doi: 10.1128/iai.62.1.215-220.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Dromer F, Aucouturier P, Clauvel JP, Saimot G, Yeni P. Cryptococcus neoformans antibody levels in patients with AIDS. Scand J Infect Dis. 1988;20(3):283–285. doi: 10.3109/00365548809032452. [DOI] [PubMed] [Google Scholar]
- 150.Fleuridor R, Lyles RH, Pirofski L. Quantitative and qualitative differences in the serum antibody profiles of human immunodeficiency virus-infected persons with and without Cryptococcus neoformans meningitis. J Infect Dis. 1999;180(5):1526–1535. doi: 10.1086/315102. [DOI] [PubMed] [Google Scholar]
- 151.Subramaniam K, French N, Pirofski LA. Cryptococcus neoformans-reactive and total immunoglobulin profiles of human immunodeficiency virus-infected and uninfected Ugandans. Clin Diagn Lab Immunol. 2005;12(10):1168–1176. doi: 10.1128/CDLI.12.10.1168-1176.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Jalali Z, Ng L, Singh N, Pirofski LA. Antibody response to Cryptococcus neoformans capsular polysaccharide glucuronoxylomannan in patients after solid-organ transplantation. Clin Vaccine Immunol. 2006;13(7):740–746. doi: 10.1128/CVI.00139-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Carsetti R, Rosado MM, Donnanno S, et al. The loss of IgM memory B cells correlates with clinical disease in common variable immunodeficiency. J Allergy Clin Immunol. 2005;115(2):412–417. doi: 10.1016/j.jaci.2004.10.048. [DOI] [PubMed] [Google Scholar]
- 154.Hart M, Steel A, Clark SA, et al. Loss of discrete memory B cell subsets is associated with impaired immunization responses in HIV-1 infection and may be a risk factor for invasive pneumococcal disease. J Immunol. 2007;178(12):8212–8220. doi: 10.4049/jimmunol.178.12.8212. [DOI] [PubMed] [Google Scholar]
- 155.Moir S, Fauci AS. B cells in HIV infection and disease. Nat Rev Immunol. 2009;9(4):235–245. doi: 10.1038/nri2524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Salinas-Carmona MC, Perez LI, Galan K, Vazquez AV. Immunosuppressive drugs have different effect on B lymphocyte subsets and IgM antibody production in immunized BALB/c mice. Autoimmunity. 2009;42(6):537–544. doi: 10.1080/08916930903019119. [DOI] [PubMed] [Google Scholar]
- 157.Devi SJ, Schneerson R, Egan W, et al. Cryptococcus neoformans serotype A glucuronoxylomannan-protein conjugate vaccines: synthesis, characterization, and immunogenicity. Infect Immun. 1991;59(10):3700–3707. doi: 10.1128/iai.59.10.3700-3707.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Devi SJ. Preclinical efficacy of a glucuronoxylomannan-tetanus toxoid conjugate vaccine of Cryptococcus neoformans in a murine model. Vaccine. 1996;14(9):841–844. doi: 10.1016/0264-410x(95)00256-z. [DOI] [PubMed] [Google Scholar]
- 159.Marshall E. Dispute slows paper on “remarkable” vaccine. Science. 1995;268(5218):1712–1715. doi: 10.1126/science.7792593. [DOI] [PubMed] [Google Scholar]
- 160•.Casadevall A, Pirofski L. Insights into mechanisms of antibody-mediated immunity from studies with Cryptococcus neoformans. Curr Mol Med. 2005;5(4):421–433. doi: 10.2174/1566524054022567. Reviews the characteristics of antibodies to glucuronoxylomannan that mediate protection against C. neoformans and provides new insights into mechanisms of antibody action. [DOI] [PubMed] [Google Scholar]
- 161.Zhong Z, Pirofski LA. Opsonization of Cryptococcus neoformans by human anticryptococcal glucuronoxylomannan antibodies. Infect Immun. 1996;64(9):3446–3450. doi: 10.1128/iai.64.9.3446-3450.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Fleuridor R, Zhong Z, Pirofski L. A human IgM monoclonal antibody prolongs survival of mice with lethal cryptococcosis. J Infect Dis. 1998;178(4):1213–1216. doi: 10.1086/515688. [DOI] [PubMed] [Google Scholar]
- 163.Maitta RW, Datta K, Chang Q, et al. Protective and nonprotective human immunoglobulin M monoclonal antibodies to Cryptococcus neoformans glucuronoxylomannan manifest different specificities and gene use profiles. Infect Immun. 2004;72(8):4810–4818. doi: 10.1128/IAI.72.8.4810-4818.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Fleuridor R, Lees A, Pirofski L. A cryptococcal capsular polysaccharide mimotope prolongs the survival of mice with Cryptococcus neoformans infection. J Immunol. 2001;166(2):1087–1096. doi: 10.4049/jimmunol.166.2.1087. [DOI] [PubMed] [Google Scholar]
- 165.Mukherjee J, Casadevall A, Scharff MD. Molecular characterization of the humoral responses to Cryptococcus neoformans infection and glucuronoxylomannan-tetanus toxoid conjugate immunization. J Exp Med. 1993;177(4):1105–1116. doi: 10.1084/jem.177.4.1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Casadevall A, Deshaw M, Fan M, Dromer F, Kozel TR, Pirofski LA. Molecular and idiotypic analysis of antibodies to Cryptococcus neoformans glucuro-noxylomannan. Infect Immun. 1994;62(9):3864–3872. doi: 10.1128/iai.62.9.3864-3872.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Maitta RW, Datta K, Lees A, Belouski SS, Pirofski LA. Immunogenicity and efficacy of Cryptococcus neoformans capsular polysaccharide glucuronoxylomannan peptide mimotope-protein conjugates in human immunoglobulin transgenic mice. Infect Immun. 2004;72(1):196–208. doi: 10.1128/IAI.72.1.196-208.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Pirofski LA. Polysaccharides, mimotopes and vaccines for fungal and encapsulated pathogens. Trends Microbiol. 2001;9(9):445–451. doi: 10.1016/s0966-842x(01)02134-5. [DOI] [PubMed] [Google Scholar]
