Next Article in Journal
Methodological Issues in Antifungal Susceptibility Testing of Malassezia pachydermatis
Next Article in Special Issue
Immune Recognition of Fungal Polysaccharides
Previous Article in Journal
Biodiversity of Pigmented Fungi Isolated from Marine Environment in La Réunion Island, Indian Ocean: New Resources for Colored Metabolites
Previous Article in Special Issue
Macrophage Migration Is Impaired within Candida albicans Biofilms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Innate Immune Responses to Cryptococcus

Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
J. Fungi 2017, 3(3), 35; https://doi.org/10.3390/jof3030035
Submission received: 24 May 2017 / Revised: 21 June 2017 / Accepted: 29 June 2017 / Published: 2 July 2017
(This article belongs to the Special Issue Host–Fungus Interactions)

Abstract

:
Cryptococcus species are encapsulated fungi found in the environment that predominantly cause disease in immunocompromised hosts after inhalation into the lungs. Even with contemporary antifungal regimens, patients with cryptococcosis continue to have high morbidity and mortality rates. The development of more effective therapies may depend on our understanding of the cellular and molecular mechanisms by which the host promotes sterilizing immunity against the fungus. This review will highlight our current knowledge of how Cryptococcus, primarily the species C. neoformans, is sensed by the mammalian host and how subsequent signaling pathways direct the anti-cryptococcal response by effector cells of the innate immune system.

1. Introduction

The encapsulated, yeast-like fungi of the genus Cryptococcus are prevalent throughout the environment worldwide. The most common species that cause disease in humans are Cryptococcus neoformans and Cryptococcus gattii. These pathogens can cause a life-threatening meningoencephalitis after acquisition through the respiratory tract and subsequent dissemination to the central nervous system (CNS). While C. gattii can infect apparently immunocompetent hosts, C. neoformans is more often an opportunistic pathogen, affecting immunocompromised patients including those with HIV/AIDS, cancer and solid organ transplantation [1].
Cryptococcal meningitis has been estimated to affect up to 1 million people worldwide each year [2,3]. Despite modern-day combination antifungal therapy, the mortality rate for cryptococcal meningitis is estimated at 15–25% [4,5], and the at-risk population is expanding with the development of new immunosuppressive regimens for autoimmunity and cancer [6]. More effective approaches to treating cryptococcosis may necessitate the incorporation of immunomodulatory therapies. Therefore, it is essential to understand the cellular and molecular mechanisms of immunity to Cryptococcus in mammalian hosts. While the adaptive immune response to Cryptococcus is an important arm of anti-cryptococcal immunity (reviewed in [1,7,8]), this review will focus on our current knowledge of innate immune responses to the species C. neoformans and identify significant questions that remain to be investigated.

2. Animal Models of Cryptococcosis

Different vertebrate and invertebrate animal models have been utilized in the study of cryptococcosis (for more comprehensive reviews see [1,9,10]). Predominantly, murine models have been used to study innate immune responses to C. neoformans due to the relative ease of genetic modification, manipulation and maintenance of this mammalian host. Therefore, results from mouse studies will comprise the majority of this review.
The use of mouse models of cryptococcosis does have its challenges. Different mouse strains develop different T helper cell (Th) responses to C. neoformans; mice that develop Th type 2 (Th2) responses are more susceptible to cryptococcosis, while those that develop Th type 1 (Th1) responses are more resistant [11,12,13,14,15]. Mouse susceptibility can further vary depending on the virulence of the C. neoformans strain, the type and amount of infectious propagule (i.e., spore versus yeast form), and the route of administration [11,12,13,14,16,17,18]. C. neoformans has two main variants: var. grubii (Serotype A), which is the most common clinical isolate, and var. neoformans (Serotype D) [1]. The most physiologic route of infection is through the respiratory tract, either intranasal or intratracheal. However, respiratory infection in mice can result in variable dissemination to the CNS, so systemic infection (intravenous or intraperitoneal) and direct inoculation into the cerebrospinal fluid have been used to study the pathology of C. neoformans in the CNS [13,19,20].
As an example of the differences between mouse models of cryptococcosis, respiratory infection of C57BL/6 mice with the highly virulent serotype A strain H99 leads to a Th2-skewed immune response that results in an acute and uniformly fatal infection [21,22,23]. On the other hand, respiratory infection of BALB/c mice with a less virulent serotype D strain like 52D leads to a Th1-skewed immune response that results in a more chronic infection that can eventually be cleared in a CD4+ T-cell-dependent manner [11,12,15,24]. A protective model of pulmonary cryptococcosis has also been established in which mice are infected with a C. neoformans strain H99-γ, that has been modified to express murine interferon gamma (IFNγ) [25].

3. Host Recognition of Cryptococcus

Fungal pathogens are typically sensed through the detection of fungal antigens, or pathogen-associated molecular patterns (PAMPs), by pattern recognition receptors (PRRs) on host immune cells. Engagement of PRRs induces signal transduction that coordinates innate immune processes like phagocytosis and cytokine production. Common fungal PAMPs include components of the cell wall, such as β-glucans, mannans, and chitin. However, C. neoformans provides an interesting challenge due to its polysaccharide capsule that can mask these potential PAMPs. Correspondingly, many PRRs that are known to detect other fungal pathogens, including members of the C-type lectin receptor (CLR) and Toll-like receptor (TLR) families, do not have similar roles in the recognition of C. neoformans. Therefore, the mechanisms by which C. neoformans is sensed by the host are still not fully defined.

3.1. C-Type Lectin Receptors

The CLRs are a large family of receptors that can recognize fungal carbohydrate ligands like β-glucans or mannans. An engaged CLR typically initiates downstream signaling pathways either through its own intracellular signaling domain, if present, or else through signaling adapters that contain an immunoreceptor tyrosine-based activation motif (ITAM), such as Fc receptor γ-chain (FcRγ or FcεRIγ chain) or DNAX activation protein of 12 kDa (DAP12). While CLRs have established roles in host innate immune responses to other pathogenic fungi (reviewed in [26]), their ability to mediate immunity to C. neoformans is less robust. There is evidence that β-glucans can be accessible on encapsulated yeast [27] and spore [28] forms of C. neoformans and that the fungal cell wall can be exposed at daughter bud sites prior to capsule assembly [29], but it is likely that the capsule is interfering with many of these potential interactions in vivo [30,31].
Mannose receptor (MR/CD206) binds to fucose and terminal mannose moieties and is known to have roles in phagocytosis as well as antigen processing and presentation as a receptor of the endocytic pathway (reviewed in [32]). MR does not have any known intracellular signaling motifs and can also exist in a soluble form [32,33], suggesting it may work in concert with other receptors for signal transduction, such as TLR2 [34]. Human MR has been shown to bind cryptococcal mannoproteins in vitro [35]. It is unclear if MR binds whole cryptococcal cells since MR-deficient murine phagocytes had no changes in binding and uptake of spores or yeast cells compared to wild-type (WT) phagocytes by microscopy [36]. Nevertheless, MR−/− mice challenged with C. neoformans in an acute respiratory infection model appear to have a moderate increase in fungal burden and susceptibility to infection [37]. Several studies have investigated the ability of MR to facilitate the priming of adaptive T cell responses by dendritic cells (DCs). MR-deficient bone marrow-derived DCs (BMDCs) from mice had no changes in uptake of cryptococcal mannoproteins and no differences in expression of maturation markers like MHCII, CD40 and CD86 [37]. In contrast, studies with human cells indicate that blocking MR can inhibit maturation marker expression by DCs in response to cryptococcal mannoproteins [38] and can inhibit fungal uptake by DCs and subsequent lymphocyte proliferation [39]. Therefore, the mechanisms by which MR mediates innate immune responses to C. neoformans warrants continued investigation.
Dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN/CD209) binds fucose and mannose residues and is involved in antigen uptake as well as cellular adhesion (reviewed in [40]). Complicating its study, DC-SIGN has eight homologs in mice, designated DC-SIGN-related proteins (SIGNR) 1-8. SIGNR3 (CD209d) is considered the closest homolog to human DC-SIGN [41], and SIGNR3 and SIGNR1 (CD209b) are the only homologs shown to bind fungal ligands [42]. Human DC-SIGN binds cryptococcal mannoproteins in vitro [43], but murine SIGNR1 does not influence the ability of splenic macrophages to internalize the cryptococcal capsular polysaccharide glucuronoxylomannan (GXM) [44]. Additional studies on the potential role of DC-SIGN or its murine homologs during cryptococcal infection are currently lacking.
Collectins are secreted carbohydrate-binding proteins and include the lung surfactant proteins (SPs) SP-A and SP-D and serum mannose binding lectin (MBL), also referred to as mannose binding protein (MBP). Collectins have been shown to engage with various fungal pathogens [45,46,47,48] and regulate cytokine responses by binding to cell surface receptors like CD14, TLR2 and TLR4 [49,50]. Interestingly, SP-D appears to be detrimental to the host, as SP-D−/− mice have improved survival after infection with C. neoformans [51]. SP-D binds to and protects C. neoformans from macrophage killing, and its activity has been correlated with increased IL-5 production and pulmonary eosinophilia [29,52]. The cryptococcal PAMP recognized by SP-D in vivo is unclear. In vitro, SP-D can bind to capsular GXM and mannoprotein 1 (MP1), but has higher affinity to pustulan, an analog of β-1,6-glucan found in the cryptococcal cell wall [29]. This higher affinity for a cell wall component correlates with the observation that acapsular C. neoformans mutants are more susceptible to agglutination and phagocytosis in the presence of SP-D compared to encapsulated strains [29,53,54]. Further studies are needed to determine which interactions and signaling mechanisms are essential for the harmful effects of SP-D on the host response. In contrast, SP-A can bind to C. neoformans but does not affect phagocytosis [55] and does not regulate murine susceptibility to infection [56]. MBL is known to bind mannose and N-acetylglucosamine (GlcNAc) and has been shown to act as an opsonin for complement activation [57]. However, soluble human MBL can only bind acapsular C. neoformans and minimally improves phagocytosis of these fungal cells by human polymorphonuclear cells in vitro [30,54,58,59]. Thus, the overall role of collectins in anti-cryptococcal responses appears to be minimal or else harmful to the host.
Other CLRs have been investigated but do not appear to have links to anti-cryptococcal immunity. Dectin-1 (CLEC7A) does not mediate immune responses in vitro or in vivo to either yeast or spore forms of C. neoformans [36,60]. Co-expression of Dectin-1 and TLR2 in vitro also does not facilitate signal transduction in response to the fungus [61]. Dectin-2 (CLEC6A/CLEC4N) is not essential in host defense against C. neoformans yeast or spore forms despite molecular evidence of increased Th2 and decreased Th1 responses in Dectin-2−/− mice [36,62]. Dectin-3 or macrophage C-type lectin (MCL/CLEC4D/CLECSF8) does not regulate murine outcomes after C. neoformans infection or phagocytosis of fungal cells [63,64] and cannot initiate signal transduction in response to C. neoformans spores [36]. Macrophage inducible C-type lectin (Mincle) does not bind C. neoformans or induce signal transduction in response to the fungus in vitro [36]. Langerin (CD207) does not bind to either encapsulated or acapsular C. neoformans [65]. Work remains to determine whether other CLRs, including novel receptors like CD23/FcεRII [66], may play a role in host recognition of C. neoformans.

3.2. Toll-Like Receptors

The potential role of TLRs as cryptococcal PRRs has been supported by evidence that myeloid differentiation primary response gene 88 (MyD88), a signaling molecule downstream of most TLRs, plays a role in murine anti-cryptococcal responses [67,68,69]. However, direct experimental evidence supporting a role for many of the TLRs in cryptococcosis is limited. Whether TLR signaling is relevant to human disease is unclear, as people with Mendelian defects in MyD88 do not have increased susceptibility to cryptococcosis [70,71].
Studies on TLR2 have had conflicting results regarding the ability of this receptor to influence infectious outcomes and to initiate signal transduction in response to C. neoformans, perhaps related to differences in experimental design. Biondo et al. demonstrated that TLR2−/− mice have increased susceptibility to systemic (intraperitoneal) infection with C. neoformans, as measured by survival, organ fungal burden and cytokine production [67]. Yauch et al. found that TLR2−/− mice have increased susceptibility to respiratory infection but not systemic (intravenous) infection; however, there were no differences in lung fungal burden or cytokine production in the TLR2−/− mice compared to WT mice [68]. Nakamura et al. also found no differences in fungal burden or cytokine production in TLR2−/− mice infected through the respiratory tract, and C. neoformans did not induce nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation through TLR2 in an in vitro cell reporter assay, even with co-expression of Dectin-1 [61].
TLR4, in conjunction with its co-receptor CD14, can respond to cryptococcal GXM in vitro by inducing NF-κB but not mitogen activated protein (MAP) kinase pathways or tumor necrosis factor alpha (TNFα) secretion, suggesting incomplete activation [72]. Monoclonal antibodies against TLR4 can inhibit Fas ligand expression [73] and partially block GXM uptake by human peripheral blood mononuclear cell (PBMC)-derived macrophages [74]. However, TLR4 has not been shown to regulate murine susceptibility to infection [67,68].
The strongest evidence for direct TLR involvement in anti-cryptococcal responses is for TLR9, an intracellular receptor of the endocytic pathway that typically recognizes unmethylated cytosine-phosphate-guanine (CpG) motifs common in the DNA of bacteria and viruses (reviewed in [75,76]). More recently, the fungus Aspergillus fumigatus was found to contain unmethylated CpG motifs that can stimulate cytokine responses by DCs in vitro in a TLR9-dependent manner [77]. Several groups have also used synthetic CpG-oligodeoxynucleotides to boost the immune response against C. neoformans [78,79,80,81]. TLR9−/− mice are more susceptible to cryptococcosis, potentially due to decreased recruitment and maturation of DCs and the development of Th2 immune responses, including alternative activation of macrophages [69,82,83,84]. Cryptococcal DNA can stimulate in vitro cytokine responses by DCs, which can be partially inhibited by deletion of TLR9 or MyD88 [84]. Subsequently, it has been shown that polymerase chain reaction (PCR) products amplified from cryptococcal genes involved in virulence including URA5, CNLAC1, and CAP59 can induce the same cytokine responses by DCs [85]. Interestingly, these genes do not contain canonical CpG motifs. Thus, cryptococcal DNA can function as a PAMP for TLR9, but the specific nucleic acid motifs involved in its recognition have not been elucidated.

3.3. Nucleotide-Binding Oligomerization Domain (NOD)-Like Receptors

The NOD-like receptors, or nucleotide-binding domain leucine-rich repeat-containing receptors (NLRs), are a family of cytoplasmic receptors that, upon activation, can form an inflammasome complex that cleaves and activates pro-IL-1β and pro-IL-18 generated after initial microbial detection induces NF-κB (reviewed in [86]). NLR family, pyrin domain-containing 3 (NLRP3) is an NLR that has been shown to play a role in immunity against A. fumigatus and the yeast Candida albicans [87,88,89], although the ligand for NLRP3 remains unidentified. Biofilms of encapsulated C. neoformans, opsonized and encapsulated C. neoformans, and acapsular yeast forms of C. neoformans stimulate formation of the NLRP3 inflammasome, and mice deficient in components of the NLRP3 inflammasome are more susceptible to infection [90,91,92]. However, additional studies will be needed to further clarify the role of NLRs and inflammasome formation in antifungal responses to C. neoformans.