- 169.Casadevall A, Cleare W, Feldmesser M, et al. Characterization of a murine monoclonal antibody to Cryptococcus neoformans polysaccharide that is a candidate for human therapeutic studies. Antimicrob Agents Chemother. 1998;42(6):1437–1446. doi: 10.1128/aac.42.6.1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Larsen RA, Pappas PG, Perfect J, et al. Phase I evaluation of the safety and pharmacokinetics of murine-derived anticryptococcal antibody 18B7 in subjects with treated cryptococcal meningitis. Antimicrob Agents Chemother. 2005;49(3):952–958. doi: 10.1128/AAC.49.3.952-958.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Bryan RA, Jiang Z, Howell RC, et al. Radioimmunotherapy is more effective than antifungal treatment in experimental cryptococcal infection. J Infect Dis. 2010;202(4):633–637. doi: 10.1086/654813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Netea MG. Toward identification of the genetic risk profile for cryptococcal disease in HIV-infected patients. mBio. 2013;4(5):e00798–00713. doi: 10.1128/mBio.00798-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ou XT, Wu JQ, Zhu LP, et al. Genotypes coding for mannose-binding lectin deficiency correlated with cryptococcal meningitis in HIV-uninfected Chinese patients. J Infect Dis. 2011;203(11):1686–1691. doi: 10.1093/infdis/jir152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Van Sorge NM, Van Der Pol WL, Van De Winkel JG. FcgammaR polymorphisms: implications for function, disease susceptibility and immunotherapy. Tissue Antigens. 2003;61(3):189–202. doi: 10.1034/j.1399-0039.2003.00037.x. [DOI] [PubMed] [Google Scholar]
- 175.Van Der Pol W, Van De Winkel JG. IgG receptor polymorphisms: risk factors for disease. Immunogenetics. 1998;48(3):222–232. doi: 10.1007/s002510050426. [DOI] [PubMed] [Google Scholar]
- 176.Gillis C, Gouel-Cheron A, Jonsson F, Bruhns P. Contribution of human FcgammaRs to disease with evidence from human polymorphisms and transgenic animal studies. Front Immunol. 2014;5:254. doi: 10.3389/fimmu.2014.00254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177•.Meletiadis J, Walsh TJ, Choi EH, et al. Study of common functional genetic polymorphisms of FCGR2A, 3A and 3B genes and the risk for cryptococcosis in HIV-uninfected patients. Med Mycol. 2007;45(6):513–518. doi: 10.1080/13693780701390140. Identified two Fc receptor polymorphisms associated with susceptibility to cryptococcosis in HIV-uninfected patients. [DOI] [PubMed] [Google Scholar]
- 178.Hu XP, Wu JQ, Zhu LP, et al. Association of Fcgamma receptor IIB polymorphism with cryptococcal meningitis in HIV-uninfected Chinese patients. PLoS ONE. 2012;7(8):e42439. doi: 10.1371/journal.pone.0042439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Parren PW, Warmerdam PA, Boeije LC, et al. On the interaction of IgG subclasses with the low affinity Fc gamma RIIa (CD32) on human monocytes, neutrophils, and platelets. Analysis of a functional polymorphism to human IgG2. J Clin Invest. 1992;90(4):1537–1546. doi: 10.1172/JCI116022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Salmon JE, Edberg JC, Brogle NL, Kimberly RP. Allelic polymorphisms of human Fc gamma receptor IIA and Fc gamma receptor IIIB. Independent mechanisms for differences in human phagocyte function. J Clin Invest. 1992;89(4):1274–1281. doi: 10.1172/JCI115712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Forthal DN, Landucci G, Bream J, Jacobson LP, Phan TB, Montoya B. FcgammaRIIa genotype predicts progression of HIV infection. J Immunol. 2007;179(11):7916–7923. doi: 10.4049/jimmunol.179.11.7916. [DOI] [PubMed] [Google Scholar]
- 182.Yuan R, Clynes R, Oh J, Ravetch JV, Scharff MD. Antibody-mediated modulation of Cryptococcus neoformans infection is dependent on distinct Fc receptor functions and IgG subclasses. J Exp Med. 1998;187(4):641–648. doi: 10.1084/jem.187.4.641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Wu J, Edberg JC, Redecha PB, et al. A novel polymorphism of FcgammaRIIIa (CD16) alters receptor function and predisposes to autoimmune disease. J Clin Invest. 1997;100(5):1059–1070. doi: 10.1172/JCI119616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Smith KG, Clatworthy MR. FcgammaRIIB in autoimmunity and infection: evolutionary and therapeutic implications. Nat Rev Immunol. 2010;10(5):328–343. doi: 10.1038/nri2762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Datta K, Pirofski LA. Towards a vaccine for Cryptococcus neoformans: principles and caveats. FEMS Yeast Res. 2006;6(4):525–536. doi: 10.1111/j.1567-1364.2006.00073.x. [DOI] [PubMed] [Google Scholar]
- 186.Huang H, Ostroff GR, Lee CK, Specht CA, Levitz SM. Characterization and optimization of the glucan particle-based vaccine platform. Clin Vaccine Immunol. 2013;20(10):1585–1591. doi: 10.1128/CVI.00463-13. [DOI] [PMC free article] [PubMed] [Google Scholar]