3.4. Scavenger Receptors

Scavenger receptors are classically known to bind and internalize oxidized low-density lipoproteins. In more recent years, they have been found to have very diverse ligands and can serve as PRRs that detect microbial PAMPs and complex with other receptors like TLRs (reviewed in [93]). In vitro studies indicate that the scavenger receptors CD36 and scavenger receptor class F member 1 (SCARF1), also known as scavenger receptor expressed by endothelial cells 1 (SREC1), can bind to and internalize encapsulated C. neoformans, thereby inducing cytokine responses that can further be enhanced by synergy with TLR2; competition assays suggest that CD36 and SCARF1 may bind to β-glucans and, to a lesser extent, mannan, although they do not contain classic lectin-binding domains [94]. In the same study, neutralizing anti-SCARF1 antibody inhibited binding of C. neoformans to alveolar macrophages in vivo, CD36−/− mice were found to be more susceptible to systemic infection with C. neoformans, and deletion of CD36 and SCARF1 orthologues in the nematode Caenorhabditis elegans resulted in increased susceptibility to fungal challenge.
Macrophage receptor with collagenous structure (MARCO) has been shown to enhance early lung recruitment of monocyte-derived immune cells and protective cytokine responses after murine respiratory infection with C. neoformans that correlate with a transient improvement in fungal clearance [95]. Interestingly, MARCO-deficient macrophages and DCs exhibit no defect in fungicidal activity though they do have decreased interactions with fungal cells [95].
Scavenger receptor A (SRA/SR-AI/II/CD204/SCARA1) has been reported to have detrimental effects on host immunity to C. neoformans. SRA−/− mice have decreases in lung fungal burden likely related to regulation of cytokine responses that influence innate immune cell recruitment and activation [96]. The potential cryptococcal ligands for SRA and MARCO and additional mechanistic details for how all these scavenger receptors influence anti-cryptococcal responses have not yet been determined.

3.5. Natural Antibodies

Natural antibodies, that are predominantly of the immunoglobulin M (IgM) isotype, are constitutively produced in mammalian hosts by an innate subset of B lymphocytes called B-1 cells; opsonization of microbial antigens with natural IgM can result in complement activation, phagocytosis by macrophages, and priming of adaptive immune responses (reviewed in [97,98]). It has been shown that IgM produced by murine B-1 cells in vitro can bind to cell wall laminarin, capsular GXM, and acapsular and heat-killed encapsulated C. neoformans [99]. Secretory IgM-deficient (sIgM−/−) mice have increased susceptibility to respiratory infection with C. neoformans compared to control mice and exhibit defects in Th1 polarization and phagocytosis of fungi by alveolar macrophages; the defect in phagocytosis can be ameliorated by administration of IgM into the lungs [100]. Additionally, depletion of B-1 cells in pulmonary infected mice increases fungal burden and decreases phagocytosis of fungal cells by alveolar macrophages compared to non-depleted controls; adoptive transfer of B-1 cells into depleted mice can restore the phenotype to that of control mice [99]. On the other hand, sIgM−/− mice infected systemically with C. neoformans have improved survival compared to control mice [101]. It was found that these sIgM−/− mice have an increased baseline number of B-1 cells [101], and, interestingly, B-1 cell derivatives may have direct fungicidal effects against C. neoformans [102]. Thus, IgM and B-1 cells may play different roles in the anti-cryptococcal response depending on the tissue compartment. X-linked immunodeficient (XID) mice, that have a defect in B cell development and IgM production due to a mutation in Bruton’s tyrosine kinase (Btk), exhibit increased susceptibility to both respiratory and systemic infection with C. neoformans [6,103]. However, adoptive transfer of B-1 cells into pulmonary infected XID mice could neither reverse this susceptibility to C. neoformans nor fully restore serum IgM levels, suggesting that B-1 cells may not be the only source of protective IgM or that additional immune mechanisms are contributing to the phenotype in this particular model [6].
Human studies support a role for IgM in protective immune responses against C. neoformans. The percentage of IgM-expressing memory B cells inversely correlates with the risk for developing cryptococcosis among HIV-positive patients [104]. In solid organ transplant recipients, pre-transplantation levels of GXM-reactive IgM inversely correlate with the development of post-transplant cryptococcosis [105]. The ability to identify B-1 cells in humans has recently been reported (reviewed in [106]), which may facilitate future studies on the role of these innate immune cells and natural antibodies in human cryptococcosis.

3.6. Complement and Other Soluble Mediators

The complement system is an important mediator for the phagocytosis of C. neoformans by innate immune cells. Opsonization by complement has been shown to improve uptake and killing of C. neoformans by phagocytes [107,108] and to mediate DC responses to C. neoformans [109]. Activation of complement can occur through three pathways: alternative, classical, and lectin (reviewed in [110]). Disruption of the alternative, but not the classical, pathway of complement reduces phagocytosis of C. neoformans in vitro and increases the mortality of guinea pigs after infection [111,112]. The lectin pathway likely does not play a significant role given minimal interactions between MBL and C. neoformans, as discussed earlier in this review. It has been shown that complement component 3 (C3) binds to the capsule of C. neoformans and then is degraded to inactivated C3b (iC3b) [113,114,115]. Phagocytosis can then proceed via the action of complement receptors (CR). Blocking CR1, CR3 and CR4 decreases the interaction between C. neoformans and human macrophages in vitro [108]. CR3 has been shown to facilitate complement-mediated phagocytosis of C. neoformans by murine macrophages [116], but CR3 and CR4 can also mediate phagocytosis independent of complement [117]. Additionally, signaling by C5a through its receptor C5aR appears to be important for neutrophil uptake and killing of C. neoformans in mice [118].
Other potential soluble mediators of anti-cryptococcal immunity have been studied. Pentraxin 3 (PTX3) expression is induced in the brains of mice infected intracerebrally with C. neoformans [119], but it is not yet known what function PTX3 may play in anti-cryptococcal responses. Recombinant rat ficolin-A can bind and facilitate uptake of acapsular mutants of C. neoformans by lung epithelial cells in vitro but does not bind encapsulated C. neoformans [120], so it is unclear if ficolins play any significant role in anti-cryptococcal immunity. Finally, production of antimicrobial peptides is increased in a protective model of cryptococcosis [121], but their specific functions in the response to C. neoformans are not understood.

3.7. Other Recognition Pathways

Additional potential cryptococcal PAMPs have been identified, but their receptors remain unclear. Chitin is a long chain polymer of GlcNAc that can also be deacetylated to chitosan; both forms are components of the cryptococcal cell wall [1] and appear to have detrimental effects on the host immune response upon recognition. The chitin content of cryptococcal cells has been shown to correlate with Th2 cell accumulation and increased mortality in the murine host [122], and a chitosan-deficient strain of C. neoformans promotes protective Th1 host responses and is avirulent in mice [123]. Cryptococcal chitin has been shown to induce IL-10 secretion from human and murine macrophages [124] and induce Th2 responses through CD11b+ conventional DCs, although this process does not seem to occur through direct sensing of chitin by DCs [122]. The PRRs for chitin and chitosan are still unknown (reviewed in [125]). Studies using C. albicans-derived chitin suggest that chitin recognition is dependent on MR, NOD2, and TLR9 [124], and purified chitosan can induce inflammasome activation [126,127]. The hypervirulent rim101Δ C. neoformans mutant, that has increased chitosan content and exposure of chito-oligomers on its cell surface, induces TNFα secretion by murine bone marrow-derived macrophages but not IL-1β, suggesting cryptococcal chitosan does not induce the inflammasome; however, the induction of TNFα appears to be dependent on the caspase recruitment domain-containing 9 (CARD9) and MyD88 signaling molecules, indicating a potential role for CLRs and TLRs [128,129,130].
Another possible source of cryptococcal PAMPs are extracellular vesicles (EVs), also referred to as exosomes, which are bilayer vesicles released by C. neoformans [131] and can contain an array of cellular components including polysaccharides, nucleic acids, and proteins (reviewed in [132,133]). Although some EVs may be able to promote the virulence of C. neoformans [134,135], cryptococcal EVs have also been shown to be internalized by macrophages and stimulate cytokine secretion, NO production, and uptake and killing of the fungus in vitro [136]. As we improve our technical capability to isolate extracellular vesicles, it will be interesting to perform further analysis of their contents under different host conditions and determine if there are specific EV-borne PAMP interactions with host PRRs.

4. Intracellular Signaling Molecules

Another approach to defining innate immune responses to C. neoformans has been to study the role of molecules that commonly integrate signals from PRRs after fungal recognition. These include CARD9, MyD88, and the signaling adapters DAP12 and FcRγ (reviewed in [137,138]).
CARD9 is best known as a downstream mediator of signaling through CLRs like Dectin-1, but it can also transmit signals from TLRs and NOD2 and can facilitate activation of NF-κB or MAP kinase pathways (reviewed in [139,140]). Respiratory infection of CARD9−/− mice with C. neoformans results in increased lung fungal burden and neutrophilia along with defective early IFNγ production by NK cells and memory T cells [141]. CARD9 may not play a direct role in phagocytic pathways, as CARD9-deficient murine phagocytes have no defect in binding or uptake of C. neoformans spores or yeast forms as evaluated by microscopy [36], but it may regulate cytokine responses. For example, TNFα production is reduced in CARD9-deficient murine macrophages in response to the chitosan-enriched, acapsular rim101Δ cap59Δ C. neoformans mutant [128]. Together, these studies suggest that CARD9 may play a role in the host response to Cryptococcus, but the signaling pathway requires further definition.
MyD88 has well-established roles in signal transduction for most TLRs but can also function downstream of the cytokine receptors IL-1R and IL-18R [142,143]. MyD88−/− mice have increased susceptibility to both systemic and respiratory infection with C. neoformans [67,68,69]. Since the increased susceptibility of TLR2−/− mice to cryptococcosis is not as pronounced as that of MyD88−/− mice [67,68], MyD88 may mediate non-TLR signaling in response to C. neoformans as well. Indeed, IL-18R−/− mice, but not IL-1R−/− mice, have increased susceptibility to respiratory infection with C. neoformans, and knockout of either receptor causes significant changes in lung cytokine production compared to WT mice [69]. Additionally, mice deficient in IL-18 have increased susceptibility to cryptococcosis [144,145]. Thus, MyD88 may integrate signals from multiple cryptococcal recognition pathways during the host innate immune response.
DAP12 is an ITAM-containing signaling adapter that pairs to a variety of carbohydrate- and protein-binding immunoreceptors on myeloid and NK cells, including CLRs and other tyrosine kinase-signaling receptors (reviewed in [138,146,147,148]). DAP12 has been shown to have roles in the regulation of macrophage activation and survival [149,150]. Interestingly, DAP12-deficient macrophages have enhanced fungal uptake and killing and TNFα production in response to C. neoformans, and DAP12−/− mice are more resistant to respiratory infection with C. neoformans than WT mice [21]. Thus, DAP12 appears to inhibit beneficial fungicidal macrophage responses to C. neoformans. Further research will be needed to identify the DAP12-associated PRRs that trigger these immunosuppressive effects and could be potential immunomodulatory targets for the treatment of cryptococcosis.
FcRγ is also an ITAM-containing signaling adapter utilized by receptors on myeloid and NK cells (reviewed in [138]). In contrast to DAP12, there is no current evidence that supports a role for FcRγ in innate immune responses to C. neoformans. Murine phagocytes from FcRγ−/− mice demonstrate no changes in binding or uptake of spores or yeast [36]. Any other potential roles of FcRγ during cryptococcosis are still unknown.
Additional important signaling molecules in fungal sensing pathways, including spleen tyrosine kinase (Syk), have not yet been investigated for their roles in cryptococcosis. As these gaps in our knowledge are filled, we may gain further insight into the signaling network that enables coordination of the innate immune response by effector cells.

5. Effector Functions of Innate Immune Cells

After a fungal pathogen is recognized by the innate immune system, signal transduction coordinates the effector functions of innate immune cells, which may include phagocytosis and the generation of inflammatory response mediators such as cytokines, fungicidal compounds and acute phase reactants. These processes can regulate clearance of the fungus or initiate the development of adaptive immune responses. In the case of C. neoformans, these pathways can also be subverted by the pathogen to suppress the host innate immune response and allow the fungus to proliferate instead.

5.1. Inflammatory Monocytes

Inflammatory monocytes are innate immune cells that are recruited from the bone marrow to sites of infection or inflammation, whereupon they can differentiate into macrophages or DCs [151,152,153]. Although monocytes from HIV-positive patients have been reported to have impaired chemotaxis and cytotoxicity [154,155], studies using human monocytes and macrophages have had conflicting results about the role of these cells during cryptococcosis. Some researchers have found that human PBMCs can kill C. neoformans in vitro [156,157,158], and blood monocyte deactivation was associated with early mortality in HIV-associated cryptococcal meningitis [159]. In other studies, human PBMCs and monocyte-derived macrophages were merely fungistatic [160] or even permissive for intracellular cryptococcal proliferation and dissemination [161,162,163], and there was no difference in antifungal activity of monocyte-derived macrophages from cryptococcosis patients compared to normal controls [161].
In mice, inflammatory monocytes are defined as cells expressing lymphocyte antigen 6 complex, locus C1 (Ly6C) and C-C chemokine receptor type 2 (CCR2) that can migrate in response to the chemokines monocyte chemoattractant protein (MCP1), also known as C-C chemokine ligand 2 (CCL2), and CCL7 (reviewed in [152]). In chronic models of respiratory cryptococcosis, inflammatory monocytes appear to be beneficial to the host because CCR2−/− mice, that have a defect in monocyte recruitment, develop Th2 responses and have increased fungal burden and decreased lung macrophages, CD11b+ DCs and CD8+ T cells [164,165,166]. Further, in response to infection with C. neoformans, Ly6Chi CCR2+ monocytes differentiate into fungicidal exudative macrophages and CD11b+ DCs that promote fungal clearance and Th1 adaptive immune responses, respectively [166,167]. However, it is interesting to note that in an acute model of respiratory cryptococcosis, enhancing Th2 responses worsens survival and correlates with increased recruitment of monocytes to the lungs [122], suggesting that monocytes and their derivatives could play different roles depending on the host environment. This theory could potentially account for the differences observed in studies on human monocyte responses to C. neoformans.

5.2. Macrophages

Macrophages are phagocytic cells that include tissue-resident, embryonic-derived cells like lung alveolar macrophages as well as monocyte-derived macrophages that are of hematopoietic cell origin [168]. Since macrophages, in the guise of alveolar macrophages, are present in the lung at the time that C. neoformans is inhaled into the lungs, they have long been considered to be the first line innate immune cell in host defense against the fungus. Indeed, fungi are seen within lung macrophages in patients with cryptococcosis [169], and in murine models, alveolar macrophages have been visualized to quickly take up cryptococcal cells after respiratory infection [170,171]. However, there have been differing results regarding the ability of macrophages to clear C. neoformans from the host. While some groups have observed that murine macrophages can kill C. neoformans in vitro [172,173], others have found that the fungus can actually replicate within these cells, which may lead to dissemination by way of a Trojan Horse mechanism [170,174,175,176]. Interestingly, clinical C. neoformans isolates that exhibit higher rates of uptake by macrophages in vitro predict poor patient outcomes [177]. In murine respiratory infection models, depletion of macrophages using liposomal clodronate reduces fungal burden [176,178]. In contrast, ablation of macrophages, along with DCs, using transgenic CD11c-diphtheria toxin receptor (DTR) mice was found to worsen survival without any differences in lung fungal burden [179]. It is important to note that the ablation protocol for CD11c-DTR mice can induce fatal toxicity, even in the absence of any infection (reviewed in [180]). Thus, it will be necessary to confirm this result using alternative strategies.
It has become apparent that macrophage polarization may be a key determinant of whether macrophages are beneficial or detrimental during cryptococcosis. M1 (classically activated) macrophages produce nitric oxide (NO) through inducible NO synthase (iNOS) expression, secrete TNFα, and are fungicidal against C. neoformans, while M2 (alternatively activated) macrophages typically express the markers arginase 1 (Arg1), chitinase-like 4 (Chil4 or Ym2), resistin like alpha (Retnla or Fizz1), and MR (CD206) and are permissive for fungal growth (reviewed in [181]). M2 polarization has been associated with severe cryptococcal disease in non-HIV patients [182], though not in HIV-positive patients [159]. In mice, alternative activation of macrophages worsens cryptococcosis in the brain [183]. In a chronic respiratory infection model in mice, lung macrophages cycle from a resting state to an M2 phenotype, that corresponds with initial proliferation of C. neoformans in the lungs, followed by an M1 phenotype, that correlates to a period of fungal clearance, and then back to a resting state; this cycling could be simulated in vitro by modifying the cytokine environment with either IFNγ (M1) or IL-4 (M2) [184,185]. IFNγ−/− mice have increased lung fungal burden and demonstrate alternative activation of macrophages after pulmonary challenge with C. neoformans [185,186]. IL4−/− mice have improved fungal clearance and demonstrate classical activation of macrophages [185,187]. C. neoformans cells weakly stimulate expression of iNOS and Arg1 in murine macrophages in vitro, suggesting that direct interaction between fungus and phagocyte is not the only determinant of macrophage polarization [184].
From a therapeutic perspective, it will be helpful to further dissect the signaling mechanisms that can influence the polarization of macrophages during cryptococcosis. Various signaling components have been identified, including DAP12 [21], heat shock protein 70 (Hsp70) [188], and signal transducer and activator of transcription 1 (STAT1) [189,190]. Studies on other intracellular pathogens suggest that TLR signaling can induce Arg1 in macrophages [191]. Understanding these processes will allow testing of the idea that macrophage polarization drives infectious outcomes in mammalian hosts and could lay the foundation for potential new immunomodulatory strategies for the treatment of cryptococcosis.

5.3. Dendritic Cells

The primary function of DCs in antifungal responses is to take up, process and present antigens to prime T cells and trigger adaptive immunity (reviewed in [192,193,194]). DCs are a heterogeneous group of cells whose classification continues to evolve. Generally, it is recognized that the main subsets of DCs include classical or conventional DCs (cDCs), monocyte-derived DCs (moDCs), plasmacytoid DCs (pDCs), and Langerhans cells (reviewed in [151,195]).
DCs appear to have roles in protective immunity against C. neoformans. Ablation of DCs, along with macrophages, using CD11c-DTR mice increases murine mortality after infection [179], although there are limitations to this mouse model as mentioned previously in this review. DCs have been shown to take up and present cryptococcal glycoantigens [43]. Researchers have found that protective adaptive immune responses to cryptococcal antigen can be mediated by CD11b+ DCs and Langerhans cells [196], and moDCs have been shown to enhance Th1 responses after respiratory infection with C. neoformans [166]. Cryptococcal cells and cryptococcal antigen have been shown to stimulate IL-12 and IL-23p40 secretion and expression of activation markers by DCs in vitro [38,197]. DCs upregulate the CD80 activation marker in response to pulmonary C. neoformans challenge in vivo and can stimulate T cell activation ex vivo [198]. In addition, DCs can phagocytose and kill C. neoformans [39,109,198]. However, CD11b+ cDCs can also mediate harmful Th2 immune responses stimulated by chitin, as discussed earlier in this review [122].
The potential role of pDCs during cryptococcosis has not been as closely examined as that of cDCs and moDCs. C. neoformans does not appear to activate pDCs in vitro [197]. Other reports suggest that pDCs phagocytose C. neoformans and limit fungal growth through a Dectin-3 and ROS-dependent mechanism [63]. However, infectious outcomes are not altered in Dectin-3−/− mice [63,64].

5.4. Neutrophils

Neutrophils are granulocytes that can phagocytose microorganisms, release antimicrobial enzymes, and produce neutrophil extracellular traps (NETs) (reviewed in [137]). Neutrophils have established roles in the innate immune response to fungal pathogens like A. fumigatus [199], but their role in anti-cryptococcal immunity remains poorly defined. Human neutrophils can kill C. neoformans in vitro [157,200], and treatment of mice with human recombinant granulocyte-colony stimulating factor (G-CSF) in combination with fluconazole improves survival from intracerebral infection [201]. At the same time, C. neoformans can inhibit human neutrophil migration [202], and its capsule blocks neutrophil binding of fungal cells [203]. Human neutrophils release NETs in response to acapsular C. neoformans mutants and the capsular polysaccharide glucoronoxylomannogalactan (GXMGal) but not in response to encapsulated C. neoformans or capsular GXM [204]. However, if already formed, NETs can kill encapsulated C. neoformans [204].
In a systemic model of murine cryptococcosis, anti-Ly6G (1A8) antibody depletion of neutrophils suggests that these cells are needed for fungal clearance in the brain and lungs [205], and neutrophils have been visualized to swarm the fungus for removal from the brain microvasculature [206,207]. In a protective model of cryptococcosis, neutrophils are the primary source of IL-17A that enhances protective immune responses, although they are not essential as γδ T cells can produce IL-17A in their absence [208]. On the other hand, after pulmonary challenge with C. neoformans, depletion of neutrophils and inflammatory monocytes with anti-Gr-1 (RB6-8C5) antibody improves murine survival and causes an overall reduction in inflammatory lung damage, suggesting a detrimental role for neutrophils [209]. In the same study, treatment with anti-Gr-1 had no effect on murine survival after systemic infection. Further supporting a harmful role for neutrophils, mice with genetically-induced neutrophilia appear to have increased susceptibility to cryptococcal disease [210]. Therefore, the role of neutrophils in anti-cryptococcal responses is still not clear and may depend on the specific host and/or tissue environment.

5.5. Natural Killer Cells

NK cells are cytotoxic lymphocytes of the innate immune system. Studies in murine models of systemic cryptococcosis suggest that NK cells may participate in early anti-cryptococcal immune responses through direct fungal interactions [211,212,213,214,215,216]. Other groups find that instead of direct cytotoxic effects against C. neoformans, NK cells may enhance the fungicidal activity of macrophages in mice by producing IFNγ [217,218]. Mice lacking NK cells have increased fungal burden, but they do not have increased susceptibility to infection [211,213].
The role of NK cells in anti-cryptococcal responses has been more closely examined in human cells. NK cells from HIV-positive patients are impaired in their growth inhibition of C. neoformans [219]. Human lymphocytes and NK cells have been shown to inhibit cryptococcal growth through direct interaction [220,221]. In studies using human primary NK cells or cell lines, Mody and colleagues have demonstrated that binding of C. neoformans by NK cells leads to signaling through the PI3K-ERK1/2 pathway [222] and triggers perforin degranulation to facilitate cryptococcal killing [223]. The natural cytotoxicity receptor NKp30, an immunoglobulin-like protein, has been identified as a human NK cell PRR for C. neoformans [224]. In the same study, blocking NKp30 impaired PI3K-ERK1/2 signaling, perforin release and ultimately fungal killing in response to C. neoformans. Additionally, it was shown that NK cells from HIV patients have decreased expression of NKp30 and decreased toxicity against C. neoformans, both of which can be reversed by IL-12 treatment in vitro. Work remains to identify any additional cryptococcal PRRs on NK cells as well as the cryptococcal ligand for NKp30. Studies on the detection of Candida glabrata by the related receptor NKp46 suggest that fungal adhesins could be potential ligands for this class of receptors [225].

5.6. Eosinophils

Eosinophils are granulocytes that are best known for their roles in allergic responses and parasitic infections (reviewed in [226]). Eosinophilia has been associated with cryptococcal disease in humans and mice [11,227,228,229,230,231,232,233,234,235] and positively correlated to murine susceptibility to cryptococcosis [11,52], but it is not clear if eosinophils have an essential role in the innate immune response to C. neoformans or if their recruitment is the byproduct of an ineffectual Th2 response. After infection with C. neoformans, eosinophil-deficient ΔdblGATA mice have enhanced Th1 and Th17 responses and decreased lung recruitment of other inflammatory cells, although fungal burden in the lung and brain are not significantly different from WT mice [236]. It is interesting to note that in rats, eosinophils can phagocytose C. neoformans and prime T and B cells in order to generate Th1 responses that are protective for the host [233,237,238]. Therefore, the role of eosinophils during cryptococcosis may depend on the particular host setting.

5.7. Other Innate Immune Cells

Innate lymphoid cells (ILCs), other than NK cells, have not been extensively studied in cryptococcosis, but type 2 ILCs may be detrimental to host anti-cryptococcal responses [239]. Derivatives of B-1 cells may have direct antifungal effects against C. neoformans [102], as discussed earlier in this review. Epithelial and endothelial cells not only serve as a physical barrier to microbial invasion, but can also participate as effector innate immune cells (reviewed in [240,241]). Lung epithelial cells can bind C. neoformans and produce cytokines in response to the fungus [242,243], and endothelial cells may enhance anti-cryptococcal activity of neutrophils [244]. The potential role of γδ T cells is still unclear. Mice deficient in γδ T cells have improved infectious outcomes after C. neoformans challenge [245], but studies in a protective model of cryptococcosis suggest that γδ T cells are a source of beneficial IL-17A in the setting of neutropenia [208].

6. Conclusions

By methodically investigating common mammalian antifungal mechanisms, researchers have established important roles for cellular PRRs, in particular MR, TLR9, and NKp30, and for signal transduction through CARD9 and MyD88 in protective immune responses against C. neoformans. Other promising PRR candidates include NLRs like NLRP3 and certain scavenger receptors. Furthermore, soluble mediators including natural IgM and complement have key functions in facilitating host recognition and immunity to C. neoformans. Many additional signaling pathways have been studied, but they either require further evaluation as to their specific anti-cryptococcal functions or appear to have limited or even detrimental roles in host responses to C. neoformans. Whether the limited findings are due to redundancies in the immune system remains to be determined [246]. Several innate immune cell types appear to have effector functions that facilitate C. neoformans clearance and prime adaptive immune responses under certain conditions, but the mechanisms that coordinate these processes require further definition. Much of the work on anti-cryptococcal immunity has been performed in vitro, so it will be important to confirm these pathways in vivo and in human hosts, when possible.
Since C. neoformans is equipped with unique virulence factors, like its polysaccharide capsule, that enable it to evade or subvert the host immune response [1], it is not unexpected that the fungus would stimulate distinct innate immune responses compared to other fungal pathogens. Thus, while it is important to study the potential roles of established antifungal pathways in the response to cryptococcosis, it is also critical to work towards identifying immune mechanisms that may be specific to C. neoformans. Identification of additional patient populations susceptible to cryptococcosis, such as those with anti-granulocyte macrophage colony-stimulating factor (GM-CSF) autoantibodies [247,248], may reveal previously unknown immune processes important for the host response to C. neoformans. Additionally, the rise of new bioinformatics approaches like next-generation sequencing [249] and tools like CRISPR-Cas gene editing [250] and fluorescent probes [251] may enable the discovery of novel pathways in anti-cryptococcal immunity.

Acknowledgments

The author thanks Tobias Hohl and Bing Zhai for helpful comments and regrets that the valuable contributions of many other researchers in the field could not be included due to space constraints. Lena J. Heung is funded by a National Institutes of Health (NIH) grant K08 AI130366, a Stony Wold-Herbert Fund Fellowship, and the Dana Foundation as a Memorial Sloan Kettering Cancer Center Clinical Scholar in Biomedical Research. This work was funded in part through the NIH/National Cancer Institute (NCI) Cancer Center Support Grant P30 CA008748.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Heitman, J.; Kozel, T.R.; Kwon-Chung, K.J.; Perfect, J.R.; Casadevall, A. Cryptococcus: From Human Pathogen to Model. Yeast, 1st ed.; ASM Press: Washington, DC, USA, 2010; pp. 1–646. [Google Scholar]
  2. Park, B.J.; Wannemuehler, K.A.; Marston, B.J.; Govender, N.; Pappas, P.G.; Chiller, T.M. Estimation of the current global burden of cryptococcal meningitis among persons living with HIV/AIDS. AIDS 2009, 23, 525–530. [Google Scholar] [CrossRef] [PubMed]
  3. Rajasingham, R.; Smith, R.M.; Park, B.J.; Jarvis, J.N.; Govender, N.P.; Chiller, T.M.; Denning, D.W.; Loyse, A.; Boulware, D.R. Global burden of disease of HIV-associated cryptococcal meningitis: An updated analysis. Lancet Infect. Dis. 2017. [Google Scholar] [CrossRef]
  4. Day, J.N.; Chau, T.T.; Wolbers, M.; Mai, P.P.; Dung, N.T.; Mai, N.H.; Phu, N.H.; Nghia, H.D.; Phong, N.D.; Thai, C.Q.; et al. Combination antifungal therapy for cryptococcal meningitis. N. Engl. J. Med. 2013, 368, 1291–1302. [Google Scholar] [CrossRef] [PubMed]
  5. Bratton, E.W.; El Husseini, N.; Chastain, C.A.; Lee, M.S.; Poole, C.; Sturmer, T.; Juliano, J.J.; Weber, D.J.; Perfect, J.R. Comparison and temporal trends of three groups with cryptococcosis: HIV-infected, solid organ transplant, and HIV-negative/non-transplant. PLoS ONE 2012, 7, e43582. [Google Scholar] [CrossRef]
  6. Szymczak, W.A.; Davis, M.J.; Lundy, S.K.; Dufaud, C.; Olszewski, M.; Pirofski, L.A. X-linked immunodeficient mice exhibit enhanced susceptibility to Cryptococcus neoformans Infection. MBio 2013, 4. [Google Scholar] [CrossRef] [PubMed]
  7. Rohatgi, S.; Pirofski, L.A. Host immunity to Cryptococcus neoformans. Future Microbiol. 2015, 10, 565–581. [Google Scholar] [CrossRef] [PubMed]
  8. Price, M.S.; Perfect, J.R. Host defenses against cryptococcosis. Immunol. Investig. 2011, 40, 786–808. [Google Scholar] [CrossRef] [PubMed]
  9. Carroll, S.F.; Guillot, L.; Qureshi, S.T. Mammalian model hosts of cryptococcal infection. Comp. Med. 2007, 57, 9–17. [Google Scholar] [PubMed]
  10. Sabiiti, W.; May, R.C.; Pursall, E.R. Experimental models of cryptococcosis. Int. J. Microbiol. 2012, 2012, 626745. [Google Scholar] [CrossRef] [PubMed]
  11. Huffnagle, G.B.; Boyd, M.B.; Street, N.E.; Lipscomb, M.F. IL-5 is required for eosinophil recruitment, crystal deposition, and mononuclear cell recruitment during a pulmonary Cryptococcus neoformans infection in genetically susceptible mice (C57BL/6). J. Immunol. 1998, 160, 2393–2400. [Google Scholar] [PubMed]
  12. Zaragoza, O.; Alvarez, M.; Telzak, A.; Rivera, J.; Casadevall, A. The relative susceptibility of mouse strains to pulmonary Cryptococcus neoformans infection is associated with pleiotropic differences in the immune response. Infect. Immun. 2007, 75, 2729–2739. [Google Scholar] [CrossRef] [PubMed]
  13. Rhodes, J.C.; Wicker, L.S.; Urba, W.J. Genetic control of susceptibility to Cryptococcus neoformans in mice. Infect. Immun. 1980, 29, 494–499. [Google Scholar] [PubMed]
  14. Carroll, S.F.; Lafferty, E.I.; Flaczyk, A.; Fujiwara, T.M.; Homer, R.; Morgan, K.; Loredo-Osti, J.C.; Qureshi, S.T. Susceptibility to progressive Cryptococcus neoformans pulmonary infection is regulated by loci on mouse chromosomes 1 and 9. Infect. Immun. 2012, 80, 4167–4176. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, G.H.; McNamara, D.A.; Hernandez, Y.; Huffnagle, G.B.; Toews, G.B.; Olszewski, M.A. Inheritance of immune polarization patterns is linked to resistance versus susceptibility to Cryptococcus neoformans in a mouse model. Infect. Immun. 2008, 76, 2379–2391. [Google Scholar] [CrossRef] [PubMed]
  16. Sukroongreung, S.; Kitiniyom, K.; Nilakul, C.; Tantimavanich, S. Pathogenicity of basidiospores of Filobasidiella neoformans var. neoformans. Med. Mycol. 1998, 36, 419–424. [Google Scholar] [CrossRef] [PubMed]
  17. Velagapudi, R.; Hsueh, Y.P.; Geunes-Boyer, S.; Wright, J.R.; Heitman, J. Spores as infectious propagules of Cryptococcus neoformans. Infect. Immun. 2009, 77, 4345–4355. [Google Scholar] [CrossRef] [PubMed]
  18. Kwon-Chung, K.J.; Edman, J.C.; Wickes, B.L. Genetic association of mating types and virulence in Cryptococcus neoformans. Infect. Immun. 1992, 60, 602–605. [Google Scholar] [PubMed]
  19. Chretien, F.; Lortholary, O.; Kansau, I.; Neuville, S.; Gray, F.; Dromer, F. Pathogenesis of cerebral Cryptococcus neoformans infection after fungemia. J. Infect. Dis. 2002, 186, 522–530. [Google Scholar] [CrossRef] [PubMed]
  20. Blasi, E.; Barluzzi, R.; Mazzolla, R.; Mosci, P.; Bistoni, F. Experimental model of intracerebral infection with Cryptococcus neoformans: Roles of phagocytes and opsonization. Infect. Immun. 1992, 60, 3682–3688. [Google Scholar] [PubMed]
  21. Heung, L.J.; Hohl, T.M. DAP12 inhibits pulmonary immune responses to Cryptococcus neoformans. Infect. Immun. 2016, 84, 1879–1886. [Google Scholar] [CrossRef] [PubMed]
  22. Olszewski, M.A.; Noverr, M.C.; Chen, G.H.; Toews, G.B.; Cox, G.M.; Perfect, J.R.; Huffnagle, G.B. Urease expression by Cryptococcus neoformans promotes microvascular sequestration, thereby enhancing central nervous system invasion. Am. J. Pathol. 2004, 164, 1761–1771. [Google Scholar] [CrossRef]
  23. Osterholzer, J.J.; Surana, R.; Milam, J.E.; Montano, G.T.; Chen, G.H.; Sonstein, J.; Curtis, J.L.; Huffnagle, G.B.; Toews, G.B.; Olszewski, M.A. Cryptococcal urease promotes the accumulation of immature dendritic cells and a non-protective T2 immune response within the lung. Am. J. Pathol. 2009, 174, 932–943. [Google Scholar] [CrossRef] [PubMed]
  24. Huffnagle, G.B.; Lipscomb, M.F. Pulmonary cryptococcosis. Am. J. Pathol. 1992, 141, 1517–1520. [Google Scholar] [PubMed]
  25. Wormley, F.L., Jr.; Perfect, J.R.; Steele, C.; Cox, G.M. Protection against cryptococcosis by using a murine γ interferon-producing Cryptococcus neoformans strain. Infect. Immun. 2007, 75, 1453–1462. [Google Scholar] [CrossRef] [PubMed]
  26. Vautier, S.; MacCallum, D.M.; Brown, G.D. C-type lectin receptors and cytokines in fungal immunity. Cytokine 2012, 58, 89–99. [Google Scholar] [CrossRef] [PubMed]
  27. Rachini, A.; Pietrella, D.; Lupo, P.; Torosantucci, A.; Chiani, P.; Bromuro, C.; Proietti, C.; Bistoni, F.; Cassone, A.; Vecchiarelli, A. An anti-β-glucan monoclonal antibody inhibits growth and capsule formation of Cryptococcus neoformans in vitro and exerts therapeutic, anticryptococcal activity in vivo. Infect. Immun. 2007, 75, 5085–5094. [Google Scholar] [CrossRef] [PubMed]
  28. Giles, S.S.; Dagenais, T.R.; Botts, M.R.; Keller, N.P.; Hull, C.M. Elucidating the pathogenesis of spores from the human fungal pathogen Cryptococcus neoformans. Infect. Immun. 2009, 77, 3491–3500. [Google Scholar] [CrossRef] [PubMed]
  29. Geunes-Boyer, S.; Oliver, T.N.; Janbon, G.; Lodge, J.K.; Heitman, J.; Perfect, J.R.; Wright, J.R. Surfactant protein D increases phagocytosis of hypocapsular Cryptococcus neoformans by murine macrophages and enhances fungal survival. Infect. Immun. 2009, 77, 2783–2794. [Google Scholar] [CrossRef] [PubMed]
  30. Cross, C.E.; Bancroft, G.J. Ingestion of acapsular Cryptococcus neoformans occurs via mannose and β-glucan receptors, resulting in cytokine production and increased phagocytosis of the encapsulated form. Infect. Immun. 1995, 63, 2604–2611. [Google Scholar] [PubMed]
  31. Fromtling, R.A.; Shadomy, H.J.; Jacobson, E.S. Decreased virulence in stable, acapsular mutants of Cryptococcus neoformans. Mycopathologia 1982, 79, 23–29. [Google Scholar] [CrossRef] [PubMed]
  32. Gazi, U.; Martinez-Pomares, L. Influence of the mannose receptor in host immune responses. Immunobiology 2009, 214, 554–561. [Google Scholar] [CrossRef] [PubMed]
  33. Taylor, P.R.; Gordon, S.; Martinez-Pomares, L. The mannose receptor: Linking homeostasis and immunity through sugar recognition. Trends Immunol. 2005, 26, 104–110. [Google Scholar] [CrossRef] [PubMed]
  34. Tachado, S.D.; Zhang, J.; Zhu, J.; Patel, N.; Cushion, M.; Koziel, H. Pneumocystis-mediated IL-8 release by macrophages requires coexpression of mannose receptors and TLR2. J. Leukoc. Biol. 2007, 81, 205–211. [Google Scholar] [CrossRef] [PubMed]
  35. Mansour, M.K.; Schlesinger, L.S.; Levitz, S.M. Optimal T cell responses to Cryptococcus neoformans mannoprotein are dependent on recognition of conjugated carbohydrates by mannose receptors. J. Immunol. 2002, 168, 2872–2879. [Google Scholar] [CrossRef] [PubMed]
  36. Walsh, N.M.; Wuthrich, M.; Wang, H.; Klein, B.; Hull, C.M. Characterization of C-type lectins reveals an unexpectedly limited interaction between Cryptococcus neoformans spores and Dectin-1. PLoS ONE 2017, 12, e0173866. [Google Scholar] [CrossRef] [PubMed]
  37. Dan, J.M.; Kelly, R.M.; Lee, C.K.; Levitz, S.M. Role of the mannose receptor in a murine model of Cryptococcus neoformans infection. Infect. Immun. 2008, 76, 2362–2367. [Google Scholar] [CrossRef] [PubMed]
  38. Pietrella, D.; Corbucci, C.; Perito, S.; Bistoni, G.; Vecchiarelli, A. Mannoproteins from Cryptococcus neoformans promote dendritic cell maturation and activation. Infect. Immun. 2005, 73, 820–827. [Google Scholar] [CrossRef] [PubMed]
  39. Syme, R.M.; Spurrell, J.C.; Amankwah, E.K.; Green, F.H.; Mody, C.H. Primary dendritic cells phagocytose Cryptococcus neoformans via mannose receptors and Fcγ receptor II for presentation to T lymphocytes. Infect. Immun. 2002, 70, 5972–5981. [Google Scholar] [CrossRef] [PubMed]
  40. Garcia-Vallejo, J.J.; van Kooyk, Y. The physiological role of DC-SIGN: A tale of mice and men. Trends Immunol. 2013, 34, 482–486. [Google Scholar] [CrossRef] [PubMed]
  41. Powlesland, A.S.; Ward, E.M.; Sadhu, S.K.; Guo, Y.; Taylor, M.E.; Drickamer, K. Widely divergent biochemical properties of the complete set of mouse DC-SIGN-related proteins. J. Biol. Chem. 2006, 281, 20440–20449. [Google Scholar] [CrossRef] [PubMed]
  42. Takahara, K.; Yashima, Y.; Omatsu, Y.; Yoshida, H.; Kimura, Y.; Kang, Y.S.; Steinman, R.M.; Park, C.G.; Inaba, K. Functional comparison of the mouse DC-SIGN, SIGNR1, SIGNR3 and langerin, C-type lectins. Int. Immunol. 2004, 16, 819–829. [Google Scholar] [CrossRef] [PubMed]
  43. Mansour, M.K.; Latz, E.; Levitz, S.M. Cryptococcus neoformans glycoantigens are captured by multiple lectin receptors and presented by dendritic cells. J. Immunol. 2006, 176, 3053–3061. [Google Scholar] [CrossRef] [PubMed]
  44. De Jesus, M.; Park, C.G.; Su, Y.; Goldman, D.L.; Steinman, R.M.; Casadevall, A. Spleen deposition of Cryptococcus neoformans capsular glucuronoxylomannan in rodents occurs in red pulp macrophages and not marginal zone macrophages expressing the C-type lectin SIGN-R1. Med. Mycol. 2008, 46, 153–162. [Google Scholar] [CrossRef] [PubMed]
  45. Madan, T.; Eggleton, P.; Kishore, U.; Strong, P.; Aggrawal, S.S.; Sarma, P.U.; Reid, K.B. Binding of pulmonary surfactant proteins A and D to Aspergillus fumigatus conidia enhances phagocytosis and killing by human neutrophils and alveolar macrophages. Infect. Immun. 1997, 65, 3171–3179. [Google Scholar] [PubMed]
  46. Lekkala, M.; LeVine, A.M.; Linke, M.J.; Crouch, E.C.; Linders, B.; Brummer, E.; Stevens, D.A. Effect of lung surfactant collectins on bronchoalveolar macrophage interaction with Blastomyces dermatitidis: Inhibition of tumor necrosis factor α production by surfactant protein D. Infect. Immun. 2006, 74, 4549–4556. [Google Scholar] [CrossRef] [PubMed]
  47. van Rozendaal, B.A.; van Spriel, A.B.; van de Winkel, J.G.; Haagsman, H.P. Role of pulmonary surfactant protein D in innate defense against Candida albicans. J. Infect. Dis. 2000, 182, 917–922. [Google Scholar] [CrossRef] [PubMed]
  48. Choteau, L.; Parny, M.; Francois, N.; Bertin, B.; Fumery, M.; Dubuquoy, L.; Takahashi, K.; Colombel, J.F.; Jouault, T.; Poulain, D.; et al. Role of mannose-binding lectin in intestinal homeostasis and fungal elimination. Mucosal Immunol. 2016, 9, 767–776. [Google Scholar] [CrossRef] [PubMed]
  49. Sano, H.; Chiba, H.; Iwaki, D.; Sohma, H.; Voelker, D.R.; Kuroki, Y. Surfactant proteins A and D bind CD14 by different mechanisms. J. Biol. Chem. 2000, 275, 22442–22451. [Google Scholar] [CrossRef] [PubMed]
  50. Ohya, M.; Nishitani, C.; Sano, H.; Yamada, C.; Mitsuzawa, H.; Shimizu, T.; Saito, T.; Smith, K.; Crouch, E.; Kuroki, Y. Human pulmonary surfactant protein D binds the extracellular domains of Toll-like receptors 2 and 4 through the carbohydrate recognition domain by a mechanism different from its binding to phosphatidylinositol and lipopolysaccharide. Biochemistry 2006, 45, 8657–8664. [Google Scholar] [CrossRef] [PubMed]
  51. Geunes-Boyer, S.; Beers, M.F.; Perfect, J.R.; Heitman, J.; Wright, J.R. Surfactant protein D facilitates Cryptococcus neoformans infection. Infect. Immun. 2012, 80, 2444–2453. [Google Scholar] [CrossRef] [PubMed]
  52. Holmer, S.M.; Evans, K.S.; Asfaw, Y.G.; Saini, D.; Schell, W.A.; Ledford, J.G.; Frothingham, R.; Wright, J.R.; Sempowski, G.D.; Perfect, J.R. Impact of surfactant protein D, interleukin-5, and eosinophilia on cryptococcosis. Infect. Immun. 2014, 82, 683–693. [Google Scholar] [CrossRef] [PubMed]
  53. van de Wetering, J.K.; Coenjaerts, F.E.; Vaandrager, A.B.; van Golde, L.M.; Batenburg, J.J. Aggregation of Cryptococcus neoformans by surfactant protein D is inhibited by its capsular component glucuronoxylomannan. Infect. Immun. 2004, 72, 145–153. [Google Scholar] [CrossRef] [PubMed]
  54. Schelenz, S.; Malhotra, R.; Sim, R.B.; Holmskov, U.; Bancroft, G.J. Binding of host collectins to the pathogenic yeast Cryptococcus neoformans: Human surfactant protein D acts as an agglutinin for acapsular yeast cells. Infect. Immun. 1995, 63, 3360–3366. [Google Scholar] [PubMed]
  55. Walenkamp, A.M.; Verheul, A.F.; Scharringa, J.; Hoepelman, I.M. Pulmonary surfactant protein A binds to Cryptococcus neoformans without promoting phagocytosis. Eur. J. Clin. Investig. 1999, 29, 83–92. [Google Scholar] [CrossRef]
  56. Giles, S.S.; Zaas, A.K.; Reidy, M.F.; Perfect, J.R.; Wright, J.R. Cryptococcus neoformans is resistant to surfactant protein A mediated host defense mechanisms. PLoS ONE 2007, 2, e1370. [Google Scholar] [CrossRef] [PubMed]
  57. Garred, P.; Genster, N.; Pilely, K.; Bayarri-Olmos, R.; Rosbjerg, A.; Ma, Y.J.; Skjoedt, M.O. A journey through the lectin pathway of complement-MBL and beyond. Immunol. Rev. 2016, 274, 74–97. [Google Scholar] [CrossRef] [PubMed]
  58. Levitz, S.M.; Tabuni, A.; Treseler, C. Effect of mannose-binding protein on binding of Cryptococcus neoformans to human phagocytes. Infect. Immun. 1993, 61, 4891–4893. [Google Scholar] [PubMed]
  59. van Asbeck, E.C.; Hoepelman, A.I.; Scharringa, J.; Herpers, B.L.; 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. [Google Scholar] [CrossRef] [PubMed]
  60. Nakamura, K.; Kinjo, T.; Saijo, S.; Miyazato, A.; Adachi, Y.; Ohno, N.; Fujita, J.; Kaku, M.; Iwakura, Y.; Kawakami, K. Dectin-1 is not required for the host defense to Cryptococcus neoformans. Microbiol. Immunol. 2007, 51, 1115–1119. [Google Scholar] [CrossRef] [PubMed]
  61. Nakamura, K.; Miyagi, K.; Koguchi, Y.; Kinjo, Y.; Uezu, K.; Kinjo, T.; Akamine, M.; Fujita, J.; Kawamura, I.; Mitsuyama, M.; et al. Limited contribution of Toll-like receptor 2 and 4 to the host response to a fungal infectious pathogen, Cryptococcus neoformans. FEMS Immunol. Med. Microbiol. 2006, 47, 148–154. [Google Scholar] [CrossRef] [PubMed]
  62. Nakamura, Y.; Sato, K.; Yamamoto, H.; Matsumura, K.; Matsumoto, I.; Nomura, T.; Miyasaka, T.; Ishii, K.; Kanno, E.; Tachi, M.; et al. Dectin-2 deficiency promotes Th2 response and mucin production in the lungs after pulmonary infection with Cryptococcus neoformans. Infect. Immun. 2015, 83, 671–681. [Google Scholar] [CrossRef] [PubMed]
  63. Hole, C.R.; Leopold Wager, C.M.; Mendiola, A.S.; Wozniak, K.L.; Campuzano, A.; Lin, X.; Wormley, F.L., Jr. Antifungal activity of plasmacytoid dendritic cells against Cryptococcus neoformans in vitro requires expression of Dectin-3 (CLEC4D) and reactive oxygen species. Infect. Immun. 2016, 84, 2493–2504. [Google Scholar] [CrossRef] [PubMed]
  64. Campuzano, A.; Castro-Lopez, N.; Wozniak, K.L.; Leopold Wager, C.M.; Wormley, F.L., Jr. Dectin-3 is not required for protection against Cryptococcus neoformans infection. PLoS ONE 2017, 12, e0169347. [Google Scholar] [CrossRef] [PubMed]
  65. De Jong, M.A.; Vriend, L.E.; Theelen, B.; Taylor, M.E.; Fluitsma, D.; Boekhout, T.; Geijtenbeek, T.B. C-type lectin langerin is a β-glucan receptor on human langerhans cells that recognizes opportunistic and pathogenic fungi. Mol. Immunol. 2010, 47, 1216–1225. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, X.; Guo, Y.; Jiang, C.; Chang, Q.; Zhang, S.; Luo, T.; Zhang, B.; Jia, X.; Hung, M.C.; Dong, C.; et al. JNK1 negatively controls antifungal innate immunity by suppressing CD23 expression. Nat. Med. 2017, 23, 337–346. [Google Scholar] [CrossRef] [PubMed]
  67. Biondo, C.; Midiri, A.; Messina, L.; Tomasello, F.; Garufi, G.; Catania, M.R.; Bombaci, M.; Beninati, C.; Teti, G.; Mancuso, G. MyD88 and TLR2, but not TLR4, are required for host defense against Cryptococcus neoformans. Eur J. Immunol. 2005, 35, 870–878. [Google Scholar] [CrossRef] [PubMed]
  68. Yauch, L.E.; Mansour, M.K.; Shoham, S.; Rottman, J.B.; Levitz, S.M. Involvement of CD14, Toll-like receptors 2 and 4, and MyD88 in the host response to the fungal pathogen Cryptococcus neoformans in vivo. Infect. Immun. 2004, 72, 5373–5382. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, J.P.; Lee, C.K.; Akalin, A.; Finberg, R.W.; Levitz, S.M. Contributions of the MyD88-dependent receptors IL-18R, IL-1R, and TLR9 to host defenses following pulmonary challenge with Cryptococcus neoformans. PLoS ONE 2011, 6, e26232. [Google Scholar] [CrossRef] [PubMed]
  70. Picard, C.; Casanova, J.L.; Puel, A. Infectious diseases in patients with IRAK-4, MyD88, NEMO, or IκBα deficiency. Clin. Microbiol. Rev. 2011, 24, 490–497. [Google Scholar] [CrossRef] [PubMed]
  71. Lanternier, F.; Cypowyj, S.; Picard, C.; Bustamante, J.; Lortholary, O.; Casanova, J.L.; Puel, A. Primary immunodeficiencies underlying fungal infections. Curr. Opin. Pediatr. 2013, 25, 736–747. [Google Scholar] [CrossRef] [PubMed]
  72. Shoham, S.; Huang, C.; Chen, J.M.; Golenbock, D.T.; Levitz, S.M. Toll-like receptor 4 mediates intracellular signaling without TNF-α release in response to Cryptococcus neoformans polysaccharide capsule. J. Immunol. 2001, 166, 4620–4626. [Google Scholar] [CrossRef] [PubMed]
  73. Monari, C.; Pericolini, E.; Bistoni, G.; Casadevall, A.; Kozel, T.R.; Vecchiarelli, A. Cryptococcus neoformans capsular glucuronoxylomannan induces expression of Fas ligand in macrophages. J. Immunol. 2005, 174, 3461–3468. [Google Scholar] [CrossRef] [PubMed]
  74. Monari, C.; Bistoni, F.; Casadevall, A.; Pericolini, E.; Pietrella, D.; Kozel, T.R.; Vecchiarelli, A. Glucuronoxylomannan, a microbial compound, regulates expression of costimulatory molecules and production of cytokines in macrophages. J. Infect. Dis. 2005, 191, 127–137. [Google Scholar] [CrossRef] [PubMed]
  75. Krieg, A.M. CpG motifs in bacterial DNA and their immune effects. Annu. Rev. Immunol. 2002, 20, 709–760. [Google Scholar] [CrossRef] [PubMed]
  76. Barber, G.N. Cytoplasmic DNA innate immune pathways. Immunol. Rev. 2011, 243, 99–108. [Google Scholar] [CrossRef] [PubMed]
  77. Ramirez-Ortiz, Z.G.; Specht, C.A.; Wang, J.P.; Lee, C.K.; Bartholomeu, D.C.; Gazzinelli, R.T.; Levitz, S.M. Toll-like receptor 9-dependent immune activation by unmethylated CpG motifs in Aspergillus fumigatus DNA. Infect. Immun. 2008, 76, 2123–2129. [Google Scholar] [CrossRef] [PubMed]
  78. Kinjo, T.; Miyagi, K.; Nakamura, K.; Higa, F.; Gang, X.; Miyazato, A.; Kaku, M.; Fujita, J.; Kawakami, K. Adjuvant effect of CpG-oligodeoxynucleotide in anti-fungal chemotherapy against fatal infection with Cryptococcus neoformans in mice. Microbiol. Immunol. 2007, 51, 741–746. [Google Scholar] [CrossRef] [PubMed]
  79. Miyagi, K.; Kawakami, K.; Kinjo, Y.; Uezu, K.; Kinjo, T.; Nakamura, K.; Saito, A. CpG oligodeoxynucleotides promote the host protective response against infection with Cryptococcus neoformans through induction of interferon-γ production by CD4+ T cells. Clin. Exp. Immunol. 2005, 140, 220–229. [Google Scholar] [CrossRef] [PubMed]
  80. Dan, J.M.; Wang, J.P.; Lee, C.K.; Levitz, S.M. Cooperative stimulation of dendritic cells by Cryptococcus neoformans mannoproteins and CpG oligodeoxynucleotides. PLoS ONE 2008, 3, e2046. [Google Scholar] [CrossRef] [PubMed]
  81. Edwards, L.; Williams, A.E.; Krieg, A.M.; Rae, A.J.; Snelgrove, R.J.; Hussell, T. Stimulation via Toll-like receptor 9 reduces Cryptococcus neoformans-induced pulmonary inflammation in an IL-12-dependent manner. Eur. J. Immunol. 2005, 35, 273–281. [Google Scholar] [CrossRef] [PubMed]
  82. Qiu, Y.; Zeltzer, S.; Zhang, Y.; Wang, F.; Chen, G.H.; Dayrit, J.; Murdock, B.J.; Bhan, U.; Toews, G.B.; Osterholzer, J.J.; Standiford, T.J.; et al. Early induction of CCL7 downstream of TLR9 signaling promotes the development of robust immunity to cryptococcal infection. J. Immunol. 2012, 188, 3940–3948. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, Y.; Wang, F.; Bhan, U.; Huffnagle, G.B.; Toews, G.B.; Standiford, T.J.; Olszewski, M.A. TLR9 signaling is required for generation of the adaptive immune protection in Cryptococcus neoformans-infected lungs. Am. J. Pathol. 2010, 177, 754–765. [Google Scholar] [CrossRef] [PubMed]
  84. Nakamura, K.; Miyazato, A.; Xiao, G.; Hatta, M.; Inden, K.; Aoyagi, T.; Shiratori, K.; Takeda, K.; Akira, S.; Saijo, S.; et al. Deoxynucleic acids from Cryptococcus neoformans activate myeloid dendritic cells via a TLR9-dependent pathway. J. Immunol. 2008, 180, 4067–4074. [Google Scholar] [CrossRef] [PubMed]
  85. Tanaka, M.; Ishii, K.; Nakamura, Y.; Miyazato, A.; Maki, A.; Abe, Y.; Miyasaka, T.; Yamamoto, H.; Akahori, Y.; Fue, M.; et al. Toll-like receptor 9-dependent activation of bone marrow-derived dendritic cells by URA5 DNA from Cryptococcus neoformans. Infect. Immun. 2012, 80, 778–786. [Google Scholar] [CrossRef] [PubMed]
  86. Plato, A.; Hardison, S.E.; Brown, G.D. Pattern recognition receptors in antifungal immunity. Semin. Immunopathol. 2015, 37, 97–106. [Google Scholar] [CrossRef] [PubMed]
  87. Gross, O.; Poeck, H.; Bscheider, M.; Dostert, C.; Hannesschlager, N.; Endres, S.; Hartmann, G.; Tardivel, A.; Schweighoffer, E.; Tybulewicz, V.; et al. Syk kinase signalling couples to the NLRP3 inflammasome for anti-fungal host defence. Nature 2009, 459, 433–436. [Google Scholar] [CrossRef] [PubMed]
  88. Hise, A.G.; Tomalka, J.; Ganesan, S.; Patel, K.; Hall, B.A.; Brown, G.D.; Fitzgerald, K.A. An essential role for the NLRP3 inflammasome in host defense against the human fungal pathogen Candida albicans. Cell Host Microbe 2009, 5, 487–497. [Google Scholar] [CrossRef] [PubMed]
  89. Said-Sadier, N.; Padilla, E.; Langsley, G.; Ojcius, D.M. Aspergillus fumigatus stimulates the NLRP3 inflammasome through a pathway requiring ROS production and the Syk tyrosine kinase. PLoS ONE 2010, 5, e10008. [Google Scholar] [CrossRef] [PubMed]
  90. Lei, G.; Chen, M.; Li, H.; Niu, J.L.; Wu, S.; Mao, L.; Lu, A.; Wang, H.; Chen, W.; Xu, B.; et al. Biofilm from a clinical strain of Cryptococcus neoformans activates the NLRP3 inflammasome. Cell Res. 2013, 23, 965–968. [Google Scholar] [CrossRef] [PubMed]
  91. Guo, C.; Chen, M.; Fa, Z.; Lu, A.; Fang, W.; Sun, B.; Chen, C.; Liao, W.; Meng, G. Acapsular Cryptococcus neoformans activates the NLRP3 inflammasome. Microbes Infect. 2014, 16, 845–854. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, M.; Xing, Y.; Lu, A.; Fang, W.; Sun, B.; Chen, C.; Liao, W.; Meng, G. Internalized Cryptococcus neoformans activates the canonical caspase-1 and the noncanonical caspase-8 inflammasomes. J. Immunol. 2015, 195, 4962–4972. [Google Scholar] [CrossRef] [PubMed]
  93. Canton, J.; Neculai, D.; Grinstein, S. Scavenger receptors in homeostasis and immunity. Nat. Rev. Immunol. 2013, 13, 621–634. [Google Scholar] [CrossRef] [PubMed]
  94. Means, T.K.; Mylonakis, E.; Tampakakis, E.; Colvin, R.A.; Seung, E.; Puckett, L.; Tai, M.F.; Stewart, C.R.; Pukkila-Worley, R.; Hickman, S.E.; et al. Evolutionarily conserved recognition and innate immunity to fungal pathogens by the scavenger receptors SCARF1 and CD36. J. Exp. Med. 2009, 206, 637–653. [Google Scholar] [CrossRef] [PubMed]
  95. Xu, J.; Flaczyk, A.; Neal, L.M.; Fa, Z.; Eastman, A.J.; Malachowski, A.N.; Cheng, D.; Moore, B.B.; Curtis, J.L.; Osterholzer, J.J.; et al. Scavenger receptor MARCO orchestrates early defenses and contributes to fungal containment during cryptococcal infection. J. Immunol. 2017, 198, 3548–3557. [Google Scholar] [CrossRef] [PubMed]
  96. Qiu, Y.; Dayrit, J.K.; Davis, M.J.; Carolan, J.F.; Osterholzer, J.J.; Curtis, J.L.; Olszewski, M.A. Scavenger receptor A modulates the immune response to pulmonary Cryptococcus neoformans infection. J. Immunol. 2013, 191, 238–248. [Google Scholar] [CrossRef] [PubMed]
  97. Baumgarth, N. B-1 cell heterogeneity and the regulation of natural and antigen-induced IgM production. Front. Immunol. 2016, 7, 324. [Google Scholar] [CrossRef] [PubMed]
  98. Panda, S.; Ding, J.L. Natural antibodies bridge innate and adaptive immunity. J. Immunol. 2015, 194, 13–20. [Google Scholar] [CrossRef] [PubMed]
  99. Rohatgi, S.; Pirofski, L.A. Molecular characterization of the early B cell response to pulmonary Cryptococcus neoformans infection. J. Immunol. 2012, 189, 5820–5830. [Google Scholar] [CrossRef] [PubMed]
  100. Subramaniam, K.S.; Datta, K.; Quintero, E.; Manix, C.; Marks, M.S.; Pirofski, L.A. The absence of serum IgM enhances the susceptibility of mice to pulmonary challenge with Cryptococcus neoformans. J. Immunol. 2010, 184, 5755–5767. [Google Scholar] [CrossRef] [PubMed]
  101. Subramaniam, K.S.; Datta, K.; Marks, M.S.; Pirofski, L.A. Improved survival of mice deficient in secretory immunoglobulin M following systemic infection with Cryptococcus neoformans. Infect. Immun. 2010, 78, 441–452. [Google Scholar] [CrossRef] [PubMed]
  102. Ghosn, E.E.; Russo, M.; Almeida, S.R. Nitric oxide-dependent killing of Cryptococcus neoformans by B-1-derived mononuclear phagocyte. J. Leukoc. Biol. 2006, 80, 36–44. [Google Scholar] [CrossRef] [PubMed]
  103. 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, 282–287. [Google Scholar] [PubMed]
  104. Subramaniam, K.; Metzger, B.; Hanau, L.H.; Guh, A.; Rucker, L.; Badri, S.; Pirofski, L.A. IgM(+) memory B cell expression predicts HIV-associated cryptococcosis status. J. Infect. Dis. 2009, 200, 244–251. [Google Scholar] [CrossRef] [PubMed]
  105. Jalali, Z.; Ng, L.; Singh, N.; Pirofski, L.A. Antibody response to Cryptococcus neoformans capsular polysaccharide glucuronoxylomannan in patients after solid-organ transplantation. Clin. Vaccine Immunol. 2006, 13, 740–746. [Google Scholar] [CrossRef] [PubMed]
  106. Rothstein, T.L.; Griffin, D.O.; Holodick, N.E.; Quach, T.D.; Kaku, H. Human B-1 cells take the stage. Ann. N. Y. Acad. Sci. 2013, 1285, 97–114. [Google Scholar] [CrossRef] [PubMed]
  107. Kozel, T.R.; Gotschlich, E.C. The capsule of Cryptococcus neoformans passively inhibits phagocytosis of the yeast by macrophages. J. Immunol. 1982, 129, 1675–1680. [Google Scholar] [PubMed]
  108. Levitz, S.M.; Tabuni, A. Binding of Cryptococcus neoformans by human cultured macrophages. Requirements for multiple complement receptors and actin. J. Clin. Investig. 1991, 87, 528–535. [Google Scholar] [CrossRef] [PubMed]
  109. Kelly, R.M.; Chen, J.; Yauch, L.E.; Levitz, S.M. Opsonic requirements for dendritic cell-mediated responses to Cryptococcus neoformans. Infect. Immun. 2005, 73, 592–598. [Google Scholar] [CrossRef] [PubMed]
  110. Zipfel, P.F.; Skerka, C. Complement regulators and inhibitory proteins. Nat. Rev. Immunol. 2009, 9, 729–740. [Google Scholar] [CrossRef] [PubMed]
  111. Diamond, R.D.; May, J.E.; Kane, M.A.; Frank, M.M.; Bennett, J.E. The role of the classical and alternate complement pathways in host defenses against Cryptococcus neoformans infection. J. Immunol. 1974, 112, 2260–2270. [Google Scholar] [PubMed]
  112. Diamond, R.D.; May, J.E.; Kane, M.; Frank, M.M.; Bennett, J.E. The role of late complement components and the alternate complement pathway in experimental cryptococcosis. Proc. Soc. Exp. Biol. Med. 1973, 144, 312–315. [Google Scholar] [CrossRef] [PubMed]
  113. Kozel, T.R.; Wilson, M.A.; Pfrommer, G.S.; Schlageter, A.M. Activation and binding of opsonic fragments of C3 on encapsulated Cryptococcus neoformans by using an alternative complement pathway reconstituted from six isolated proteins. Infect. Immun. 1989, 57, 1922–1927. [Google Scholar] [PubMed]
  114. Kozel, T.R.; Pfrommer, G.S. Activation of the complement system by Cryptococcus neoformans leads to binding of iC3b to the yeast. Infect. Immun. 1986, 52, 1–5. [Google Scholar] [PubMed]
  115. Pfrommer, G.S.; Dickens, S.M.; Wilson, M.A.; Young, B.J.; Kozel, T.R. Accelerated decay of C3b to iC3b when C3b is bound to the Cryptococcus neoformans capsule. Infect. Immun. 1993, 61, 4360–4366. [Google Scholar] [PubMed]
  116. Zaragoza, O.; Taborda, C.P.; 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, 1957–1967. [Google Scholar] [CrossRef] [PubMed]
  117. Taborda, C.P.; Casadevall, A. CR3 (CD11b/CD18) and CR4 (CD11c/CD18) are involved in complement-independent antibody-mediated phagocytosis of Cryptococcus neoformans. Immunity 2002, 16, 791–802. [Google Scholar] [CrossRef]
  118. Sun, D.; Zhang, M.; Liu, G.; Wu, H.; Zhu, X.; Zhou, H.; Shi, M. Real-time imaging of interactions of neutrophils with Cryptococcus neoformans demonstrates a crucial role of complement C5a-C5aR signaling. Infect. Immun. 2015, 84, 216–229. [Google Scholar] [CrossRef] [PubMed]
  119. Polentarutti, N.; Bottazzi, B.; di Santo, E.; Blasi, E.; Agnello, D.; Ghezzi, P.; Introna, M.; Bartfai, T.; Richards, G.; Mantovani, A. Inducible expression of the long pentraxin PTX3 in the central nervous system. J. Neuroimmunol. 2000, 106, 87–94. [Google Scholar] [CrossRef]
  120. Schelenz, S.; Kirchhof, N.; Bidula, S.; Wallis, R.; Sexton, D.W. Opsonizing properties of rat ficolin-A in the defence against Cryptococcus neoformans. Immunobiology 2013, 218, 477–483. [Google Scholar] [CrossRef] [PubMed]
  121. Wozniak, K.L.; Hole, C.R.; Yano, J.; Fidel, P.L., Jr.; Wormley, F.L., Jr. Characterization of IL-22 and antimicrobial peptide production in mice protected against pulmonary Cryptococcus neoformans infection. Microbiology 2014, 160, 1440–1452. [Google Scholar] [CrossRef] [PubMed]
  122. Wiesner, D.L.; Specht, C.A.; Lee, C.K.; Smith, K.D.; Mukaremera, L.; Lee, S.T.; Lee, C.G.; Elias, J.A.; Nielsen, J.N.; Boulware, D.R.; et al. Chitin recognition via chitotriosidase promotes pathologic type-2 helper T cell responses to cryptococcal infection. PLoS Pathog. 2015, 11, e1004701. [Google Scholar] [CrossRef] [PubMed]
  123. Upadhya, R.; Lam, W.C.; Maybruck, B.; Specht, C.A.; Levitz, S.M.; Lodge, J.K. Induction of protective immunity to cryptococcal infection in mice by a heat-killed, chitosan-deficient strain of Cryptococcus neoformans. MBio 2016, 7. [Google Scholar] [CrossRef] [PubMed]
  124. Wagener, J.; Malireddi, R.K.; Lenardon, M.D.; Koberle, M.; Vautier, S.; MacCallum, D.M.; Biedermann, T.; Schaller, M.; Netea, M.G.; Kanneganti, T.D.; et al. Fungal chitin dampens inflammation through IL-10 induction mediated by NOD2 and TLR9 activation. PLoS Pathog. 2014, 10, e1004050. [Google Scholar] [CrossRef] [PubMed]
  125. Bueter, C.L.; Specht, C.A.; Levitz, S.M. Innate sensing of chitin and chitosan. PLoS Pathog. 2013, 9, e1003080. [Google Scholar] [CrossRef] [PubMed]
  126. Bueter, C.L.; Lee, C.K.; Rathinam, V.A.; Healy, G.J.; Taron, C.H.; Specht, C.A.; Levitz, S.M. Chitosan but not chitin activates the inflammasome by a mechanism dependent upon phagocytosis. J. Biol. Chem. 2011, 286, 35447–35455. [Google Scholar] [CrossRef] [PubMed]
  127. Bueter, C.L.; Lee, C.K.; Wang, J.P.; Ostroff, G.R.; Specht, C.A.; Levitz, S.M. Spectrum and mechanisms of inflammasome activation by chitosan. J. Immunol. 2014, 192, 5943–5951. [Google Scholar] [CrossRef] [PubMed]
  128. Ost, K.S.; Esher, S.K.; Leopold Wager, C.M.; Walker, L.; Wagener, J.; Munro, C.; Wormley, F.L., Jr.; Alspaugh, J.A. Rim pathway-mediated alterations in the fungal cell wall influence immune recognition and inflammation. MBio 2017, 8. [Google Scholar] [CrossRef] [PubMed]
  129. O'Meara, T.R.; Holmer, S.M.; Selvig, K.; Dietrich, F.; Alspaugh, J.A. Cryptococcus neoformans Rim101 is associated with cell wall remodeling and evasion of the host immune responses. MBio 2013, 4. [Google Scholar] [CrossRef] [PubMed]
  130. O'Meara, T.R.; Norton, D.; Price, M.S.; Hay, C.; Clements, M.F.; Nichols, C.B.; Alspaugh, J.A. Interaction of Cryptococcus neoformans Rim101 and protein kinase A regulates capsule. PLoS Pathog. 2010, 6, e1000776. [Google Scholar] [CrossRef] [PubMed]
  131. Rodrigues, M.L.; Nimrichter, L.; Oliveira, D.L.; Frases, S.; Miranda, K.; Zaragoza, O.; Alvarez, M.; Nakouzi, A.; Feldmesser, M.; Casadevall, A. Vesicular polysaccharide export in Cryptococcus neoformans is a eukaryotic solution to the problem of fungal trans-cell wall transport. Eukaryot. Cell 2007, 6, 48–59. [Google Scholar] [CrossRef] [PubMed]
  132. Brown, L.; Wolf, J.M.; Prados-Rosales, R.; Casadevall, A. Through the wall: Extracellular vesicles in gram-positive bacteria, mycobacteria and fungi. Nat. Rev. Microbiol. 2015, 13, 620–630. [Google Scholar] [CrossRef] [PubMed]
  133. Schorey, J.S.; Cheng, Y.; Singh, P.P.; Smith, V.L. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep. 2015, 16, 24–43. [Google Scholar] [CrossRef] [PubMed]
  134. Rodrigues, M.L.; Nakayasu, E.S.; Oliveira, D.L.; Nimrichter, L.; Nosanchuk, J.D.; Almeida, I.C.; Casadevall, A. Extracellular vesicles produced by Cryptococcus neoformans contain protein components associated with virulence. Eukaryot. Cell 2008, 7, 58–67. [Google Scholar] [CrossRef] [PubMed]
  135. Panepinto, J.; Komperda, K.; Frases, S.; Park, Y.D.; Djordjevic, J.T.; Casadevall, A.; Williamson, P.R. Sec6-dependent sorting of fungal extracellular exosomes and laccase of Cryptococcus neoformans. Mol. Microbiol. 2009, 71, 1165–1176. [Google Scholar] [CrossRef] [PubMed]
  136. Oliveira, D.L.; Freire-de-Lima, C.G.; Nosanchuk, J.D.; Casadevall, A.; Rodrigues, M.L.; Nimrichter, L. Extracellular vesicles from Cryptococcus neoformans modulate macrophage functions. Infect. Immun. 2010, 78, 1601–1609. [Google Scholar] [CrossRef] [PubMed]
  137. Drummond, R.A.; Gaffen, S.L.; Hise, A.G.; Brown, G.D. Innate defense against fungal pathogens. Cold Spring Harb. Perspect. Med. 2014, 5. [Google Scholar] [CrossRef] [PubMed]
  138. Hamerman, J.A.; Ni, M.; Killebrew, J.R.; Chu, C.L.; Lowell, C.A. The expanding roles of ITAM adapters FcRγ and DAP12 in myeloid cells. Immunol. Rev. 2009, 232, 42–58. [Google Scholar] [CrossRef] [PubMed]
  139. Colonna, M. All roads lead to CARD9. Nat. Immunol. 2007, 8, 554–555. [Google Scholar] [CrossRef] [PubMed]
  140. Ruland, J. CARD9 signaling in the innate immune response. Ann. N. Y. Acad. Sci. 2008, 1143, 35–44. [Google Scholar] [CrossRef] [PubMed]
  141. Yamamoto, H.; Nakamura, Y.; Sato, K.; Takahashi, Y.; Nomura, T.; Miyasaka, T.; Ishii, K.; Hara, H.; Yamamoto, N.; Kanno, E.; et al. Defect of CARD9 leads to impaired accumulation of γ interferon-producing memory phenotype T cells in lungs and increased susceptibility to pulmonary infection with Cryptococcus neoformans. Infect. Immun. 2014, 82, 1606–1615. [Google Scholar] [CrossRef] [PubMed]
  142. Adachi, O.; Kawai, T.; Takeda, K.; Matsumoto, M.; Tsutsui, H.; Sakagami, M.; Nakanishi, K.; Akira, S. Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function. Immunity 1998, 9, 143–150. [Google Scholar] [CrossRef]
  143. Burns, K.; Martinon, F.; Esslinger, C.; Pahl, H.; Schneider, P.; Bodmer, J.L.; di Marco, F.; French, L.; Tschopp, J. MyD88, an adapter protein involved in interleukin-1 signaling. J. Biol. Chem. 1998, 273, 12203–12209. [Google Scholar] [CrossRef] [PubMed]
  144. Kawakami, K.; Koguchi, Y.; Qureshi, M.H.; Miyazato, A.; Yara, S.; Kinjo, Y.; Iwakura, Y.; Takeda, K.; Akira, S.; Kurimoto, M.; et al. IL-18 contributes to host resistance against infection with Cryptococcus neoformans in mice with defective IL-12 synthesis through induction of IFN-γ production by NK cells. J. Immunol. 2000, 165, 941–947. [Google Scholar] [CrossRef] [PubMed]
  145. Kawakami, K.; Koguchi, Y.; Qureshi, M.H.; Kinjo, Y.; Yara, S.; Miyazato, A.; Kurimoto, M.; Takeda, K.; Akira, S.; Saito, A. Reduced host resistance and Th1 response to Cryptococcus neoformans in interleukin-18 deficient mice. FEMS Microbiol. Lett. 2000, 186, 121–126. [Google Scholar] [CrossRef] [PubMed]
  146. Takaki, R.; Watson, S.R.; Lanier, L.L. DAP12: An adapter protein with dual functionality. Immunol. Rev. 2006, 214, 118–129. [Google Scholar] [CrossRef] [PubMed]
  147. Turnbull, I.R.; Colonna, M. Activating and inhibitory functions of DAP12. Nat. Rev. Immunol. 2007, 7, 155–161. [Google Scholar] [CrossRef] [PubMed]
  148. Lanier, L.L. DAP10- and DAP12-associated receptors in innate immunity. Immunol. Rev. 2009, 227, 150–160. [Google Scholar] [CrossRef] [PubMed]
  149. Otero, K.; Turnbull, I.R.; Poliani, P.L.; Vermi, W.; Cerutti, E.; Aoshi, T.; Tassi, I.; Takai, T.; Stanley, S.L.; Miller, M.; et al. Macrophage colony-stimulating factor induces the proliferation and survival of macrophages via a pathway involving DAP12 and β-catenin. Nat. Immunol. 2009, 10, 734–743. [Google Scholar] [CrossRef] [PubMed]
  150. Turnbull, I.R.; Gilfillan, S.; Cella, M.; Aoshi, T.; Miller, M.; Piccio, L.; Hernandez, M.; Colonna, M. Cutting edge: TREM-2 attenuates macrophage activation. J. Immunol. 2006, 177, 3520–3524. [Google Scholar] [CrossRef] [PubMed]
  151. Schlitzer, A.; McGovern, N.; Ginhoux, F. Dendritic cells and monocyte-derived cells: Two complementary and integrated functional systems. Semin Cell. Dev. Biol. 2015, 41, 9–22. [Google Scholar] [CrossRef] [PubMed]
  152. Shi, C.; Pamer, E.G. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 2011, 11, 762–774. [Google Scholar] [CrossRef] [PubMed]
  153. Serbina, N.V.; Jia, T.; Hohl, T.M.; Pamer, E.G. Monocyte-mediated defense against microbial pathogens. Annu. Rev. Immunol. 2008, 26, 421–452. [Google Scholar] [CrossRef] [PubMed]
  154. Smith, P.D.; Ohura, K.; Masur, H.; Lane, H.C.; Fauci, A.S.; Wahl, S.M. Monocyte function in the acquired immune deficiency syndrome. Defective chemotaxis. J. Clin. Investig. 1984, 74, 2121–2128. [Google Scholar] [CrossRef] [PubMed]
  155. Szelc, C.M.; Mitcheltree, C.; Roberts, R.L.; Stiehm, E.R. Deficient polymorphonuclear cell and mononuclear cell antibody-dependent cellular cytotoxicity in pediatric and adult human immunodeficiency virus infection. J. Infect. Dis. 1992, 166, 486–493. [Google Scholar] [CrossRef] [PubMed]
  156. Diamond, R.D.; Root, R.K.; Bennett, J.E. Factors influencing killing of Cryptococcus neoformans by human leukocytes in vitro. J. Infect. Dis. 1972, 125, 367–376. [Google Scholar] [CrossRef] [PubMed]
  157. Miller, M.F.; Mitchell, T.G. Killing of Cryptococcus neoformans strains by human neutrophils and monocytes. Infect. Immun. 1991, 59, 24–28. [Google Scholar] [PubMed]
  158. Levitz, S.M.; Farrell, T.P.; Maziarz, R.T. Killing of Cryptococcus neoformans by human peripheral blood mononuclear cells stimulated in culture. J. Infect. Dis. 1991, 163, 1108–1113. [Google Scholar] [CrossRef] [PubMed]
  159. Scriven, J.E.; Graham, L.M.; Schutz, C.; Scriba, T.J.; Wilkinson, K.A.; Wilkinson, R.J.; Boulware, D.R.; Urban, B.C.; Meintjes, G.; Lalloo, D.G. The CSF immune response in HIV-1-associated cryptococcal meningitis: Macrophage activation, correlates of disease severity and effect of antiretroviral therapy. J. Acquir. Immune Defic. Syndr. 2017, 75, 299–307. [Google Scholar] [CrossRef] [PubMed]
  160. Levitz, S.M.; Farrell, T.P. Growth inhibition of Cryptococcus neoformans by cultured human monocytes: Role of the capsule, opsonins, the culture surface, and cytokines. Infect. Immun. 1990, 58, 1201–1209. [Google Scholar] [PubMed]
  161. Diamond, R.D.; Bennett, J.E. Growth of Cryptococcus neoformans within human macrophages in vitro. Infect. Immun. 1973, 7, 231–236. [Google Scholar] [PubMed]
  162. Alvarez, M.; Burn, T.; Luo, Y.; Pirofski, L.A.; Casadevall, A. The outcome of Cryptococcus neoformans intracellular pathogenesis in human monocytes. BMC Microbiol. 2009, 9, 51. [Google Scholar] [CrossRef] [PubMed]
  163. Sorrell, T.C.; Juillard, P.G.; Djordjevic, J.T.; Kaufman-Francis, K.; Dietmann, A.; Milonig, A.; Combes, V.; Grau, G.E. Cryptococcal transmigration across a model brain blood-barrier: Evidence of the Trojan horse mechanism and differences between Cryptococcus neoformans var. grubii strain H99 and Cryptococcus gattii strain R265. Microbes Infect. 2016, 18, 57–67. [Google Scholar] [PubMed]
  164. Traynor, T.R.; Kuziel, W.A.; Toews, G.B.; Huffnagle, G.B. CCR2 expression determines T1 versus T2 polarization during pulmonary Cryptococcus neoformans infection. J. Immunol. 2000, 164, 2021–2027. [Google Scholar] [CrossRef] [PubMed]
  165. Osterholzer, J.J.; Curtis, J.L.; Polak, T.; Ames, T.; Chen, G.H.; McDonald, R.; Huffnagle, G.B.; Toews, G.B. CCR2 mediates conventional dendritic cell recruitment and the formation of bronchovascular mononuclear cell infiltrates in the lungs of mice infected with Cryptococcus neoformans. J. Immunol. 2008, 181, 610–620. [Google Scholar] [CrossRef] [PubMed]
  166. Osterholzer, J.J.; Chen, G.H.; Olszewski, M.A.; Curtis, J.L.; Huffnagle, G.B.; Toews, G.B. Accumulation of CD11b+ lung dendritic cells in response to fungal infection results from the CCR2-mediated recruitment and differentiation of Ly-6Chigh monocytes. J. Immunol. 2009, 183, 8044–8053. [Google Scholar] [CrossRef] [PubMed]
  167. Osterholzer, J.J.; Chen, G.H.; Olszewski, M.A.; Zhang, Y.M.; Curtis, J.L.; Huffnagle, G.B.; Toews, G.B. Chemokine receptor 2-mediated accumulation of fungicidal exudate macrophages in mice that clear cryptococcal lung infection. Am. J. Pathol. 2011, 178, 198–211. [Google Scholar] [CrossRef] [PubMed]
  168. Ginhoux, F.; Jung, S. Monocytes and macrophages: Developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 2014, 14, 392–404. [Google Scholar] [CrossRef] [PubMed]
  169. Gal, A.A.; Koss, M.N.; Hawkins, J.; Evans, S.; Einstein, H. The pathology of pulmonary cryptococcal infections in the acquired immunodeficiency syndrome. Arch. Pathol. Lab. Med. 1986, 110, 502–507. [Google Scholar] [PubMed]
  170. Feldmesser, M.; Kress, Y.; Novikoff, P.; Casadevall, A. Cryptococcus neoformans is a facultative intracellular pathogen in murine pulmonary infection. Infect. Immun. 2000, 68, 4225–4237. [Google Scholar] [CrossRef] [PubMed]
  171. Goldman, D.; Lee, S.C.; Casadevall, A. Pathogenesis of pulmonary Cryptococcus neoformans infection in the rat. Infect. Immun. 1994, 62, 4755–4761. [Google Scholar] [PubMed]
  172. Levitz, S.M.; DiBenedetto, D.J. Paradoxical role of capsule in murine bronchoalveolar macrophage-mediated killing of Cryptococcus neoformans. J. Immunol. 1989, 142, 659–665. [Google Scholar] [PubMed]
  173. Bolanos, B.; Mitchell, T.G. Killing of Cryptococcus neoformans by rat alveolar macrophages. J. Med. Vet. Mycol. 1989, 27, 219–228. [Google Scholar] [CrossRef] [PubMed]
  174. Alvarez, M.; Casadevall, A. Phagosome extrusion and host-cell survival after Cryptococcus neoformans phagocytosis by macrophages. Curr. Biol. 2006, 16, 2161–2165. [Google Scholar] [CrossRef] [PubMed]
  175. Ma, H.; Croudace, J.E.; Lammas, D.A.; May, R.C. Expulsion of live pathogenic yeast by macrophages. Curr. Biol. 2006, 16, 2156–2160. [Google Scholar] [CrossRef] [PubMed]
  176. 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, 120–127. [Google Scholar] [CrossRef] [PubMed]
  177. Sabiiti, W.; Robertson, E.; Beale, M.A.; Johnston, S.A.; Brouwer, A.E.; Loyse, A.; Jarvis, J.N.; Gilbert, A.S.; Fisher, M.C.; Harrison, T.S.; et al. Efficient phagocytosis and laccase activity affect the outcome of HIV-associated cryptococcosis. J. Clin. Investig. 2014, 124, 2000–2008. [Google Scholar] [CrossRef] [PubMed]
  178. Shao, X.P.; Mednick, A.; Alvarez, M.; van Rooijen, N.; Casadevall, A.; Goldman, D.L. An innate immune system cell is a major determinant of species-related susceptibility differences to fungal pneumonia. J. Immunol. 2005, 175, 3244–3251. [Google Scholar] [CrossRef] [PubMed]
  179. Osterholzer, J.J.; Milam, J.E.; Chen, G.H.; Toews, G.B.; Huffnagle, G.B.; Olszewski, M.A. Role of dendritic cells and alveolar macrophages in regulating early host defense against pulmonary infection with Cryptococcus neoformans. Infect. Immun. 2009, 77, 3749–3758. [Google Scholar] [CrossRef] [PubMed]
  180. Van Blijswijk, J.; Schraml, B.U.; Reis e Sousa, C. Advantages and limitations of mouse models to deplete dendritic cells. Eur. J. Immunol. 2013, 43, 22–26. [Google Scholar] [CrossRef] [PubMed]
  181. Leopold Wager, C.M.; Wormley, F.L., Jr. Classical versus alternative macrophage activation: The Ying and the Yang in host defense against pulmonary fungal infections. Mucosal Immunol. 2014, 7, 1023–1035. [Google Scholar] [CrossRef] [PubMed]
  182. Panackal, A.A.; Wuest, S.C.; Lin, Y.C.; Wu, T.; Zhang, N.; Kosa, P.; Komori, M.; Blake, A.; Browne, S.K.; Rosen, L.B.; et al. Paradoxical immune responses in non-HIV cryptococcal meningitis. PLoS Pathog. 2015, 11, e1004884. [Google Scholar] [CrossRef] [PubMed]
  183. Stenzel, W.; Muller, U.; Kohler, G.; Heppner, F.L.; Blessing, M.; McKenzie, A.N.; Brombacher, F.; Alber, G. IL-4/IL-13-dependent alternative activation of macrophages but not microglial cells is associated with uncontrolled cerebral cryptococcosis. Am. J. Pathol. 2009, 174, 486–496. [Google Scholar] [CrossRef] [PubMed]
  184. Davis, M.J.; Tsang, T.M.; Qiu, Y.; Dayrit, J.K.; Freij, J.B.; Huffnagle, G.B.; Olszewski, M.A. Macrophage M1/M2 polarization dynamically adapts to changes in cytokine microenvironments in Cryptococcus neoformans infection. MBio 2013, 4. [Google Scholar] [CrossRef] [PubMed]
  185. Arora, S.; Olszewski, M.A.; Tsang, T.M.; McDonald, R.A.; Toews, G.B.; Huffnagle, G.B. Effect of cytokine interplay on macrophage polarization during chronic pulmonary infection with Cryptococcus neoformans. Infect. Immun. 2011, 79, 1915–1926. [Google Scholar] [CrossRef] [PubMed]
  186. Arora, S.; Hernandez, Y.; Erb-Downward, J.R.; McDonald, R.A.; Toews, G.B.; Huffnagle, G.B. Role of IFN-γ in regulating T2 immunity and the development of alternatively activated macrophages during allergic bronchopulmonary mycosis. J. Immunol. 2005, 174, 6346–6356. [Google Scholar] [CrossRef] [PubMed]
  187. Hernandez, Y.; Arora, S.; Erb-Downward, J.R.; McDonald, R.A.; Toews, G.B.; Huffnagle, G.B. Distinct roles for IL-4 and IL-10 in regulating T2 immunity during allergic bronchopulmonary mycosis. J. Immunol. 2005, 174, 1027–1036. [Google Scholar] [CrossRef] [PubMed]
  188. Eastman, A.J.; He, X.; Qiu, Y.; Davis, M.J.; Vedula, P.; Lyons, D.M.; Park, Y.D.; Hardison, S.E.; Malachowski, A.N.; Osterholzer, J.J.; et al. Cryptococcal heat shock protein 70 homolog SSA1 contributes to pulmonary expansion of Cryptococcus neoformans during the afferent phase of the immune response by promoting macrophage M2 polarization. J. Immunol. 2015, 194, 5999–6010. [Google Scholar] [CrossRef] [PubMed]
  189. Leopold Wager, C.M.; Hole, C.R.; Wozniak, K.L.; Olszewski, M.A.; Mueller, M.; Wormley, F.L., Jr. STAT1 signaling within macrophages is required for antifungal activity against Cryptococcus neoformans. Infect. Immun. 2015, 83, 4513–4527. [Google Scholar] [CrossRef] [PubMed]
  190. Leopold Wager, C.M.; Hole, C.R.; Wozniak, K.L.; Olszewski, M.A.; Wormley, F.L., Jr. STAT1 signaling is essential for protection against Cryptococcus neoformans infection in mice. J. Immunol. 2014, 193, 4060–4071. [Google Scholar] [CrossRef] [PubMed]
  191. El Kasmi, K.C.; Qualls, J.E.; Pesce, J.T.; Smith, A.M.; Thompson, R.W.; Henao-Tamayo, M.; Basaraba, R.J.; Konig, T.; Schleicher, U.; Koo, M.S.; et al. Toll-like receptor-induced arginase 1 in macrophages thwarts effective immunity against intracellular pathogens. Nat. Immunol. 2008, 9, 1399–1406. [Google Scholar] [CrossRef] [PubMed]
  192. Ramirez-Ortiz, Z.G.; Means, T.K. The role of dendritic cells in the innate recognition of pathogenic fungi (A. fumigatus, C. neoformans and C. albicans). Virulence 2012, 3, 635–646. [Google Scholar] [CrossRef] [PubMed]
  193. Eastman, A.J.; Osterholzer, J.J.; Olszewski, M.A. Role of dendritic cell-pathogen interactions in the immune response to pulmonary cryptococcal infection. Future Microbiol. 2015, 10, 1837–1857. [Google Scholar] [CrossRef] [PubMed]
  194. Leopold Wager, C.M.; Hole, C.R.; Wozniak, K.L.; Wormley, F.L., Jr. Cryptococcus and phagocytes: Complex interactions that influence disease outcome. Front. Microbiol. 2016, 7, 105. [Google Scholar] [CrossRef] [PubMed]
  195. Merad, M.; Sathe, P.; Helft, J.; Miller, J.; Mortha, A. The dendritic cell lineage: Ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 2013, 31, 563–604. [Google Scholar] [CrossRef] [PubMed]
  196. Bauman, S.K.; Nichols, K.L.; Murphy, J.W. Dendritic cells in the induction of protective and nonprotective anticryptococcal cell-mediated immune responses. J. Immunol. 2000, 165, 158–167. [Google Scholar] [CrossRef] [PubMed]
  197. 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, 417–427. [Google Scholar] [CrossRef] [PubMed]
  198. Wozniak, K.L.; Vyas, J.M.; Levitz, S.M. In vivo role of dendritic cells in a murine model of pulmonary cryptococcosis. Infect. Immun. 2006, 74, 3817–3824. [Google Scholar] [CrossRef] [PubMed]
  199. Espinosa, V.; Rivera, A. First line of defense: Innate cell-mediated control of pulmonary aspergillosis. Front. Microbiol. 2016, 7, 272. [Google Scholar] [CrossRef] [PubMed]
  200. Qureshi, A.; Subathra, M.; Grey, A.; Schey, K.; del Poeta, M.; Luberto, C. Role of sphingomyelin synthase in controlling the antimicrobial activity of neutrophils against Cryptococcus neoformans. PLoS ONE 2010, 5, e15587. [Google Scholar] [CrossRef] [PubMed]
  201. Graybill, J.R.; Bocanegra, R.; Lambros, C.; Luther, M.F. Granulocyte colony stimulating factor therapy of experimental cryptococcal meningitis. J. Med. Vet. Mycol. 1997, 35, 243–247. [Google Scholar] [CrossRef] [PubMed]
  202. Walenkamp, A.M.; Ellerbroek, P.; Scharringa, J.; Rijkers, E.; Hoepelman, A.I.; Coenjaerts, F.E. Interference of Cryptococcus neoformans with human neutrophil migration. Adv. Exp. Med. Biol. 2003, 531, 315–339. [Google Scholar] [PubMed]
  203. Richardson, M.D.; White, L.J.; McKay, I.C.; Shankland, G.S. Differential binding of acapsulate and encapsulated strains of Cryptococcus neoformans to human neutrophils. J. Med. Vet. Mycol. 1993, 31, 189–199. [Google Scholar] [CrossRef] [PubMed]
  204. Rocha, J.D.; Nascimento, M.T.; Decote-Ricardo, D.; Corte-Real, S.; Morrot, A.; Heise, N.; Nunes, M.P.; Previato, J.O.; Mendonca-Previato, L.; DosReis, G.A.; Saraiva, E.M.; Freire-de-Lima, C.G. Capsular polysaccharides from Cryptococcus neoformans modulate production of neutrophil extracellular traps (NETs) by human neutrophils. Sci. Rep. 2015, 5, 8008. [Google Scholar] [CrossRef] [PubMed]
  205. Sun, D.; Zhang, M.; Liu, G.; Wu, H.; Li, C.; Zhou, H.; Zhang, X.; Shi, M. Intravascular clearance of disseminating Cryptococcus neoformans in the brain can be improved by enhancing neutrophil recruitment in mice. Eur. J. Immunol. 2016, 46, 1704–1714. [Google Scholar] [CrossRef] [PubMed]
  206. Sun, D.; Shi, M. Neutrophil swarming toward Cryptococcus neoformans is mediated by complement and leukotriene B4. Biochem. Biophys. Res. Commun. 2016, 477, 945–951. [Google Scholar] [CrossRef] [PubMed]
  207. Zhang, M.; Sun, D.; Liu, G.; Wu, H.; Zhou, H.; Shi, M. Real-time in vivo imaging reveals the ability of neutrophils to remove Cryptococcus neoformans directly from the brain vasculature. J. Leukoc. Biol. 2016, 99, 467–473. [Google Scholar] [CrossRef] [PubMed]
  208. Wozniak, K.L.; Kolls, J.K.; Wormley, F.L., Jr. Depletion of neutrophils in a protective model of pulmonary cryptococcosis results in increased IL-17A production by γΔ T cells. BMC Immunol. 2012, 13, 65. [Google Scholar] [CrossRef] [PubMed]
  209. Mednick, A.J.; 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, 1744–1753. [Google Scholar] [CrossRef] [PubMed]
  210. Wiesner, D.L.; Smith, K.D.; Kashem, S.W.; Bohjanen, P.R.; Nielsen, K. Different lymphocyte populations direct dichotomous eosinophil or neutrophil responses to pulmonary Cryptococcus infection. J. Immunol. 2017, 198, 1627–1637. [Google Scholar] [CrossRef] [PubMed]
  211. Lipscomb, M.F.; Alvarellos, T.; Toews, G.B.; Tompkins, R.; Evans, Z.; Koo, G.; Kumar, V. Role of natural killer cells in resistance to Cryptococcus neoformans infections in mice. Am. J. Pathol. 1987, 128, 354–361. [Google Scholar] [PubMed]
  212. Salkowski, C.A.; Balish, E. Role of natural killer cells in resistance to systemic cryptococcosis. J. Leukoc. Biol. 1991, 50, 151–159. [Google Scholar] [PubMed]
  213. Hidore, M.R.; Murphy, J.W. Natural cellular resistance of beige mice against Cryptococcus neoformans. J. Immunol. 1986, 137, 3624–3631. [Google Scholar] [PubMed]
  214. Hidore, M.R.; Murphy, J.W. Murine natural killer cell interactions with a fungal target, Cryptococcus neoformans. Infect. Immun. 1989, 57, 1990–1997. [Google Scholar] [PubMed]
  215. Murphy, J.W.; Hidore, M.R.; Nabavi, N. Binding interactions of murine natural killer cells with the fungal target Cryptococcus neoformans. Infect. Immun. 1991, 59, 1476–1488. [Google Scholar] [PubMed]
  216. Nabavi, N.; Murphy, J.W. In vitro binding of natural killer cells to Cryptococcus neoformans targets. Infect. Immun. 1985, 50, 50–57. [Google Scholar] [PubMed]
  217. Kawakami, K.; Koguchi, Y.; Qureshi, M.H.; Yara, S.; Kinjo, Y.; Uezu, K.; Saito, A. NK cells eliminate Cryptococcus neoformans by potentiating the fungicidal activity of macrophages rather than by directly killing them upon stimulation with IL-12 and IL-18. Microbiol. Immunol. 2000, 44, 1043–1050. [Google Scholar] [CrossRef] [PubMed]
  218. Zhang, T.; Kawakami, K.; Qureshi, M.H.; Okamura, H.; Kurimoto, M.; Saito, A. Interleukin-12 (IL-12) and IL-18 synergistically induce the fungicidal activity of murine peritoneal exudate cells against Cryptococcus neoformans through production of γ interferon by natural killer cells. Infect. Immun. 1997, 65, 3594–3599. [Google Scholar] [PubMed]
  219. Horn, C.A.; Washburn, R.G. Anticryptococcal activity of NK cell-enriched peripheral blood lymphocytes from human immunodeficiency virus-infected subjects: Responses to interleukin-2, interferon-γ, and interleukin-12. J. Infect. Dis. 1995, 172, 1023–1027. [Google Scholar] [CrossRef] [PubMed]
  220. Levitz, S.M.; Dupont, M.P.; Smail, E.H. Direct activity of human T lymphocytes and natural killer cells against Cryptococcus neoformans. Infect. Immun. 1994, 62, 194–202. [Google Scholar] [PubMed]
  221. Murphy, J.W.; Hidore, M.R.; Wong, S.C. Direct interactions of human lymphocytes with the yeast-like organism, Cryptococcus neoformans. J. Clin. Investig. 1993, 91, 1553–1566. [Google Scholar] [CrossRef] [PubMed]
  222. Wiseman, J.C.; Ma, L.L.; Marr, K.J.; Jones, G.J.; Mody, C.H. Perforin-dependent cryptococcal microbicidal activity in NK cells requires PI3K-dependent ERK1/2 signaling. J. Immunol. 2007, 178, 6456–6464. [Google Scholar] [CrossRef] [PubMed]
  223. Ma, L.L.; Wang, C.L.; Neely, G.G.; Epelman, S.; Krensky, A.M.; Mody, C.H. NK cells use perforin rather than granulysin for anticryptococcal activity. J. Immunol. 2004, 173, 3357–3365. [Google Scholar] [CrossRef] [PubMed]
  224. Li, S.S.; Kyei, S.K.; Timm-McCann, M.; Ogbomo, H.; Jones, G.J.; Shi, M.; Xiang, R.F.; Oykhman, P.; Huston, S.M.; Islam, A.; 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, 387–397. [Google Scholar] [CrossRef] [PubMed]
  225. Vitenshtein, A.; Charpak-Amikam, Y.; Yamin, R.; Bauman, Y.; Isaacson, B.; Stein, N.; Berhani, O.; Dassa, L.; Gamliel, M.; Gur, C.; et al. NK cell recognition of Candida glabrata through binding of NKp46 and NCR1 to fungal ligands Epa1, Epa6, and Epa7. Cell Host Microbe 2016, 20, 527–534. [Google Scholar] [CrossRef] [PubMed]
  226. Rosenberg, H.F.; Dyer, K.D.; Foster, P.S. Eosinophils: Changing perspectives in health and disease. Nat. Rev. Immunol. 2013, 13, 9–22. [Google Scholar] [CrossRef] [PubMed]
  227. Marwaha, R.K.; Trehan, A.; Jayashree, K.; Vasishta, R.K. Hypereosinophilia in disseminated cryptococcal disease. Pediatr. Infect. Dis. J. 1995, 14, 1102–1103. [Google Scholar] [PubMed]
  228. Pfeffer, P.E.; Sen, A.; Das, S.; Sheaff, M.; Sivaramakrishnan, A.; Simcock, D.E.; Turner, B. Eosinophilia, meningitis and pulmonary nodules in a young woman. Thorax 2010, 65, 1066–1085. [Google Scholar] [CrossRef] [PubMed]
  229. Yamaguchi, H.; Komase, Y.; Ikehara, M.; Yamamoto, T.; Shinagawa, T. Disseminated cryptococcal infection with eosinophilia in a healthy person. J. Infect. Chemother. 2008, 14, 319–324. [Google Scholar] [CrossRef] [PubMed]
  230. Epstein, R.; Cole, R.; Hunt, K.K., Jr. Pleural effusion secondary to pulmonary cryptococcosis. Chest 1972, 61, 296–298. [Google Scholar] [CrossRef] [PubMed]
  231. Jensen, W.A.; Rose, R.M.; Hammer, S.M.; Karchmer, A.W. Serologic diagnosis of focal pneumonia caused by Cryptococcus neoformans. Am. Rev. Respir. Dis. 1985, 132, 189–191. [Google Scholar] [PubMed]
  232. Brewer, G.E.; Wood, F.C. XII. Blastomycosis of the spine: Double lesion: Two operations: Recovery. Ann. Surg. 1908, 48, 889–896. [Google Scholar] [CrossRef] [PubMed]
  233. Feldmesser, M.; Casadevall, A.; Kress, Y.; Spira, G.; Orlofsky, A. Eosinophil-Cryptococcus neoformans interactions in vivo and in vitro. Infect. Immun. 1997, 65, 1899–1907. [Google Scholar] [PubMed]
  234. Jain, A.V.; Zhang, Y.; Fields, W.B.; McNamara, D.A.; Choe, M.Y.; Chen, G.H.; Erb-Downward, J.; Osterholzer, J.J.; Toews, G.B.; Huffnagle, G.B.; et al. Th2 but not Th1 immune bias results in altered lung functions in a murine model of pulmonary Cryptococcus neoformans infection. Infect. Immun. 2009, 77, 5389–5399. [Google Scholar] [CrossRef] [PubMed]
  235. Rivera, J.; Casadevall, A. Mouse genetic background is a major determinant of isotype-related differences for antibody-mediated protective efficacy against Cryptococcus neoformans. J. Immunol. 2005, 174, 8017–8026. [Google Scholar] [CrossRef] [PubMed]
  236. Piehler, D.; Stenzel, W.; Grahnert, A.; Held, J.; Richter, L.; Kohler, G.; Richter, T.; Eschke, M.; Alber, G.; Muller, U. Eosinophils contribute to IL-4 production and shape the T-helper cytokine profile and inflammatory response in pulmonary cryptococcosis. Am. J. Pathol. 2011, 179, 733–744. [Google Scholar] [CrossRef] [PubMed]
  237. Garro, A.P.; Chiapello, L.S.; Baronetti, J.L.; Masih, D.T. Eosinophils elicit proliferation of naive and fungal-specific cells in vivo so enhancing a T helper type 1 cytokine profile in favour of a protective immune response against Cryptococcus neoformans infection. Immunology 2011, 134, 198–213. [Google Scholar] [CrossRef] [PubMed]
  238. Garro, A.P.; Chiapello, L.S.; Baronetti, J.L.; Masih, D.T. Rat eosinophils stimulate the expansion of Cryptococcus neoformans-specific CD4(+) and CD8(+) T cells with a T-helper 1 profile. Immunology 2011, 132, 174–187. [Google Scholar] [CrossRef] [PubMed]
  239. Flaczyk, A.; Duerr, C.U.; Shourian, M.; Lafferty, E.I.; Fritz, J.H.; Qureshi, S.T. IL-33 signaling regulates innate and adaptive immunity to Cryptococcus neoformans. J. Immunol. 2013, 191, 2503–2513. [Google Scholar] [CrossRef] [PubMed]
  240. Whitsett, J.A.; Alenghat, T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat. Immunol. 2015, 16, 27–35. [Google Scholar] [CrossRef] [PubMed]
  241. Mai, J.; Virtue, A.; Shen, J.; Wang, H.; Yang, X.F. An evolving new paradigm: Endothelial cells—Conditional innate immune cells. J. Hematol. Oncol. 2013, 6, 61. [Google Scholar] [CrossRef] [PubMed]
  242. Guillot, L.; Carroll, S.F.; Badawy, M.; Qureshi, S.T. Cryptococcus neoformans induces IL-8 secretion and CXCL1 expression by human bronchial epithelial cells. Respir. Res. 2008, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  243. Teixeira, P.A.; Penha, L.L.; Mendonca-Previato, L.; Previato, J.O. Mannoprotein MP84 mediates the adhesion of Cryptococcus neoformans to epithelial lung cells. Front. Cell. Infect. Microbiol. 2014, 4, 106. [Google Scholar] [CrossRef] [PubMed]
  244. Roseff, S.A.; Levitz, S.M. Effect of endothelial cells on phagocyte-mediated anticryptococcal activity. Infect. Immun. 1993, 61, 3818–3824. [Google Scholar] [PubMed]
  245. Uezu, K.; Kawakami, K.; Miyagi, K.; Kinjo, Y.; Kinjo, T.; Ishikawa, H.; Saito, A. Accumulation of γΔ 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, 7629–7634. [Google Scholar] [CrossRef] [PubMed]
  246. Nish, S.; Medzhitov, R. Host defense pathways: Role of redundancy and compensation in infectious disease phenotypes. Immunity 2011, 34, 629–636. [Google Scholar] [CrossRef] [PubMed]
  247. Rosen, L.B.; Freeman, A.F.; Yang, L.M.; Jutivorakool, K.; Olivier, K.N.; Angkasekwinai, N.; Suputtamongkol, Y.; Bennett, J.E.; Pyrgos, V.; Williamson, P.R.; et al. Anti-GM-CSF autoantibodies in patients with cryptococcal meningitis. J. Immunol. 2013, 190, 3959–3966. [Google Scholar] [CrossRef] [PubMed]
  248. Saijo, T.; Chen, J.; Chen, S.C.; Rosen, L.B.; Yi, J.; Sorrell, T.C.; Bennett, J.E.; Holland, S.M.; Browne, S.K.; Kwon-Chung, K.J. Anti-granulocyte-macrophage colony-stimulating factor autoantibodies are a risk factor for central nervous system infection by Cryptococcus gattii in otherwise immunocompetent patients. MBio 2014, 5. [Google Scholar] [CrossRef] [PubMed]
  249. Chen, S.; Yan, H.; Zhang, L.; Kong, W.; Sun, Y.; Zhang, W.; Chen, Y.; Deng, A. Cryptococcus neoformans infection and immune cell regulation in human monocytes. Cell. Physiol. Biochem. 2015, 37, 537–547. [Google Scholar] [CrossRef] [PubMed]
  250. Pelletier, S.; Gingras, S.; Green, D.R. Mouse genome engineering via CRISPR-Cas9 for study of immune function. Immunity 2015, 42, 18–27. [Google Scholar] [CrossRef] [PubMed]
  251. Heung, L.J.; Jhingran, A.; Hohl, T.M. Deploying FLAREs to visualize functional outcomes of host-pathogen encounters. PLoS Pathog. 2015, 11, e1004912. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Heung, L.J. Innate Immune Responses to Cryptococcus. J. Fungi 2017, 3, 35. https://doi.org/10.3390/jof3030035

AMA Style

Heung LJ. Innate Immune Responses to Cryptococcus. Journal of Fungi. 2017; 3(3):35. https://doi.org/10.3390/jof3030035

Chicago/Turabian Style

Heung, Lena J. 2017. "Innate Immune Responses to Cryptococcus" Journal of Fungi 3, no. 3: 35. https://doi.org/10.3390/jof3030035

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop